The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (2025)

Table of Contents
P001 AUTOLOGOUS VERSUS HAPLOIDENTICAL DONOR STEM CELL TRANSPLANTATION FOR FAVORABLE-AND INTERMEDIATE-RISK ACUTE MYELOID LEUKEMIA IN FIRST REMISSION AND UNDETECTABLE MINIMAL RESIDUAL DISEASE: A MULTI-CENTER, PROSPECTIVE STUDY Yiyang Ding1, Yuhua Ru1, Jinjin Zhu1, Xi Zhang2, Lin Liu3, Erlie Jiang4, He Huang5, Jishi Wang6, Jiong Hu7, Yanming Zhang8, Yajing Xu9, Mingzhen Yang10, Jia Chen 1, Depei Wu1 P002 OUTCOME OF ALLOGRAFTED ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS TREATED WITH THE PEDIATRIC-INSPIRED, MRD-ORIENTED GIMEMA LAL 1913 PROTOCOL IN THE REAL-LIFE: A CAMPUS ALL STUDY Gianluca Cavallaro 1, Davide Lazzarotto2, Chiara Pavoni1, Francesca Valsecchi1, Cristina Papayannidis3, Marco Cerrano4, Sabina Chiaretti5, Nicola Fracchiolla6, Fabio Giglio7, Michelina Dargenio8, Monia Lunghi9, Silvia Imbergamo10, Maria Ilaria Del Principe11, Silvia Trappolini12, Monica Fumagalli13, Patrizia Zappasodi14, Prassede Salutari15, Mario Delia16, Crescenza Pasciolla17, Federico Mosna18, Barbara Scappini19, Fabio Forghieri20, Patrizia Chiusolo21, Cristina Skert22, Benedetta Cambò23, Marzia Defina24, Giuseppe Lanzarone25, Mauro Endri26, Massimiliano Bonifacio27, Carla Mazzone28, Lidia Santoro29, Antonino Mulè30, Valentina Mancini31, Paola Minetto32, Giorgia Battipaglia33, Alessandro Cignetti34, Lara Aprile35, Robin Foà5, Anna Candoni2,20, Federico Lussana1,36 P003 FREQUENCY AND IMPACT OF SOMATIC MUTATIONS ON OUTCOMES OF ACUTE MYELOID LEUKEMIA PATIENTS RECEIVING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION: FROM THE EBMT ACUTE LEUKEMIA WORKING PARTY Ali Bazarbachi 1, Jacques-Emmanuel Galimard2, Myriam Labopin2, Iman Abou Dalle1, Jaime Sanz3, Huang He4, Jiri Mayer5, Carlos Solano6, Celestine Simand7, Laimonas Griskevicius8, Johan Maertens9, Maija Itäla-Remes10, Ain Kaare11, Maria-Pilar Gallego-Hernanz12, Gesine Bug13, Josep-Maria Ribera14, Alain Gadisseur15, Christoph Schmid16, Mi Kwon17, Xavier Poiré18, Paola Coccia19, Manuel Jurado Chacón20, Frédéric Baron21, Eolia Brissot22, Arnon Nagler23, Fabio Ciceri24, Mohamad Mohty25 P004 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR ELDERLY ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS: A STUDY FROM THE SOCIÉTÉ FRANCOPHONE DE GREFFE DE MOELLE ET THÉRAPIE CELLULAIRE (SFGM-TC) Yves Chalandon 1, Raynier Devillier2, Ariane Boumendil3, Stephanie Nguyen4, Claude-Eric Bulabois5, Patrice Ceballos6, Eolia Brissot7, Marie-Thérèse Rubio8, Hélène Labussière-Wallet9, Johan Maertens10, Patrice Chevallier11, Natacha Maillard12, Xavier Poiré13, Cristina Castilla-Llorente14, Yves Beguin15, Jérôme Cornillon16, Sébastien Maury17, Tony Marchand18, Etienne Daguindau19, Jacques-Olivier Bay20, Pascal Turlure21, Amandine Charbonnier22, Anne-Lise Menard23, Karin Bilger24, Gaelle Guillerm25, Sylvie François26, Ali Bazarbachi27, Sylvain Chantepie28, Philippe Lewalle29, Ambroise Marçais30, Michael Loschi31, Malek Benakli32, Paul Chauvet33, Edouard Forcade34, Anne Huynh35, Marie Robin36, Stavroula Masouridi-Levrat1 P005 ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION FOR OLDER PATIENTS WITH PHILADELPHIA-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA. A SURVEY BY THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT Sebastian Giebel 1, Myriam Labopin2, Ryszard Swoboda1, Didier Blaise3, Ibrahim Yakoub-Agha4, Stephanie Nguyen5, Eva Maria Wagner-Drouet6, Cristina Castilla-Llorente7, Panagiotis Kottaridis8, Thomas Schroeder9, Renato Fanin10, Jakob Passweg11, Jurjen Versluis12, Charles Crawley13, Ludovic Gabellier14, Stephan Mielke15, Xavier Poiré16, Erfan Nur17, Carlos Pinho Vaz18, Matthias Eder19, Riccardo Saccardi20, Peter Dreger21, Zinaida Peric22, Mohamad Mohty2, Fabio Ciceri23 P006 OUTCOME OF PATIENTS WITH IDH-MUTATED AML FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION Thomas Schroeder 1, Sarah Flossdorf1, Caroline Pabst2, Michael Stadler3, Johannes Schetelig4, Claudia Wehr5, Claudia Schuh6, Matthias Stelljes7, Elisa Sala8, Andreas Burchert9, Jennifer Kaivers1, Christian Reinhardt1, Nicolaus Kroeger10, Katharina Fleischhauer1, Christina Rautenberg1 P007 CD22-TARGETED IMMUNOTHERAPY FOR ADULT PATIENTS WITH RELAPSED OR REFRACTORY B-ALL WHO PREVIOUSLY EXPOSED TO CD19-TARGETED IMMUNOTHERAPY Mingming Zhang1,2, Yongxian Hu1,2, Peihua Lu3,4, Liang Huang5, Shan Fu1,2, Jingjing Feng1,2, Ruimin Hong1,2, Alex H. Chang6, He Huang 1,2 P008 COMPARISON OF THE PROGNOSTIC IMPACT BETWEEN ELN2022 AND ELN2017 RISK CLASSIFICATION IN ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS WITH AML Weihao Chen1, Yeqian Zhao1,2,3,4, Jimin Shi1,2,3,4, Yi Luo1,2,3,4, Jian Yu1,2,3,4, Yamin Tan5,6, Xiaoyu Lai1,2,3,4, Lizhen Liu1,2,3,4, Huarui Fu1,2,3,4, Yishan Ye1,2,3,4, Luxin Yang1,2,3,4, Congxiao Zhang1,2,3,4, He Huang1,2,3,4, Yanmin Zhao 1,2,3,4 P009 EFFICACY AND SAFETY OF THE THIRD-GENERATION TKI OLVEREMBATINIB IN ADULT PH + ACUTE LYMPHOBLASTIC LEUKEMIA WITH RELAPSED DISEASE OR PERSISTENT MRD BRIDGING TO ALLO-HSCT XiaoYu Zhang 1, Yanhong Zhao1, Rongli Zhang1, Erlie Jiang1 P010 INTRACELLULAR CYTOKINE ASSAYS IN COMBINATION WITH DEGRANULATION ASSAY CONTRIBUTE SIGNIFICANTLY TO DETECT AND QUANTIFY LEUKEMIA SPECIFIC IMMUNE CELLS IN AML PATIENTS’ BLOOD OR CULTURE SETTINGS Olga Schutti1, Lara Klauer2,1, Tobias Baudrexler2,1, Christoph Schmid3, Andreas Rank3, Joerg Schmohl4, Markus Hentrich5, Doris Kraemer6, Helga Schmetzer 1 P011 TOTAL MARROW AND LYMPHOID IRRADIATION IN COMBINATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE-BASED GRAFT VERSUS HOST DISEASE PROPHYLAXIS CONFERS FAVORABLE GVHD-FREE/RELAPSE-FREE SURVIVAL IN PATIENTS WITH ACUTE MYELOID LEUKEMIA Anthony Stein 1, Monzr Al Malki1, Dongyun Yang1, Joycelynne Palmer1, Ni-Chun Tsai1, Ibrahim Aldoss1, Haris Ali1, Ahmed Aribi1, Andrew Artz1, Savita Dandapani1, Len Farol1,2, Susanta Hui1, An Liu1, Ryotaro Nakamura1, Vinod Pullarkat1, Eric Radany1, Amandeep Salhotra1, James Sanchez1, Ricardo Spielberger1,2, Guido Marcucci1, Stephen Forman1, Jeffrey Wong1 P012 INCREASED MESENCHYMAL STROMAL CELLS SENESCENCE AND HIGH CXCL14 LEVELS IN BONE MARROW ARE LINKED WITH THE OCCURRENCE OF GVHD AFTER HEMATOPOIETIC STEM CELLS TRANSPLANTATION Alexandra Guelton 1, Meriem El Ouafy1, Romain Perouf1, Naceur Charif1, Simona Pagliuca1, Marie-Thérèse Rubio1, Natalia de Isla1 P013 CORRELATION OF THE OVERALL SURVIVAL IN MIXED-PHENOTYPE ACUTE LEUKAEMIA AND ACUTE LYMPHOBLASTIC LYMPHOMA WITH THE REFINED DISEASE RISK INDEX APPLIED IN AN UPPER-MIDDLE INCOME COUNTRY Naty López-Córdova 1, Jessica Meza-Liviapoma1, Jule Vasquez-Chavez1, Claudio Flores-Flores2, Shirley Quintana-Truyenque1 P014 SIGNIFICANT IMPROVEMENT OF SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN ACUTE LYMPHOBLASTIC LEUKEMIA OVER TWO DECADES – A SINGLE CENTER RETROSPECTIVE ANALYSIS Ben-Niklas Baermann1, Paula Kessler 1, Astrid Tautges2, Paul Sebastian Jäger1, Mustafa Kondakci3, Kathrin Nachtkamp1, Sascha Dietrich1, Guido Kobbe1 P015 OUTCOMES OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOLLOWING BLINATUMOMAB IN CHILDREN AND YOUNG ADULTS WITH B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA Mattia Algeri 1, Michele Massa2, Federica Galaverna2, Daria Pagliara2, Ilaria Pili2, Francesca Del Bufalo2, Marco Becilli2, Emilia Boccieri2, Roberto Carta2, Francesco Quagliarella2, Chiara Rosignoli2, Carmen Dolores De Luca2, Barbarella Lucarelli2, Pietro Merli2, Franco Locatelli3 P016 THE ROLE OF ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN CONSOLIDATION OF MINIMAL RESIDUAL DISEASE-NEGATIVE REMISSION IN RELAPSED/REFRACTORY B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA IN ADULTS AFTER PHARMACOLOGIC IMMUNOTHERAPY Kseniia Afanaseva1, Ivan Moiseev 1, Bella Ayubova1, Elena Babenko1, Ildar Barkhatov1, Tatiana Gindina1, Anna Smirnova1, Olesya Smykova1, Yulia Vlasova1, Sergey Bondarenko1, Alexander Kulagin1 P017 FLAG-IDA/VENETOCLAX IS ASSOCIATED WITH HIGHER REMISSION RATES AND IMPROVED POST-TRANSPLANT SURVIVAL COMPARED TO VENETOCLAX/AZACITIDINE IN RELAPSED/REFRACTORY AML PATIENTS Evgeny Klyuchnikov1, Anita Badbaran1, Radwan Massoud 1, Normann Steiner1, Petra Freiberger1, Franziska Modemann1, Martin Schönrock1, Sophia Cichutek1, Walter Fiedler1, Micha Peeck2, Nico Gagelmann1, Christine Wolschke1, Francis Ayuk1, Ulrike Bacher3, Nicolaus Kröger1 P018 COMPARISON OF OUTCOMES OF HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION USING POST-TRANSPLANTATION CYCLOPHOSPHAMIDE AND EX-VIVO TCRΑΒ + CD19 DOUBLE-NEGATIVE SELECTED GRAFT IN ACUTE MYELOID LEUKEMIA László Gopcsa 1, Hajnalka Andrikovics1, Alexandra Balogh1, Anikó Barta1, Judit Bogyó2, Zoltán Csukly1, Katalin Dobos1, János Dolgos1, Apor Hardi1, János Fábián1, Laura Giba-Kiss1, József Harasztdombi1, Kinga Kerner1, Ágnes Király1, Gergely Lakatos1, Viktor Lakatos1, Lilla Lengyel1, Nóra Meggyesi1, Noémi Németh1, Melinda Paksi1, Laura Regáli1, Marienn Réti1, János Sinkó1, Bálint Szabó1, Anikó Szilvási2, Éva Torbágyi1, Andrea Várkonyi1, Nikolett Wohner1, István Vályi-Nagy1, Péter Reményi1 P019 AZACITIDINE AND VENETOCLAX WITH OR WITHOUT DLI IN RELAPSED MYELOID MALIGNANCIES AFTER ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION – A RETROSPECTIVE MULTICENTER STUDY OF THE SFGM-TC Turot Mélanie 1, Loschi Michael2, Chantepie Sylvain3, Arnautou Pierre4, Poire Xavier5, Maillard Natacha6, Chalandon Yves7, El-Cheikh Jean8, Ceballos Patrice9, Devillier Raynier10, Alani Mustafa11, Fatrara Thomas12, Rubio Thérèse13, Daguindau Etienne14, Klemencie Marion15, Beauvais David16, Huynh Anne17, Marchand Tony18, Volpari Victoria19, Barette Lauren19, Pivert Cécile20, Maury Sébastien21, Suarez Felipe22, Fuseau Charline23, Lauron Sandrine24, Uzunov Madalina25, Castilla Cristina26, Forcade Edouard27, Bay Jacques-Olivier1, Ravinet Aurélie1 P020 EFFICACY AND SAFETY ANALYSIS OF PROPHYLACTIC BRENTUXIMAB VEDOTIN ADMINISTRATION AFTER PEDIATRIC ACUTE LEUKEMIA HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION Zhu Huili 1, Cao Xingyu1, Lu Yue1, Zhang Jianping1, Zhao Yanli1, Liu Deyan1, Xiong Min1, Sun Ruijuan1, Liu Hongxing1, Wei Zhijie1 P021 OUTCOMES AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR ACUTE LEUKEMIA IN YOUNG ADULTS COMPARED WITH CHILDREN, ADOLESCENTS AND ELDERLY PATIENTS IN GERMANY Jochen J. Frietsch 1,2, Sarah Flossdorf3,4, Ashrafossadat Ahmadian3,4, Claudia Schuh3, Thomas Schroeder5, Igor-Wolfgang Blau6, Matthias Stelljes7, Nicolaus Kröger3,8, Katharina Egger-Heidrich9, Matthias Eder10, Peter Dreger11, Johanna Tischer12, Eva Wagner-Drouet13, Rita Beier14,15, Peter Bader16, Martin Sauer15, Barbara Meissner14,15, Katharina Fleischhauer3,17, Inken Hilgendorf2 P022 PHASE I/II STUDY OF CYCLOPHOSPHAMIDE AND BENDAMUSTINE AS POST-TRANSPLANTATION GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS IN REFRACTORY MYELOID NEOPLASMS Ivan Moiseev 1, Olga Epifanovskaya1, Ksenia Afanasyeva1, Anastasia Beynarovich1, Dmitrii Zhogolev1, Mikhail Kanunnikov1, Yulia Rogacheva1, Tatyana Rudakova1, Nikita Volkov1, Zhamshidbek Khudaiberdiev1, Azamjon Meliboev1, Yulia Vlasova1, Elena Morozova1, Sergei Bondarenko1, Alexander Kulagin1 P023 REAL-WORLD OUTCOME OF ACUTE MYELOID LEUKEMIA PATIENTS TREATED WITH ALLOGENEIC STEM CELL TRANSPLANTATION WITHOUT STANDARD INDUCTION CHEMOTHERAPY Francesca Biavasco 1, Kristina Maas-Bauer1, Jesus Duque-Afonso1, Ralph Waesch1, Michael Luebbert1, Justus Duyster1, Robert Zeiser1, Juergen Finke1, Claudia Wehr1 P024 DURING CB TRANSPLANT IN MRD-POSITIVE AML, RELAPSE IS USUALLY EARLY, IS REDUCED BY ACUTE GVHD AND IS INFLUENCED BY CICLOSPORIN EXPOSURE: A MULTICENTER NATIONAL EXPERIENCE Srividhya Senthil 1, Abdul Moothedath1, Archana Rauthan2, Ioannis Peppas3, Sandeep Potluri4, Pamela Evans5, Valerie Broderick5, Sarah Lawson4, Emma Barrett6, Caroline Furness7, Kanchan Rao2, Robert Wynn1 P025 IMPACT OF RESPONSE TO THE FIRST INDUCTION COURSE IN AML PATIENTS ON ALLOHSCT OUTCOMES WITH PTCY Dmitrii Zhogolev1, Bella Aybova1, Anna Smirnova1, Yulia Vlasova1, Daria Chernyshova1, Elena Babenko1, Tatyana Gindina1, Ildar Barkhatov1, Elena Morozova1, Ivan Moiseev 1, Sergey Bondarenko1, Alexander Kulagin1 P026 POST-TRANSPLANT TYROSINE KINASE INHIBITOR MAINTENANCE IN PH-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA DELIVERS BENEFIT TOWARD IMPROVED OVERALL, RELAPSE-FREE, GVHD/RELAPSE-FREE SURVIVAL AND DECREASED NON-RELAPSE MORTALITY WITHOUT REDUCED RELAPSE Eshrak Al-Shaibani 1, Carol Chen1, Igor Novitzky-Basso1,2, Ivan Pasic1,2, Auro Viswabandya1,2, Rajat Kumar1,2, Wilson Lam1,2, Arjun Law1,2, Armin Gerbitz1,2, Jonas Mattson1,2, Fotios V. Michelis1,2, José-Mario Capo-chichi3, Dennis D. Kim1,2 P027 IMPACT OF DONOR AND STEM-CELL SOURCE IN ADULTS WITH HIGH-RISK PHILADELPHIA-NEGATIVE ACUTE LYMPHOBLASTIC LEUKEMIA TRANSPLANTED IN FIRST COMPLETE REMISSION: A STUDY FROM GRAALL AND SFGM-TC Matthieu Jestin1, Sébastien Maury2, Anne Huynh3, Edouard Forcade4, Ibrahim Yakoubagha5, Cristina Castilla-Llorente6, Jean-Baptiste Mear7, Urs Schanz8, Sylvain Chantepie9, Jakob Passweg10, Nathalie Contentin11, Yves Chalandon12, Nicole Raus13, Véronique Lheritier13, Natacha Maillard14, Marie-Thérèse Rubio15, Raynier Devillier16, Patrice Ceballos17, Sylvie François18, Stéphanie Nguyen-Quoc19, Patrice Chevallier20, Hélène Labussière-Wallet13, Hervé Dombret1, Nicolas Boissel1, Nathalie Dhédin 1 P028 HOW DISEASE STATUS IMPACTS ON LONG-TERM SURVIVAL AFTER ALLOGENEIC TRANSPLANT IN ACUTE MYELOID LEUKEMIA: ANALYSIS ON 456 PATIENTS Alessandro Bruno1, Matteo Giovanni Carrabba1, Simona Piemontese1, Raffaella Greco1, Sarah Marktel1, Sara Mastaglio1, Daniela Clerici1, Francesca Farina1, Elisa Diral1, Luca Vago1, Lorenzo Lazzari1, Edoardo Campodonico1, Giulia Furnari1, Alessandro Criscimanna1, Chiara Secco1, Magda Marcatti1, Consuelo Corti1, Massimo Bernardi1, Jacopo Peccatori1, Fabio Ciceri1,2, Maria Teresa Lupo-Stanghellini 1 P029 TLS::ERG FUSION GENE PREDICTS A POOR PROGNOSIS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN ACUTE MYELOID LEUKEMIA: A SINGLE CENTER STUDY Gele Tong 1, Yanli Zhao1, Jianping Zhang1, Min Xiong1, Xingyu Cao1, Deyan Liu1, Ruijuan Sun1, Zhijie Wei1, Jiarui Zhou1, Yue Lu1 P030 HIGH TOLERABILITY OF PROLONGED AZACITIDINE MAINTENANCE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH AML LACKING A TARGETABLE MUTATION Cuong An Do 1, Anne-Claire Mamez1, Amandine Pradier1,2, Sarah Morin1, Chiara Bernardi1,2, Sarah Prati-Perdikis1, Yan Beauverd1, Thomas Longval1, Maria Mappoura1, Sarah Noetzlin1, Laetitia Dubouchet1, Evgenia Laspa1, Marie Maulini1, Thien-An Tran1, Carmen de Ramon Ortiz1, Stavroula Masouridi-Levrat1, Federico Simonetta1,2, Yves Chalandon1,2, Federica Giannotti1 P031 OUTCOME OF ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN ACUTE MYELOID LEUKAEMIA FLT3 IN THE ERA OF FLT3 INHIBITORS – STUDY OF THE POLISH ADULT LEUKAEMIA GROUP (PALG) Elzbieta Patkowska1, Anna Czyz2, Alicja Sadowska-Klasa3, Andrzej Szczepaniak4, Lukasz Bolkun5, Marta Sobas2, Michal Gorka6, Edyta Subocz7, Anna Koclega8, Renata Guzicka-Kazimierczak9, Jan Zaucha3, Lidia Gil4, Marta Libura6, Agnieszka Pluta10, Dorota Bartosinska11, Ewa Lech-Maranda1, Agnieszka Wierzbowska10, Sebastian Giebel12, Barbara Nasilowska-Adamska 1 P032 CLINICAL CHARACTERISTICS AND PROGNOSIS OF ADULT T CELL ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS WITH ACTIVATING RAS MUTATIONS Mimi Xu 1,2,3, Yuqing Tu1,2,3, Mengqi Xiang1,2,3, Yi Fan1,2,3, Xiang Zhang1,2,3, Tiemei Song1,2,3, Lian Bai4, Xiaoli Li5, Yang Xu1,2,3, Yuejun Liu1,2,3, Depei Wu1,2,3 P033 PREVENTION AND TREATMENT OF ACUTE MYELOID LEUKEMIA RELAPSE FOLLOWING ALLOGENEIC STEM CELL TRANSPLANT: A NATIONAL SURVEY BY GITMO (GRUPPO ITALIANO TRAPIANTO MIDOLLO OSSEO) Francesco Saraceni1, Eliana Degrandi2, Simona Piemontese3, Francesco Saglio4, Giorgia Battipaglia 5, Marta Lisa Battista6, Patrizia Chiusolo7, Jacopo Mariotti8, Stefania Bramanti8, Anna Grassi9, Fabio Benedetti10, Manuela Tumino11, Giorgia Saporiti12, Salvatore Leotta13, Sadia Falcioni14, Martina Chiarucci15, Gladis Bortoletto16, Elisabetta Terruzzi17, Annalisa Imovilli18, Carlo Borghero19, Eugenia Piras20, Francesco Zallio21, Vincenzo Federico22, Irene Maria Cavattoni23, Andrea Gilioli24, Alessandra Picardi25, Domenico Pastore26, Anna Paola Iori27, Valentina Giudice28, Carmen Di Grazia29, Luca Castagna30, Michele Malagola31, Attilio Olivieri1, Francesca Bonifazi32, Massimo Martino33 P034 TRENDS IN THE USE OF HEMATOPOIETIC CELL TRANSPLANTATION FOR ADULTS WITH ACUTE LYMPHOBLASTIC LEUKEMIA: A REPORT FROM THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT Sebastian Giebel 1, Myriam Labopin2, Frederic Baron3, Ali Bazarbachi4, Eolia Brissot5, Gesine Bug6, Jordi Esteve7, Norbert-Claude Gorin8, Francesco Lanza9, Arnon Nagler10, Annalisa Ruggeri11, Jaime Sanz12, Bipin Savani13, Christoph Schmid14, Roni Shouval15, Alexandros Spyridonidis16, Jurjen Versluis17, Zinaida Peric18, Mohamad Mohty5, Fabio Ciceri19 P035 ALLOGENEIC STEM CELL TRANSPLANTATION FOR ADULT ACUTE LYMPHOBLASTIC LEUKEMIA - A SINGLE CENTER 25-YEARS EXPERIENCE Veronika Válková 1,2, Cyril Šálek1,2, Antonín Vítek1, Markéta Marková1,2, Ludmila Nováková1, Mariana Koubová1, Michal Kolář1, Barbora Čemusová1, Hana Bartáková1, Petr Cetkovský1,2, Jan Vydra1,2 P036 STRUCTURAL ANALYSIS OF LEUKEMIA-ASSOCIATED PROTEIN PTPN21 REVEALS A DOMINANT-NEGATIVE EFFECT OF THE FERM DOMAIN ON ITS PHOSPHATASE ACTIVITY Zijun Qian 1,2, Lu Chen1,2, Jie Zhang2, Yuyuan Zheng1, Chun Zhou1,2, Haowen Xiao1,2 P037 EARLY DETECTION OF RELAPSE AND PROMPT TREATMENT WITH DLI FACILITATED BY PROPHYLACTIC LYMPHOCYTE STORAGE PERI-TRANSPLANT RESULTS IN EFFECTIVE DISEASE CONTROL IN ACUTE LEUKAEMIA AND MDS Daire Quinn 1, Anne Parker1, Grant McQuaker1, Anne-Louise Latif1, David Irvine1, Ailsa Holroyd1, Dimitris Galopoulos1, Andrew Clark1 P038 THIOTEPA, BUSULFAN, AND FLUDARABINE CONDITIONING REGIMEN IN T-CELL REPLETE HLA-HAPLOIDENTICAL BONE MARROW TRANSPLANTATION FOR ADULTS WITH ACUTE MYELOBLASTIC LEUKEMIA IN SECOND COMPLETE REMISSION Ahmad Ibrahim 1, Ahmad Khalil2, Kamal Al Zahran3, Mohamad Hachem4, Pamela Sfeir5, Amin Abyad6, Jad Ibrahim7, Hussein Abou Abbas8, Ali Youssef8, Charbel Khalil2, Tamima Jisr9 P039 REDUCED INTENSITY CHEMOTHERAPY WITH TYROSINE KINASE INHIBITOR FOLLOWED BY ALLOGENEIC STEM CELL TRANSPLANTATION WAS EFFECTIVE IN PATIENTS WITH PHILADELPHIA CHROMOSOME POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA Dong Won Baek 1, Jung Min Lee1, Hyukjin Choi1, Joon Ho Moon1, Sang Kyun Sohn1 P040 IMPACT OF CO-MUTATIONS ON THE TRANSPLANT OUTCOMES IN AML PATIENTS WITH DNMT3A MUTATION Feng-ming Tien1, Xavier Cheng-Hong Tsai 1, Min-Yen Lo1, Hsin-An Hou1, Hwei-Fang Tien1 P041 SALVAGE TREATMENTS HAVE EXTREMELY POOR OUTCOMES FOR PATIENTS WITH RELAPSED T-ACUTE LYMPHOBLASTIC LEUKAEMIA POST TRANSPLANT Alexandros Rampotas 1, Fotios Panitsas2, Maximillian Brodermann1, Sridhar Chaganti3, Styliani Bouziana4, Emma Nicholson5, Samantha Drummond6, Sharon Allen7, Andrew King7, Henry Crosland3, Anne-Louise Latif6, Daniele Avenoso4, Claire Roddie1 P042 FEASIBILITY OF ALLOGENEIC STEM CELL TRANSPLANTATION (HSCT) IN PATIENTS WITH ACUTE MYELOID LEUKEMIA PREVIOUSLY TREATED WITH CPX-351: REPORT FROM A SINGLE CENTER Sabrina Giammarco 1, Elisabetta Metafuni1, Maria Assunta Limongiello1, Federica Sorá2, Eugenio Galli1, Filippo Frioni2, John Marra2, Patrizia Chiusolo2, Simona Sica2 P043 COMPARISON OF VENETOCLAX-BASED NON-ANTHRACYCLINE INDUCTION THERAPY VERSUS 3 + 7 FOLLOWED BY HEMATOPOIETIC STEM CELL TRANSPLANTATION IN NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA PATIENTS Tran-Der Tan 1, Lun-Wei Chiou1 P044 MYELOFIBROSIS GRADE II-III IS AN INDEPENDENT RISK FACTOR FOR RELAPSE OF ACUTE MYELOID LEUKEMIA AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Haixiao Zhang1, Erlie Jiang 2 P045 INCIDENCE AND MANAGEMENT OF ADULT ACUTE LYMPHOBLASTIC LEUKAEMIA RELAPSE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN LIMITED RESOURCE COUNTRIES Rihab Ouerghi1, Nour Ben Abdeljelil 1, Sabrine Mekni1, Insaf Ben Yaiche1, Ines Turki1, Rim Dachraoui1, Dorra Belloumi1, Lamia Torjemane1, Rimmel Kanoun1, Saloua Ladeb1, Tarek Ben Othman1 P046 IMPACT OF BODY MASS INDEX ON OUTCOME IN ADULT PATIENTS WITH ACUTE MYELOID AND ACUTE LYMPHOBLASTIC LEUKEMIA UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Iveta Oravcova1,2, Zuzana Rusinakova1, Eva Mikuskova1, Miriam Ladicka1, Ludmila Demitrovicova1, Alica Slobodova1, Vanda Mikudova1, Jana Spanikova1, Radka Vasickova1, Denis Urban1, Kristina Bandurova1, Lubos Drgona1,2, Silvia Cingelova 1 P047 VENETOCLAX COMBINED WITH HMAS-BASED THERAPY AS PRE-EMPTIVE TREATMENT FOR ACUTE MYELOID LEUKEMIA PATIENTS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Mimi Xu 1,2,3, Yuqing Tu1,2,3, Mengqi Xiang1,2,3, Yi Fan1,2,3, Xiang Zhang1,2,3, Tiemei Song1,2,3, Lian Bai4, Xiaoli Li5, Yang Xu1,2,3, Yuejun Liu1,2,3, Jia Chen1,2,3, Depei Wu1,2,3 P048 VENETOCLAX, AZACITIDINE, COMBINED WITH LOW-DOSE CYTARABINE IMPROVE THE REMISSION RATE IN OLDER OR UNFIT PATIENTS WITH NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA Xiao Han1, Qingxiao Song 1, Kai Wan1, Mengyun Zhang1, Xue Liu1, Hongju Yan1, Cheng Zhang1, Qin Wen1, Xi Zhang1 P049 TARGET THERAPY IN ACUTE LEUKAEMIA RELAPSES AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Riccardo Boncompagni 1, Chiara Camerini1, Ilaria Cutini1, Mirella Giordano1, Antonella Gozzini1, Chiara Nozzoli1, Edoardo Simonetti1, Riccardo Saccardi1 P050 ALLOGENEIC STEM CELL TRANSPLANTATION IN THE MANAGEMENT OF ADULT ACUTE LYMPHOBLASTIC LEUKAEMIA: A TEN YEAR EXPERIENCE IN IRELAND’S NATIONAL ADULT ALLOGENEIC TRANSPLANT CENTRE Conor Browne 1, Micheal Brennan1, Mairead Ni Chonghaile1, Sarah Maher1, Greg Lee1, Deirdre Waldron1, Nicola Gardiner1, Michelle Regan1, Lorraine Brennan1, Carmel Waldron1, Catherine M. Flynn1, Eibhlin Conneally1, Patrick J. Hayden1, Robert Henderson1, Elisabeth Vandenberghe1, Christopher L. Bacon1 P051 FLT3-MUTATED AML: MANAGEMENT AND LONG-TERM OUTCOME OVER 24 YEARS AT A SINGLE CENTER Saveria Capria 1, Silvia Maria Trisolini1, Lorenzo Torrieri2, Elena Amabile2, Giovanni Marsili3, Alfonso Piciocchi3, Walter Barberi1, Daniela Diverio1, Daniela Carmini1, Massimo Breccia2, Maurizio Martelli2, Clara Minotti1 P052 THE ROLE OF TOLEROGENIC DENDRITIC CELLS IN SUPPRESSING AND DCLEU IN INCREASING ANTILEUKEMIC CYTOTOXICITY A.S. Hartz1, L. Li1, H. Aslan1, E. Pepeldjiyska1, E. Rackl1, T. Baudrexler1, P. Bojko2, J. Schmohl3, A. Rank4, C. Schmid4, H.M. Schmetzer 1 P053 TREATMENT EFFICACY OF BLINATUMOMAB FOR MAINTENANCE THERAPY OF PATIENTS WITH B-LINEAGE ACUTE LYMPHOBLASTIC LEUKEMIA RECEIVING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION Jiayu Huang1, Luxiang Wang1, Chuanhe Jiang1, Zilu Zhang1, Zengkai Pan1, Ling Wang2, Jieling Jiang2, Jiong Hu2, Jun Zhu3, Lijing Shen4, Suning Chen5, Yang Cao6, Xiaoxia Hu 1 P054 EFFICACY, SAFETY AND TOLERABILITY OF MITOXANTRONE HYDROCHLORIDE LIPOSOME CONTAINING REGIMEN IN TREATING RELAPSED ACUTE MYELOID LEUKEMIA AFTER ALLOGENIC STEM CELL TRANSPLANTATION Li Liu 1, Yigeng Cao1, Ni Lu1, Erlie Jiang1 P055 STUDY ON THE EFFICACY AND SAFETY OF BLINATUMOMAB MAINTENANCE THERAPY OF HIGH RISK PH NEGATIVE ALL AFTER ALLO-HSCT Zhang Jianping1, Li Nannan1 P056 THE IMPACT OF PRE-TRANSPLANT MDS-ASSOCIATED SOMATIC MUTATIONS AND CO-MUTATIONS IN PATIENTS WITH AML ON POST-TRANSPLANT OUTCOMES Khalid Halahleh 1, Ayat Taqash1, Isra Muradi2, Farah Alul1, Osama Alsmadi1, Mohamad Ma’koseh1 P057 TRIALS IN PROGRESS: A PHASE 1B SINGLE-ARM, OPEN-LABEL STUDY OF EMAVUSERTIB IN COMBINATION WITH AZACITIDINE AND VENETOCLAX IN AML PATIENTS IN COMPLETE RESPONSE WITH MRd Adolfo de la Fuente 1, Claudio Cerchione2, Sebastian Scholl3, Jan Moritz Middeke4, Gaurav S. Choudhary5, Dora Ferrari5, Reinhard von Roemeling5, Uwe Platzbecker6 P058 PREVALENCE OF ELIGIBILITY FOR GERMLINE MUTATION TESTING IN PATIENTS WITH MDS/AML UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT IN A US ACADEMIC MEDICAL CENTER Tyler Fugere 1, Samuil Ivanovsky1, Gomathy Nageswaran1, Brad Fugere1, Urooj Hudda1, Sravani Gundarlapalli1, Lakshmi Yarlagadda1, Zhongning “Jim” Chen1, Cesar Gentille1 P059 REFINED DISEASE RISK INDEX APPLIED IN AN UPPER-MIDDLE INCOME COUNTRY AS A TOOL TO ESTIMATE OVERALL SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN HAEMATOLOGICAL MALIGNANCIES Shirley Quintana-Truyenque1, Naty López-Córdova 1, Jessica Meza-Liviapoma1, Lourdes López-Chavez1, Cindy Alcarraz-Molina1, Victor Mallma-Soto1, Claudio Flores-Flores2, Jule Vasquez-Chavez1 P060 PREEMPTIVE DONOR LYMPHOCYTE INFUSION AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN PATIENTS WITH ACUTE MYELOID LEUKEMIA – SINGLE CENTRE ANALYSIS OF EFFECTIVENESS AND SAFETY Anna Łojko Dankowska 1, Ewa Bembnista1, Paula Matuszak1, Magdalena Matuszak1, Bartosz Małecki1, Andrzej Szczepaniak1, Anna Wache1, Dominik Dytfeld1, Lidia Gil1 P061 IN CHILDREN WITH RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA GIVEN ALLOGENEIC-HSCT EMPLOYING REDUCED-INTENSITY CONDITIONING, GVHD IS LESS FREQUENT IF THE PROCEDURE IS CONDUCTED ON AN OUTPATIENT BASIS Edgar Jared Hernández-Flores1, Moisés Manuel Gallardo-Pérez2, Max Robles-Nasta2, Daniela Sánchez-Bonilla2, Merittzel Abigail Montes-Robles3, Guillermo Ocaña-Ramm1, Olivia Lira-Lara4, Juan Carlos Olivares-Gazca2, David Gómez-Almaguer5, Óscar González Llano5, Yajaira Valentine Jiménez-Antolinez5, Guillermo José Ruiz-Delgado 2,1, Guillermo José Ruiz-Arguelles2,1 P062 IMPLEMENTATION OF NON-TBI CONDITIONING AND POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR PEDIATRIC ACUTE LYMPHOBLASTIC LEUKEMIA IN RESOURCE-LIMITED SETTINGS Lusine Krmoyan 1,2, Inga Khalatiani1,2, Mane Gizhlaryan1, Karen Meliksetyan1 P063 RELAPSE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENT WITH ACUTE MYELOBLASTIC LEUKEMIA Insaf Ben Yaiche1, Nour Ben Abdejelil 1, Rihab Ouerghi1, Ines Turki1, Sabrine Mekni1, Lamia Torjemane1, Dorra Belloumi1, Rimmel Kanoun1, Saloua Ladab1, Tarek Ben Othman1 P064 IAMP21 ACUTE LYMPHOBLASTIC LEUKEMIA WITH DELETION AROUND SUBTELOMERIC REGION OF CHROMOSOME 21: REPORT OF ONE CASE AND REVIEW OF LITERATURE Jiantuo Liu 1, Hongrui Li1, Xiangjun Chen2, Yanli He2 P065 RESULTS OF HSCT WITH TCRΑΒ AND CD19-DEPLETION FROM MATCHED RELATED DONORS AND INFUSIONS OF CD45RA DEPLETED DONOR LYMPHOCYTES IN PEDIATRIC SEVERE APLASTIC ANEMIA Daria Shasheleva1, Larisa Shelikhova1, Anna Bogoyavlenskaya1, Rimma Khismatullina1, Svetlana Radygina1, Dmitriy Balashov1, Yakov Muzalevsky1, Tatiana Salimova1, Dina Baidildina1, Elena Kurnikova1, Dmitriy Pershin 1, Pavel Trakhtman1, Galina Novichkova1, Alexei Maschan1, Michael Maschan1 P066 MITOCHONDRIAL METABOLISM IN FANCONI ANEMIA PATIENTS POST HEMATOPOIETIC STEM CELL TRANSPLANTATION Filomena Pierri1, Enrico Cappelli1, Stefano Regis1, Silvia Ravera2, Federica Grilli1, Gianluca Dell’Orso1, Stefano Giardino1, Maura Faraci 1 P067 ATLG HAS BETTER SAFETY THAN RATG IN THE TREATMENT OF ADULT AA WITH ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Zhang Jianping 1, Lu Yue1, Zhao Yanli1, Xiong Min1, Liu Deyan1, Cao Xingyu1, Wei Zhijie1, Sun Ruijuan1, Zhou Jiarui1 P068 TREATMENT WITH DARATUMUMAB IN 4 PATIENTS WITH IMMUNE ANEMIA AFTER HSCT WITH ABO INCOMPATIBILITY OR OTHER IMMUNE-ERYTHROCYTE DISCREPANCIES Marina Aranguren Ostolaza1, Candela Ceballos Bolaños 1, Itziar Oiartzabal Ormategui1, Nerea Arratibel Zalacain1, Pamela Millacoy Austenrritt1, Mónica Fernández Pérez1, Maria Cruz Viguria Alegria1, Carlos Manuel Panizo Santos1, Irene Sánchez Prieto1, Monserrat Lozano Lobato1 P069 UPFRONT T-REPLETE HAPLO-IDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR CHILDREN WITH SEVERE APLASTIC ANEMIA: A SINGLE CENTER EXPERIENCE FROM JORDAN Ayad Ahmed Hussein 1, Nour Awni Ghanem1, Bayan Majed Alaaraj1, Tariq Ahmed Abdelghani1, Dina Mohammad Abu Assab1, Hadeel Hassan Al-Zoubi1 P070 PERIPHERAL BLOOD AS GRAFT SOURCE FOR APLASTIC ANEMIA TRANSPLANT: OUTCOMES OF YOUNG ADULTS AND ADULTS IN A MEXICAN PUBLIC HOSPITAL Guillermo Sotomayor Duque 1, Severiano Baltazar Arellano1, Roxana Saldaña Vazquez1, Humberto Guerra Ramos1, Raul Ramos1, Alma Fabiola Alvarado Charles1, Karen Machuca Adame1, Luis Omar Gudiño Cobos1, Victor Valerio Bugarin1, Rosa Elva De leon Cantú1, Roberto Hernandez Valdez1, Jose Marcelino Chavez Garcia1 P071 UPFRONT HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) IN CLASSICAL PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH) Lorie Gandhi 1, Col (Dr) Rajiv Kumar1, Brig(Dr) Rajan Kapoor1 P072 A REAL-WORLD ANALYSIS OF TREATMENT ADHERENCE AND CLINICAL OUTCOMES FOR PATIENTS WITH PAROXYSMAL NOCTURNAL HAEMOGLOBINURIA (PNH) RECEIVING PEGCETACOPLAN Koo Wilson 1, Carly Rich1, Zalmai Hakimi1, Regina Horneff1, Jesse Fishman2, Jennifer Mellor3, Lucy Earl3, Yasmin Taylor3, Alice Simons3 P073 EXPERIENCE OF SALVAGE HAPLO-HSCT WITH PTCY FOR ACQUIRED SEVERE APLASTIC ANEMIA Yuliya Mareika1, Nina Minakovskaya1, Natalia Kirsanava 1, Dmitriy Prudnikov1, Mariya Naumovich1, Lubov Zherko1, Aliaksei Kakunin1, Aliaksei Lipnitski1, Volha Mishkova1, Katsiaryna Vashkevich1, Anzhalika Solntsava1 P074 THYMIC SIZE LONGITUDINAL CHANGES ON CHEST HIGH-RESOLUTION COMPUTED TOMOGRAPHY AFTER AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR DIFFUSE CUTANEOUS SYSTEMIC SCLEROSIS Gregory Pugnet 1, Samia Collot1, Antoine Petermann1, Pauline Lansiaux2, Gwenaëlle Lorillon3, Nassim Ait Abdallah2, Mathieu Resche-Rigon4, Cécile Borel1, Zora Marjanovic5, Dominique Farge2 P075 LONG TERM PERSISTENCE OF T MEMORY STEM CELLS FOLLOWING AUTOLOGOUS HAEMATOPOIETIC STEM CELL TRANSPLANTATION FOR MULTIPLE SCLEROSIS Melissa Khoo1,2, Carole Ford1, Jennifer Massey1,2,3, Kevin Hendrawan4, Malini Visweswaran1,2, John Zaunders1,2, Ian Sutton3, Barbara Withers3, David Ma1,2,3, John Moore 1,2,3 P076 EFFECTS OF HIGH DOSE IMMUNOSUPPRESSIVE THERAPY WITH AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN MULTIPLE SCLEROSIS FROM PATIENT’S PERSPCTIVE: LONG-TERM QUALITY OF LIFE OUTCOMES Denis Fedorenko 1, Vladimir Melnichenko1, Anatoly Rukavitsin1, Nikolai Vasilev1, Tatiana Nikitina2, Natalia Porfirieva3, Tatiana Ionova2 P077 THE MATHEC-SFGM-TC REGISTRY FOR CELL THERAPY IN AUTOIMMUNE DISEASES: A DEDICATED TOOL FOR REAL-WORLD DATA COLLECTION Pauline Lansiaux1, Manuela Badoglio2, Grégory Pugnet3, Mathieu Puyade4, Emmanuel Chatelus5, Thierry Martin5, Louis Terriou6, Alexandre Maria7, Marc Ruivard8, Jacques-Olivier Bay8, Bertrand Dunogué9, Matthieu Allez1, Hélène Zéphir10, Arsène Mékinian11, Eric Deconinck12, Sabine Berthier13, Françoise Sarrot-Reynaud14, Frédéric Garban14, Nicolas Maubeuge4, Guillaume Mathey15, Cristina Castilla-Llorente16, Céline Labeyrie17, Régis Peffault de la Tour18, Marie Robin19, Zora Marjanovic11, Dominique Farge 1 P078 ATG AND OTHER SEROTHERAPY IN CONDITIONING REGIMENS FOR ASCT IN AUTOIMMUNE DISEASES: A SURVEY OF THE EBMT AUTOIMMUNE DISEASES WORKING PARTY (ADWP) Azza Ismail1, Rosamaria Nitti 2, Basil Sharrack1, Manuela Badoglio3, Pascale Ambron3, Myriam Labopin3, Tobias Alexander4, John Snowden5, Greco Raffaella2 P079 ADOPTING A STANDARD OF CARE: AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR SCLERODERMA Ernesto Ayala 1, Madiha Iqbal1, Hemant Murthy1, James Foran1, Vivek Roy1, Mohamed Kharfan-Dabaja1 P080 FLUDARABINE AND CYCLOPHOSPHAMIDE AS A SAFE AND EFFECTIVE LYMPHOABLATIVE CONDITIONING REGIMEN FOR MULTIPLE SCLEROSIS (MS) Denis Fedorenko 1, Anatoly Rukavitsin1, Nikolai Vasilev1, Vladimir Melnichenko1, Tatiana Ionova2 P081 IMMUNE RESET AND GRAFT COMPOSITION IN AUTOLOGOUS TRANSPLANTATION FOR MULTIPLE SCLEROSIS: EARLY RECOVERY OF NATURAL KILLER CELLS MAY BE IMPORTANT IN EFFECTING DURABLE RESPONSES Latoya Reid 1, Oliver Gittner2, Malia Begley3, Rowayda Peters3, Andy Drake3, Matthias Klammer3 P082 REDUCED PRO-INFLAMMATORY INTERLEUKINS IL-6 AND IL-8 SECRETED BY SKIN FIBROBLASTS IN SYSTEMIC SCLEROSIS PATIENTS AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION Gunter Assmann 1,2, Jan Weghorn1, Michael Schmidt1, Joerg Henes3, Claudia Pfoehler4, Frank Neumann2 P083 THE QUALITY OF LIFE OF PERSONS WITH MULTIPLE SCLEROSIS IMPROVES AFTER AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION Olivia Lira-Lara1, Moisés Manuel Gallardo-Pérez2, Miranda Melgar-de-la-Paz3, Paola Negrete-Rodríguez4, Luis Enrique Hamilton-Avilés5, Guillermo Ocaña-Ramm5, Max Robles-Nasta2, Daniela Sánchez-Bonilla2, Juan Carlos Olivares-Gazca2, Guillermo José Ruiz-Delgado 2,5, Guillermo José Ruiz-Arguelles2,5 P084 AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN THE TREATMENT OF MULTI-REFRACTORY STIFF PERSON SYNDROME: A CASE STUDY Tamim Alsuliman 1, Dimitri Psimaras2, Nicolas Stocker1, Simona Sestili1, Anne Banet1, Zoé Van de Wyngaert1, Agnès Bonnin1, Manuela Badoglio3, Mathieu Puyade4,5, Dominique Farge6,5, Mohamad Mohty1, Zora Marjanovic1,5 P085 BRIDGING THERAPY PRIOR TO TREATMENT WITH ANTI-CD19 CAR T CELLS FOR RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA IN CHILDREN AND YOUNG ADULTS - A MULTINATIONAL RETROSPECTIVE ANALYSIS Maike Breidenbach1, Peter Bader2, Andishe Attarbaschi3, Claudia Rossig4, Roland Meisel5, Markus Metzler6, Marion Subklewe1, Fabian Mueller7, Paul-Gerhard Schlegel8, Irene Teichert von Lüttichau9, Jean-Pierre Bourquin10, Gabriele Escherich11, Gunnar Cario12, Peter Lang13, Ramona Krauss1, Arend von Stackelberg14, Semjon Willier15, Christina Peters3, Tobias Feuchtinger 16,1 P086 SAFETY AND EFFICACY COMPARISON OF HUMANIZED CD19 CAR-T VERSUS BLINATUMOMAB THERAPY FOR RELAPSED/REFRACTORY B-ALL PATIENTS Kexin Wang 1,2,3,4, Songfu Jiang5, Yongxian Hu1,2,3,4, He Huang1,2,3,4 P087 PREDICTIVE VALUE OF PRE-TREATMENT CIRCULATING TUMOR DNA GENOMIC LANDSCAPE IN PATIENTS WITH R/RMM UNDERGOING ANTI-BCMA CAR-T THERAPY: INSIGHTS FROM TUMOR CELLS AND T CELLS Rongrong Chen 1, Chunxiang Jin1, Tingting Yang1, Kai Liu1, Mengyu Zhao1, Mingming Zhang1, Pingnan Xiao1, Jingjing Feng1, Ruimin Hong1, Shan Fu1, Jiazhen Cui1, Simao Huang1, Guoqing Wei1, He Huang1, Yongxian Hu1 P088 FINAL RESULTS OF PROSPECTIVE CLINICAL TRIAL EVALUATING OUTPATIENT ADMINISTRATION OF AXICABTAGENE CILOLEUCEL IN HIGH-GRADE B CELL LYMPHOMA Bhagirathbhai Dholaria1, Shakthi Bhaskar1, Vivek Patel1, Eden Biltibo1, Salyka Sengsayadeth1, Andrew Jallouk1, James Jerkins1, Brittney Baer1, Nur Ali1, David Morgan1, Muhamed Baljevic1, Bipin Savani1, Adetola Kassim1, Olalekan Oluwole 1 P089 EUPLAGIA-1: SEVEN-DAY VEIN-TO-VEIN POINT-OF-CARE MANUFACTURED GLPG5201 ANTI-CD19 CAR-T CELLS DISPLAY EARLY PHENOTYPES IN RELAPSED/REFRACTORY CLL INCLUDING RT Esmée P. Hoefsmit 1, Sandra Blum2, Claire Vennin1, Kirsten Van Hoorde3, Sergi Betriu4, Leticia Alserawan4, Julio Delgado4, Nadia Verbruggen5, Anna D.D. van Muyden1, Henriëtte Rozema1, Ruiz Astigarraga1, Margot J. Pont1 P090 INCIDENCES AND FACTORS ASSOCIATED WITH EARLY HEMATOTOXICITY AFTER CAR T-CELL THERAPY ASSESSED BY EHA/EBMT ICAHT CRITERIA Emily Liang 1,2, Aya Albittar1, Andrew Portuguese1,2, Jennifer Huang1,2, Natalie Wuliji1,2, Qian Wu1, Joseph De Los Reyes1,2, Nikki Pin1, Aiko Torkelson1, Delaney Kirchmeier1, Abigail Chutnik1, Barbara Pender1, Joshua Hill1,2, Noam Kopmar1,2, Rahul Banerjee1,2, Andrew Cowan1,2, Damian Green1,2, Ajay Gopal1,2, Christina Poh1,2, Mazyar Shadman1,2, Alexandre Hirayama1,2, Brian Till1,2, Erik Kimble1,2, Lorenzo Iovino1,2, Aude Chapuis1,2, Folashade Otegbeye1,2, Ryan Cassaday1,2, Filippo Milano1,2, Cameron Turtle3, David Maloney1,2, Jordan Gauthier1,2 P091 COMPARISON OF IMMUNOPHENOTYPIC AND FUNCTIONAL PROPERTIES OF ANTI-CD19 CAR-T CELL PRODUCTS MANUFACTURED USING CLINIMACS PRODIGY AND G-REX PLATFORMS Ekaterina Malakhova1, Dmitriy Pershin 1, Viktoria Vedmedskaia1, Mariia Fadeeva1, Elena Kulakovskaya1, Oyuna Lodoeva1, Tatiana Sozonova1, Elvira Musaeva1, Yakov Muzalevskii1, Alexei Kazachenok1, Vladislav Belchikov1, Anastasia Melkova1, Larisa Shelikhova1, Olga Molostova1, Michael Maschan1 P092 ALLOGENEIC HSCT AFTER CAR T-CELL THERAPY HAD DELAYED PLATELET ENGRAFTMENT, HIGHER RISK OF CYTOMEGALOVIRUS VIRUS VIREMIA AND THROMBOTIC MICROANGIOPATHY COMPARED TO CHEMOTHERAPY Luxin Yang 1, Xiaoyu Lai1, Lizhen Liu1, Jimin Shi1, Yanmin Zhao1, Jian Yu1, Huarui Fu1, Yongxian Hu1, Mingming Zhang1, He Huang1, Yi Luo1 P093 AUTOMATED GRADING OF IMMUNE EFFECTOR CELL-ASSOCIATED HEMATOTOXICITY Emily Liang 1,2, Kai Rejeski3, Sandeep Raj3, Aya Albittar1, Jennifer Huang1,2, Andrew Portuguese1,2, Natalie Wuliji1,2, Qian Wu1, Aiko Torkelson1,2, Delaney Kirchmeier1,2, Abigail Chutnik1, Barbara Pender1, Joshua Hill1,2,2, Noam Kopmar1,2, Rahul Banerjee1,2, Andrew Cowan1,2, Christina Poh1,2, Mazyar Shadman1,2, Alexandre Hirayama1,2, Erik Kimble1,2, Lorenzo Iovino1,2, Roni Shouval3, Jordan Gauthier1,2 P094 RELEVANT STUDIES ON CYTOKINES LEVEL AND CAR-T EXPANSION ASSOCIATED TO CD7 CAR-T THERAPY Dongchu Wang1, Hui Wang1, Man Chen 1 P095 AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION COMBINED WITH CAR-T CELL THERAPY SIGNIFICANTLY IMPROVES PROGRESSION-FREE SURVIVAL IN RELAPSED REFRACTORY CENTRAL NERVOUS SYSTEM LYMPHOMA Rui Liu 1, Fan Yang1, Fei Xue1, Zhonghua Fu1, Yuelu Guo1, Shaomei Feng1, Peihao Zheng1, Lixia Ma1, Hui Shi1, Biping Deng1, Xiaoyan Ke1, Kai Hu1 P096 SAFETY AND EFFICACY OF BENDAMUSTINE VERSUS FLUDARABINE-CYCLOPHOSPHAMIDE LYMPHODEPLETION PRIOR TO CAR-T CELL THERAPY WITH AXICABTAGENE CILOLEUCEL FOR NON-HODGKIN LYMPHOMA: A SINGLE-INSTITUTION STUDY Anthony Stack 1, Yuliya Shestovska1, Rachel Thomas2, Matthew Hamby1, Asya Varshavsky-Yanovsky1, Peter Abdelmessieh1, Michael Styler1, Henry Fung1, Rashmi Khanal1 P097 PSEUDOPROGRESSION: A FREQUENT AND CLINICALLY RELEVANT ADVERSE EVENT OF CHIMERIC ANTIGEN RECEPTOR T CELL THERAPY Torsten Schroeder 1, Tjark Martens1, Guranda Chitadze1, Fatih Yalcin1, Heiko Trautmann1, Britta Kehden1, Monika Brüggemann1, Christine Heinen1, Philipp Nakov,1, Nicolas Spath1, Lukas Sprenger1, Anca-Maria Albici1, Laura-Jane Kramp1, Matthias Ritgen1, Dominique Wellnitz1, Sebastian Lippross1, Maciej Simon1, Ingram Iaccarino Idelson1, Nico Gagelmann2, Wolfram Klapper1, Lars Fransecky1, Thomas Valerius1, Natalie Schub1, Christiane Pott1, Katja Weisel2, Winfried Alsdorf2, Fabian Müller3, Claudia Baldus1, Friedrich Stölzel1 P098 CAR-T CELL TREATMENT OF LARGE B-CELL LYMPHOMA (LBCL): DO PROGRAM DURATION AND TREATMENT LINE MATTER? Maria-Luisa Schubert 1, Michael Schmitt1, Andrea Bondong1, Mandy Wegner1, Simon Renders1, Isabelle Krämer1, Carsten Müller-Tidow1, Peter Dreger1 P099 REAL-WORLD EXPERIENCE OF CAR T-CELL THERAPY: A SINGLE CENTRE EXPERIENCE Francys Lopez Llanos1, Inês Rocha2, Gilda Teixeira1, Isabelina Ferreira1, Pedro Sousa1, Maria João Gutierrez1, Susana Pereira 1, Ana Carolina Freitas1, Carla Espadinha1, Nuno Miranda1 P100 IMPACT OF ACUTE KIDNEY INJURY IN LYMPHOMA PATIENTS TREATED WITH AXICABTAGENE CILOLEUCEL Cristina Soler1, Rafael Hernani 1, Ariadna Pérez1, Jose Luis Piñana1, Juan Carlos Hernández-Boluda1, Ana Benzaquen1, Rosa Goterris1, Montse Gómez1, Paula Amat1, Blanca Ferrer1, Francesc Moncho1, Isidro Torregrosa1, Jose Luis Górriz1, María José Terol1, Carlos Solano1 P101 CHARACTERISTICS OF CENTRAL NERVOUS SYSTEM RELAPSE AFTER CD19 CAR-T CELL THERAPY IN R/R NHL Mengyu Zhao 1, Tingting Yang1, YeTian Dong1, Jiazhen Cui1, Delin Kong1, Wenfa Huang2, Lei Wang2, Lixin Wang2, He Huang1, Yongxian Hu1 P102 DAY7-CART COUNT ENHANCES PREDICTIVE POWER OF PET30 AND POSSIBLY ANTICIPATES CD19 LOSS IN RELAPSE AFTER CART Fabian Müller 1,2, Viktoria Blumenberg3,2, Veit L. Bücklein3,2, Ralph A. Bundschuh4,2, Dennis Harrer5,2, Klaus Hirschbühl4,2, Johannes Jung6,2, Wolfgang Kunz3,2, Igor Yakushev7,2, Anna Lena Illert6,2, Simon Völkl1,2, Rainer Claus4,2, Leo Hansmann5,2, Judith Hecker6,2, Dirk Hellwig8,2, Andreas Mackensen1,2, Marion Subklewe3,2, Michael Beck1,2 P103 EXCELLENT OUTCOME OF CAR-T CELL THERAPY IN CHILDREN WITH PB-ALL RELAPSING POST HSCT, WHO HAD VERY LOW MRD AT LYMPHODEPLETING CHEMOTHERAPY – POLISH CENTRES’ EXPERIENCE Karolina Liszka 1, Paweł Marschollek1, Iwona Dachowska - Kałwak1, Monika Mielcarek-Siedziuk1, Jowita Frączkiewicz1, Anna Panasiuk1, Igor Olejnik1, Natalia Haze1, Monika Richert-Przygońska2, Krzysztof Czyżewski2, Robert Dębski2, Jan Styczyński2, Krzysztof Kalwak1 P104 THE DARK SIDE OF CORTICOSTEROIDS IN CD19. CAR-T CELL THERAPY Marcello Roberto 1,2, Francesco Iannotta1, Francesco De Felice1,2, Beatrice Casadei1, Gianluca Storci1, Serena De Matteis1, Noemi Laprovitera1, Francesca Vaglio1, Barbara Sinigaglia1, Enrica Tomassini1, Enrico Maffini1, Mario Arpinati1, Francesco Barbato1,2, Margherita Ursi1,2, Salvatore Nicola Bertuccio1, Daria Messelodi1, Irene Salamon1, Maria Naddeo1, Elisa Dan1, Luca Zazzeroni1, Cinzia Pellegrini1, Matteo Carella1,2, Marianna Gentilini2,1, Pier Luigi Zinzani1,2, Massimiliano Bonafè1,2, Francesca Bonifazi1 P105 MATCHING-ADJUSTED INDIRECT COMPARISON OF EFFECTIVE CHARACTERISTICS AMONG DIFFERENT BCMA TARGETING CAR-T IN TREATMENT OF RELAPSED OR REFRACTORY MULTIPLE MYELOMAS Chunrui Li 1, Lu-Gui Qiu2, Di Wang1, Yongping Song3, He Huang4, Jianyong Li5, Bing Chen6, Jing Liu7, Xi Zhang8, Yu-Jun Dong9, Kai Hu10, Peng Liu11, Jian-Qing Mi12, Kaiyang Ding13, Zhenyu Li14, Qi’e Dong15, Fuyuan Zhang15, Guang Hu15, Wen Wang15 P106 IMPACT OF EARLY CAR– CD4+ T-LYMPHOCYTES RECOVERY FOLLOWING CAR-T CELL INFUSION FOR RELAPSED OR REFRACTORY B CELL LYMPHOMAS Massimiliano Gambella 1,2, Simona Carlomagno2, Rosa Mangerini1, Nicoletta Colombo1, Alessia Parodi1, Chiara Ghiggi1, Livia Giannoni1, Elisa Coviello1, Chiara Setti2, Silvia Luchetti1, Alberto Serio1, Antonella Laudisi1, Monica Passannante1, Alessandra Bo1, Elisabetta Tedone1, Simona Sivori2, Emanuele Angelucci1, Anna Maria Raiola1 P107 ACUTE INFECTION AND BASELINE C-REACTIVE PROTEIN – KEY MARKERS OF PROGRESSION IN RELAPSED REFRACTORY DLBCL PATIENTS RECEIVING CAR-T CELL THERAPY Jayachandran Perumal Kalaiyarasi 1, Khalil Begg1, Luminita Keating1, Louise Corbett1, Paul Boraks1, Brendan O’Sullivan1, Andrew King1, Charles Crawley1, Benjamin Uttenthal1, Ram Malladi1 P108 THIRD-LINE CAR-T IN PATIENTS WITH EARLY RELAPSE FOLLOWING FIRST-LINE TREATMENT FOR AGGRESSIVE B-CELL LYMPHOMA: STILL A VALUABLE OPTION Massimiliano Gambella 1, Anna Maria Raiola1, Livia Giannoni1, Elisa Coviello1, Chiara Ghiggi1, Anna Ghiso1, Riccardo Varaldo1, Stefania Bregante1, Carmen Di Grazia1, Alida Dominietto1, Antonella Laudisi1, Monica Passannante1, Silvia Luchetti1, Alberto Serio1, Alessandra Bo1, Emanuele Angelucci1 P109 EASIX SCORE AS A PROGNOSTIC FACTOR OF CYTOKINE RELEASE SYNDROME IN CAR-T PATIENTS - A RETROSPECTIVE ANALYSIS OF MM AND LBCL COHORTS Jaromir Tomasik 1, Batia Avni2, Sigal Grisariu3, Shlomo Elias3, Eran Zimran3, Polina Stepensky3, Grzegorz Basak1 P110 HEMATOTOXICITY POST CAR-T CELL THERAPY: REAL-WORLD DATA ON RISK FACTORS AND OUTCOMES Zoi Bousiou 1, Ioannis Kyriakou1, Anna Vardi1, Despoina Mallouri1, Eleni Gavriilaki1, Christos Demosthenous1, Nikolaos Spyridis1, Alkistis-Kyra Panteliadou1, Eleni Papchianou1, Georgios Karavalakis1, Evangelia Yannaki1, Ioannis Batsis1, Ioanna Sakellari1 P111 DONOR-DERIVED CLL-1 CHIMERIC ANTIGEN RECEPTOR T-CELLS FOR RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA BRIDGING TO ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A CASE REPORT UPDATE Xiaojuan Miao1, Yanrong Shuai1, Ying Han1, Yilan Liu1, Nan Zhang1, Hao Yao1, Xiao Wang1, Guangcui He 1, Dan Chen1, Fangyi Fan1, Alex H. Chang2, Yi Su1, Hai Yi1 P112 A RETROSPECTIVE REVIEW TO IDENTIFY OPTIMAL LYMPHODEPLETION PRIOR TO TISAGENLECLEUCIL IN PATIENTS WITH REFRACTORY OR RELAPSED LARGE B-CELL LYMPHOMA IN SCOTLAND’S NATIONAL CAR-T CENTRE Daire Quinn 1, Jennifer McLellan1, Sinead Connolly1, Grant McQuaker1, Anne Parker1, Dimitris Galopoulos1, Ailsa Holroyd1, Anne-Louise Latif1, David Irvine1 P113 METAGENOMIC NEXT-GENERATION SEQUENCING VISUALLY DISPLAYS A FALSE-POSITIVE DETECTION OF HIV NUCLEIC ACID RELATED TO CART TREATMENT: A CASE STUDY IN A CHILD M. I. Zhou 1, Lili Huang1, Zhenjiang Bai1, Shuiyan Wu1, Jun Lu1, Shaoyan Hu1 P114 IMPACT OF SERUM INTERLEUKIN 1-BETA LEVELS DURING CAR-T CELLS THERAPY ON CLINICAL OUTCOME: A SINGLE-CENTER EXPERIENCE Ilaria Cutini 1, Riccardo Boncompagni1, Chiara Camerini1, Mirella Giordano1, Antonella Gozzini1, Alessio Mazzoni1, Gaia Mirabella1, Chiara Nozzoli1, Edoardo Simonetti1, Riccardo Saccardi1 P115 THE SCOTTISH EXPERIENCE WITH CAR-T CELL THERAPY: A FOCUS ON DLBCL David Hopkins 1, Anne Parker1, Dimitris Galopoulos1, Grant McQuaker1, Andrew Clark1, Ailsa Holroyd1, Anne-Louise Latif1, David Irvine1 P116 DONOR-DERIVED LYMPHOID ANTIGEN-DIRECTED CAR-T CELLS FOR RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA IN CHILDREN AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: REPORT OF 17 CASES Olga Molostova1, Larisa Shelikhova1, Rimma Khismatullina1, Evelina Lyudovskikh1, Yuliya Abugova1, Dmitriy Pershin 1, Maria Klimentova1, Irina Shipitsina1, Ekaterina Malakhova1, Maria Fadeeva1, Alena Kulakovskaya1, Lili Khachatryan1, Viktoria Vedmedskaya1, Galina Novichkova1, Alexei Maschan1, Michael Maschan1 P117 CARDIOVASCULAR COMPLICATIONS OF CHIMERIC ANTIGEN RECEPTOR (CAR)-T CELL THERAPY Giorgia Mancini 1, Alice Frangione1, Edlira Rrapaj1, Ilaria Scortechini1, Francesco Saraceni1, Roberta Domizi1, Simona Pantanetti1, Abele Donati1, Antonio Dello Russo1, Federico Guerra1, Attilio Olivieri1 P118 CORRELATION OF IL-6 LEVELS WITH THE EXPANSION OF LYMPHOCYTIC POPULATIONS, SEVERITY OF CYTOKINE RELEASE SYNDROME AND NEUROTOXICITY FOLLOWING CAR-T CELL THERAPY Maria Liga 1, Dimitris Tsokanas1, Memnon Lysandrou1, Dionysia Kefala1, Evangelia Triantafyllou1, Angeliki Georgopoulou1, Eleftheria Sagiadinou1, Nikolaos Savvopoulos1, Vassiliki Zacharioudaki1, Alexandros Spyridonidis1 P119 EFFICACY OF ANTI-CD19 C AR-T CELL TREATMENT IN MANTLE-CELL LYMPHOMA HARBORING P53 ALTERATION: A MONOCENTRIC RETROSPECTIVE STUDY Daniele Mannina1, Chiara De Philippis1, Jacopo Mariotti 1, Barbara Sarina1, Daniela Taurino1, Stefania Bramanti1, Armando Santoro1 P120 OUTCOMES OF COMMERCIAL AND IN-HOUSE ANTI CD19 CAR T-CELL THERAPY FOR RELAPSED/REFRACTORY B-CELL LYMPHOMA IN SINGAPORE Shin Yeu Ong1, Cindy Krisnadi 1, Samuel Sherng Young Wang1, Yun Xin Chen1, Michaela Su-fern Seng2, Shui Yen Soh2, Esther Hian Li Chan3, William Ying Khee Hwang1, Aloysius Yew Leng Ho1, Francesca Lorraine Wei Inng Lim1 P121 CHOICE OF COMMERCIALLY AVAILABLE CAR T-CELLS PRODUCT FOR AGGRESSIVE LYMPHOMA: A SURVEY OF BEHALF OF THE CELLULAR THERAPY & IMMUNOBIOLOGY WORKING PARTY (CTIWP) OF THE EBMT Urban Novak1, Jarl E. Mooyaart2, Michael Daskalakis1, Christof Scheid3, Anne Sirvent4, Ibrahim Yakoub-Agha5, Ron Ram6, Edouard Forcade7, Lucía López Corral8, Emma Nicholson9, Eugenio Galli10, Friedrich Stölzel11, Wolfgang Bethge12, Eva Maria Wagner-Drouet13, Jorinde D. Hoogenboom14, Stephan Mielke15, Caroline Arber Barth16, Federico Simonetta17, Christian Chabannon18, Jürgen Kuball19, Annalisa Ruggeri20, Florent Malard 21 P122 ASSESSMENT OF MALNUTRITION DURING PREHABILITATION FOR PATIENTS UNDERGOING CAR-BASED CELLULAR THERAPY Juliet Morgan 1, Helen Long2 P123 LEUKAPHERESIS FOR AUTOLOGOUS CHIMERIC ANTIGEN RECEPTOR T CELL THERAPY IN CHILDREN: REPORT FROM A SINGLE PEDIATRIC CENTER June Iriondo 1,2, Josune Zubicaray1,2, Guzmán López de Hontanar3,1,2, Elena Sebastián1,2, Josefa Navarrete1, Ana Torre1, Rocio Rico1, Ana Castillo1, Blanca Herrero1,2, Alba Rubio1,2, Susana Buendía1,2, Alejandro Sanz1,2, Ana de la Cruz1,2, Almudena Galán1,2, Jesus Gonzalez de Pablo2, Manuel Ramirez1,2, Julián Sevilla1,2 P124 SEVERE HHV-6 RELATED ENCEPHALITIS FOLLOWING ANTI-CD19 CAR-T TREATMENT: A CASE STORY FROM A POINT OF CARE PRODUCTION FACILITY Nina Marie Birk1, Katrine Kielsen 1,2, Özcan Met3, Inge Marie Svane3, Eva Haastrup4, Lisbeth Pernille Andersen4, Kristian Schønning5, Jonas Nielsen6, Søren Lykke Petersen7, Marianne Ifversen1 P125 UNLOCKING POSSIBILITIES: THE IMPACT OF CYTOGENETIC ANALYSIS ON PATIENTS UNDERGOING CAR-T CELL THERAPY Christos Varelas 1, Ioannis Kyriakou1, Eleni Gavriilaki2, Zoi Bousiou1, Ioannis Batsis1, Nikolaos Spyridis1, Evangelia Yannaki1, Giorgos Papaioannou1, Maria Gkaitatzi1, Michail Iskas1, Ioanna Sakellari1, Anastasia Athanasiadou1 P126 T-CELL LARGE GRANULAR LYMPHOCYTE POPULATION INVOLVING CHIMERIC ANTIGEN RECEPTOR-MODIFIED T (CAR T) CELLS IN PATIENTS WITH CYTOPENIA AFTER CD19-TARGETED CAR T-CELL THERAPY: CASE SERIES Aya Albittar 1, Alireza Torabi2, Emily Liang1,2, Andrew Portuguese1,2, Jennifer Huang1,2, Cecilia Yeung1,2, Erik Kimble1,2, Delaney Kirchmeier1, Aiko Torkelson1, Abigail Chutnik1, Ryan Cassaday1,2, Aude Chapuis1,2, Hans-Peter Kiem1,2, Filippo Milano1,2, Folashade Otegbeye1,2, Mazyar Shadman1,2, Brian Till1,2, David Maloney1,2, Cameron Turtle1,2,3, Jordan Gauthier1,2, Alexandre Hirayama1,2 P127 TREATMENT WITH TISAGENLECLEUCEL OF RELAPSE REFRACTORY DIFFUSE LARGE B CELL LYMPHOMA – SINGLE CENTER EXPERIENCE UHC ZAGREB Barbara Dreta 1, Sandra Bašić-Kinda1, Dino Dujmović1, Neno Živković1, Josip Batinić1,2, Ida Hude Dragičević1, Klara Dubravčić1, Margareta Dobrenić1, Ivana Ilić1, Snježana Dotlić2,1, Lea Galunić-Bilić1, Ivana Franić-Šimić1, Igor Aurer1,2 P128 SERUM AMYLOID-A IN CAR-T CELL THERAPY: A NEW ACUTE PHASE REACTANT Eugenio Galli 1, Ilaria Pansini2, Patrizia Chiusolo1,2, Stefan Hohaus1,2, Anna Modoni1, Luca Montini1, Nicola Piccirillo1,2, Federica Sorà1,2, Simona Sica1,2 P129 CHALLENGES IN CHIMERIC ANTIGEN RECEPTOR-T CELL PRODUCT ADMINISTRATION IN A HIGH TUMOR-BURDEN ELDERLY PATIENT WITH MANTLE-CELL LYMPHOMA Umberto Pizzano1,2, Francesca Patriarca1,2, Marta Lisa Battista 1, Giuseppe Petruzzellis1,2, Gabriele Facchin1, Giuseppe Di Renzo1,2, Renato Fanin1,2 P130 REGIONAL REPRESENTATION OF THE OUTCOMES OF A COHORT OF PATIENTS WITH B-CELL ALL TREATED WITH TISAGENLECLEUCEL FROM A CENTRE IN THE MIDDLE EAST Fahad Bahkali MD 1, Ali Alahmari MD1, Riad El Fakih1, Amr Hanbali1, Rashed Al Bakr1, Abdulwahab Albabtain1, Naeem Chaudhri1, Walid Rasheed1, Ayman Saad1, Saud Alhayli1, Ahmed Abdrabou1, Samar Alfaqaawi1, Yazeed Bajuaifer1, Ahmed Bin Salman1, Alfadil Haroon1, Amal Hejab1, Taimoor Hussain1, Heba Madien1, Mostafa Saleh1, Momen Nassani1, Marwa Nassar1, Sara Samarkandi1, Mohamed Sharif1, Reem Alasbali1, Emad Ghabashi1, Majed Altareb1, Shaykhah Alotaibi1, Shihab Ahmed1, Reena Ulahannan1, Alfadel Alshaibani1, Hanan Alkhaldi1, Abdullah Alamer1, Mansour Alfayez1, Feras Alfraih1, Ahmad Alotaibi1, Fahad Alsharif1, Marwan Shaheen1, Fahad Almohareb1, Hazza Alzahrani1, Mahmoud Aljurf1, Syed Ahmed1 P131 DELAYED ICANS IN POST BCMA CAR-T CELL THERAPY CHALLENGES Mohamed Abuhaleeqa 1, Yara Afifi1, Sheima Mahmoud Ali1, Fatema Al Kaabi1, Inas El-Najjar1, Jayakumar David Dennison1, Mansi Sachdev1, Nameer Al Saadawi1, Ruqqia Mir1, Lev Brylev1, Mohamed Mostafa1, Yendry Ventura1, Krina Patel2 P132 CAR T CELLS TARGETING CD28 SHOW PRECLINICAL ACTIVITY AGAINST T-ALl Semjon Willier1, Julian Fäber1, Theodora Ispyrlidou2, Sophia Nikolaides2, Jonas Wilhelm2, Paulina Ferrada-Ernst2, Franziska Blaeschke2, Fiona Becker-Dettling2, Maike Breidenbach2, Theresa Käuferle2, Annika Peters2, Dana Stenger2, Christoph Klein2, Tobias Feuchtinger 1 P133 HAMBURG REAL-WORLD EXPERIENCE WITH AXICABTAGENE CILOLEUCEL IN AGGRESSIVE B-CELL LYMPHOMA: IN-VIVO EXPANSION CORRELATES WITH TREATMENT OUTCOME Susanna Carolina Berger1, Anita Badbaran1, Evgeny Klyuchnikov1, Kristin Rathje1, Nico Gagelmann1, Maria Geffken1, Dominic Wichmann1, Christian Frenzel1, Dietlinde Janson1, Christine Wolschke1, Nicolaus Kröger1, Boris Fehse1, Francis Ayuk 1 P134 CD45-DIRECTED CAR-T CELLS WITH CD45 KNOCKOUT EFFICIENTLY KILL MYELOID LEUKEMIA AND LYMPHOMA CELLS IN-VITRO EVEN AFTER EXTENDED CULTURE Maraike Harfmann 1, Tanja Schröder1, Dawid Glow1, Maximilian Jung1, Almut Uhde1, Nicolaus Kröger1, Stefan Horn1, Kristoffer Riecken1, Boris Fehse1, Francis Ayuk1 P135 COMPARISON OF CD123- AND CD33-CAR-NK CELL PREPARATIONS IN A XENOGRAFT MOUSE MODEL FOR TREATMENT OF ACUTE MYELOID LEUKEMIA Fenja Gierschek 1,2, Sabine Müller3, Julia Kostyra3, Jan Kuska3, Tobias Bexte1,2,4,5, Katja Stein1,2, Lisa Marie Reindl1,2, Sophia Thul1,2, Alina Moter1,2, Philipp Wendel1,2,6,7, Hadeer Mohamed Rasheed8,9, Ningyu Li8, Juliane Schlüter1, Sabine Hünecke1, Claudia Cappel1, Olaf Penack8,10, Thomas Oellerich2,4,6, Jan-Henning Klusmann1,2, Nina Möker3, Congcong Zang3, Evelyn Ullrich1,2,4,6 P136 HELP FOR UKRAINIAN HEMATOLOGY PATIENTS (HUP) – A GLOBAL INITIATIVE SUPPORTING THE ESTABLISHMENT OF HEMATOLOGY CENTERS AND HEMATOPOIETIC CELL TRANSPLANTATION PROGRAMS IN DIFFICULT SITUATIONS Irina Korenkova 1, Sergiy Klymenko2, Oleksandr Lysytsia3, Menachem Bitan4, Olena Lukianets5, Jan M. Zaucha6, Joannis Mytilineos7, Andreas Hochhaus8, Mats Heyman9, Francois Guilhot10, Natacha Bolanos11, Roman Kuts12, Hildegard T. Greinix13, Isabel Sanchez-Ortega14, Mickey Boon Chai Koh15, Bohdan Medvediev1, Susan Landgraf16, Rüdiger Hehlmann17, Mahmoud Aljurf18, Katherina Filonenko19, Reguia Belkhedim18, Torsten Haferlach*20, Arnold Ganser*21, Anna Sureda Balari*22, Damiano Rondelli*23, Dietger Niederwieser*24,25,26 P137 A RETROSPECTIVE ANALYSIS COMPARING ORCA-T TO POST-TRANSPLANT CYCLOPHOSPHAMIDE BASED ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT IN PATIENTS WITH MATCHED UNRELATED DONORS RECEIVING MYELOABLATIVE CONDITIONING Amandeep Salhotra 1, Alexandra Gomez Arteaga2, Caspian Oliai3, Sagar S. Patel4, Jeremy Pantin5, Arpita Gandhi6, Bhagirathbhai Dholaria7, Edmund K. Waller8, Samer A. Srour9, Anna Pavlova10, Irene Agodoa10, J. Scott McClellan10, Nathaniel B. Fernhoff10, Mehrdad Abedi11, Everett H. Meyer12 P138 PHASE I/II STUDY ON INFUSION OF ALLOREACTIVE OR EX VIVO IL-15 STIMULATED NK CELLS AFTER HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS WITH ACUTE LEUKEMIA Carmen Mestre1,2, Odelaisy León Triana1,2,3, Alfonso Navarro Zapata1,2, David Bueno4, Luisa Sisinni4, Isabel Badell Serra5, Marta González Vicent6, Cristina Beléndez7, Laura Clares Villa1,2, Yasmina Mozo6, Karima Al-Akioui1,2, Victor Galán4, Pilar Guerra1,4, Carlos Echecopar4, Halin Bareke1,2, Cristina Aguirre1,2, Antonio Pérez Martínez 1,2,4,8 P139 HARNESSING CIK CELL INTERVENTION FOR ACUTE LEUKEMIA OR MDS RELAPSE POST ALLO-HSCT Eva Rettinger 1, Marie Luedtke1, Emilia Salzmann-Manrique1, Sabine Huenecke1, Melanie Bremm1, Claudia Cappel1, Gesine Bug1, Johann Greil2, Roland Meisel3, Eva Maria Wagner-Drouet4, Jan-Henning Klusmann1, Halvard Bonig5, Peter Bader1 P140 RISK FACTORS FOR THE DEVELOPMENT OF GRAFT-VERSUS-HOST-DISEASE FOLLOWING DONOR LYMPHOCYTE INFUSION AFTER ALLOGENEIC STEM CELL TRANSPLANTATION Eva Koster1, Peter von dem Borne1, Peter van Balen1, Erik Marijt1, Jennifer Tjon1, Tjeerd Snijders2, Daniëlle van Lammeren3, Hendrik Veelken1, Frederik Falkenburg1, Liesbeth de Wreede1, Constantijn Halkes 1 P141 RELAPSE AFTER HSCT: MAY DLI/CIK CELLS STILL PLAY A ROLE IN THE CELLULAR THERAPY ERA? DLI/CIK AS A RESCUE STRATEGY FOR CHILDREN WITH POST-TRANSPLANT MRD-POSITIVITY Francesca Limido1,2, Alex Moretti2,3, Riccardo Carnevale1,2, Sara Napolitano2, Francesca Vendemini2, Sonia Bonanomi2, Andrea Biondi1,2,3, Giuseppe Gaipa3, Adriana Balduzzi 1,2 P142 PHASE I/II PRELIMINARY RESULTS OF HUMAN PRO-T-CELLS MANUFACTURING IN VITRO TO ACCELERATE IMMUNE RECONSTITUTION AFTER UMBILICAL CORD BLOOD TRANSPLANTATION IN ADULT PATIENT WITH HEMATOLOGIC MALIGNANCIES Elisa Magrin1, Jean-Sébastien Diana 1, Clotilde Aussel1, Eden Schwartz1, Jinmi Baek1, Naim Bouazza2, Aurélie Gabrion1, Alice Girardot1, Axelle Maulet1, Brigitte Ternaux1, Aurore Touzart1, Caroline Tuchmann-Durand3, Ludovic Lhermitte1, Vahid Asnafi1, Edouard Forcade4, Huynh Anne5, Jean-Marc Treluyer1,2, Olivier Hermine1,6,3, Marina Cavazzana1,6,3 P143 INNATE DONOR EFFECTOR ALLOGENEIC LYMPHOCYTES AFTER ALLOGENEIC STEM CELL TRANSPLANTATION - THE IDEAL TRIAL Lia Minculescu 1, Lone Smidstrup Friis1, Soeren Lykke Petersen1, Brian Thomas Kornblit1, Niels Smedegaard Andersen1, Ida Schjoedt1, Helle Lesley Andersen1, Eva Haastrup1, Lisbeth Pernille Andersen1, Henrik Sengeloev1 P144 CD45RA DEPLETION PRESERVES PROPORTIONS AND CLONAL BREATH OF VIRUS-SPECIFIC T CELLS Amandine Pradier 1,2, Astrid Melotti1, Chiara Bernardi1,2, Simona Pagliuca3, Sisi Wang1, Sarah Morin2, Federica Giannotti2, Stavroula Marouridi-Levrat2, Yves Chalandon1,2, Federico Simonetta1,2, Anne-Claire Mamez2 P145 LAYING THE FOUNDATIONS FOR AN IMMUNOPOTENCY TEST FOR ALLOGENEIC MESENCHYMAL STROMAL CELLS María Esther Martínez-Muñoz 1,2, Trinidad Martín-Donaire2, Rocío Sánchez2, Elvira Ramil2, Rosalía Alonso-Trillo2, Nuria Panadero1, Rocío Zafra2, Enrique Andreu3, Gloria Carmona4, Ana María García-Hernández5, Miriam López-Parra6, Gustavo Melen7, Luciano Rodríguez-Gómez8, Rosa Yáñez9, Cristina Avendaño-Solá1,2, Jose María Moraleda5, María Eugenia Fernández10, Rafael Francisco Duarte1,2 P146 CHARACTERIZATION OF CYTOKINE RELEASE SYNDROME AND NEUROTOXICITY IN COMMERCIALLY AVAILABLE BISPECIFIC ANTIBODIES AND CHIMERIC ANTIGEN RECEPTOR T-CELLS IN MULTIPLE MYELOMA Eden Biltibo1 P147 TRIAL IN PROGRESS: OBSERVATIONAL, POST-AUTHORISATION SAFETY STUDY TO DESCRIBE SAFETY AND EFFECTIVENESS OF TABELECLEUCEL IN PATIENTS WITH EBV + PTLD IN A REAL-WORLD SETTING IN EUROPE François Denjean1, Asmaa Zkik1, Constance Battin1, Sophie Blanchet1, Florence Carrere1, Mathias Domostoj1, Valérie Cassan2, Pavan Randhawa 3, Roberta Valenti1 P148 CD45RO CELLS ADDBACK IN TCR ΑΒ/CD45RA DEPLETED HAPLOIDENTICAL HSCT LEADS TO SUPERIOR OUTCOMES IN INFANTS WITH INBORN ERRORS OF IMMUNITY: EXPERIENCE FROM A DEVELOPING COUNTRY! Vimal Kumar Gunasekaran 1, Rishab Bharadwaj1, Meena Sivasankaran2, Niranjan Hegde2, Deepti Sachan1, Deenadayalan Munirathnam1 P149 IMPACT OF MYELOFIBROSIS ON PATIENTS WITH MYELODYSPLASTIC SYNDROMES FOLLOWING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Panpan Zhu 1,2,3,4, Xiaoyu Lai4,2,3,4, Lizhen Liu1,2,3,4, Jimin Shi1,2,3,4, Jian Yu1,2,3,4, Yanmin Zhao1,2,3,4, Luxin Yang1,2,3,4, Weiyan Zheng1,2,3,4, Jie Sun1,2,3,4, Wenjun Wu1,2,3,4, He Huang1,2,3,4, Yi Luo1,2,3,4 P150 MANAGEMENT AND TOXICITY OF DONOR LYMPHOCYTE INFUSIONS IN PEDIATRIC PATIENTS WITH ONCOLOGICAL DISEASES AFTER ALLOGENIC STEM CELL TRANSPLANTATION IN A SINGLE INSTITUTION Luisa Paschke 1, Marie Sophie Knape1, Katrin Vollmer1, Katja Gall1, Angela Wawer1, Jan-Henning Klusmann2, Dirk Reinhardt3, Hans-Jochem Kolb4, Belinda Simoes5, Stefan EG Burdach1, Irene Teichert von Luettichau1, Julia Hauer1, Uwe Thiel1 P151 IMPROVING OUTCOMES IN MDS/MPN: TREOSULFAN-BASED CONDITIONING FOR ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Alessandro Bruno1, Lorenzo Lazzari 1, Elisa Diral1, Sara Mastaglio1, Daniela Clerici1, Sarah Marktel1, Francesca Lunghi1, Simona Piemontese1, Daniele Sannipoli1, Camilla Gariazzo1, Gianluca Scorpio1, Gregorio Bergonzi1, Consuelo Corti1, Matteo Giovanni Carrabba1, Massimo Bernardi1, Luca Vago1, Maria Teresa Lupo-Stanghellini1, Fabio Ciceri1,2, Jacopo Peccatori1, Raffaella Greco1 P152 PREVIOUS RESPONSE TO TKIS AS A FAVORABLE PROGNOSTIC FACTOR FOR SURVIVAL IN HSCT FOR CML: LONG TERM RESULTS OF A SINGLE CENTER IN BRAZIL Vaneuza Funke 1, Giuliana Rosendo1, Daniela Setubal1, Caroline Bonamin Sola1, Samir Nabhan1, Rafael Marchesine1, Glaucia Tagliari1, Isabela Menezes1, Ana Lucia Mion1, Ricardo Pasquini1 P153 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT FOR MYELOFIBROSIS: UPDATED RESULTS FROM BRAZILIAN REGISTRY Vaneuza Funke 1, Alberto Cardoso Martins Lima1, Nelson Hamersclak2, Vergilio Colturato3, Afonso Vigorito4, Gustavo Machado Teixeira5, Vanderson Rocha6, Livia Mariano6, Decio Lerner7, George Mauricio Navarro Barros8, Alessandra Paz9, Celso Arrais10, Claudia Astigarra11, Fabio Pires12, Fernando Barroso Duarte13, Ricardo Pasquini1, Mary Flowers14 P154 RESULTS OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA AFTER THERAPY WITH TYROSINE KINASE INHIBITORS Larisa Kuzmina1, Vera Vasilyeva 1, Zoya Konova1, Marya Dovydenko1, Olga Koroleva1, Olga Pokrovskaya1, Darya Mironova1, Elena Parovichnikova1 P155 ALLOGENEIC STEM CELL TRANSPLANTATION IN CHRONIC MYELOID LEUKEMIA IN ERA OF TYROSINE KINASE INHIBITORS: A 13-YEAR SINGLE CENTRE EXPERIENCE FROM INDIA Mehak Trikha1, Lingaraj Nayak 1, Sachin Punatar1, Anant Gokarn1, Akanksha Chichra1, Sumeet Mirgh1, Nishant Jindal1, P.G. Subramaniam1, Nikhil Patkar1, Prashant Tembhare1, Gourav Chatterjee1, Sweta Rajpal1, Navin Khattry1 P156 ATG-TARGETED DOSING STRATEGY REDUCED CMV/EBV REACTIVATION AND IMPROVED SURVIVAL WITHOUT INCREASING GVHD AFTER URD-PBSCT: A SINGLE-CENTER, PROSPECTIVE, SINGLE-ARM, CLINICAL TRIAL Sheng Chen 1, Jishan Du1, Haitao Wang2, Nan Wang1, Liping Dou2, Daihong Liu2 P157 IMPACT OF FLUDARABINE DOSE ON OUTCOME AFTER ALLO-HCT WITH REDUCED INTENSITY CONDITIONING FOR OLDER PATIENTS WITH AML: A STUDY FROM THE ALWP OF THE EBMT Guillaume Dachy 1, Myriam Labopin2, Gerard Socié3, Cristina Castilla-Llorente4, Edouard Forcade5, Igor Wolfgang Blau6, Patrice Ceballos7, Eric Deconinck8, David Burns9, Claude Eric Bulabois10, Radovan Vrhovac11, Anne Huynh12, Didier Blaise13, Johan Maertens14, Thomas Schroeder15, Jacques-Olivier Bay16, Bipin Savani17, Alexandros Spyridonidis18, Fabio Ciceri19, Mohamad Mohty20 P158 TOXICITY COMPARISON OF TWO REDUCED INTENSITY CONDITIONING (RIC) TRANSPLANT REGIMENS FOR ADULT ALL IN CR1/2: RESULTS FROM THE RANDOMISED PHASE 2 MULTICENTRE ALL-RIC TRIAL Anna Castleton 1, Rebekah Weston2, Aimee Jackson2, Matthew Beasley3, Rebecca Bishop2, Scott Chapman2, Patricia Diez4, Mohamed Elhaneid2, Adele Fielding5, Andrea Hodgkinson2, Stephanie Lane2, George Mikhaeel6, Nick Morley7, Eduardo Olavarria8, Eleni Tholouli9, Bela Patel10, Ronjon Chakraverty11, David Marks12 P159 CXCR4 DIRECTED ENDORADIOTHERAPY WITH [177LU] PENTIXATHER ADDED TO TBI IS FEASIBLE AND SPARES THE HEMATOPOIETIC NICHE IN PATIENTS WITH RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA Krischan Braitsch 1, Martina Rudelius2, Maike Hefter1, Katrin Koch1, Katharina Nickel1, Katharina S. Götze1, Wolfgang Weber1, Florian Bassermann1, Matthias Eiber1, Mareike Verbeek1, Peter Herhaus1 P160 EVALUATION OF THE TRANSPLANT CONDITIONING INTENSITY (TCI) SCORE FOR SECOND ALLOGENEIC STEM CELL TRANSPLANTATION (ALLO-SCT2) - A STUDY OF THE ALWP OF THE EBMT Giuliano Filippini Velázquez 1, Jacques-Emmanuel Galimard2, Alexandros Spyridonidis3, Jakob Passweg4, Didier Blaise5, Nikolaus Kröger6, Igor Wolfgang Blau7, Patrice Chevallier8, Tobias Gedde-Dahl9, Panagiotis Kottaridis10, Johan Maertens11, Emma Nicholson12, Arancha Bermudez Rodriguez13, Peter Dreger14, Gerald G. Wulf15, Mareike Verbeek16, Emanuele Angelucci17, Mathias Eder18, Uwe Platzbecker19, Bipin Savani20, Christoph Schmid1, Fabio Ciceri21, Mohamad Mohty2 P161 VENETOCLAX COMBINED WITH FLUDARABINE AND MELPHALAN CONDITIONING REGIMEN IN PATIENTS > 50 YEARS WITH MYELOID MALIGNANCIES: INTERIM ANALYSIS OF A PROSPECTIVE, MULTICENTER, PHASE II TRIAL Panpan Zhu 1,2,3,4, Xiaoyu Lai1,2,3,4, Lizhen Liu1,2,3,4, Yibo Wu1,2,3,4, Jimin Shi1,2,3,4, Jian Yu1,2,3,4, Yanmin Zhao1,2,3,4, Yishan Ye1,2,3,4, Guifang Ouyang5, Yi Chen6, Weiyan Zheng1,2,3,4, Jie Sun1,2,3,4, He Huang1,2,3,4, Yi Luo1,2,3,4 P162 FLUDARABINE/TREOSULFAN VS. FLUDARABINE/TBI CONDITIONING FOR ALLOSCT FOR AML AND MDS: COMPARABLE SURVIVAL RATES BUT DIVERGENT IMMUNE RECONSTITUTION Lina Kolloch1, Philipp Berning1, Christian Reicherts1, Julian Ronnacker1, Simon Call1, Julia Marx1, Matthias Floeth1, Eva Esseling1, Jan-Henrik Mikesch1, Christoph Schliemann1, Georg Lenz1, Matthias Stelljes 1 P163 CONDITIONING FOR ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN PATIENTS WITH PERIPHERAL T-CELL LYMPHOMA (PTCL): INTERMEDIATE-DOSE TBI DOES THE JOB Isabelle Krämer1, Thomas Luft 1, Ute Hegenbart1, Stefan Schönland1, Peter Stadtherr1, Linda Selberg1, Laila König1, Carsten Müller-Tidow1, Peter Dreger1 P164 5-YEAR TRANSPLANT SUCCESS AFTER TREOSULFAN CONDITIONING Rohtesh S. Mehta 1, Joachim Deeg1, Ted Gooley1, Stephanie J. Lee1, Laurel Thur1, Filippo Milano1 P165 EFFICACY ANALYSIS OF HAPLOIDENTICAL TRANSPLANTATION USING BUSULFAN/CYCLOPHOSPHAMIDE AND MELPHALAN AS CONDITIONING REGIMEN FOR RELAPSED AND REFRACTORY ACUTE MYELOID LEUKEMIA Fei Pan1, Guanlan Yue1, Kang Gao1, Kun Wang1, Huili Zhu1, Yujie Wang1, Zheren Wang1, Zhijie Wei 1 P166 POST-TRANSPLANT CYCLOPHOSPHAMIDE RESULTS IN LOW INCIDENCE OF CHRONIC GRAFT-VERSUS-HOST DISEASE IN UNRELATED OR MATCHED-SIBLING DONOR STEM CELL TRANSPLANTATION WITHOUT COMPROMISING IMMUNE RECONSTITUTION AND GRAFT-VERSUS-LEUKEMIA EFFECT Ioannis Tsonis 1, Ifigeneia Tzannou1, Tatiana Tzenou1, Natali El Gkotmi1, Maria Katsareli1, Ismini Darmani1, Vasiliki Bampali1, Evridiki Theodorou1, Maria-Eleni Karatza1, Maria Garofalaki1, Zoi Poulopoulou1, Marina Papageorgiou1, Soultana Tryfonidou1, Panagiotis Siamatas1, Eirini Tziotziou1, Eirini Grispou1, Stavros Gigantes1, Ioannis Baltadakis1, Aikaterini Souravla1 P167 CONDITIONING INTENSITY IN STEM CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR ACUTE MYELOID LEUKEMIA: A STUDY FROM THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT Jaime Sanz 1, Myriam Labopin2, Jurjen Versluis3, Didier Blaise4, Jacoppo Peccatori5, Juan Montoro1, Gwendolyn Van Gorkom6, Peter von dem Borne7, Hélène Labussière-Wallet8, Montserrat Rovira9, Péter Reményi10, Patrice Chevallier11, Mi Kwon12, Matthias Eder13, Jan Vydra14, Eolia Brissot15, Alexandros Spyridonidis16, Simona Piemontese5, Mohamad Mohty15, Fabio Ciceri5 P168 EVALUATION OF A BALTIMORE MODIFIED NON-TBI CONDITIONING BEFORE ALLOGENEIC TRANSPLANT FROM HAPLOIDENTICAL OR MISMATCHED UNRELATED DONORS, RESULTS FROM A MONOCENTRIC RETROSPECTIVE STUDY Victoria Volpari 1, Claude Eric Bulabois1, Anne Thiebaut-Bertrand1, Sophie Park1, Martin Carre1, Mathieu Meunier1 P169 EXPLORING THE IMPACT OF CONDITIONING REGIMEN INTENSITY ON ALLOGENEIC STEM CELL TRANSPLANT OUTCOMES: INSIGHTS FROM A SINGLE-CENTER STUDY Vera Radici 1, Riccardo Marnoni1, Mirko Farina1, Eugenia Accorsi Buttini1, Sofia Terlizzi1, Lorenzo Masina1, Enrico Morello1, Gabriele Magliano1, Alessandro Leoni2, Federica Re2, Katia Bosio2, Simona Bernardi2, Domenico Russo1, Michele Malagola1 P170 IMPACT OF PRETRANSPLANT MRD IN PATIENTS ALLOGRAFTED FOR AML AFTER TREOSULFAN-BASED CONDITIONING Jacob Pyka 1, Nils Leimkühler1, Artur Schneider1, Jennifer Kaivers1, Rudolf Trenschel1, Annemarie Mohring1, Christian Reinhardt1, Thomas Schroeder1, Christina Rautenberg1 P171 FLUDARABINE TREOSULFAN REDUCED-INTENSITY CONDITIONING REGIMEN PRIOR HAPLOIDENTICAL HEMATOPOIETIC CELL TRANSPLANTATION WITH POST TRANSPLANTATION CYCLOPHOSPHAMIDE IN FRAIL/OLDER AML PATIENTS: PRELIMINARY RESULTS OF A SINGLE CENTER EXPERIENCE Benjamin Bouchacourt1, Anne-Charlotte Le Floch1, Sabine Fürst1, Sylvain Garciaz1, Samia Harbi1, Yosr Hicheri1, Thomas Pagliardini1, Boris Calmels1, Faezeh Legrand1, Claude Lemarie1, Federico Pagnussat1, Christian Chabannon1, Pierre-Jean Weiller1, Marie-Anne Hospital1, Norbert Vey1, Didier Blaise1, Raynier Devillier 1 P172 PHARMACOKINETIC ANALYSIS OF TARGETED DOSING OF ANTITHYMOCYTE GLOBULIN IN ADULTS: A PROSPECTIVE ANALYSIS Jishan Du 1, Haitao Wang1, Nan Wang1, Chao Ma1, Fei Li1, Lu Wang1, Liping Dou1, Daihong Liu1 P173 THE COMBINATION OF FLUDARABINE, BUSULFAN AND ANTI-T LYMPHOCYTE GLOBULIN IS WELL TOLERATED AND OFFERS EFFECTIVE REDUCED INTENSITY CONDITIONING THERAPY FOR PATIENTS WITH MYELOID DISEASE Chris Armstrong 1, Hayley Foy-Stones1, Nicola Gardiner1, Catherine Flynn1, Eibhlin Conneally1, Nina Orfali1 P174 COMPARING PTCY AND GIAC IN HAPLOIDENTICAL SETTINGS: UPDATED ANALYSIS FROM A NATIONWIDE TRANSPLANTATION REGISTRY Yi-Wei Lee 1,2, Xavier Cheng-Hong Tsai2,3, Tzu-Ting Chen4, Tung-Liang Lin5, Yi-Chang Liu6, Chi-Cheng Li7, Ming Yao2,3, Bor-Sheng Ko2,3,8 P175 THE RESULTS OF ALLOGENEIC HSCT AFTER TREOSULFAN–BASED CONDITIONING REGIMEN IN CHILDREN WITH AML: MULTICENTER RETROSPECTIVE STUDY OF THE POLISH PEDIATRIC STUDY GROUP FOR HSCT Agnieszka Sobkowiak-Sobierajska 1, Olga Zając-Spychała1, Maksymilian Deręgowski1, Monika Mielcarek-Siedziuk2, Krzysztof Kałwak2, Agnieszka Zaucha-Prażmo3, Katarzyna Drabko3, Robert Dębski4, Jan Styczyński4, Jolanta Goździk5, Jacek Wachowiak1 P176 OUTCOME OF ALLOGENEIC STEM CELL TRANSPLANTATION IN AML PATIENTS AFTER CONDITIONING WITH FLUDARABINE/TOTAL BODY IRRADIATION(2GY) WITH OR WITHOUT ANTI-THYMOCYTE GLOBULINE OR POST-TRANSPLANT CYCLOPHOSPHAMIDE Thomas Heinicke 1, Myriam Labopin2, Emmanuelle Polge3, Annoek EC Broers4, Gwendolyn van Gorkom5, Michel Schaap6, Jürgen Kuball7, Goda Choi8, Peter von dem Borne9, Gitte Olesen10, Henrik Sengeloev11, Uwe Platzbecker12, Giang Lam Vuong13, Alexandros Spyridonidis14, Bipin N. Savani15, Fabio Ciceri16, Mohamad Mohty17 P177 CONFIRMING BETTER OUTCOMES WITH TBI-BASED MYELOABLATIVE CONDITIONING FOR ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION EVEN IN SECOND COMPLETE REMISSION Mario Delia 1, Vito Pier Gagliardi1, Corinne Contento2, Daniela Di Gennaro2, Olga Battisti2, Camilla Presicce2, Immacolata Attolico1, Paola Carluccio1, Francesco Albano1,2, Pellegrino Musto1,2 P178 POST TRANSPLANT CYCLOPHOSPHAMIDE PLUS ABATACEPT LEADS TO DECREASED DAY + 30 CHIMERISM LEVELS Peter Abdelmessieh 1, Henry Fung1, Yuliya Shestovska1, Michael Styler1, Khanal Rashmi1, Asya Varshavsky-Yanovsky1 P179 HEMATOPOIETIC STEM CELL TRANSPLANT WITH REGULATORY AND CONVENTIONAL T CELL ADOPTIVE IMMUNOTHERAPY IN TP53-MUTATED/DELETED AML Rebecca Sembenico1, Loredana Ruggeri1, Simonetta Saldi1, Sara Tricarico2, Antonella Mancusi1, Valerio Viglione1, Francesco Zorutti1, Tiziana Zei2, Roberta Iacucci Ostini2, Eleonora Cristofani1, Roberto Limongello1, Cristina Mecucci1, Andrea Velardi1, Massimo Fabrizio Martelli1, Cynthia Aristei1, Maria Paola Martelli1, Alessandra Carotti2, Antonio Pierini 1 P180 LOMUSTINE, ETOPOSIDE, CYTARABINE, MELPHALAN (LEAM) PROTOCOL AS CONDITIONING REGIMEN BEFORE AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION (ASCT) IN PATIENTS WITH LYMPHOMAS: A SINGLE CENTER EXPERIENCE Željko Prka 1, Anamarija Vrkljan Vuk1, David Čičić1, Ena Sorić1, Zdravko Mitrović1,2, Marko Lucijanić1,2, Marija Ivić Čikara1, Željko Jonjić1, Mario Piršić1, Vlatko Pejša1,2, Ozren Jakšić1,2 P181 SAFETY AND EFFICACY OF BUSULFAN 9.6 BASED RIC (AUGMENTED RIC) IN OLDER PATIENTS WITH AML WHO ARE NOT SUITABLE FOR MAC Kalina Abrol 1, Ram Vasudevan Nampoothiri1, Michael Kennah1, Natasha Kekre1, David Allan1, Harold Atkins1, Lothar Huebsch1, Trang Doan1, Carolina Cieniak1, Sheryl McDiarmid1, Tim Ramsay1, Christopher Bredeson1, Ashish Narayan Masurekar1 P182 TOTAL MARROW AND LYMPHOID IRRADIATION (TMLI) IN COMBINATION WITH CYCLOPHOSPHAMIDE AND ETOPOSIDE IMPROVES THE OUTCOME OF PATIENTS WITH POOR-RISK ACUTE LEUKEMIA Anthony Stein 1, Monzr Al Malki1, Yan Wang1, Joycelynne Palmer1, Ibrahim Aldoss1, Haris Ali1, Ahmed Aribi1, Andrew Artz1, Brian Ball1, Len Farol1,2, An Liu1, Guido Marcucci1, Ryotaro Nakamura1, Vinod Pullarkat1, Eric Radany1, Firoozeh Sahebi1,2, Amandeep Salhotra1, Karamjeet Sandhu1, Eileen Smith1, Ricardo Spielberger1,2, Susanta Hui1, Stephen Forman1, Jeffrey Wong1 P183 TREOSULFAN-FLUDARABINE CONDITIONING REGIMEN WITH POST-TRANSPLANT HIGH-DOSE CYCLOPHOSPHAMIDE: A RETROSPECTIVE ANALYSIS Aura Arola 1, Lotta Tapana2, Maija Itälä-Remes1 P184 CONDITIONING WITH I.V. BUSULFAN, ONCE VERSUS FOUR TIMES DAILY IN ADULTS: IMPACT ON PHARMACOKINETICS, ORGAN TOXICITIES AND SURVIVAL AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Claire Seydoux1, Michael Medinger1, Joerg Halter1, Dominik Heim1, Katharina M. Rentsch1, Jakob R. Passweg 1 P185 NON MYELOABLATIVE VERSUS REDUCED INTENSITY CONDITIONING IN ELDERLY PATIENTS WITH PTCY. A MULTICENTRE RETROSPECTIVE COHORT ANALYSIS FROM GATMO-TC Mariano Berro 1, Jorge Arbelbide2, Ana Lisa Basquiera3, Adriana Vitriu4, Silvina Palmer5, Martin Saslavsky6, Patricio Duarte7, Sebastian Yantorno8, Martin Castro9, Ruben Burgos10, Gabriela Sturich3, Gonzalo Bentolila11, Sol Jarchum12, Juan Real13, Gonzalo Ferini2, Georgina Bendek2, Maria M. Rivas1, Gustavo Kusminsky1 P186 IMPACT OF TRANSPLANT CONDITIONING INTENSITY ON BOTH ACUTE AND CHRONIC GVHD IN HAPLO-PTCY TRANSPLANT: TWO GITMO CENTRES’ EXPERIENCE Umberto Pizzano1,2, Simona Piemontese3, Gabriele Facchin1, Raffaella Greco3, Marta Lisa Battista1, Jacopo Peccatori3, Renato Fanin1,2, Fabio Ciceri3,4, Francesca Patriarca1,2, Maria Teresa Lupo-Stanghellini 3 P187 CHANGES IN PROGNOSTIC VARIABLES FOR ALLOGENEIC TRANSPLANTATION WITH REDUCED INTENSITY CONDITIONING IN ELDERLY PATIENTS OVER TIME Seong Kyu Park 1, Se Hyung Kim1, Jina Yun1, Chan Kyu Kim1, Jong Ho Won2 P188 COMPARISON OF FLUDARABINE-MELPHALAN AND FLUDARABINE-TREOSULFAN AS CONDITIONING REGIMENS PRIOR TO ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Maria Liga 1, Dimitris Tsokanas1, Eleftheria Sagiadinou1, Memnon Lysandrou1, Angeliki Georgopoulou1, Evangelia Triantafyllou1, Vassiliki Zacharioudaki1, Anastasia Christopoulou1, Alexandros Spyridonidis1 P189 OUTCOMES OF PATIENTS WITH HIGH-RISK ACUTE LYMPHOBLASTIC LEUKEMIA IN CR1 UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION THROUGH A FIXED TRANSPLANTATION POLICY INCLUDING A CHEMOTHERAPY-BASED CONDITIONING Enrico Santinelli1, William Arcese1, Gottardo De Angelis2, Benedetta Mariotti2, Giulia Ciangola2, Giuseppe Avvisati1, Laura Cudillo3, Paolo De Fabritiis4, Andrea Mengarelli5, Agostino Tafuri6, Antonio Bruno2, Ilaria Mangione2, Gaspare Adorno2, Adriano Venditti2, Alessandra Picardi7, Raffaella Ceretti 2 P190 DESENSITISATION TREATMENT FOR HLA-DONOR SPECIFIC ANTIBODIES IN EX-VIVO T CELL DEPLETED HAPLOIDENTICAL ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANT – A MULTI-CENTRE RETROSPECTIVE STUDY IN SINGAPORE Christopher S W Tham1, Yeh Ching Linn1, Liang Piu Koh2, Jeffrey KS Quek1, Aloysius Y L. Ho1, Francesca WI Lim1, William Y.K. Hwang 1, Yeow Tee Goh1, Jing Jing Lee1, Hein Than1 P191 PROMISING OUTCOMES IN HAPLOIDENTICAL TRANSPLANTATION USING ABATACEPT IN COMBINATION WITH POST TRANSPLANT CYCLOPHOSPHAMIDE IN TREOSULFAN BASED CONDITIONING REGIMEN Mohammed Debes1, Gabe Toth1, Tom Seddon1, Shahid Iqbal1, Leah Credidio1, Hannah Williams2, Fotini Partheniou2, Sophie Hughes1, Priscilla Hetherington1, Thomas Seddon1, Muhammad Saif 1 P192 FOLLOW-UP OF TRANSPLANT-ELIGIBLE MULTIPLE MYELOMA RECEIVED BUSULFAN-BASED CONDITIONING VERSUS HIGH-DOSE MELPHALAN Sungnam Lim1, Byeong-Sok Sohn2, Seonyang Park 3 P193 SEQUENTIAL CONDITIONING REGIMEN WITH THIOTEPA, CLOFARABINE AND BUSULFAN (TEC/CLOB2A2) AND POST-TRANSPLANT CYCLOPHOSPHAMIDE IN ADULTS WITH REFRACTORY AML: A RETROSPECTIVE MONOCENTRIC STUDY Amandine Le Bourgeois1, Alice Garnier1, Pierre Peterlin1, Maxime Jullien1, Chloe Antier1, Thierry Guillaume1, Patrice Chevallier 1 P194 CLINICAL OUTCOMES OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION USING BUSULFAN/CYCLOPHOSPHAMIDE AND MELPHALAN OR THIOTEPA AS CONDITIONING REGIMEN IN 37 PATIENTS WITH ACUTE MEGAKARYOBLASTIC LEUKEMIA Fei Pan 1, Guanlan Yue1, Kang Gao1, Kun Wang1, Huili Zhu1, Yujie Wang1, Zheren Wang1, Zhijie Wei1 P195 RETROSPECTIVE EVALUATION OF NUTRITIONAL STATUS PRE AND POST HEMATOPOIETIC STEM CELL TRANSPLANTATION: A SINGLE CENTRE EXPERIENCE Yuva Vishalini Ravindran 1, Maria Losa Maroto2, Amrith Mathew2, Sunrit Majumder2, Lewis David Oxley2, Venkatesh Karthikeyan3, Nitin Ramanathan4, David Waldron2 P196 BEAM/LEAM VERSUS LACE CONDITIONING FOR AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN LYMPHOMAS – A RETROSPECTIVE, SINGLE-CENTRE ANALYSIS OF CLINICAL OUTCOMES AND TOXICITY PROFILE Germano Glauber de Medeiros Lima1, André Dias Américo1, Eurides Leite da Rosa2, Pedro Paulo Faust Machado1, Juliana Matos Pessoa1, Hegta Taina Rodrigues Figueiroa 1, Fauze Lutfe Ayoub1, Isabella Silva Pimentel Pittol1, José Ulysses Amigo Filho1, Phillip Scheinberg1, Fabio Rodrigues Kerbauy1 P197 REAL-WORLD, SINGLE-CENTRE OUTCOME DATA OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION UTILISING TREOSULFAN-BASED CONDITIONING REGIMENS Mohamed Debes1, Yeong Lim1, Thomas Seddon1, James Clarke1, Clare Hawkins1, Anna Smielewska2, Alex Howard3, Julie Grant1, Sajid Pervaiz1, Shahid Iqbal1, Arpad Toth1, Muhammad Saif 1 P198 IMPACT OF CO-ADMINISTERED ACETAMINOPHEN ON BUSULFAN PHARMACOKINETICS Rutvij Khanolkar 1, Shahbal Kangarloo1, Jan Storek1 P199 TREOSULFAN-BASED CONDITIONING REGIMEN IN PEDIATRIC ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A CASE SERIES OF SINGLE CENTER EXPERIENCE Ilgen Sasmaz 1,2, Ali Antmen1, Utku Ayguneş1, Barbaros Karagun1, Duygu Turksoy1 P200 IMPLEMENTATION OF MACHINE LEARNING TO MEASURE THE IMPACT OF OVER- AND UNDER-EXPOSURE, AND PREDICT BUSULFAN’S PHARMACOKINETICS, IN PATIENTS RECEIVING CONDITIONING PRIOR TO HSCT Dorian Protzenko1, Laurent Bourguignon2, Benjamin Bouchacourt 3, Raynier Devillier3, Joseph Ciccolini1 P201 MITOCHONDRIAL COPY NUMBER VARIATION AS A SURROGATE MARKER OF ACQUIRED RESISTANCE TO THE BIFUNCTIONAL ELECTROPHILIC CHEMOTHERAPEUTIC DRUG BUSULFAN Vid Mlakar1, Simona Jurkovic Mlakar1, Yvonne Gloor1, Isabelle Dupanloup 1, Yoann Sarmiento1, Denis Marino1, Mary Boudal-Khosbheen1, Chakradhara Rao Satyanarayana Uppugunduri1, Marc Ansari2,1 P202 CLOFARABINE, MELPHALAN AND THIOTEPA REDUCED-INTENSITY CONDITIONING CHEMOTHERAPY ALONG WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE-BASED GRAFT VERSUS HOST DISEASE PROPHYLAXIS FOR ALLOGENEIC STEM CELL TRANSPLANT Alejandro Del Rio Verduzco1, Aileen Go1, Murali Kodali1, Matthew Ulrickson1, Nicolas De Padova1, Eric Smith1, Ivan Komerdelj1, Hayley Heers1, Jennifer Brookes1, Haley Pebler1, Rachael Yasuda1, Rajneesh Nath 1 P203 THE EFFICACY OF MODIFIED MELPHALAN AND BUSULFAN-BASED CONDITIONING REGIMEN FOR ASCT IN LOW-RISK AND INTERMEDIATE-RISK AML PATIENTS Shulian Chen1, Xiaoyu Zhang1, Yi He1, Sizhou Feng1, Mingzhe Han1, Xingli Zhao2, Jie Bai3, Lijuan Li4, Zhihua Zhang5, Ying Wang1, Jianxiang Wang1, Weihua Zhai 1, Erlie Jiang1 P204 SECOND ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH REDUCED-INTENSITY CONDITIONING AND DONOR CHANGES IN HEMATOLOGICAL NON-MALIGNANCIES AFTER THE FIRST TΑΒ + CELL DEPLETED HAPLO-HSCT Jianyun Liao 1, Chaoke Bu1, Lan He1, Jujian He1, Weiwei Zhang1, Zongxin Feng1, Yanfeng Zhou1, Bo Lyu1, Wenqian Shi1, Jing Wang1, Yuelin He1, Chunfu Li1 P205 FIRST ALLOGENIC STEM CELL TRANSPLANTATION IN A PATIENT WITH ROIFMAN SYNDROME AND EBV-ASSOCIATED PLASMABLASTIC LYMPHOMA RELAPSE USING AUC CONTROLLED BUSULFAN BASED MYELOABLATIVE CONDITIONING Marie Sophie Knape1, Katja Gall1, Katrin Vollmer1, Florian Ressle1, Felicitas Ferrari von Klot1, Angela Wawer1, Michael H. Albert2, Chaim M. Roifman3, Britta Eiz-Vesper4, Birgit Burkhardt5, Wilhelm Woessmann6, Irene Teichert von Lüttichau1, Julia Hauer1, Uwe Thiel 1 P206 HIGH GRAFT FAILURE WITH FLUDARABINE, TREOSULPHAN CONDITIONING REGIMEN IN MYELOID MALIGNANCIES Taner Tan1, Sinem Civriz Bozdağ 1, Ümit Barbaros Üre1, Elif Birtaş Ateşoğlu2, Emre Osmanbaşoğlu1, Ahmet Burhan Ferhanoğlu1, Hakan Kalyon1, Meltem Olga Akay1 P207 ALLOGENEIC TRANSPLANTATION USING MYELOABLATIVE CONDITIONING REGIMES IS SAFE AND EFFECTIVE IN ELDERLY PATIENTS OVER 60 YEARS WITH AML AND MDS Yan-Li Zhao 1, Yue Lu1, Xing-Yu Cao1, Jian-Ping Zhang1, Jia-Rui Zhou1, Zhi-Jie Wei1, Min Xiong1, De-Yan Liu1, Rui-Juan Sun1 P208 COMPARISON BETWEEN DIFFERENT REDUCED INTENSITY CONDITIONING REGIMENS IN AML OR MDS José Emilio Salinas 1, Yorman Flores1, Patricio Rojas1, Catherine Gutierrez1, Marcela Vidal1, Verónica Jara1, Elizabeth Rivera1, James Campbell1, María José Garcia1, Vicente Sandoval1, Maximiliano Vergara1, Mauricio Ocqueteau1, Felipe Palacios1, Pablo Sandoval1, Mauricio Sarmiento1 P209 PHASE 1 TRIAL RESULTS FOR PATIENTS WITH ADVANCED HEMATOLOGIC MALIGNANCIES TREATED WITH ORCA-T CELL THERAPY WITH REDUCED INTENSITY CONDITIONING AND SINGLE AGENT TACROLIMUS Alejandro Villar-Prados 1, Katherine Sutherland1, Robert Negrin1, Sally Arai1, Matthew Frank1, Laura Johnston1, Robert Lowsky1, David Miklos1, Lori Muffly1, Saurabh Dahiya1, Andrew Rezvani1, Surbhi Sidana1, Parveen Shiraz1, Judith Shizuru1, Wen-Kai Weng1, Melody Smith1, Sushma Bharadwaj1, John Tamaresis1, Anna Pavlova2, Scott McClellan2, Everett Meyer1 P210 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH CONDITIONING INCLUDING DONOR CAR-T CELLS FOR R/R B-CELL LYMPHOMA AND R/R MULTIPLE MYELOMA FAILING CAR T-CELL THERAPY Fan Yang1, Rui Liu 1, Zhonghua Fu1, Teng Xu1, Hui Shi1, Lixia Ma1, Guoai Su1, Biping Deng2, Tong Wu1, Xiaoyan Ke1, Kai Hu1 P211 SMADCAM-1 IS DECREASED AFTER ALLOHCT, ALONG WITH GUT MICROBIOTA DYSBIOSIS, AND IS ASSOCIATED WITH HEMATOPOIETIC RECOVERY Karen Fadel 1, Lama Siblany1, Razan Mohty1, Nicolas Stocker1, Eolia Brissot1, Rémy Dulery1, Anne Banet1, Simona Sestili1, Zoe van de Wyngaert1, Agnes Bonnin1, Ramdane Belhocine1, Tounes Ledraa1, Antoine Capes1, Mohamad Mohty1, Béatrice Gaugler1, Florent Malard1 P212 PENTOSTATIN AND CYCLOPHOSPHAMIDE DECREASE GRAFT REJECTION BUT INCREASE TRANSPLANT-RELATED COMPLICATIONS IN HAPLOIDENTICAL HCT FOR SICKLE CELL DISEASES Emily Limerick1, Matthew Hsieh 1, Julia Varga1, Mary Lacy Grecco1, Jennifer Brooks1, Wynona Coles1, Jackie Queen1, Neal Jeffries1, Courtney Fitzhugh1 P213 GRANULOCYTES TO INDUCE DONOR-DERIVED T CELL EXPANSION AFTER T REPLETE, MISMATCHED CB IN POST-TRANSPLANT RELAPSED AND REFRACTORY PAEDIATRIC ACUTE LEUKAEMIA: RESULTS OF GRANS TRIAL Srividhya Senthil 1, Roisin Borrill1, Denise Bonney1, Ramya Hanasoge-Nataraj1, Madeleine Powys1, Omima Mustafa1, Robert Wynn1 P214 GENOME EDITING OF LONG-TERM REPOPULATING HEMATOPOIETIC STEM CELLS USING CCR5-UCO-HETTALEN Valeriia Laushkina1, Alena Shakirova1, Olga Epifanovskaya1, Vladislav Sergeev1, Kirill Lepik1, Marina Popova1, Alexander Kulagin 1 P215 ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION IMPROVES PROGNOSIS IN AML PATIENTS WITH CSF3R MUTATION AND CEBPA DOUBLE MUTATION Jiaxin Cao1, Yawei Zheng 1, Aiming Pang1, Erlie Jiang1 P216 CAST REGIMEN FOR GVHD PROPHYLAXIS YIELDS LOWER RELAPSE RATE AND IMPROVED DISEASE-FREE SURVIVAL: A CIBMTR PROPENSITY SCORE MATCHED CASE-CONTROL ANALYSIS Samer Al-Homsi1, Todd DeFor2, Kelli Cole1, Frank Cirrone1, Maher Abdul-Hay1, Stephanie Wo1, Andres Suarez-Londono1, Sharon Gardner1, Jingmei Hsu1, George Yaghmour3, Caitrin Bupp2, Stephen Spellman 2 P217 SEQUENTIAL INTRAVENOUS UMBILICAL CORD DERIVED MESENCHYMAL STROMAL CELLS (MSCS) FOR SALVAGE TREATMENT OF STEROID-REFRACTORY ACUTE GVHD (RS-AGVHD): A PHASE IB/II STUDY Yanmin Zhao 1,2,3,4, Yi Luo1,2,3, Jimin Shi1,2,3,4, Jian Yu1,2,3,4, Lizhen Liu1,2,3,4, Xiaoyu Lai1,2,3,4, Huarui Fu1,2,3,4, Yishan Ye1,2,3,4, Nainong Li5,6,7, Erlie Jiang8, Qiong Xie9, Jundong Gu9, Zhibo Han9, Zhongchao Han9, He Huang1,2,3,4 P218 NOVEL GVHD PROPHYLAXIS WITH POST-TRANSPLANT INTERMEDIATE DOSE CYTOXAN AND ABATACEPT – LOW RATES OF ACUTE GVHD AND TRANSPLANT-RELATED TOXICITY Asya Varshavsky Yanovsky1, Yuliya Shestovska 1, Shalina Joshi1, Dzhirgala Mandzhieva1, Peter Abdelmessieh1, Rashmi Khanal1, Michael Styler1, Henry Fung1 P219 REDUCING PTCY DOSE IN PATIENTS RECEIVING CAST FOR GVHD PREVENTION RESULTS IN ACCELERATED ENGRAFTMENT AT THE COST OF HIGHER INCIDENCE OF GVHD J. Andres Suarez-Londono 1, Kelli Cole1, Frank Cirrone1, Stephanie Wo1, Maher Abdul-Hay1, Jingmei Hsu1, Sharon Gardner1, Mohammad Abu-Zaid1, Gloria Contreras Yametti1, Benjamin Levinson1, Judith Goldberg1, A. Samer Al-Homsi1 P220 RUXOLITINIB IN ACUTE AND CHRONIC GRAFT-VERSUS-RECEPTOR DISEASE: LONG-TERM REAL-LIFE EXPERIENCE IN A MULTI-CENTRE STUDY Virginia Escamilla Gómez 1, Lucía López Corral2, Beatriz Astibia Mahillo3, Patricia Alcalde Mellado1, Francisco Manuel Martín Domínguez1, Marina Acera Gómez2, Pedro Asensi Cantó4, Juan Montoro Gómez4, Melissa Torres Ochando5, Asunción Borrego Borrego5, Leslie González Pinedo5, María Teresa Zudaire Ripa6, Marta González Vicent7, Ana Benzaquén Vallejos8, Isabel Izquierdo9, Irene García Cadenas10, Sara Redondo Velao10, Alberto Musseti11, Eloi Cañamero Giro12, Pilar Palomo Moraleda13, Guillermo Orti Pascual14, Pedro Antonio González Sierra15, Lucía García16, Valentín García Gutiérrez3, José Antonio Pérez-Simón1 P221 PHASE 1 TRIAL INVESTIGATING THE SAFETY AND TOLERABILITY OF LEFLUNOMIDE IN PATIENTS WITH STEROID-DEPENDENT CHRONIC GRAFT-VERSUS-HOST DISEASE (CGVHD) AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT (ALLOHCT) Amandeep Salhotra 1, Dongyun Yang1, Sally Mokhtari1, Badri Modi1, Monzr Al-Malki1, Haris Ali1, Idoroenyi Amannam1, Nicole Karras1, Karamjeet Sandhu1, Ahmed Aribi1, Salman Otoukesh1, Andrew Artz1, Guido Marcucci1, Stephen Forman1, Eileen Smith1, De-Fu Zhang1, Steven Rosen1, Ryotaro Nakamura1 P222 COMPARISON OF EFFICACY AND TOLERABILITY OF RUXOLITINIB ALONE OR RUXOLITINIB WITH EXTRACORPOREAL PHOTOPHERESIS OR EXTRACORPOREAL PHOTOPHERESIS MONOTHERAPY FOR STEROID REFRACTORY AGVHD: A GITMO RETROSPECTIVE STUDY Marta Lisa Battista 1, Francesca Patriarca1,2, Miriam Isola2, Gabriele Facchin1, Patrizia Chiusolo3,4, Domenico Russo5, Attilio Olivieri6, Bruna Puglisi7, Ilaria Scortechini7, Alessandro Rambaldi8, Irene Maria Cavattoni9, Albana Lico10, Riccardo Saccardi11, Angela Cuoghi12, Marco Ladetto13, Vincenzo Federico14, Maria Caterina Micò15, Antonella Geromin1, Anna Maria Gallina16, Elena Oldani8, Eliana Degrandi17, Renato Fanin1,2, Fabio Ciceri18, Massimo Martino15 P223 MINI-DOSE METHOTREXATE COMBINED WITH METHYLPREDNISOLONE FOR INITIAL TREATMENT OF MILD ACUTE GVHD: A MULTI-CENTER, RANDOMIZED TRIAL Yu Wang 1, Qi-Fa Liu2, De-Pei Wu3, Zheng-Li Xu1, Ting-Ting Han1, Yu-Qian Sun1,4, Fen Huang2, Zhi-Ping Fan2, Na Xu2, Feng Chen3, Ye Zhao3, Yuan Kong1, Xiao-Dong Mo1, Lan-Ping Xu1, Xiao-Hui Zhang1, Kai-Yan Liu1, Xiao-Jun Huang1,5 P224 IMPROVED LONG-TERM OUTCOME OF RUXOLITINIB PLUS ECP VS RUXOLITINIB ALONE IN STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST DISEASE (SR-AGVHD) Iryna Lastovytska1, Evgeny Klyuchnikov1, Silke Heidenreich1, Normann Steiner1, Radwan Massoud1, Niko Gagelmann1, Johanna Richter1, Christian Niederwieser1, Kristin Rathje1, Tatiana Urbanowicz1, Ameya Kunte1, Janik Engelmann1, Christina Ihne1, Cecilia Lindhauer1, Franziska Elisabeth Marquard1, Mirjam Reichard1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schäfersküpper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Ina Rudolph1, Gabriele Zeck1, Rolf Krause1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger 1 P225 COMPARISON BETWEEN POSTTRANSPLANT CYCLOPHOSPAMIDE AND ATG BASED REGIMENS AS GVHD PROPYLAXIS IN ONE-ANTIGEN MISMATCH UNRELATED DONOR TRANSPLANTATION Gabriele Facchin1, Marta Lisa Battista 1, Maria De Martino2, Miriam Isola2, Antonella Geromin1, Chiara Savignano3, Valentina Simeon1,2, Alessandra Soravia1,2, Giuseppe Di Renzo1,2, Umberto Pizzano1,2, Martina Pucillo1,2, Renato Fanin1,2, Francesca Patriarca1,2 P226 PROSPECTIVE EVALUATION OF PLASMA INFLAMMATORY PROTEINS FOR PREDICTION OF SEVER GRAFT-VERSUS-HOST DISEASE AFTER TRANSPLANT Mingyang Wang1, Erlie Jiang 1 P227 LYMPHOPENIA BUT NOT ABSOLUTE LYMPHOCYTE COUNT BEFORE ANTI-T-LYMPHOCYTE GLOBULIN (ATLG) ADMINISTRATION IMPACTS TRANSPLANT OUTCOMES IN MATCHED UNRELATED PERIPHERAL BLOOD STEM CELL ALLOGENEIC STEM CELL TRANSPLANTATION Radwan Massoud 1, Evgeny Klyuchnikov1, Normann Steiner1, Christian Niederwieser1, Ameya Shirinian Kunte1, Silke Heidenreich1, Kristin Rathje1, Nico Gagelmann1, Tetiana Perekhrestenko1, Gaby Zeck1, Rolf Krause1, Christine Wolschke1, Dietlinde Janson1, Francis Ayuk1, Nicolaus Kröger1 P228 A PROSPECTIVE, MULTI-CENTER, RANDOMIZED, CONTROLLED CLINICAL TRIAL TO EXPLORE THE ROLE OF MESENCHYMAL STEM CELLS IN PREVENTING GRAFT VERSUS HOST DISEASE Han Yao 1, Xiaoqi Wang1, Ruihao Huang1, Haixia Fu2, Ren Lin3, Yimei Feng1, Xiaojuan Deng1, Ting Chen1, Lidan Zhu1, Jia Liu1, Yuqing Liu1, Lu Zhao1, Lu Wang1, Shichun Gao1, Huanfeng Liu1, Cheng Zhang1, Peiyan Kong1, Li Gao1, Qifa Liu3, Xiaohui Zhang2, Xi Zhang1,4,5 P229 RUXOLITINIB AS SECOND-LINE TREATMENT FOR BOS AFTER HEMATOPOIETIC CELL TRANSPLANTATION Feng Chen 1, Yuqing Tu1,2,3, Jia Chen1,2,3, Shushu Xu1,2,3, Yi Fan1,2,3, Mimi Xu1,2,3, Mengqi Xiang1,2,3, Tiemei Song1,2,3, Xiang Zhang1,2,3, Xiaoli Li4, Lian Bai5, Depei Wu1,2,3 P230 POST-TRANSPLANTATION CYCLOPHOSPHAMIDE WITH OR WITHOUT PROPHYLACTIC DONOR LYMPHOCYTE INFUSION AFTER HLA-MATCHED ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN MDS/AMl Joost van der Hem 1, Jurjen Versluis2, Iris Erpelinck2, Chantal Mouws2, Peter Von dem Borne1, Annoek Broers2, Stijn Halkes1 P231 IMPROVED GRFS WHEN ATG/ATLG IS USED IN ALLO-PBSCT FROM MATCHED SIBLING DONORS - AN EBMT REGISTRY STUDY BY THE TRANSPLANT COMPLICATIONS WORKING PARTY Agnieszka Piekarska 1, Mouad Abouqateb2, William Boreland2, Christophe Peczynski2, Jan Maciej Zaucha1, Nicolaus Kröger3, Robert Zeiser4, Fabio Ciceri5, Thomas Schroeder6, Thomas Luft7, Jakob Passweg8, Desiree Kunadt9, Matthias Stelljes10, Igor Wolfgang Blau11, Uwe Platzbecker12, Ibrahim Yakoub-Agha13, Didier Blaise14, Anna Maria Raiola15, Johanna Tischer16, Eva Maria Wagner-Drouet17, Julia Winkler18, Christoph Schmid19, Gerald Wulf20, Matthias Edinger21, Johan Maertens22, Friedrich Stölzel23, Jan Vydra24, Pavel Zak25, Ivan Moiseev26, Hélène Schoemans22, Olaf Penack11, Zinaida Perić27 P232 NAVIGATING GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS IN MATCHED SIBLING DONOR ALLOGENEIC TRANSPLANTS: A COMPARATIVE ANALYSIS OF POST-TRANSPLANT CYCLOPHOSPHAMIDE VERSUS DIVERSE REGIMENS – INSIGHTS FROM A SINGLE-CENTER EXPERIENCE Majed Altareb1, Carol Chen1, Mats Remberger2, Armin Gerbitz1,3, Dennis Kim 1,3, Rajat Kumar1,3, Wilson Lam1,3, Ivan Pasic1,3, Fotios Michelis1,3, Igor Novitzky-Basso1,3, Auro Viswabandya1,3, Jonas Mattsson1,3, Arjun Datt Law1,3 P233 POST TRANSPLANT CYCLOPHOSPHAMIDE AS GVHD PROPHYLAXIS IN PATIENTS RECEIVING HLA MISMATCHED (7/8) UNRELATED DONOR FOR MYELOID MALIGNANCIES: RESULTS OF THE GITMO PROSPECTIVE PHYLOS TRIAL Anna Maria Raiola 1, Benedetto Bruno2, Antonio Maria Risitano3, Federico Mosna4, Irene Maria Cavattoni4, Francesco Onida5, Giorgia Saporiti5, Francesca Patriarca6, Marta Lisa Battista6, Vincenzo Pavone7, Anna Mele7, Patrizia Chiusolo8, Simona Sica8, Barbara Loteta9, Carmen Di Grazia1, Angelo Michele Carella10, Dalila Salvatore10, Enrico Morello11, Alessandro Leoni11, Luisa Giaccone2, Paolo Bernasconi12, Elisabetta Terruzzi13, Nicola Mordini14, Carlo Borghero15, Francesco Zallio16, Franco Narni17, Anna Grassi18, Attilio Olivieri19, Adriana Vacca20, Nicoletta Sacchi21, Giovannino Ciccone22, Anna Castiglione22, Emanuele Angelucci1, Francesca Bonifazi23, Fabio Ciceri24, Eliana Degrandi25, Massimo Martino9 P234 EXTRACORPOREAL PHOTOPHERESIS AS SALVAGE TREATMENT IN PEDIATRIC PATIENTS WITH STEROID-REFRACTORY ACUTE GVHD: A MULTICENTER, RETROSPECTIVE STUDY Daria Pagliara1, Valentina Sofia1, Claudia Del Fante2, Manuela Tumino3, Francesco Saglio4, Giovanna Leone1, Gianluca Viarengo2, Mattia Algeri 1, Giovanna Giorgiani2, Giovanna Del Principe1, Barbarella Lucarelli1, Pietro Merli1, Anna Colpo3, Cesare Perotti2, Marco Zecca2, Alessandra Biffi3, Franca Fagioli4, Franco Locatelli1,5 P235 BELUMOSUDIL PHASE 1 DOSE-ESCALATION STUDY IN HEALTHY VOLUNTEERS AND PHASE 2 STUDY IN PATIENTS WITH CGVHD WHO FAILED ≥1 LINE OF SYSTEMIC THERAPY IN CHINA Ying Wang 1, Depei Wu1, Xiang Zhang1, Yuhua Li2, Yanjie He2, Qifa Liu3, Li Xuan3, Zhenyu Li4, Kunming Qi4, Yuqian Sun5, Shunqing Wang6, Wenjian Mo6, Lei Gao7, Ye Hua8, Yu Wang8, Ying Zhang8, Nico Fu9 P236 IMPACT OF RESPIRATORY FUNCTION TESTS IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Francisco Manuel Martin Dominguez 1, Rocío Parody-Porras1, Virginia Escamilla-Gómez1, Nancy Rodríguez-Torres1, Cristina Blázquez-Goñi1, Javier Delgado-Serrano1, Juan Luis Reguera-Ortega1, Ildefonso Espigado-Tocino1, José Antonio Rodríguez-Portal2, José Antonio Pérez-Simón1 P237 MANAGEMENT OF CORTICOSTEROIDS IN PATIENTS RECEIVING RUXOLITINIB TREATMENT FOR CHRONIC GRAFT-VERSUS-HOST DISEASE: A POST HOC ANALYSIS FROM THE RANDOMIZED PHASE 3 REACH3 STUDY Emily Kintsch1, Zhenyi Xue1, John Galvin1, Franco Locatelli2,3, Robert Zeiser4, Valkal Bhatt 1 P238 A PROGNOSTIC MODEL FOR NON-RELAPSE MORTALITY RISK BASED ON MACHINE LEARNING IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM-CELL TRANSPLANTATION Jingjing Yang1, Xiawei Zhang 1, Liping Dou1, Daihong Liu1 P239 A HIGH FREQUENCY OF TYPE 3 DENDRITIC CELLS IN CD1C + DC ON DAY + 28 AFTER HSCT COULD BE AN EFFECTIVE INDICATOR OF SEVERE GVHD IN PEDIATRIC PATIENTS Xin Liu 1, Bohan Li1, Di Yao1, Minyuan Liu1, Peifang Xiao1, Jun Lu1, Jie Li1, Yuanyuan Tian1, Shaoyan Hu1 P240 HAPLOIDENTICAL HEMATOPOIETIC CELL TRANSPLANTATION WITH UNRELATED CORD BLOOD ENHANCES SURVIVAL AND REDUCES THE INCIDENCE OF AGVHD FOR PEDIATRIC ACUTE MYELOID LEUKEMIA Fei Pan1, Jing Long2, Guanlan Yue1, Kang Gao1, Kun Wang1, Huili Zhu1, Yujie Wang1, Zheren Wang1, Yuan Feng1, Lili Shi1, Zhijie Wei 1, Kaiyan Liu2 P241 RUXOLITINIB AND ANTI-TNF-ALPHA THERAPY SUBSTANTIALLY IMPROVE SURVIVAL IN PEDIATRIC PATIENTS WITH SEVERE STEROID REFRACTORY GVHD Jaspar Kloehn1, Manon Queudeville1, Johanna Schrum1, Ysabel Alessa Schwietzer1, Ingo Müller 1 P242 IMMUNOPHENOTYPE OF EXTRACELLULAR VESICLES AS A BIOMARKER OF CHRONIC GRAFT-VERSUS-HOST-DISEASE: A PILOT STUDY Giuseppe Lia1,2, Federica Ferrando1,1, Michele Dicataldo1,2, Andrea Evangelista3, Alessia Cargnino1,2, Irene Dogliotti1,2, Stefania Bruno4, Maria Chiara Deregibus4, Aurora Martin1,2, Jessica Gill1,2, Chiara Maria Dellacasa5, Alessandro Busca5, Giovanni Camussi4, Benedetto Bruno1,2, Luisa Giaccone 1,2 P243 T-CELL DEPLETED ALLOGENEIC STEM CELL TRANSPLANTATION WITH ALEMTUZUMAB IS A VIABLE ALTERNATIVE TO POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR PATIENTS WITH ACUTE MYELOID LEUKEMIA AND MYELODYSPLASTIC SYNDROME Alexandra Rojek 1, Mylove Mortel1, Andrew Artz2, Richard Larson1, Hongtao Liu3, Mariam Nawas1, Adam Duvall1, Toyosi Odenike1, Gregory Roloff1, Wendy Stock1, Michael Bishop1, Satyajit Kosuri1 P244 EXTENDED ABATACEPT USE WITH SEROTHERAPY TO REDUCE ACUTE GVHD INCIDENCE IN PEDIATRIC PATIENTS TRANSPLANTED FROM HLA-MISMATCHED UNRELATED DONORS Francesco Quagliarella1, Jochen Buechner2, Mattia Algeri1, Ingvild Heier2, Francesca Del Bufalo1, Phoi Phoi Diep2, Daria Pagliara1, Marco Becilli1, Barbarella Lucarelli1, Emilia Boccieri1, Federica Galaverna1, Ilaria Pili1, Roberto Carta1, Chiara Rosignoli1, Tiziana Corsetti1, Pietro Merli 1, Franco Locatelli1,3 P245 ENDOTHELIAL DYSFUNCTION AND RISK OF ACUTE GRAFT-VERSUS-HOST DISEASE AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: A PRELIMINARY ANALYSIS OF A SINGLE CENTRE STUDY Francesco Romano1, Antonio Bianchessi1, Irene Defrancesco1, Giulia Losi 1, Beatrice Ferrari2, Virginia Ferretti2, Cristina Picone2, Erica Consensi2, Mara De Amici2, Giorgia Testa2, Daniela Caldera2, Caterina Zerbi2, Valentina Zoboli2, Luca Arcaini1,2, Nicola Polverelli2 P246 EXTENDED ABATACEPT DOSING MAY REDUCE RATES OF CHRONIC GRAFT VS HOST DISEASE IN PEDIATRIC PATIENTS UNDERGOING BONE MARROW TRANSPLANT FOR UNDERLYING HEMATOLOGICAL DISEASES Eleanor Cook1, Pooja Khandelwal1, Ashley Teusink-Cross1, Michael Grimley 1 P247 COMPARISON OF THE EFFICACY OF MYCOPHENOLATE MOFETIL AFTER ORAL OR INTRAVENOUS ADMINISTRATION IN THE PREVENTION OF ACUTE GRAFT-VERSUS-HOST DISEASE Xinhui Zheng1, AiMing Pang1, Erlie Jiang 1 P248 NEAR ELIMINATION OF GVHD WITH AN ABBREVIATED CALCINEURON AND M-TOR INHIBITOR-FREE GVHD PREVENTION REGIMEN A. Samer Al-Homsi 1, Kelli Cole1, Frank Cirrone1, Stephanie Wo1, J. Andres Suarez-Londono1, Jingmei Hsu1, Mohammad Abu-Zaid1, Maher Abdul-Hay1 P249 EFFICACY AND SAFETY OF REDUCED-DOSES OF POST-TRANSPLANT CYCLOPHOSPHAMIDE IN PERIPHERAL BLOOD STEM CELL TRANSPLANTATION OUTSIDE THE HAPLOIDENTICAL SETTING Sara Redondo 1, Anna Arrufat1, Albert Esquirol1, Silvana Saavedra1, Miguel Arguello-Tomas1, Ana Carolina Caballero1, Guadalupe Oñate1, Ana Garrido1, Jordi Lopez1, Sara Miqueleiz1, J.M. Portos1, Eva Iranzo1, M.E. Moreno-Martinez2, Mireia Riba2, Jorge Sierra1, Javier Briones1, Irene García-Cadenas1, Rodrigo Martino1 P250 A LOW TRAJECTORY OF PLATELETS IN THE EARLY STAGES AFTER TRANSPLANTATION IS LINKED TO THE DEVELOPMENT OF SEVERE ACUTE GRAFT-VERSUS-HOST DISEASE (AGVHD) Dan Feng1, Erlie Jiang 1 P251 GVHD PROPHYLAXIS WITH THREE VERSUS FIVE DRUGS COMBINATION IN AML PATIENTS UNDERGOING ALLOGENEIC TRANSPLANT FROM HLA MISMATCHED UNRELATED OR HAPLOIDENTICAL RELATED DONOR Alessandra Picardi 1,2, Stella Santarone3, Massimo Martino4, Gottardo De Angelis2, Mariangela Pedata1, Stefania Leone1, Annalisa Natale3, Benedetta Mariotti2, Barbara Loteta4, Serena Marotta1, Doriana Vaddinelli3, Maria Celentano1, Giulia Ciangola2, Maria Caterina Micò4, William Arcese5,2, Raffaella Cerretti2 P252 CYTOKINE SECRETION DURING REDUCED INTENSITY CONDITIONING HAEMATOPOIETIC STEM CELL TRANSPLANT FOR MYELOID DISEASE, AN OPPORTUNITY FOR OUTCOME PREDICTION Chris Armstrong 1, Hayley Foy-Stones1, Gardiner Nicola1, Doherty Derek2, Anthony McElligott2, Fiona O’Connell2, Eibhlin Conneally1, Catherine Flynn1, Nina Orfali1 P253 INTERFERON GAMMA ACTIVITY IN IMPAIRED HAEMATOPOIETIC STEM CELL (HSC) PROLIFERATION AND GRAFT-VERSUS-HOST DISEASE Régis Peffault de Latour 1,2, Pietro Merli3, Emmanuel Monnet4, Malin Löfqvist4, Franco Locatelli3 P254 DELETIONS OF COMPLEMENT FACTOR H-RELATED GENES CFHR-1 AND CFHR-3 AND ACUTE GRAFT-VERSUS-HOST DISEASE AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION Lars Klingen Gjærde 1, Maja Milojevic1, Niels Smedegaard Andersen1, Lone Smidstrup Friis1, Brian Kornblit1, Marietta Nygaard1, Søren Lykke Petersen1, Ida Schjødt1, Jens D. Lundgren1,2, Daniel D. Murray1, Henrik Sengeløv1,2 P255 EXTERNAL VALIDATION OF SAINT LOUIS SCORE TO PREDICT ACUTE GRAFT-VERSUS-HOST DISEASE SEVERITY Alessandro Criscimanna 1,2, Annalisa Ruggeri1, Elisa Diral1, Raffaella Greco1, Alessandro Bruno1, Luca Canziani1,2, Sarah Marktel1, Francesca Farina1, Sara Mastaglio1, Daniela Clerici1, Simona Piemontese1, Andrea Acerbis1,2, Chiara Secco1, Magda Marcatti1, Matteo Giovanni Carrabba1, Massimo Bernardi1, Consuelo Corti1, Jacopo Peccatori1, Fabio Ciceri1,2, Maria Teresa Lupo-Stanghellini1 P256 EVALUATION OF RISK FOR BRONCHIOLITIS OBLITERANS SYNDROME AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION WITH MYELOABLATIVE CONDITIONING REGIMENS Kristina Maas-Bauer 1, Paraschiva Rassner1, Kristin Walther1, Claudia Wehr1, Reinhard Marks1, Ralph Wäsch1, Robert Zeiser1, Jürgen Finke1, Jesus Duque Afonso1 P257 THE MACROPHAGE ACTIVATION MARKER SCD163 AND ACUTE AND CHRONIC GRAFT-VERSUS-HOST DISEASE IN ALLOGENEIC HSCT Maja Munk Kristoffersen 1, Elisa Peen1, Henning Grønbæk2, Holger Jon Møller2, Henrik Sengeløv1, Klaus Müller1, Lars Klingen Gjærde1, Katrine Kielsen1 P258 ETIOLOGY OF DIARRHEA IN PATIENTS UNDERGOING COLONOSCOPIC BIOPSIES POST ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT FOR SUSPECTED ACUTE GUT GRAFT VERSUS HOST DISEASE Niket Mantri 1, Sachin Punatar1, Mukta Ramadwar1, Prachi Patil1, Shaesta Mehta1, Aditya Kale1, Shridhar Epari1, Sridhar Sundaram1, Koustubh Shekar1, Anant Gokarn1, Sumeet Mirgh1, Akanksha Chichra1, Nishant Jindal1, Lingaraj Nayak1, Libin Mathew1, Shesheer Kumar2, Rachana Tripathi2, Bhausaheb Bagal1, Navin Khattry1 P259 ABATACEPT ADDED TO POST TRANSPLANT CYCLOPHOSPHAMIDE-BASED GVHD PROPHYLAXYS IN ALLOGENEIC HEMATOPOYETIC STEM CELL TRANSPLANTATION: ONE SINGLE CENTER EXPERIENCE Paula Fernández- Caldas González 1,2, Rebeca Bailén1,2, Pilar Lancho1, Ignacio Gómez-Centurión1,2, Lucía Castilla1, Cristina Muñoz1, Santiago Sabell1, Javier Anguita1,2,3, Mi Kwon1,2,3 P260 SAFETY AND EFFICACY OF THE ROCK2-INHIBITOR BELUMOSUDIL IN CGVHD TREATMENT – A RETROSPECTIVE, MULTICENTER REAL-WORLD DATA ANALYSIS Silke Heidenreich 1, Katharina Egger-Heidrich2, Jörg P. Halter3, Friedrich Stölzel4, Matthias Edinger5, Wolfgang Herr5, Nicolaus Kröger1, Daniel Wolff5, Francis Ayuk1, Matthias A. Fante5 P261 EFFICACY AND SAFETY OF IBRUTINIB FOR CHRONIC GRAFT-VERSUS-HOST DISEASE: A SYSTEMATIC REVIEW Damai Santosa1, Daniel Rizky 1, Kevin Tandarto1, Ika Kartiyani1, Vina Yunarvika1, Desta Nur Ewika Ardini1, Budi Setiawan1, Eko Adhi Pangarsa1, Catharina Suharti1 P262 WHEN SHOULD RUXOLITINIB BE CEASED? A SINGLE CENTRE, RETROSPECTIVE, REAL-WORLD ANALYSIS OF RUXOLITINIB IN GRAFT VS HOST DISEASE POST ALLOGENEIC STEM CELL TRANSPLANT Harshita Rajasekariah 1,2, Barbara Withers1,2, Keith Fay1,3, Sam Milliken1,2, Nada Hamad1,2, John Moore1,2, Jacinta Perram1,2 P263 CLINICAL RESPONSE AND IMMUNOLOGICAL RECOVERY IN PATIENTS WITH STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST DISEASE (AGVHD) TREATED WITH THE ASSOCIATION OF EXTRACORPOREAL PHOTOPHERESIS (ECP) AND RUXOLITINIB Chiara Marcon1,2, Chiara Savignano1, Eleonora Toffoletti 1,2, Angela Michelutti1, Margherita Cavallin1, Giovanni Barillari1, Renato Fanin1,2, Francesca Patriarca2,1 P264 GVHD PROPHYLAXIS WITH POST-TRANSPLANT HIGH DOSE CYTOXAN OVERCOMING THE BARRIER OF ALLOGENEIC HCT IN FUNCTIONAL HIGH-RISK PATIENTS Asya Varshavsky Yanovsky1, Yuliya Shestovska 1, Matthew Hamby1, Dzhirgala Mandzhieva1, Shalina Joshi1, Peter Abdelmessieh1, Rashmi Khanal1, Michael Styler1, Henry Fung1 P265 MESENCHYMAL STROMAL CELLS IN PATIENTS WITH III-IV GRADE STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST-DISEASE: DAY 14 RESPONSE IS PREDICTIVE OF SURVIVAL Adomas Bukauskas 1, Renata Jucaitienė1, Artūras Slobinas1,2, Inga Šlepikienė1, Andrius Žučenka1,2, Linas Davainis1, Valdas Pečeliūnas2, Igoris Trociukas1, Laimonas Griškevičius1,2 P266 JAK-2 INHIBITOR RUXOLITINIB FOR THERAPY OF ACUTE STEROID-REFRACTORY GRAFT-VERSUS-HOST DISEASE AFTER ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH HEMATOLOGICAL MALIGNANT DISEASES Nikita Levkovsky1, Olesya Paina1, Anna Zvyagintseva1, Anna Osipova1, Alexandra Laberko1, Ivan Moiseev1, Tatyana Bykova 1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1 P267 THE IMPACT OF MEASURED RENAL FUNCTION BY IOTHALAMATE CLEARANCE ON ALLOGENEIC STEM CELL TRANSPLANT OUTCOMES IN PATIENTS RECEIVING POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR GVHD PROPHYLAXIS Jade Kutzke 1, Christopher Cahoon2, Kristin Mara1, Jodi Taraba1, Anmol Baranwal1, Mark Litzow1, William Hogan1, Abhishek Mangaonkar1, Mehrdad Hefazi Torghabeh1, Aasiya Matin1, Mithun Shah1, Urshila Durani1, Saad Kenderian1, Robert Wolf1, Gabriel Bartoo1, Hassan Alkhateeb1 P268 RUXOLITINIB TREATMENT OUTCOMES IN ACUTE GRAFT-VERSUS-HOST DISEASE (AGVHD) IN REAL-WORLD SETTING IN FINLAND Eeva Martelin 1, Arttu Kuikka2, Hanna Rajala1, Tuomas Ruohonen2, Hannu Mönkkönen3, Johanna Vikkula4, Kristiina Uusi-Rauva4, Urpu Salmennniemi1, Maija Itälä-Remes2 P269 VON WILLEBRAND FACTOR AS A DIAGNOSTIC AND ACTIVITY INDICATOR OF CHRONIC GRAFT-VERSUS-HOST DISEASE Antonela Lelas1, Lana Desnica 1, Ivan Sabol2, Desiree Coen Herak1,3, Marija Milos1,4, Ana Zelic Kerep1, Ante Vulic1, Nadira Durakovic1,3, Ranka Serventi Seiwerth1, Zinaida Peric5,6, Radovan Vrhovac1,3, Steven Zivko Pavletic7, Drazen Pulanic1,3 P270 A PROSPECTIVE PILOT STUDY OF GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS WITH POSTBIOTICS IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Jong-Ho Won 1, Seug Yun Yoon1, Sun Young Jeong1, Min-Young Lee1, Kyoung Ha Kim1, Namsu Lee1 P271 POST-TRANSPLANT CYCLOPHOSPHAMIDE WITH ANTI-THYMOCYTE GLOBULIN LOWERED SERUM IL-6 LEVEL COMPARED TO POST-TRANSPLANT CYCLOPHOSPHAMIDE ALONE AFTER HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION Jeong Suk Koh1, Wonhyoung Seo1, Sora Kang1, Chul Hee Kim1, Myung-Won Lee1, Hyewon Ryu1, Hyo-Jin Lee1, Hwan-Jung Yun1, Deog-Yeon Jo1, Ik-Chan Song 1 P272 USE OF RUXOLITINIB IN STEROID DEPENDENT/REFRACTORY ACUTE AND CHRONIC GRAFT VERSUS HOST DISEASE: A SINGLE CENTRE EXPERIENCE Dimitris Galopoulos 1, David Irvine1, Anne Parker1, Grant McQuaker1, Anne-Louise Latif1, Ailsa Holroyd1, Lorna Welsh1, Andrew Clark1 P273 ALPHA-1-ANTITRYPSIN TREATMENT OF STEROID-RESISTANT ACUTE AND CHRONIC GRAFT-VERSUS-HOST DISEASE, SINGLE CENTER EXPERIENCE Şebnem İzmir Güner1 P274 EFFICACY AND SAFETY OF INFLIXIMAB THERAPY IN THE SETTING OF STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST DISEASE Khalid Halahleh 1, Jana Al Shurman2, Ayat Taqash1, Mohammad Ma’koseh1, Ahmad Abu Khader1, Isra Muradi3, Waleed Da’na1, Rawad Rihani1 P275 THE EFFICACY AND SAFETY OF EXTRACORPOREAL PHOTOPHERESIS IN STEROID-DEPENDENT-REFRACTORY/ GVHD: COHORT STUDY FROM COMPREHENSIVE ADULT BONE MARROW TRANSPLANTATION PROGRAM IN JORDAN Khalid Halahleh 1, Raneem Al zubi1, Mohammad Bssol1, Husam Abu-Jazar1, Mohammad Ma’koseh1, Ehab Omari1, Isra Muradi2, Rawad Rihani1 P276 COMPARING THE OUTCOMES OF MATCHED AND MISMATCHED UNRELATED ALLOHCT WITH DIFFERENT ANTI-THYMOCYTE GLOBULIN FORMULATIONS. A RETROSPECTIVE ANALYSIS ON BEHALF OF POLISH ADULT LEUKEMIA GROUP Ugo Giordano1, Monika Mordak2, Małgorzata Sobczyk-Kruszelnicka3, Sebastian Giebel3, Lidia Gil4, Justyna Pilch1, Jarosław Dybko 5 P277 RITUXIMAB FOR THE TREATMENT OF STEROID-REFRACTORY ORAL CHRONIC GRAFT-VERSUS-HOST DISEASE AFTER ALLO-HEMATOPOIETIC STEM CELL TRANSPLANTATION Wei Ma 1, Xing-Yu Cao1, Song Xue2, Lei Dong1 P278 EXTRACORPOREAL PHOTOPHERESIS FOR THE TREATMENT OF GVHD IN CHILDREN IN THE ERA OF TRANSPLANT FROM PARTIALLY MATCHED DONORS Marianna Maffeis 1, Elena Soncini1, Giulia Baresi1, Stefano Rossi1, Chiara Gorio1, Emilio Ferrari2, Arnalda Lanfranchi3, Fulvio Porta1 P279 POST-TRANSPLANTATION CYCLOPHOSPHAMIDE IN HAPLOIDENTICAL TRANSPLANT: A REPORT IN LOW AND MIDDLE INCOME COUNTRY Araceli Leal-Alanis1, Juan Luis Ontiveros-Austria1, Maria Alejandra Nuñez-Atahualpa1, Anghela Milenka Mendoza-Sanchez1, Bogar Pineda-Terreros1, Rubén Solis Armenta 1, Brenda Lizeth Acosta-Maldonado1, Manuel Valero Saldaña1 P280 EFFICACY AND SAFETY OF RUXOLITINIB FOR PEDIATRIC ACUTE AND CHRONIC GRAFT-VERSUS-HOST DISEASE - A SINGLE CENTER EXPERIENCE Shu-Wei Chou 1, Meng-Yao Lu1, Hsiu-Hao Chang1, Yung-Li Yang1, Shiann-Tarng Jou1 P281 SYSTEMATIC LITERATURE REVIEW ON THE SAFETY, EFFICACY, AND EFFECTIVENESS OF EATG VERSUS COMPARATORS FOR GVHD Erin Sheffels1, Kevin Kallmes1, Keith Kallmes1, John Pederson1, Barbara Możejko-Pastewka 2, Raj Gokani2, Judith Hey-Hadavi2, Andres Quintero2 P282 A SERIES OF STEROID-REFRACTORY, SEVERE CHRONIC GRAFT- VERSUS- HOST DISEASE CASES, TREATED WITH RUXOLITINIB- A SINGLE-CENTER EXPERIENCES Ewa Karakulska-Prystupiuk 1, Agnieszka Tomaszewska1, Piotr Boguradzki1, Piotr Kacprzyk1, Grzegorz Władysław Basak1 P283 BELUMOSUDIL FOR SEVERE, REFRACTORY CHRONIC GVHD IN PEDIATRIC PATIENT: CASE REPORT Agnieszka Sobkowiak-Sobierajska 1, Maksymilian Deręgowski1, Sandra Rutkowska1, Jacek Wachowiak1 P284 CLINICAL EVOLUTION OF PATIENTS WITH GRAFT-VERSUS-HOST DISEASE - THE EXPERIENCE OF A SINGLE CENTER Ana Salselas 1, Pedro Leite-Silva2,1, Sara Lopes1, Lucia Vieira1, Susana Roncon1 P285 USE OF MYCOPHENOLATE MOFETIL FOR GRAFT VERSUS HOST DISEASE PROPHYLAXIS IN PATIENTS UNDERGOING HEMATOPOIETIC STEM CELLS TRANSPLANTATION FROM UNRELATED DONORS Davide Pio Abagnale1, Andrea Cacace1, Giuseppe Gaeta1, Maria Luisa Giannattasio1, Linda Piccolo1, Francesco Grimaldi1, Simona Avilia1, Mara Memoli1, Lucia Ammirati2, Valentina Maglione2, Alessandro Severino1, Catello Califano2, Fabrizio Pane1, Giorgia Battipaglia 1 P286 RETROSPECTIVE ANALYSIS OF RUXOLITINIB TREATMENT FOR THE MANAGEMENT OF STEROID REFRACTORY GRAFT VERSUS HOST DISEASE IN ADULTS WITH HEMATOLOGIC MALIGNANCIES Bogar Pineda Terreros1, Araceli Leal Alanis1, Brenda Lizeth Acosta Maldonado1, Luis Manuel Valero Saldaña1, Rubén Solís Armenta 1, María Alejandra Núñez Atahualpa1, Anghela Milenka Mendoza Sánchez1, Liliana Mey Len Rivera Fong1 P287 DR3 AND GITR AGONISTS BOOST ENDOGENOUS TREG POOL TO PREVENT ACUTE GVHD Juan Fernando Gamboa Vargas 1, Isabell Lang2, Svetlana Stepanzow2, Olena Zaitseva2, Estibaliz Arellano Viera1, Carolin Graf1, Haroon Shaikh1, Muhammad Daud1, Hermann Einsele3, Harald Wajant2, Andreas Beilhack1 P288 ROLE OF BCL2 IN GRAFT VERSUS HOST DISEASE José Antonio Bejarano García1,2, Melanie Nuffer1,2, Mª José Palacios Barea1,2, Luzalba Sanoja Flores1,2, Mª Reyes Jiménez León1,2, Javier Delgado Serrano2, Teresa Caballero Velázquez2,1,3, Alfonso Rodríguez Gil1,2,3, José Antonio Pérez Simón 2,1,3 P289 DIMETHYL FUMARATE INHIBITS TFH DIFFERENTIATION AND CGVHD VIA NRF2 Huanle Gong 1, Fulian Lv1, Yang Xu1, Depei Wu1 P290 SPECIFIC INHIBITION OF PATHOLOGICAL ANGIOGENESIS TO DECREASE ACUTE GVHD Katarina Riesner 1, Lydia Verlaat1, Sarah Mertlitz1, Angela Jacobi2, Hadeer Mohamed Rasheed1,3, Pedro Vallecillo Garcia1, Beate Anahita Jung1, Ningyu Li1, Martina Kalupa1, Claudine Fricke1, Bertina Mandzimba-Maloko1, Laura Barrero1, Lars Bullinger1, Olaf Penack1 P291 ENDOTHELIAL-TO-MESENCHYMAL TRANSITION IN CHRONIC GVHD Katarina Riesner 1, Lydia Verlaat1, Sarah Mertlitz1, Hadeer Mohamed Rasheed1,2, Pedro Vallecillo Garcia1, Beate Anahita Jung1, Ningyu Li1, Martina Kalupa1, Claudine Fricke1, Bertina Mandzimba-Maloko1, Lars Bullinger1, Bruce Blazar3, Olaf Penack1 P292 RAPID ADAPTATION OF DONOR REGULATORY T CELLS (TREG) TO RECIPIENT TISSUES FOR THE CONTROL OF ACUTE GRAFT-VERSUS-HOST DISEASE (GVHD) Franziska Pielmeier 1, David J. Dittmar1, Nicholas Strieder2, Alexander Fischer1, Michael Herbst1, Hanna Stanewsky1, Niklas Wenzl2, Eveline Röseler2, Rüdiger Eder1, Claudia Gebhard2, Lucia Schwarzfischer-Pfeilschifter1, Christin Albrecht1, Wolfgang Herr1, Michael Rehli1,2, Petra Hoffmann1,2, Matthias Edinger1,2 P293 INHIBITION OF MYELOPEROXIDASE AMELIORATES MURINE ACUTE GRAFT-VERSUS-HOST DISEASE Michelle Klesse 1, Sebastian Schlaweck1, Oliver Schanz1, Peter Brossart1, Annkristin Heine1 P294 IFN-GAMMA INDUCES ACUTE GRAFT-VERSUS-HOST DISEASE BY PROMOTING HMGB1-MEDIATED NUCLEAR-TO-CYTOPLASM TRANSLOCATION AND AUTOPHAGIC DEGRADATION OF P53 Xu Yajing 1, Wang Shiyu1, Cheng Tingting1, Chen Xu1, Zeng Cong1, Qin Wei1 P295 DEFINING THE TRANSCRIPTOMIC LANDSCAPE OF ACUTE SKIN GVHD AT SINGLE CELL RESOLUTION Callum Wright 1, Jason Lam1, Lucas Cortes1, Paul Milne1, Jamie Macdonald1, Laura Jardine1, Rafiqul Hussain1, Jonathan Coxhead1, Rachel Queen1, Megan Hasoon1, Adrienne Unsworth1, Michael McCorkindale1, Erin Hurst2, Amy Publicover2, Venetia Bigley1, Matthew Collin1 P296 IMPROVING THE THERAPEUTIC EFFECT OF HUMAN UMBILICAL CORD MESENCHYMAL STEM CELLS ON ACUTE GRAFT-VERSUS-HOST DISEASE BY CRISPLD2-OVEREXPRESSION Qing Xu1,2, Ya Zhou1, Yuxi Xu1, Rui Wang1, Xiaoqi Wang1, Shijie Yang1, Qingxiao Song 1,2, Xi Zhang1,2 P297 RUXOLITINIB USE DURING DONOR STEM CELL MOBILIZATION RESULTS IN RECIPIENTS IMMUNOMODULATION POST BONE MARROW TRANSPLANT IN MURINE MODEL AND MIGHT REDUCE GRAFT VERSUS HOST DISEASE Vamsi Kota 1, Ahmet Alptekin1, Hasanul Chowdhury1, Mahrima Parvin1, Evila Sales2, Amanda Barrett3, Babak Baban2, Ali Arbab1, Ravindra Kolhe3 P298 ROS DEFICIENCY OF RECEIPT DERIVED MDSCS EXACERBATES ALLO-REACTIVE T CELLS RESPONSES DURING HYPERACUTE GVHD Yigeng Cao1, Jiali Wang1, Zihan Zhao1, Erlie Jiang 1, Yuanfu Xu1 P299 DIAGNOSIS OF CUTANEOUS ACUTE GRAFT-VERSUS-HOST THROUGH CIRCULATING PLASMA MIR-638, MIR-6511B-5P, MIR-3613-5P, MIR-455-3P, MIR-5787, AND MIR-548A-3P AS PROSPECTIVE NONINVASIVE BIOLOGICAL INDICATORS IN ACUTE MYELOID LEUKEMIA Marjan Yaghmaie 1, Marzieh Izadifard1, Mohammad Ahmadvand1, Hossein Pashaiefar1, Kamran Alimoghadam1, Amir Kasaeian1, Maryam Barkhordar1, Ghazal Seghatoleslami1, Mohammad Vaezi1, Ardeshir Ghavamzadeh2 P300 THE IMPORTANCE OF CYTOGENETIC BONE MARROW TESTS IN POST-TRANSPLANT CARE OF PATIENTS AFTER ALLO-HSCT DUE TO ACUTE MYELOID LEUKEMIA OR MYELODYSPLASTIC SYNDROME- A SINGLE-CENTER ANALYSIS Ewa Karakulska-Prystupiuk 1, Agnieszka Stefaniak1, Anna Kulikowska1, Krzysztof Madry1, Agnieszka Tomaszewska1, Piotr Boguradzki1, Tomasz Stokłosa1, Grzegorz Władysław Basak1 P301 INFECTION BY HUMAN HERPES VIRUS 6 IN ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN THE POST-TRANSPLANT CYCLOPHOSPHAMIDE ERA Paola Charry1, Virginia Rodríguez1, Ares Guardia1, Teresa Solano1, Jordi Arcarons1, Joan Cid1, Miquel Lozano1, Laura Rosiñol1, Carmen Martínez1, María Queralt Salas1, Francesc Fernandez-Avilés1, Mª Ángeles Marcos1, Montserrat Rovira1, María Suárez-Lledó 1 P302 SECOND HAPLOIDENTICAL HAEMATOPOIETIC STEM CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE AS RESCUE STRATEGY FOR HIGH RISK CHILDREN WITH LEUKEMIA Giorgio Ottaviano1, Maria Barazzoni2, Federica Acone2, Francesca Vendemini1, Giulia Prunotto1, Alex Moretti2, Pietro Casartelli1, Sara Napolitano1, Marta Verna1, Sonia Bonanomi1, Adriana Balduzzi 1,2 P303 DISTINCT CLINICO-BIOLOGICAL FEATURES IN NPM1-MUTATED ACUTE MYELOID LEUKEMIA PATIENTS FOLLOWING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION Xiaolin Yuan 1,2,3,4, Yibo W1,2,3,4, Xiaoyu Lai1,2,3,4, Lizhen Liu1,2,3,4, Jimin Shi1,2,3,4, Weiyan Zheng1,2,3,4, Jian Yu1,2,3,4, Yanmin Zhao1,2,3,4, Jie Sun1,2,3,4, Zhen Cai1,2,3,4, He Haung1,2,3,4, Yi Luo1,2,3,4 P304 DIFFERENT SOURCES OF HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH THALASSEMIA MAJOR: RELATIONSHIP BETWEEN IMMUNE RECONSTITUTION, GRAFT-VERSUS-HOST DISEASE, AND VIRAL INFECTIONS Jianyun Liao 1, Chaoke Bu1, Lan He1, Huaying Liu1, Zhiyong Peng1, Jujian He1, Weiwei Zhang1, Yuqian Xia1, Yuelin He1, Chunfu Li1 P305 COMBINATION OF POST-TRANSPLANT CYCLOPHOSPHAMIDE WITH ANTITHYMOCYTE GLOBULIN AFTER HAPLOIDENTICAL ALLOGENEIC HEMATOPOIETIC STEM-CELL TRANSPLANTATION: A SINGLE CENTER ANALYSIS Chiara Bernardi 1,2, Amandine Pradier1,2, Anne-Claire Mamez1, Federica Giannotti1, Sarah Morin1, Sisi Wang1,2, Astrid Melotti1,2, Yves Chalandon1, Federico Simonetta1,2, Starvoula Masouridi-Levrat1 P306 TRIPLE STEM CELL INFUSION ALLEVIATE GRAFT VERSUS HOST DISEASE AND IMPROVE OUTCOME IN UNMANIPULATED HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION Fang Hua1,2, Shan Zhang1, Xiao-Mei Zhang1, Yan Deng1, Ying Han1, Si-Han Lai1, Ying He1, Lei Ma1, Xu-Pai Zhang1, Dan Chen1, Yi Su1, Ling Zhang1, Hui Yang1, Rong Huang1, Guang-Cui He 1, Hao Yao1, Hai Yi1 P307 HIGH-DOSE CHEMOTHERAPY WITH AUTOLOGOUS BONE MARROW TRANSPLANTATION IN MULTI-REFRACTORY TESTICULAR GERM CELL CANCER: A RETROSPECTIVE SINGLE INSTITUTIONAL ANALYSIS Andrea Tamayo1, Adolfo Sáez1, Reyes Mas1, Lucia Medina1, Guillermo Ramos1, Esther Parra1, Alberto Blanco1, Clara Cuellar1, Paula Lázaro1, Ana Jimenez1, Rafael Alonso1, Tycho Baumann1, Jose Sanchez Pina 1, Pilar Martinez1, Enrique Gonzalez1, Joaquin Martinez1, Maria Calbacho1 P308 KIR-MISMATCH IMPACTS THE OUTCOMES AFTER HLA-HAPLOIDENTICAL STEM-CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE Jairo Eduardo Niño-Ramirez1, Francisco Javier Gil-Etayo1, Marta Fonseca Santos1, Isabel Jiménez-Hernaz1, Ariadna Vicente-Parra1, Pilar Terradillos-Sánchez1, Ana Balanzategui1, Francisco Boix1, Miguel Alcoceba1, Almudena Navarro-Bailón1, Estefanía Pérez-López1, Mónica Cabrero1, Fermín Sanchez-Guijo1, Ramón García-Sanz1, Lucía López-Corral 1, Amalia Tejeda-Velarde1 P309 NOVEL AUTOMATIC APPLICATION FOR ENRICHMENT OF CD34 + CELLS FROM APHERESIS PRODUCTS USING THE CLINIMACS PRODIGY Julia Dzionek 1, Stephanie Soltenborn1, Valeriya Olevska1, Svenja Oberbörsch1, Christin Bosbach1, Felix Hebbeker1, Burgund Kauling1, Juliane Raasch1, Carina Wenzel1, Heike Lahnor2, Katharina Krämer2, René Meißner2, Eleni Papanikolaou1, Andreas Bosio1 P310 OUTCOMES OF ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH WISKOTT-ALDRICH SYNDROME Samin Sharafian1,2, Su Han Lum1,3, Christo Tsilifis1,3, Terry Flood1, Eleri Williams1, Stephen Owens1, Sophie Hambleton1,3, Andrew Gennery1,3, Mary Slatter 1,3, Zohreh Nademi1,3 P311 STEM CELL TRANSPLANT ACTIVITY IN KYIV NATIONAL CANCER INSTITUTE IN THE CONDITIONS OF FULL-SCALE RUSSIAN AGGRESSION IN UKRAINE Nazar Shokun 1, Alevtyna Burtna1, Maryna Bushuieva1, Moiseienko Kateryna1, Irina Kryachok1, Yana Stepanishyna1 P312 HOME-BASED HEMATOPOIETIC CELL TRANSPLANTATION IS FEASIBLE AND SAFER IN TERMS OF NUTRITIONAL PARAMETERS, TOXICITY AND LENGTH OF HOSPITALIZATION STAY Javier Cornago Navascués1, Laura Pardo Gambarte 1, Juan Carlos Caballero Hernáez1, María Carolina Dassen1, Ana Rodríguez-Calvo Parra1, Alberto Sánchez Donaire1, Jesús Ignacio Merlo Luis1, José Luis López Lorenzo1, Amalia Domingo González1, Pilar Llamas Sillero1, Laura Solán Blanco1 P313 A RISK-ADAPTED STRATEGY TO SELECT AUTOLOGOUS OR ALLOGENEIC TRANSPLANT CONSOLIDATION IS EFFECTIVE IN MATURE T-CELL LYMPHOMA: A SINGLE CENTER EXPERIENCE Alessio Maria Edoardo Maraglino1, Simona Sammassimo1, Viviana Amato1, Anna Vanazzi1, Paulina Maria Nierychlewska1, Ginevra Lolli1, Simonetta Viviani1, Tommaso Radice1, Fabio Giglio 1, Federica Gigli1, Rocco Pastano1, Corrado Tarella1, Enrico Derenzini1,2 P314 EFFECTS OF THE PERIPHERAL EXPANSION OF CYTOTOXIC T LYMPHOCYTES ON OUTCOMES FOLLOWING HAPLOIDENTICAL ALLOGENEIC STEM CELL TRANSPLANTATION Lucía García Tomás1, Oriana Jimena López Godino 1, Cristina Aroca Valverde1, Inmaculada Heras Fernando1, María Luisa Lozano Almela1 P315 OUTCOME OF ALLO-HSCT IN THERAPY RELATED MYELOID NEOPLASMS WITH A HISTORY OF OVARIAN CANCER PREVIOUSLY TREATED WITH CHEMOTERAPY AND PARP INHIBITORS Alessio Maria Edoardo Maraglino1, Simona Sammassimo1, Patrizia Chiusolo2, Cristina Papayannidis3, Federica Gigli1, Filippo Frioni2, Chiara Sartor4, Viviana Amato1, Rocco Pastano1, Elisabetta Todisco5, Corrado Tarella1, Enrico Derenzini1,6, Fabio Giglio 1 P316 EFFECTS OF LOW-DOSE HYPOMETHYLATING AGENTS ON THE PREVENTION OF RELAPSE AFTER TRANSPLANTATION IN PEDIATRIC HIGH-RISK AML Qingwei Wang1, Shengqin Cheng1, Wei Gao1, Qi Ji1, Peifang Xiao1, Jun Lu1, Jie Li1, Shaoyan Hu 1 P317 OUTCOMES OF PEDIATRIC CHRONIC MYELOID LEUKEMIA IN ADVANCED PHASE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH REDUCED-INTENSITY CONDITIONING REGIMEN Polina Sheveleva1, Elena Morozova1, Anna Osipova1, Olesya Paina1, Olga Slesarchuk1, Tatyana Gindina1, Ekaterina Izmailova1, Ildar Barkhatov1, Tatyana Bykova 1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1 P318 UPDATE ON DONOR DERIVED NK CELLS INFUSION FOLLOWING HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANT IN PEDIATRIC HIGH-RISK AND RELAPSED/REFRACTORY LEUKEMIA Emanuela Rossitti1, Fabiana Cacace2, Flavia Antonucci1, Valeria Caprioli2, Maria Rosaria D’amico2, Giuseppina De Simone2, Maria Simona Sabbatino2, Mario Toriello2, Giovanna Maisto2, Roberta Penta de Vera d’aragona2, Francesco Paolo Tambaro 2 P319 COMPARISONS OF SAFERTY AND EFFICACY OF ALLO-HSCT AFTER CAR T-CELL OR CHEMOTHERAPY-BASED COMPLETE REMISSION IN PEDIATRIC T-ALL Ruijuan Sun 1, Jianping Zhang1, Yue Lu1, Yanli Zhao1, Deyan Liu1, Xingyu Cao1, Min Xiong1, Jiarui Zhou1, Zhijie Wei1 P320 EFFECT OF CYTOMEGALOVIRUS REACTIVATION ON RELAPSE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN ACUTE LYMPHOBLASTIC LEUKEMIA Mekni Sabrine1, Ben Abdeljelil Nour 1, Rihab Ouerghi1, Turki Ines1, Khayati Malek1, Ben Yaiche Insaf1, Frigui Siwar1, Torjemane Lamia1, Belloumi Dorra1, Kanoun Rimmel Yosra1, Ladeb Saloua1, Ben Othman Tarek1 P321 LOWER INCIDENCE OF CHRONIC GVHD WITH ATLG AS GVHD PROPHYLAXIS IN ALL PATIENTS IN FIRST COMPLETE REMISSION UNDERGOING ALLOGENEIC SCT COMPARED TO POST-TRANSPLANT CYCLOPHOSPHAMIDE Normann Steiner 1, Radwan Massoud1, Evgeny Klyuchnikov1, Nico Gagelmann1, Johanna Richter1, Christian Niederwieser1, Kristin Rathje1, Ameya Kunte1, Jannik Engelmann1, Christina Ihne1, Tatiana Urbanowicz1, Iryna Lastovytska1, Cecilia Lindhauer1, Franziska Marquard1, Mirjam Reichard1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schäfersküpper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Silke Heidenreich1, Gabriele Zeck1, Ina Rudolph1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger1 P322 LONGITUDINAL FOLLOW-UP ON HUMORAL AND T CELL IMMUNE RESPONSES AFTER COVID-19 VACCINATION IN ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTED PATIENTS Alma Wegener 1, Line Dam Heftdal1, Sebastian Rask Hamm1, Cecilie Bo Hansen1, Laura Pérez-Alós1, Andreas Runge Poulsen1, Dina Leth Møller1, Kamille Fogh2, Mia Pries-Heje1, Rasmus Bo Hasselbalch2, Sisse Rye Ostrowski1, Ruth Frikke-Schmidt1, Erik Sørensen1, Linda Hilsted1, Marietta Nygaard1, Niels Smedegaard Andersen1, Lone Smidstrup Friis1, Søren Lykke Petersen1, Ida Schjødt1, Brian Kornblit1, Henrik Sengeløv1, Henning Bundgaard1, Peter Garred1, Kasper Iversen2, Kirsten Grønbæk1, Susanne Dam Nielsen1, Lars Klingen Gjærde1 P323 HAPLOIDENTICAL DONOR PERIPHERAL BLOOD STEM CELL TRANSPLANTATION USING THIRD-PARTY CORD BLOOD COMPARED WITH MATCHED UNRELATED DONOR TRANSPLANTATION FOR PATIENTS WITH HEMATOLOGIC MALIGNANCIES Xia Ma1, Yan Chen1, Yi Liu1, Tingting Cheng1, Cong Zeng1, Xu Chen1, Shiyu Wang1, Juan Hua1, Yajing Xu 1 P324 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH PERIPHERAL T-CELL LYMPHOMAS: A RETROSPECTIVE MULTICENTER STUDY IN CHINA Hongye Gao1, Jiali Wang1, Zhuoxin Zhang1, Hao Zhang2, Xin Du3, Dehui Zou1, Xianmin Song4, Erlie Jiang 1 P325 SAFETY AND EFFICACY OF POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR MUD, MMUD AND HAPLOIDENTICAL STEM CELL TRANSPLANTS IN PAEDIATRIC POPULATION Gloria Miguel 1, Julia Marsal1, Maria Trabazo1, Cristina Rivera1, Montserrat Rovira2, Laura Jimenez1, Paula Mazorra1, Julio Ropero3, Ana Maria Infante4 P326 BEYOND BORDERS: HSCT’S GLOBAL SITUATIONS AND INDONESIA’S PURSUIT OF HEALTH EQUITY Daniel Rizky 1, Ridho M. Naibaho2, Mohammed Bader Obeidat3, Mabruratussania Maherdika1, Budi Setiawan1, Eko Adhi Pangarsa1, Damai Santosa1, Catharina Suharti4, Marco A. Salvino5,6, Abel Santos Carreira7, Annalisa Paviglianiti7, Marta Peña7, Alberto Mussetti7, Anna Sureda7 P327 EFFECTIVENESS OF THE SIMPLIFIED COMORBIDITY INDEX COMPARED TO THE HCT-CI AS A PREDICTOR OF TRANSPLANT RELATED MORTALITY AND OVERALL SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION Maria Alejandra Nunez Atahualpa 1, Ruben Solis Armenta1, Anghela Milenka Mendoza Sánchez1, Araceli Leal Alanis1, Bogar Pineda Terreros1, Liliana Rivera Fong1, Brenda Lizeth Acosta Maldonado1, Luis Manuel Velero Saldaña1 P328 8 VS 12 GRAY TBI IN ALLOGENEIC SCT FOR ALL PATIENTS RESULTS IN SIMILAR SURVIVAL, BUT IMPROVED SURVIVAL FOR MRD + PATIENTS AFTER 12 GRAY Normann Steiner 1, Radwan Massoud1, Evgeny Klyuchnikov1, Nico Gagelmann1, Johanna Richter1, Christian Niederwieser1, Kristin Rathje1, Ameya Kunte1, Tatiana Urbanowicz1, Jannik Engelmann1, Christina Ihne1, Iryna Lastovytska1, Cecilia Lindhauer1, Franziska Marquard1, Mirjam Reichard1, Gabriele Zeck1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schäfersküpper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Silke Heidenreich1, Ina Rudolph1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger1 P329 POST-TRANSPLANT MAINTENANCE THERAPY WITH AZACITIDINE FOR HIGH-RISK MYELOID MALIGNANCIES: WHO BENEFITS THE MOST? Linli Lu1, Xin Li 1, Qian Cheng1 P330 PHYSICIAN PERSPECTIVES ON THE BURDEN AND CLINICAL MANAGEMENT OF CYTOMEGALOVIRUS (CMV) INFECTION IN TRANSPLANT RECIPIENTS IN CENTRAL AND EASTERN EUROPE Radovan Vrhovac 1, Alicja Dębska-Ślizień2, Tina Roblek3, Aleksandar Biljić Erski4, Milena Todorović Balint5 P331 THE DOSES OF POST-TRANSPLANTATION CYCLOPHOSPHAMIDE IN HAPLOIDENTICAL STEM CELL ALLOGRAFTS CAN BE REDUCED Juan Carlos Olivares-Gazca1, María de Lourdes Pastelín-Martínez2,1, Merittzel Abigail Montes-Robles2,1, Moisés Manuel Gallardo-Pérez1, Edgar Jared Hernández-Flores1,3, Max Robles-Nasta1,3, Daniela Sánchez-Bonilla1, César Homero Gutiérrez-Aguirre4, Andrés Gómez-De-León4, David Gómez-Almaguer4, Guillermo José Ruiz-Delgado 1,3, Guillermo José Ruiz-Arguelles1,3 P332 OUTCOMES WITH ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN THERAPY RELATED ACUTE MYELOID LEUKEMIA: A SYSTEMATIC REVIEW AND META-ANALYSIS Moazzam Shahzad 1, Muhammad Kashif Amin2, Muhammad Fareed Khalid3, Iqra Anwar2, Michael Jaglal1 P333 OUTCOME OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR THERAPY RELATED ACUTE MYELOID LEUKEMIA: MULTICENTER REAL-LIFE EXPERIENCE Alessandra Sperotto1, Matteo Leoncin2, Chiara Cigana3, Roberta De Marchi1, Carla Filì4, Albana Lico5, Corinna Greco5, Marco Petrella6, Endri Mauro7, Eleonora De Bellis8, Ilaria Tanasi9, Federico Mosna10, Giulia Ciotti1, Cristina Skert2, Mariagrazia Michieli3, Alberto Tosetto5, Carmela Gurrieri6, Francesco Zaja8, Mauro Krampera9, Michele Gottardi1, Costanza Fraenza 2 P334 OUTCOMES OF SECOND ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR HEMATOLOGIC DISEASES: A SINGLE-CENTER REAL-WORLD EXPERIENCE Marta Castelli 1, Anna Grassi2, Alessandra Algarotti2, Maria Caterina Micò2, Federico Lussana1, Maria Chiara Finazzi1, Chiara Pavoni2, Gianluca Cavallaro2, Federico Mazzon1, Giuliana Rizzuto2, Benedetta Rambaldi2, Alessandro Rambaldi1 P335 PERSPECTIVES OF GENERAL HEMATOLOGISTS ON A PROPOSED SHARED CARE MODEL FOR HSCT IN SAUDI ARABIA FOR SICKLE CELL DISEASE Abdulrahman Alsultan 1, Mohsen Alzahrani2, Mohammed Essa3, Abdullah Aljefri4, Hatoon Ezzat5, Wasil Jastaniah6 P336 DOES AGE MATTER? IMPACT OF DONOR AGE IN ENGRAFTMENT AND GRAFT FUNCTION AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION Oana Enache1, Lavinia Eugenia Lipan2,1, Oana Gabriela Craciun2, Andra Georgiana Stoica2, Adela Ionela Ranete2, Ileana Constantinescu2,1, Alina Daniela Tanase 2,1 P337 COMPARABLE RESULTS OF HAPLOIDENTICAL AND MATCHED RELATED HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR PATIENTS WITH BLAST PHASES CHRONIC MYELOID LEUKEMIA Darina Zammoeva1, Renat Badaev 1, Yuliya Alekseeva1, Dmitriy Motorin2, Elza Lomaia1 P338 ANALYSIS OF THE EFFICACY OF LUSPATERCEPT IN THE TREATMENT OF ANEMIA AFTER ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION Lei Dong 1, Wei Ma1, Liu Chan-Chan1, Zhang Fang-Fang1, Tian Yue-Feng1, Liu Ji-Cong1, Zhao Xiao-Zhen1, He Yang2, Cao Xing-Yu1 P339 THE IMPACT OF THE TRAVELED DISTANCE TO THE TRANSPLANT CENTER ON THE CLINICAL OUTCOME OF ALLOGENEIC BONE MARROW TRANSPLANT PATIENTS Jean El-Cheikh 1, Mustafa Saleh1, Khodr Terro1, Radwan Massoud2, Ghassan Bidaoui1, Layal Sharrouf1, Sara Chehayeb1, Toufik Sebai3, David Karam1, Ammar Zahreddine1, Rita Nehme1, Iman Abou Dalle1, Nour Moukalled1, Ali Bazarbachi1 P340 THE CLINICAL ANALYSIS OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN THERAPY-RELATED MYELOID NEOPLASMS Shu Yan1, Deyan Liu1, Fang Xu 1 P341 HAPLOIDENTICAL TRANSPLANTATION FOR PATIENTS WITH FANCONI ANEMIA HAPLOIDENTICAL TRANSPLANTATION FOR PATIENTS WITH FANCONI ANEMIA Eman Khattab 1, Hasan Hashem1, Mayada Abu Shanap1, Iyad Sultan1, Rawad Rihani1 P342 NK-CELL INFUSIONS TO PREVENT RELAPSE FOLLOWING HEMATOPOIETIC STEM CELL TRANSPLANTATION Maryia Navumovich1, Tatsiana Shman1, Yuliya Mareika1, Volha Mishkova1, Katsiaryna Vashkevich1, Natalia Kirsanava1, Iryna Pakhomova1, Viktoryia Belabokava1, Dmitriy Prudnikov1, Lubov Zherko1, Nina Minakovskaya 1, Anzhalika Solntsava1 P343 SURVIVAL OF PATIENTS WITH MULTIPLE MYELOMA WITH ASCT IN ONCOSALUD - AUNA BETWEEN 2015 AND 2021 Alonso Diaz1, Claudio J. Flores1, Shirley Quintana 2, Cesar Samanez2 P344 RESOLUTION OF VOD/SOS WITH DEFIBROTIDE AFTER HSCT AND PERSISTENT HEPATIC DEMAGE: MONOCENTRIC PEDIATRIC EXPERIENCE Fabiana Cacace1, Emanuela Rossitti2, Valeria Caprioli1, Flavia Antonucci2, Maria Rosaria D’amico1, Giuseppina De Simone1, Maria Simona Sabbatino1, Francesco Paolo Tambaro 1 P345 ACCURATE DIETARY EVALUATION AND ADEQUATE NUTRITIONAL THERAPY ARE INDISPENSABLE FOR BETTER OUTCOMES OF ALLO-HSCT Yuki Mori 1, Koichi Nakase1, Toshimi Noma1, Tomoya Katsuta1, Ryuichiro Hiyama1, Yuki Goto1, Ryo Ueda1, Risa Hashida1, Kyoko Itakusu1, Kyosuke Saeki1, Masakazu Mori1, Yuichiro Nawa1 P346 ALLOGENOUS BONE MARROW TRANSPLANT IN HIGH-RISK CHRONIC MYELOID LEUKEMIA IN THE ERA TKI INHIBITOR: A SYSTEMATIC REVIEW Nia Novianti Siregar 1, Resti Mulya Sari1, Andree Kurniawan2, Felix Wijovi2, Devina Adella Halim2, Patricia Angel2, Rivaldo Steven Heriyanto2 P347 LONELINESS AND RUMINATIONS IN CANCER PATIENT-CAREGIVER DYADS – ANALYSIS OF DAILY FLUCTUATIONS Małgorzata Sobczyk-Kruszelnicka1, Aleksandra Kroemeke2 P348 PATIENTS WITH FOLLICULAR LYMPHOMA AND CANDIDATES FOR AUTOLOGOUS TRANSPLANTATION: IS THE COLLECTION OF HEMATOPOIETIC PROGENITORS EFFICIENT AFTER THE FIRST LINE OF TREATMENT? María Casado Sánchez1, Sara Hormaza de Jauregui1, Juan José Mateos Mazón1, Javier Arzuaga Méndez1, Xabier Martín Martitegui1, Paula María Zoco Gallardo 1, Laura Aranguren del Castillo1, Ariadna García Ascacibar1, Gorka Pinedo Martín1, María Elena Amutio Díez1, Juan Carlos García Ruiz1 P349 HAPLOIDENTICAL BONE MARROW TRANSPLANTATION IN A COLOMBIAN BMT UNIT: CLINICAL DESCRIPTION AND OUTCOMES Jorge Cuervo sierra 1, Germán A. Velasquez-Quintero1, Juan Felipe Jaramillo-Alvarez1, Wang Nataly Montoya-Arbelaez1, Sandra Gisella Martinez-Jimenez1, Luz Maridier Tobon-Tobon1, Jurany Andrea Marin-Montoya1, Deisy Johana Giraldo-Gomez1, Luisa Fernanda Castro-Quintero1, Maira Alejandra Mesa-Giraldo1, Maribel Londoño-Buitrago1 P350 HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN FANCONI ANEMIA: A SINGLE CENTER EXPERIENCE Guzmán López de Hontanar 1,2,3, June Iriondo2,3, Josune Zubicaray2,3, Elena Sebastián2,3, Blanca Molina2,3, Sara Vinagre2,3, Marta González-Vicent2,3, Julián Sevilla2,3 P351 EARLY AUTOLOGOUS AND/OR ALLOGENEIC STEM CELL TRANSPLANTATION FOR ADULT PATIENTS WITH ADVANCED STAGE T- LYMPHOBLASTIC LEUKEMIA/LYMPHOMA OR BURKITT LYMPHOMA. A RETROSPECTIVE SINGLE-CENTRE ANALYSIS Normann Steiner 1, Katherina Baier1, Denise Ritter1, Gabriele Hetzenauer1, Jakob Rudzki1, Stefan Köck1, Eberhard Gunsilius1, Brigitte Kircher1, Dominik Wolf1, David Nachbaur1 P352 MONOCYTES IN ALLO-HSCT WITH AGED DONORS SECRET IL-1/IL-6/TNF TO INCREASE THE RISK OF GVHD AND DAMAGE THE AGED HSCS Xia Li 1, Wanying Zhang2, Yanan Wang2, Chentao Li2, Yibo Wu1, Xiaoyu Lai1, Yi Luo1, Pengxu Qian1, He Huang1 P353 SETTING UP THE FIRST PEDIATRIC BONE MARROW TRANSPLANTATION UNIT IN THE UAE AND THE EXPERIENCE OF COMPLETING THE FIRST 30 PEDIATRIC BMTS WITHOUT ANY MORTALITY Zainul Aabideen 1, Sagar Mohan Nivargi1, Abdullah Odat1, Mohammed Alfar1, Charbel Khalil1, Rakeshkumar Shah1, Kesava Ramakrishnan1, Rajesh Phatak2, Ahmed Elsheikh1, Mohamed Abdelsamad1, Krishnappa Venkatesh1, Ghulam Mujtaba1, Dr. Georgey Koshy1, Amro El Saddik3, Humaid Alshamsi4, Fulvio Porta5, Lawrence Faulkner6, Mohammed Hamid1 P354 POST-TRANSPLANT CYCLOPHOSPHAMIDE ± ANTI-THYMOCYTE GLOBULIN IN PATIENTS WITH AML/MDN UNDERGOING ALLOGENEIC HCT WITH PBSC FROM HAPLOIDENTICAL DONOR: A SINGLE-CENTER EXPERIENCE Francesca Cavallaro 1, Marta Canzi1,2, Fabio Serpenti1, Maria Cecilia Goldaniga1, Kordelia Barbullushi1, Giulia Galassi3, Francesco Onida4,2, Francesco Passamonti1,2, Giorgia Natascia Saporiti1 P355 DESCRIPTION OF PATIENTS WITH RELAPSED/REFRACTORY HODGKIN LYMPHOMA WHO UNDERWENT TO ALLOGENEIC STEM CELL TRANSPLANTATION IN A REFERENCE CENTER IN BOGOTA, COLOMBIA Carlos Fernando Gómez Calcetero 1, Angie Paola Guarin Castañeda1, Oscar Javier Peña Ardila1, Gloria Elena Mora Figueroa1, Cesar Lamadrid Sastre1, Enrique Pedraza Mesa1, Gerson Menoyo Caballero1, Gustavo Adolfo Martinez Salazar1, Licet Villamizar Gómez1 P356 TREOSULFAN-BASED HAPLOIDENTICAL T CELL DEPLETED HSCT FOR ADOLESCENTS AND YOUNG ADULTS (AYA) WITH ADVANCED STAGE SICKLE CELL DISEASE AND THALASSEMIA Anja Troeger 1, Juergen Foell1, Katharina Kleinschmidt1, Gina Penkivech1, Tarek Hanafee-Alali1, Andreas Brosig1, Robert Offner1, Daniel Wolff1, Selim Corbacioglu1 P357 COMPARISON OF THE UNRELATED TRANSPLANTATION RESULTS OF AN HLA FULLY MATCHED DONOR AND A HOMOZYGOUS MM DONOR AT ONE LOCUS IN PATIENTS WITH BETA THALASSEMIA Koray Yalçın 1,2, Safiye Suna Çelen1, Seda Öztürkmen3, Hayriye Daloğlu4, Suleimen Zhumatayev5, Vedat Uygun6, Gülsün Karasu5, Akif Yeşilipek3 P358 CLINICAL OUTCOMES AND HEALTHCARE RESOURCE USE IN PATIENTS WITH SICKLE CELL DISEASE WITH RECURRENT VASO-OCCLUSIVE CRISES WHO RECEIVED HEMATOPOIETIC STEM CELL TRANSPLANTS IN FRANCE Jessica Baldwin 1, Chuka Udeze1, Naxin Li1, Lyes Boulmerka1, Lila Dahal1, Giancarlo Pesce2, Nadia Quignot2, Heng Jiang2, Frederic Galactéros3 P359 SINGLE-CENTER EXPERIENCE OF STEM CELL TRANSPLANTATION IN PATIENTS WITH SEVERE SICKLE CELL DISEASE Miriam P. Klahr 1, Amer Assal1, Christian Gordillo1, Ran Reshef1, Monica Bhatia1, Diane George1, Markus Y. Mapara1 P360 HAEMATOPOIETIC STEM CELL TRANSPLANTATION IN A LOW RESOURCE COUNTRY NIGERIA. A 10 YEARS REPORT FROM A SINGLE CENTER Nosakahare Bazuaye 1, Nancy Ojiemhangbe2 P361 IMPACT OF THE FIRST ONLINE WEBINAR OF THE WBMT NURSING GROUP IN COLLABORATION WITH THE LAGOS UNIVERSITY TEACHING HOSPITAL AND SICKLE CELL FOUNDATION OF NIGERIA Adetola Kassim 1, Reggie Belkhedim2, Sebastian Galeano3, Dietger Niederwieser3, Damiano Rondelli4, Abijah Princy B5, Amala Lucas6, Ainy Azra7, Cristina Vogel8, Diana Villalobos9, Eugenia Arjona10, Felicia Michael11, Francisca Negrete12, Gloria Ceballo13, John Murray14, Jorge Morales15, Josephine Suganya6, Kishwar Sultana16, Komal Mundhe17, Latha Gracelin6, Lu Yin18, Megan Hogg11, Michelle Kenyon19, Niti Lotey20, Nouf Shwikan21, Paola Llamas22, Rita Nehme23, Sandeep Elizabeth24, Suman Kubal17, Trish Joyce25, Yvonne Panek-Hudson25, Adriana Seber26, Annette Akinsete27, Edamisan Temiye28, Oluseye Akinstete28, Mahmoud Aljurf21 P362 MATCHED-RELATED HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR INTERMEDIATE AND HIGH RISK THALASSEMIA PATIENTS USING REDUCED INTENSITY CONDITIONING REGIMEN WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE AS GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS. A SINGLE CENTER EXPERIENCE FROM JORDAN Ayad Ahmed Hussein 1, Bayan Majed Alaaraj1, Tariq Ahmed Abdelghani1, Nour Awni Ghanem1, Hadeel Hassan Al-Zoubi1, Dina Mohammad Abu Assab1 P363 ABO INCOMPATIBILITY DID NOT IMPACT TRANSPLANT OUTCOMES FOLLOWING REDUCED INTENSITY HAPLOIDENTICAL BONE MARROW TRANSPLANTATION FOR PATIENTS WITH SICKLE CELL DISEASE: MULTICENTER EXPERIENCE Danielle Murphy 1, Pavina Akhom1, Karina Wilkerson1, Katie Gatwood1, Lindsay Orton1, Jim Connelly1, Reena Jayani1, Olalekan Oluwole1, Bhagirathbhai Dholaria1, Nancy Clayton-Long1, Brittney Baer1, Bipin Savani1, James Jerkins1, Andrew Jallouk1, Mohsen Alzahrani2, Ali D. Alhamari3, Josu de la Fuenta4, Erfan Nur5, Heidi Chen1, Adetola Kassim1 P364 DURABLE ENGRAFTMENT AFTER PHARMACOLOGICAL PRE-TRANSPLANT IMMUNE SUPPRESSION FOLLOWED BY REDUCED-TOXICITY MYELOABLATIVE HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN HIGHLY HLA-IMMUNIZED ADULTS WITH SICKLE CELL DISEASE Sabine Fürst 1, Emmanuelle Bernit2, Faezeh Legrand1, Angela Granata1, Samia Harbi1, Raynier Devillier1, Benjamin Bouchacourt1, Thomas Pagliardini1, Valerio Maisano1, Djamel Mokart1, Claude Lemarié1, Boris Calmels1, Christophe Picard3, Agnès Basire3, Borje S. Andersson4, Didier Blaise1 P365 CLINICAL OUTCOMES AND HEALTHCARE RESOURCE USE IN PATIENTS WITH TRANSFUSION-DEPENDENT Β-THALASSEMIA WHO RECEIVED HEMATOPOIETIC STEM CELL TRANSPLANTS IN FRANCE Jessica Baldwin1, Chuka Udeze 1, Nanxin Li1, Lyes Boulmerka1, Lila Dahal1, Giancarlo Pesce2, Nadia Quignot2, Heng Jiang2, Frederic Galactéros3 P366 THE IMPACT OF PANEL REACTIVE ANTIBODIES IN PEDIATRIC BETA THALASSEMIA PATIENTS UNDERGOING STEM CELL TRANSPLANTATION Gülay Sezgin1 P367 SICKLE CELL DISEASE WITH SEVERE ACUTE CHEST SYNDROME AND HEPATOPATHY: CAN ALLOGENEIC SCT BE PERFORMED IN SUCH DESPERATE CLINICAL SITUATIONS? A CASE REPORT Celia Kaffenberger 1, Eva Rettinger1, Shahrzad Bakhtiar1, André Willasch1, Roland Schrewe1, Michael Merker1, Birgit Knoppke2, Jean-Hugues Dalle3, Jan-Henning Klusmann1, Peter Bader1, Andrea Jarisch1 P368 OUTCOMES FOLLOWING HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR MONOGENIC INFLAMMATORY BOWEL DISEASE Xiaowen Zhai 1, Ping Wang1, Wenjin Jiang1, Hongsheng Wang1, Ying Huang1, Xiaowen Qian1 P369 CLINICAL CHARACTERISATION AND OUTCOME OF 125 CASES WITH GRISCELLI SYNDROME Adriel Roa-Bautista1, Mahreen Sohail2, Reem Elfeky3, Maaike Kusters 3 P370 OUTCOMES FOR ALLOGENEIC SCT FOR PATIENTS WITH INBORN ERRORS OF IMMUNITY UTILIZING REDUCED INTENSITY CONDITIONING REGIMEN WITH TARGETED BUSULFAN Hannah Lust1, Olatundun Williams2, Morris Kletzel1, Reggie Duerst1, Jennifer Schneiderman1, William Tse3, Sonali Chaudhury 1 P371 OUTCOME OF THE PATIENTS DIAGNOSED WITH SEVERE COMBINED IMMUNODEFICIENCY THROUGH NEWBORN SCREENING IN CATALONIA (2017-2023) Laura Alonso1, Andrea Martin-Nalda1, Maria Isabel Benitez Carabante 1, Ana Argudo-Ramirez2, Luz Uria-Ofialdegui1, Judit Garcia-Villoria2, Melissa Panesso1, Jacques Gabriel Riviere1, Pere Soler-Palacin1, Cristina Diaz de Heredia1 P372 HSCT AS A CURATIVE TREATMENT OPTION IN PATIENTS WITH A PRIMARY IMMUNODEFICIENCY DUE TO IL2RB-DEFECT: DESCRIPTION OF A SMALL COHORT Ommo E. Mauss 1, Christo Tsilifis2,3, Eva-Maria Jacobsen1, Manfred Hönig1, Ansgar Schulz1, Mary A. Slatter2,3, Sophie Hambleton2,3, Klaus-Michael Debatin1, Klaus Schwarz4,5, Ulrich Pannicke4,5, Andrew R. Gennery2,3, Mehtap Sirin1 P373 WHICH ALTERNATIVE TO CHOOSE? - A COMPARATIVE STUDY BETWEEN MATCHED UNRELATED AND HAPLOIDENTICAL DONOR TRANSPLANTS IN CHILDREN WITH INBORN ERRORS OF IMMUNITY IN LOW-MIDDLE-INCOME COUNTRIES Anuraag Reddy Nalla 1, Revathi Raj1, Ramya Uppuluri1, Venkateswaran Swaminathan1, Kavitha Ganesan1, Suresh R D1, Anupama Nair1, Vijayshree M1, Indira Jayakumar1, Ravindra Prasad Thokala1, Usha Suriyanarayanan1 P374 MUD-HSCT IN A RARE COMBINED IMMUNODEFICIENCY WITH ARPC1B DEFICIENCY-CASE REPORT Devyani Surange 1, Rajiv Kumar1, Rajan Kapoor1, Sanjeev Khera1 P375 ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION (HCT) IN TWO PEDIATRIC PATIENTS WITH CENTRAL NERVOUS SYSTEM-RESTRICTED FAMILIAL HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS Alison Cusmano1, Ariane Soldatos1, Jessenia Campos1, Katherine Townsend1, Corina Gonzalez1, Allison Grimes2, Sara Makkeyah3, Maha Mohammed3, Hoda Tomoum3, Luigi Notarangelo1, Laila Selim4, Jennifer Kanakry1, Dimana Dimitrova 1 P376 SIL2-RΑ AND ST2 LEVELS BEFORE HEMATOPOIETIC STEM CELL TRANSPLANTATION ARE ASSOCIATED WITH MORTALITY IN HLH PATIENTS Anne B. Verbeek 1, Anja M. Jansen-Hoogendijk1, Erik G J. von Asmuth1, Marco W. Schilham1, Arjan C. Lankester1, Emilie P. Buddingh1 P377 HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN FOR PRIMARY IMMUNE DEFICIENCIES: THE EXPERIENCE OF SINGLE CENTER Utku Aygunes1, Ali Antmen 1, Ilgen Sasmaz1,2, Barbaros Karagun1, Duygu Turksoy1 P378 THE OUTCOMES OF UMBILICAL CORD BLOOD STEM CELL TRANSPLANTATION FOR COMBINED IMMUNODEFICIENCY DISEASE IN CHILDREN WITH CD40LG MUTATION Xiaowen Qian 1, Ping Wang1, Wenjin Jiang1, Xiaowen Zhai1 P379 BACILLE CALMETTE-GUÉRIN (BCG) VACCINE-ASSOCIATED COMPLICATIONS FOLLOWING STEM CELL TRANSPLANTATION IN PATIENTS WITH INBORN ERRORS OF IMMUNITY (IEI) – SINGLE TERTIARY CENTRE EXPERIENCE FROM SOUTH INDIA Stalin Ramprakash 1, C P. Raghuram1, P. Anoop1, Neha Singh1, Jyothi Janardhanan1, Sagar Bhattad1 P380 UNSUCCESSFUL HSCT IN 2 ADA2 DEFICIENCY PATIENTS ASSOCIATED WITH HIGH LEVELS OF ANTI-HLA ANTIBODIES Amit Dotan 1, Marganit Benish1, Hila Rosenfeld Keidar1, David Hagin1, Sabina Edelman1, Ronit Elhasid1 P381 FATAL NEUROLOGICAL COMPLICATION POST HSCT IN A PEDIATRIC PATIENT WITH IMMUNODEFICIENCY DUE TO NFKB2 MUTATION Paula Catalán 1,2, Cristián Sotomayor2, Cecilia Poli3,4, María Angélica Wietstruck1,2 P382 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) TO CHILDREN WITH PRIMARY IMMUNODEFICIENCY –THE HELLENIC EXPERIENCE Eleni Ioannidou 1, Aikaterini Kaissari1, Anna Paissiou1, Christina Oikonomopoulou1, Anna Komitopoulou1, Michalis Kastamoulas1, Georgia Stavroulaki1, Ioannis Grafakos1, Vassiliki Kitra1, Ioulia Peristeri1, Stylianos Grafakos1, Georgios Vessalas1, Maria Theodosaki1, Eftychia Petrakou1, Irene Sfougataki1, Dimitra Kattemi1, Alexandra Papasarantopoulou1, Evangelia Farmaki2, Rediona Cane3, Marianna Tzanoudaki3, Evgenios Goussetis1 P383 POSTTRANSPLANT COMPLICATIONS IN A CASE OF LATE-ONSET FAMILIAL HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS TYPE 2 Delfien Bogaert 1, Siel Daelemans2, Tessa Wassenberg2, Filomeen Haerynck1, Victoria Bordon1, Catharina Dhooge1 P384 HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PRIMARY IMMUNODEFICIENCY DISEASES: SINGLE CENTRE EXPERIENCE Nayera El-Sherif 1, Heba G.A. Ali1, Sara Makkeyah1, Safa Sayed1, Nesrine Radwan2, Dalia ElGhoneimy2, Fatma S.E. Ebeid1, Sally Gouda2 P385 HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR INBORN ERRORS OF IMMUNITY (IEI): GAP BETWEEN NEED AND REALITY IN A LIMITED RESOURCE SETTING: A FIVE-YEAR SINGLE-CENTER EXPERIENCE Stalin Ramprakash 1, Neha Singh1, Jyothi Janardhanan1, Harish Kumar1, Chetan Ginegeri1, C P. Raghuram1, P. Anoop1, Sagar Bhattad1 P386 AN INNOVATIVE PLATFORM APPROACH FOR THE DEVELOPMENT OF EX-VIVO HEMATOPOIETIC STEM AND PROGENITOR CELL-GENE THERAPY FOR THE TREATMENT OF LYSOSOMAL STORAGE DISEASES WITH SKELETAL INVOLVEMENT Stefania Crippa1, Pamela Quaranta1, Margherita Berti1, Luca Basso-Ricci1, Ludovica Santi1, Giada De Ponti1, Raisa Jofra Hernandez1, Claudia Forni1, Ilaria Visigalli1, Paola Albertini1, Rossella Parini1, Serena Scala1, Alessandro Aiuti2, Maria Ester Bernardo 2 P387 HEMATOPOETIC STEM CELL TRANSPLANTATION WITH CURRENT PERSPECTIVE IS SAFE FOR MUCOPOLYSACCHARIDOSIS TYPE VI PATIENTS Vedat Uygun1, Gülsün Karasu2, Koray Yalçin 3,4, Seda Öztürkmen5, Safiye Suna Çelen3, Hayriye Daloğlu6, Suleimen Zhumatayev2, Akif Yesilipek5 P388 HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM MATCHED AND MISMATCHED DONORS WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE AND ANTITHYMOCYTE GLOBULIN IN CHILDREN WITH PRIMARY IMMUNODEFICIENCIES Alexandr Bazaev1, Alexandra Laberko 1,2, Yulia Skvortsova1, Elena Gutovskaya1, Svetlana Kozlovskaya1, Anna Vasilieva1, Larisa Shelikhova1, Galina Novichkova1, Alexey Maschan1, Michael Maschan1, Dmitry Balashov1 P389 CONDITIONING FOR SECOND HSCTS FROM THE SAME DONOR AFTER MYELOID AUTOLOGOUS RECONSTITUTION IN PATIENTS WITH IEI AND THALASSEMIA: IMMUNOSUPPRESSION IS NOT ALWAYS REQUIRED Sarah Bauer1, Beatrice Zwiebler1, Michael H. Albert2, Jörn-Sven Kühl3, Mehtap Sirin1, Kerstin Felgentreff1, Ingrid Furlan1, Klaus-Michael Debatin1, Eva-Maria Jacobsen1, Ansgar Schulz1, Manfred Hoenig 1 P390 SINGLE CENTER OBSERVATION ON CLINICAL PRESENTATION, TRANSPLANTATION AND OUTCOME OF SEVEN PATIENTS WITH ZAP-70 DEFICIENCY Felix Immanuel Maier 1, Philipp Friederichsen1,2, Ansgar Schulz1, Eva-Maria Jacobsen1, Klaus-Michael Debatin1, Katharina Kleinschmidt3, Selim Corbacioglu3, Jürgen Föll3, Klaus Schwarz4,5, Ulrich Pannicke4, Manfred Hönig1 P391 POLYMORPHISM OR RISK ALLELE? PRF1 A91V IN TRANS WITH A “SEVERE” PRF1 MUTATION Oliver Wegehaupt 1,2, Oleg Borisov3, Florian Oyen4, Jasmin Mann1, Despina Moshous5, Geneviève de Saint Basile5, Kimberly Gilmour6, Wenying Zhang7, Rebecca Marsh7, Eberhard Gunsilius8, Laine Hosking9, Sharon Choo9, Katharina Wustrau10, Sujal Gosh11, Kai Lehmberg4, Anna Köttgen3, Stephan Ehl1 P392 IMMUNE-MEDIATED CYTOPENIA AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN CHILDREN WITH INHERITATED METABOLIC DISORDERS Hirotoshi Sakaguchi 1, Tetsumin So1, Yoshihiro Gochio1, Akihiro Iguchi1, Takao Deguchi1, Daisuke Tomizawa1, Motomichi Kosuga1, Kimikazu Matsumoto1 P393 PROPHYLACTIC ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR CSF1R-RELATED LEUKOENCEPHALOPATHY- A CASE REPORT Malu Lian Hestdalen 1, Morten Andreas Horn2, Camilla Dao Vo1, Ingerid Weum Abrahamsen1, Tor Henrik Anderson Tvedt1, Tobias Gedde-Dahl1, Anders Eivind Myhre1 P394 CLINICAL PROFILE AND TRANSPLANT OUTCOMES OF 41 PATIENTS WITH INBORN ERRORS OF IMMUNITY: A SINGLE CENTER EXPERIENCE FROM SOUTH INDIA Stalin Ramprakash 1, Neha Singh1, C P. Raghuram1, P. Anoop1, Jyothi Janardhanan1, Harish Kumar1, Chetan Ginigeri1, Fulvio Porta2, Sagar Bhattad1 P395 HEMATOPOIETIC CELL TRANSPLANTATION AND SPLIT LIVER TRANSPLANTATION IN A PATIENT WITH ERYTHROPOIETIC PROTOPORPHYRIA - A CASE REPORT Lasse Jost 1, Ulrich Stölzel2, Daniel Seehofer3, Katharina Egger-Heidrich4, Kristina Hölig4, Thomas Stauch5, Desiree Kunadt4, Detlef Schuppan6, Johannes Schetelig4, Nils Wohmann2, Martin Bornhäuser4, Friedrich Stölzel1 P396 SUCCESSFUL THIRD ALLOGENEIC STEM CELL TRANSPLANTATION FOLLOWING TWO GRAFT REJECTIONS IN A PATIENT WITH CHRONIC GRANULOMATOUS DISEASE AND HEPATIC/PARAPHRENIC ABSCESSES Daniela Sperl 1, Martin Benesch1, Ursula Posch1, Konrad Rosskopf1, Volker Strenger1, Sebastian Tschauner1, Markus G. Seidel1, Wolfgang Schwinger1 P397 CHRONIC GRANULOMATOUS DISEASE DO NOT NEED MOST OF THE TIMES NGS FOR DIAGNOSIS Marianna Maffeis 1, Marta Comini2, Alessandra Beghin2, Federica Bolda2, Elena Soncini1, Giulia Baresi1, Stefano Rossi1, Fulvio Porta1, Arnalda Lanfranchi2 P398 BLINATUMOMAB IS AN EFFECTIVE AND LOW TOXICITY BRIDGE TREATMENT TO HEMATOPOIETIC STEM CELL TRANSPLANTATION IN ACUTE LYMPHOBLASTIC LEUKEMIA ASSOCIATED WITH SHWACHMAN-DIAMOND SYNDROME Fabiana Cacace1, Giovanna Giagnuolo1, Pio Stellato1, Emanuela Rossitti2, Valeria Caprioli1, Flavia Antonucci2, Maria Rosaria D’amico1, Giuseppina De Simone1, Maria Simona Sabbatino1, Rosanna Parasole1, Giuseppe Menna1, Francesco Paolo Tambaro 1 P399 BREAKTHROUGH INVASIVE FUNGAL DISEASE IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHINA: A MULTICENTER EPIDEMIOLOGICAL STUDY (CAESAR 2.0) Yu-Qian Sun 1, Chuan Li1, Dan-Ping Zhu1, Jia Chen2, Xiao-Yu Zhu3, Nai-Nong Li4, Wei-Jie Cao5, Zhong-Ming Zhang6, Hai-Long Yuan7, Xiao-Xia Hu8, Xiao-Sheng Fang9, Hong-Tao Wang10, Yue Yin11, Ye-Hui Tan12, Xiao-Jun Huang1 P400 CHANGES IN THE EPIDEMIOLOGY OF INVASIVE FUNGAL DISEASES IN HSCT OVER ALMOST A QUARTER OF A CENTURY Marina Popova 1, Yuliya Rogacheva1, Valentin Zagranichnov2, Olga Pinegina3, Vladislav Markelov1, Aleksander Siniaev1, Irina Baranova1, Yulia Rodneva1, Alisa Volkova1, Ilya Nikolaev1, Alena Zaytseva1, Alexander Shvetsov1, Anna Spiridonova2, Oleg Golochtapov1, Yulia Vlasova1, Anna Smirnova1, Natalia Mikhaylova1, Maria Vladovskaya1, Sergey Bondarenko1, Alexander Kulagin1 P401 OUTCOMES FROM BRONCHOSCOPY AND BRONCHOALVEOLAR LAVAGE IN HEMATOPOIETIC CELL TRANSPLANT RECIPIENTS Colin Hayes 1, Benison Lau1, Kengo Inagaki1, Mike Triebwasser1, John Magenau1, Mary Riwes1, Mark Vander Lugt1, Ghada Abusin1, Sung Choi1, Sarah Anand1, Monalisa Ghosh1, John Maciejewski1, Attaphol Pawarode1, Gregory Yanik1 P402 LETERMOVIR PROPHYLAXIS FOR CYTOMEGALOVIRUS INFECTION IS THE POTENTIAL RISK OF EBV-POSITIVE POSTTRANSPLANT LYMPHOPROLIFERATIVE DISORDERS AFTER HAPLOIDENTICAL STEM-CELL TRANSPLANTATION Xuying Pei 1, Xiaojun Huang1 P403 DIAGNOSIS OF MUCORMYCOSIS IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION USING METAGENOMIC NEXT-GENERATION SEQUENCING: A SINGLE-CENTER CLINICAL STUDY Fang Xu 1, Jianping Zhang1, Min Xiong1, Yanli Zhao1, Deyan Liu1, Xingyu Cao1, Zhijie Wei1, Jiarui Zhou1, Ruijuan Sun1, Yue Lu1 P404 MARIBAVIR USE FOR REFRACTORY CMV INFECTION/DISEASE: THE RESULTS FROM THE FRENCH COMPASSIONATE PROGRAMME Catherine Cordonnier 1, Nassim Kamar2, Philippe Gatault3, Faouzi Saliba4, Fanny Vuotto5, Lionel Couzi6, Cinira Lefevre7, Michèle Maric7, Sophie Alain8 P405 COMPREHENSIVE SCREENING AND SENSITIVITY-GUIDED TREATMENT SIGNIFICANTLY REDUCES THIRTY-DAY MORTALITY OF METALLO-Β-LACTAMASES PRODUCING ENTEROBACTERALES (MBL-E) INFECTION FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION: INSIGHTS FROM A PROSPECTIVE STUDY Yi-Han Yang1, Meng Lv1, Qi Wang1, Jing Liu1, Xiao-Lu Zhu1, Xiao-Dong Mo1, Yu-Qian Sun 1, Yu Wang1, Lan-Ping Xu1, Xiao-Hui Zhang1, Xiao-Jun Huang1,2 P406 PENTAGLOBIN® AS EARLY ADJUVANT TREATMENT FOR FEBRILE NEUTROPENIA IN HEMATOLOGICAL PATIENTS COLONIZED BY CARBAPENEM-RESISTANT ENTEROBACTERIACEAE OR PSEUDOMONAS AERUGINOSA: INTERIM ANALYSIS OF GITMO STUDY “PENTALLO” Daniela Clerici 1, Alessandra Picardi2, Patrizia Chiusolo3, Nicola Di Renzo4, Anna Paola Iori5, Mario Delia6, Ilaria Cutini7, Francesca Bonifazi8, Michele Malagola9, Raffaella Cerretti10, Maria Goldaniga11, Francesca Patriarca12, Daniele Vallisa13, Attilio Olivieri14, Roberto Sorasio15, Stefania Bregante16, Angelo Michele Carella17, Alessandro Busca18, Consuelo Corti1, Eliana Degrandi19, Fabio Ciceri1, Massimo Martino20, Corrado Girmenia5 P407 MUCOSITIS-ASSOCIATED BLOODSTREAM INFECTIONS IN HAEMATOLOGY PATIENTS WITH FEVER DURING NEUTROPENIA Nick de Jonge 1,2,3, Jeroen Janssen1,2,3, Paula Ypma4, Alexandra Herbers5, Arne de Kreuk6, Wies Vasmel6, Jody van den Ouweland7, Aart Beeker8, Otto Visser9, Sonja Zweegman1,3, Nicole Blijlevens2, Michiel van Agtmael1, Jonne Sikkens1 P408 DESCRIPTIVE ANALYSES OF PULMONARY FUNCTION CHANGES BEFORE AND AFTER COVID-19 IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS. AN INFECTIOUS DISEASE WORKING PARTY STUDY Jose-Luis Piñana 1, Per Ljungman2, Stephan Mielke2, Gloria Tridello3, Helene Labussiere4, Carlos Solano1, Anna Torrent5, Regis Peffault De Latour6, Nicolaus Kroeger7, Maria Jesus Pascual Gascon8, Adriana Balduzzi9, Lutz Peter-Muller10, Jürgen Kuball11, Amjad Hayat12, Urpu Salmenniemi13, Fabio Benedetti14, Dina Averbuch15, Rafael De La Camara16 P409 EPSTEIN-BARR VIRUS-RELATED POSTTRANSPLANT LYMPHOPROLIFERATIVE DISORDERS IN THE LETERMOVIR ERA Jingtao Huang1, Luxiang Wang1, Zengkai Pan1, Zilu Zhang1, Jiayu Huang1, Chuanhe Jiang1, Xiaoxia Hu 1 P410 PATTERN OF SENSITIVITY OF STOOL SURVEILLANCE OVER 13 YEARS AND IMPACT OF STOOL SURVEILLANCE GUIDED SELECTION OF ANTIBIOTIC IN ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT PATIENTS Keshav Garg1, Akanksha Chichra1, Davinder Paul1, Anant Gokarn1, Sachin Punatar1, Sumeet Mirgh1, Nishant Jindal1, Lingaraj Nayak1, Sujata Lall1, Vivek Bhat1, Sadhana Kannan1, Bhausaheb Bagal1, Navin Khattry1, Laxma Reddy 2 P411 CYTOMEGALOVIRUS (CMV) LOAD PREDICTS CMV DISEASE AND MORTALITY INDEPENDENT OF POSTTRANSPLANT IMMUNOSUPPRESSION AFTER HEMATOPOIETIC CELL TRANSPLANTATION (HCT) Alicja Sadowska-Klasa 1,2, Danniel Zamora1, Hu Xie1, Elizabeth R. Duke1, Margaret L. Green3, Masumi Ueda Oshima1,3, Alpana Waghmare1,3,4, Joshua A. Hill1,3, Brenda M. Sandmaier1,3, Keith R. Jerome1,3, Wendy M. Leisenring1,3, Michael Boeckh1,3 P412 ADENOVIRUS INFECTION IN ADULT ALLOGENEIC TRANSPLANT RECIPIENTS WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE Maria Chiara Quattrocchi 1, Giorgio Orofino1, Edoardo Campodonico1, Alessandro Bruno1, Lorenzo Lazzari1, Daniela Clerici1, Francesca Farina1, Simona Piemontese1, Elisa Diral1, Oltolini Chiara1, Raffaele Dell’Acqua1, Sara Racca1, Raffaella Milani1, Elisabetta Xue1, Sara Mastaglio1, Consuelo Corti1, Jacopo Peccatori1, Maria Teresa Lupo-Stanghellini1, Antonella Castagna1, Fabio Ciceri1, Raffaella Greco1 P413 NON-TUBERCULOUS MYCOBACTERIAL INFECTIONS AMONG HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS: A MULTICENTER EUROPEAN CASE-CONTROL STUDY BY THE INFECTIOUS DISEASES WORKING PARTY OF EBMT Maria Stamouli 1, John Snowden2, Alienor Xhaard3, Awatif AlAnazi4, Antonio Perez Martinez5, Moshe Yeshurun6, Nicolaus Kröger7, Jürgen Kuball8, Fanourios Kontos1, Jakob Passweg9, Stephan Mielke10, Mohsen Al Zahrani11, Luis Miguel Juarez-Salcedo12, Joanna Drozd-Sokolowska13, Fabio Benedetti14, Lucrecia Yañez Sansegundo15, Alessandra Biffi16, Maria Teresa Lupo Stanghellini17, Pavel Jindra18, Alexander Kulagin19, Nuno Miranda20, Urpu Salmenniemi21, Petr Sedlacek22, Gloria Tridello23, Inge Verheggen23, Dina Averbuch24, Rafael de la Camara12 P414 BACTEREMIA IN THE FIRST + 100 DAYS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: MICROORGANISMS, RESISTANCE AND PROGNOSTIC IMPACT Merce Aren1, Rosa Pacheco2, Mireia Morgades1, Nadia Güell1, Gerard García-Cirera1, Cristina Blanco-Montes1, Vitor Botafogo1, Maria Izquierdo1, Anna Torrent1, Christelle Ferrà3, Maria Josefa Jiménez1, Josep Maria Ribera1, Juan Manuel Sancho1, Maria Dolores Quesada2, Maria Huguet 1 P415 ISAVUCONAZOLE THERAPEUTIC DRUG MONITORING-DRIVEN TREATMENT IN A COHORT OF PAEDIATRIC PATIENTS UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANTATION: A SINGLE-CENTRE EXPERIENCE Carmen Dolores De Luca 1,2, Chiara Rossi1, Chiara Rosignoli1, Federica Galaverna1, Francesca Del Bufalo1, Marco Becilli1, Michele Massa1,2, Raffaele Simeoli1, Emilia Boccieri1, Barbarella Lucarelli1, Antonio Torelli1, Daria Pagliara1, Bianca Maria Goffredo1, Mattia Algeri1, Franco Locatelli1,3, Pietro Merli1 P416 THINK TO CHECK PREVIOUS SEROLOGIES WHEN CMV OR TOXOPLASMA SEROLOGIES ARE NEGATIVE AT TRANSPLANT Christine Robin 1, Ludovic Cabanne1, Florence Beckerich1, Anne Le Bouter1, Francoise Foulet1, Catherine Cordonnier1 P417 IMMUNE POPULATIONS PREDICT CONTROL OF CMV REACTIVATION AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Paula Díaz-Fernández 1,2,3, Valle Gómez García de Soria1,2,3, Javier Sevilla-Montero1,2, Ana Marcos-Jiménez1,2, José María Serra López-Matencio1,2, Laura Cardeñoso1, Ángela Figuera Álvarez1,2,3, Rafael de la Cámara1,2, Cecilia Muñoz-Calleja1,2,3 P418 NEW ANTIMICROBIAL STEWARDSHIP PROGRAM TOOL TO REDUCE THE EMPIRICAL ANTIBACTERIAL THERAPY IN HAPLO-HSCT Yuliya Rogacheva1, Marina Popova 1, Alexandr Siniaev1, Yuliya Vlasova1, Ivan Moiseev1, Sergey Bondarenko1, Alexander Kulagin1 P419 LETERMOVIR IS SAFE AND EFFECTIVE FOR TREATMENT AND PREVENTION OF CMV REACTIVATION IN PAEDIATRIC ALLOGENEIC STEM CELL TRANSPLANT Nathanael Lucas 1, Abbey Forster1, Malcolm Guiver1, Ramya Nataraj1, Madeleine Powys1, Noor Barotchi1, Omima Mustafa1, Srividhya Senthil1, Robert Wynn1 P420 DUAL METAGENOMICS NEXT-GENERATION SEQUENCING FOR FIRST-LINE DIAGNOSIS OF BLOODSTREAM INFECTION IN HEMATOLOGIC PATIENTS WITH FEBRILE NEUTROPENIA: A MULTICENTER PROSPECTIVE STUDY Yuqian Sun 1, Jingrui Zhou1, Yue Yin2, Jianping Zhang3, Meixiang Zhang4, Yun He1, Wei Gai5, Xiaohui Zhang1, Yu Wang1, Lanping Xu1, Kaiyan Liu1, Xiaojun Huang1 P421 CYTOMEGALOVIRUS-SPECIFIC T CELL IMMUNITY RECONSTITUTION AFTER LETERMOVIR WITHDRAWAL IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION RECIPIENTS Luxiang Wang1, Jingtao Huang1, Jiayu Huang1, Zilu Zhang1, Zengkai Pan1, Chuanhe Jiang1, Sujiang Zhang1, Xiaoxia Hu 1 P422 CMV REACTIVATION AND ITS IMPACT ON SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN LETERMOVIR ERA – A SINGLE CENTRE STUDY Lana Desnica 1, Nadira Durakovic1, Zinaida Peric2, Zrinka Bosnjak1, Ranka Serventi Seiwerth1, Dubravka Sertic1, Ante Vulic1, Mirta Mikulic1, Violeta Rezo Vranjes1, Pavle Roncevic1, Drazen Pulanic1, Nurka Rustan1, Zorana Grubic1, Radovan Vrhovac1 P423 IMPACT OF QUANTIFERON-CMV (QNF-CMV) ON LATE ONSET CLINICALLY SIGNIFICANT CMV INFECTION (CSCMVI) AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) Chiara Maria Dellacasa 1, Roberto Passera1, Carolina Secreto1, Luisa Giaccone1, Irene Dogliotti1, Jessica Gill1, Davide Stella1, Aurora Martin1, Federica Ferrando1, Sofia Zompi1, Alessandro Busca1 P424 THERAPEUTIC DRUG MONITORING (TDM) OF PIPERACILLIN IN PATIENTS WITH NEUTROPENIC FEVER AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Sabrina Kraus1, Prisca Rauen1, Josip Zovko1, Hermann Einsele1, Güzin Surat 1, Torsten Steinbrunn1 P425 ESTABLISHING EBV LOAD THRESHOLDS FOR PREDICTING PERSISTING EBV-DNAEMIA AND ASSOCIATED DISEASE IN HAEMATOPOIETIC CELL TRANSPLANT RECIPIENTS Svenia Schmid 1, Andrea Erba1, Rainer Gosert1, Hans H. Hirsch2, Jakob Passweg1, Joerg Halter1, Nina Khanna1, Karoline Leuzinger1 P426 CLINICAL OUTCOMES OF STRONGYLOIDIASIS IN HEMATOPOIETIC CELL TRANSPLANT (HCT) RECIPIENTS Avneet Kaur 1, Deepa Nanayakkara1, Ryotaro Nakamura1, Randy Taplitz1, Jana Dickter1, Stephen Forman1, Monzr Malki1, Sanjeet Dadwal1 P427 MONITORING OF CMV-SPECIFIC CELL-MEDIATED IMMUNITY - PROGNOSTIC FACTORS AND CLINICAL VALUE Jonas Wißkirchen 1, Diana Wolff1, Isabelle Ries1, Oliver Kriege1, Pascal Wölfinger1, Beate Hauptrock1, Matthias Theobald1, Eva Wagner-Drouet1 P428 USE OF LETERMOVIR IN PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS UNDER AGE OF 12: A RETROSPECTIVE MULTI-CENTRE STUDY ON BEHALF OF THE IEWP AND PDWP Katharina Kleinschmidt 1, Giovanna Lucchini2, Jacques-Emmanuel Galimard3, Adriana Balduzzi4, Krzysztof Czyzewski5, Jowita Frączkiewicz6, Francesco Paolo Tambaro7, Jaroslava Adamcakova8, Andrea Urtasun9, Matthias Woelfl10, Maria Ester Bernardo11, Alessandra Biffi12, Gergely Kriván13, Iván Lopez Torija14, Petr Sedlacek15, Wolfgang Holter16, Tamara Diesch17, Michael H. Albert18, Agnieszka Sobkowiak-Sobierajska19, Marta Gonzalez Vicent20, Elif Ince21, Sarah May Johnson2, Roland Meisel22, Arnaud Dalissier3, Selim Corbacioglu1, Bénédicte Neven23, Krzysztof Kalwak6 P429 CONVENTIONAL VERSUS NGS-BASED PATHOGEN DIAGNOSTICS IN FEBRILE ALLOGENEIC STEM CELL TRANSPLANT PATIENTS: A PROSPECTIVE SINGLE-CENTRE STUDY Sophie Weil 1, Madlen Amersbach1, Jochen J. Frietsch1, Sabrina Kraus1, Christoph Schoen2, Johannes Forster2, Philipp Reuter-Weissenberger2, Philip Stevens3, Oliver Kurzai2, Hermann Einsele1, Daniel Teschner1,4 P430 MULTINATIONAL STUDY ASSESSING TREATMENT PATTERNS, OUTCOMES, AND HEALTHCARE RESOURCE UTILIZATION IN HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS WITH CYTOMEGALOVIRUS INFECTION AND INTOLERANCE TO ANTI-CYTOMEGALOVIRUS THERAPIES Genovefa Papanicolaou1,2, Robin Avery3, María Laura Fox 4,5, Karl S. Peggs6, Luís Veloso7, Tien Bo8, Ishan Hirji8, Kimberly Davis8 P431 EFFICACY OF LETERMOVIR IN PREVENTING CYTOMEGALOVIRUS REACTIVATION AFTER CORD BLOOD TRANSPLANTATION Naoki Okada 1, Hiroyuki Muranushi1, Kazuya Okada1, Takayuki Sato1, Takeshi Maeda1, Tatsuhito Onishi1, Yasunori Ueda1 P432 SARS-COV-2 CYCLE THRESHOLD LEVELS AND COVID-19 OUTCOME IN ALLOGENEIC STEM CELL TRANSPLANTATION. AN INFECTIOUS DISEASE WORKING PARTY STUDY Jose Luis Piñana Sanchez 1, Per Ljungman2, Alexander Kulagin3, Maria Teresa Lupo Stanghellini4, Carlos Solano5, Gloria Tridello6, Jiri Mayer7, Maria Suarez-Lledó8, Hélène Labussière-Wallet9, Robert Zeiser10, Jennifer Clay11, Anna Bergendahl Sandstedt12, Inmaculada Heras13, Krzysztof Kalwak14, Jose Luis Lopez15, Judith Schaffrath16, Adriana Balduzzi17, Fabio Benedetti18, Amjad Hayat19, David Gallardo20, Harry Koene21, Jose Rifón22, Paul Gerard Schlegel23, Nina Knelange24, Dina Averbuch25, Rafael De la Camara26 P433 SINGLE-DAY TRIMETHOPRIM-SULFAMETHOXAZOLE PROPHYLAXIS FOR PNEUMOCYSTIS PNEUMONIAE AND TOXOPLASMOSIS INFECTION IN HEMATOPOIETIC STEM CELL TRANSPLANTATION PEDIATRIC PATIENTS Maria Speranza Massei 1, Ilaria Capolsini1, Elena Mastrodicasa1, Grazia Gurdo1, Carla Cerri1, Francesco Arcioni1, Maurizio Caniglia1, Katia Perruccio1 P434 AN INTERIM ANALYSIS OF FOVOCIP: A MULTICENTER RANDOMIZED TRIAL OF FOSFOMYCIN VERSUS CIPROFLOXACIN FOR FEBRILE NEUTROPENIA PREVENTION IN HEMATOLOGIC PATIENTS Ahinoa Fernández Moreno1, Ana Julia GonzalezHuerta2, Paula López de Ugarriza1, Cristina Muñoz3, Rebeca Rodríguez Veiga4, Laura Solán Blanco5, Maria Luisa Calabuig6, Karem Humala7, Juan Manuel Bergua Burgués3, Marta Polo Zarzuela8, Maria Luz Amigo9, María Izquierdo de Miguel3, Eva González Barberá4, Marina Medel Plaza5, Guillermo Ruiz Carrascoso7, Laura López Gonzalez8, Irene Weber9, Pau Montesinos Fernández4, Ana Maria Fernández Verdugo2, Javier Fernández Dominguez2, Teresa Bernal 1 P435 IMPACT OF LETERMOVIR PRIMARY AND SECONDARY PROPHYLAXIS IN A PEDIATRIC COHORT Francesca Vendemini1, Francesca Romani1,2, Sonia Bonanomi1, Giorgio Ottaviano1, Paola de Lorenzo3, Maria Grazia Valsecchi2, Sergio Maria Ivano Malandrin4, Marta Verna 1, Giulia Prunotto1, Pietro Casartelli1, Adriana Cristina Balduzzi1,2 P436 EPSTEIN-BARR VIRUS REACTIVATION AND POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER (PTLD) AFTER IMPLEMENTATION OF ANTITHYMOCYTE GLOBULIN AS GVHD PROPHYLAXIS. A SINGLE CENTER EXPERIENCE Tor Henrik Anderson Tvedt1, Mats Remberger1,2,3, Ingerid Weum Abrahamsen1, Camilla Dao Vo 1, Anders Eivind Leren Myhre1, Tobias Gedde-Dahl1 P437 IMPACT OF INVASIVE FUNGAL DISEASE UPDATED DEFINITIONS ON ALLOGENEIC HEMATOPOIETIC TRANSPLANTATION OUTCOME IN ADULT PATIENTS - POST-HOC ANALYSIS OF NATION-WIDE CROSS-SECTIONAL 2012-2014 Patrycja Mensah-Glanowska 1, Agnieszka Piekarska2, Monika Adamska3, Joanna Drozd-Sokolowska4, Anna Waszczuk-Gajda4, Agnieszka Tomaszewska5,4, Agnieszka Wierzbowska6, Marek Hus7, Joanna Manko5, Sylwia Bartzel-Palinska8, Lidia Gil3 P438 SURVEILLANCE BLOOD CULTURES FROM TUNNELLED CENTRAL VENOUS CATHETERS ON ADMISSION ARE OFTEN POSITIVE, GUIDE MANAGEMENT AND HELP INFORM REPORTING OF HOSPITAL ACQUIRED INFECTIONS Debbie Barrow1, Krzysztof Ciesielski1, Seamus McDermott1, Grant McQuaker1, David Irvine1, Annie Latif1, Ailsa Holroyd1, Christine Peters1, Dimitris Galopoulos1, Andrew Clark 1 P439 ROLE OF HUMAN HERPESVIRUS 6 REACTIVATION IN T-DEPLETE TREG/TCON ALLOGENEIC STEM CELL TRANSPLANTATION Valerio Viglione 1, Loredana Ruggeri2, Antonella Mancusi1, Sara Tricarico2, Alessandra Cipiciani1, Rebecca Sembenico1, Matteo Caridi1, Zorutti Francesco1, Anna Castaldo1, Tiziana Zei2, Roberta Iacucci Ostini2, Simonetta Saldi1, Roberto Castronari2, Silvia Bozza1, Cynthia Aristei1, Antonella Mencacci1, Maria Paola Martelli1, Alessandra Carotti2, Antonio Pierini1 P440 INTERLEUKIN-6 IS SIGNIFICANTLY INCREASED IN SEVERE PNEUMONIA AFTER ALLO-HSCT AND MIGHT INDUCE LUNG INJURY VIA IL-6/SIL-6R/JAK1/STAT3 PATHWAY Jingrui Zhou1, Leqing Cao1, Rui Ma1, Yun He1, Danping Zhu1, Na Li1, Xiaosu Zhao1, Xiaojun Huang1, Yuqian Sun 1 P441 ECHINOCANDIN PROPHYLAXIS IS ASSOCIATED WITH BREAKTHROUGH TRICHOSPORON AND CANDIDA INFECTIONS AND A LIMITED ROLE OF GALACTOMANNAN MONITORING AMONG ALLOGENEIC STEM CELL TRANSPLANT RECIPIENTS André Dias Américo 1, João Antônio Gonçalves Garreta Prats1, Hegta Tainá Rodrigues Figueiroa1, Juliana Matos Pessoa1, Eurides Leite da Rosa1, Fauze Lutfe Ayoub1, Moyses Antônio Porto Soares1, Isabella Silva Pimentel Pittol1, Fabio Rodrigues Kerbauy1,2, Phillip Scheinberg1, José Ulysses Amigo Filho1 P442 REAL WORLD EXPERIENCE OF EPSTEIN-BARR VIRUS(EBV) SCREENING FOR PERSISTENT VIRAEMIA POST HAEMATOPOIETIC STEM CELL TRANSPLANT (HSCT) Abigail Downing 1, David Davies1, Keith Wilson1,2, Emma Kempshall1, Sophie Bertorelli1, Serena Linley-Adams1, Damith Dhanasekara1, Rey Consolacion1, Susannah Froude3, Samantha Ray3, Wendy Ingram1 P443 EPIDEMIOLOGY OF VIRAEMIA IN CHILDREN UNDERGOING CORD BLOOD STEM CELL TRANSPLANT (CB-SCT) WITH NON-MALIGNANT DISEASES: A RETROSPECTIVE COHORT STUDY Patel Zeeshan Jameel 1, Denise Bonney1, Omima Mustafa1, Malcolm Guiver1, Madeleine Powys1, Ramya Nataraj1, Srividhya Senthil1, Robert Wynn1 P444 PREVALENCE OF VZV REACTIVATION AND EFFECTIVENESS OF VACCINATION WITH RECOMBINANT ADJUVANTED ZOSTER VACCINE IN ALLOGENEIC HEMATOPOIETIC STEM CELL RECIPIENTS: A SINGLE-CENTER ANALYSIS Ewa Karakulska-Prystupiuk 1, Magdalena Feliksbrot-Bratosiewicz1, Maria Król1, Agnieszka Tomaszewska1, Grzegorz Władysław Basak1 P445 BK VIRUS-ASSOCIATED HEMORRHAGIC CYSTITIS IN POSTTRANSPLANT CYCLOPHOSPHAMIDE-BASED ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION (HCT) FOR IMMUNE DEFICIENCY OR DYSREGULATION Dimana Dimitrova 1, Christi McKeown1, Scott Napier1, Anita Stokes1, Alison Cusmano1, Ruby Sabina1, Jennifer Sponaugle1, Elisabetta Xue1, Jeffrey Cohen1, Alexandra Freeman1, Luigi Notarangelo1, Gulbu Uzel1, Vladimir Valera Romero1, Christopher Kanakry1, Jennifer Kanakry1 P446 INFECTION PREVENTION MEASURES TAKEN FOR ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT PATIENTS DURING THE COVID-19 PANDEMIC AND THEIR IMPACT ON CLINICAL OUTCOMES Sarah Noetzlin1, Michael Bader2, Andriyana Bankova3, Dominik Schneidawind3, Jakob Passweg2, Anne-Claire Mamez1, Federica Giannotti1, Sarah Morin 1, Federico Simonetta1, Stavroula Masouridi-Levrat1, Dionysios Neofytos1, Yves Chalandon1 P447 EFFECT OF EARLY ADMINISTRATION OF ANTI-MRSA DRUGS ON THE COURSE OF FEBRILE NEUTROPENIA IN ALLO-SCT Anna Akaogi 1, Junya Kanda1, Fumiya Wada1, Yasuyuki Arai1, Chisaki Mizumoto1, Toshio Kitawaki1, Kouhei Yamashita1, Akifumi Takaori-Kondo1 P448 VISCERAL TOXOCARIAS IN THE EARLY POST-TRANSPLANT COURSE Iacopo Bellani 1, Francesca Vendemini2, Sonia Bonanomi2, Sergio Foresti3, Guglielmo Marco Migliorino3, Gaia Kullmann3, Alex Moretti2, Sergio Maria Ivano Malandrin4, Adriana Balduzzi2,1 P449 DE-ESCALATION ANTIBIOTIC STRATEGY IN MULTIDRUG-RESISTANCE BACTERIAL COLONIZED PATIENTS AFTER ALLOGENEIC STEM CELL TRANSPLANTATION. A 3-YEAR RETROSPECTIVE STUDY Roberto Bono1, Giuseppe Sapienza 1, Stefania Tringali1, Cristina Rotolo1, Alessandra Santoro2, Laura Di Noto3, Orazia Di Quattro4, Caterina Patti5, Luca Castagna1 P450 MARIBAVIR FOR TREATMENT OF REFRACTORY/RECURRENT CYTOMEGALOVIRUS INFECTION IN ALLOGENEIC HSCT RECIPIENTS Aleksandr Siniaev1, Ivan Moiseev1, Marina Popova 1, Yulia Rogacheva1, Yulia Vlasova1, Sergey Bondarenko1, Alexander Kulagin1 P451 REAL LIFE MANAGEMENT OF ANTIBIOTIC THERAPY IN HSCT RECIPIENTS – FOCUS ON DE-ESCALATION IN PRE-ENGRAFTMENT NEUTROPENIA, THE STUDY FROM THE EBMT INFECTIOUS DISEASES WORKING PARTY (IDWP) Malgorzata Mikulska 1, Lotus Wendel2, Gloria Tridello2, Alexander Kulagin3, Anna Czyz4, Nabil Yafour5, Fabio Ciceri6, Burak Deveci7, Simona Sica8, Kristina Carlson9, Matthias Eder10, Agnieszka Tomaszewska11, Tunc Fisgin12, Michel Schaap13, Michal Karas14, Anastasia Pouli15, Elisabetta Calore16, Marta Gonzalez Vicent17, Mariagrazia Michieli18, Maria Jesús Pascual Cascon19, Alessandra Picardi20, Gergely Krivan21, Anna Torrent Catarineu22, Tessa Kerre23, Pedro Chorão24, Fabio Benedetti25, Alessandra Carotti26, Melissa Gabriel27, Rik Schots28, Amjad Hayat29, Antonio Perez Martinez30, Domenico Pastore31, Mauro Turrini32, Diana Averbuch33, Jan Styczynski34, Rafael de la Camara35 P452 INFECTIOUS COMPLICATIONS IN 549 BMTS DONE WITHOUT CENTRAL HEPA FILTRATION OR POSITIVE PRESSURE? Rajpreet Soni1,2, Lawrence Faulkner 3,4, Priya Marwah1,2, Harshita Agarwal1, Deepa Trivedi5, Vaibhav Shah5, Mohan Reddy6, Neema Bhatt6, Stalin Ramprakash4, Rajat Kumar Agarwal4,7, Rachna Narain1,2, Ganesh Narain Saxena1,2 P453 COVID-19 IN PATIENTS WITH HAEMATOLOGICAL DISEASES: THE ASSOCIATION OF RNAEMIA WITH CLINICAL OUTCOMES IN VACCINATED PATIENTS Francisco Manuel Martin Dominguez 1, Sonsoles Salto-Alejandre2, Carmen Infante-Domínguez2, Mónica Carretero-Ledesma2, Natalia Maldonado-Lizarazo3, Pedro Camacho-Martínez2, Inmaculada Tallón-Ruiz4, Zaira Palacios-Baena3, Patricia Pérez-Palacios3, Rocío Álvarez-Marín2, José Antonio Lepe-Jiménez2, José Miguel Cisneros2, María Elisa Cordero2, Jerónimo Pachón2, Javier Sánchez-Céspedes2, José Antonio Pérez-Simón1, Manuela Aguilar-Guisado2 P454 COST-EFFECTIVENESS OF MARIBAVIR FOR POST-TRANSPLANT CYTOMEGALOVIRUS INFECTION THAT IS REFRACTORY TO ALTERNATIVE TREATMENTS FROM THREE HEALTHCARE SYSTEM PERSPECTIVES: ITALY, UK AND US Stacey L. Amorosi1, Emtiyaz Chowdhury2, Riccardo Ressa2, Peter Cain3, Simone Corinti4, Bob G. Schultz5, Tien Bo 1 P455 INVASIVE FUNGAL INFECTIONS IN PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A 4-YEAR SINGLE INSTITUTION ANALYSIS WITH FOCUS ON PROPHYLAXIS Federica Gironi1, Benedetta Elena Di Majo2,1, Ilaria Castelli1, Sonia Bonanomi1, Francesca Vendemini1, Giorgio Ottaviano1, Annalisa Cavallero1, Sergio Maria Malandrin1, Irene Maria Sciabica1, Guglielmo Marco Migliorino1, Sergio Foresti1, Marta Verna 1, Adriana Balduzzi2,1 P456 ENHANCING SARS-COV-2 IMMUNITY EARLY AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION: A THREE-DOSE RBD–TT-CONJUGATED VACCINE APPROACH Maryam Barkhordar 1, Mohammad Vaezi1, Leyla Sharifi Aliabadi1, Mohammad Ahmadvand1, Ardeshir Ghavamzadeh2 P457 USE OF LETERMOVIR IN PEDIATRIC POPULATION POST BONE MARROW TRANSPLANT Cristina Rivera-Pérez1, Nuria Mas Malagarriga1, Silvia Simó Nebot1, Maria Trabazo del Castillo1, Gloria Miguel Llordes1, Laura Jimenez Prat1, Montserrat Rovira Tarrats1,2, Julia Marsal Ricoma 1, Cristina Rivera P458 REAL-WORLD OUTCOMES AND TREATMENT PATTERNS OF CMV INFECTIONS IN HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS, REFRACTORY OR INTOLERANT TO TREATMENTS IN EUROPE, CANADA, ISRAEL: INTERIM ANALYSIS Johan Maertens 1, Matthew Pellan Cheng2, Avichai Shimoni3, Andreas Braun4, Nawal Bent-Ennakhil4, Irmgard Andresen4 P459 THE RESPECT STUDY DESIGN: COMPARISON OF ONCE-WEEKLY REZAFUNGIN AGAINST STANDARD ANTIMICROBIAL PROPHYLAXIS IN ADULTS UNDERGOING ALLOGENEIC BLOOD AND MARROW TRANSPLANTATION Johan Maertens 1,2, Federica Sora3,4, Drew J. Winston5, Shariq Haider6, Lourdes Vazquez Lopez7, Christine Robin8, Alexander Schauwvlieghe9, Dominik Selleslag9, Taylor Sandison10, Kieren A. Marr11,12,13 P460 NO INFERIOR OUTCOME OF OMITTING FLUOROQUINOLONE PROPHYLAXIS IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Vanessa Nehrbaß 1, Krischan Braitsch1, Katharina Nickel1, Maike Hefter1, Katrin Koch1, Kathrin Rothe1, Florian Bassermann1, Jochen Schneider1, Katharina S. Götze1, Peter Herhaus1, Mareike Verbeek1 P461 OUTCOMES, CHOICE OF ANTIBIOTICS AND THEIR EFFECTIVENESS IN ALLOGENEIC-HSCT RECIPIENTS COLONIZED WITH MULTIRESISTANT GRAM-NEGATIVE BACTERIA: A SINGLE-CENTER RETROSPECTIVE ANALYSIS Igor Age Kos 1, Jana Speer2, Onur Cetin1, Vadim Lesan1, Angelika Bick1, Konstantinos Christofyllakis1, Manfred Ahlgrimm1, Sigrun Smola3, Andreas Link1, Torben Rixecker1, Anna K. H. Hirsch4,5, Joerg Thomas Bittenbring1, Fabian Berger3, Sören Becker6, Lorenz Thurner1, Moritz Bewarder1 P462 LETERMOVIR AS TREATMENT AND SECONDARY PROPHYLAXIS OF CMV REACTIVATION IN LOW-WEIGHT PEDIATRIC PATIENTS Chiara Mainardi 1, Cecilia Liberati1, Marica De Pieri1, Antonio Marzollo1, Manuela Tumino1, Maria Gabelli1, Marta Pillon1, Elisabetta Calore1, Alessandra Biffi1,2 P463 COMPARISON OF BLOOD STREAMING INFECTIONS BETWEEN TUNNELED CENTRAL VENOUS CATHETERS AND NON-TUNNELED CENTRAL VENOUS CATHETERS IN ACUTE LEUKEMIA REMISSION INDUCTION – A SINGLE CENTER EXPERIENCE Cheongin Yang 1, Seong Hyun Jeong1, Joon Seong Park1 P464 INCIDENCE OF INFECTIONS AND QUALITY OF IMMUNE RECONSTITUTION FOLLOWING CRYOPRESERVATION OF PERIPHERAL BLOOD STEM CELLS FROM HLA-IDENTICAL DONOR Luca Barabino1, Stefania Bregante2, Massimiliano Gambella 2, Anna Ghiso2, Livia Giannoni2, Silvia Lucchetti2, Alberto Serio2, Riccardo Varaldo2, Antonella Laudisi2, Monica Passannante2, Roberta Murru3, Andrea Galitzia1, Alessandra Bo2, Emanuele Angelucci2, Giovanni Caocci1, Anna Maria Raiola2 P465 DETECTION OF RESPIRATORY VIRUSES AMONG PAEDIATRIC HSCT RECIPIENTS – A TEN YEARS SINGLE-CENTRE EXPERIENCE Petr Hubacek 1,2, Petr Riha2, Ales Briksi2, Petra Keslova2, Renata Formankova1,2, Daniela Janeckova2, Miroslav Zajac2, Zdenek Kepka2, Petr Sedlacek1,2 P466 MUTUAL IMPACT AND CHARACTERISTICS OF VIRAL REACTIVATIONS AND IMMUNE RECONSTITUTION AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN. A SINGLE CENTER EXPERIENCE Cristian Jinca 1,2, Andrada Oprisoni1,2, Anca Isac2, Andreea Pascalau2, Loredana Balint-Gib2, Margit Serban2, Mihaela Baica2, Smaranda Arghirescu1,2 P467 CMV MONITORING AND LETERMOVIR PROPHYLAXIS IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A SINGLE CENTER EXPERIENCE Yuta Katayama 1, Yu Kochi1, Takuya Nunomura1, Riichiro Ikeda1, Kenjiro Hino1, Ryota Imanaka1, Kohei Kyo1, Takeshi Okatani1, Mitsuhiro Itagaki1, Shinya Katsutani1, Tsuyoshi Muta1 P468 LETERMOVIR COMBINED WITH GANCICLOVIR AS A PREEMPTIVE OR TREATMENT FOR CYTOMEGALOVIRUS INFECTION FOLLOWING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION, A SINGLE-CENTER RETROSPECTIVE REVIEW Han Yao 1,2, Yimei Feng1,2, Ting Chen1,2, Lu Zhao1,2, Yuqing Liu1,2, Lidan Zhu1,2, Jia Liu1,2, Lu Wang1,2, Shichun Gao1,2, Huanfeng Liu1,2, Lei Gao1,2, Peiyan Kong1,2, Xi Zhang1,2,3 P469 THE CLINICAL MANIFESTATION, PROGNOSTIC FACTORS, AND OUTCOMES OF ADENOVIRUS PNEUMONIA AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Yuewen Wang 1, Xiaohui Zhang1, Lanping Xu1, Yu Wang1, Chenhua Yan1, Huan Chen1, Yuhong Chen1, Fangfang Wei1, Wei Han1, Fengrong Wang1, Jingzhi Wang1, Xiaojun Huang1, Xiaodong Mo1 P470 OPTIMIZING CMV PREVENTION IN STEM CELL TRANSPLANTS: HYBRID STRATEGY DELIVERS PROMISING REDUCTION IN REACTIVATION RATES Amirabbas Rashidi1,2, Tanaz Sayar Bahri1,2, Maryam Barkhordar 1,2, Seied Asadollah Mousavi1,2, Ahmad Khajeh-Mehrizi3, Mohammad Vaezi1,2 P471 STRATIFICATION AND IMMUNOLOGIC ANALYSIS OF PATIENTS WITH SARS-COV-2 INFECTION AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION Man Chen 1, Wei Zhao1, Hui Wang1 P472KLEBSIELLA PNEUMONIAE INFECTION BEFORE AND AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN THE ERA OF NOVEL ANTIBIOTICS: A COMPARATIVE STUDY Eleni Gavriilaki 1, Damianos Sotiropoulos2, Ioannis Batsis2, Despina Mallouri2, Alkistis Panteliadou2, Nikolaos Spyridis2, Giorgos Karavalakis2, Paschalis Evangelidis1, Vasiliki Kanava2, Giannis Kyriakou2, Eleni Papchianou2, Christos Demosthenous2, Zoi Bousiou2, Anna Vardi2, Ioanna Sakellari2 P473 OUTCOMES OF HYPERBARIC OXYGEN THERAPY IN LATE-ONSET HEMORRRHAGIC CYSTITS AFTER ALLO-HSCT: A RETROSPECTIVE ANALYSIS Mariana Trigo Miranda 1, Joana Arana Ribeiro2, Diogo Alpuim Costa3,4,3, Carla D’Espiney Amaro4,3, Andreia Teixeira5,6, Inês Portugal Rodrigues7, Clara Gaio-Lima7, Carlos Pinho Vaz8, Óscar Camacho7 P474 INVASIVE FUNGAL DISEASES IN PATIENTS WITH CLL TREATED WITH BRUTON TYROSINE KINASE INHIBITORS, A MONO-CENTRIC RETROSPECTIVE STUDY Yuri Vanbiervliet 1, Robina Aerts1, Finn Segers1, Johan Maertens1, Ann Janssens1 P475 PREDICTORS OF INVASIVE FUNGAL INFECTION AND IMPACT OF OUTCOME AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION: SINGLE CENTER EXPERIENCE FROM THE KINGDOM OF SAUDI ARABIA Lama AlHmaly 1, Ahmad Alharbi1, Moussab Damlaj1,2,3, Ahmed Alaskar1,2,3,4, Ayman Hejazi1,2,3, Hind Salama1,2,3, Abdulrahman Al raizah1,2,3, Abdullah S. Al Saleh1,2,3, Ayman Ibrahim1,2,3, Ayel Yahya1,2,3, Mohammed Bakkar1,2,3, Inaam Shehabeddine1, Suha Alkhraisat1, Amani Alharbi1, Isam Mahasneh1, Maybelle Ballili1, Mazen Ahmed1, Husam Alsadi1,2,3, Mohsen Alzahrani1,2,3,4, Bader Alahmari1,2,3 P476 THE ROLE OF COLONIZATION WITH RESISTANT G-BACTERIA IN THE TREATMENT OF FEBRILE NEUTROPENIA AFTER STEM CELL TRANSPLANTATION Tereza Sokolova1, Pavla Paterova1, Alzbeta Zavrelova1, Benjamin Visek1, Pavel Zak1, Jakub Radocha 1 P477 OUTCOME OF THE USE OF LETERMOVIR AS PROPHYACTIC TREATMENT FOR HSCT RECIPIENTS: A SINGLE CENTER EXPERIENCE Giulia Baresi1, Marianna Maffeis 2, Elena Soncini1, Stefano Rossi1, Giulia Albrici1, Marta Comini3, Federica Bolda3, Alessandra Beghin3, Arnalda Lanfranchi3, Fulvio Porta1 P478 COMPARATIVE EVALUATION OF THE EFFECTIVENESS OF LETERMOVIR SINCE ITS USE IN PREVENTION OF CYTOMEGALOVIRUS (CMV) INFECTIONS IN ALLOGENEIC TRANSPLANT PATIENTS HEMATOLOGY DEPARTMENT OF CAEN UNIVERSITY HOSPITAL Tchuanga Djialeu Yannick Noel 1, Gandhi Damaj Laurent2, Johnson Ansah Hyacinthe Atchroue2, Dina J.2, Parienti Jean Jacques2, Chantepie Sylvain2 P479 EXPERIENCE WITH THE USE OF MARIBAVIR IN HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS WITH REFRACTORY OR RESISTANT CYTOMEGALOVIRUS (CMV) Mónica Fernández Pérez1, Candela Ceballos Bolaños 1, Itziar Oiartzabal Ormategui1, Nerea Arratibel Zalacain1, Pamela Millacoy Austenrritt1, Marina Aranguren Ostolaza1, Maria Cruz Viguria Alegria1, Carlos Manuel Panizo Santos1 P480 IS THERE A PLACE FOR ANTI-HCMV HUMAN IMMUNOGLOBULIN AS PRE-EMPTIVE THERAPY IN T-DEPLETE TREG/TCON ALLOGENEIC STEM CELL TRANSPLANTATION? Valerio Viglione 1, Loredana Ruggeri2, Antonella Mancusi1, Sara Tricarico2, Alessandra Cipiciani1, Rebecca Sembenico1, Matteo Caridi1, Francesco Zorutti1, Anna Castaldo1, Simonetta Saldi1, Roberto Castronari2, Silvia Bozza1, Cynthia Aristei1, Antonella Mencacci1, Maria Paola Martelli1, Antonio Pierini1, Alessandra Carotti2 P481 FREQUENCY AND OUTCOME OF CYTOMEGALOVIRUS INFECTION IN RECIPIENTS OF HAEMATOPOIETIC STEM CELL TRANSPLANTATION – AN EXPERIENCE FROM PAKISTAN Natasha Ali 1, Zurrya Khan1, Mohammad Usman Shaikh1, Zehra Fadoo1, Salman Adil1 P482 CENTRAL VENOUS ACCESS DEVICE IN AUTOLOGOUS STEM CELL TRANSPLANT PATIENTS Danah Chakfeh 1, Antonette Amao1, Baaba Bentil Tumi1, Josephine Crowe1 P483 FACTORS AFFECTING OUTCOME OF SALVAGE CHEMOTHERAPY AND AUTOLOGOUS STEM CELL TRANSPLANT IN PATIENTS WITH CLASSICAL HODGKIN LYMPHOMA RELAPSING OR PROGRESSING AFTER A FRONT-LINE PET-ADAPTED THERAPY Simonetta Viviani1, Anna Vanazzi1, Samuele Frassoni2, Chiara Rusconi3, Andrea Rossi4, Lessandra Romano5, Caterina Patti6, Corrado Schiavotto7, Roberto Sorasio8, Vincenzo Marasco3, Laura Lissandrini7, Davide Rapezzi8, Daniela Gottardi9, Federica Cocito10, Antonio Mulé11, Salvatore Leotta5, Guido Gini12, Marco Sorio13, Enrico Derenzini1, Alessandro Rambaldi 4,14, Vincenzo Bagnardi2, Corrado Tarella1,14 P484 REDUCED RELAPSE INCIDENCE AFTER CHECK-POINT INHIBITORS RELATIVE TO BRENTUXIMAB VEDOTIN AS SALVAGE THERAPY BEFORE ALLOGENEIC STEM CELL TRANSPLANTATION FOR REFRACTORY/RELAPSED HODGKIN LYMPHOMA: A RETROSPECTIVE ANALYSIS Jacopo Mariotti 1, Chiara Pinton1, Chiara de Philippis1, Daniele Mannina1, Barbara Sarina1, Daniela Taurino1, Armando Santoro1, Stefania Bramanti1 P485 LOW NON-RELAPSE MORTALITY ASSOCIATED WITH ALLOGENEIC HAEMATOPOIETIC CELL TRANSPLANTATION AFTER CAR-T FAILURE IN PATIENTS AFFECTED BY LARGE B CELL LYMPHOMA Chiara de Philippis1, Massimiliano Gambella2, Jacopo Mariotti 1, Anna Dodero3, Patrizia Chiusolo4, Alessia Castellino5, Laura Giordano6, Barbara Sarina1, Daniela Taurino1, Daniele Mannina1, Anna Maria Raiola2, Simona Sica4, Carmelo Carlo-Stella1, Emanuele Angelucci2, Armando Santoro1, Paolo Corradini3, Stefania Bramanti1 P486 TREOSULFAN-BASED CONDITIONING AND SIROLIMUS-PTCY GVHD PROPHYLAXIS IN ALLOGENEIC STEM CELL TRANSPLANTATION FOR AGGRESSIVE B-CELL NHL Lorenzo Lazzari 1, Alessandro Bruno1, Simona Piemontese1, Federico Erbella1, Piera Angelillo1, Maria Teresa Lupo Stanghellini1, Andrés Ferreri1,2, Fabio Ciceri1,2, Raffaella Greco1, Jacopo Peccatori1 P487 OUTCOME OF HIGH-DOSE CHEMOTHERAPY FOLLOWED BY AUTOLOGOUS HEMATOPOIETIC CELL TRANSPLANTATION IN PATIENTS WITH PERIPHERAL T-CELL LYMPHOMA AT A SINGLE INSTITUTION OVER 20 YEARS Joanna Romejko-Jarosińska 1, Ewa Paszkiewicz-Kozik1, Marcin Szymanski1, Lidia Poplawska1, Anna Borawska1, Anna Dabrowska-Iwanicka1, Katarzyna Domanska-Czyz1, Agnieszka Druzd-Sitek1, Robert Konecki1, Martyna Kotarska1, Ewa Mroz-Zycińska1, Wlodzimierz Osiadacz1, Beata Ostrowska1, Monika Swierkowska1, Joanna Tajer1, Lukasz Targonski1, Elzbieta Wojciechowska-Lampka1, Jan Walewski1 P488 IMPROVING RESULTS OF ALLO-HCT FOR PATIENTS WITH RELAPSED/REFRACTORY HODGKIN LYMPHOMA. A SINGLE CENTER EXPERIENCE Andrzej Frankiewicz 1, Małgorzata Ociepa-Wasilkowska1, Tomasz Czerw1, Monika Dzierżak-Mietła1, Magdalena Głowala-Kosińska1, Jerzy Hołowiecki1, Małgorzata Krawczy-Kuliś1, Włodzimierz Mendrek1, Iwona Mitrus1, Jacek Najda1, Maria Saduś-Wojciechowska1, Małgorzata Sobczyk-Kruszelnicka1, Sebastian Giebel1 P489 THE SINGLE-CELL LANDSCAPE EXPLORING ABNORMAL T CELL STATES AND DEVELOPMENTAL TRAJECTORIES IN HETEROGENEOUS NON-HODGKIN LYMPHOMA Yuqing Wang 1, Cong Wang1, Chanmin Xiao1, Zheng Wang1, Xi Zhang1 P490 BEYOND TRANSPLANTATION: BRENTUXIMAB’S IMPACT ON LONG-TERM OUTCOMES AFTER AUTOLOGOUS STEM CELL TRANSPLANT (ASCT) IN ROMANIAN TRANSPLANT CENTERS Lavinia Eugenia Lipan1,2, Andrei Colita3,2, Angela Dascalescu4, Anca Colita1,2, Zsofia Varady1, Laura Diana Stefan1, Miruna Tirnovan1, Alina Daniela Tanase 1,2 P491 ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION FOR T CELL LYMPHOMAS: A RETROSPECTIVE ANALYSIS FROM THE POLISH LYMPHOMA RESEARCH GROUP (PLRG) Malgorzata Sobczyk-Kruszelnicka 1, Tomasz Czerw1, Joanna Drozd-Sokołowska2, Patrycja Mensah-Glanowska3, Agnieszka Piekarska4, Jarosław Dybko5, Anna Łojko-Dankowska6, Anna Czyż7, Jan Maciej Zaucha4, Sebastian Giebel1 P492 OPTIMIZING OUTCOMES: THE VITAL SIGNIFICANCE OF TRANSPLANTATION IN RESCUING PATIENTS DISCONTENTED WITH INITIAL PERIPHERAL T-CELL LYMPHOMA TREATMENT Hongye Gao1, Zhuoxin Zhang1, Jiali Wang1, Yannan Jia2, Hao Zhang3, Xin Du4, Xianmin Song2, Yao Liu5, Dehui Zou1, Erlie Jiang 1 P493 LOW DOSE NIVOLUMAB IN COMBINATION WITH SALVAGE CHEMOTHERAPY BEFORE AUTO-HSCT IN PRIMARY REFRACTORY HODGKIN LYMPHOMA – A PILOT STUDY Rakesh Reddy Boya 1, Pradeep Ventrapati1, Veni Prasanna Gedala1, Chandrasekhar Bendi2 P494 PREDICTORS OF DELAYED PLATELET ENGRAFTMENT AFTER AUTOLOGOUS STEM CELL TRANSPLANTATION IN NON-HODGKIN LYMPHOMA Rita Costa e Sousa 1, Cátia Almeida1, João Gaião Santos1, Ricardo Ferreira1, Mafalda Urbano1, Maria Carolina Afonso1, Marília Gomes1, Adriana Roque1, Catarina Geraldes1 P495 NON-MYELOABLATIVE ALLOGENEIC TRANSPLANTATION AFTER TOTAL SKIN ELECTRON BEAM THERAPY (TSEBT) IN CUTANEOUS T-CELL LYMPHOMA. A SINGLE CENTER EXPERIENCE Carlos De Miguel 1, Belén Navarro1, Irma Zapata1, Mercedes Hospital1, Guiomar Bautista1, Ana Bocanegra1, José Antonio García-Vela1, María Esther Martínez-Muñoz1, Silvia Monsalvo1, José Luis Bueno1, Isabel Salcedo1, Luis Gastón Roustan1, Jesús Romero1, Rafael Duarte1 P496 ROLE OF AUTOLOGOUS STEM CELL TRANSPLANT IN PRIMARY AND SECONDARY CENTRAL NERVOUS SYSTEM LYMPHOMA – A SINGLE CENTER EXPERIENCE Meredith Tan 1,2, Tertius Tuy1,2, Melinda Tan1,2, Chieh Hwee Ang1,2, Jeffrey Quek1,2, Than Hein1,2, Yunxin Chen1,2, Francesca Lim1,2, Chandramouli Nagarajan1,2, Jordan Hwang1, Jing Jing Lee1, Lalitha Krishnan1, Lyn Lee Wong1, Zi Jing Seng1, Yeow Tee Goh1,2, Yeh Ching Linn1,2, Colin Diong1,3, Yuh Shan Lee1,3, William Hwang1,2, Aloysius Ho1,2, Lawrence Ng1,2 P497 HIGH DOSE THERAPY FOLLOWED BY AUTOLOGOUS STEM CELL TRANSPLANTATION IN 236 PATIENTS WITH HODGKIN LYMPHOMA ON 25 YEARS PERIOD (1998-2022) Sabrina Akhrouf 1, Hanane Bouarab1, Rihab Benouattas1, Farih Mehdid1, Nadia Rahmoun1, Mounira Baazizi1, Dina Ait ouali1, Nacera Ait Amer1, Farida Tensaout1, Rose Marie Hamladji1, Redhouane Ahmed Nacer1, Malek Benakli1 P498 NON-CRYOPRESERVED AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN HODGKIN LYMPHOMA. ELEVEN-YEAR SINGLE CENTRE EXPERIENCE FROM ORAN. ALGERIA Nabil Yafour 1, Kamila Amani1, Nawel Bounoua1, Fatima Aoudia1, Amel Bendimerad1, Mohamed Amine Benaissa1, Nour El Houda Hassam1, Mounir Serradj1, Manel Maarouf1, Leila Charef1, Badra Enta Soltane1, Soufi Osmani1, Rachid Bouhass1, Abdessamad Arabi1, Mohamed Brahimi1, Mohamed Amine Bekadja1 P499 AUTOLOGOUS AND ALLOGENEIC STEM CELL TRANSPLANTATION IN MATURE NK/T-CELL LYMPHOMA: A TWO-CENTER RETROSPECTIVE REAL-LIFE ANALYSIS Michele Wieczorek 1, Giorgia Bonetto1, Beatrice Bugnotto2,1, Alessia Moioli2, Albana Lico1, Francesca Elice1, Marcello Riva1, Cristina Tecchio2, Mauro Krampera2, Carlo Borghero1, Alberto Tosetto1 P500 PRETREATMENT WITH RITUXIMAB IS SUSPECTED TO INCREASE RISK FOR OCCURRENCE OF VENO-OCCLUSIVE DISEASE IN PATIENTS WITH AGGRESSIVE LYMPHOMA AFTER ALLOGENEIC TRANSPLANT Ahmet Elmaagacli 1, Mathis Samuel Bittermann1, Farouk Dahmash1, Anju Singh1, Yana Shikova1, Vitaly Varyushkin1, Mathias Vierbuchen1, Hans Salwender1, Christian Jehn1 P501 GENETIC PREDICTORS OF PROGNOSIS IN CHRONIC LYMPHOCYTIC LEUKEMIA: INSIGHTS FROM NEXT-GENERATION SEQUENCING Emilia Jaskuła1,2, Anna Sobczyńska-Konefał2,1, Iga Jendrysik2, Marzena Wojtaszewska3, Monika Mordak-Domagała2, Krzysztof Suchnicki2, Mariola Sędzimirska2, Monika Jasek1, Lidia Karabon1, Jarosław Dybko 2 P502 A REAL WORLD EXPERIENCE WITH PD1 INHIBITORS IN RELAPSED/REFRACTORY HODGKINS LYMPHOMA Disha Kakkar 1, Narendra Agarwal1, Tribikram Panda1, Aakanksha Singh1, Rohan Halder1, Roy J. Palatty1, Dinesh Bhurani1 P503 PEGFILGRASTIM AFTER CONDITIONING WITH BEAM AND AUTOLOGOUS HEMATOPOIETIC PERIPHERAL BLOOD STEM CELL TRANSPLANTATION IN LYMPHOMA PATIENTS Barbara Loteta 1, Giovanni Tripepi2, Pitino Annalisa3, Mercedes Gori3, Gaetana Porto1, Giovanna Utano1, G. Policastro1, Ludovica Santoro1, Maria Caterina Micò1, Massimo Martino1 P504 BEGEV AS SECOND-LINE SALVAGE THERAPY IN RELAPSED/REFRACTORY HODGKIN LYMPHOMA: A REAL‐LIFE EXPERIENCE Ant Uzay 1, Elif Şenocak Taşçı1, Arda Ulaş Mutlu1, Barış Koşan2, Aybüke Görkem Koç1, Bülent Küçük2, S. Sami Kartı2 P505 EVALUATION IN SARDINIAN GENETIC UNIQUENESS POPOLATION: STEREOTYPED SUBSETS# AND MOLECULAR PROGNOSTIC MARKERS, CATEGORIZATION IN U-MUTATED POOR PROGNOSTIC GROUP OF CHRONIC LYMPHOCYTIC LEUKEMIA (CLL) Fabio Culurgioni 1, Roberta Murru2, Alba Piras2, Wedad Salem Hamdi1, Manal Yaghmour1, Abdallah Alkhoujah1, Sara Alromaihi1, Aisha Alkaseri1, Nasimeh Azadi1, Giorgio La Nasa2, Einas Al Kuwari1 P506 A NATIONWIDE STUDY OF CHRONIC LYMPHOCYTIC LEUKEMIA INCIDENCE AND MORTALITY IN THE REPUBLIC OF KOREA Min Ji Jeon1, Hoonji Oh1, Kunye Kwak1, Eun Sang Yu1, Dae Sik Kim1, Chul Won Choi1, Byung-Hyun Lee1, Se Ryeon Lee1, Hwa Jung Sung1, Yong Park1, Byung Soo Kim1, Ka-Won Kang 1 P507 AUTOLOGOUS HEMATOPOETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH RELAPSED AND REFRACTORY PRIMARY MEDIASTINAL LARGE B-CELL LYMPHOMA Alexander Galimov1, Andrey Kozlov1, Ilya Kazantsev1, Tatiana Yuhta1, Polina Tolkunova1, Natalia Mihailova1, Yuri Punanov1, Vadim Baykov1, Ivan Moiseev1, Alexander Kulagin1, Lyudmila Zubarovskaya 1 P508 HAPLOIDENTICAL TRANSPLANTATION OF HEMATOPOIETIC STEM CELLS FOR PATIENT WITH POST-TRANSPLANT LYMPHOPROLIFERATIVE DISEASE Vera Vasilyeva 1, Larisa Kuzmina1, Olga Aleshina1, Mariya Dovydenko1, Mikhail Drokov1, Irina Lukyanova1, Vera Troitskaya1, Elena Parovichnikova1 P509 ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANT IN LYMPHOMA: A SINGLE-CENTRE EXPERIENCE; A CASE SERIES Jan Miko Aaron Baybay 1, Francisco Vicente Lopez1 P510 THE FIRST CASE OF ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION AT DHARMAIS HOSPITAL – INDONESIAN NATIONAL CANCER CENTER Dwi Wahyunianto Hadisantoso 1, Resti Mulya Sari1, Edel Herbitya1, Lyana Setiawan1, Hubertus Hosti Hayuanta1, Muhammad Raya Kurniawan1, Yanto Ciputra1, Annisa Annisa1, I Gusti Ngurah Agastya1, Della Manik Worowerdi Cintakaweni1 P511 PRE-EMPTIVE DONOR LYMPHOCYTE INFUSION AS OPTIMAL TREATMENT FOR RELAPSED ACUTE MYELOID LEUKEMIAS AND MYELODYSPLASTIC SYNDROMES FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION:A FRENCH-ITALIAN EXPERIENCE WITH 103 PATIENTS Eugenia Accorsi Buttini1, Cristina Doran2, Michele Malagola 1, Vera Radici1, Mirko Farina1, Marco Galli1, Gabriele Magliano1, Alessandro Leoni3, Federica Re3, Simona Bernardi3, Mohamad Mohty2, Domenico Russo1, Eolia Brissot2 P512 HLA-DR+REGULATORY T CELLS ARE ASSOCIATED WITH THE ONSET AND SEVERITY OF ACUTE GRAFT-VERSUS-HOST DISEASE AFTER HEMATOPOIETIC CELL TRANSPLANTATION Kinga Hosszu 1, Devin McAvoy1, Moises Garcia-Rosa2, Charlie White1, Matthew Thomsen1, Evangelos Ntrivalas1, Elizabeth Klein1, Abdulrahman Alsultan1, Audrey Mauguen1, Kevin J. Curran1, Maria Cancio1, Andromachi Scaradavou1, Andrew Kung1, Joseph H. Oved1, Miguel-Angel Perales1, Andrew C. Harris1, Jaap Jan Boelens1 P513 MAINTENANCE THERAPY WITH OLAPARIB AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH TP53 MUTATED HEMATOLOGIC MALIGNANCIES Zhihui Li 1, Teng Xu1, Yipei Guo1, Xianxuan Wang1, Xiaopei Wen1, Lei Wang1, Jingjing Wang1, Yanzhi Song1, Yongqiang Zhao1, Tong Wu1 P514 PERSISTENCE OF MINOR HISTOCOMPATIBILITY ANTIGEN- AND VIRUS-SPECIFIC T CELLS IN LEUKEMIC REMISSION AFTER HEMATOPOIETIC ALLOGENEIC STEM CELL TRANSPLANTATION Lisa Marie Schulz 1, Debora Basilio-Queiros1, Susanne Luther-Wolf1, Elke Dammann1, Michael Stadler1, Eva Mischak-Weissinger1 P515 SERIAL LINEAGE CHIMERISM ANALYSIS IMPROVES EARLY DIAGNOSIS OF GRAFT FAILURE AFTER ALLOGENEIC HAPLOIDENTICAL HSCT Pilar Lancho Lavilla1, Ignacio Gómez Centurión1,2, Rebeca Bailén Almorox 1,2, Paula Fernández-Caldas1,2, Asunción Escudero1, Lucía Castilla1,2, Javier Anguita1,2,3, Ismael Buño1,2,4,3, Mi Kwon1,2,3, Carolina Martínez-Laperche1,2,4 P516 PREDICTING MEASURABLE RESIDUAL DISEASE FOR ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS POSTTRANSPLANTATION Yue-Wen Wang 1, Guo-Mei Fu1, Lan-Ping Xu1, Yu Wang1, Yi-Fei Cheng1, Yuan-Yuan Zhang1, Xiao-Hui Zhang1, Yan-Rong Liu1, Kai-Yan Liu1, Xiao-Jun Huang1, Ying-Jun Chang1 P517 CYTOKINE PROFILES ASSOCIATED WITH GRAFT VERSUS HOST DISEASE AND RECENT THYMIC EMIGRANT T CELL RECONSTITUTION AFTER HEMATOPOIETIC CELL TRANSPLANTATION Devin McAvoy 1, Kinga Hosszu1, Matthew Thomsen1, Evangelos Ntrivalas1, Elizabeth Klein1, Katina Singh1, Esther Vidal1, Kevin Curran1, Maria Cancio1, Andromachi Scaradavou1, Joseph Oved1, Miguel-Angel Perales1, Andrew Harris1, Andrew Kung1, Jaap Jan Boelens1 P518 FAVOURABLE IMPACT OF POST-TRANSPLANT MINIMAL DISEASE NEGATIVITY ASSESSED BY FLOW CYTOMETRY ON SURVIVAL IN PATIENTS WITH MYELODYSPLASTIC SYNDROMES Evgeny Klyuchnikov1, Anita Badbaran1, Tetiana Perekhrestenko2, Normann Steiner1, Radwan Massoud 1, Petra Freiberger1, Christine Wolschke1, Francis Ayuk1, Ulrike Bacher3, Nicolaus Kröger1 P519 FLOW-CYTOMETRIC AND TRANSCRIPTIONAL CHARACTERIZATION OF RECENT THYMIC EMIGRANTS AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) IN CHILDREN WITH ACUTE LYMPHOBLASTIC LEUKEMIA (ALL) Silvia Nucera1,2, Francesca Limido1,2,3, Marco Maria Sindoni 1,2, Cristina Bugarin1, Grazia Fazio1, Andrea Biondi1,2,3, Adriana Balduzzi2,3, Giuseppe Gaipa1 P520 LOW PERCENTAGES OF EARLY B-CELL PRECURSORS IN THE BONE MARROW MAY PREDICT THE DEVELOPMENT OF CGVHD FOLLOWING PEDIATRIC HSCT Elisa Christine Peen 1, Klaus Gottlob Müller1, Claus Henrik Nielsen1, Katrine Kielsen1, Hanne Vibeke Marquart1 P521 MEASURABLE RESIDUAL DISEASE (MRD) AND T-CELL CHIMERISM ARE PROGNOSTIC BIOMARKERS IN ACUTE MYELOID LEUKEMIA (AML) PATIENTS UNDERGOING ALLOGENEIC STEM CELLS TRANSPLANTATION (ASCT) Elisa Meddi 1, Raffaele Palmieri1, Giovanni Marsili2, Federico Moretti1, Flavia Mallegni1, Alfonso Piciocchi2, Luca Maurillo1, Maria Ilaria Del Principe1, Giovangiacinto Paterno1, Raffaella Cerretti1, Gottardo De Angelis1, Benedetta Mariotti1, Maria Irno Consalvo1, Mariadomenica Divona1, Tiziana Ottone1, Sara Gargiulo1, Giulia Colafranceschi1, Maria Teresa Voso1, Adriano Venditti1, Francesco Buccisano1 P522 THE NUMBER OF CENTRAL MEMORY CD8 + T CELLS INFUSED AND THEIR EARLY RECOVERY AFTER ALLOGENEIC HSCT WAS ASSOCIATED WITH A DECREASED RISK OF DISEASE RELAPSE Valle Gómez García de Soria 1,2,3, Paula Díaz-Fernández1,2,3, Ana Marcos-Jiménez1,2, Nuria Montes-Casado1,2, Javier Sevilla-Montero1,2, Itxaso Portero-Sainz1,2, Yaiza Pérez-García1,2, Ángela Figuera Álvarez1,2,3, Rafael de la Cámara1,2, Cecilia Muñoz-Calleja1,2,3 P523 CONTRASTING GENE EXPRESSION PROFILES AND SIGNALING PATHWAYS BETWEEN CIRCULATING DONOR-AND HOST-DERIVED MONOCYTES DURING TOLERANCE IN MIXED CHIMERISM FOLLOWING UNRELATED DONOR CADAVERIC BMT AND LUNG TRANSPLANT Paul Szabolcs 1, Evelyn Garchar1, Dhivyaa Rajasundaram1, Xiaohua Chen1 P524 MEASURABLE RESIDUAL DISEASE AND CHIMERISM ANALYSIS INTERPLAY: IMPACT IN PROGNOSIS OF ACUTE MYELOID LEUKEMIA PATIENTS UNDERGOING ALLOGENIC STEM CELL TRANSPLANTATION Manuel Jorge Fernandez-Villalobos1, Ignacio Gomez-Centurion1,2, Rebeca Bailen 1,2, Paula Fernandez-Caldas1,2, Lucia Castilla1, Ana Pérez-Corral1,2, Carolina Martinez-Laperche1,2, Mi Kwon1,2,3 P525 THE INFLUENCE OF GRAFT IMMUNE COMPOSITION ON IMMUNE RECOVERY AND TRANSPLANT OUTCOME: A SINGLE CENTER EXPERIENCE Stefania Leone 1, Serena Marotta1, Maria Celentano1, Mariangela Pedata1, Cristina Luise1, Angela Carobene1, Ilaria Migliaccio1, Alfonso Fiumarella2, Aldo Leone2, Mario Toriello3, Daniela Graziano1, Mirella Alberti1, Simona Maria Muggianu Muggianu1, Mafalda Caputo1, Assunta Viola1, Roberta Penta3, Claudio Falco1, Antonio Meles1, Alessandra Picardi1,4 P526 RELAPSE MONITORING BY NPM1-PCR AND ITS PROGNOSTIC VALUE BEFORE AND AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Oliver Kriege 1, Johannes Barucha2,1, Markus Radsak3,1, Jonas Wißkirchen1, Pascal Wölfinger1, Beate Hauptrock1, Eva-Maria Wagner-Drouet1 P527 GUIDING POST-TRANSPLANT RELAPSE TREATMENT IN MYELOID MALIGNANCIES WITH QPCR MONITORING OF CHIMERISM DYNAMIC FROM PERIPHERAL WHOLE BLOOD – SINGLE CENTRE EXPERIENCE Ana Bošković1, Tadeja Dovč Drnovšek2, Primož Rožman2, Klara Šlajpah1, Njetočka Gredelj Šimec1, Matjaž Sever1, Polona Novak 1 P528 THE COMBINATION OF DISEASE BIOLOGY WITH PRE-HSCT MRD STATUS COULD BETTER STRATIFY RELAPSE RISK AFTER HSCT AMONG ADULT AML PATIENTS IN FIRST COMPLETE REMISSION Margherita Ursi1,2, Francesco Barbato1,2, Francesco De Felice1,2, Enrico Maffini2, Marcello Roberto 1,2, Gianluca Storci2, Salvatore Nicola Bertuccio2, Daria Messelodi2, Serena De Matteis2, Noemi Laprovitera2, Maria Naddeo2, Irene Salamon2, Francesco Iannotta2, Elisa Dan2, Luca Zazzeroni1, Barbara Sinigaglia2, Enrica Tomassini2, Massimiliano Bonafè1,2, Mario Arpinati2, Francesca Bonifazi2 P529 IMPACT OF ABSOLUTE LYMPHOCYTE COUNT RECOVERY AT DIFFERENT TIME-POINTS ON CLINICAL OUTCOMES AFTER THE ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Nour Ben Abdeljelil 1, Hana Ben Hammamia1, Ines Jemaa1, Rihab Ouerghi1, Insaf Ben Yaiche1, Ines Turki1, Lamia Torjemane1, Sabrine Mekni1, Rimmel Kanoun1, Dorra Belloumi1, Saloua Ladeb1, Tarek Ben othman1 P530 PHENOTYPES OF BONE MARROW MONOCYTES IN HEMATOPOIETIC CELL TRANSPLANTATION FOR ACUTE MYELOID LEUKEMIA: A DESCRIPTIVE PILOT STUDY Gül Yavuz Ermiş1, Klara Dalva1, Ekin Kırcalı2, Şenay İpek1, Nihal Okul1, Güldane Cengiz Seval1, Pervin Topçuoğlu1, Selami Koçak Toprak1, Meltem Kurt Yüksel 1 P531 AVAILABLE METHODS FOR MONITORING AML NPM1 + PATIENTS POST ALLOGENEIC TRANSPLANTATION CANNOT PREDICT OUTCOME Apostolia Papalexandri 1, Eleni Gavriilaki2, Vassiliki Kanava1, Panayiotis Dolgyras1, Fotini Kika1, Tasoula Touloumenidou1, Aggeliki Paleta1, Georgia Konstantinidou1, Panayiota Zerva1, Lamprini Vachtsetzi1, Christos Demosthenous1, Zoi Boussiou1, Anna Vardi1, Maria Papathanasiou1, Ioannis Batsis1, Anastasia Athanasiadou1, Anastasia Marvaki1, Chrisavgi Lalayianni1, Ioanna Sakellari1 P532 DONOR LYMPHOCYTE INFUSION, A SINGLE-CENTER RETROSPECTIVE SAFETY ANALYSIS Birgitte Strand Bergland 1, Anders Eivind Myhre1, Camilla Dao Vo1, Ingerid Weum Abrahamsen1, Tobias Gedde-Dahl1,2, Tor Henrik Anderson Tvedt1 P533 WT1 MONITORING AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: A NEW IDEA OF DYNAMIC ASSESSMENT THROUGH THE PARAMETER “DELTA-WT1 THRESHOLD”. A SINGLE-CENTRE RETROSPECTIVE STUDY Beatrice Manghisi 1, Paola Perfetti1, Marilena Fedele1, Elisabetta Terruzzi1, Andrea Aroldi1, Sonia Palamini2, Martina Venegoni2, Carlo Gambacorti Passerini3, Matteo Parma1 P534 CHIMERISM DYNAMICS POST ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A TOUGH NUT TO CRACK Fatma SE Ebeid1, Sara Makkeyah1, Safa Matbouly1, Heba G.A. Ali1, Nayera HK Elsherif 1 P535 ANALYSIS OF THE EXTENDED T-LYMPHOCYTES PHENOTYPE REVEALS DIFFERENCES IN THE EARLY IMMUNE RECONSTITUTION AMONG PEDIATRIC PATIENTS RECEIVING MATCHED-UNRELATED DONOR OR HAPLOIDENTICAL HSCT Alessandro Di Gangi 1,2, Giorgio Costagliola3, Annalisa Legitimo4, Eva Parolo3, Elisa Costa4, Chiara Lardone4, Gabriella Casazza3, Mariacristina Menconi3 P536 CLINICAL FACTORS ASSOCIATED WITH IMMUNOGLOBULINS AFTER ALLO-SCT Alberto Blanco Sánchez1, José María Sánchez Pina 1, Adolfo Sáez Marín1, Guillermo Ramos Moreno1, Esther Parra Virto1, Andrea Tamayo Soto1, Lucía Alba Medina1, Reyes Más Babio1, María Calbacho Robles1, Joaquín Martínez-López1 P537 REAL-WORLD OUTCOMES OF UPFRONT AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN NEWLY DIAGNOSED MULTIPLE MYELOMA PATIENTS WITH DEL (17P) Curtis Marcoux 1, Oren Pasvolsky2, Denái R. Milton2, Hina Khan3, Mark R. Tanner2, Amna Ahmed4, Qaiser Bashir2, Samer Srour2, Neeraj Saini2, Paul Lin2, Jeremy Ramdial2, Yago Nieto2, Guilin Tang2, Yosra Aljawai2, Hans C. Lee2, Krina K. Patel2, Partow Kebriaei2, Sheeba K. Thomas2, Donna M. Weber2, Robert Z. Orlowski2, Elizabeth J. Shpall2, Richard E. Champlin2, Muzaffar H. Qazilbash2 P538 IMPACT OF DARA-VTD INDUCTION THERAPY ON HEMATOPOIETIC STEM CELL COLLECTION AND ENGRAFTMENT IN MULTIPLE MYELOMA PATIENTS ELIGIBLE FOR ASCT: RESULTS OF THE REAL-LIFE PRIMULA STUDY Vanda Strafella1, Immacolata Attolico 2, Francesco Tarantini1,2, Paola Carluccio2, Paola Curci2, Nicola Sgherza2, Rita Rizzi1,2, Angelo Ostuni2, Gabriele Buda3, Maria Livia Del Giudice3, Viviana Beatrice Valli4, Giuseppe Mele5, Candida Rosaria Germano6, Angela Maria Quinto7, Giulia Palazzo8, Massimiliano Arangio Febbo9, Lorella Melillo10, Nicola Di Renzo11, Francesco Albano1,2, Pellegrino Musto1,2 P539 SAFETY AND EARLY EFFICACY OF TANDEM AUTOLOGOUS STEM-CELL TRANSPLANTATION AFTER DARATUMUMAB VTD INDUCTION IN NEWLY DIAGNOSED MULTIPLE MYELOMA Carmine Liberatore1, Elena Rossi2, Lara Malerba3, Francesca Fioritoni1, Francesca Di Landro2, Francesca Fazio4, Silvia Ferraro5, Giusy Antolino6, Laura De Padua7, Ugo Coppetelli8, Velia Bongarzoni9, Stefano Pulini1, Patrizia Chiusolo2, Doriana Vaddinelli1, Annalisa Natale 1, Giuseppe Visani3, Valerio De Stefano2, Mauro Di Ianni1 P540 RESPONSE AND SURVIVAL IMPROVEMENT WITH AUTOLOGOUS STEM CELL TRANSPLANTATION IN PATIENTS WITH NEWLY DIAGNOSED MULTIPLE MYELOMA: DIFFERENT INDUCTION THERAPY IMPACT OVER THE LAST 30 YEARS José Miguel Mateos Pérez 1,2, Francesc Fernández-Avilés3,2, Carlos Fernández de Larrea1,2, Maria Teresa Cibeira1,2, Natalia Tovar1,2, Carmen Martínez3,2, Luis Gerardo Rodrígez-Lobato1,2, Maria Suárez-Lledó3,2, Maria Queralt Salas3,2, Montserrat Rovira3,2, Joan Bladé1,2, Laura Rosiñol1,3,2 P541 OUTCOMES OF FRAIL PATIENTS RECEIVING HIGH-DOSE CHEMOTHERAPY/AUTOLOGOUS STEM CELL TRANSPLANTATION FOR MULTIPLE MYELOMA Stephanie Yohay 1, Temitope Oloyede2, Binod Dhakal1, Anita D’Souza1, Ayesha Aijaz3, Meera Mohan1, Ravi Narra1, Marcelo Pasquini1,2, Mehdi Hamadani1,2, Ciara Louise Freeman4, Othman Salim Akhtar1,2 P542 BASELINE INVESTIGATIONS AND AUTOLOGOUS STEM CELL TRANSPLANT OUTCOMES: A RETROSPECTIVE REVIEW OF ECHOCARDIOGRAPHY AND PULMONARY FUNCTION TESTS Yousif Badri 1, Amany Ihab Mohamed1, Gordon Cook1, Roger Owen1, Simon Bulley1, Sylvia Feyler1, Christopher Parrish1, Frances Seymour1 P543 IMPROVED SURVIVAL OF MULTIPLE MYELOMA PATIENTS IN RELAPSE AFTER AUTOLOGOUS STEM CELL FOR THOSE WHO PREVIOUSLY ACHIEVED COMPLETE REMISSION AFTER TRANSPLANT Nour Moukalled 1, Ammar Zahreddine1, Iman Abou Dalle1, Jean El Cheikh1, Ali Bazarbachi1 P544 NON-CRYOPRESERVED PERIPHERAL BLOOD STEM CELL AUTOLOGOUS TRANSPLANTATION FOR MULTIPLE MYELOMA AS A SAFE ALTERNATIVE IN COUNTRIES WITH LOW RESOURCES: A NINE-YEARS OF BICENTRIC EXPERIENCE Siham Ahchouch 1, Othman Doghmi1, Selim Jennane1, El Mehdi Mahtat1, Sara Bougar2, Saadia Zafad3, Hicham El Maaroufi1, Kamal Doghmi1 P545 UP-FRONT HIGH DOSE MELPHALAN AND AUTOLOGOUS STEM CELL TRANSPLANTATION FOR NEWLY DIAGNOSED MULTIPLE MYELOMA - SINGLE CENTER REAL-WORLD ANALYSIS OF 165 CASES Nobuhiro Tsukada 1, Taku Kikuchi1, Kodai Kunisada1, Yuki Oda1, Moe Yogo1, Tomomi Takei1, Kota Sato1, Mizuki Ogura1, Yu Abe1, Kenshi Suzuki1, Tadao Ishida1 P546 SETTING UP A HEMATOPOIETIC STEM CELL TRANSPLANT PROGRAM IN BAHRAIN -EXPERIENCE FROM BAHRAIN ONCOLOGY CENTER Shruti Prem Sudha1, Hazem Afify 1, Nabil Abdelfattah1, Volkan Kahraman1, Salih Aksu1, Aly Rashed1, Cigdem Ozturk1 P547 SALVAGE SECOND AUTOLOGOUS STEM CELL TRANSPLANTS FOR PATIENTS WITH MYELOMA: A 24-YEAR RETROSPECTIVE AUDIT IN A NATIONAL TERTIARY REFERRAL CENTRE (1999-2022) Micheal Brennan 1, Patrick Hayden1, James Fey1, Catherine Ronayne1, Orla Fallon1, Greg Lee1, Nicola Gardiner1, Ezzat El Hassadi2, Meegahage Perera3, Helen Enright4, Johnny McHugh4, Philip Murphy5, Patrick Thornton5, John Quinn5, Jeremy Sargent5, Mary McCloy6, Peter O’Gorman7, Denis O’Keeffe8, Hilary O’Leary8, Mark Gurney1 P548 NOVEL DRUGS INCLUDING MONOCLONAL ANTIBODIES AND/OR DONOR LYMPHOCYTE INFUSIONS CONFERRED LONG TERM SURVIVAL TO MULTIPLE MYELOMA PATIENTS RELAPSED AFTER ALLOGENEIC STEM CELL TRANSPLANTATION Chiara Nozzoli 1, Martina Pucillo2, Massimo Martino3, Luisa Giaccone4, Alessandro Rambaldi5, Edoardo Benedetti6, Domenico Russo7, Nicola Mordini8, Silvia Mangiacavalli9, Pietro Enrico Pioltelli10, Paola Carluccio11, Piero Galieni12, Marco Ladetto13, Simona Sica14, Miriam Isola15, Maria De Martino15, Elena Oldani5, Eliana Degrande16, Elisabetta Antonioli17, Renato Fanin2, Riccardo Saccardi1, Fabio Ciceri18, Francesca Patriarca2 P549 HAPLOIDENTICAL ALLOGENEIC CELL TRANSPLANTATION IN RELAPSED/REFRACTORY MULTIPLE MYELOMA Julia Frimmel 1, Anke Morgner2, Claudia Brogsitter3, Karolin Trautmann-Grill3, Desiree Kunadt3, Raphael Teipel3, Christoph Röllig3, Mathias Hänel2, Johannes Schetelig3,4, Friedrich Stölzel1, Martin Bornhäuser3,5,6 P550 ANTI-HLA ANTIBODIES AND INCIDENCE OF PLATELET TRANSFUSION IN PATIENTS WITH MULTIPLE MYELOMA AFTER AUTO-HSCT Elena Kuzmich1, Irina Pavlova1, Ivan Kostroma1, Sergey Gritsaev 1 P551 AUTOLOGOUS PERIPHERAL BLOOD STEM CELL HARVESTING AND UTILIZATION IN MULTIPLE MYELOMA PATIENTS IN LUHS KAUNAS CLINICS, LITHUANIA 2015-2022 Titas Tiskevicius 1,2, Domas Vaitiekus1,2, Rolandas Gerbutavicius1,2, Milda Rudzianskiene1,2, Ruta Dambrauskiene1,2, Migle Kulboke1,2, Ignas Gaidamavicius1,2, Diana Remeikiene1,2, Birute Sabaniene1, Ieva Stakaitiene1, Egidija Kukarskyte2, Jonas Surkus1,2, Ruta Leksiene1,2, Elona Juozaityte1,2, Dietger Niederwieser3 P552 AUTOLOGOUS PERIPHERAL BLOOD STEM CELL TRANSPLANTATION IN MULTIPLE MYELOMA AT DHARMAIS HOSPITAL – INDONESIAN NATIONAL CANCER CENTER Resti Mulya Sari 1, Dwi Wahyunianto Hadisantoso1, Edel Herbitya1, Lyana Setiawan1, Hubertus Hosti Hayuanta1, Muhammad Raya Kurniawan1, Yanto Ciputra1, Annisa Annisa1, I Gusti Ngurah Agastya1, Della Manik Worowerdi Cintakaweni1 P553 OUTCOMES OF HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM HAPLOIDENTICAL DONORS IN PATIENTS WITH MYELODYSPLASTIC SYNDROMES – A SINGLE-CENTER RETROSPECTIVE ANALYSIS Marketa Stastna Markova 1, Ludmila Novakova1, Mariana Koubova1, Barbora Cemusova1, Veronika Valkova1, Antonin Vitek1, Petr Cetkovsky1, Jan Vydra1 P554 CHALLENGES AND REALITIES OF HEMATOPOIETIC CELL TRANSPLANTATION OF MYELODYSPLASTIC SYNDROMES PATIENTS IN LATIN AMERICA: A SURVEY Fernando Barroso Duarte 1, Rodolfo Daniel de Almeida Soares2, Abrahão Elias Hallack Neto3, Anderson João Simione4, Talyta Ellen de Jesus dos Santos Sousa5, Erika Oliveira de Miranda Coelho6, Vaneuza Araujo Moreira Funke7, Nelson Hamerschlak8, Rodolfo Froes Calixto9, Maria Claudia Rodrigues Moreira10, Alicia Enrico11, Marco Aurelio Salvino12, Eduardo José de Alencar Paton13, Mariana Stevenazzi14, Neysimelia Costa Villela15, Carmem Bonfim7, Gisele Loth16, Breno Moreno Gusmão17, Maria Cristina Martins de Almeida Macedo18, Isabella Araújo Duarte19, Vergílio Antônio Rensi Colturato4 P555 OUTCOMES OF RELAPSE POST HEMATOPOIETIC CELL TRANSPLANTATION IN MYELODYSPLASTIC SYNDROMES FROM THE LATIN AMERICAN REGISTER Fernando Duarte 1, Talyta Ellen de Jesus dos Santos Sousa1, Vaneuza Araújo Moreira Funke2, Nelson Hamerschlak3, Neysimélia Costa Villela4, Maria Cristina Martins de Almeida Macedo5, Afonso Celso Vigorito6, Rodolfo Daniel de Almeida Soares7, Alessandra Paz8, Lilian Diaz9, Mariana Stevenazzi9, Abrahão Elias Hallack Neto10, Gustavo Bettarello11, Breno Moreno Gusmão12, Marco Aurélio Salvino13, Rodolfo Froes Calixto14, Maria Cláudia Rodrigues Moreira15, Gustavo Machado Teixeira16, Cinthya Corrêa Silva3, Eduardo José de Alencar Paton17, Vanderson Rocha18, Alicia Enrico19, Carmem Bonfim20, Ricardo Chiattone21, Anderson João Simioni22, Celso Arrais23, Erika Oliveira de Miranda Coelho24, Vergílio Antônio Rensi Colturato22 P556 OUTCOME OF HEMATOPOIETIC STEM CELL TRANSPLANTATION OF IN CHILDREN WITH A NPM1 MUTATION AND MYELODYSPLASTIC SYNDROME WITH EXCESS BLASTS Ayami Yoshimi 1, Miriam Erlacher1, Peter Noellke1, Senthilkumar Ramamoorthy1, Gudrun Göhring2, Shlomit Barzilai-Birenboim3, Ivana Bodova4, Jochen Buechner5, Albert Catala6, Valérie De Haas7, Barbara De Moerloose8, Michael Dworzak9, Henrik Hasle10, Kirsi Jahnukainen11, Krisztian Kallay12, Marko Kavcic13, Paula Kjollerstrom14, Franco Locatelli15, Riccardo Masetti16, Sophia Polychronopoulou17, Markus Schmugge18, Owen Smith19, Jan Stary20, Dominik Turkiewicz21, Marek Ussowicz22, Marcin Wlodarski23, Christian Thiede24, Brigitte Strahm1, Charlotte Niemeyer1 P557 SYSTEMATIC LITERATURE REVIEW ON THE SAFETY, EFFICACY, AND EFFECTIVENESS OF EATG VERSUS COMPARATORS FOR MDS Erin Sheffels1, Kevin Kallmes1, Keith Kallmes1, Barbara Możejko-Pastewka2, Raj Gokani 2, Judith Hey-Hadavi1, Andres Quintero2 P558 TCRALPHABETA/CD19 DEPLETED ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN COMBINATION WITH POST-TRANSPLANT RUXOLITINIB FOR MYELOFIBROSIS Flores Weverling1, Yousra van der Leest1, Frances Verheij1, Iris Brinkman1, Anna van Rhenen1, Lotte van der Wagen1, Laura Daenen1, Kasper Westinga2, Henk-Jan Prins2, Lisette van de Corput3, Reinier Raymakers1, Anke Janssen1, Jurgen Kuball1, Moniek de Witte 1 P559 MINIMAL RESIDUAL DISEASE MONITORING OF DRIVER MUTATION BY DIGITAL DROPLET PCR IS PREDICTIVE OF RELAPSE AFTER ALLOGENEIC STEM CELL TRANSPLATION IN MYELOFIBROSIS Maria Chiara Finazzi 1,2, Roberta Stavola1, Francesca Valsecchi2, Alessia Civini2, Chiara Pavoni2, Matteo Raviglione2, Anna Grassi2, Alessandra Algarotti2, Maria Caterina Micò2, Federico Lussana1,2, Benedetta Rambaldi2, Gianluca Cavallaro2, Giuliana Rizzuto2, Orietta Spinelli2, Alessandro Rambaldi1,2, Silvia Salmoiraghi2 P560 EVALUATION AND IMPLICATIONS OF PORTAL AND PULMONARY HYPERTENSION IN MYELOFIBROSIS PRIOR TO TRANSPLANT: A PRACTICE-BASED SURVEY OF THE CHRONIC MALIGNANCIES WORKING PARTY OF THE EBMT Giorgia Battipaglia 1, Nicola Polverelli2, Joe Tuffnell3, Patrizia Chiusolo4, Marie Robin5, Massimiliano Gambella6, Annoek Broers7, Elisa Sala8, Jakob Passweg9, Sabine Furst10, Henrik Sengeloev11, Remy Dulery12, Moniek de Witte13, Ibrahim Yakoub-Agha14, Maria Chiara Finazzi15, Claudia Wehr16, Arnon Nagler17, Deborah Richardson18, Wolfgang Bethge19, Andrew Clark20, Joanna Drozd-Sokolowska21, Kavita Raj22, Tomasz Czerw23, Juan Carlos Hernández-Boluda24, Donal P. McLornan22 P561 IMPACT OF NUMBER OF CD34 + CELLS INFUSED ON OVERALL SURVIVAL IN MYELOFIBROSIS PATIENTS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Filippo Frioni 1, Sabrina Giammarco2, Elisabetta Metafuni2, Maria Assunta Limongiello2, Nicola Piccirillo2, John Donald Marra1, Luca Di Marino1, Luciana Teofili2, Andrea Bacigalupo1, Simona Sica2, Patrizia Chiusolo2 P562 AVAPRITINIB IN THE POST-ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT SETTING IN PATIENTS WITH ADVANCED SYSTEMIC MASTOCYTOSIS Deborah Christen 1, Johannes Luebke2, Juliana Schwaab2, Anne Kaiser1, Svetlana Rylova3, Saša Dimitrijević3, Ilda Bidollari3, Jens Peter Panse1, Andreas Reiter2,1 P563 ASCIMINIB: A NEW TKI BEFORE AND AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHRONIC MYELOID LEUKEMIA Iuliia Vlasova 1, Elena Morozova1, Elza Lomaia2, Tamara Chitanava2, Ksenia Tsvirko1, Tatiana Rudakova1, Nikita Volkov1, Tatiana Gindina1, Dmitriy Motorin3, Yulia Alexeeva2, Valeriya Katerina1, Ivan Moiseev1, Alexander Kulagin1 P564 EXCELLENT LONG TERM SURVIVAL DESPITE LOW RATES OF GVHD IN PATIENTS WITH MYELOFIBROSIS FOLLOWING ALLOGENIC HSCT: A SINGLE CENTRE EXPERIENCE Mariam Amer 1, Michael Nathan1, Christopher Dalley1, Kim Orchard1, Sara Main1, Amy Creighton1, Jane Lamb1, Helen Snow1, Annie Major1, Sarah Holtby1, Linda Jarvis1, Mathew Lee1, Muhammad Maqbool1, Hwai Jing Hiew1, Fiona Duggan1, Deborah Richardson1 P565 VENETOCLAX PLUS AZACITIDINE VERSUS AZACITIDINE ALONE AS POST-TRANSPLANT MAINTENANCE TREATMENT IN HIGH-RISK AML AND MDS Yigeng Cao1, Wenwen Guo1, Erlie Jiang 1 P566 CURRENT PRACTICE WHEN TRANSPLANTING PATIENTS WITH MYELOFIBROSIS: A SURVEY ON BEHALF OF THE BRITISH SOCIETY OF BLOOD AND MARROW TRANSPLANTATION AND CELLULAR THERAPY (BSBMTCT) Emma Kempshall 1, Donal McLornan2, Anjum Khan3, Pramilla Krishnamurthy4, Daniele Avenoso4, Sebastian Francis5, Matthew Collin6 P567 LONG-TERM OUTCOME OF ALLOGENEIC TRANSPLANTATION IN PRIMARY AND SECONDARY MYELOFIBROSIS Alzbeta Zavrelova 1,1, Benjamin Visek1, Miriam Lanska1, Pavel Zak1, Petra Lukesova1, Jakub Radocha1 P568 FIRST-IN-HUMAN PHASE I/II CLINICAL TRIAL OF IG-TREGS FOR GVHD PREVENTION Memnon Lysandrou1, Dionysia Kefala1, Panagiota Christofi1,2, Maria Liga1, Antonis Miggos3, Charys Papagregoriou4, Elisavet Vlachonikola3, Nikolaos Savvopoulos1, Vassiliki Zacharioudaki 1, Ioanna Vallianou2, Eleutheria Sagiadinou1, Dimitris Tsokanas1, Rodanthy Theodorellou5, Anastasia Papadopoulou2, Evi Triantafyllou1, Ioanna Sakellari2, Paul Costeas4,6, Anastasia Chatzidimitriou3, Evangelia Yannaki2, Alexandros Spyridonidis1 P569 GILTERITINIB WITH VENETOCLAX, ACTINOMYCIN D, AND LOW-DOSE CYTARABINE AS BRIDGING TO ALLOGENEIC STEM CELL TRANSPLANTATION IN RELAPSED OR REFRACTORY FLT3 MUTATED ACUTE MYELOID LEUKEMIA Andrius Žučenka1,2, Guoda Daukėlaitė1,2, Vilmantė Vaitekėnaitė1,2, Regina Pileckytė1,2, Igoris Trociukas2, Adomas Bukauskas1,2, Rita Čekauskienė 1,2, Laimonas Griškevičius1,2 P570 EFFICACY OF SALVAGE THERAPY INCLUDING NOVEL AGENTS IN ADULT B-CELL ACUTE LYMPHOBLASTIC LEUKAEMIA (B-ALL) PROGRESSING AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: A REAL-WORLD UK STUDY Alexandros Rampotas 1, Fotios Panitsas2, Maximillian Brodermann1, Sridhar Chaganti3, Bouziana Styliani4, Emma Nicholson5, Samanth Drummond6, Sharon Allen7, Andrew King7, Anne-Louise Latif6, Henry Crosland3, Daniele Avenoso4, Claire Roddie1 P571 TREOSULFAN PLUS FLUDARABINE “REDUCED INTENSITY CONDITIONING” WITH POSTTRANSPLANT CYCLOPHOSPHAMIDE IN PATIENTS WITH ACUTE MYELOID LEUKEMIA OLDER THAN 65 YEARS Vincenzo Federico 1,2, Rosella Matera1, Dalila Salvatore3, Filippo Antonio Canale4, Manuela Merla3, Daniela Valente3, Corine Contento1, Giulia Campagna1, Doriana Vaddinelli5, Annalisa Natale5, Davide Seripa1, Stella Santarone5, Tiziana Grassi2, Francesco Bagordo2, Nicola Di Renzo1, Massimo Martino4, Angelo Michele Carella3 P572 ORAL IPTACOPAN MONOTHERAPY MAINTAINS EFFICACY AND SAFETY OVER 48 WEEKS IN COMPLEMENT INHIBITOR-NAÏVE PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH) IN THE PHASE III APPOINT-PNH TRIAL Antonio M. Risitano 1,2, Bing Han3, Yasutaka Ueda4, Jaroslaw P. Maciejewski5, Rong Fu6, Li Zhang7, Austin Kulasekararaj8,9,10, Alexander Röth11, Lee Ping Chew12, Jun Ho Jang13,14, Lily Wong Lee Lee15, Jens Panse16,17, Eng-Soo Yap18, Luana Marano1,2, Flore Sicre de Fontbrune19,20, Chen Yang3, Hui Liu6, Roochi Trikha8,9,10, Navin Mahajan21, Tomasz Lawniczek21, Zhixin Wang22, Christine Thorburn23, Shujie Li22, Marion Dahlke21, Régis Peffault de Latour19,20 P573 T CELLS DIRECTED AGAINST THE METASTATIC DRIVER CHONDROMODULIN-1 IN EWING SARCOMA: COMPARATIVE ENGINEERING WITH CRISPR/CAS9 VS. RETROVIRAL GENE TRANSFER FOR ADOPTIVE TRANSFER Uwe Thiel 1, Kristina von Heyking1, Busheng Xue1, Hendrik Gaßmann1, Melanie Thiede1, Kilian Schober2, Josef Mautner3, Julia Hauer4, Jürgen Ruland5, Dirk H. Busch6, Stefan E.G. Burdach7 P574 UTILITY OF ROMIPLOSTIM IN PAEDIATRIC ALLOGENEIC STEM CELL TRANSPLANT ASSOCIATED POOR GRAFT FUNCTION, IMMUNE MEDIATED CYTOPENIA AND IMMUNE MEDIATED GRAFT FAILURE Srividhya Senthil 1, Abdul Moothedath2, Abbey Forster1, Ramya Hanasoge-Nataraj1, Madeleine Powys1,1, Omima Mustafa1,1, Robert Wynn1,1 P575 RAG2>-/-ΓC-/- MICE HUMANIZED WITH CD34+ HEMATOPOIETIC STEM CELLS ARE A SUITABLE TOOL TO SCREEN FOR FUNCTIONAL EWING SARCOMA-SPECIFIC T CELLS IN VIVO Uwe Thiel 1, Sabine Heim1, Hendrik Gaßmann1, Kristina von Heyking1, Sebastian J. Schober1, Melanie Thiede1, Markus Niemeyer2, Dirk Busch3, Ropert Oostendorp4, Irene Esposito5, Julia Hauer1, Günther HS Richter6 P576 REAL-WORLD EFFECTIVENES OF VENETOCLAX COMBINED WITH IPOMETILATING-AGENTS AS A BRIDGE TO ALLOGENEIC TRANSPLANTATION IN ACUTE MIELOID LEUKEMIA: RETROSPECTIVE AND MULTICENTRIC STUDY OF APULIAN HEMATOLOGICAL NETWORK “REP05” Vincenzo Federico 1,2, Rosella Matera1, Domenico Pastore3, Giuseppe Tarantini4, Lorella Melillo5, Angelo Michele Carella6, Attilio Guarini7, Caterina Buquicchio4, Lucia Ciuffreda5, Mariachiara Abbenante6, Paola Carluccio8, Crescenza Pasciolla7, Mariapaolo Fina1, Alessandro Spina3, Marina Urbano3, Daniela Carlino1, Mariangela Lecciso1, Michelina Dargenio1, Davide Seripa1, Pellegrino Musto8,9, Giorgina Specchia10, Nicola Di Renzo1 P577 T CELL RECEPTOR IDENTIFICATION AGAINST EWING SARCOMA ASSOCIATED DICKKOPF 2 (DKK2) DERIVED ANTIGEN USING SINGLE-CELL RNA SEQUENCING Sophia Laube 1, Kristina von Heyking1, Melanie Thiede1, Hendrik Gassmann1, Sebastian Johannes Schober1, Jennifer Eck1, Hannah Hüls1, Carolin Prexler1, Sabrina Wagner2, Elvira D’Ippolito2, Julia Hauer1, Stefan E G. Burdach3, Dirk H. Busch2, Uwe Thiel1 P578 TARGETED CD8+ T CELL THERAPY IN ADVANCED EWING SARCOMA: EXPLORING LIPI, GPR64, PAX7, AND CHM1 AS TARGETS FOR ADOPTIVE TRANSFER APPROACHES Hannah Hüls 1, Kristina von Heyking1, Melanie Thiede1, Hendrik Gassmann1, Sebastian Johannes Schober1, Jennifer Eck1, Sophia Laube1, Carolin Prexler1, Sabrina Wagner2, Elvira D’Ippolito2, Julia Hauer1, Stefan E G. Burdach3, Dirk H. Busch2, Uwe Thiel1 P579 COMPARATIVE EFFECTIVENESS OF IPTACOPAN VERSUS C5 INHIBITORS IN COMPLEMENT INHIBITOR-NAÏVE PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA FROM THE PHASE III APPOINT-PNH TRIAL AND REAL-WORLD APPEX COHORT Matthew Holt 1, Richard J. Kelly1, Jilles M. Fermont2, Georgina Bermann2, Ariel Chernofsky2, Jens Haenig2, Marion Dahlke2, Régis Peffault de Latour3,4 P580 TREATMENT WITH BLAST-MODULATORY RESPONSE MODIFIERS INDUCED SPECIFIC IMMUNE RESPONSES AND LED TO DISEASE STABILIZATION IN A PATIENT WITH REFRACTORY AML AFTER SECOND ALLOGENEIC SCT Philipp Anand1, Giuliano Fillipini Velazquez 2, Joudi Abdulmajid1, Xiaojia Feng1, Klaus Hirschbühl2, Anwesha Sinha1, Helga Schmetzer1, Christoph Schmid2 P581 REDUCED PRESENCE OF MESENCHYMAL STEM CELLS IN BONE MARROW ASPIRATES OF FEMALE PATIENTS PERSISTS AFTER ALLOGENEIC STEM CELL TRANSPLANTATION Judith Schaffrath 1, Ole Vollstädt2, Jana Lützkendorf1, Cornelia Baum1, Sabine Edemir1, Kirstin Lauer1, Lutz P. Müller1 P582 EFFICACY AND SAFETY OF A CORD BLOOD-DERIVED PLATELET CONCENTRATE FOR THE TREATMENT OF OCULAR SURFACE DISEASES: RESULTS FROM A SINGLE-CENTER PROSPECTIVE STUDY Camilla Delponte1, Laura Mazzucco2, Maria Rosa Astori3, Lucia Brunello3, Sara Butera3, Paolo Rivela3, Valeria Balbo2, Davide Dealberti4, Riccardo Dondolin5, Monia Lunghi5, Francesca Pollis2, Marco Ladetto3, Francesco Zallio 3 P583 IMPACT OF DIMENSIONALITY AND EXTRACELLULAR MATRIX ON MESENCHYMAL STEM CELL PHENOTYPE Jeong Jun Kim1, Ji Hye Lee1, Hee Hoon Yoon1, Jun Eun Park 2 P584 PROTECTING HLA-A AND HLA-B ANTIGENS THROUGH EPLETS ANALYSIS IN THE SETTING OF PLATELET TRANSFUSIONS IN HAPLOIDENTICAL HEMATOPOIETIC STEM CELL ALLOGRAFT CANDIDATES Magalie Joris1, Amandine Charbonnier1, Delphine Lebon1, Judith Desoutter1, Etienne Paubelle1, Nicolas Guillaume 1 P585 CLINICAL SIGNIFICANCE OF CYTOKINE RELEASE SYNDROME FOLLOWING HLA-MISMATCHED HEMATOPOIETIC STEM CELL TRANSPLANTATION: A RETROSPECTIVE MONOCENTRIC STUDY ON 250 TRANSPLANTS Filippo Frioni 1, Eugenio Galli2, Sabrina Giammarco2, Elisabetta Metafuni2, Federica Sorà2, Maria Assunta Limongiello2, Roberto Maggi1, John Donald Marra1, Luca Di Marino1, Andrea Mattozzi1, Andrea Bacigalupo1, Simona Sica2, Patrizia Chiusolo2 P586 INVESTIGATING VASCULAR ENDOTHELIAL DYSFUNCTION BY SYNDECAN-1 FOLLOWING PEDIATRIC HSCT Sarah Weischendorff 1, Denise Elbæk Horan1, Katrine Kielsen1, Maria Ebbesen Sørum1, Marianne Ifversen1, Pär Ingemar Johansson1, Klaus Mûller1,2 P587 ASSESSING THE IMPACT OF FLUID OVERLOAD ON OUTCOMES AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Ana Belen Bocanegra 1, David Jaimovich2, Irene Solano1, Javier Martinez-Costa1, Jorge Verdejo1, Maria Valdenebro1, Carlos De Miguel1, Guiomar Bautista1, Sangeeta Hingorani3, Rafael Francisco Duarte1 P588 BEYOND PAIN MANAGEMENT: ASSESSING THE NEGATIVE IMPACT OF INTENSE OPIOID USE ON ALLOHCT SURVIVAL Tommy Alfaro Moya 1, Igor Novitzky Basso1, Shiyi Chen1, Mats Remberger2, Refik Saskin3, Jonas Mattsson1 P589 VENO-OCCLUSIVE DISEASE/SINUSOIDAL OBSTRUCTION SYNDROME (VOD/SOS)-ASSOCIATED MORBIDITY IN CHILDREN POST HEMATOPOIETIC STEM CELL TRANSPLANTATION - A HINT FOR AN ADEQUATE PROPHYLAXIS? Zofia Szmit 1, Zuzanna Gamrot1, Anna Król1, Jowita Frączkiewicz1, Monika Mielcarek-Siedziuk1, Karolina Liszka1, Igor Olejnik1, Tomasz Jarmoliński1, Selim Corbacioglu2, Krzysztof Kałwak1 P590 OUTCOME AND COMPLEMENT SYSTEM INVOLVEMENT IN PATIENTS WITH THROMBOTIC MICROANGIOPATHY AFTER ALLOGENEIC STEM CELL TRANSPLANTATION Jacopo Mariotti 1, Massimo Cugno2, Luigi Porcaro3, Daniela Taurino1, Barbara Sarina1, Chiara de philippis1, Daniele Mannina1, Armando Santoro1, Gianluigi Ardissino4, Stefania Bramanti1 P591 BONE REMODELLING ARE SIGNIFICANTLY IMPAIRED AFTER PEDIATRIC HSCT AND INFLUENCED BY DIAGNOSIS, CONDITIONING REGIMEN AND ACUTE GRAFT-VERSUS-HOST DISEASE Katrine Kielsen 1, Kathrine Fogelstrøm1, Anne Nissen1, Marianne Ifversen1, Bolette Hartmann2, Klaus Müller1,2 P592 IDIOPATHIC UPPER GASTROINTESTINAL BLEEDING AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: AN UNPRECEDENTED COMPLICATION Juan Eirís 1, Marina Pérez-Bravo2, Nuria Rausell1, Carla Satorres1, Marta Villalba1, Pedro Chorão1, Juan Montoro1,3, Pedro Asensi1, Pablo Granados1, David Martínez-Campuzano1, Alberto Louro1, Marta Henriques4, Ana Facal5, Mª Consejo Ortí-Verdet6, Miguel Sanz1, Javier de la Rubia1,3, Jaime Sanz1, Aitana Balaguer-Roselló1 P593 ACUTE KIDNEY INJURY AND CRONIC KIDNEY DISEASE AFTER ALLOGENEIC HCT. A SINGLE-CENTER EXPERIENCE Ana Belén Bocanegra 1, Jaimovich David2, Irene Solano3, Javier Martinez-Costa3, Jorge Verdejo3, Maria Valdenebro3, Ana Muñoz Sanchez3, Carlos De Miguel3, Guiomar Bautista3, Sangeeta Hingorani4, Rafael Francisco Duarte3 P594 SINUSOIDAL OBSTRUCTION SYNDROME/VENO-OCCLUSIVE DISEASE: A RETROSPECTIVE STUDY OF THE SPANISH HEMATOLOGY ASSOCIATION-HEMATOPOIETIC STEM CELL TRANSPLANTATION GROUP (GETH) Cristina Blázquez Goñi 1, Francisco Manuel Martin Dominguez1, Silvia García Canale1, Cristina Díaz de Heredia2, Melissa Panesso Romero2, Maribel Benítez Carabante2, María Luz Uría2, David Bueno3, Annalisa Paviglianiti4, Pilar Palomo Moraleda5, Ana Isabel Gallardo Morillo6, Antonia Pascual Martínez6, Marina Acera Gómez7, Alexandra Regueiro8, Carlos Vallejo Llamas9, Gillen Oarbeascoa10, Diana Campos11, Leslie González Pinedo12, Melissa Torres12, Julia Marsal Ricomà13, Marta González Vicent14, María José Jiménez Lorenzo15, Estefanía García Torres16, Mónica López Duarte17, Andrés Sánchez Salinas18, Pedro González Sierra19, José Luis López Lorenzo20, Amaya Zabalza21, Sara Redondo Velao22, Oriana López-Godino23, Alejandro Luna de Abia24, Cristina Beléndez10, Beatriz Aguado25, María Teresa Artola Urain26, Patricia Jiménez Guerrero27, Agustín Nieto Vázquez28, Francisca Almagro Torres29, José Antonio Sanz30, Beatriz de Rueda31, Rocío Picón González32, José Antonio Pérez Simón1 P595 SAFETY AND BENEFIT OF LIVER BIOPSY AFTER ALLOGENIC STEM CELL TRANSPLANTATION: A RETROSPECTIVE SINGLE-CENTER STUDY Marie Maulini 1, Aurélie Bornand1, Federico Simonetta1, Anne-Claire Mamez1, Chiara Bernardi1, Federica Giannotti1, Yan Beauverd1, Thomas Longval1, Maria Mappoura1, Sarah Noetzlin1, Laetitia Dubouchet1, Sarah Perdikis-Prati1, Evgenia Laspa1, Cuong-An Do1, Thien-An Tran1, Carmen De Ramon Ortiz1, Amandine Pradier1, Sarah Morin1, Stavroula Masouridi-Levrat1, Simon Maulini2, Laura Rubbia-Brandt1, Laurent Spahr1, Yves Chalandon1 P596 INVESTIGATION OF LABORATORY PARAMETERS BEFORE THE ONSET OF HEPATIC SINUSOIDAL OBSTRUCTION SYNDROME IN CHILDREN AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION Bernd Gruhn 1, Lorena Johann1 P597 OUTCOME OF ALLOGENEIC HCT RECIPIENTS COMPARED TO OTHER PATIENTS WITH HEMATOLOGICAL MALIGNANCIES ADMITTED TO THE INTENSIVE CARE UNIT. A SINGLE CENTRE STUDY Ana Belen Bocanegra 1, Marta Perez Calle1, Jorge Verdejo1, Alicia Segura1, Ana María Bellón1, Rosalia Alonso1, Ana Amaro Harpigny1, Patricia Enciso1, Inmaculada Tendero1, Maria Esther Martinez-Muñoz1, Carlos De Miguel1, Guiomar Bautista1, Ines Lipperheide1, Daniel Ballesteros1, Rafael Francisco Duarte1 P598 ADVERSE PROGNOSTIC IMPACT OF PRETRANSPLANT HEPCIDIN AND FERRITIN ON OUTCOMES FOLLOWING HEMATOPOIETIC STEM CELL TRANSPLANTATION Michelle Pirotte 1, Marianne Fillet2, Laurence Seidel3, Evelyne Willems1, Sophie Servais1, Frédéric Baron1, Yves Beguin1 P599 IMPACT OF GRANULOCYTE-COLONY STIMULATING FACTOR (G-CSF) ON CLINICAL OUTCOMES IN ALLOGENEIC STEM CELL TRANSPLANTATION: INSIGHTS FROM A SINGLE-CENTER EXPERIENCE AT PRINCESS MARGARET CANCER CENTRE Ahmed Alnughmush1,2, Ayman Sayyed1,2, Mohammed Kawari 2, Mats Remberger3, Carol Chen1, Caden Chiarello1, Ivan Pasic1,2, Igor Novitzky-Basso1,2, Arjun Datt Law1,2, Wilson Lam1,2, Dennis (Dong Hwan) Kim1,2, Fotios V. Michelis1,2, Armin Gerbitz1,2, Auro Viswabandya1,2, Rajat Kumar1,2, Jonas Mattsson1,2,4 P600 VALIDATION OF EBMT 2023 VOD CRITERIA: IMPACT ON EARLY DIAGNOSIS AND SENSITIVITY/ESPECIFICITY OF PROBABLE VOD CATEGORY Monica Cabrero 1,2, Maria Cortes-Rodriguez3,2, Almudena Cabero1,2, Estefania Perez-Lopez1,2, Ana Africa Martin-Lopez1,2, Monica Baile1,2, Alejandro Avendaño1,2, Ana Garcia-Bacelar1,2, Lorena Hernandez-Medina2, Marina Acera2, Lourdes Vazquez1,2, Fermin Sanchez-Guijo1,2, Lucia Lopez-Corral1,2 P601 EFFICACY AND SAFETY OF NETUPITANT/PALONOSETRON COMBINATION (NEPA) AND LOW DOSE OF DEXAMETHASONE IN PREVENTING NAUSEA AND VOMITING IN PATIENTS UNDERGOING ALLOGENEIC STEM CELL TRANSPLANTATION Alessandro Spina 1, Marina Aurora Urbano1, Claudia Schifone1, Francesca Merchionne1, Maria Laura Di Noi1, Gianluca Guaragna1, Erminia Rinaldi1, Giovanni Quintana1, Giuseppe Mele1, Domenico Pastore1 P602 CYTOKINE RELEASE SYNDROME AFTER HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION AND ITS IMPACT ON OVERALL SURVIVAL Isabella Silva Pimentel Pittol 1, Camila Carminatti Isoppo2, André Dias Américo1, Germano Glauber de Medeiros Lima1, Eurides Leite da Rosa3, João Antônio Gonçalves Garreta Prats1, Hegta Tainá Rodrigues Figueroa1, Juliana Matos Pessoa1, Phillip Scheinberg1, Fauze Lutfe Ayoub1, Fabio Rodrigues Kerbauy1 P603 EXPLORING THE SAFETY PROFILE OF NORETHINDRONE IN POST-MENARCHAL STEM CELL TRANSPLANT PATIENTS: ADDRESSING VOD CONCERNS AND CAUTIONARY CONSIDERATIONS Mahvish Rahim1, Jodi Skiles1, Devin Dinora1, Jessica Harrison1, Ryanne Green1, Allie Carter1, April Rahrig 1 P604 POST-TRANSPLANT DE NOVO ANTI-HLA DONOR SPECIFIC ANTIBODIES MAY CONTRIBUTE TO POOR GRAFT FUNCTION AFTER HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION Xinyu Ji 1, Luxin Yang1,2, Xiaoyu Lai1,2, Yishan Ye1,2, Yibo Wu1,2, Shipei Xiang1, Yi Luo1,2, Lizhen Liu1,2 P605 IMPACT OF EARLY CYCLOSPORINE INITIATION ON CYTOKINE RELEASE SYNDROME AND TRANSPLANT OUTCOMES IN PEDIATRIC T CELL REPLETE PERIPHERAL BLOOD HAPLO-IDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANT Veerendra Patil 1, Pavan Kumar Boyella1, Pallavi Ladda1, Rakesh Pinninti1, Rohan Tewani1, Senthil Rajappa1 P606 USE OF GRANULOCYTE COLONY-STIMULATING FACTORS IS FEASIBLE IN ALL DONOR SETTING WITHOUT INCREASING THE RISK OF ENGRAFTMENT SYNDROME Maria Luisa Giannattasio 1, Linda Piccolo1, Andrea Cacace1, Davide Pio Abagnale1, Giuseppe Gaeta1, Francesco Grimaldi1, Simona Avilia1, Mara Memoli1, Lucia Ammirati2, Valentina Maglione2, Catello Califano2, Claudia Andretta3, Ermanno Badi3, Pasqualino Correale3, Patrizia Ricci4, Domenica Borzacchiello4, Marco Picardi1, Antonio Feliciello4, Antonio Leonardi3, Fabrizio Pane1, Giorgia Battipaglia1 P607 THE UTILITY OF EXTRACORPOREAL ULTRASONOGRAPHY SCORE (HOKUS-10) IN PEDIATRIC SOS/VOD Atsushi Narita 1, Hideki Muramatsu1, Ryo Maemura1, Daiki Yamashita1, Daichi Sajiki1, Yusuke Tsumura1, Ayako Yamamori1, Manabu Wakamatsu1, Kotaro Narita1, Shinsuke Kataoka1, Yoshiyuki Takahashi1 P608 RETROSPECTIVE REVIEW OF TRANSPLANT AND TRANSFUSION OUTCOMES IN PATIENTS WITH PRE-TRANSPLANT RED CELL ALLOANTIBODIES Samantha Drummond 1, Jennifer Laird2, David Irvine3 P609 TWO GLOBAL PHASE 3 TRIALS OF THE EFFICACY AND SAFETY OF RAVULIZUMAB IN ADULT AND PEDIATRIC PATIENTS WITH THROMBOTIC MICROANGIOPATHY AFTER HEMATOPOIETIC STEM CELL TRANSPLANT Carolyn Hahn 1, Elsa Konig1, Jonathan Monteleone1, Edward Wang1 P610 RITUXIMAB, BORTEZOMIB, PLASMA EXCHANGE COMBINED WITH IVIG FOR DESENSITIZATION DURING HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN PATIENTS WITH A POSITIVE DONOR-SPECIFIC ANTI-HLA ANTIBODY Fang Liu 1, Cunbang Wang1, Yecheng Li1, Min Chen1, Jialin Duan1, Xinyu Wei1, Yongli Li1, Jing Wu1, Xiaofei Shen1, Ying Zhou1, Jinwei Li1, Yaoling Fu1, Lingling Yu1 P611 PREDICTIVE FACTORS OF GRAFT FAILURE IN ADULT AND PAEDIATRIC PATIENTS WITH MALIGNANT AND NON-MALIGNANT DISEASE UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANT (HSCT) Pietro Merli 1, Régis Peffault de Latour2,3, Emmanuel Monnet4, Malin Löfqvist4, Franco Locatelli1 P612 EARLY LOW DOSE CYCLOSPORINE AND MYCOPHENOLATE REDUCE THE RISK OF CYTOKINE RELEASE SYNDROME AFTER HLA-HAPLOIDENTICAL PERIPHERAL BLOOD STEM CELL TRANSPLANTATION WITH PTCY BASED GVHD PROPHYLAXIS Jan Vydra 1, Ludmila Nováková1, Veronika Válková1, Markéta Šťastná Marková1, Mariana Koubová1, Anna Dobrovolná1, Barbora Čemusová1, Antonín Vítek1, Petr Cetkovský1 P613 INCIDENCE, RISK FACTORS, AND OUTCOMES OF TRANSPLANT-ASSOCIATED THROMBOTIC MICROANGIOPATHY IN PEDIATRIC PATIENTS AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION Kyung-Nam Koh 1, Su Hyun Yun1, Eun Seok Choi1, Sung Han Kang1, Hyery Kim1, Ho Joon Im1 P614 STEM CELL MOBILIZATION EFFECTS ON HEART SIZES AND FUNCTION Domas Vaitiekus 1, Ignas Gaidamavicius1, Audrone Vaitiekiene1, Migle Kulboke1, Monika Bieseviciene1, Benas Kireilis1, Ruta Dambrauskiene1, Milda Rudzianskiene1, Antanas Jankauskas1, Elona Juozaityte1, Dietger Niederwieser1, Jolanta Justina Vaskelyte1, Gintare Sakalyte1, Rolandas Gerbutavicius1 P615 EASIX SCORE AS INDEPENDENT PREDICTOR FOR OVERALL SURVIVAL IN PATIENTS WITH ACUTE LEUKEMIA AND MDS UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION Penka Ganeva1, Andriyana Bankova 1, Georgi Vasilev1, Krasen Venkov1, Kameliya Milcheva1, Victoria Yankova1, Margarita Guenova1, Georgi Mihaylov1 P616 EARLY CICLOSPORINE TO EVEROLIMUS CONVERSION POST HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR PATIENT WITH CALCINEURIN INHIBITORS INDUCED TOXICITIES Benjamin Bouchacourt 1, Raynier Devillier1,2, Didier Blaise1,2, Thomas Pagliardini1, Sabine Furst1, Samia Harbi1, Faezeh Legrand1, Charlotte Nykolyszyn1, Federico Pagnussat1, Pierre Jean Weiller1, Claude Lemarie1,3, Boris Calmels1,3, Christian Chabannon1,3,2 P617 IMMUNE-MEDIATED CYTOPENIA IN PEDIATRIC PATIENTS FOLLOWING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: ON BEHALF OF THE RBC DISORDER WORKING PARTY OF THE KOREAN SOCIETY OF HEMATOLOGY Hyoung Soo Choi 1, Jeong-A Park2, Hee Won Chueh3, Hee-Jo Baek4, Hoon Kook4 P618 ENGRAFTMENT SYNDROME IN PATIENTS UNDERGOING AUTOLOGOUS STEM CELL TRANSPLANTATION FOR MULTIPLE MYELOMA: A SINGLE-CENTER DATA ANALYSIS OF CLINICAL FEATURES AND RISK FACTORS Shanshan Liu 1, Luxin Yang2, Lizhen Liu2, Yan Gao1, Xianqi Feng1, He Huang2, Yi Luo2 P619 DONOR SPECIFIC ANTI-HLA ANTIBODIES IN HAPLOIDENTICAL PERIPHERAL STEM CELL TRANSPLANTATION: SINGLE CENTER EXPERIENCE Alessandro Spina 1, Marina Aurora Urbano1, Claudia Schifone1, Maria Laura Di Noi1, Francesca Merchionne1, Maria Rosaria Coppi1, Maria Patrizia D’Errico1, Maria Antonietta Miccoli2, Domenico Pastore1 P620 EARLY ONSET OF EPSTEIN-BARR VIRUS POSITIVE MULTIPLE MYELOMA TYPE OF POSTTRANSPLANT LYMPHOPROLIFERATIVE DISORDER – A CASE OF INFREQUENT COMPLICATION AFTER ALLOGENIC PERIPHERAL BLOOD STEAM CELL TRANSPLANTATION Kamila Kruczkowska-Tarantowicz1, Piotr Rzepecki1 P621 THE ROLE OF DARATUMUMAB IN POST-TRANSPLANT COMPLICATIONS: A PROSPECTIVE STUDY ON PRCA AND AIHA Sabrina Giammarco 1, Maria Assunta Limongiello1, Luca Di Marino2, Elisabetta Metafuni1, Maggi Roberto2, Federica Sorá2, Eugenio Galli1, Patrizia Chiusolo2, Simona Sica2 P622 TRANSPLANT ASSOCIATED THROMBOTIC MICROANGIOPATHY IS AN IMPORTANT CAUSE OF HIGH EARLY MORTALITY IN ALLOGENIC BONE MARROW TRANSPLANT Pawan Kumar Singh 1, Isha Gambhir1, Ravi Shanker1, Rasika Setia1, Anil Handoo1 P623 IMMUNE DYSREGULATION AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION BEYOND GVHD: A COMPREHENSIVE PEDIATRIC CASE REPORT - FROM DIAGNOSTICS TO TAILORED BTK INHIBITION BY IBRUTINIB Peter Švec 1, Maria Füssiová1, Jaroslava Adamčáková1, Ivana Boďová1, Júlia Horáková1, Dominika Dóczyová1, Miroslava Pozdechová1, Tomáš Sýkora1, Alexandra Kolenova1 P624 A RARE CASE OF MULTIPLY RELAPSED POST-ALLOGENEIC STEM CELL TRANSPLANT IMMUNE THROMBOTIC THROMBOCYTOPENIC PURPURA IN A YOUNG PATIENT WITH GATA-2 POSITIVE MYELODYSPLASTIC SYNDROME Angela Dăscălescu 1, Antohe Ion2, Elena Dolachi3, Roxana Dumitru4 P625 CENTRAL NERVOUS SYSTEM TOXICITY WITH EXCLUSIVE INVOLVEMENT OF THE BRAIN WHITE MATTER LIKELY RELATED TO TOXIC/IMMUNOLOGIC CAUSES. A PRESENTATION OF 4 SIMILAR CASES Valentina Sangiorgio1, Elena Agostani 1, Marilena Fedele2, Paola Perfetti2, Cristina Capraro2, Elisabetta Terruzzi2, Andrea Aroldi2, Carlo Gambacorti Passerini1, Matteo Parma2 P626 EVALUATING AN UPDATED ANTIEMETIC PROTOCOL IN HAEMATOPOIETIC STEM CELL TRANSPLANTATION Katie Robertson1, Muhammad Saif 1, Ann Griffiths1, Thomas Sanders1, Daniel Monnery1 P627 ECULIZUMAB TREATMENT OF HEMATOPOIETIC STEM CELL TRANSPLANTATION-ASSOCIATED THROMBOTIC MICROANGIOPATHY: SINGLE CENTER EXPERIENCE Tatiana Rudakova1, Julia Vlasova1, Olesya Paina1, Olga Slesarchuk1, Marina Gorodnova1, Tatyana Schegoleva1, Oleg Goloshchapov1, Tatyana Bykova 1, Elena Morozova1, Lyudmila Zubarovskaya1, Ivan Moiseev1, Alexander Kulagin1 P628 IMMUNE THROMBOCYTOPENIA FOLLOWING ALLOGENEIC STEM CELL TRANSPLANT : RESULTS FROM A FRENCH MULTICENTRIC RETROSPECTIVE STUDY OF 35 CASES Antoine Gondé 1,2, Aude Legal3, Flore Sicre de Fontbrune4, Mathieu Puyade5, Maud d’Aveni6, Yves Béguin7, Carmen Botella Garcia8, Éolia Brissot9, Étienne Daguindau10, Bertrand Godeau1, Marc Michel1, Thibault Comont3 P629 AT-HOME AUTOLOGOUS HEMATOPOIETIC CELL TRANSPLANT FOR ADULTS WITH HEMATOLOGICAL MALIGNANCIES. HOW FRAILTY SYNDROME IMPACTS AND EVOLVES DURING HCT PROCEDURE Juan Ortiz 1, Maria Teresa Solano2, Cristina Galleago2, Nuria Ballestar2, Anna Serrahima2, Noemi de llobet2, Raquel Salinas2, Alexandra Patricia Martinez2, María Suárez-Lledó2, Beatriz Merchán2, Paola Charry2, Joan Cid2, Miquel Lozano2, Laura Rosiñol2, Carmen Martinez2, Montserrat Rovira2, Enric Carreras2, Francesc Fernandez2 P630 FRAILTY ASSESSMENT IN ADULTS UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION OUTCOMES. PROSPECTIVE STUDY ON BEHALF OF THE GRUPO ESPAÑOL DE TRASPLANTE HEMATOPOYÉTICO Y TERAPIA CELULAR (GETH-TC) Maria Queralt Salas 1,2, Maria Teresa Solano1,2, Mónica Baile-González3,2, Marina Acera-Gómez3,2, Laura Fox4,2, Maria del Mar Pérez-Artigas4,2, Ana Santamaría5,2, María del Carmen Quintela-González5,2, Andrés Sánchez-Salinas6,2, Joaquina M. Salmerón-Camacho6,2, Verónica Illana-Álvaro7,2, Zahra Abdallahi-Lefdil7,2, Javier Cornago-Navascues78,2, Laura Pardo8,2, Sara Fernández-Luis9,2, Leddy Patricia Vega-Suárez9,2, Sara Villar10,2, Patricia Beorlegui-Murillo10,2, Albert Esquirol11,2, Isabel Izquierdo-García12,2, Sonia Rodríguez González13,2, Alberto Mussetti13,2, Esperanza Lavilla14,2, Javier López-Marin15,2, Ángel Cedillo2, Leyre Bento16,2, Anna Sureda13,2 P631 LATE EFFECTS AFTER PEDIATRIC ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Raquel García Ruiz1, Sara Fernández-Luis1, Miriam Sánchez Escamilla1, María Terán Díaz1, Juan Manuel Cerezo Martín1, Juan José Dominguéz-García1, Arancha Bermúdez Rodríguez1, Enrique M. Ocio San Miguel1, Mónica López Duarte 1 P632 OUTCOMES AND RISK FACTORS FOR SURVIVAL OF LUNG TRANSPLANTATION AFTER PRIOR ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION: A STUDY FROM THE EBMT TRANSPLANT COMPLICATIONS WORKING PARTY Saskia Bos1, Juan Montoro 2, Christophe Peczynski3, Pascale Ambron3, Manuela Badoglio3, Robin Vos4, Tobias Gedde-Dahl5, Mahmoud Aljurf6, Werner Rabitsch7, David Ma8, Harshita Rajasekariah8, Jaime Sanz2, Johannes Schetelig9, Anna Paola Iori10, Christian Koenecke11, Mar Bellido12, Juergen Finke13, Montserrat Rovira14, Angela Figuera15, Rabah Redjoul16, Ivan Moiseev17, Olaf Penack18, Hélène Schoemans19, Zina Peric20 P633 DEPRESSION IN SURVIVORS OF HEMATOPOIETIC STEM CELL TRANSPLANTATION COMPARED WITH A MATCHED GENERAL POPULATION COMPARISON SAMPLE: THE MOSA STUDY Bianca Wauben 1, M.W.M. van der Poel1, M. van Greevenbroek2, S. Koehler2, H.C. Schouten1, N. van Yperen2 P634 PREGNANCY OUTCOMES IN WOMEN FOLLOWING HAEMATOPOIETIC STEM CELL TRANSPLANT FOR HAEMATOLOGICAL CONDITIONS Michele Robinson1, Melanie Davies1, Anna L. David1, Panagiotis Kottaridis 1 P635 SAFETY AND EFFICACY OF IMMUNE CHECKPOINT INHIBITORS FOR SOLID ORGAN MALIGNANCIES AFTER ALLOGENEIC HSCT: A RETROSPECTIVE, MULTICENTRIC STUDY FROM THE EBMT TRANSPLANT COMPLICATIONS WORKING PARTY Jan Brijs 1, Christophe Peczynski2,3, William Boreland2,3, Angela Cuoghi4, Johan Maertens1, Mohamad Mohty5, Nicolaus Kröger6, Philipp Nakov7, Annoek E.C. Broers8, Matthias Eder9, Concepcion Herrera Arroyo10, Martin Kaufmann11, Ron Ram12, Michel Schaap13, Olaf Penack3,14, Christian Könecke3,9, Ivan Moiseev3,15, Helene Schoemans1,3,16, Zinaida Peric3,17 P636 POST-TRANSPLANT CYCLOPHOSPHAMIDE IS ASSOCIATED WITH REDUCED RATE OF SECONDARY MALIGNANCIES POST ALLOGENEIC STEM CELL TRANSPLANT Nihar Desai 1, Mariana Pinto Pereira1, Mats Remberger2, Rajat Kumar1, Dennis Kim1, Auro Viswabandya1, Arjun Law1, Wilson Lam1, Ivan Pasic1, Armin Gerbitz1, Igor Novitzky-Basso1, Jonas Mattsson1, Fotios Michelis1 P637 MOBILE HEALTH IN THE MANAGEMENT OF ALLOGENIC STEM CELL TRANSPLANT RECIPIENTS (MY-MEDULA STUDY): PRELIMINARY RESULTS OF THE CLINICAL TRIAL Sara Redondo 1, Anna De Dios1, Marina Vallve1, Jordi Real1, Merce Triquell1, Olga Aso1, Albert Esquirol1, Me Moreno-Martinez1, Mireia Riba1, Julia Ruiz1, Eva Tobajas1, Jorge Sierra1, Javier Briones1, Rodrigo Martino1, Mar Gomis-Pastor1, Irene Garcia-Cadenas1 P638 HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR PATIENTS FROM AREAS OF CONFLICT: THE KING HUSSEIN CANCER CENTER TRANSPLANTATION PROGRAM EXPERIENCE Salwa Saadeh 1,2, Haya Hamarsha1, Razan Odeh3, Mohammad Makoseh1,2, Zaid Abdel Rahamn1 P639 AUTOIMMUNE NEUTROPENIA AFTER ALLOGENIC BONE MARROW TRANSPLANTATION DUE TO ANTIBODIES SPECIFICALLY TARGETING THE HNA-1A ANTIGEN IN PEDIATRIC PATIENTS Maria Luz Uria Oficialdegui 1, Enric Casanovas2, Carme Canals2, Nuria Nogues2, Marta Rodriguez2, Cecilia Gonzalez2, Melissa Panesso1, Laura Alonso1, Lucia Garcia3, Monica Linares2, M. Isabel Benitez-Carabante1, Cristina Diaz-de-Heredia1 P640 IONA - INTERDISCIPLINARY ONCOLOGICAL FOLLOW-UP CLINIC IN VIENNA Alexandra Böhm 1, Sabine Burger1, Anna Pucher2, Sabina Ziomek2, Arno Hraschan2, Sophie Lindermann2, Barbara Schröder-Aranyosy2, Felix Keil1 P641 RAISING FOLLOW-UP AFTER CELL THERAPY TO THE NEXT LEVEL: A TRANS-REGIONAL, TRANS-SECTORAL, INTERDISCIPLINARY RESEARCH PROJECT Katharina Egger-Heidrich 1, Martin Schneider1, Gabriele Müller1, Roman Schmädig1, Franziska Schmidt1, Lynn Leppla2,3, Sabina De Geest3, Alexandra Teynor4, Markus Wolfien1,5, Martin Sedlmayr1,5, Mathias Hänel6, Anke Morgner6, Vladan Vucinic7, Jochen Schmitt1, Johannes Schetelig1, Uwe Platzbecker7, Martin Bornhäuser1, Jan Moritz Middeke1 P642 SEVERE POLYAUTOIMMUNITY AFTER ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: POTENTIAL ROLE OF THE GENETIC BACKGROUND AND CHIMERISM LOSS Giorgio Costagliola1, Alessandro Di Gangi1, Eva Parolo 1, Chiara Lardone1, Sayla Bernasconi1, Sofia D’Elios1, Nina Tyutyusheva1, Gabriella Casazza1, Mariacristina Menconi1 P643 A CASE REPORT OF A REFRACTORY EVANS SYNDROME AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION Viktoria Yankova 1, Krasen Venkov1, Andriyana Bankova1, Kameliya Milcheva1, Penka Ganeva1, Jonka Lazarova1, Georgi Mihaylov1, Galya Kondeva1 P644 PLASMA REG3Α AND CITRULLINE LEVELS PREDICT GASTROINTESTINAL AGVHD BEFORE CLINICAL ONSET IN PEDIATRIC HSCT Nakisa Kamari-Kany1, Sarah Weischendorff 1,2, Christian Enevold1, Marianne Ifversen1, Katrine Kielsen1, Klaus Müller1,2 P645 THE OUTCOME OF HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS WITH CHEMOREFRACTORY ACUTE MYELOID LEUKEMIA Maria Ilushina1, Larisa Shelikhova1, Daria Shasheleva1, Yuliya Skvortsova1, Sergey Blagov1, Dmitriy Balashov1, Galina Novichkova1, Alexei Maschan 1, Michael Maschan1 P646 OUTCOMES AFTER HEMATOPOIETIC CELL TRANSPLANTATION FOR HURLER SYNDROME AFTER IMPLEMENTATION NEWBORN SCREENING IN US AND EUROPE Jaap Jan Boelens 1, Caroline Lindemans2, Peter van Hasselt3, Klaas Koop3, Maria I. Cancio1, Paul J. Orchard4, Troy C. Lund4 P647 ADDITION OF DINUTUXIMAB-BETA ON DAY -1 TO IMPROVE GRAFT-VERSUS-NEUROBLASTOMA EFFECTS OF HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN RELAPSED HIGH-RISK NEUROBLASTOMA Christina Lämmle 1, Michaela Döring1, Johannes H. Schulte1, Tim Flaadt1, Florian Heubach1, Peter Lang1,2,3, Christian M. Seitz1,2,3 P648 HHV-6 INFECTION MAY IMPAIR IMMUNE RECONSTITUTION FOLLOWING PEDIATRIC HAPLO-HSCT Katrine Kielsen 1, Eva Kannik Haastrup1, Lisbeth Pernille Andersen1, Tania Masmas1, Marianne Ifversen1 P649 FINAL HEIGHT AND IMPACT OF GROWTH HORMONE TREATMENT IN CHILDREN RECEIVING TBI-BASED CONDITIONING REGIMEN BEFORE HSCT FOR ACUTE LYMPHOBLASTIC LEUKEMIA Charlotte Calvo1,2, Coline Mornet3, Caroline Storey3, Carine Halfon-Domenech4, Cécile Pochon5, Marie-Dominique Tabone6, Guy Leverger6, André Baruchel1, Catherine Paillard7, Charlotte Jubert8, Marilyne Poirée9, Dominique Plantaz10, Justyna Kanold11, Virginie Gandemer12, Anne Sirvent13, Sandrine Thouvenin14, Marlène Pasquet15, Julie Berbis16, Pascal Auquier17, Jean-Hugues Dalle 1,2, Gérard Michel18, Paul Saultier19 P650 ENGRAFTMENT SYNDROME AND ACUTE GVHD AFTER HAPLOIDENTICAL POST-TRANSPLANT CYCLOPHOSPHAMIDE: A PAEDIATRIC MULTICENTRIC EXPERIENCE Maura Faraci 1, Francesco Saglio2, Francesca Baudi1, Filomena Pierri1, Francesca Bagnasco1, Stefano Giardino1, Francesca Gottardi3, Davide Leardini3, Franca Fagioli2,4, Riccardo Masetti3 P651 PULMONARY COMPLICATIONS IN CHILDREN FOLLOWING HAEMATOPOIETIC STEM CELL TRANSPLANT Hannah Walker 1, Diane Hanna1, Theresa Cole1, Gabrielle Haeusler1, Shivanthan Shanthikumar1, Melanie Neeland2 P652 EXCELLENT SURVIVAL USING T-REPLETE UMBILICAL CORD BLOOD TRANSPLANT IN HIGH RISK PAEDIATRIC MYELOID LEUKAEMIA, INCLUDING IN REFRACTORY DISEASE Kate Davies 1, Rob Wynn1 P653 TCRΑΒ + /CD19 + -DEPLETION IN HEMATOPOIETIC STEM CELLS TRANSPLANTATION FROM MATCHED UNRELATED AND HAPLOIDENTICAL DONORS IN CHILDREN WITH HIGH-RISK ACUTE MYELOBLASTIC LEUKEMIA Larisa Shelikhova 1, Olga Molostova1, Maria Ilyushina1, Yuliya Skvortsova1, Irina Shipitsina1, Rimma Khismatullina1, Sergey Blagov1, Svetlana Kozlovskaya1, Irina Kalinina1, Dina Baidildina1, Elena Gytovskaya1, Dmitriy Balashov1, Alexey Kazachenok1, Alexander Popov1, Ekaterina Mikhailova1, Julia Olshanskaya1, Dmitriy Litvinov1, Galina Novichkova1, Alexey Maschan1, Michael Maschan1 P654 IMPLEMENTATION OF CYP3 A5 GENOTYPING TO INDIVIDUALISE TACROLIMUS DOSE IN PAEDIATRIC PATIENTS UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANTATION Iván López Torija 1, Laura Gras Martín1, Pablo Escribano Sanz1, Pau Riera Armengol1, Carla Miñarro Chacón1, Sara Bernal Noguera1, Montserrat Torrent Español1, Esther Rojas Rodriguez1, Susana Boronat Guerrero1, Edurne Fernández de Gamarra Martínez1 P655 >EPIDEMIOLOGY OF GUT COLONIZATION AND INFECTION BY CARBAPENEM-RESISTANT GRAM-NEGATIVE BACILLI (CR-GNB) IN PEDIATRIC HEMATO-ONCOLOGICAL AND TRANSPLANTED PATIENTS Adriana Balduzzi 1,2, Iacopo Bellani2, Bianca Monti2, Marta Adavastro2, Francesca Limido2, Sonia Bonanomi1, Arianna De Buglio2, Marianna Rossi1, Francesca Vendemini1, Sergio Foresti1, Andrea Biondi1,2, Paolo Bonfanti1,2, Marco Guglielmo Migliorino1, Sergio Malandrin1, Francesca Iannuzzi1 P656 ALPHA/BETA-DEPLETION AND EARLY TIMING OF SEROTHERAPY PROVIDES RELIABLE ENGRAFTMENT AND LOW TRANSPLANT RELATED MORBIDITY IN CHILDREN UNDERGOING HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR MUCOPOLYSACCHARIDOSIS Annika Ewert 1, Anna Zychlinsky Scharff1, Rita Beier1, Britta Maecker-Kolhoff1, Isolde Schridde1, Kirsten Mischke1, Karl Walter Sykora1, Martin Sauer1 P657 CARDIORESPIRATORY FITNESS, PHYSICAL PERFORMANCE, AND METABOLIC SYNDROME IN ADULT SURVIVORS OF PAEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANTATION Anne Nissen 1, Tina Gerbek1, Kathrine Fogelstrøm1, Peter Schmidt-Andersen1, Kaspar Sørensen1, Abigail L. Mackey2,3, Martin Kaj Fridh1, Klaus Müller1,3,4 P658 HEMATOPOIETIC TRANSPLANTATION WITH CD45RA+ DEPLETION AND ADOPTIVE THERAPY WITH CD45RO- T-LYMPHOCYTES AND NK CELLS IN PEDIATRIC PATIENTS DIAGNOSED WITH HEMATOLOGICAL MALIGNANCIES AND NON-MALIGNANT DISEASES Mercedes Gasior Kabat 1, David Bueno1, Luisa Sisinni1, Yasmina Mozo1, Raquel De Paz1, Dolores Corral1, Mikel Fernandez Artazcoz1, Ana Belen Romero1, Antonio Marcos1, Victor Jimenez-Yuste1, Antonio Pérez-Martínez1 P659 PSYCHOLOGICAL IMPACT OF HEMATOPOIETIC STEM CELL DONATION ON PEDIATRIC SIBLING DONORS Sara Mostafa Makkeyah 1, Nihal Hussien Aly1, Eslam Elsayed Elhawary2, Sally Abdelmonsif Mohammed1, Salwa Amin Abdelhamid1 P660 AUTOLOGOUS STEM CELL TRANSPLANTATION IN HIGH-RISK NEUROBLASTOMA: A LONG-TERM FOLLOW-UP Ana Pinto1, Isabelina Ferreira 1, Gilda Teixeira1, Ana Sofia Jorge1, Pedro Sousa1, Maria João Gutierrez1, Bárbara Marques2, Ana Forjaz Lacerda1, Fernando Leal-da-Costa1, Nuno Miranda1 P661 OUTCOMES OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH DIAMOND-BLACKFAN ANEMIA – REPORT OF THE POLISH PEDIATRIC STUDY GROUP FOR HEMATOPOIETIC STEM CELL TRANSPLANTATION Anna Pieczonka 1, Olga Zajac-Spychala1, Krzysztof Kałwak2, Marek Ussowicz2, Jolanta Goździk3, Katarzyna Drabko4, Jan Styczynski5, Agnieszka Sobkowiak-Sobierajska1, Katarzyna Derwich1, Jacek Wachowiak1 P662 OUTCOMES OF ALLO-HSCT FROM MISMATCHED DONORS WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE IN CHILDREN WITH NON- MALIGNANT DISEASES Tatyana Bykova 1, Anna Osipova1, Olga Slesarchuk1, Oleg Goloshchapov1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1 P663 CARDIOVASCULAR RISK FACTORS AND SUBCLINICAL ORGAN DAMAGE ARE COMMON AND INCREASING OVER TIME IN CHILDREN AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION Damaris Werner 1, Jeannine von der Born1, Elena Lehmann1, Nima Memaran1, Britta Maecker-Kolhoff1, Martin Sauer1, Anette Melk1, Rita Beier1 P664 POSITIVE PRE-TRANSPLANT RESPIRATORY VIRAL PCR IS ASSOCIATED WITH INCREASED DAY 100 TRANSPLANT-RELATED MORTALITY IN PEDIATRIC HSCT RECIPIENTS Jane Trainor 1, Benjamin Hanisch1, Kelly Lyons Snelling1, Robert Podolsky1, David Alex Jacobsohn1 P665 HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS WITH MYELODYSPLASTIC SYNDROME Anna Osipova1, Tatiana Bykova 1, Olga Slesarchuk1, Olesya Paina1, Oleg Goloshchapov1, Tatiana Gindina1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1 P666 COMPARISONS OF PROGNOSIS OF ALLO-HSCT AFTER CAR-T CELL THERAPY OR CHEMOTHERAPY IN PAEDIATRIC PATIENTS OF REFRACTORY/RELAPSED B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA : A MULTICENTER STUDY CCCG-ALL-2015 Bohan Li 1, Jing Chen2, Chengjuan Luo2, Shaoyan Hu1 P667 TCR-ΑΒ DEPLETED HAPLOIDENTICAL TRANSPLANT WITH CD 45 RA- DEPLETED MEMORY T CELL ADD-BACK IN PAEDIATRIC NON-MALIGNANT DISEASES IN A SINGLE PAEDIATRIC CENTER: 10-YEAR RESULTS Tan Ah Moy 1,2, Michaela Seng1,2, Pham Ngi Thoc Anh1, Vijayakumari K1, Mya Soe Nwe1, Prasad Iyer1,2, Shui Yen Soh1,2, Joyce Lam1,2, Wing Leung1,3 P668 AIEOP EXPERT BOARD RECOMMENDATIONS FOR MANAGEMENT OF POST-HCT HEMORRHAGIC CYSTITIS IN PEDIATRIC SETTING Gianluca Dell’Orso 1, Simone Cesaro2, Marcello Carlucci1, Evelina Olcese1, Adriana Balduzzi3, Francesca Vendemini3, Massimo Catti4, Francesco Saglio4, Francesca Compagno5, Natalia Maximova6, Marco Rabusin6, Maria Cristina Menconi7, Katia Perruccio8, Elena Soncini9, Francesco Paolo Tambaro10, Veronica Tintori11, Daria Pagliara12, Maura Faraci1 P669 EXCELLENT SURVIVAL AND LONG TERM OUTCOMES IN INFANTS RECEIVING TOTAL BODY IRRADIATION FOR CONDITIONING PRIOR TO ALLOGENEIC TRANSPLANT Hannah Lust1, Stephanie Powell1, Karina Danner Koptik1, Erin Kaseda1, Jennifer Schneiderman1, Sonali Chaudhury 1 P670 BCG-VACCINE COMPLICATIONS IN CHILDREN WITH INFANT ACUTE LYMPHOBLASTIC LEUKEMIA Mikhail Lemeshev1, Alexandra Laberko 1, Larisa Shelikhova1, Veronika Fominykh1, Lili Khachatryan1, Galina Solopova1, Natalia Myakova1, Dmitry Balashov1 P671 WORKFORCE CONSENSUS RECOMMENDATIONS FOR PAEDIATRIC HSCT CENTRES ON BEHALF OF THE PAEDIATRIC SUB-COMMITTEE OF THE BRITISH SOCIETY OF BLOOD, BONE MARROW TRANSPLANTATION CELLULAR THERAPY (BSBMTCT) Reem Elfeky 1, Valerie Broderick2, Anna- Marie Ewins3, Elizabeth Rivers1, Wing Roberts4, Mary Slatter4, Kanchan Rao1, Beki James5, Brenda Gibson3 P672 HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH A CONTROLLED NUMBER OF CD3 + CELLS AND POST-TRANSPLANT CYCLOPHOSPHAMIDE: EXPERIENCE FROM A SINGLE PEDIATRIC CENTER Giulia Albrici1, Giulia Baresi1, Stefano Rossi1, Elena Soncini1, Elisa Bertoni1, Marta Comini2, Federica Bolda2, Alessandra Beghin2, Arnalda Lanfranchi2, Fulvio Porta1, Marianna Maffeis 1 P673 PLASMA AMINO ACIDS AND FECAL CALPROTECTIN USAGE FOR THE PREDICTION OF INTESTINAL INJURY AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN Igne Kairiene 1, Jelena Rascon1, Ramune Vaisnore1 P674 MULTIPLE MINOR PHLEBOTOMIES IN EPP INVOLVING THE LIVER: IS THERE A WAY TO AVOID NEEDING A LIVER AND HAEMATOPOIETIC STEM CELL TRANSPLANT? Rita Beier 1, Jasmin Barman-Aksözen2, Franziska van Breemen2, Hagen Ott3, Katharina Becker1, Annika Ewert1, Ulrich Baumann1, Eva Pfister1, Martin Sauer1, Elisabeth Minder2, Britta Maecker-Kolhoff1, Anna-Elisabeth Minder2 P675 COMPARISON OF BUSULFAN-BASED AND TREOSULFAN-BASED HIGH-DOSE CHEMOTHERAPY AND AUTOLOGOUS STEM CELL TRANSPLANTATION IN HIGH-RISK NEUROBLASTOMA Kyung-Nam Koh 1, Young Kwon Koh2, Aejin Kang1, Ji Young Kim1, Su Hyun Yun1, Sung Han Kang1, Hyery Kim1, Ho Joon Im1 P676 USE OF BLINATUMOMAB TO ACHIEVE REMISSION AND CONSOLIDATION WITH HAPLOIDENTICAL TRANSPLANT WITH CYCLOPHOSPHAMIDE FOR THE TREATMENT OF CHILDREN WITH REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA Alberto Olaya Vargas 1, Haydee Salazar-Rosales2, Martin Peréz-Gárcia3, Yadira Melchor-Vidal4, Annecy Herver-Olivares5, Gerardo López- Hernández2, Nideshda Ramírez-Ortiz2, Cesar Galván-Diaz2, Alberto Olaya-Nieto2 P677 HEMORRHAGIC CYSTITIS IN CHILDREN AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: INCIDENCE, TREATMENT, OUTCOME, AND RISK FACTORS Nur Ayca Celik 1, Talia Ileri2, Elif Ince2, Hasan Fatih Cakmaklı2, Berk Burgu1, Zeynep Ceren Karahan1, Seda Kaynak Sahap1, Ebru Dumlupınar1, Mehmet Ertem2 P678 TRANSPLANT-ASSOCIATED THROMBOTIC MICROANGIOPATHY (TA-TMA) IN CHILDREN TREATED WITH ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (ALLO-HSCT): ONE CENTER STUDY Tomasz Jarmoliński 1, Monika Rosa1, Iwona Bil-Lula1, Joanna Korlaga1, Krzysztof Kałwak1, Marek Ussowicz1 P679 DISTANCE MAKES THE CELLS GROW FONDER- PAEDIATRIC ALLOGENIC TRANSPLANT AND CELLULAR THERAPY REFERRAL PROGRAM, THE SOUTH AUSTRALIAN EXPERIENCE Matthew O’Connor 1,2, Catherine Mitchell1, Laura Chapman2, Karen McCleary2, Adam Nelson2, Richard Mitchell2 P680 INCREASING TIM-3 + T CELLS IN PERIPHERAL BLOOD OF PEDIATRIC PATIENTS WITH RELAPSE B-ALL AFTER HAPLOIDENTICAL ALLO-HSCT Liubov Tsvetkova 1, Olga Epifanovskaya1, Elena Babenko1, Olesya Paina1, Polina Kozhokar’1, Zhemal Rakhmanova1, Olesya Yudinceva1, Anna Osipova1, Sabina Ryabenko1, Elena Dobrovolskaya1, Ivan Moiseev1, Alexandr Kulagin1, Ludmila Zubarovskaya1 P681 IMMUNE-MEDIATED CYTOPENIA AFTER PEDIATRIC HEMATOPOIETIC CELL TRANSPLANTATION – A RETROSPECTIVE ANALYSIS Mária Füssiová 1, Júlia Horáková1, Ivana Boďová1, Jaroslava Adamčáková1, Dominika Dóczyová1, Miroslava Pozdechová1, Tomáš Sýkora1, Peter Švec1, Alexandra Kolenová1 P682 SEQUENTIAL KIDNEY-STEM CELL TRANSPLANTATION IN PATIENT WITH SCHIMKE IMMUNO-OSSEOUS DYSPLASIA: CASE REPORT Alexandra Shutova1, Alexandr Bazaev1, Yulia Skvortsova1, Alexandr L. Rumyantsev2, Diana Khalikova2, Galina Novichkova1, Michael Maschan 1, Dmitry Balashov1, Alexey Maschan1 P683 CYCLOSPORINE PLUS METHOTREXATE VERSUS CYCLOSPORINE ALONE FOR GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS IN PEDIATRIC PATIENTS UNDERGOING HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM HLA-IDENTICAL SIBLING Vincenzo Apolito 1, Valeria Ceolin1, Manuela Spadea1,2, Marta Barone1, Marco Basiricò1, Francesco Saglio1, Franca Fagioli1,2 P684 NUTRITIONAL DYNAMICS IN PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A RETROSPECTIVE MONOCENTRIC STUDY ASSESSING BMI PERCENTILES AND CLINICAL OUTCOMES Timo Stetter1, Michaela Döring 1, Christian Seitz1, Nora Rieflin1, Thomas Baumgarten1, Léa Thérond1, Pia Glogowski1, Johannes Schulte1, Rupert Handgretinger1, Peter Lang1, Karin M. Cabanillas Stanchi1 P685 INTEGRATING CAR T-CELL THERAPY AND ALLOGENEIC STEM CELL TRANSPLANTATION IN CHILDREN WITH HIGH RISK ACUTE LYMPHOBLASTIC LEUKEMIA. THE EXPERIENCE OF A SINGLE CENTER Blanca Molina 1, Marta Gonzalez Vicent1, Sara Vinagre1, Blanca Herrero1, David Diaz1, Miguel Angel Diaz1 P686 ASSESSMENT OF THE ENDOTHELIAL GLYCOCALYX BY VIDEO MICROSCOPY IN CHILDREN WITH ACUTE LEUKEMIA AFTER ALLOGENEIC STEM CELL TRANSPLANTATION Zarina Khaerova1, Zhemal Rakhmanova1, Olesya Paina1, Timur Vlasov2, Elena Semenova1, Ivan Moiseev 1, Alexander Kulagin1, Ludmila Zubarovskaya1 P687 INTRODUCTION OF BODY IMPEDENCE ANAYSIS TO MEASURE BODY COMPOSITION CHANGES AND FLUID STATUS OF PAEDIATRIC PATIENTS UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANTATION Rebecca Fisher 1, Alice Brewer1, Belinda Meares1, Sheridan Collins1, Sarah Slack1, Melissa Gabriel1 P688 TRANSPLANTATION OUTCOME OF HIGH RISK ACUTE MYELOID LEUKEMIA AND MYELODYSPLASTIC SYNDROME IN CHILDREN: SINGLE CENTER EXPERIENCE IN KOREA Young Tak Lim 1, Eu Jeen Yang1, Jun Young Oh1 P689 BUILDING A PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANT UNIT IN PARAGUAY: LESSON LEARNT Marta Verna 1, Maria Liz Benitez2, Attilio Maria Rovelli1, Jabibi Noguera2, Marta Canesi1, Valentino Conter1, Susy Thiel Figueredo3, Andrea Biondi1,4 P690 HIGH-DOSE CHEMOTHERAPY WITH STEM CELL TRANSPLANTATION IN ADULT PRIMARY MEDIASTINAL GERM CELL TUMORS. A RETROSPECTIVE STUDY OF THE EBMT, CELLULAR THERAPY & IMMUNOBIOLOGY WORKING PARTY Paolo Pedrazzoli1, Manuela Badoglio2, Myriam Labopin2, Giovanni Rosti1, Matthieu Delaye3, Carsten Bokemeyer4, Christoph Seidel4, Jane Apperley5, Elisabetta Metafuni6, Juergen Finke7, Jean-Henri Bourhis8, Christos Kosmas9, Florent Malard10, Jurgen Kuball11, Christian Chabannon12, Annalisa Ruggeri13, Simona Secondino 1 P691 SURVIVAL OUTCOMES IN PANCREATIC CANCER: EVALUATING THE IMPACT OF WIPPLE’S SURGERY AND HAEMATOPOIETIC CELL TRANSPLANTATION Olle Ringden 1, Johan Permert2, Johan Törlen1, Stephan Mielke1, Behnam Sadeghi1 P692 SUCCESSFUL HIGH DOSE CHEMOTHERAPY WITH STEM CELL RESCUE IN DISSEMINATED OVARIAN CHORIOCARCINOMA COMPLICATED WITH CHORIOCARCINOMA SYNDROME Abdulaziz Bin Mesained 1, Abdulwahab Alharthi1, Walid Hazem1, Omar Alsharif1, Nawaf Alkhayat1, Mohammad Alshahrani1, Hasna Hamzi1, Amal Binhassan1, Yasser Elborai1 P693 DONOR-SPECIFIC ANTI-HLA ANTIBODIES (DSAS) IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM MISMATCHED DONORS, ON BEHALF OF GITMO AND AIBT Ursula La Rocca1,2, Roberto Ricci1, Alfonso Piciocchi3, Walter Barberi1, Elena Oldani4, Alida Dominietto5, Raffaella Cerretti6, Alessandra Picardi6, Francesca Bonifazi7, Riccardo Saccardi8, Maura Faraci9, Giovanni Grillo10, Lucia Farina11, Benedetto Bruno12,13, Anna Grassi4, Anna Proia14, Elena Tagliaferri15, Giuseppina De Simone16, Michele Malagola17, Michela Cerno18, Simone Cesaro19, Paolo Bernasconi20, Lucia Prezioso21, Paola Carluccio22, Nicola Mordini23, Matteo Pelosini24, Attilio Olivieri25, Patrizia Chiusolo26, Stella Santarone27, Michele Cimminiello28, Roberto Crocchiolo10, Franco Papola29, Gianni Rombolà30, Nicoletta Sacchi31, Valeria Miotti32,33, Lia Mele34,33, Benedetta Mazzi35,33, Fabio Ciceri36, Massimo Martino37, Anna Paola Iori 1 P694 DETERMINING THE PREDICTIVE IMPACT OF DONOR PARITY ON THE OUTCOMES OF HUMAN LEUKOCYTE ANTIGEN MATCHED HEMATOPOIETIC STEM CELL TRANSPLANTS: A RETROSPECTIVE, SINGLE-CENTER STUDY Tanaz Bahri1, Maryam Barkhordar 1, Mojtaba Azari1, Hosein Kamranzadeh1, Seied Asadollah Mousavi1, Soroush Rad1, Sahar Tavakoli Shiraji1, Mohammad Vaezi1 P695 POST-TRANSPLANTATION CYCLOPHOSPHAMIDE IMPROVES GRAFT-VERSUS-HOST DISEASE-FREE, RELAPSE-FREE SURVIVAL IN HLA-DPB1 MISMATCHED UNRELATED DONOR ALLOGENEIC TRANSPLANT Shannon R. McCurdy1, Scott R. Solomon2, Brian C. Shaffer3, Meilun He4, Yung-Tsi Bolon 4, Amanda G. Blouin3, Alla Keyzner5, Francisco A. Socola6, Uroosa Ibrahim5, Jun Zou7, Hana Safah6, Nakhle Saba6, Shahinaz Gadalla8, Miguel-Angel Perales9, Sophie Paczesny10, Steven Marsh11, Effie W. Petersdorf12, Tao Wang13, Ephraim J. Fuchs14, Stephanie J. Lee12 P696 THE RESCUE TRANSPLANTATION STRATEGY FOR GRAFT FAILURE: DONOR SELECTION AND GVHD PROPHYLAXIS IN THE CURRENT ERA Mizuki Watanabe 1,2, Junya Kanda1, Kentaro Fukushima3, Kaito Harada4, Naoyuki Uchida5, Makoto Onizuka4, Noriko Doki6, Ken-ichi Matsuoka7, Takahiro Fukuda2, Satoshi Yoshihara8, Toshiro Kawakita9, Shingo Yano10, Masatsugu Tanaka11, Yasushi Onishi12, Fumihiko Ishimaru13, Tatsuo Ichinohe14, Yoshiko Atsuta15, Kimikazu Yakushijin16, Hideki Nakasone17,18 P697 UNRELATED DONOR SELECTION FOR STEM CELL TRANSPLANT USING POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR ACUTE MYELOID LEUKEMIA: A STUDY FROM THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT Jaime Sanz 1, Myriam Labopin2, Goda Choi3, Alexander Kulagin4, Jacopo Peccatori5, Jan Vydra6, Péter Reményi7, Jurjen Versluis8, Montserrat Rovira9, Didier Blaise10, Hélène Labussière-Wallet11, Juan Montoro12, Simona Sica13, Ellen Meijer14, Maija Itäla-Remes14, Nicolaas Schaap15, Claude Eric Bulabois16, Simona Piemontese5, Mohamad Mohty17, Fabio Ciceri5 P698 PATIENT HLA GENOTYPE AND REGISTRY DIVERSITY STRONGLY INFLUENCE DONOR SEARCH DURATION AND SUCCESS Stefanie V. Dorner 1, Henning Baldauf2, Carina Rave2, Christine Urban3, Thilo Mengling3, Johannes Schetelig2, Alexander H. Schmidt3,1, Julia Pingel1 P699 FEASIBILITY AND SAFETY OF GRANULOCYTE COLONY-STIMULATING FACTOR (G-CSF) PERIPHERAL BLOOD STEM CELL MOBILIZATION AND COLLECTION BY APHERESIS IN RELATED DONORS AGED 65 AND OVER Kahina Amokrane 1, Kamelia Alexandrova1, Cristina Castilla-Llorente1, Sylvain Pilorge1, Tereza Coman1, Jean-Henri Bourhis1, Valerie Lapierre1 P700 HAPLOIDENTICAL VERSUS MISMATCHED UNRELATED ALLOGENEIC STEM CELL TRANSPLANTATION FOR MYELOID MALIGNANCIES: AN ANALYSIS ON 1413 PATIENTS FROM THE GERMAN REGISTRY Andrea Gantner 1, Daniel Fürst2,3, Aysenur Arslan2,3, Svenja Labuhn4, Mark Ringhoffer5, Johannes Schetelig6,7, Thomas Schroeder8, Gesine Bug9, Elisa Amann2,3, Christine Neuchel2,3, Sandra Schmeller4, Jan Beyersmann4, Hubert Schrezenmeier2,3, Joannis Mytilineos10, Nicolaus Kröger11, Elisa Sala1 P701 THE POST-PANDEMIC HAEMATOPOIETIC STEM CELL (HSC) GRAFT CRYOPRESERVATION TRENDS AND THEIR EFFECTS ON UNRELATED DONOR REGISTRIES: THE UK EXPERIENCE Ruta Barkauskiene1, Sara Lozano Cerrada1, Angharad Pryce1, Joanne Badger1, Paul Johnson1, Ann O’Leary1, Robert Danby 1, Chloe Anthias1 P702 ALLOGENIC STEM CELL TRANSPLANTATION AND PERI TRANSPLANT STRATEGIES IN PATIENTS WITH MISMATCH DONORS – A SINGLE CENTER EXPERIENCE IN 151 PATIENTS Paul Jäger1, Benno Biermann 1, Felicitas Schulz1, Kathrin Nachtkamp1, Patrick Tressin1, Ben-Niklas Baermann1, Annika Kasprzak1, Felix Matkey1, Ulrich Germing1, Sascha Dietrich1, Guido Kobbe1 P703 SURVIVAL PROGNOSTIC FACTORS AFTER A SECOND ALLOGENEIC HEMATOPOIETIC TRANSPLANTATION Rodrigo Cantera Estefanía1, Raquel García Ruiz1, Miriam Sánchez Escamilla1, Leddy Patricia Vega Suárez1, María Terán1, Dianis Escorcio Fariab2, Sara Fernández Luis1, Juan José Domínguez García1, Tatiana Fernández Barge 1, Ana Gea Peña1, Irene Francés Alexandre1, Ana Tobalina García1, María Oviedo Madrid1, Irene Gorostidi Álvarez1, Julia Bannatyne Undabeitia1, Juan Manuel Cerezo Martín1, Mercedes Colorado Araujo1, Mónica López Duarte1, Guillermo Martín Sánchez1, Lucrecia Yáñez San Segundo1, Noemí Fernández Escalada1, Andrés Insunza Gaminde1, Monserrat Briz del Blanco1, Jose Iñigo Romón Alonso1, Gala Aglaia Méndez Navarro1, Enrique Ocio San Miguel1, Arancha Bermúdez Rodríguez1 P704 QUALITY OF ENGRAFTMENT AND INCIDENCE OF ACUTE GVHD GRADE II – IV POST PERIPHERAL HAEMOPOIETIC CELLS CRYOPRESERVATION IN PATIENTS UNDERGOING ALLOGENEIC TRANSPLANTATION FROM HLA IDENTICAL DONORS Anna Maria Raiola 1, Massimiliano Gambella1, Barabino Luca2, Stefania Bregante1, Carmen Di Grazia1, Alida Dominietto1, Anna Ghiso1, Livia Giannoni1, Alberto Serio1, Silvia Luchetti1, Riccardo Varaldo1, Monica Passannante1, Antonella Laudisi1, Alessandra Scalas2, Paola Contini3, Federico Ivaldi3, Raffaele De Palma3, Alessandra Bo1, Emanuele Angelucci1 P705 WHAT WORKS AND WHAT DOES NOT? THE IMPACT OF DONOR CHARACTERISTICS IN T REPLETE HAPLOIDENTICAL STEM CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE IN CHILDREN Anuraag Reddy Nalla 1, Revathi Raj1, Ramya Uppuluri1, Venkateswaran Vellaichamy1, Suresh Duraisamy1, Kavitha Ganesan1, Anupama Nair1, Vijayshree Muthukumar1, Indira Jayakumar1, Ravindra Prasad1, Usha Surianarayanan1 P706 OUTCOMES OF OLDER ADULTS UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE BASED PROPHYLAXIS Victoria Murillo Cortés 1, Paola Charry2, María Suárez-Lledó3,4, María Teresa Solano4, Anna Serrahima4, Joan Cid2,4, Miquel Lozano2,4, Laura Rosiñol3,4, Jordi Esteve3,4, Álvaro Urbano-Ispizua3,4, Enric Carreras5, Francesc Fernandez-Avilés3,4, Carmen Martínez3,4, Montserrat Rovira3,4, María Queralt Salas3,4 P707 COMPARING THE CLINICAL OUTCOMES OF ALLOGENEIC STEM CELL TRANSPLANTATION FROM MATCHED-SIBLING DONORS AND ALTERNATIVE DONORS IN PATIENTS WITH SEVERE THALASSEMIA : SYSTEMATIC REVIEW AND META-ANALYSIS Sirichai Srichairatanakool 1, Wuttipat Kiratipaisarl1, Sarunsorn Krintratun1, Adisak Tantiworawit1, Chatree Chai-Adisaksopha1 P708 IMPACT OF DONOR BLOOD GROUP AND CMV MATCH ON ALLOGENEIC STEM CELL TRANSPLANTATION: A SINGLE CENTER LONG TERM FOLLOW UP STUDY Mohamed Debes1, James Clarke1, Yeong Lim1, Shahid Iqbal1, Leah Credidio1, Hannah Williams2, Jennifer Gibson1, Sajid Pervaiz1, Angela Milner1, Thomas Seddon1, Arpad Toth1, Muhammad Saif 1 P709 DIFFERENCES IN OVERALL SURVIVAL AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT WITH IDENTICAL AND HAPLOIDENTICAL DONORS IN MEXICO Marco Alejandro Jiménez-Ochoa 1, Elsa Avila-Arreguin2, Severiano Baltazar-Arellano3, María Margarita Contreras-Serratos1, Martha Leticia González Bautista1, Alba Morales-Hernandez2, Uendy Pérez-Lozano4, María Guadalupe Rodríguez González2, Guillermo Sotomayor-Duque3, Elizabeth Urbina-Escalante1 P710 ANTI-T LYMPHOCYTE GLOBULIN (ATLG) TO PREVENT GRAFT FAILURE AFTER HAPLOIDENTICAL STEM CELL TRANSPLANTATION WITH DONOR SPECIFIC ANTIBODIES (DSA) Christian Niederwieser 1, Radwan Massoud1, Evgeny Klyuchnikov1, Gagelmann Nico1, Johanna Richter1, Kristin Rathje1, Tatiana Urbanowicz1, Ameya Kunte1, Janik Engelmann1, Christina Ihne1, Iryna Lastovytska1, Cecilia Lindhauer1, Franziska Marquard1, Mirjam Reichard1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schaeferskuepper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Silke Heidenreich1, Ina Rudolph1, Normann Steiner1, Julia Kienast1, Maroly Bohorquezmanjarres1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger1 P711 HAPLOIDENTICAL TRANSPLANT WITH POST- CY WITHOUT T DEPLETION. PEDIATRIC EXPERIENCE IN HIGH-RISK LEUKEMIAS FROM GATMO [ARGENTINIAN GROUP OF BONE MARROW TRANSPLANT] Maria Laura Rizzi 1, Natalia Gonzalez1, Evelin Colombo1, Sol Jarchum1, Emilia Mas2, Victoria Suen2, Sandra Formisano3, Vera Milovik4, Rquel Staciuk5, Pablo Longo6, Silvina Palmer3, German Stemelin3, Silvina Wilberger4, Valeria Santidrian5 P712 DONOR SELECTION CRITERIA IN ALLOGENEIC STEM CELL TRANSPLANTATION FOR ACUTE MYELOID LEUKAEMIA: RETROSPECTIVE SINGLE-CENTER EVALUATION Ursula La Rocca1,2, Saveria Capria 1, Claudia Lucci1, Roberto Ricci1, Adele Delli Paoli1, Martina Salvatori1, Maurizio Martelli1, Anna Paola Iori1, Walter Barberi1 P713 IRANIAN DONORS SAVE LIVES: COLLABORATING WITH COMMUNITY ADVOCATES TO ADDRESS RACIAL DISPARITY IN ACCESS TO UNRELATED DONORS FOR ALLOGENEIC TRANSPLANTATION Farnaz Farahbakhsh 1,2,3, Warren Fingrut1,4,5, Bonnie Lu1 P714 INVESTIGATE THE POTENTIAL ASSOCIATION OF HLA ALLELES WITH HEMATOPOIETIC STEM CELL TRANSPLANTATION OUTCOMES Fatma Al Lawati 1,2, Murtadha Alkhbouri3, Salma Al Harrasi2, Aliya Al Ansari1 P715 SUCCESSFUL OUT-PATIENT SELF-ADMINISTRATION OF MOBILISATION AGENTS FOR HAEMATOPOIETIC STEM CELL TRANSPLANT Yona Skosana 1, Emma Smith1, Bridged Mofokeng1, Thabo Gcayiya1, Tanya Glatt1,2,3 P716 ANALYSIS OF BONE MARROW TRANSPLANTATION OUTCOMES BETWEEN JAPANESE AND NON-JAPANESE DONORS AND RECIPIENTS Ryu Yanagisawa 1, Michiho Shindo2, Akihito Shinohara2, Yachiyo Kuwatsuka3, Koichi Nakase4, Fumihiko Kimura5, Naoki Shingai6, Tetsuya Nishida7, Takahiro Fukuda8, Masatoshi Sakurai9, Mineo Kurokawa10, Takashi Koike11, Shuichi Ota12, Satoru Takada13, Makoto Onizuka11, Tatsuo Ichinohe14, Yoshiko Atsuta15,16, Junya Kanda17, Hideki Nakasone18,19, Tomomi Toubai20 P717 PSYCHOLOGICAL ASSESSMENT OF HEMATOPOIETIC STEM CELL DONORS UNDERGOING PERIPHERAL BLOOD STEM CELL MOBILISATION Alka Khadwal 1, Ritwik Lahiri1, Sandeep Grover1, Rekha Hans1, Nabhajit Mallik1, Charanpreet Singh1, Arihant Jain1, Deepesh Lad1, Gaurav Prakash1, Aditya Jandial1, Pankaj Malhotra1 P718 IMPACT AND EFFICACY OF DONOR TYPE IN ALLOGENEIC STEM CELL TRANSPLANTATION IN PATIENTS WITH ACUTE LEUKEMIA, SINGLE CENTER EXPERIENCE Lazar Chadievski 1, Irina Panovska Stavridis1, Sanja Trajkova1, Nevenka Ridova1, Milche Cvetanoski1, Simona Stojanovska1, Bozidar Kocoski1, Lidija Chevreska1, Svetlana Krstevska Balkanov1, Aleksandra Pivkova Veljanovska1 P719 THE EFFECT OF GRAFT MANIPULATION AND CRYOPRESERVATION ON DIFFERENT GRAFT COMPONENT AND ITS POTENTIAL CLINICAL APPLICATIONS Mohammed Kawari 1,2, Emily Fu1, Shiyi Chen1, Mileidys Alvarez1, Jessica McLeod1, Muhammad Badawi1, Arpita Parikh1, Rashied Kawshermolla1, Racheljihye Kim1, Bramdeo Motiram1, Lynn Jean1, Miyada Himmat1, Lydia Morrison1, Keanu Herzog1, Madhavi Gerbitz3, Megan Nelles1, Eunyoung Cho1, Ahmed Najemeldin1, Igor Novitzky-Basso1,2, Armin Gerbitz1,2,3 P720 A SIMPLE PREDICTIVE ALGORITHM FOR ESTIMATION OF THE BLOOD VOLUME TO BE PROCESSED TO YIELD TARGET CELL NUMBERS FOR MANUFACTURING OF TISA-CEL® AND AXI-CEL® Stephanie Winter 1, Andreas Tanzmann1, Manuela Branka1, Patricija Rajsp1, Philipp Wohlfarth1, Philipp Staber1, Ulrich Jaeger1, Antonia Mueller1, Nina Worel1 P721 REAL-WORLD MOBILIZATION AND HARVEST OUTCOMES IN NEWLY DIAGNOSED MULTIPLE MYELOMA PATIENTS RECEIVING D-VTD: UK EXPERIENCE Jose Aspa Cilleruelo 1, Christianne Bourlon2, Elisa Roldan Galvan1, Reuben Benjamin2, Kirsty Cuthill2, Arief Gunawan2, Majid Kazmi3, Pramila Krishnamurthy2, Victoria Potter2, Robin Sanderson2, Michelle Kenyon2, Mili Shah2, Matthew Streetly3, Fabio Serpenti2, Benjamin Robinson1, Andrew Chantry1, Stella J. Bowcock2, Lalita Banerjee4, John Robert Jones5, Sabia Rashid6, Ana Isabel Duran Maestre6, Satyajit Sahu6, Sudarshan Gurung7, Lianwea Chia8, Haili Cui2, Alison Pratt2, Maheen Ahsan2, Stella Bouziana2, John A. Snowden1, Katharine Elizabeth Bailey2 P722 FACTORS AFFECTING CD34 + CRUDE COLLECTION EFFICIENCY IN MOBILIZED DONORS UNDERGOING LEUKOCYTAPHERESIS Jesús Fernandez 1, Monica Linares1, David Gomez-Vives1, Sara Lewandowski2, Hugo Fabre3, Ana Garcia-Buendia1, Marina Rierola1, Veronica Pons1, Julia Ayats1, Juan Porras1, Carmen Montanero1, Teresa Contijoch1, Isabel Amaya1, Esperanza Ribas1, Marc Vilches1, Marta Guerrero1, Alba Acedo1, Sofia Alonso1, Nerea Castillo-Flores1, Sergi Querol1, Rafael Parra1 P723 EFFICACY OF PROPHYLACTIC ANTIBIOTICS FOR THE PREVENTION OF NEUTROPENIC FEVER IN PATIENTS WITH MULTIPLE MYELOMA RECEIVING HIGH-DOSE CYCLOPHOSPHAMIDE FOR STEM CELL MOBILIZATION Liqiong Hou1, Juan Li 1 P724 EFFICACY OF PLERIXAFOR IN CD34+ STEM CELL MOBILIZATION IN HEALTHY PEDIATRIC STEM CELL DONORS Rishab Bharadwaj1, Vimal Kumar 1, Deenadayalan Munirathnam1 P725 PEGFILGRASTIM FOR STEM CELL MOBILIZATION FOLLOWED BY FRESH STEM CELL INFUSION IN MULTIPLE MYELOMA Hegta Tainá Rodrigues FIgueiroa 1, Pedro Paulo Faust1, Phillip Scheinberg1, Juliana Matos Pessoa1, Fauze Lutfe Ayoub1, André Dias Américo1, Dimitra Delijaicov1, Breno Moreno Gusmão1, André Larrubia1, Eurides Leite da Rosa1, Ana Cynira Franco Marret1, Arlette Edna Lazar1, Germano Glauber de Medeiros Lima1, Isabella Silva Pimentel Pitol1, Fabio Rodrigues Kerbauy1,2 P726 A MID-APHERESIS CD34 COUNT UTILITY IN EARLY STOPPAGE OF APHERESIS IN PATIENTS/DONORS IN A PBSCT PROCEDURE – ANALYSIS OF EXPERIENCE IN TWO SOUTH INDIAN CANCER CENTRES Veni Prasanna Gedala 1, Rakesh Reddy Boya1, Pradeep Ventrapati1, Anuradha Dasari1, Chandrasekhar Bendi2, Praveena Voonna2 P727 PLERIXAFOR AS A MOBILIZING AGENT FOR SALVAGE HIGH-DOSE CHEMOTHERAPY IN RELAPSED/REFRACTORY TESTICULAR CANCER: AN EXPERIENCE OF 22 CASES Sara Bleve1, Maria Concetta Cursano1, Cecilia Menna1, Caterina Gianni1, Giuseppe Schepisi1, Valentina Gallà2, Chiara Casadei1, Serena Mangianti3, Stefano Baravelli4, Accursio Fabio Augello4, Francesco Lanza5, Simona Secondino6, Paolo Pedrazzoli6, Giovanni Rosti1, Ugo De Giorgi 1 P728 ASSESSING AND REFINING PREDICTIVE MODELS OF POTENTIAL POOR MOBILIZERS Nelson Chan1, Kai Wan 2, Sinju Thomas1, Sandra Loaiza1, Noora Buti1, Sophie Zemenides1, Eitan Mirvis1, Lucy Cook1,2, Edward Bataillard1, Edward Kanfer1, Maria Atta2, Aristeidis Chaidos2,1, Jane Apperley2,1, Edurado Olavarria2,1, Renuka Palanicawandar1 P729LEFT OUT IN THE COLD: STEM CELL GRAFTS ARE STILL FUNCTIONAL AFTER MORE THAN 20 YEARS IN CRYOSTORAGE Rebecca Axelsson Robertson 1,2, Lyda Osorio Fernandez1, Michael Uhlin2 P730 SAFETY AND EFFICACY OF PLERIXAFOR IN POOR MOBILIZERS: AN INTERIM ANALYSIS AND MULTICENTER PROSPECTIVE STUDY Weiping Liu1, Yao Liu2, Hui Liu3, Mingxing Zhong4, Zengjun Li5, Caixia Li6, Yuerong Shuang7, Jun Luo8, Jian Zhou9, Jie Ji 10, Sujun Gao11, Wenzheng Yu12, Xiaopei Wang1, Yuqin Song1, Jun Zhu1 P731 BONE MARROW HARVESTING IN ALLOGENEIC DONORS: HOW TO PREDICT SUCCESSFUL AND SATISFYING BM PRODUCT Tigran Torosian 1, Alicja Wozniak1, Grzegorz Hensler1, Katarzyna Szymanska1, Iwona Tomanek1 P732 COMPARISON OF MOBILIZATION REGIMENS USED IN LYMPHOMA AND FACTORS THAT INFLUENCE PERIPHERAL BLOOD STEM CELL MOBILIZATION Laxma Reddy 1,2, Anant Gokarn1,2, Akanksha Chichra1,2, Sachin Punatar1,2, Sumeet Mirgh1,2, Nishant Jindal1,2, Lingaraj Nayak1,2, Navin Khattry1,2, Sumathi Hiregoudar1,2, Minal Poojary1,2, Suryatapa Saha1,2, Shashank Ojha1,2, Bhausaheb Bagal1,2 P733 THIRTEEN-YEAR EXPERIENCE WITH PLERIXAFOR IN CHILDREN FROM A SINGLE CENTRE: TOLERABILITY, GRAFT QUALITY AND ENGRAFTMENT Lucy Metayer1, Charlotte Rigaud1, Valerie Lapierre1, Sylvie Psyche1, Angelique Benoit-Tricoire1, Claudia Pasqualini1, Christelle Dufour1, Kamélia Alexandrova 1 P734 AN ALTERNATIVE USE OF A COLLOIDAL SUBSTITUTE (SUCCINYLATED GELATINE) FOR QUALITY CONTROLS IN PERIPHERAL BLOOD STEM TRANSPLANTATION Claudia Del Fante 1, Eugenio Barone1, Simona De Vitis1, Rosalia Cacciatore1, Daniela Troletti1, Cesare Perotti1 P735 ANTI-CD38 TREATMENT AS A PREDICTIVE FACTOR FOR STEM CELL MOBILIZATION FAILURE AND DELAYED GRAFT RECOVERY IN MULTIPLE MYELOMA: A TERTIARY HOSPITAL EXPERIENCE Paula Zoco Gallardo 1, Ariadna García Ascacibar1, Juan Mateos Mazón1, Laura Posada Alcón1, Javier Arzuaga Méndez1, Julia Arrieta Aguilar1, Amaia Uresandi Iruin1, Amaia Balerdi Malcorra1, Maria Elena Amutio Díez1, María Casado Sánchez1, Sara Hormaza De Jauregui1, Gorka Pinedo Martín1, Laura Aranguren Del Castillo1, Ana Lobo Olmedo1, Maria Elena Amutio Díez1, Juan Carlos García Ruiz1 P736 INCREASED DOSE OF G-CSF OVERCOMES AGE AS A RISK FACTOR FOR POOR MOBILIZATION Yuliya Shestovska 1, Shalina Joshi1, Dzhirgala Mandzhieva1, Regina Mullaney1, Ey Luu1, Rashmi Khanal1, Asya Varshavsky Yanovsky1, Michael Styler1, Henry Fung1, Peter Abdelmessieh1 P737 THE IMPACT OF TRANSPORTATION TIME ON STEM HEMATOPOIETIC CELL VIABILITY AFTER CRYOPRESERVATION Iwona Mitrus 1, Marcin Wilkiewicz1, Agata Kawulok1, Wojciech Fidyk1, Małgorzata Sobczyk-Kruszelnicka1, Sebastian Giebel1 P738 LENOGRASTIM AND FILGRASTIM: REAL-LIFE COMPARISON IN MOBILIZATION OF ALLOGENEIC HEMATOPOIETIC STEM CELL DONORS Rosamaria Nitti 1, Filippo Magri1, Alessia Orsini1, Michela Giulia Tassara1, Raffaella Milani1, Salvatore Gattillo2, Milena Coppola1, Luca Santoleri1, Fabio Ciceri3 P739 NIVOLUMAB-IFOSFAMIDE-CARBOPLATIN-ETOPOSIDE (NICE) AS MOBILIZING REGIMEN FOR RELAPSED/REFRACTORY HODGKIN LYMPHOMA (R/R HL) Maria K. Angelopoulou1, Eleni Lalou1, Athanasios Liaskas 1, Aikaterini Benekou1, Maria-Aikaterini Lefaki1, Ioannis Vasilopoulos1, Angeliki Georgopoulou1, Chrysovalantou Chatzidimitriou1, Alexandros Machairas1, Alexia Piperidou1, Kalliopi Zerzi1, Konstantinos Keramaris1, Marina P. Siakantaris1, Panayiotis Panayiotidis1, Theodoros P. Vassilakopoulos1 P740 IMPACT OF DARATUMUMAB, BORTEZOMIB, THALIDOMIDE DEXAMETHASONE INDUCTION THERAPY ON STEM CELL MOBILIZATION: A REAL WORLD EXPERIENCE Giuseppe Sapienza 1, Marika Porrazzo1, Fabrizio Accardi1, Roberto Bono1, Stefania Tringali1, Cristina Rotolo1, Cirino Botta2, Caterina Patti1, Alessandra Santoro1, Laura Di Noto1, Lucia Sbriglio1, Luca Castagna1 P741 INSTITUTIONAL THRESHOLD OF PREAPHERESIS PERIPHERAL BLOOD CD34+ CELL COUNT: A SINGLE-CENTER EXPERIENCE Yandy Marx Castillo Aleman 1,2, Carlos Agustin Villegas Valverde1, Antonio Alfonso Bencomo Hernandez1, Yendry Ventura Carmenate1,2, Fatema Mohammed Al Kaabi1,2, Shinnette Lumame2, Charisma Castelo2, Nameer Abdul Al-Saadawi1,2, Inas El-Najjar2, Mohamed Ibrahim Abu Haleeqa1,2, Jayakumar David Dennison2, Mansi Sachdev2, Fatma Abdou1, Shadi Shamat1, Anil Kumar Sarode1, Dina El Mouzain1,2, Hiba Mohamad Abdelrahman2, Rene Antonio Rivero Jimenez1 P742 APPLICATION OF GRANULOCYTE COLONY STIMULATING FACTOR DURING CYTAPHERESIS LEADS TO BETTER STEM CELL YIELDS Ivan Tonev 1, Stanislav Simeonov1, Milcho Mincheff1 P743 ADAPTATION OF THE VOLUMETRIC METHOD ON THE FACSLYRIC CYTOMETER FOR THE ENUMERATION OF CD34 + STEM CELLS Mohamed Brahimi 1, Amel Mihoubi2, Kamilia Amani2, Nawel Bounoua2, Aoudia Fatima2, Benaissa Mohamed Amine2, Amel Bendimerad2, Nabil Yafour1 P744 COMPARISON OF CE CHEMOTHERAPY PLUS G-CSF VS. G-CSF ALONE FOR AUTOLOGOUS HEMATOPOIETIC STEM CELL MOBILIZATION IN PATIENTS WITH MULTIPLE MYELOMA: A SINGLE CENTER RETROSPECTIVE ANALYSIS Martin Hildebrandt1, Veronika Dill 1, Philipp Blüm2, Anja Lindemann1, Alexander Biederstädt3, Marion Högner1, Katharina Götze1, Florian Basssermann1 P745 BLOOD TRANSFUSION NEEDS DURING PERIPHERAL BLOOD STEM CELL COLLECTION: A SINGLE-CENTER RETROSPECTIVE STUDY Liliana Fonseca1, Catarina Almeida2, Pedro Leite-Silva3, Ana Salselas 3, Susana Roncon3 P746 ENHANCING PRECISION IN PERIPHERAL BLOOD HAEMATOPOIETIC PROGENITOR CELL APHERESIS: A NOVEL APPROACH TO INTEGRATE COLLECTION EFFICIENCY VARIANCE FOR PRECISE CD34 + YIELD PREDICTION Nelson Chan1, Kai Wan 1, Sinju Thomas1, Sandra Loaiza1, Shab Uddin1, Edward Aina1, Emma Bray1, Kaivalya Kulkarni1, Romeo Valeza1, Ruth Emhazion1, Jane Apperley2,1, Edurado Olavarria2,1, Renuka Palanicawandar1 P747 STEM CELL MOBILIZATION IN MULTIPLE MYELOMA: DESCRIPTIVE ANALYSIS AND PREDICTIVE FACTORS FOR STEM CELL MOBILIZATION FAILURE Laura Pardo Gambarte 1, Isabel Iturrate Basarán1, Begoña Pérez de Camino Gaisse1, Soledad Sánchez Fernández1, Javier Cornago Navascués1, Laura Solán Blanco1, Jose Luis López Lorenzo1, Elham Askari1, Amalia Domingo González1, Elena Prieto Pareja1, Alberto Velasco Valdazo2, Rafael Martos Martínez3, Daniel Naya Errea4, Pilar Llamas Sillero1 P748 MOBILIZATION AND HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH MULTIPLE MYELOMA WITH DARATUMUMAB-BASED INDUCTION TREATMENS Isabel Izquierdo 1, Ana Gomez2, Ana Godoy1, Juan Gimeno1, Beatriz Martinez3, Vicente Carrasco4 P749 STEM CELL MOBILIZATION IN MULTIPLE MYELOMA PATIENTS: A SINGLE CENTER STUDY IN TUNISIA Imen Bellalah1, Karima Kacem 1, Malek Sayadi1, Dorra Jabeur1, Raoudha Mansouri1, Raihane Banlakhal1 P750 CHALLENGES OF HEMATOPOIETIC STEM CELL COLLECTION IN BIG PEDIATRIC TRANSPLANT CENTER: SINGLE CENTER EXPERIENCE Nara Stepanyan 1, Kirill Kirgizov1, Elena Machneva2, Ramil Fatkhullin1, Timur Aliev1, Irina Kostareva1, Nune Matinyan1, Vladimir Zhogov1, Marina Rubanskaya1, Olga Romantsova1, Natalia Batmanova1, Timur Valiev1, Tatiyana Gorbunova1, Vladimir Polyakov1, Svatlana Varfolomeeva1 P751 IMMUNOPHENOTYPING OF PERIPHERAL BLOOD MONONUCLEAR CELLS COLLECTED BY APHERESIS Carlos Agustin Villegas-Valverde 1, Antonio Bencomo-Hernandez1, Yandy Max Castillo-Aleman1, Fatrma Abdou1, Shadi Shamat1, Anil Kumar1, Yendry Ventura-Carmenate2, Fatema Al Kaabi3, Inas El-Najjar3, Shinnette Lumame3, Charisma Castelo3, Nameer Abdul Al Saadawi3, Dina El Mouzain3, Hiba Abdelrahman3, Jayakumar Dennison3, Mansi Sachdev3, Rene Antonio Rivero-Jimenez1 P752 REAPPRAISING CORD BLOOD AS A PRIORITY CELL SOURCE IN ALTERNATE DONOR TRANSPLANT AND IN DIFFICULT-TO-CURE PAEDIATRIC HEMATOLOGIC MALIGNANCY Abdul Wajid Moothedath 1, Denise Bonney1, Madeleine Powys1, Ramya Nataraj1, Omima Mustafa1, Srividhya Senthil1, Robert Wynn1 P753 CORTICOSTEROIDS IMPAIR HEMATOPOIETIC MICROENVIRONMENT VIA INDUCING SENESCENCE OF THE MESENCHYMAL STROMAL NICHE Yuqing Wang 1, Cong Wang1, Zheng Wang1, Xi Zhang1 P754 THE CORD BLOOD POST THAW CLINIC – HELPING YOU WORK OUT “WHAT GOOD LOOKS LIKE” Roger Horton 1, Ben Cash2, Kennedy Davies2, Irina Evseeva3, Phoebe Groves3, Sophie Macleod3, Alex Ross2, Claire Simpson1, Daniel Gibson1 P755 USE ON NON-CRYOPRESERVED HEMATOPOIETIC STEM CELLS FOR AUTOLOGOUS HSCT IN ADULTS - MULTICENTER STUDY, FROM THE IDWP AND TCWP EBMT – PRELIMINARY RESULTS Emilian Snarski 1, Gloria Tridello2, Lotus Wendel2, Zinaida Peric3, William Boreland4, Dina Averbuch5, Rafael de la Camara6 References

14 – 17 April, 2024 Hybrid Meeting

Copyright: Modified and published with permission from https://www.ebmt.org/annual-meeting

Sponsorship Statement: Publication of this supplement is sponsored by the European Society for Blood and Marrow Transplantation. All content was reviewed and approved by the EBMT Committee, which held full responsibility for the abstract selections.

19: Acute Leukaemia

P001 AUTOLOGOUS VERSUS HAPLOIDENTICAL DONOR STEM CELL TRANSPLANTATION FOR FAVORABLE-AND INTERMEDIATE-RISK ACUTE MYELOID LEUKEMIA IN FIRST REMISSION AND UNDETECTABLE MINIMAL RESIDUAL DISEASE: A MULTI-CENTER, PROSPECTIVE STUDY

Yiyang Ding1, Yuhua Ru1, Jinjin Zhu1, Xi Zhang2, Lin Liu3, Erlie Jiang4, He Huang5, Jishi Wang6, Jiong Hu7, Yanming Zhang8, Yajing Xu9, Mingzhen Yang10, Jia Chen 1, Depei Wu1

1National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China, 2Xinqiao Hospital, Third Military Medical University, Chongqing, China, 3The First Affiliated Hospital of Chongqing Medical University, Chongqing, China, 4Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China, 5Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 6Affiliated Hospital of Guizhou Medical University, Guiyang, China, 7Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 8Huai’an Hospital Affiliated to Xuzhou Medical College and Huai’an Second People’s Hospital, Huai’an, China, 9XiangYa Hospital, Central South University, Changsha, China, 10the First Affiliated Hospital of Anhui Medical University, Hefei, China

Background: In the absence of HLA-matched donor, HLA-haploidentical donor stem cell transplantation (haplo-SCT) and autologous SCT (ASCT) are both valid postremission therapy with well-documented effects in patients with favorable-and intermediate-risk acute myeloid leukemia (AML). Our previous retrospective study has suggested similar survival following ASCT compared to haplo-SCT for favorable- and intermediate-risk AML patients in the first complete remission (CR1). Hence, we designed a prospective, multi-center study to further confirm this conclusion.

Methods: A prospective study was conducted in 10 Chinese centers. The main inclusion criteria were: 1) adult patients >=18 years old; 2) diagnosis as AML with intermediate-risk according to ELN 2010 or 2017; 3) ASCT or haplo-SCT underwent between 2014-2019; 4) in CR1 and MRD negative before transplant. The Primary endpoint was overall survival (OS). Secondary endpoints were progression-free survival (PFS), cumulative incidence of relapse (CIR), treatment-related mortality (TRM), and graft-versus-host disease-free and relapse-free survival (GRFS).

Results: A total of 368 patients diagnosed with favorable-and intermediate-risk AML were included from 10 institutions between January 2014 and December 2019, in accordance with the European Leukemia Net criteria (Table 1). Of these, 184 patients underwent ASCT, and 184 underwent haplo-SCT. In the overall cohort analysis, the CIR in the ASCT group was offset by reduced TRM when compared with the haplo-SCT group, resulting in similar PFS (70.5% versus 74.9%, P = 0.880) and improved OS (83.0% versus 77.0%, P = 0.044). Furthermore, ASCT patients exhibited a higher GRFS (70.5% versus 57.4%, P<0.001) attributed to the absence of GVHD. The OS trend was accentuated in the favorable-risk cohort, while comparable OS was observed in the intermediate-risk cohort. Multivariate analyses indicated that haplo-SCT, advanced age, MLL-AF9 rearrangement, graft cell dosage, and hematopoietic engraftment time independently impacted outcomes.

Table 1. Patient Characteristics.

Auto (n = 184)

Haplo (n = 184)

P

Median age (range)

36 (18-64)

35 (18-60)

0.078

Gender

Male

104

95

0.346

Female

80

89

Risk stratification

Favorable

94

85

0.348

Intermediate

90

99

WBC at diagnosis (median, 109/L)

8.83

15.42

0.003

Abnormal karyotype at diagnosis

51

81

0.013

Abnormal molecular at diagnosis

FLT3-ITD

4

11

0.065

MLL-AF9

3

5

0.475

Induction courses

1

134

133

0.907

>1

50

51

Consolidation courses

≤2

67

139

<0.001

>2

117

45

HiDAC before transplant

91

18

<0.001

Conclusions: In conclusion, both ASCT and haplo-SCT proved to be viable approaches for the post-remission treatment of patients with favorable-and intermediate-risk AML. Notably, ASCT demonstrated superior outcomes in the favorable-risk cohort.

Clinical Trial Registry: Haplo-SCT vs ASCT With or Without Decitabine in AML CR1 - Full Text View - ClinicalTrials.gov (ClinicalTrails.gov Identifier: NCT02059720)

Disclosure: Nothing to declare

19: Acute Leukaemia

P002 OUTCOME OF ALLOGRAFTED ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS TREATED WITH THE PEDIATRIC-INSPIRED, MRD-ORIENTED GIMEMA LAL 1913 PROTOCOL IN THE REAL-LIFE: A CAMPUS ALL STUDY

Gianluca Cavallaro 1, Davide Lazzarotto2, Chiara Pavoni1, Francesca Valsecchi1, Cristina Papayannidis3, Marco Cerrano4, Sabina Chiaretti5, Nicola Fracchiolla6, Fabio Giglio7, Michelina Dargenio8, Monia Lunghi9, Silvia Imbergamo10, Maria Ilaria Del Principe11, Silvia Trappolini12, Monica Fumagalli13, Patrizia Zappasodi14, Prassede Salutari15, Mario Delia16, Crescenza Pasciolla17, Federico Mosna18, Barbara Scappini19, Fabio Forghieri20, Patrizia Chiusolo21, Cristina Skert22, Benedetta Cambò23, Marzia Defina24, Giuseppe Lanzarone25, Mauro Endri26, Massimiliano Bonifacio27, Carla Mazzone28, Lidia Santoro29, Antonino Mulè30, Valentina Mancini31, Paola Minetto32, Giorgia Battipaglia33, Alessandro Cignetti34, Lara Aprile35, Robin Foà5, Anna Candoni2,20, Federico Lussana1,36

1ASST Papa Giovanni XXIII, Bergamo, Italy, 2Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy, 3IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, 4AOU Città della Salute e della Scienza – Presidio Molinette, Torino, Italy, 5Sapienza University, Roma, Italy, 6IRCCS Ca’ Granda Ospedale Maggiore Policlinico di Milano, Milano, Italy, 7IRCCS Ospedale San Raffaele, Milano, Italy, 8Ospedale Vito Fazzi, Lecce, Italy, 9University of Eastern Piedmont, Novara, Italy, 10Azienda Ospedale Università di Padova, Padova, Italy, 11University of Rome Tor Vergata, Roma, Italy, 12Azienda Ospedaliero - Universitaria Ospedali Riuniti Umberto I, Ancona, Italy, 13Ospedale San Gerardo, Monza, Italy, 14Fondazione IRCCS Policlinico San Matteo, Pavia, Italy, 15Azienda USL di Pescara, Pescara, Italy, 16Azienda Ospedaliero-Universitaria Consorziale Policlinico di Bari, Bari, Italy, 17IRCCS Istituto Tumori Giovanni Paolo II, Bari, Italy, 18Azienda Sanitaria dell’Alto Adige, Bolzano, Italy, 19Azienda Ospedaliera Universitaria Careggi, Firenze, Italy, 20Azienda Ospedaliero Universitaria di Modena, Modena, Italy, 21Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy, 22Ospedale dell’Angelo, Venezia, Italy, 23University of Parma, Parma, Italy, 24Azienda Ospedaliero Universitaria Senese, Siena, Italy, 25A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy, 26Ospedale Cà Foncello, Treviso, Italy, 27Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy, 28St. Eugenio Hospital, Roma, Italy, 29Azienda Ospedaliera San Giuseppe Moscati, Avellino, Italy, 30A.O. Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy, 31ASST Grande Ospedale Metropolitano Niguarda, Milano, Italy, 32IRCCS Ospedale Policlinico San Martino, Genova, Italy, 33Azienda Ospedaliero-Universitaria Federico II, Napoli, Italy, 34Azienda Ospedaliera Ordine Mauriziano, Torino, Italy, 35Ospedale San Giuseppe Moscati, Taranto, Italy, 36University of Milan, Milano, Italy

Background: The GIMEMA LAL 1913 was a pediatric-inspired and minimal residual disease (MRD)-oriented phase 2 chemotherapy protocol that included peg-asparaginase for the frontline treatment of adult Philadelphia-negative acute lymphoblastic leukemia (Ph- ALL) patients (Bassan et al, Blood Advances, 2023). This protocol represents the standard treatment for adult Ph- ALL in Italy. The outcome of patients treated with the same protocol in the real-life setting and who underwent an allogeneic stem cell transplant (SCT) was the aim of this study.

Methods: Within the framework of the Campus ALL network in Italy, we analyzed the post-transplant data of newly diagnosed adult Ph- ALL patients treated according to the LAL 1913 protocol and who underwent a SCT between August 2016 and January 2023 in 35 Italian centers. Patients in 1st remission (CR1) were considered eligible for a SCT as consolidation based on the joint assessment of the disease risk profile at diagnosis (high white blood cell, immunophenotype or adverse cytogenetics/molecular biology) and post-consolidation MRD status. We also included relapsed patients in 2nd CR (CR2). Endpoints were the 2-year overall survival (OS), disease-free survival (DFS), non-relapse mortality (NRM) and cumulative incidence of relapse (CIR). Outcomes were analyzed from the day of SCT.

Results: Among 322 consecutive newly diagnosed Ph- ALL, 201 patients who underwent a SCT were included in this analysis (Table 1). The median age at diagnosis was 38 years (18-66). Median follow-up was 28 months. Patients allografted in CR1 were associated with a significantly superior 2-year OS (77% vs 32%, p<0.001), DFS (67% vs 35%, p = 0.003) and CIR (24% vs 47%, p = 0.006) compared to patients transplanted in CR2. Pre-transplant MRD negativity was strongly associated with a superior OS (81% vs 50%, p<0.001), DFS (71% vs 44%, p = 0.003) and CIR (21% vs 43%, p = 0.006). MRD negativity also impacted favorably on the prognosis of patients transplanted in 2nd CR in terms of OS and disease-free survival (Fig. 1). The outcome was similar between B- and T-lineage ALL, patients’ age (< or > 55 years) and baseline risk class. A myeloablative conditioning was adopted in 156 (79%) patients and total-body irradiation (TBI) in 125 (66%). There was no significant difference when using TBI 800 cGy vs TBI 1200 cGy and TBI-fludarabine vs TBI-alkylating agents. The use of ATG in sibling and unrelated donors did not affect CIR. The 2-year NRM was 11% and it was not significantly affected by pre- and post-transplant factors.

Table 1.

Patients’ characteristics

N = 201

Age at diagnosis, n(%)

<55

163 (81.1)

>=55

38 (18.9)

Cell lineage, n(%)

B-ALL

97 (48.3)

T-ALL

104 (51.7)

Baseline risk, n(%)

Standard Risk

55 (27.4)

High Risk

25 (12.4)

Very High Risk

121 (60.2)

Disease status at transplant, n(%)

CR1

171 (85.1)

CR2

29 (14.4)

Pre transplant MRD, n(%)

Negative

130 (68.8)

Positive

58 (30.7)

Donor, n(%)

Sibling

61 (30.7)

Matched unrelated donor

68 (34.2)

Aploidentical

37 (18.6)

Mismatched unrelated donor

33 (16.6)

Conditioning regimen, n(%)

Myeloablative

156 (78.8)

Reduced intensity

42 (21.2)

TBI, n(%)

No

64 (33.9)

Yes

125 (66.1)

Figure 1: 2-years DFS

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (1)

Conclusions: Our study on a series of patients treated with a modern therapeutic strategy confirms the higher risk of relapse in patients with pre-transplant MRD positivity. Given the availability of pre-transplant immunotherapies, all efforts should be made to eradicate MRD prior to undergoing the SCT, or with post-transplant pre-emptive treatment. An advantage in survival was observed when transplant was performed in CR1 compared to CR2, highlighting the importance of an early identification of high-risk patients candidate to transplant consolidation. Importantly, a significant improvement in NRM outcome was recorded.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P003 FREQUENCY AND IMPACT OF SOMATIC MUTATIONS ON OUTCOMES OF ACUTE MYELOID LEUKEMIA PATIENTS RECEIVING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION: FROM THE EBMT ACUTE LEUKEMIA WORKING PARTY

Ali Bazarbachi 1, Jacques-Emmanuel Galimard2, Myriam Labopin2, Iman Abou Dalle1, Jaime Sanz3, Huang He4, Jiri Mayer5, Carlos Solano6, Celestine Simand7, Laimonas Griskevicius8, Johan Maertens9, Maija Itäla-Remes10, Ain Kaare11, Maria-Pilar Gallego-Hernanz12, Gesine Bug13, Josep-Maria Ribera14, Alain Gadisseur15, Christoph Schmid16, Mi Kwon17, Xavier Poiré18, Paola Coccia19, Manuel Jurado Chacón20, Frédéric Baron21, Eolia Brissot22, Arnon Nagler23, Fabio Ciceri24, Mohamad Mohty25

1American University of Beirut Medical Center, Beirut, Lebanon, 2EBMT Statistical Unit, Sorbonne University, Saint-Antoine Hospital, Paris, France, 3University Hospital La Fe, Valencia, Spain, 4First Affiliated Hospital of Zhejiang University School of Medicine, Kunming, China, 5University Hospital Brno, Brno, Czech Republic, 6Hospital Clínico Universitario. University of Valencia, Valencia, Spain, 7ICANS, Strasbourg, France, 8Hôpitaux Universitaires Strasbourg, Strasbourg, France, 9University Hospital Gasthuisberg, Leuven, Belgium, 10Turku University Hospital, Turku, Turkey, 11Tartu University Hospital, Tartu, Estonia, 12Hopital La Miletrie, Poitiers, France, 13Goethe-Universitaet, Frankfurt Main, Germany, 14ICO-Hospital Universitari Germans Trias i Pujol, Josep Carreras Research Institute, Badalona, Spain, 15Antwerp University Hospital (UZA), Antwerp E, Belgium, 16University Hospital and Medical Faculty, University of Augsburg, Augsburg, Germany, 17Hospital General Universitario Gregorio Marañón, Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain, 18Cliniques Universitaires St. Luc, Brussels, Belgium, 19Azienda Ospedali Riuniti di Ancona, Ancona, Italy, 20Hospital Univ. Virgen de las Nieves, Granada, Grenada, 21University of Liege, Liege, Belgium, 22Sorbonne University, Paris, France, 23Hematology Division, Chaim Sheba Medical Center, Tel-Hashomer, Israel, 24IRCCS Ospedale San Raffaele, Milano, Italy, 25Sorbonne University, Saint-Antoine Hospital, Paris, France

Background: Acute myeloid leukemia (AML) is a very heterogeneous hematological malignancy, which includes numerous genetically defined subsets. In the context of allogeneic hematopoietic cell transplantation (allo-HCT), the frequency and prognostic value of different gene-gene interactions has not been studied and may differ from that of patients treated with chemotherapy alone.

Methods: This is a registry-based analysis from the EBMT with the approval of the EBMT Acute Leukemia Working Party. Adult patients aged more than 18 years with a diagnosis of AML who received an allo-HCT between 2013-2022, with an available genetic profile determined at diagnosis by next generation sequencing (NGS) were included.

Results: We identified 836 allografted AML patients who had NGS performed at diagnosis. Most of these patients had de novo AML (88%), with a median age of 53 years (range: 18-78 years). Karyotype was favorable in 7%, intermediate in 69% and adverse in 24% of patients. At transplant, 74% of patients were in first remission (CR1) and 13% in CR2. The most frequent detectable mutations by frequency were DNMT3A (33%), FLT3 (29%), NPM1 (29%), TET2 (28%), NRAS (23%), RUNX1 (22%), WT1 (22%), BCOR (19%), ASXL1 (17%), IDH2 (17%), IDH1 (15%), SRSF2 (13%), STAG2 (12%), CEBPA (11%), TP53 (10%), KRAS (10%), and PTPN11 (10%). By multiple correspondence analysis, two independent groups of co-occurring mutations were identified, the first group included DNMT3A, NPM1 and FLT3, the second group included ASXL1, SRSF2 and RUNX1. Outcome analysis was performed on the subset of 298 patients allografted in CR1 with available data for DNMT3A, NPM1, FLT3, ASXL1, SRSF2 and RUNX1. Most of these patients had de novo AML (90%), with a median age of 53 years (range: 19-75 years). Seventy percent had intermediate-risk cytogenetics, while 27% were adverse-risk. Median follow up was 2.5 years. When outcome analysis was performed according to the presence of single mutations, none of the six mutations significantly affected RI or LFS. The 2-year OS was positively affected by the presence of NPM1 mutation (78% vs 66%; p = 0.02) and FLT3 (79% vs 65%, p = 0.01) but not significantly affected by the other 4 mutations. When mutations were investigated in groups, the 2-year RI, LFS and OS were 24%, 70% and 78%, respectively, for patients with NPM1 mutation regardless of other co-mutations, 35%, 56% and 69% for patients with FLT3-ITD and/or DNMT3A mutation, wild type NPM1, regardless of other co-mutations, 17%, 70% and 74% for patients with RUNX1 and/or ASXL1 and/or SRSF2 mutation without FLT3-ITD and with wild type NPM1 and wild type DNMT3A, and 20%, 56% and 61% for patients with all six genes unmutated.

Conclusions: NGS at diagnosis can be extremely useful in risk stratification of AML patients undergoing allo-HSCT, potentially allowing adequate post-transplant interventions. Notably, the 2-year LFS of 70% for patients harboring RUNX1 and/or ASXL1 and/or SRSF2 mutation indicates that allo-HSCT can overcome the adverse risk associated with these somatic mutations at diagnosis.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P004 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR ELDERLY ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS: A STUDY FROM THE SOCIÉTÉ FRANCOPHONE DE GREFFE DE MOELLE ET THÉRAPIE CELLULAIRE (SFGM-TC)

Yves Chalandon 1, Raynier Devillier2, Ariane Boumendil3, Stephanie Nguyen4, Claude-Eric Bulabois5, Patrice Ceballos6, Eolia Brissot7, Marie-Thérèse Rubio8, Hélène Labussière-Wallet9, Johan Maertens10, Patrice Chevallier11, Natacha Maillard12, Xavier Poiré13, Cristina Castilla-Llorente14, Yves Beguin15, Jérôme Cornillon16, Sébastien Maury17, Tony Marchand18, Etienne Daguindau19, Jacques-Olivier Bay20, Pascal Turlure21, Amandine Charbonnier22, Anne-Lise Menard23, Karin Bilger24, Gaelle Guillerm25, Sylvie François26, Ali Bazarbachi27, Sylvain Chantepie28, Philippe Lewalle29, Ambroise Marçais30, Michael Loschi31, Malek Benakli32, Paul Chauvet33, Edouard Forcade34, Anne Huynh35, Marie Robin36, Stavroula Masouridi-Levrat1

1Hôpitaux Universitaire Genève (HUG) and Faculty of Medicine, Geneva, Switzerland, 2Institut Paoli-Calmettes, Marseille, France, 3SFGM-TC, Paris, France, 4Hôpital de la Pitié Salpêtrière, AP-HP, Paris, France, 5CHU Grenoble-Alpes, Grenoble, France, 6CHU de Montpellier, Montpellier, France, 7Hôpital Saint-Antoine, AP-HP, Paris, France, 8CHRU de Nancy-Brabois, Nancy, France, 9Hôpital Lyon Sud, Lyon, France, 10UZ Leuven, Leuven, Belgium, 11CHU Nantes, Nantes, France, 12CHU Poitiers, Poitiers, France, 13Cliniques Universitaires Saint-Luc, Brussels, Belgium, 14Gustave Roussy Cancer Campus, Villejuif, France, 15CHU de Liège, Liège, Belgium, 16CHU St-Etienne, Saint-Etienne, France, 17Hôpital Henri-Mondor, AP-HP, Créteil, France, 18CHU Rennes, Rennes, France, 19CHU de Besançon, Besançon, France, 20CHU de Clermont-Ferrand, Clermont-Ferrand, France, 21Limoges University Hospital, Limoges, France, 22CHU Amiens, Amiens, France, 23Centre Henri Becquerel, Rouen, France, 24CHRU de Strasbourg, Strasbourg, France, 25CHRU de Brest, Brest, France, 26CHU Angers, Angers, France, 27American University of Beirut, Beirut, Lebanon, 28CHU de Caen, Caen, France, 29Hôpital Universitaire de Bruxelles (HUB), Institut Jules Bordet, Brussels, Belgium, 30Hôpital Necker, AP-HP, Paris, France, 31CHU de Nice, Nice, France, 32CPMC d’Alger, Alger, Algeria, 33CHU de Lille, Université de Lille, Lille, France, 34CHU Bordeaux, Bordeaux, France, 35CHU Toulouse, Toulouse, France, 36Hôpital Saint-Louis, AP-HP, Paris, France

Background: There are very scarce data regarding the outcome of elderly patients with ALL who received alloHSCT as consolidation therapy in CR. Thus, the optimal conditioning regimen still need to be determined. We here present the outcome of ALL patients older than 59 years from the SFGM-TC.

Methods: The primary outcome of this registry study was OS. Secondary outcomes were PFS, NRM, RI, aGvHD grade II-IV, cGvHD, engraftment and GRFS. Competing risks analyses were performed to analyze NRM, aGvHD grade II-IV, cGvHD and neutrophil engraftment. Univariable analyses were performed using the log-rang test for OS and PFS, while Gray’s test was used for CI. Multivariable analyses were performed using the Cox proportional hazards regression model including age, ALL subtype, time from diagnosis to alloHSCT, disease status at alloHSCT, donor to patient CMV status, donor to patient sex, ATG use, MAC, TBI use.

Results: A total of 316 patients ≥ 60 years old transplanted for ALL from 2012 to 2022 in 36 participating centers were included. Patient’s characteristics are described in Table 1.

With a median follow up of 34.5 months (IQR 29.5-38.8), 3-year OS was 46% (95% CI 40-53%) with disease status at transplant impacting negatively OS, 53% in CR1, 32% in CR2, 29% in advanced disease, p = 0.002 and ALL subtype, 59% in Ph+ ALL, 40% in Ph- ALL, 34% in T-ALL, 20% in other/NA, p<0.001.

3-year PFS was 41% (95% CI 35-48%) with disease status at transplant impacting negatively, 49% in CR1, 26% in CR2, 22% in advanced disease, p<0.001, ALL subtype, 51% in Ph+ ALL, 41% in Ph- ALL, 21% in T-ALL, 21% in other/NA, p<0.001, year of HSCT worse < 2018, p = 0.007, CMV -/- worse, p = 0.033, absence of TBI worse, p = 0.018. 3-year NRM was 23% (95% CI 18-28%). 3-year RI was 36% (95% CI 31-42%) with disease status at transplant impacting negatively, 29% in CR1, 50% in CR2, 56 in advanced disease, p = 0.0042, ALL subtype, 26% in Ph+ ALL, 43% in Ph- ALL, 57% in T-ALL, 42% in other/NA, p = 0.0064, year of HSCT worse < 2018, p = 0.0216, CMV -/- worse, p<0.001, absence of TBI worse, p = 0.0069, MRD worse, p = 0.0111. 3-year GRFS was 30% (95% CI 25-37%) with disease status at transplant impacting negatively, 35% in CR1, 22% in CR2, 23% in advanced disease, p = 0.029, ALL subtype, 37% in Ph+ ALL, 33% in Ph- ALL, 15% in T-ALL, 17% in other/NA, p = 0.0029, year of HSCT worse < 2018, p<0.001.

CI of aGVHD grade II-IV was 33% (95% CI 28-38%), grade III-IV 11% (95% CI 8-15%), cGVHD 35% (95% CI30-41%), extensive cGVHD 21% (95% CI 16-26%).

Multivariable analyses confirmed a worse OS and PFS for advanced disease, with a HR of 1.79 (95% CI 1.22-2.64), p = 0.00322 and ALL subtype with a HR for other than Ph+ ALL of 1.99 (95%CI 1.42-2.79).

Characteristics

N = 316

Age at HSCT median (IQR)

63.82 (61.8-66.5)

Age at diagnosis median (IQR)

63.02 (61-65.4)

Time from diagnosis to HSCT median (IQR) months

7.05 (5.7-10.3)

Year of HSCT median (IQR)

2017.5 (2015-2020)

ALL subtype

B-ALL Ph-

81 (25.9%)

B-ALL Ph+

156 (49.8%)

T-ALL

42 (13.4%)

Other/unknown

37 (11.7%)

Patient’s sex

Female

156 (49.4%)

Male

160 (50.6%)

Disease status at HSCT

CR1

224 (70.9%)

CR2 and +

74 (23.4%)

Not in CR

18 (5.7%)

Molecular CR

Yes

121 (38.3%)

No

79 (25%)

Missing

116 (36.7%)

Type of donor

Haploidentical

56 (17.7%)

MSD

83 (26.3%)

MUD

142 (44.9%)

MMUD

23 (7.2%)

Missing

12 (3.8%)

Source of stem cell

BM

22 (7%)

PBSC

294 (93%)

Donor to patient’s sex

Female to male

44 (14%)

Other

271 (86%)

Missing

1

CMV donor/patient

Neg/Neg

84 (26.8%)

Neg/Pos

93 (29.7%)

Pos/Neg

24 (7.7%)

Pos/Pos

112 (35.8%)

Missing

3

ATG

Yes

220 (69.6)

No

96 (30.4%)

Conditioning

RIC

204 (64.6%)

MAC

112 (35.4%)

TBI

No

203 (64.2%)

Yes

113 (35.8%)

TBI & Dose

No TBI

203 (64.4%)

TBI < 8 Gy

43 (13.7%)

TBI 8 Gy

69 (21.9%)

Missing

1

Disease Risk Index

Intermediate

224 (71.3%)

High

72 (22.9%)

Very high

18 (5.7%)

Missing

2

Comorbidity index (Sorror)

>=3

76 (32.5%)

1-2

53 (22.6%)

105 (44.9%)

Missing

82

Conclusions: This study suggests that alloHSCT is a reasonable option for elderly ALL patients without any impact of age but advanced disease and ALL subtype other than Ph+ ALL negatively influenced the outcome.

Disclosure: YC: Y.C. has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution.

EF: EF received funding for congress participation from Alexion, Gilead, Jazz, MSD, Novartis; and also honoraria for boards and speaker bureau participation from Alexion, GSK, Novartis, Gilead, Astellas.

ML: honorari : Alexion, Astra Zeneca, BMS Celgene, Gilead, GSK, Jazz, Kartos, Medac, MSD, Novartis, Pfizer, Sanofi, Sobi, Telios

TM: Consulting fees : Servier, Jazz Pharmaceutical, Sobi, Astellas. Travel support from Servier, Jazz Pharmaceutical

AH : AH has received consulting fees advisory board from Jazz, Pfizer, Servier, Novartis, Astellas; Travel support from Medac, Jazz, Neovii all via the Institution.

All the others do not have any conflict of interest.

19: Acute Leukaemia

P005 ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION FOR OLDER PATIENTS WITH PHILADELPHIA-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA. A SURVEY BY THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT

Sebastian Giebel 1, Myriam Labopin2, Ryszard Swoboda1, Didier Blaise3, Ibrahim Yakoub-Agha4, Stephanie Nguyen5, Eva Maria Wagner-Drouet6, Cristina Castilla-Llorente7, Panagiotis Kottaridis8, Thomas Schroeder9, Renato Fanin10, Jakob Passweg11, Jurjen Versluis12, Charles Crawley13, Ludovic Gabellier14, Stephan Mielke15, Xavier Poiré16, Erfan Nur17, Carlos Pinho Vaz18, Matthias Eder19, Riccardo Saccardi20, Peter Dreger21, Zinaida Peric22, Mohamad Mohty2, Fabio Ciceri23

1Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland, 2EBMT Unit, Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), AP-HP, Saint Antoine Hospital, Paris, France, 3Programme de Transplantation & Therapie Cellulaire, Marseille, France, 4CHU de Lille, University of Lille, INSERM U1286, Infinite, Lille, France, 5Universite Paris IV, Hopital la Pitié-Salpêtrière, Paris, France, 6University Medical Center Mainz, Meinz, Germany, 7Gustave Roussy Cancer Campus, Villejuif, France, 8University College London Hospital, London, United Kingdom, 9University Hospital Essen, Essen, Germany, 10Azienda Ospedaliero Universitaria di Udine, Udine, Italy, 11University Hospital | Basel, Basel, Switzerland, 12Erasmus University Medical Center Cancer Institute, Rotterdam, Netherlands, 13Addenbrookes Hospital Cambridge, Cambridge, United Kingdom, 14University Hospital of Montpellier, Montpellier, France, 15Karolinska University Hospital, Stockholm, Sweden, 16Cliniques Universitaires St. Luc, Brussels, Belgium, 17Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, Netherlands, 18Inst. Português de Oncologia do Porto, Porto, Portugal, 19Hannover Medical School, Hannover, Germany, 20Azienda Ospedaliera Universitaria Careggi, Firenze, Italy, 21University of Heidelberg, Heidelberg, Germany, 22University Hospital Center Rijeka and School of Medicine, University of Rijeka, Rijeka, Croatia, 23Ospedale San Raffaele s.r.l., Haematology and BMT, Milano, Italy

Background: Tyrosine kinase inhibitors (TKIs) with or without chemotherapy, followed by allogeneic hematopoietic cell transplantation (allo-HCT) are the standard of care for patients with Philadelphia-positive acute lymphoblastic leukemia (Ph+ ALL). In older patients, intensity of the conditioning regimens is frequently reduced, which may result in increased risk of relapse. However, introduction of alternative approaches using 2nd or 3rd generation TKIs in combination with blinatumomab may restrict the role of allo-HCT. The goal of this study was to evaluate results of recent allo-HCTs for patients aged ≥55 years, and to identify prognostic factors.

Methods: This was a retrospective analysis based on data from the EBMT registry, including 566 patients with Ph+ ALL treated with allo-HCT in first complete remission between the years 2016 and 2022.

Results: Median recipient age was 60 (range, 55-76) years. Allo-HCT was performed using either a matched sibling (n = 138, 24%), unrelated (n = 343, 61%) or haploidentical (n = 85, 15%) donor. Conditioning regimen was myeloablative in 47% of cases and based on total body irradiation (TBI) in 49% of patients. Status of measurable residual disease was reported as negative in 59.5% of patients.

The probability of overall survival (OS) and leukemia-free survival (LFS) at 2 years was 71% and 59.5%, respectively. The incidence of relapse (RI) and non-relapse mortality (NRM) was 18% and 22.5%, respectively. The rate of graft-versus-host disease (GVHD)-free, relapse-free survival (GRFS) was 50%. The incidence of grade 3-4 acute GVHD was 9% while that of extensive chronic GVHD it was 15%.

In univariate analysis the use of TBI-based as compared to chemotherapy-based regimens was associated with reduced RI (15% vs. 20.5%, p = 0.04), improved LFS (67% vs. 53%, p = 0.002) and OS (77% vs. 66%, p = 0.01), as well as a tendency toward reduced incidence of NRM (18% vs. 27%, p = 0.1). On the other hand, it was also associated with increased incidence of both overall and extensive chronic GVHD (46% vs. 27.5%, p = 0.001 and 21% vs. 10%, p = 0.001, respectively). In a multivariant model, the use of TBI was the only factor affecting outcomes, being associated with reduced risk of NRM (hazard ratio, [HR] = 0.46, p = 0.004), improved LFS (HR = 0.53, p<0.001) and OS (HR = 0.47, p<0.001) as well as increased risk of chronic GVHD (HR = 1.68, p = 0.009) and extensive chronic GVHD (HR = 1.84, p = 0.04).

Conclusions: Based on this large-scale analysis we conclude that results of allo-HCT for patients with Ph+ ALL aged ≥55 years in the last few years are very encouraging with approximately 60% LFS at 2 years. TBI-based conditioning appears preferable in this patient population. Our data can serve as reference for results of prospective trials.

Clinical Trial Registry: Not applicable

Disclosure: SG received honoraria from Angelini and Novartis

19: Acute Leukaemia

P006 OUTCOME OF PATIENTS WITH IDH-MUTATED AML FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION

Thomas Schroeder 1, Sarah Flossdorf1, Caroline Pabst2, Michael Stadler3, Johannes Schetelig4, Claudia Wehr5, Claudia Schuh6, Matthias Stelljes7, Elisa Sala8, Andreas Burchert9, Jennifer Kaivers1, Christian Reinhardt1, Nicolaus Kroeger10, Katharina Fleischhauer1, Christina Rautenberg1

1University Hospital Essen, Essen, Germany, 2University Hospital Heidelberg, Heidelberg, Germany, 3MHH Hannover, Hannover, Germany, 4University Hospital Dresden, Dresden, Germany, 5University Hospital Freiburg, Freiburg, Germany, 6DRST, Ulm, Germany, 7University Hospital Muenster, Muenster, Germany, 8University Hospital Ulm, Ulm, Germany, 9University Hospital Marburg, Marburg, Germany, 10University Hospital Hamburg, Hamburg, Germany

Background: Somatic mutations in the isocitrate dehydrogenase 1 and 2 genes (IDH1 and IDH2) are found in about 15% to 20% of patients with acute myeloid leukemia (AML). While the prognostic impact of these mutations is still controversial, IDH inhibitors have been developed and introduced as targeted treatment for patients with IDH-mutated AML. They are currently under investigation also as maintenance therapy after allogeneic transplantation (allo-SCT), although outcomes of IDH1- and IDH2-mutated (IDHmut) AML patients after allo-SCT have not been well described.

Methods: To address this, we retrospectively analyzed the outcome of IDHmut patients, who were allografted between 2014 and 2021, had available data collected within the German Registry for hematopoetic Stem Cell Transplantation and cellular therapy (DRST) and a follow-up of at least 6 months. Overall (OS) and event-free survival (EFS) were estimated by Kaplan–Meier method and logrank tests were applied for univariable comparisons. Relapse incidence and non-relapse mortality (NRM) were considered as competing risks and calculated using cumulative incidence (CI) estimates employing Gray test for univariable comparisons.

Results: Overall, 356 IDH-mutated AML patients (IDH1 n = 142 40%, IDH2 n = 214 60%) with a median age of 60 (18 to 76) received a first allo-SCT from a related (30%) or unrelated (70%) donor following myeloablative (50%) or reduced intensity (50%) conditioning. According to ELN 2016 genetic risk stratification 21% belonged to the favorable, 42% to intermediate and 27% to adverse category (missing 10%), with 71% of patients being in CR at transplant. Of 262 patients with available information 10 (4%) had received an IDH inhibitor outside clinical trials prior transplantation. After a median follow-up of 24 months, IDH1mut and IDH2mut patients had similar estimated 3-year probabilities of overall survival (78% vs 70%, p = .64). Non-relapse mortlity was comparable (10% vs 14%, p= .48). Three-year probabilities of relapse were numerically higher for IDH1mut compared with IHD2mut patients (34% vs 24%, p= .18) without reaching statistical significance, and concomitantly a numerically lower 3-year event-free survival (56% vs 62% respectively; p= .14).

Conclusions: Taken together, these data from a large multicenter cohort provide benchmarks for analysis and interpretation of results emerging from clinical trials evaluating maintenance IDH1 and IDH2 inhibitor therapy for AML patients after allo-SCT

Clinical Trial Registry: not applicable

Disclosure: Thomas Schroeder: Advisory Boards:BMS; lecture fees BMS, research funding: BMS

Elisa Sala: honoraria for consultancy or travel support from Gilead, Novartis, BMS, Jazz, Neovii, Therakos/Mallinckrodt, MSD, Priothera.

Johannes Schetelig: Advisory Boards: Abbvie, AstraZeneca, BeiGene, BMS, Janssen, and MSD und Lecture Fees: Astellas, AstraZeneca, BeiGene, BMS, Novartis, Eurocept, Medac and Janssen

Claudia Wehr: Travel support/honoraria from Takeda, Jazz Pharmaceuticals.

Christian Reinhardt: consulting and lecture fees from AbbVie, AstraZeneca, Roche, Janssen-Cilag, Novartis, Vertex and Merck. H.C.R. received research funding from Gilead and AstraZeneca. H.C.R. is a co-founder of CDL Therapeutics GmbH.

19: Acute Leukaemia

P007 CD22-TARGETED IMMUNOTHERAPY FOR ADULT PATIENTS WITH RELAPSED OR REFRACTORY B-ALL WHO PREVIOUSLY EXPOSED TO CD19-TARGETED IMMUNOTHERAPY

Mingming Zhang1,2, Yongxian Hu1,2, Peihua Lu3,4, Liang Huang5, Shan Fu1,2, Jingjing Feng1,2, Ruimin Hong1,2, Alex H. Chang6, He Huang 1,2

1Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Hangzhou, China, 3Hebei Yanda Lu Daopei Hospital, Langfang, China, 4Lu Daopei Institute of Hematology, Beijing, China, 5Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 6Shanghai YaKe Biotechnology, Shanghai, China

Background: CD19-targeted immunotherapy effectively induce remission in relapsed or refractory (r/r) B-cell acute lymphoblastic leukemia (B-ALL). Patients who fail or relapse after CD19-targeted immunotherapy have an extremely poor prognosis, and immunotherapy targeting CD22 is one of the few possible therapeutic options, but there is currently limited data.

Methods: We conducted a multicenter, retrospective study of CD22-targeted immunotherapy for adult patients with r/r B-ALL who previously exposed to CD19-targeted immunotherapy. Primary objective was complete remission (CR) rate at 1 month.

Results: A total of 30 patients were included, and the median age was 32 (18-67) years old. Four (13.3%) patients were Ph-positive, 10 (33.3%) patients were primary refractory and 10 (33.3%) patients relapsed after allogeneic stem cell transplantation. Six (20.0%) patients were exposed to blinatumomab only, 20 (66.7%) patients were exposed to CD19 CAR-T cell therapy only, and 4 (13.3%) patients were exposed to both. 20 (66.7%) patients received CD22 CAR-T cell therapy, and 10 (33.3%) patients received inotuzumab ozogamicin. In the target expression analysis before CD22-targeted immunotherapy, all patients had CD22-positive expression except one patient was CD22-dim. As for the expression of CD19, 13 (43.3%) patients were CD19-positive, 12 (40.0%) patients were CD19-negative, and 5 (16.7%) patients were CD19-dim or partially expression. Totally 17 (56.7%) patients achieved CR at 1 month after CD22-targeted immunotherapy. The CR rate was 55.0% (11/20) in the CD22 CAR-T group, and 60.0% (6/10) in the inotuzumab ozogamicin group. There was no significant difference between the two groups (P = 0.79). The CR rate for patients exposed to both blinatumomab and CD19 CAR-T seemed to be low and was 25% (1/4). Other factors including primary refractory, post-transplantation relapse, and CD19 expression status had no effect on CR rate. Of the 17 patients who achieved CR, 10 had subsequent relapses, 2 lost to follow-up, 1 died of a post-CAR-T infection, and 1 died of complications of transplantation. As of December 15, 2023, excluding the 2 patients lost to follow-up, only 7 patients remained in MRD negative CR.

Conclusions: CD22-targeted immunotherapy is an effective treatment option for patients with r/r B-ALL who previously exposed to CD19-targeted immunotherapy. However, the relapse rate post CD22-targeted immunotherapy remains high. Therefore, new strategies are still needed to improve the prognosis of this group of patients.

Disclosure: Alex H. Chang is an employee of Shanghai YaKe Biotechnology Ltd., the other authors declare no conflict of interest.

19: Acute Leukaemia

P008 COMPARISON OF THE PROGNOSTIC IMPACT BETWEEN ELN2022 AND ELN2017 RISK CLASSIFICATION IN ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS WITH AML

Weihao Chen1, Yeqian Zhao1,2,3,4, Jimin Shi1,2,3,4, Yi Luo1,2,3,4, Jian Yu1,2,3,4, Yamin Tan5,6, Xiaoyu Lai1,2,3,4, Lizhen Liu1,2,3,4, Huarui Fu1,2,3,4, Yishan Ye1,2,3,4, Luxin Yang1,2,3,4, Congxiao Zhang1,2,3,4, He Huang1,2,3,4, Yanmin Zhao 1,2,3,4

1Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China, 3Institute of Hematology, Zhejiang University, Hangzhou, China, 4Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China, 5Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China, 6Institute of Cancer and Basic Medicine(IBMC), Chinese Academy of Sciences, Hangzhou, China

Background: Risk classification based on genetic aberrations plays a pivotal role in predicting prognosis and guiding treatment decisions in patients with acute myeloid leukemia (AML). In 2022, the European LeukemiaNet (ELN) issued an updated recommendations for diagnosis and risk classification, building upon the ELN2017 guidelines. While many studies have validated and compared the two versions of ELN risk classifications in chemotherapy cohorts, there remains a scarcity of research comparing the prognostic impact of ELN2017 and ELN2022 criteria in the context of allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Methods: We conducted a retrospective study which compared the the prognostic impact between the 2017 and 2022 ELN risk classifications in a real-life cohort of de novo AML patients who underwent allo-HSCT (n = 641) in our center from January 2015 to January 2022. We enrolled patients who had sufficient clinical and genetic information available for ELN risk classification. Patients diagnosed with acute promyelocytic leukemia or whose who received a second transplantation were excluded. Gene mutations or fusion genes detection were performed using polymerase chain reaction before 2017 and next-generation sequencing after 2017.

Results: 623 patients were included in this retrospective study, with a median follow-up time of 2.8 years after allo-HSCT. The median age at allo-HSCT was 40 years (range 10-67). According to ELN2017 risk categories, 32% (n = 202) patients at diagnosis were classified into ELN2017 favorable group, 43% (n = 264) into ELN2017 intermediate group, and 25% (n = 157) into ELN2017 adverse group. In ELN2022, the classification was 32% (n = 202) in favorable, 45% (n = 277) in intermediate, and 23% (n = 144) in adverse group. Patients in adverse group had significantly lower RFS (relapse-free survival) and OS (overall survival) after allo-HSCT compared to those in favorable and intermediate group, regardless in ELN2017 or ELN2022. Nonetheless, RFS of ELN2022 intermediate patients was worse than favorable patients (68.8 vs 82.3%, P = 0.015), and we did not obverse this statistic difference in ELN2017. The c-statistic (area under the curves [AUC]) from ELN2022 for RFS after allo-HSCT was not obviously better compared to ELN2017 (AUCELN22 = 0.637 vs AUCELN17 = 0.616, P = 0.096). The univariate analysis indicated that adverse ELN classification, NR status at allo-HSCT, and CR patients with MRD positivity at allo-HSCT might be associated with worse RFS and OS after allo-HSCT. Therefore, we further refined the ELN2022 system into four categories by incorporating the remission and MRD status at allo-HSCT (favorable: ELN2022 favorable with MRDneg, intermediate: ELN2022 favorable with MRDpos and ELN2022 intermediate with MRDneg, adverse: ELN2022 intermediate with MRDpos and ELN2022 adverse with MRDneg, and very adverse: NR patients at allo-HSCT and ELN2022 adverse with MRDpos). The refined ELN risk classification showed that there was statistical significance between groups for RFS after allo-HSCT, and the AUCRefined ELN22 was significantly better than AUCELN22 (0.703 VS 0.637, P<0.001).

Table 1. Univariate analysis of clinical characteristics in RFS and OS

Variables

RFS

OS

HR (95% CI)

P Value

HR (95% CI)

P Value

Patient age at HSCT

1.01(1.00-1.02)

0.150

1.01(1.00-1.03)

0.066

WBC at diagnosis

1.00(0.99-1.00)

0.734

1.00(0.99-1.00)

0.981

ELN2017 risk classification

<0.001

<0.001

Favorable

1.00[Reference]

1.00[Reference]

Intermediate

1.22(0.83-1.80)

0.316

1.31(0.84-2.03)

0.232

Adverse

2.52(1.71-3.70)

<0.001

2.63(1.70-4.07)

<0.001

ELN2022 risk classification

<0.001

<0.001

Favorable

1.00[Reference]

1.00[Reference]

Intermediate

1.64(0.98-2.74)

0.058

1.45(0.82-2.59)

0.205

Adverse

4.58(2.90-7.23)

<0.001

4.40(2.65-7.28)

<0.001

Remission status at time of HSCT

<0.001

<0.001

CR1

1.00[Reference]

1.00[Reference]

≥CR2

1.05(0.70-1.57)

0.826

1.25(0.81-1.94)

0.312

NR

2.91(2.02-4.20)

<0.001

3.44(2.31-5.11)

<0.001

Pretransplant MRD status in CR patents

<0.001

<0.001

Negative

1.00[Reference]

1.00[Reference]

Positive

2.88(1.95-4.26)

2.70(1.74-4.19)

HLA match, n (%)

0.053

0.127

HLA-identical sibling transplant

1.00[Reference]

1.00[Reference]

Haploidentical transplant

0.85(0.56-1.29)

0.455

0.90(0.56-1.44)

0.669

Unrelated transplant

1.45(0.84-2.49)

0.185

1.48(0.80-2.74)

0.185

Conclusions: The updated ELN2022 risk classification did not demonstrate improved predictive performance for outcomes after allo-HSCT compared to ELN2017. We refined the stratification system into four groups by incorporating disease status at transplant, aiming to more accurately identify patients with different outcomes following allo-HSCT.

Disclosure: Nothing to declare

19: Acute Leukaemia

P009 EFFICACY AND SAFETY OF THE THIRD-GENERATION TKI OLVEREMBATINIB IN ADULT PH + ACUTE LYMPHOBLASTIC LEUKEMIA WITH RELAPSED DISEASE OR PERSISTENT MRD BRIDGING TO ALLO-HSCT

XiaoYu Zhang 1, Yanhong Zhao1, Rongli Zhang1, Erlie Jiang1

1Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China

Background: To investigate the efficacy and safety of an olverembatinib regimen in adult Ph/BCR-ABL1 + ALL patients with relapsed disease or persistent MRD prior to allo-HSCT.

Methods: We retrospectively enrolled Ph+ ALL patients with relapsed disease or persistent MRD who were treated with an olverembatinib-based regimen before bridging to HSCT between February 2022 and February 2023.

Results: Seventeen Ph+ ALL patients were treated with olverematinib because of disease recurrence (n = 3) and persistent molecular positivity (n = 14). BCR: ABL1 p190 and p210 fusions were found in nine and eight patients, respectively. Six patients harbored a T315I mutation. In all, 13 patients achieved CMR, and the overall CMR rate by PCR was 76.47%, whereas the MRD neg rate by flow cytometry was 100%. All patients successfully underwent bridging allo-HSCT. With a median follow-up time of 342 (60-509) days post-HSCT, the one-year overall survival rate and recurrence-free survival rate were 94.1±5.71% (95 % CI 83.6~100%) and 88.2±7.81% (95% CI 74.2~100%), respectively. Only one patient experienced extramedullary relapse on day 129. One patient died due to CNS complications.

Conclusions: The findings of this study suggest that in Ph+ ALL patients with disease recurrence and persistent MRD positivity, olverembatinib showed a profound molecular response rate and was well-tolerated in MRD clearance prior to allo-HSCT

Disclosure: The authors declare that they have no competing interests.

19: Acute Leukaemia

P010 INTRACELLULAR CYTOKINE ASSAYS IN COMBINATION WITH DEGRANULATION ASSAY CONTRIBUTE SIGNIFICANTLY TO DETECT AND QUANTIFY LEUKEMIA SPECIFIC IMMUNE CELLS IN AML PATIENTS’ BLOOD OR CULTURE SETTINGS

Olga Schutti1, Lara Klauer2,1, Tobias Baudrexler2,1, Christoph Schmid3, Andreas Rank3, Joerg Schmohl4, Markus Hentrich5, Doris Kraemer6, Helga Schmetzer 1

1Klinikum Grosshadern, Ludwig-Maximilians-University, Working-Group Immune-Modulation, Munich, Germany, 2Contributed Equally, Munich, Germany, 3University Hospital of Augsburg, Augsburg, Germany, 4University Hospital of Tuebingen, Tuebingen, Germany, 5Red Cross Hospital of Munich, Munich, Germany, 6University Hospital of Oldenburg, Oldenburg, Germany

Background: Novel (immune) therapies are needed to stabilize remissions or the disease in AML. Leukemia derived dendritic cells (DCleu) can be generated ex vivo from AML patients’ blasts in whole blood using approved drugs (GM-CSF and PGE-1 (Kit-M)). After T cell enriched, mixed lymphocyte culture (MLC) with Kit-M pretreated (vs. untreated WB) antileukemically directed immune cells of the adaptive and innate immune system were already shown to be significantly increased.

Aims: To evaluate the use of leukemiaspecific assays intracellular cytokine production (INFy, TNFa) INCYT, degranulation (CD107a) DEG for a detailed quantification of leukemiaspecific cells in correlation with functional cytotoxicity and patients’ clinical data.

Methods: Whole blood (WB) samples were collected from 26 AML patients at first diagnosis, during persisting disease, or at relapse after allogeneic stem cell transplantation (SCT), and from 18 healthy volunteers. WB samples were treated with vs. without Kit-M to generate DC/DCleu. After MLC with Kit-M treated vs. untreated WB antigenspecific/ anti-leukemic effects were assessed through INCYT, DEG and a cytotoxicity fluorolysis assay. Quantification of cell subtypes was performed via flowcytometry.

Results: We generated significantly higher frequencies of (mature) DCleu without induction of blast proliferation in Kit-M treated vs. untreated samples. After MLC with Kit-M treated vs. untreated WB, frequencies of immunoreactive cells (e.g. non-naive T-cells, Tprol) as well as in INCYT/DEG ASSAYS leukemiaspecific adaptive - (e.g. B, T(memory)) or innate immune cells (e.g. NK, CIK) were significantly increased. The results of the intracellular production of INFy and TNFa were comparable. The cytotoxicity fluorolysis assay revealed a significantly enhanced blast lysis in Kit-M treated vs. untreated WB. Significant correlations could be shown between induced leukemiaspecific cells of several lines and improved blast lysis.

Conclusions: We successfully detected and quantified immunoreactive cells at a single-cell level using the functional assays (DEG, INCYT, and CTX) ex vivo, allowing us to evaluate the impact of Kit-M pretreated (DC/DCleu containing) WB on the provision of leukemia specific immune cells. Kit-M pretreatment (vs no pretreatment) was shown to significantly increase leukemia-specific IFNγ and TNFɑ producing, degranulating cells and to improve blast-cytotoxicity after MLC.

In vivo treatment of AML patients with Kit-M may lead to anti-leukemic effects and contribute to stabilize the disease or remissions. INCYT and DEG assays qualify to quantify potentially leukemia specific cells on a single cell level and to predict the clinical course of patients under treatment.

Disclosure: Conflict of interest: Modiblast Pharma GmbH (Oberhaching, Germany) holds the European Patent 15 801 987.7-1118 and US Patent 15-517627 ‘Use of immunomodulatory effective compositions for the immunotherapeutic treatment of patients suffering from myeloid leukemias’, with whom H.M.S. is involved with.

19: Acute Leukaemia

P011 TOTAL MARROW AND LYMPHOID IRRADIATION IN COMBINATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE-BASED GRAFT VERSUS HOST DISEASE PROPHYLAXIS CONFERS FAVORABLE GVHD-FREE/RELAPSE-FREE SURVIVAL IN PATIENTS WITH ACUTE MYELOID LEUKEMIA

Anthony Stein 1, Monzr Al Malki1, Dongyun Yang1, Joycelynne Palmer1, Ni-Chun Tsai1, Ibrahim Aldoss1, Haris Ali1, Ahmed Aribi1, Andrew Artz1, Savita Dandapani1, Len Farol1,2, Susanta Hui1, An Liu1, Ryotaro Nakamura1, Vinod Pullarkat1, Eric Radany1, Amandeep Salhotra1, James Sanchez1, Ricardo Spielberger1,2, Guido Marcucci1, Stephen Forman1, Jeffrey Wong1

1City of Hope, Duarte, United States, 2Southern California Permanente Medical Group, Los Angeles, United States

Background: We have conducted a phase 2 study of a conditioning regimen of total marrow and lymphoid irradiation (TMLI) at 2000cGy, together with post-transplant cyclophosphamide (PTCy) for patients with acute myeloid leukemia in first or second complete remission (CR). This approach employs PTCy to attenuate the risk of chronic graft-versus-host-disease (GVHD), while using escalated targeted radiation conditioning before allografting to offset the possible increased risk of relapse.

Methods: We evaluated to date the first 25 patients with a median follow-up of ≥1yr (Table) who enrolled between 2018–2023. Key criteria: ages 18–60, first or second CR, minimal residual disease negative by multi-color flow cytometry, and normal organ function. TMLI was administered on days -4 to 0 without addition of chemotherapy. The radiation dose for all patients (n = 25) was 2000cGy, delivered in 200cGy fractions twice daily. The radiation dose delivered to the liver and brain was kept at 1200cGy. Remaining organs were considered non-targeted. All patients received peripheral blood stem cells on day 0. Cyclophosphamide was given on days +3 and +4, 50mg/kg each day for GVHD prevention. Tacrolimus, 1mg continuous infusion adjusted to maintain levels from 5–10ng/mL, was given from day +5 to day +90, and G-CSF 5µg/kg daily was administered at day +5 until recovery of neutrophil counts.

Endpoints included toxicity, engraftment, overall survival (OS), non-relapse mortality (NRM), and GVHD/relapse-free survival (GRFS) at 1yr. Toxicities were defined according to the Bearman and CTCAE 4.03 scales, the latter for hematologic toxicity. A patient safety lead-in segment (n = 6) was conducted to ensure there were no unexpected toxicities. GRFS was defined as the absence of grade 3-4 acute GvHD, chronic GvHD requiring systemic treatment, relapse, or death (from any cause).

Results:

Variable

Median (range) or N (%)

Age at transplant (yrs)

39 (19-56)

Cytogenetic risk (ELN criteria)

favorable

1 (4%)

intermediate

9 (36%)

unfavorable

8 (32%)

missing

7 (28%)

KPS at HSCT

80

3 (12%)

90

17 (68%)

100

5 (20%)

HCT-CI

16 (64%)

1

6 (24%)

≥2

3 (12%)

Donor source

MSD

11 (44%)

MUD

14 (56%)

Female donor to male recipient

Yes

4 (16%)

No

21 (84%)

CMV

Negative

8 (32%)

Positive

17 (68%)

  1. Patient characteristics. Abbreviations: ELN, European LeukemiaNet; HCT-CI, Hematopoietic Cell Transplantation-specific Comorbidity Index; KPS, Karnofsky Performance Score; MSD, matched sibling donor; MUD, matched unrelated donor; CMV, cytomegalovirus.

The median follow up was 47.1 months (range: 5.9-59.9) for surviving patients (n = 22). Bearman toxicity data are available for all patients. Among these patients, grade 2 toxicities were bladder toxicity and stomatitis. No grade 3-4 toxicities or toxicity-related deaths were observed. All patients engrafted. Median times to neutrophil and platelet recovery were 14 days (range: 13-32) and 20 days (range: 11-49), respectively.

Acute GVHD (aGVHD) developed in 2 patients (100-day Grade II-IV aGVHD: 8.0%, 95%CI: 1.3-22.9); of those, only 1 patient developed Grades III-IV (100-day Grade III-IV aGVHD: 4.0%, 95%CI: 0.3-17.4). Six patients developed chronic GVHD (2-year cGVHD rate: 27%, 95%CI: 11%-47%), which was mild in 5 patients and moderate/severe in 1 patient.

Two-year estimates of OS and leukemia-free survival were 89% (95%CI: 62-97) and 84% (95%CI: 62-94). Disease relapse at 2yrs was 16% (95%CI: 4.9-33). The estimate of NRM at 2yrs was 0%. Relapsed disease after transplant occurred in 5 patients, which led to 3 deaths from relapsed disease. The GRFS rate at 2yrs was 61% (95%CI: 37-78).

Conclusions: 1) This chemotherapy-free conditioning regimen, together with PTCy and tacrolimus, is safe and feasible, with no NRM. 2) All patients achieved engraftment. 3) Participants with ≥1yr follow-up have discontinued immunosuppressive therapy, reducing financial burden and leading to improved quality of life. The results to date suggest a favorable GRFS rate.

Clinical Trial Registry: clinicaltrials.gov, NCT#03467386

Disclosure: Anthony Stein: Sanofi: Current Employment, Current holder of stock options.

19: Acute Leukaemia

P012 INCREASED MESENCHYMAL STROMAL CELLS SENESCENCE AND HIGH CXCL14 LEVELS IN BONE MARROW ARE LINKED WITH THE OCCURRENCE OF GVHD AFTER HEMATOPOIETIC STEM CELLS TRANSPLANTATION

Alexandra Guelton 1, Meriem El Ouafy1, Romain Perouf1, Naceur Charif1, Simona Pagliuca1, Marie-Thérèse Rubio1, Natalia de Isla1

1University of Lorraine, Vandoeuvre-les-Nancy, France

Background: The acute myeloid leukemia (AML) is an hematological malignancy with a bad pronostic with an estimated survival of 50% at 1 year, and 27% at 5 years. The allogenic hematopoietic stem cells transplantation (allo-HSCT) is the only curative treatment of AML, marked by two major adverse effects: graft versus host disease and relapse. Although medullary microenvironment is composed of many cellular actors, mescenchymal stromal stem cells (MSC) constitute key players, both for their role in hematopoiesis and for their immunomodulatory capacities, particulary toward T cells. The aim of our work was to study the link between medullary microenvironment (MSC, cytokines…) in pre-transplantation, and the occurrence of GVHD.

Methods: We conducted a prospective, single-center study at the Nancy University Hospital, involving HSC allograft patients with AML. Inclusion criteria were: graft conditioning including a combination of fludarabine and busulfan, homogeneous graft-versus-host disease (GVHD) prophylaxis (anti-lymphocytic serum, cyclosporin and mycophenolate mofetil. Medullary MSC from bone marrow of patients before HSCT were isolated and cultured. MSC used as control provided from patients without AML, and were obtained after surgical hip arthroplasty. MSC were characterized based on the International Society of Cellular Therapie (ISCT) criteria as their adhesion to plastic, their surface phenotype (CD73 + , CD90+, HLA-DR−, CD45−, CD34−) and their self-renewal capacity.

Then, MSC senescence was studied thanks to β-galactosidase test coupled with EdU proliferation test (5-Methyl-2′-deoxycytidine), to distinguish senescents and proliferative cells, and avoid false positives. Moreover, proteins involved in cell senescence (p16, p21, p53, p38) were evaluated by Western-Blot. Then, many cytokines involved in cell senescence (IL10, IL6, IL1, IL13…) and in immune surveillance (CXCL14) were quantified by CBA and ELISA method. In parallel, MSC immunomodulatory properties were determined by coculture experiments between MSC and T cells from healthy donnors.

T statistical tests and Pearson correlation tests was performed thanks to GraphPad Prism. Principal component analysis (PCA) was performed thanks to XLSTAT.

Results: Our results showed that high levels of medullary MSC senescence from AML patients before HSCT, correlated with a decrease of MSC immunomodulatory properties toward T cells, was associated with the occurrence of GVHD, three months after HSCT. Moreover, a positive correlation between the level of medullary MSC senescence and the medullary concentration of CXCL14 was observed. Among the other cytokines studied, no correlation was shown between their concentration and medullary MSC senescence.

Conclusions: Our results showed that a pro inflammatory microenvironment before HSCT, characterized by the presence of senescent MSC with disturbed immunomodulatory properties, and the presence of CXCL14, would be favorable to the occurrence of GVHD after HSCT.

Disclosure: Nothing to declare

19: Acute Leukaemia

P013 CORRELATION OF THE OVERALL SURVIVAL IN MIXED-PHENOTYPE ACUTE LEUKAEMIA AND ACUTE LYMPHOBLASTIC LYMPHOMA WITH THE REFINED DISEASE RISK INDEX APPLIED IN AN UPPER-MIDDLE INCOME COUNTRY

Naty López-Córdova 1, Jessica Meza-Liviapoma1, Jule Vasquez-Chavez1, Claudio Flores-Flores2, Shirley Quintana-Truyenque1

1Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru, 2Universidad Peruana San Juan Bautista, Lima, Peru

Background: The Refined Disease Risk Index (R-DRI; 4 risk groups: low [LR], intermediate [IR], high [HR] and very high [V-HR]) is a score used to estimate overall survival after allogeneic hematopoietic stem cell transplantation (Allo-HSCT) in patients with haematological malignancies, however, both in its original version and in the R-DRI does not consider certain diagnoses, such as mixed-phenotype acute leukaemia (MPAL) and acute lymphoblastic lymphomas (LBL). These diagnoses constitute an important population since they have a very aggressive course and fatal outcomes in most cases. The objective of the study is to determine the correlation of overall survival in patients with MPAL and LBL after Allo-HSCT, in relation to R-DRI. These results will allow these patients to be assigned to one of the existing risk groups in order to have an estimate of their post HSCT prognosis.

Methods: We reviewed retrospectively the data of 166 patients who had an Allo-HSCT between 2012 and 2023 in the Bone Marrow Transplant (BMT) unit of the National Institute of Neoplastic Diseases (INEN). The information was extracted from the unit registry and was completed through the electronic medical records. Patients diagnosed and transplanted for MPAL and LBL were subjected to the analysis and their overall survival was evaluated, which was compared with the data of patients with acute lymphoblastic leukaemia (ALL) and acute myeloid leukaemia (AML). Survival was estimated using the Kaplan-Meier method and comparisons were made using the Log-Rank test.

Results: The median age at HSCT was 26 years (range: 15 to 63), 84 patients were men (50.6%). 85 patients had ALL; 41, AML; 11, Chronic myeloid leukaemia (CML); 13, MPAL; and 16, others (Myelodysplastic syndrome: 8, LBL: 6, Hodgkin lymphoma: 1, aggressive T/NK cell leukaemia: 1). According to the R-DRI classification (does not include MPAL and LBL), 4.8% of patients were LR, 38.1% IR, 35.4% HR, and 21.8% V-HR. Median follow-up was 3.9 years and median overall survival (OS) was not reached for the entire cohort. According to R-DRI, the 4-year OS rate was 100% in LR, 57.4% in IR, 53.5% in HR, and 38.3% in V-HR, respectively. The 4-year OS in patients with MPAL was 76.9%, and in LBL 44.4%. The OS of patients with MPAL was superior to those with IR, presenting a significant difference in relation to the HR group (p = 0.015).

Conclusions: The OS of patients with MPAL is superior to those with IR, presenting a significant difference in relation to those with V-HR. These results suggest that there would be a group that is similar to that of IR, but with a better prognosis than that described in the R-DRI and that could give rise to a new “low-intermediate” group in which the MPAL could be included.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P014 SIGNIFICANT IMPROVEMENT OF SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN ACUTE LYMPHOBLASTIC LEUKEMIA OVER TWO DECADES – A SINGLE CENTER RETROSPECTIVE ANALYSIS

Ben-Niklas Baermann1, Paula Kessler 1, Astrid Tautges2, Paul Sebastian Jäger1, Mustafa Kondakci3, Kathrin Nachtkamp1, Sascha Dietrich1, Guido Kobbe1

1Medical Faculty and University Hospital Düsseldorf, Düsseldorf, Germany, 2Office for general practice Dirk Röhlich, Trier, Germany, 3Lukas Hospital Solingen, Solingen, Germany

Background: Acute lymphoblastic leukemia (ALL) treatment landscape has dramatically changed within the last two decades. Especially tyrosine kinase inhibitors in Philadelphia positive (BCR-ABL) ALL as well as Blinatumomab, Inotuzumab and chimeric antigen receptor t-cell therapy for B-cell precursor ALL (B-ALL) were the most prominent pharmacologic milestones. Moreover the german multicenter ALL study group (GMALL) made strong efforts to improve conventional and transplant strategies to reduce non relapse mortality.

Methods: We retrospectively analyzed clinical outcome of 146 patients (61% male, 39% female, median age at transplant 36 years, range 17-74) with acute lymphoblastic leukemia (B-ALL 76%, T-ALL 24%) receiving allogeneic stem cell transplantation (alloHSCT) at the UKD, Heinrich Heine University of Düsseldorf between 1989 and 2022. Of Patients with B-ALL 33% were Philadelphia chromosome positive.

The majority of patients (92%) received myeloablative conditioning almost all containing total body irradiation (TBI) with at least 8 Gy. Matched related donors were used in 32%, 40% had matched unrelated donors and 27% mismatched or haploidentical donors. Median time from diagnosis to alloHSCT was 6 months (range 2-80). Haematologic remission could be achieved in 95% prior to transplant.

Results: Median follow up after alloHSCT was 5.6 years for surviving patients. Median overall survival (OS) from transplant for all patients was 2.8 years (95% CI 0.0-6.1 years). OS after two and five years were 51% (95% CI 43-59) and 47% (95% CI 39-55). For T-ALL median OS was not reached, for B-ALL median OS was 1.3 years (95% CI 0-2.9 years).

For patients with first diagnosis from 2000 to 2010, median OS from alloHSCT was 1.2 years, whereas after 2010 median OS is not reached with a significant difference between these subgroups (p<0.05). OS after two and five years for patients with first diagnosis after 2010 were 66 % (95% CI 55-78) and 64 % (95% CI 52-76). These findings can be reproduced for BCR-ABL positive as well as negative B-ALL subgroups but not for patients with T-ALL.

MRD persistence before alloHSCT was associated with significant worse OS even in haematologic complete remission (median OS 1.4 years vs. not reached, p < 0.05), not significantly differing for patients with first diagnosis before or after 2010.

Median OS after alloHSCT could not be improved by application of higher doses of TBI (12Gy vs. 8Gy, median OS 2.9 years vs. not reached, p = 0.5).

Conclusions: Prognosis of B-lineage acute lymphoblastic leukemia after alloHSCT significantly improved over time and reducing TBI toxicity by dose reduction did not negatively influence OS. Achieving MRD negativity before alloHSCT especially with new targeted therapies seems to play an important role. Nevertheless worse OS in patients with MRD persistence could not be overcome in recent years.

Disclosure: Ben-Niklas Baermann: Travel support: Medac, Kite-Gilead; Advisory Role or Speaker Honoraria: Kite-Gilead, Incyte

Paula Kessler: Nothing to declare.

Astrid Tautges: Nothing to declare

Paul Sebastian Jäger: Nothing to declare

Mustafa Kondakci: Nothing to declare

Sascha Dietrich: Nothing to declare

Kathrin Nachtkamp: Nothing to declare.

Guido Kobbe: Advisory Role or Speaker Honoraria: MSD, Pfizer, Amgen, Novartis, Gilead, BMS-Celgene, Abbvie, Biotest, Takeda, Eurocept; Financing of Scientific Research: BMS-Celgene, Amgen, Abbvie, Eurocept, Medac.

19: Acute Leukaemia

P015 OUTCOMES OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOLLOWING BLINATUMOMAB IN CHILDREN AND YOUNG ADULTS WITH B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA

Mattia Algeri 1, Michele Massa2, Federica Galaverna2, Daria Pagliara2, Ilaria Pili2, Francesca Del Bufalo2, Marco Becilli2, Emilia Boccieri2, Roberto Carta2, Francesco Quagliarella2, Chiara Rosignoli2, Carmen Dolores De Luca2, Barbarella Lucarelli2, Pietro Merli2, Franco Locatelli3

1IRCCS Bambino Gesù Children’s Hospital - Magna Graecia University, Rome - Catanzaro, Italy, 2IRCCS Bambino Gesù Children’s Hospital, Rome, Italy, 3IRCCS, Bambino Gesù Children’s Hospital, Catholic University of the Sacred Heart, Rome, Italy

Background: Blinatumomab, a CD3-CD19 bispecific T-cell engager, demonstrated high efficacy in inducing and consolidating remission in relapsed/refractory (r/r) B-cell acute lymphoblastic leukemia (B-ALL). Available evidence suggests that r/r B-ALL patients have a better outcome if they received allogeneic hematopoietic stem cell transplantation (HSCT) after blinatumomab. However, the effect of blinatumomab on HSCT outcomes in children and young adults (YA) remains to be fully elucidated.

Methods: We retrospectively analyzed 78 children and YA with B-ALL who underwent allogeneic HSCT at our Institution, from February 2016 to June 2023, after receiving blinatumomab as last therapy before transplant either as part of reinduction/consolidation or as MRD-clearing strategy. Most patients (n = 55) received a single 28-day course. Median interval from blinatumomab discontinuation to HSCT was 23 days (IQR 19-31.75).

Median age at HSCT was 5 years (range 1-24). Patients received unmanipulated grafts from HLA-identical related (MFD, n = 13) or matched unrelated donors (MUD, n = 33), or αβT-cell depleted HSCT from HLA-haploidentical relatives (TCD-Haplo, n = 32) after TBI- (n = 72, 92.3%) or chemo-based myeloablative conditioning regimen (n = 6, 7.7%). All subjects were in complete remission (CR) at time of HSCT, associated with MRD-negativity (<10^-4) in 94% of cases. Thirty-one (39.7%) patients harbored at least one very high-risk (VHR) disease feature (Table 1). Further patient and transplant characteristics are reported in Table 1.

Sex (female/male)

31 (40)/47(60)

CR number at Blinatumomab2

• CR1

15 (19.2)

• CR2

58 (74.4)

• CR3

5 (6.4)

Age group

• ≤ 4 years

17 (21.8)

• 5-11 years

38 (48.7)

• ≥12 year

23 (29.5)

Very high-risk disease features

• TCF3-Rearranged

2 (2.6)

• Hypodiploidy

2 (2.6)

• KMT2A/AF4

5 (6.4)

• IKZF1plus and poor MRD response after induction

4 (5.1)

• No CR at Day33 without cytogenetic abnormalities

2 (2.6)

• Very early relapse (<18 months from diagnosis) for CR2 patients

21 (26.9)

Therapy before HSCT:

• Chemotherapy + Blinatumomab

49 (62.9)

• Chemotherapy + Inotuzumab + Blinatumomab

14 (17.9)

• Chemo-free treatment of relapse (Inotuzumab OR TKI + Blinatumomab)

15 (19.2)

Results: All patients achieved sustained donor engraftment, median time to neutrophil and platelet recovery being 17 (IQR 14-19) and 15 (IQR 11-20) days, respectively. After a median follow-up of 26 months, disease-free survival (DFS) of the whole cohort was 74.2% (95%CI 63.3-85.1), overall survival (OS) 84.3% (95%CI 74.5-94.1) and non-relapse mortality (NRM) 2.6% (95%CI 0-6.2). Cumulative incidence (CI) of grade II-IV acute graft-versus-host disease (aGvHD) was 17.9% (95%CI 9.08-26.6), with only two cases of grade III; CI of extensive chronic GvHD (cGvHD) was 4.6% (95%CI 0-9.7). No unexpected toxicities were observed. Patients aged ≤4 years had significantly lower DFS as compared to those aged 5-11 years or ≥12 years (45.6% 95%CI 20.1-71.1, 82% 95%CI 70-100 and 85.6% 95%CI 68.8-95.5, respectively, p = 0.02), due to a higher cumulative incidence of relapse (CIR 51.5% 95%CI 24-79.1, 18% 95%CI 4.9-31.1, 18.6 95%CI 8.6-28.9, respectively, p = 0.05). Neither the use a specific conditioning, nor MRD status and presence of VHR features influenced the probability of DFS. A trend toward better DFS was observed in CR1 patients (94.1%, 95%CI 82.95-100) versus CR2-3 (69.5%, 95CI 57-83.04, p = 0.18) and in those receiving unmanipulated matched grafts (80.1% 95%CI 64.6-95.6) versus TCD-Haplos (64.9%, 95%CI 48.2-81.5, p = 0.058).

Sixteen patients relapsed after HSCT, all with CD19-positive recurrence, after a median time of 9 months following transplantation (range 1-23). Of them, 10 received anti-CD19 CAR-T cells while 2 received inotuzumab, the 2-year OS after relapse being 52.7% (95%CI 27.1-78,3).

Conclusions: HSCT following blinatumomab in children/YA with r/r B-ALL is safe and highly effective, with remarkably low NRM (only 2 patients died for NRM) and absence of unexpected toxicities. These findings indicate that blinatumomab represents a suitable strategy to achieve long-term disease eradication and that it doesn’t preclude successful salvage with CAR-T cells in case of relapse.

Disclosure: Mattia Algeri: Vertex: Membership on an entity’s Board of Directors or advisory committees; Neovii: Speakers Bureau.

Pietro Merli: Sobi: Membership on an entity’s Board of Directors or advisory committees; Jazz: Membership on an entity’s Board of Directors or advisory committees; Miltenyi: Speakers Bureau; Amgen: Speakers Bureau.

Franco Locatelli: Miltenyi, Jazz Pharm, Medac, Sobi, Gilead, BluebirdBio: Speakers Bureau; Sanofi, Vertex: Membership on an entity’s Board of Directors or advisory committees; Bellicum, Amgen, Neovii, Novartis. Sanofi, SOBI, Vertex: Membership on an entity’s Board of Directors or advisory committees, Speakers Bureau

19: Acute Leukaemia

P016 THE ROLE OF ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN CONSOLIDATION OF MINIMAL RESIDUAL DISEASE-NEGATIVE REMISSION IN RELAPSED/REFRACTORY B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA IN ADULTS AFTER PHARMACOLOGIC IMMUNOTHERAPY

Kseniia Afanaseva1, Ivan Moiseev 1, Bella Ayubova1, Elena Babenko1, Ildar Barkhatov1, Tatiana Gindina1, Anna Smirnova1, Olesya Smykova1, Yulia Vlasova1, Sergey Bondarenko1, Alexander Kulagin1

1RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russian Federation

Background: Allogeneic hematopoietic cell transplantation (allo-HCT) is a standard consolidation strategy for B-cell precursor acute lymphoblastic leukemia (BCP-ALL) patients with relapsed/refractory (r/r) disease and persistent minimal residual disease (MRD). Long-term outcomes of pharmacologic immunotherapy in r/r BCP-ALL is relatively modest, but MRD-negative patients have better prognosis, which was not directly compared to the results of allo-HCT in large studies.

Methods: The study included 101 BCP-ALL patients who achieved MRD-negative complete remission (CR) with two consecutive evaluations after either blinatumomab or inotuzumab ozogamicin treatment from November 2014 to September 2023 (Table 1). Median age was 22 years (range, 18 - 67 years). MRD response was assessed both by flow cytometry and molecular analysis for patients with known molecular markers. Median blinatumomab courses were 1 (range, 1-7), inotuzumab ozogamicin – 1 (range, 1-3). Consolidation with allo-HCT was performed in 62 patients, not performed in 39 patients within the frame of the routine clinical practice. Long-term outcomes after immunotherapy according to allo-HCT consolidation were estimated by landmark analysis.

Table 1. Patients’ characteristics of the study group

Characteristics

Immunotherapy followed by allo-HCT

N = 62

Immunotherapy only

N = 39

P-value

Blinatumomab/Inotuzumab ozogamicin

40 (65%)/22 (35%)

32 (82%)/7 (18%)

0,06

High cytogenetic risk

(yes/no)

23 (37%)/39 (63%)

11 (28%)/28 (72%)

0,36

Extramedullary disease in r/r

(yes/no)

17 (27%)/45 (73%)

3 (8%)/36 (92%)

0,02

Indication to immunotherapy (MRD/r/r*)

16 (26%)/46 (74%)

19 (49%)/20 (51%)

0,02

*R/R status

(refractory/1st relapse/>1 relapse)

5 (11%)/24 (52%)/17 (37%)

0 (0%)/8 (40%)/12 (60%)

0,12

Previous allo-HCT

(yes/no)

4 (6%)/58 (94%)

15 (38%)/24 (62%)

0,001

Subsequent immunotherapy

(yes/no)

10 (16%)/52 (84%)

8 (21%)/31 (79%)

0,58

Donor

(Haploidentical/other)

21 (34%)/41 (66%)

-

-

Results: Median follow-up time in the group of patients who received allo-HCT after immunotherapy was 55 months (range, 2-95), in immunotherapy without allo-HCT group – 56 months (range, 5 - 101) for those who survived by follow-up. Median time from achievement of MRD-negative CR to allo-HCT was 2 months (range, 2 - 27). Median time from achievement of MRD-negative CR to relapse was 12 months (range, 2 - 72) in immunotherapy followed by allo-HCT group and 13 months (range, 2 – 61) in immunotherapy without HCT group. In a landmark analysis of 5-year overall survival (OS) and relapse-free survival (RFS) by allo-HCT at 2 months after MRD-negative CR establishment no difference between outcomes in two groups was demonstrated: OS was 44,1% (95% CI 29,3 – 66,3) in allo-HCT group and 49,6% (95% CI 38,4 – 64,1) in immunotherapy without allo-HCT group, p = 0,19; RFS was 31% (95% CI 17,9 – 53,7) and 36,1% (95% CI 25,3 – 51,5), p = 0,25, respectively. Results were similar when assessed for 4-months (p = 0,46 for OS and p = 0,67 for RFS) and 6-months landmark points (p = 0,61 for OS and p = 0,83 for RFS). Subgroup univariate analysis showed no influence of any predictors (blinatumomab versus inotuzumab ozogamicin, high cytogenetic risk, extramedullary disease in relapse, MRD or relapse as an indication to immunotherapy, previous allo-HCT) for RFS in immunotherapy without subsequent allo-HCT group, while extramedullary disease in relapse and haploidentical donor increased risk of RFS after allo-HCT, which was confirmed in a multivariate model (HR 2,8 (95% CI 1,38 – 5,6), p = 0,004 and HR 2,3 (95% CI 1,06 – 4,9), p = 0,03).

Conclusions: The study demonstrated that responders to pharmacologic immunotherapy with r/r BCP-ALL with sustainable MRD-negative status have no benefit from all-HCT consolidation in the general group. Nonetheless, heterogeneity in allo-HCT outcomes indicates that further cooperative studies and consolidation of multicenter data is required to define the group which will benefit from allo-HCT in MRD-negative CR.

Disclosure: Nothing to declare

19: Acute Leukaemia

P017 FLAG-IDA/VENETOCLAX IS ASSOCIATED WITH HIGHER REMISSION RATES AND IMPROVED POST-TRANSPLANT SURVIVAL COMPARED TO VENETOCLAX/AZACITIDINE IN RELAPSED/REFRACTORY AML PATIENTS

Evgeny Klyuchnikov1, Anita Badbaran1, Radwan Massoud 1, Normann Steiner1, Petra Freiberger1, Franziska Modemann1, Martin Schönrock1, Sophia Cichutek1, Walter Fiedler1, Micha Peeck2, Nico Gagelmann1, Christine Wolschke1, Francis Ayuk1, Ulrike Bacher3, Nicolaus Kröger1

1University Cancer Center of Hamburg, Hamburg, Germany, 2Agaplesion Diakonie Clinic, Rotenburg, Germany, 3Bern University Hospital, Bern, Switzerland

Background: Patients (pts) with relapsed/refractory (R/R) AML experience poor remission rates and short durations of response. Combinations of BCL2 inhibition have proven to be synergistic with low intensity and intensive chemotherapy, leading to high remissions. In this monocentric study, we compared post-transplant outcomes in R/R AML pts who received low (azacitidine [AZA]) or intensive (FLAG-Ida) regimen combined with venetoclax [VEN] as “bridging” strategies before allogeneic stem cell transplantation (allo-SCT).

Methods: 90 pts (male, n = 53; median, 59 years [range, 19-75]) with R/R AML who received allo-SCT (related, n = 58) between 2019-2023 at the Department of Stem Cell Transplantation (University Medical Centre Hamburg) were included if they received VEN-based “bridging” strategies. The measurable residual disease (MRD) detection was performed by flow cytometry (“different from normal”; ELN guidelines). Primary endpoint was the difference in overall survival (OS) from allo-SCT. Secondary endpoints were differences in leukaemia-free survival (LFS), relapses and non-relapse mortality (NRM) from allo-SCT.

Results: The majority of allografts were performed after myeloablative conditioning (n = 57, 63%) with ATG as GvHD prophylaxis (n = 65, 72%). 62 pts underwent 1st whereas 26 (29%) 2nd and two 3rd allo-SCT. At allo-SCT,62 pts (69%) were in complete remission (CR). Bridging therapy consisted of AZA-VEN (one or two cycles) in 41 pts or FLAG-Ida-VEN in 49 pts. Pre-transplant MRD data were available in 49 pts. The rate of MRDnegCR (16/27, 59% vs 11/27, 41%) and MRDpos CR (16/22, 73% vs 6/22, 27%) was higher in the FLAG-Ida-VEN group. The rate of non-CR (17/28, 61% vs 11/28, 39%, p = 0.055) was higher in the AZA-VEN group.

During a median follow up of 13 months (2-140), there were 34 mortalities, 27 relapses and 13 NRM events. The 2-year OS (61%, 43-77% vs 46%, 29-64%, p = 0.089) and LFS (46%, 27-66% vs 39%, 24-52%, p = 0.045) were better in FLAG-Ida-VEN. The relapse and NRM rates were similar between two groups. Other factors, significant in univariate analysis included remission status and donor type. In the multivariate analysis, CR at allo-SCT had significant independent impact on OS (HR 0.17, 0.08-0.35, p<0.0001), LFS (HR 0.18, 0.1-0.35, p<0.0001) and relapses (HR 0.19, 0.08-0.43, p<0.0001).

In addition, we performed an analysis (n = 77) focusing on remission status augmented by MRD. The 1 y OS for MRDneg CR, MRDpos CR and pts with non-CR was 93% (77-98%), 63% (40-81%) and 30% (16-49%, p<0.001), respectively. The 1-y LFS for MRDneg CR, MRDpos CR and pts with non-CR was 92% (76-98%), 59% (37-78%) and 21% (11-51%, p<0.001), respectively. The relapse rate at 1 y for MRDneg CR, MRDpos CR and pts with non-CR was 9% (2-39%), 21% (8-44%) and 55% (36-72%, p<0.001), respectively. In multivariate analysis, MRDposCR and non-CR had adverse impact on OS (HR 5.2, 1.1-24.4, p = 0.035; HR 11.5, 2.7-49.9, p = 0.001; Wald test, p = 0.002), LFS (HR 2.9, 0.9-9.2, p = 0.076; HR 8.6, 2.9-25, p<0.001; Wald test, p<0.001) and relapses (HR 2.6, 0.5-14.2, p = 0.24; HR 10.1, 2.5-39.8, p<0.001; Wald test, p<0.001) compared to MRDnegCR, respectively.

Conclusions: Intensive venetoclax-based “bridging” regimen with FLAG-Ida is associated with higher CR rates than AZA-VEN, leading to overall better outcomes.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P018 COMPARISON OF OUTCOMES OF HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION USING POST-TRANSPLANTATION CYCLOPHOSPHAMIDE AND EX-VIVO TCRΑΒ + CD19 DOUBLE-NEGATIVE SELECTED GRAFT IN ACUTE MYELOID LEUKEMIA

László Gopcsa 1, Hajnalka Andrikovics1, Alexandra Balogh1, Anikó Barta1, Judit Bogyó2, Zoltán Csukly1, Katalin Dobos1, János Dolgos1, Apor Hardi1, János Fábián1, Laura Giba-Kiss1, József Harasztdombi1, Kinga Kerner1, Ágnes Király1, Gergely Lakatos1, Viktor Lakatos1, Lilla Lengyel1, Nóra Meggyesi1, Noémi Németh1, Melinda Paksi1, Laura Regáli1, Marienn Réti1, János Sinkó1, Bálint Szabó1, Anikó Szilvási2, Éva Torbágyi1, Andrea Várkonyi1, Nikolett Wohner1, István Vályi-Nagy1, Péter Reményi1

1Central Hospital of Southern-Pest, National Institute of Hematology and Infectious Diseases, Budapest, Hungary, 2The Hungarian National Blood Transfusion Service, Budapest, Hungary

Background: Haploidentical hematopoietic stem cell transplantation (Haplo-HSCT) represents a curative therapeutic option for the treatment of patients suffering from acute leukemia. Moreover, the incidence of severe acute and extensive chronic GVHD in the case of Haplo-HSCT has proven to be more favorable compared to the other donor types. Currently, Haplo-HSCT with post-transplantation cyclophosphamide (PTCY) GVHD prophylaxis is considered the “gold standard” procedure. However, a less common approach to performing Haplo-HSCT involves ex-vivo T-cell depletion, employing various methods, with the most recognized being the use of a graft produced through TCRαβ + CD19 double-negative depletion (TCDαβ + CD19). Experiencies with pediatric HSCT using haploidentical grafts generated through TCDαβ + CD19 have been notable for their compelling outcomes. There is limited clinical experience in adult HSCT practice with this method.

Methods: In our retrospective analysis, we compared the outcomes of Haplo-HSCT performed between 2015 and 2022 for AML using PTCY (N = 60) and TCDαβ + CD19 (N = 28).

Results: In the PTCY group, 37 males (62%) and 23 females (38%) were included, with a median age of 50.5 years, while in the TCDαβ + CD19 group, there were 15 males (54%) and 13 females (46%) with a median age of 50 years. Mostly myeloablative conditioning regimens were used in both groups, primarily involving Thiotepa-Busulfan-Fludarabine and Thiotepa-Treosulfan-Fludarabine. All patients in the PTCY group received PTCY+tacrolimus+MMF GVHD prophylaxis. In the TCDαβ + CD19 group, 64% of patients did not received post-graft immunosuppression, however, GVHD prophylaxis was administered in 36% of cases (18% with MMF, 11% with tacrolimus, and 7% with ruxolitinib). The incidence of acute GVHD in the PTCY and TCDαβ + CD19 groups was 39% and 25%, respectively, with severe cases being 7% and 4%. The majority of acute GVHD cases were grade 2 and responded well to steroid treatment. The incidence of chronic GVHD in the PTCY and TCDαβ + CD19 groups was 31% and 21%, respectively. The extensive chronic GVHD was similar in the two groups (14% versus 10,5%). The relapse rates were low in both groups: 13% and 11%. Prolonged poor graft function was observed in 22% of patients receiving PTCY. Non-relapse mortality (NRM) was identical in both groups: PTCY and TCDαβ + CD19 34% and 28%, respectively. The overall survival (OS), disease-free survival (DFS), and refined GVHD-free and relapse-free survival (rGRFS) were more favorable in the TCDαβ + CD19 group: 61% and 53%, 61% and 51%, 53% and 46%.

Conclusions: The presented results are unique in terms of comparing both PTCY and TCDαβ + CD19 methods within one center. The OS, DFS, and GRFS were favorable with the TCDαβ + CD19 method, but larger studies are necessary for confirmation. Unlike in ALL, the occurrence of rejection with the TCDαβ + CD19 method does not significantly impact long-term outcomes. Therefore, the use of TCDαβ + CD19 method in AML can be safely employed, resulting in faster engraftment, shorter hospital stays, and a clearly lower incidence of poor graft function. The relapse rate was low in both patient groups, indicating a significant graft-versus-leukemia effect. The incidence of severe acute GVHD was low. Moreover, the use of post-graft maintenance therapeutic strategies in AML can further enhance long-term patient survival.

Disclosure: Disclosure of conflict of interest: None.

19: Acute Leukaemia

P019 AZACITIDINE AND VENETOCLAX WITH OR WITHOUT DLI IN RELAPSED MYELOID MALIGNANCIES AFTER ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION – A RETROSPECTIVE MULTICENTER STUDY OF THE SFGM-TC

Turot Mélanie 1, Loschi Michael2, Chantepie Sylvain3, Arnautou Pierre4, Poire Xavier5, Maillard Natacha6, Chalandon Yves7, El-Cheikh Jean8, Ceballos Patrice9, Devillier Raynier10, Alani Mustafa11, Fatrara Thomas12, Rubio Thérèse13, Daguindau Etienne14, Klemencie Marion15, Beauvais David16, Huynh Anne17, Marchand Tony18, Volpari Victoria19, Barette Lauren19, Pivert Cécile20, Maury Sébastien21, Suarez Felipe22, Fuseau Charline23, Lauron Sandrine24, Uzunov Madalina25, Castilla Cristina26, Forcade Edouard27, Bay Jacques-Olivier1, Ravinet Aurélie1

1Clermont-Ferrand University Hospital, Clermont-Ferrand, France, 2Nice University Hospital, Nice, France, 3Basse-Normandie Institute of Hematology, Caen, France, 4Percy Army Training Hospital, Clamart, France, 5Saint-Luc University Clinic, Uclouvain Bruxelles Woluwe, Belgium, 6Poitiers University Hospital, Poitiers, France, 7Geneva University Hospitals, Geneva, Switzerland, 8AUB Medical Center, Beyrouth, Lebanon, 9Montpellier University Hospital, Montpellier, France, 10Paoli-Calmettes Institute, Marseille, France, 11Henri Becquerel Center, Rouen, France, 12Saint-Etienne University Hospital, Saint-Etienne, France, 13Nancy Regional and University Hospital, Nancy, France, 14Besançon University Hospital, Besançon, France, 15Anger University Hospital, Anger, France, 16Lille University Hospital, Lille, France, 17Toulouse Cancer University Institute, Toulouse, France, 18Rennes University Hospital, Rennes, France, 19Grenoble Alpes University Hospital, Grenoble, France, 20Public Assistance - Paris Hospitals, Paris, France, 21Henri-Mondor Hospital Public Assistance - Paris Hospitals, Créteil, France, 22Necker Hospital Public Assistance - Paris Hospitals, Paris, France, 23Strasbourg Europe Cancer Institute, Strasbourg, France, 24Edouard Herriot Hospital-Lyon Civil Hospices, Lyon, France, 25La Pitié Salpêtrière Hospital Public Assistance - Paris Hospitals, Paris, France, 26Gustave Roussy, Villejuif, France, 27Bordeaux University Hospital, Bordeaux, France

Background: Despite the recent therapeutic progress, between 30 to 40 % patients relapse after allogeneic hematopoietic stem cell transplantation (alloHSCT) for acute myeloid leukemia (AML) or myelodysplastic syndromes (MDS). While Azacitidine and Venetoclax (VenAza) combination has been approved in previously untreated patients with AML ineligible for intensive induction therapy, VenAza associated with donor lymphocyte infusion (DLI) is also considered post-transplant as salvage therapy. However, limited data are available regarding outcomes with this specific setting.

Methods: To evaluate efficiency and safety of VenAza with/without DLI in relapsed myeloid malignancies after alloHSCT, we retrospectively collected data from January 1, 2016 to September 1, 2022 in 29 centers from the Société Francophone de Greffe de Moelle et Thérapie Cellulaire (SFGM-TC). We included 173 patients aged 18 years or older treated with the association of VenAza as salvage therapy for AML or MDS after one or more alloHSCT. The data were extracted from the ProMISe database, an international registry coordinated in part by the SFGM-TC.

Results: The median follow up after the transplant was 14 months. Out of the 173 patients, 152 (87,86%) had an AML including 11 cases of secondary acute myeloid leukemia, and 21 (12,14%) had a MDS. The median age at transplant was 53,78 (17,9 – 74,86). DLI was administered to 43 patients (24,86%) who relapsed after alloHSCT.76 patients (44,19%) experienced a relapse within 6 months after alloHSCT (defined as “early relapse”) and 96 patients (55,8%) relapsed more than 6 months after alloHSCT (“late relapse”).117 patients were treated with VenAza after their 1st relapse,39 after their 2nd and 6 after 3 or more relapses. The median number of VenAza cycles per patient was 2 (1-13) (Table 1). The median overall survival time estimate was 18,99 months with a 1-year overall survival (OS) of 62,45% (95% CI 55,52 - 70,23%) and a 2-years OS of 40,32 % (95% CI 33,21 – 48,95%). The median survival was longer (p = 0,001) for patients who were treated by VenAza for a late relapse with a 2-years OS of 62,96% (95% CI 53,60 – 73,96%) and 7,17% (95% CI 2,58-19,89%) for early relapses. The OS and EFS (P<0,001) were better in the DLI group. In multivariate analysis, survival remains better in DLI group. (HR = 0,57 (95% CI 0,36 – 0,92)). A total of 82,75 % patients relapsed after post-transplant VenAza and 10,90 % had a complete remission. Additionally, 17 patients (10,43%) developed chronic graft-versus-host disease (GVHC) after alloHCST, including 6 patients after venAza. The main causes of death were relapse or progression of the original disease (61,81%) and infections (21,18%).

Table 1. Patients’ characteristics.

n = 173

Patient sex, (n) %

Female

76 (43,96)

Male

97 (56,07)

Median age, years (range)

53,78 (17,19; 74,86)

Diagnosis, (n) %

AML

152 (87,86)

De novo AML

142 (92,76)

Secondary AML

11 (7,23)

MDS

21 (12.14)

Score HCT-CI, (n) %

Score > 2

34 (21,79)

Type of transplantation, (n) %

Match related donor

52 (30.06)

Match unrelated donor

82 (47.40)

Haplo identical donor

39 (22.55)

Transplantation conditioning, (n) %

MAC

48 (28.24)

TBI

10 (5.88)

ATG

124 (73.81)

GVHD prophylaxis, (n) %

170 (98.27)

Cyclosporine

31 (18.24)

Cyclosporine and Methotrexate

37 (21.76)

Cyclosporine and Mycophenolate mofetil

54 (31.77)

Cyclosporine, Mycophenolate mofetil and Cyclophosphamide

34 (20,00)

Others

14 (8,24)

DLI before VenAza, (n) %

23 (13.30)

1

18 (78,26)

2

4 (17,39)

3

1 (4,35)

Acute GVHD, (n) %

79 (45.66)

Grade >2

9 (11,38)

Chronic GVHD, (n) %

32 (18,50)

Score NIH >3

5 (10,32)

Relapses after HSCT, (n) %

First relapse

117 (72,22)

Second relapse

39 (24,07)

Three or more relapse

6 (3,70)

Median follow-up after transplant, months (range)

14,08 (1,74; 65,91)

VenAza cycles, (n) % (range)

2 (1;13)

DLI in association to VenAza, (n) %

43 (24,86)

1

26 (60,47)

2

7 (16,28)

3

8 (18,60)

4

2 (4,65)

Relapse after HSCT, (n) %

Early relapse < 6 months

76 (44.19)

Late relapse > 6 months

96 (55.81)

Status post VenAza, (n) %

Complete remission

17 (10,90)

Relapse

113 (85,26)

Conclusions: Notwithstanding the retrospective nature, the results suggest that VenAza is an acceptable salvage therapy after alloHSCT and that the addition of DLI appears to improve survival with superior OS and EFS compared with non-DLI group, according to univariate and multivariate analysis.

Disclosure: Yves Chalandon. has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution.

19: Acute Leukaemia

P020 EFFICACY AND SAFETY ANALYSIS OF PROPHYLACTIC BRENTUXIMAB VEDOTIN ADMINISTRATION AFTER PEDIATRIC ACUTE LEUKEMIA HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION

Zhu Huili 1, Cao Xingyu1, Lu Yue1, Zhang Jianping1, Zhao Yanli1, Liu Deyan1, Xiong Min1, Sun Ruijuan1, Liu Hongxing1, Wei Zhijie1

1Hebei Yanda Lu Daopei Hospital, Langfang, China

Background: Acute graft-versus-host disease (aGVHD) is a common complication following allogeneic hematopoietic stem cell transplantation. Currently, the reported incidence of moderate to severe aGVHD in domestic cases is 13%-47%, while the incidence of grade III/IV aGVHD after haploidentical transplantation ranges from 7.9% to 13.8%. This study aims to analyze whether the administration of Brentuximab vedotin after transplantation can reduce the incidence of aGVHD without increasing disease relapse rates and viral infections.

Methods: A retrospective analysis was conducted on 50 pediatric leukemia patients who underwent haploidentical hematopoietic stem cell transplantation at our hospital between June 2017 and July 2022. These patients received prophylactic treatment with Brentuximab vedotin (20mg) on day +1 and day +4 post-transplantation. The median age of the patients was 4.1 years (range: 0.6-16.0 years). Among the cases, 22 were acute lymphoblastic leukemia (ALL), and 28 were acute myeloid leukemia (AML). Before transplantation, 28 patients were in CR1, 14 in CR2, 2 in CR3, and 6 in NR. All patients received myeloablative conditioning regimens, with 33 receiving Bu/Cy, 15 receiving TBI/Cy, 1 receiving Bu/Flu, and 1 receiving TBI/Flu. The median follow-up time was 29.1 months (range: 3.1-70.1 months). Detailed patient data are presented in Table 1.

Results: All 50 patients achieved 100% engraftment of white blood cells, with a median time of 15 days (range: +10 to +21 days). Platelet engraftment occurred in a median time of 9 days (range: +5 to +78 days), except for one patient who did not achieve platelet engraftment. The incidence of aGVHD after transplantation was 44%, with 28% experiencing grade I-II and 16% experiencing grade III-IV aGVHD. The incidence of CMV viremia was 40%, while the incidence of EBV viremia was 8%. During the follow-up period, five patients experienced disease relapse. Seven patients died during the follow-up, including 4 with AML and 3 with ALL. The causes of death were disease relapse in 3 cases, infection in 2 cases, TMA in 1 case, and cGVHD in 1 case. The estimated 5-year overall survival (OS) and leukemia-free survival (LFS) rates were 86%, and the 5-year cumulative relapse rate was 10%. In comparison, a control group of 44 pediatric acute T-cell lymphoblastic leukemia patients who underwent haploidentical transplantation without prophylactic Brentuximab vedotin treatment between January 5, 2021, and June 30, 2022, were analyzed. In the control group, the incidence of aGVHD was 56.8% (P = 0.007), with 36.4% experiencing grade I-II and 20.4% experiencing grade III-IV aGVHD. The incidence of CMV viremia was 68.2% (P = 0.006), and the incidence of EBV viremia was 29.5% (P = 0.007). The relapse rate was 15.9%, and the mortality rate was 27.3%.

Characteristics

Number of Cases

Total Number of Cases

50

Median Age(Range) Years

4.1 (0.6-16 years old)

Gender, Male (Number of Cases, %)

25 (50%)

Disease Diagnosis

Number of Cases

ALL (B-cell)

18 (including 4 cases with CNS involvement)

ALL (T-cell)

4 (including 1 case with CNS involvement)

AML

28 (including 7 cases of acute megakaryocytic leukemia)

Pre-transplant Status (Number, %)

CR1

28 (56%)

≥CR2

16 (32%)

NR

6 (12%)

Pre-conditioning Regimen (Number, %)

Bu/Cy

33 (66%)

TBI/Cy

15 (30%)

Bu/Flu

1 (2%)

TBI/Flu

1 (2%)

Infusion of MNC (Mean, ×108/kg)

14.9

Infusion of CD34+ (Mean, ×106/kg)

7.9

Infusion of CD3+ (Mean, ×108/kg)

3.2

Conclusions: For pediatric acute leukemia haploidentical HSCT, the administration of Brentuximab vedotin (20mg) on days +1 and +4 after transplantation, in addition to conventional aGVHD prophylaxis (CsA+MMF+sMTX), significantly reduced the overall incidence of aGVHD and grade I-II aGVHD. Administering Brentuximab vedotin on days +1 and +4 did not increase the rates of CMV or EBV infection nor did it increase the risk of disease relapse.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P021 OUTCOMES AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR ACUTE LEUKEMIA IN YOUNG ADULTS COMPARED WITH CHILDREN, ADOLESCENTS AND ELDERLY PATIENTS IN GERMANY

Jochen J. Frietsch 1,2, Sarah Flossdorf3,4, Ashrafossadat Ahmadian3,4, Claudia Schuh3, Thomas Schroeder5, Igor-Wolfgang Blau6, Matthias Stelljes7, Nicolaus Kröger3,8, Katharina Egger-Heidrich9, Matthias Eder10, Peter Dreger11, Johanna Tischer12, Eva Wagner-Drouet13, Rita Beier14,15, Peter Bader16, Martin Sauer15, Barbara Meissner14,15, Katharina Fleischhauer3,17, Inken Hilgendorf2

1Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany, 2Klinik für Innere Medizin II, Hämatologie und internistische Onkologie, Universitätsklinikum Jena, Jena, Germany, 3Deutsches Register für hämatopoetische Stammzelltransplantation und Zellherapie e.V., Ulm, Germany, 4Institut für Medizinische Informatik, Biometrie und Epidemiologie, Uniklinikum Essen, Essen, Germany, 5Klinik für Hämatologie und Stammzelltransplantation, Universitätsklinikum Essen, Essen, Germany, 6Medizinische Klinik mit Schwerpunkt Hämatologie, Onkologie und Tumorimmunologie, Campus Virchow-Klinikum, Berlin, Germany, 7Medizinische Klinik A, Hämatologie, Hämostaseologie, Onkologie und Pneumologie, Universitätsklinikum Münster, Münster, Germany, 8Klinik für Stammzelltransplantation, Universitätsklinikum Hamburg-Eppendorf, Hamburg, Germany, 9Medizinische Klinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany, 10Klinik für Hämatologie, Hämostaseologie, Onkologie und Stammzelltransplantation, Medizinische Hochschule Hannover, Hannover, Germany, 11Medizinische Klinik V, Universitätsklinikum Heidelberg, Heidelberg, Germany, 12Medizinische Klinik und Poliklinik III, Klinikum der Ludwig-Maximilians-Universität München, München, Germany, 13III. Medizinische Klinik und Poliklinik, Universitätsmedizin Mainz, Mainz, Germany, 14Pädiatrisches Register für Stammzelltransplantationen und Zelltherapien, Hannover, Germany, 15Klinik für Pädiatrische Hämatologie und Onkologie, Medizinische Hochschule Hannover, Hannover, Germany, 16Klinik für Kinder- und Jugendmedizin, Studienzentralen der Schwerpunkte Onkologie, Hämatologie und Hämostaseologie sowie Stammzelltransplantation, Immunologie und Intensivmedizin, Universitätsklinikum Frankfurt, Frankfurt, Germany, 17Institut für Zelltherapeutische Forschung, Universitätsklinikum Essen, Essen, Germany

Background: The survival after allogeneic hematopoietic SCT is influenced by the patients’ age, diagnosis, donor type and achieved remission. Nevertheless, with a 5-year overall survival (OS) probability of 53%, the outcome of young adults (YA) aged 18 to 39 years is sobering. We sought to provide more detail on outcome by focusing on acute leukemia (AL).

Methods: The German Registry for Hematopoietic Cell Transplantation and Cellular Therapy (DRST) and the German Pediatric Registry for Stem Cell Transplants and Cell Therapies (PRSZT) were screened for patients suffering from AL who received 1st SCT between 2011 and 2019. OS rates were determined by Kaplan-Meier analysis and differences between were evaluated using Score-tests (corresponding to the log-rank test, computed when Hazards are not crossing). Gray’s test was applied to cumulative incidence of NRM using competing risk analysis. To separate relapse from progression, patients not being in complete remission at the time of SCT or within 100 days were excluded from competing risk analysis. A p-value of <0.05 was considered statistically significant.

Results: Altogether 11258 patients were included, with 573, 269, 2158, 4667 and 3591 in the age groups 0-12, 13-17, 18-39, 40-59 and 60-72 years at the time of SCT, respectively. 2424 (21.5%) suffered from acute lymphoblastic leukemia (ALL) and 8834 (78.5%) from acute myeloid leukemia (AML). The majority (89.6%, 10089) of the predominantly male (56.2%, 6331) patients received peripheral blood SCT from a related (28.4%, 3192) or an unrelated (71.6%, 8066) donor with varying degrees of histocompatibility. During the observation period, 4380 (38.9%) patients deceased.

The probability of 5-year OS decreased for AML with increasing patient age: 0-12: 69%; 13-17: 60%; 18-39: 57%; 40-59: 49%; and 60-72 years: 37% and for ALL: 0-12: 73%; 13-17: 71%; 18-39: 53%; 40-59: 51%; and 60-72 years: 37%. Statistically significant differences in the probability of 5-year OS were observed between related and unrelated donors for AML (48% vs. 46%, p = 0.0018), but not for ALL (52% vs. 57%). No statistically significant differences could be observed concerning gender for AML (♀ 49% vs. ♂ 45%, p = 0.0508) and ALL (♀ 57% vs. ♂ 55%).

The 5-year-NRM increased with patient age for both ALL (0-12: 7%; 13-17: 10%; 18-39: 19%; 40-59: 26% and 60-72: 30%; p<0.001) and AML (5%; 14%; 12%; 20% and 30%; p<0.001). The 5-year relapse rate in both entities was lower for 13-17 year old adolescents (AML/ALL 13-17: 26%/18%) than for the other age groups (0-12: 33%/28%, 18-39: 40%/31%, 40-59: 34%/28%, 60-72: 35%/32%).

Conclusions: In this preliminary large retrospective multicenter analysis we show that NRM increases with patient age which also negatively impacts OS. The heterogeneity of the cohort was taken into account by considering diseases separately and age in groups. As differences in patient outcome are being reflected by a distinct disease biology, further investigations are necessary to fully elucidate differences between AML and ALL in age-dependent survival. The increasing NRM with respect to age underlines the relevance of interdisciplinary collaboration between pediatricians and non-pediatricians for the improvement of aftercare concepts, especially for children, adolescents and YA.

Disclosure: IH is chairperson of the board of trustees of the German Foundation for Young Adults with Cancer, received research funding from the H.W. and J. Hector-Foundation and honoraria from AbbVie, Medac, Novartis and Takeda. JF received honoraria from Novartis. All other authors do not declare any conflicts of interest. The Cooperative German Transplant Study group is acknowledged for project development and support.

19: Acute Leukaemia

P022 PHASE I/II STUDY OF CYCLOPHOSPHAMIDE AND BENDAMUSTINE AS POST-TRANSPLANTATION GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS IN REFRACTORY MYELOID NEOPLASMS

Ivan Moiseev 1, Olga Epifanovskaya1, Ksenia Afanasyeva1, Anastasia Beynarovich1, Dmitrii Zhogolev1, Mikhail Kanunnikov1, Yulia Rogacheva1, Tatyana Rudakova1, Nikita Volkov1, Zhamshidbek Khudaiberdiev1, Azamjon Meliboev1, Yulia Vlasova1, Elena Morozova1, Sergei Bondarenko1, Alexander Kulagin1

1RM Gorbacheva Research Institute, Pavlov University, Saint Petersburg, Russian Federation

Background: Efficacy of allogeneic hematopoietic stem cell transplantation (alloHCT) in disease progression of myeloid neoplasms resistant to chemotherapy or targeted therapy is low. Our group recently demonstrated an enhanced graft-versus-leukemia (GVL) effect when replacing cyclophosphamide with bendamustine in a graft-versus-host disease (GVHD) prophylaxis regimen (Moiseev IS et al., TCT, 2021), but there was significant toxicity from this regimen from a poorly controlled cytokine release syndrome (CRS). To reduce toxicity while preserving GVL, we conducted a pilot single-center study of graft-versus-host disease (GVHD) prevention with a combination of cyclophosphamide and bendamustine in refractory myeloid neoplasms.

Methods: Prospective single-center Phase I/II study (NCT04943757) enrolled 50 patients (table 1). Inclusion criteria were myeloid neoplasm, available hematopoietic cell donor, >5% clonal blasts at the start of conditioning, disease refractory to at least one course of induction therapy. Main exclusion criteria: absence of uncontrolled infection and severe concomitant co-morbidities. The protocol included the administration of bendamustine 50 mg/kg/day on days +3, + 4, cyclophosphamide 25 mg/kg/day on days +3, + 4 (PTBC), tacrolimus 0.03 mg/kg from day+5 to day+ 100 day and mycophenolate mofetil 30 mg/kg/day on days 5-35. Patients received FB2 or FB3 conditioning regimen according to their age and performance status.

Characteristics of the study group

N = 50

Diagnosis

Acute myeloid leukemia

37

Myelodysplastic syndrome

9

Chronic myeloid leukemia

2

Chronic myelomonocytic leukemia

1

Atypical chronic myeloid leukemia

1

Age, median years (range)

48 (18-69)

Karnofsky index, median (range)

80% (60-90%)

Gender

Males

34

Females

16

Matched related donor

10

Matched unrelated donor

26

Haploidentical donor

14

Conditioning

FB2 conditioning

33

FB3 conditioning

13

Other conditioning

4

Active antimicrobial therapy at the time of HCT

20

Second HCT

4

Co-morbidity index

HTC-CI 0

24

HTC-CI 1

17

HTC-CI 2

6

HTC-CI >2

3

Primary refractory disease

26

Secondary refractory disease

24

Complex karyotype

10

High-risk somatic mutations

22

Median % of blasts at the start of conditioning (range)

12% (5-86%)

Results: The cumulative engraftment rate was 86%. Only 1 patient died before engraftment assessment. The median time to recovery of white blood cells was 18 days (range 12-35), platelets - 14 days (range 9-104). Remission was achieved in 88% of patients, with 76% of patients negative for minimal residual disease (MRD). Overall survival (OS) was 33% (95%CI 19-48%), event-free survival (EF) - 21% (95%CI 10-35%). Non-relapse mortality (NRM) was 19% (95%CI 9-31%), cumulative relapse incidence was 60% (95%CI 43-74%). Toxicity included development of CRS in 30% of patients, including 2 patients with grade 4-5 CRS. The most common manifestation of CRS was fever, increased ferritin and abnormal liver function tests (24% of patients). The cumulative incidence of grade II-IV acute GVHD was 20% (95%CI 10-33%), grade III-IV – 18% (95%CI 7-31%), moderate and severe chronic GVHD - 18% (95%CI 8 -31%). Grade II-IV acute GVHD was observed predominantly in patients with CRS grades 2-5 (31% vs. 16%, p = 0.02). Survival subanalysis showed that unrelated or haploidentical donor transplantation was associated with better OS (40% vs 0%, p = 0.008). Unique tolerance mechanisms were revealed with a massive expansion of PD-1L positive monocytes by day +30 (17.7±10.9% of all nucleated cells [NC]) with a decline to 7.1±4.3% and 6.9±6.5% of NC by days +60 and +100 (p<0.001). The level of PD-1L granulocytes was also high: 13.2±10.9%, 8.1±9.4% and 9.0±9.0% of NC on days 30, 60 and 100 (p = 0.4). This was accompanied by rapid expansion of effector T-cells (p<0.001), effector memory T-cells (p<0.001), central memory T-cells (p<0.001), naive T-cells (p<0.0001) and CD197-positive activated T-cells (p<0.0001) early post-transplant. In several patients abnormally high levels of monocytes and lymphocytes persisted for 2 years after HCT.

Conclusions: We developed a promising alloHCT platform for refractory myeloid neoplasms with a unique immunological recovery profile.

Clinical Trial Registry: NCT04943757, clinicaltrials.gov

Disclosure: The study was supported by RSF grant №23-15-00327.

19: Acute Leukaemia

P023 REAL-WORLD OUTCOME OF ACUTE MYELOID LEUKEMIA PATIENTS TREATED WITH ALLOGENEIC STEM CELL TRANSPLANTATION WITHOUT STANDARD INDUCTION CHEMOTHERAPY

Francesca Biavasco 1, Kristina Maas-Bauer1, Jesus Duque-Afonso1, Ralph Waesch1, Michael Luebbert1, Justus Duyster1, Robert Zeiser1, Juergen Finke1, Claudia Wehr1

1University Hospital Freiburg, Freiburg, Germany

Background: Allogeneic hematopoietic cell transplantation (allo-HCT) has become an accessible curative option in elderly patients with acute myeloid leukemia (AML) after development of reduced-intensity conditioning regimen and the introduction of hypomethylating agents (HMA) with venetoclax as induction therapy for elderly unfit patients. While a recent prospective randomised trial showed non-inferior efficacy and better tolerance of HMA over standard chemotherapy in elderly patients, only half of the studied cohort underwent allo-HCT and venetoclax was not included in the study (Luebbert et al., Lancet Haematol. 2023 Nov;10(11):e879-e889). Therefore, real-world data on outcomes of patients not eligible for intensive chemotherapy who received allo-HCT in the venetoclax era are needed.

Methods: We retrospectively analysed data of 106 adult AML patients who underwent allo-HCT without receiving intensive chemotherapy at our center between 2010 and 2023.

Results: Forty-two patients received HMA alone, 24 patients received HMA-venetoclax induction therapy, and 40 patients underwent allo-HCT without previous therapy (upfront) due to personalised clinical decision. Patients who received allo-HCT upfront were younger (median 59.1 years) than HMA (64.9 years) and HMA-venetoclax patients (70.9 years respectively, p<0.0001). The groups did not differ in Karnofsky performance status, ELN risk distribution and time from diagnosis to allo-HCT. Forty-three of the HMA treated (83%) and 15 of the HMA-venetoclax treated patients (62%) did not achieve CR before allo-HCT, resulting in a similar mean blast count before transplantation among groups (41.4 for upfront, 26.5 for HMA and 32.2 for HMA-venetoclax). With a median follow-up of 85.5, 75.4 and 18.9 months in upfront, HMA treated and HMA-venetoclax treated patients, the overall survival at median follow-up time (19 months) was 75.7%, 54.8% and 89.0% respectively, showing a better survival for patients treated with addition of BCL-2 inhibitor. Interestingly, the difference in survival was not reflected in marked difference of progression free survival, which was similar among groups (19-months progression free survival 77.8% upfront, 72.3% HMA and 73.9% HMA-venetoclax). Of note, overall survival benefits for HMA-venetoclax treated patients persisted in the subgroup of patients who did not reach a complete remission before transplant (19-months overall survival for HMA 51.5%, for HMA-venetoclax 82.3%). As expected though, the achievement of a complete remission before allo-HCT was associated with a better survival in both groups (19-months overall survival HMA CR 66.7%, HMA non-CR 51.5% vs. HMA-venetoclax CR 100%, HMA-venetoclax non-CR 82.3%).

Conclusions: These real-world retrospective data on the avoidance of intensive chemotherapy in pre-transplant setting suggest that HMA-venetoclax could be the best therapeutic option for patients with AML. Additionally, the advantage of HMA-venetoclax was independent from achieving CR, even though the achievement of CR is associated with better survival. Confirmatory data of prospective randomised trials are urgently needed to establish the best possible pre-transplant treatment for non-highly proliferating AML patients.

Disclosure: K. M-B is supported by a fellowship of the Faculty of Medicine, Freiburg University (Berta-Ottenstein-Programm) and by the Else-Kröner-Fresenius-Stiftung Nr: 2021_EKEA.131

R.W. received research fundings from Janssen, Sanofi, consultancy: from Amgen, BMS/Celgene, Janssen, Kite/Gilead, Novartis, Pfizer, Sanofi, Takeda, honoraria from Abbvie, Alexion/Astra Zeneca, Amgen, BMS/Celgene, Janssen, Kite/Gilead, Novartis, Pfizer, Sanofi, Takeda and travel support from Janssen, Kite/Gilead, Pfizer, BMS.

C.W. received honoraria/travel grant from Takeda and Jazz Pharmaceuticals.

19: Acute Leukaemia

P024 DURING CB TRANSPLANT IN MRD-POSITIVE AML, RELAPSE IS USUALLY EARLY, IS REDUCED BY ACUTE GVHD AND IS INFLUENCED BY CICLOSPORIN EXPOSURE: A MULTICENTER NATIONAL EXPERIENCE

Srividhya Senthil 1, Abdul Moothedath1, Archana Rauthan2, Ioannis Peppas3, Sandeep Potluri4, Pamela Evans5, Valerie Broderick5, Sarah Lawson4, Emma Barrett6, Caroline Furness7, Kanchan Rao2, Robert Wynn1

1Royal Manchester Children’s Hospital, Manchester, United Kingdom, 2Great Ormond Street Hospital, London, United Kingdom, 3Kings College Hospital NHS Foundation Trust, London, United Kingdom, 4Birmingham Children’s Hospital, Birmingham, United Kingdom, 5Royal Crumlin Hospital, Dublin, Ireland, 6Manchester University Foundation Trust, Manchester, United Kingdom, 7Royal Marsden Hospital, Manchester, United Kingdom

Background: Ciclosporin forms the major GVHD prophylaxis in allogeneic stem cell transplantation. It might be expected to reduce the beneficial graft-versus-leukemia [GVL] effect, and previous studies have reported a relationship between ciclosporin exposure and risk of post-transplant relapse in leukaemia. T cell replete Cord blood (TRCBT) HSCT might be considered the preferred donor cell source in AML with superior GVHD-free, Relapse-free survival (RFS). We report a multicentric experience of the relationship between ciclosporin exposure and time to relapse in a large cohort of paediatric AML patients receiving such a graft.

Methods: It is a retrospective study, data was collected from 5 paediatric transplant centers in the UK on the patients who had a TRCBT for HR AML between 2013 and 2023. Case records were analyzed to get patient, donor, transplant characteristics, GVHD prophylaxis, trough ciclosporin levels in the first 8 weeks and the transplant outcomes including GVHD and its treatment, relapse and transplant related mortality (TRM). The primary endpoint was time to disease relapse, and the relationship between this endpoint and ciclosporin AUC, use of additional immunosuppressive treatment (IST) and its total duration was investigated by Cox regression analysis.

Results:

Demographics and outcomes

No of patients / %

Disease Burden:

MRD-positive

64 (59.25%)

MRD-negative

44 (40.7%)

Severe aGVHD:

Skin

25 (23%)

Gut

28 (26%)

Required systemic IST

56 (51.8%)

Mortality in MRD-positive group:

32

Relapse associated

22(68.7%)

TRM

10 (31.2%)

Mortality in MRD-negative group:

12

Relapse associated

5 (41.6%)

TRM

7 (58.3%)

GVHD related deaths:

4

MRD-positive group

2

MRD-negative group

2

Relapse:

Very early (<6 months)

21 (61.7%)

Early (6-12 months)

11 (32.3%)

Late (>12 months)

2 (5.8%)

The cohort included 108 patients, of whom 64 were MRD-positive pre-transplant, with a median follow up of 737.1 days (range: 24-3540 days). All were given ciclosporin and MMF as GVHD prophylaxis for a variable duration post-transplant according to each centers’ protocol. The severe aGVHD was treated with IST which included steroids in all with or without second agents.

The mortality rate was 40.7% of which nearly three-quarters occurred in the MRD-positive group. Overall, relapse formed the leading cause of death, causing 68.7% of the total deaths in MRD-positive group. It constituted only 41.6% of the deaths in the MRD negative group. The median time to relapse was 180 days (range: 32 to 949) and nearly a third occurred very early within 6 months. TRM was 13% and accounted for the most prominent cause of mortality in MRD-negative group. The GVHD related mortality was very low.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (2)

The RFS is significantly greater in MRD-negative group without severe aGVHD than in those without. By comparison, the RFS is significantly lower in the MRD-positive group without severe aGVHD than in those with it. The COX proportional hazards model using 95% CI confirmed a significant inverse relationship of AUC of Ciclosporin in the first 8 weeks on the time to relapse (p<0.02). However, it did not find any effect of IST or the duration of IST on time to relapse (p = 0.579 and p = 0.623 respectively).

Conclusions: This is the largest homogenous cohort of TRCBT in AML investigating the role of ciclosporin prophylaxis on relapse. It confirms the importance of GVL effect and its relation to GVHD in MRD-positive patients as RFS is significantly better in those who experience aGVHD. Most relapse is early and increased ciclosporin exposure in the first 2 months of HSCT reduces the time to relapse in AML. This study supports the development of novel protocols and alternative IS regimens during CBT in the highest risk AML.

Clinical Trial Registry: Not applicable

Disclosure: The authors have no conflict of interest.

19: Acute Leukaemia

P025 IMPACT OF RESPONSE TO THE FIRST INDUCTION COURSE IN AML PATIENTS ON ALLOHSCT OUTCOMES WITH PTCY

Dmitrii Zhogolev1, Bella Aybova1, Anna Smirnova1, Yulia Vlasova1, Daria Chernyshova1, Elena Babenko1, Tatyana Gindina1, Ildar Barkhatov1, Elena Morozova1, Ivan Moiseev 1, Sergey Bondarenko1, Alexander Kulagin1

1RM Gorbacheva Research Institute, St. Petersburg, Russian Federation

Background: For the majority of patients with acute myeloid leukemia (AML), allogeneic hematopoietic stem cell transplantation (alloHSCT) remains as the sole curative option. Recent advancements in transplant technologies, such as post-transplant cyclophosphamide (PTCy)-based GVHD prophylaxis, have enhanced the overall outcomes of alloHSCT. It is well-established that patients refractory to 2 induction courses, even with subsequent remission achievement, exhibit poorer outcomes after alloHSCT. However, the impact of refractoriness to the first induction course remains unclear, particularly within the context of PTCy.

Methods: A total of 260 adult patients with AML in first complete remission (CR1), who underwent alloHSCT at RM Gorbacheva Research Institute (CIC 725) from 2013 to 2022 were included in this retro- and prospective study. All participants received the “7 + 3” regimen as their initial induction therapy. Overall survival (OS), leukemia-free survival (LFS), and GVHD-free, relapse-free survival (GRFS) were assessed using the Kaplan-Meier method and log-rank test. Multivariate analysis was conducted utilizing a Cox regression model. To estimate relapse incidence (RI) and non-relapse mortality (NRM) within a competing risk framework, cumulative incidence functions were applied.

Results: Achievement of CR1 occurred after 1 induction course in 158 patients, after 2 induction courses in 68 patients, and after more than 2 induction courses in 34 patients. Groups were balanced on key parameters, with the exception of a higher incidence of secondary AML in the non-refractory group (p = 0.026) and a higher prevalence of unfavorable cytogenetics in the 2 courses refractory group (p = 0.041) (Table 1).

Table 1. Patient, disease and transplant characteristics.

Non-refractory

Refractory to 1 induction course

Refractory to 2 induction courses

p. overall

N = 158

N = 68

N = 34

Gender, n(%)

0.332

Female

74 (46.8)

39 (57.4)

16 (47.1)

Male

84 (53.2)

29 (42.6)

18 (52.9)

Diagnosis, n(%)

0.026

De novo AML

133 (84.2)

64 (94.1)

33 (97.1)

Secondary AML

25 (15.8)

4 (5.9)

1 (2.9)

Cytogenetics, n(%)

0.041

Fav/Int

138 (87.3)

59 (86.8)

24 (70.6)

Adv

20 (12.7)

9 (13.2)

10 (29.4)

Age at transplant, median (range)

38 (18-66)

33 (19-65)

36 (18-61)

0.185

MRD status at transplant, n(%)

0.442

Negative

101 (63.9)

43 (63.2)

19 (55.9)

Positive

38 (24.1)

21 (30.9)

10 (29.4)

Missing

19 (12)

4 (5.9)

5 (14.7)

Donor, n(%)

0.122

MRD

38 (24.1)

16 (23.5)

2 (5.8)

MUD

68 (43)

25 (36.8)

14 (41.2)

MMUD

27 (17.1)

18 (26.5)

9 (26.5)

HID

25 (15.8)

9 (13.2)

9 (26.5)

Conditioning intensity, n(%)

0.073

MAC

17 (10.8)

15 (22.1)

6 (17.6)

RIC

141 (89.2)

53 (77.9)

28 (82.4)

GVHD prophylaxis, n(%)

0.105

CyTxMMF

116 (73.4)

51 (75)

26 (76.5)

MonoCy

32 (20.3)

12 (17.6)

2 (5.9)

CyRuxo

10 (6.3)

5 (7.4)

6 (17.6)

Graft source, n(%)

0.539

PBSC

116 (73.4)

50 (73.5)

28 (82.4)

BM

42 (26.6)

18 (26.5)

6 (17.6)

Median follow up after alloHSCT in survivors was 42.3 (3-108.5) months. Engraftment on d28 was attained in 86% (95%CI, 80-91), 90% (95%CI, 79-95), and 88% (95%CI, 70-96) in the non-refractory, refractory to 1, and refractory to 2 courses groups, respectively (p = 0.8). Three-year OS rates were 81.7% (95%CI, 75.7-88.1), 66.5% (95%CI, 55.6-79.5), and 60.1% (95%CI, 44.5-81.2) (p = 0.038), LFS – 78.6% (95%CI, 72.2-85.5), 66.8% (95%CI, 56-79.8), and 40.5% (95%CI, 25.3-64.8) (p<0.001), GRFS – 69.3% (95%CI, 62.2-77.2), 49.5% (95%CI, 38.6-63.6), and 36.5% (95%CI, 22.3-59.8) (p<0.001). The cumulative incidence of NRM at three years was 14% (95%CI, 8.7-19), 16% (95%CI, 8-26), and 19% (95%CI, 7.3-34) (p = 0.8), RI – 7.9% (95%CI, 4.1-13), 17% (95%CI, 9.1-28), and 41% (95%CI, 22-59) (p<0.001). There was no significant statistical difference in aGVHD grades 2-4 (p = 0.22) and grades 3-4 (p = 0.074), cGVHD grades 1-3 (p = 0.93) and grades 2-3 (p = 0.61) incidence.

In the multivariate analysis, failure to achieve remission after one “7 + 3” course was associated with lower OS (HR 1.97, p = 0.02) and GRFS (HR 1.96, p = 0.004) after alloHSCT, with a tendency for lower LFS (HR 1.72, p = 0.056). Age and refractoriness to 2 induction courses were also associated with lower OS, LFS, and GRFS.

Conclusions: This study highlights the failure to respond to one “7 + 3” course as a negative prognostic factor for both OS and GRFS after alloHSCT, when performed with PTCy. The findings underscore the necessity for novel treatment approaches to enhance survival outcomes in these patients. Additionally, there is a crucial need for improved strategies in patients with primary refractory AML and older patients.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P026 POST-TRANSPLANT TYROSINE KINASE INHIBITOR MAINTENANCE IN PH-POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA DELIVERS BENEFIT TOWARD IMPROVED OVERALL, RELAPSE-FREE, GVHD/RELAPSE-FREE SURVIVAL AND DECREASED NON-RELAPSE MORTALITY WITHOUT REDUCED RELAPSE

Eshrak Al-Shaibani 1, Carol Chen1, Igor Novitzky-Basso1,2, Ivan Pasic1,2, Auro Viswabandya1,2, Rajat Kumar1,2, Wilson Lam1,2, Arjun Law1,2, Armin Gerbitz1,2, Jonas Mattson1,2, Fotios V. Michelis1,2, José-Mario Capo-chichi3, Dennis D. Kim1,2

1Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Canada, 2University of Toronto, Toronto, Canada, 3University Health Network and University of Toronto, Toronto, Canada

Background: Post-allogenic hematopoietic cell transplantation (HCT) maintenance therapy with tyrosine kinase inhibitor (TKI) in Philadelphia positive acute lymphoblastic leukemia (Ph+ALL) is expected to reduce the risk of relapse. However, clinical benefit from the post-HCT TKIs maintenance (PTM) in Ph+ALL remains still on debate due to conflicting results from the retrospective studies and lack of randomized controlled trial. Herein, we report the long-term outcomes of the patients who received PTM following allogeneic HCT versus not in Ph+ALL.

Methods: We have retrospectively reviewed 80 patients with Ph+ ALL underwent first HCT in complete remission at Princess Margaret Cancer Center from 2000 till 2022. Relapse was defined as morphologic recurrence of ≥5% blasts in bone marrow or by the presence of extramedullary disease. Overall (OS), graft-versus host disease (GVHD)/relapse-free (GRFS) and relapse-free survival (RFS) was calculated by the Kaplan-Meier method and analyzed using a log-rank test, respectively. Cumulative incidence of relapse (CIR), non-relapse mortality (NRM) and chronic GVHD (cGVHD) were calculated considering competing events and analyzed using Fine-Gray model.

Results: Patients’ and disease characteristic are summarized in Table 1. With a median follow-up duration of 26 months among survivors, 16 patients (20%) had progressed following HCT. Fifty-eight patients (72%) did not receive PTM while 22 (28%) patients received PTM. The median time to start PTM was 5.3 months (range; 2.6-9.2) with imatinib (n = 15), dasatinib (n = 5) or ponatinib (n = 4). ABL1 kinase-domain mutation (KDM) was detected in 8 patients: T315I (n = 2), F317L (n = 3), E255K (n = 1), Y253H (n = 1), Q252H (n = 1).

The analysis for OS, GRFS, RFS, NRM, cGVHD and CIR at 2 years showed improved outcomes in favor of PTM: OS, 81% vs 43.4% (p = 0.01); GRFS, 40% vs 5.3% (p = 0.003); RFS, 81% vs 43.4% (p = 0.02); NRM, 4.8% vs 45.8% (p = 0.003); cGVHD 27.7% vs 41.3% (p = 0.54); CIR, 14.3% vs 10.8% (p = 0.19).

Univariate analysis showed PTM improved OS (HR; 0.39 [0.19-0.85], P = 0.02), RFS (HR: 0.54 [0.29-0.99], P = 0.03), GRFS (HR: 0.41 [0.23-0.75], P = 0.004) and NRM (HR: 0.045 [0.003-0.20], P = 0.003). However, it was not associated with relapse risk (HR: 0.143 [0.03-0.33], P = 0.19).

Besides PTM, age >40 was associated with OS (HR: 2.08 [1.11-3.9], P = 0.02), RFS (HR: 1.85 [1.01-3.37], P = 0.05) and NRM (HR: 0.41 [0.27-0.55], P = 0.04). Time -dependent cGVHD found to be associated with improved OS (HR: 0.36 [0.16-0.78], P = 0.009), RFS (HR: 0.31 [0.14-0.68], P = 0.003) and CIR (HR: 0.1522 [0.04-0.54], P = 0.004). Detectable ABL-KDM at ant time prior-HCT increased risk of relapse (HR: 0.38 [0.07-0.69], P = 0.0005).

Multivariable analysis confirmed that PTM improved OS (HR; 0.28 [0.13-0.62], P = 0.02], RFS (HR: 0.32 [0.15- 0.68], P = 0.003), GRFS (HR: 0.41 [0.23-0.75], P = 0.004) and NRM (HR:0.18 [0.06-0.57], P = 0.003). cGVHD also found to improve OS (HR: 0.28 [0.13-0.64], P = 0.002), PFS (HR: 0.23 [0.10-0.51], P = 0.003) and decreased CIR (HR: 0.33 [0.09-1.14], P = 0.08).

Table 1: patients characteristics

Variables

TKI maintenance

No TKI-maintenance

P

N = 22

N = 58

Median age (range)

54 (19-63)

44 (19- 65)

0.2

Gender, N (%)

0.09

Female

14 (63.6)

24 (41.4)

Male

8 (36.4)

34 (58.6)

Disease status, N (%)

0.04

CR1

15 (68.2)

52 (89.7)

CR2

7 (31.8)

6 (10.3)

BCR/ABL pre-HCT, N (%)

0.57

MMR

18 (81.8)

43 (74.1)

No MMR

4 (18.2)

15 (25.9)

ABL Kinase mutation

0.03

Positive

5 (22.7)

3 (5.2)

Negative

17 (77.3)

55 (94.8)

Conditioning regimen, N (%)

0.09

MAC

13 (59.1)

46 (79.3)

RIC

9 (40.9)

12 (20.7)

GVHD prophylaxis, N (%)

0.005

Dual T-cell depletion

8 (36.4)

5 (8.6)

Others

14 (63.6)

53 (91.4)

PTCY based GVHD, N (%)

<0.001

Yes

10 (45.5)

5 (8.6)

No

12 (54.5)

53 (91.4)

Donor type, N (%)

0.17

MRD

9 (40.9)

27 (46.6)

MUD

10 (45.5)

23 (39.7)

MMUD

6 (10.3)

Haploidentical

3 (13.6)

2 (3.4)

Stem sources

0.49

PBSC

20 (90.9)

48 (82.8)

BMSC

2 (9.1)

10 (17.2)

OS

0.02

Alive

14 (63.6)

19 (32.8)

Death

8 (36.4)

39 (67.2)

NRM

0.005

Alive

19 (86.4)

28 (48.3)

Death

3 (13.6)

30 (51.7)

RFS

0.04

No

13 (59.1)

18 (31)

Yes

9 (40.9)

40 (69)

Relapse

0.49

No

16 (72.7)

48 (82.8)

Yes

6 (27.3)

10 (17.2)

Acute GVHD

0.79

Yes

14 (63.6)

39 (67.2)

No

8 (36.4)

19 (32.8)

Chronic GVHD

0.79

Yes

7 (31.8)

22 (37.9)

No

15 (68.2)

36 (62.1)

  1. Abbreviation: BM: bone marrow, CR: complete remission, HCT: allogenic hematopoietic cell transplantation, MAC: myeloablative conditioning, MMR: Major molecular response, MRD: matched related donor, MUD: matched unrelated donor, MMUD: mismatch unrelated donor, NRM: non-relapse mortality, OS: overall survival, PBSC: peripheral blood stem cell transplant, PFS: progressive-free survival, PTCY: post-transplant cyclophosphamide, RIC: reduce intensity regimen.

Conclusions: PTM in Ph+ALL delivers a clinical benefit toward better OS, improved RFS, decreased NRM and better GRFS. However, it was not confirmed whether PTM can reduce the risk of relapse.

Disclosure: Nothing to declare

19: Acute Leukaemia

P027 IMPACT OF DONOR AND STEM-CELL SOURCE IN ADULTS WITH HIGH-RISK PHILADELPHIA-NEGATIVE ACUTE LYMPHOBLASTIC LEUKEMIA TRANSPLANTED IN FIRST COMPLETE REMISSION: A STUDY FROM GRAALL AND SFGM-TC

Matthieu Jestin1, Sébastien Maury2, Anne Huynh3, Edouard Forcade4, Ibrahim Yakoubagha5, Cristina Castilla-Llorente6, Jean-Baptiste Mear7, Urs Schanz8, Sylvain Chantepie9, Jakob Passweg10, Nathalie Contentin11, Yves Chalandon12, Nicole Raus13, Véronique Lheritier13, Natacha Maillard14, Marie-Thérèse Rubio15, Raynier Devillier16, Patrice Ceballos17, Sylvie François18, Stéphanie Nguyen-Quoc19, Patrice Chevallier20, Hélène Labussière-Wallet13, Hervé Dombret1, Nicolas Boissel1, Nathalie Dhédin 1

1Hôpital Saint Louis, Paris, France, 2Hôpital Henri Mondor, Créteil, France, 3Institut universitaire du Cancer de Toulouse, Toulouse, France, 4CHU de Bordeaux, Bordeaux, France, 5CRHU Lille, Lille, France, 6Institut Gustave Roussy, Villejuif, France, 7CHU de Rennes, Rennes, France, 8Hôpital Universitaire de Zurich, Zurich, Switzerland, 9CHU de Caen, Caen, France, 10Hôpital Universitaire de Bâle, Bâle, Switzerland, 11Centre Henri Becquerel, Rouen, France, 12Hôpitaux Universitaires de Génève, Génève, Switzerland, 13CHU de Lyon, Lyon, France, 14CHU de Poitiers, Poitiers, France, 15CHRU de Nancy, Nancy, France, 16Institut Paoli-Calmettes, Marseille, France, 17CHU de Montpellier, Montpellier, France, 18CHU d’Angers, Angers, France, 19Hôpital Pitié-Salpétrière, Paris, France, 20CHU de Nantes, Nantes, France

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) stands as a cornerstone in managing high-risk (HR) Philadelphia chromosome (Ph)-negative acute lymphoblastic leukemia (ALL) in the first complete remission (CR1). Whereas unrelated donor (UD) has become a standard option in absence of matched sibling donor (MSD), peripheral blood (PB) is mostly used despite a higher incidence of graft-versus-host disease (GVHD). The aim of this study was to evaluate the impact of donor and stem cell source in adults with HR Ph-negative ALL in CR1 prospectively included in the GRAALL-2003 and -2005 trials.

Methods: Between November 2003 and March 2014, 311 patients with HR Ph-negative ALL in CR1 were included in the GRAALL-2003/05 trials and received a HSCT from MSD or UD after a myeloablative conditioning (MAC) regimen. Cord blood (n = 17) or haploidentical transplant (n = 2) were excluded. The primary objective was to assess the impact of donor and stem cell source on acute grade III-IV or chronic extensive GVHD-free relapse-free survival (GRFS). Type of donor (MSD versus UD), use of anti-thymocyte globulin (ATG) and total body irradiation (TBI), stem cell source (PB versus bonne marrow (BM)) and age at transplant were included in univariate and multivariate analyses for GRFS, overall survival (OS), event-free survival (EFS), cumulative incidence of relapse (CIR), non-relapse mortality (NRM), incidence of aGVHD grade III-IV and of extensive cGVHD.

Results: Among the 311 patients, 100 had a T-ALL and 211 a Ph-negative BCP-ALL. Median age at transplant was 31.8 years (IQR, 29.7 – 34.5) with 60% of male. All received a MAC regimen, 293 (94%) a TBI-based, 14 (5%) a busulfan-based, and 4 another regimen. Transplant was performed from a MSD in 156 patients (50%) and from a 9/10 or 10/10 matched UD in 155 patients (50%). PB and BM were used in 105 patients (34%) and 206 patients (66%), respectively. ATG was part of the conditioning regimen in 58 patients (19%). In univariate and multivariate analysis for GFRS, after adjustment on age at transplant, use of ATG and TBI, and type of donor, stem cell source (PB vs BM) emerges as the sole variable significantly associated with GRFS (Table). Of note, the type of donor and the use of ATG did not show significant association with GRFS, and also lacked significant correlation with OS, EFS, CIR, TRM, cGVHD and aGVHD. In multivariate analysis using the same covariates, PB was significantly associated with an increased risk of NRM (SHR 2.03, 95%CI(1.19-3.48)), an increased incidence of cGVHD (SHR 3.20, 95%CI(1.79-5.76)) and a decreased CIR (SHR 0.51, 95%CI(0.27-0.96)), but did not correlate with OS and EFS. Interestingly, in sub-groups analysis, GRFS was significantly improved using BM versus PB from UD (HR 2.25, 95%CI(1.46-3.47)) whereas no impact of stem cell source was observed in transplant from MSD.

Variables

Hazard ratio value

95%CI

p

Age at HSCT (y)

1.00

0.99-1.01

0.95

UD vs MSD

1.11

0.79-1.56

0.56

ATG

0.68

0.43-1.08

0.10

PB vs BM

1.54

1.11-2.14

0.01

TBI

0.95

0.60-1.18

0.87

Conclusions: In adults with HR Ph-negative ALL in CR1, PB and BM are two acceptable options when a MSD is available. Despite the absence of impact of stem cell source on OS, the use of UD BM should be preferred over PB, since it improves long-term quality of life.

Clinical Trial Registry: NCT00327678

Disclosure: Nothing to declare.

19: Acute Leukaemia

P028 HOW DISEASE STATUS IMPACTS ON LONG-TERM SURVIVAL AFTER ALLOGENEIC TRANSPLANT IN ACUTE MYELOID LEUKEMIA: ANALYSIS ON 456 PATIENTS

Alessandro Bruno1, Matteo Giovanni Carrabba1, Simona Piemontese1, Raffaella Greco1, Sarah Marktel1, Sara Mastaglio1, Daniela Clerici1, Francesca Farina1, Elisa Diral1, Luca Vago1, Lorenzo Lazzari1, Edoardo Campodonico1, Giulia Furnari1, Alessandro Criscimanna1, Chiara Secco1, Magda Marcatti1, Consuelo Corti1, Massimo Bernardi1, Jacopo Peccatori1, Fabio Ciceri1,2, Maria Teresa Lupo-Stanghellini 1

1IRCCS Ospedale San Raffaele, Milan, Italy, 2Università Vita-Salute San Raffaele, Milan, Italy

Background: Acute myeloid leukemia (AML) is the most frequent indication for allogeneic hematopoietic stem cell transplant (allo-HSCT). The number of patients undergoing allo-HSCT for AML has steadily increased in the last decades, with improving outcomes. Both the incidence of disease relapse and treatment related mortality (TRM) decrease over time after transplant. While risk factors for disease relapse and TRM in the first years after allo-HSCT are well known, their correlation with long-term survivorship is unclear.

Methods: We retrospectively collected data of 456 consecutive patients (193 females and 263 males) with a diagnosis of AML who received a first allogeneic hematopoietic stem cell transplant in our centre between 2004 and 2019. Ninety-six patients had a matched related donor, 221 a mismatched related donor, 120 an unrelated donor and 19 underwent allo-HSCT from a cord blood unit. Sixty-four (14%) patients had secondary AML. Patients were included regardless of conditioning intensity and graft-versus-host disease (GvHD) prophylaxis. Two hundred and four patients (44%) received allo-HSCT in first complete remission (CR1), 62 patients (14%) in CR>1, and 190 patients (42%) in active disease (AD). Factors impacting on long-term survival after allo-HSCT were evaluated through a multivariate analysis using Cox proportional-hazards model in the whole cohort and subsequently in landmark analysis at 1, 2 and 3 years after allo-HSCT.

Results: Two-hundred and thirty-nine patients (52%) were alive 2 years after allo-HSCT, with a median follow-up of 5.1 years. In this subset, 51 patients died (21%) during subsequent follow-up: 31 of progressive disease (13%), 18 of treatment related complications (8%) and two due to unknown causes (1%), while 15 patients were lost at follow-up (6%). As expected, a better 5-years overall survival (OS) was observed in the overall cohort among patients in CR1 (63%) in comparison to patients in CR>1 (38%) or AD (25%). Nevertheless, in the landmark analysis the impact of disease status on long-term survival was progressively less meaningful: for example, for patients alive at 2 years landmark, the probability of being alive for 3 more years was 86% for patients in CR1, 72% for patients in CR>1 and 79% for patients in AD, with no statistically significant difference between CR>1 and AD. Similar results were found for progression free survival (PFS), TRM and relapse risk. Of note, better long-term outcomes in the overall cohort were associated with younger patient age, allo-HSCT performed after 2013 and using a female donor for a male recipient. Only being younger was still significantly associated with better outcomes in the various landmark analysis. Furthermore, our study confirmed that relapse is still the main cause of death even for long-term survivors: 13% of patients alive at 2 years eventually relapsed.

Conclusions: In conclusion, the negative impact on outcomes of a more advanced disease at allo-HSCT seems to decline progressively after transplant, fostering the importance of strategies that may enhance the anti-leukemic effect of allo-HSCT in the following first months and years.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P029 TLS::ERG FUSION GENE PREDICTS A POOR PROGNOSIS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN ACUTE MYELOID LEUKEMIA: A SINGLE CENTER STUDY

Gele Tong 1, Yanli Zhao1, Jianping Zhang1, Min Xiong1, Xingyu Cao1, Deyan Liu1, Ruijuan Sun1, Zhijie Wei1, Jiarui Zhou1, Yue Lu1

1Hebei Yanda Lu Daopei Hospital, Langfang, China

Background: Acute myeloid leukemia (AML) patients with the TLS::ERG fusion gene tend to show poor outcomes to traditional chemotherapy. Allogeneic hematologic stem cell transplantation (allo-HSCT) may serve as a potential curative treatment, aiming to enhance the overall survival (OS). However, the long-term safety and efficacy of this treatment for these patients require further exploration.

Methods: A total of 30 AML patients with the TLS::ERG fusion gene were consecutively analyzed, who underwent allo-HSCT in our center between August 2012 and December 2022.

Results: The median age for these patients was 13(2-53) years, with 18 patients 18 years or younger. Before HSCT, the median disease course was 6(3-60) months. Of these patients, 17 (56%) had complex karyotypes, 10(30%) exhibited extramedullary disease, and 10(30%) had additional gene mutations. Prior to HSCT, the disease status was as follows: 18 patients in the first complete remission(CR1),6 in ≥CR2, and 6 in non-remission (NR). In 24 of morphologic CR patients, regular monitoring of TLS::ERG transcript levels before HSCT revealed that 10 patients were TLS::ERG negative, and 14 were TLS::ERG positive. Twenty-seven patients received intensified myeloablative conditioning regimens and the remaining 3 received standard regimens. The transplant donors were haploidentical (n = 23), matched unrelated (n = 5), and matched related (n = 2). In total, eight patients received celluar immutherapy to prophylaxis relapse after transplantation, of them 4 patients received received modified donor lymphocyte infusion (DLI) and another four received IFN-α.eight patients received TLS::ERG positive guided preemptive therapy after transplantation, of them 6 patients received received chemotherapy plus DLI and another two received IFN-α. The median follow-up time for the survivors was 42(12-86)months. The 5-year cumulative incidence of relapse (CIR), and 5-year leukemia-free survival (LFS) and overall survival(OS) after HSCT were 81.6%(95%CI:73.3-89.9),13.2%(95%CI:6.5-19.9), and 13.4%(95%CI:6.4-20.4%), respectively. Relapse occurred in 22 patients with a median relapsed time post-HSCT of 6(3-18) months, among whom 11 simultaneously merged with the extramedullary disease (EMD), among them, twenty patients died from relapse, one patient received therapy and achieved CR, the other one was in relapse status but still alive. Two patients died from treatment-related mortality(TRM). According to disease status (CR/TLS::ERG negative vs. CR/ TLS-ERG positive vs. NR) before allo-HSCT, no significant differences were observed in the 5-year CIR [83.1%(69.8-97.4%) vs. 71.4%(59.3-83.5) vs. 100%), p = 0.602], LFS[15.0%(2.2-27.8%) vs. 21.4%(10.4-32.4) vs 0%), p = 0.548], and OS[18.0%(2.9-33.1%) vs. 16.3%(5.8-26.8%) vs. 16.7%(1.5-31.9%), p = 0.568. Univariate analysis also showed that other factors did not affect the HSCT outcomes.

TABLE1 patient characteristics

Parameters(at diagnosis) n = 30

Median age, y(range) 13(2-53)

≤18y 18(60%)

Cytogenetics:

Complex karyotype 7(56%)

With other gene mutation 10(30%)

Disease status when receiving HSCT

CR1 18(60%)

≥2st CR 6(20%)

NR 6(20%)

With extramedullary disease 10(30%)

HCT-CI

0 27(90%)

1 2(6%)

2 1(3%)

Median disease course(months) before HSCT 6(3-60)

Donor type

HID 23(76%)

MUD 5(16%)

MSD 2(6%)

Conditioning regimen (Intensified MAC) 27(90%)

Conclusions: The presence of TLS::ERG fusion gene in AML is strongly associated with complex karyotype and EMD. Although allo-HSCT is a promising treatment option, the prognosis remains poor due to a high early relapse rate, even in patients with negative TLS::ERG transcript levels before HSCT.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P030 HIGH TOLERABILITY OF PROLONGED AZACITIDINE MAINTENANCE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH AML LACKING A TARGETABLE MUTATION

Cuong An Do 1, Anne-Claire Mamez1, Amandine Pradier1,2, Sarah Morin1, Chiara Bernardi1,2, Sarah Prati-Perdikis1, Yan Beauverd1, Thomas Longval1, Maria Mappoura1, Sarah Noetzlin1, Laetitia Dubouchet1, Evgenia Laspa1, Marie Maulini1, Thien-An Tran1, Carmen de Ramon Ortiz1, Stavroula Masouridi-Levrat1, Federico Simonetta1,2, Yves Chalandon1,2, Federica Giannotti1

1Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland, 2Translational Research Center for Oncohematology, University of Geneva, Geneva, Switzerland

Background: Maintenance therapy after allogeneic hematopoietic stem cell transplantation (HSCT) aims to reduce relapse. Targeted maintenance is available for only about 25% of acute myeloid leukemia (AML). Studies employing hypomethylating agents for maintenance in patients without targetable mutations have shown conflicting results.

Methods: We retrospectively analyzed outcomes and tolerability of azacitidine as post-transplant maintenance in a cohort of 23 adult patients with FLT3neg AML, transplanted at our institution between March 2018 and September 2021. Azacitidine was started at hematological reconstitution from day 60 post-transplant, in patients without active GVHD, evidence of relapse or MRD + . It was administrated subcutaneously at 32.5-50 mg/m2 for 5 days every 4 weeks for 2 years, if tolerated. Till May 2020, patients eligible for azacitidine maintenance were FLT3neg AML patients with MRD+ or active disease at transplant. Thereafter, we proposed azacytidine maintenance for all FLT3neg AML. Overall, 48 patients were screened: 25 did not start azacitidine due to MRD positivity (n = 7), relapse (n = 2), prolonged cytopenia (n = 7), GVHD (n = 5), death (n = 1), insurance refusal (n = 2) and physician choice (n = 1). Tolerability was evaluated by total number of cycles administrated, hematological toxicity, infections or other adverse events leading to withdrawal or delay of maintenance schedule.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (3)

Results: Median follow-up was 30 months (range 11-62). Among 23 patients, one had active disease and 10 were MRD+ before transplant. Median age at transplant was 57 years (range 39-74). Median time from transplant to azacitidine initiation was 102 days (range 66-244). The median number of cycles was 11 (range 1-25). Overall, 7 patients completed the 2-years schedule and 1 is still ongoing. Among these 8, one extended administration intervals to 6 weeks due to cytopenias occasionally requiring G-SCF, another developed recurrent grade 3 neutropenia at higher doses of azacitidine (50 mg/m2), rapidly resolved with G-CSF. One patient on ruxolitinib had grade 3 neutropenia requiring G-CSF once. None of these 3 patients developed infections related to hematological toxicity. Three patients stopped maintenance for mild adverse events (fatigue, myalgias and pruritis) after 11, 12 and 21 cycles, respectively. Among the remaining 12 patients, 3 permanently stopped maintenance due to GVHD (2 chronic moderate, 1 late acute grade 2) after a median of 2 cycles; 9 switched to pre-emptive or curative treatment due to MRD+ occurrence (n = 8) or relapse (n = 1), after a median of 3 cycles. Overall, 5 patients had GVHD or infections not related to azacitidine leading to temporary interruptions (>28 days) for a median time of 73 days (range 49-168). The probability of 2-years OS and PFS were 82.6% (95%CI 68.5%-99.6%) and 69.6% (95%CI 53.1%-91.2%), respectively. Overall, 7 patients died, 1 of lung cancer and 6 of relapse. The 2-years CI of relapse and NRM were 30%±9.85% and 0%±0% respectively.

Conclusions: In our cohort of AML patients lacking targetable mutations, prolonged post-transplant maintenance with azacitidine was relatively well tolerated and outcomes were encouraging. These data support azacitidine maintenance in AML when targeted therapy is not an option.

Disclosure: Y.C. has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution.

F.S. has received institutional consulting fees from BMS/Celgene, Incyte, Kite/Gilead; Speaker fees from Kite/Gilead, Incyte; Travel support from Kite/Gilead, Novartis, AstraZeneca, Neovii, Janssen; Research funding from Kite/Gilead, Novartis, BMS/Celgene.

19: Acute Leukaemia

P031 OUTCOME OF ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN ACUTE MYELOID LEUKAEMIA FLT3 IN THE ERA OF FLT3 INHIBITORS – STUDY OF THE POLISH ADULT LEUKAEMIA GROUP (PALG)

Elzbieta Patkowska1, Anna Czyz2, Alicja Sadowska-Klasa3, Andrzej Szczepaniak4, Lukasz Bolkun5, Marta Sobas2, Michal Gorka6, Edyta Subocz7, Anna Koclega8, Renata Guzicka-Kazimierczak9, Jan Zaucha3, Lidia Gil4, Marta Libura6, Agnieszka Pluta10, Dorota Bartosinska11, Ewa Lech-Maranda1, Agnieszka Wierzbowska10, Sebastian Giebel12, Barbara Nasilowska-Adamska 1

1Institute of Hematology and Transfusion Medicine, Warsaw, Poland, 2Wroclaw Medical University, Wroclaw, Poland, 3Gdansk Medical University, Gdansk, Poland, 4Poznan University of Medical Sciences, Poznan, Poland, 5Medical University of Bialystok, Bialystok, Poland, 6Warsaw Medical University, Warsaw, Poland, 7Independent Public Health Care Ministry of the Interior of Warmia and Mazury Oncology Center, Olsztyn, Poland, 8Medical University of Silesia, Katowice, Poland, 9Pomeranian Medical University, Szczecin, Poland, 10Medical University of Lodz, Lodz, Poland, 11Centre of Postgraduate Medical Education, Warsaw, Poland, 12National Institute of Oncology, Gliwice, Poland

Background: Allogeneic hematopoietic cell transplantation (allo-HCT) offers a potential survival benefit for patients with FLT3-positive acute myeloid leukemia (AML), particularly those harboring the FLT3-internal tandem duplication mutation (FLT3-ITDmut), which is associated with a poorer prognosis compared to point mutations in the tyrosine kinase domain (FLT3-TKDmut). Little is known about the impact of FLT3-TKDmut in the context of allo-HCT, especially with the use of FLT3 inhibitors. This study aims to analyze the outcomes of allo-HCT in FLT3-positive AML patients, considering the type of FLT3 mutation and post-transplant maintenance treatment.

Methods: This retrospective analysis includes FLT3-positive AML patients, encompassing FLT3-ITDmut and/or FLT3-TKDmut, who received intensive chemotherapy combined with midostaurin and underwent allo-HCT between 2017 and 2023.

Results: The study involved 88 patients, with a median age of 50.3 years (IQR 37.1-62.2), predominantly female (n = 58; 65.91%). FLT3-ITDmut was observed in 67 (76.1%) patients, FLT3-TKDmut in 15 (17%), and both mutations in 6 (6.8%) patients. All patients received intensive chemotherapy with midostaurin, initiated either after the first or second induction in 70 (80%) patients or after consolidation in 18 (20%) patients. The median follow-up period post allo-HCT was 15.7 months (IQR: 8.8-28.7). Allo-HCT was performed in the first complete remission (CR) in 68 (77%) patients, subsequent CR in 12 (13.63%) patients, or active disease in 8 (9%) patients. Most patients received myeloablative conditioning (MAC), including TBI-based MAC (n = 5; 5.6%), with grafts from matched related donors (MRD) (n = 24; 27%), matched unrelated donors (MUD) (n = 45; 51%), mismatched unrelated donors (mMUD) (n = 6; 6.8%), or family haploidentical donors (n = 13; 15%). The median overall survival (OS) post allo-HCT was 11 months, with a 2-year OS probability rate of 62%. Subgroup analysis of 60 patients who initiated midostaurin after the first or second induction and were tested for both FLT3 mutations showed a 2-year OS advantage for AML FLT3-TKDmut and AML FLT3-TKDmut co-occurring with FLT3-ITDmut patients compared to AML FLT3-ITDmut patients (rates: 81% vs 80% vs 67%, respectively), although this difference was not statistically significant (p = 0.289). Sorafenib was administered as maintenance treatment post allo-HCT to 22 (30%) of 73 AML FLT3-ITDmut patients. Univariate analysis of OS for the entire group identified age as the only significant prognostic factor, with better survival in patients under 60 years of age (HR = 0.44; 95% CI 0.2-0.93). In the FLT3-ITDmut subgroup, sorafenib maintenance (HR = 0.08; 95% CI 0.01-0.58), age under 60 years (HR 0.36; 95% CI 0.16-0.82) and male gender (HR 0.33; 95% CI 0.11-0.97) were prognostic factors associated with improved OS in univariate analysis. However, in the multivariate analysis of this subgroup, only sorafenib maintenance remained a favourable prognostic factor for improved OS (HR 0.11; 95% CI 0.01-0.89).

Conclusions: Allo-HCT emerges as an effective curative treatment option, providing satisfactory survival outcomes for both AML FLT3-ITDmut and FLT3-TKDmut patients. In the entire patient group, age stands out as the only significant prognostic factor for OS. However, among FLT3-ITDmut patients, post allo-HCT maintenance treatment with sorafenib remains the sole favorable prognostic factor for OS.

Disclosure: E. Patkowska: KCR- consultancy, Astellas Pharma, Servier, Amgen, Novartis - Honoraria.

19: Acute Leukaemia

P032 CLINICAL CHARACTERISTICS AND PROGNOSIS OF ADULT T CELL ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS WITH ACTIVATING RAS MUTATIONS

Mimi Xu 1,2,3, Yuqing Tu1,2,3, Mengqi Xiang1,2,3, Yi Fan1,2,3, Xiang Zhang1,2,3, Tiemei Song1,2,3, Lian Bai4, Xiaoli Li5, Yang Xu1,2,3, Yuejun Liu1,2,3, Depei Wu1,2,3

1National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China, 2Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China, 3Key Laboratory of Stem Cells and Biomedical, Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China, 4Canglang of Suzhou Hospital, Suzhou, China, 5Soochow Hopes Hematonosis Hospital, Suzhou, China

Background: Several studies indicated that RAS mutations were associated with a poorer outcome in T cell acute lymphoblastic leukemia (T-ALL) patients. However, data regarding the clinical outcomes of adult RAS mutated T-ALL patients undergoing different consolidation treatment are limited. This study was conducted to analyze the clinical characteristics and prognosis of adult T-ALL patients with RAS mutations.

Methods: From January 2016 to December 2022, 137 newly diagnosed adult T-ALL patients were explored for activating RAS mutations in our center. We retrospectively analyzed the clinical characteristics of adult RAS mutated T-ALL patients and compare the efficacy of allogeneic hematopoietic stem cell transplantation (allo-HSCT) with chemotherapy in these patients. About statistical analysis, overall survival (OS) and event free survival (EFS) were estimated by the Kaplan-Meier method and the log-rank test. The cumulative incidence of relapse (CIR) was estimated using Gray’s test, with non-relapse morality considered a competing risk. The covariates with a P value < 0.2 in univariate analyses were used in the multivariate analyses (Fine-Gray model). A P value < 0.05 was considered statistically significant. Statistical analyses were carried out with SPSS 20.0 and R 4.3.0.

Results: KRAS and NRAS mutations were identified in four (2.9%) of 137 and 15 (11%) of 137 patients, respectively. Overall, 19 (13.9%) of 137 T-ALLs harbored activating RAS mutations, without any accompanying PTEN gene changes. The early precursor T (ETP) was the most common immunophenotype (68.4%), with chromosomal abnormalities accounting for 36.8%, and 3 cases of SET-NUP214 fusion gene positivity. The complete response (CR) rate in one course of treatment for 19 patients was 78.9%, and the total CR rate in two courses was 93.3%, one patient with continuous non-remission died on 5.7 months after diagnosis. The median follow-up of survivals was 26.2 months. The 3-year OS rate was (60±12)%, EFS rate was (41±12)%, and CIR was (61.6±1.9)%. Six patients who only received chemotherapy died within 2 years after diagnosis; The 3-year OS rate and EFS rate of 13 patients who underwent allo-HSCT as consolidation were (90.0±9.5)% and (62.3±15.2)%, respectively. Multivariate analyses revealed that achieved CR after first induction chemotherapy (P = 0.036, HR = 0.075, 95%CI: 0.007-0.849) and allo-HSCT (P = 0.008, HR = 0.031, 95%CI: 0.002-0.398) were independent good factors affecting CIR.

Table 1 Characteristics of 19 adult T-ALL patients with activating RAS mutations.

Total

(n = 19)

Allo-HSCT

(n = 13)

Chemothreapy

(n = 6)

P value

Gender, male/female, n

13/6

10/3

3/3

0.241

Age at diagnosis [years, M (range)]

44(21-67)

38(21-55)

60(44-67)

0.001

WBC at diagnosis [×109/L, M (range)]

5.18(0.62-204.7)

4.82(0.62-204.7)

8.81(1.55-95.09)

0.831

Blasts in bone marrow [%, M (range)]

72.5(24-97.5)

84(47.5-97.5)

54.5(24-89)

0.244

Immunophenotype, n(%)

0.018

Pro-T

3(15.8)

0(0.0)

3(50.0)

Pre-T

3(15.8)

2(15.4)

1(16.7)

ETP

13(68.4)

11(84.6)

2(33.3)

Chromosomal, n(%)

0.829

Normal

12(63.2)

8(61.5)

4(66.7)

Abnormal

7(36.8)

5(38.5)

2(33.3)

Fusion gene, n(%)

0.200

SET-NUP214

3(15.8)

3(23.1)

0(0.0)

Negative

16(84.2)

10(76.9)

6(100.0)

RAS mutations, n(%)

0.126

NRAS mutation

15(78.9)

9(69.2)

6(100.0)

KRAS mutation

4(21.1)

4(30.8)

0(0.0)

Accompany by NOTCH1/FBXW7 mutations, n(%)

12(63.2)

7(53.8)

5(83.3)

0.216

Conclusions: We concluded that RAS mutations were common in adult T-ALL without accompanying PTEN changes, and had more frequently an immature immunophenotype. Adult RAS mutated T-ALL patients had a high induction response rate, but were prone to relapse. Allo-HSCT might effectively reduce the recurrence rate and improve their survival.

Disclosure: Nothing to declare

19: Acute Leukaemia

P033 PREVENTION AND TREATMENT OF ACUTE MYELOID LEUKEMIA RELAPSE FOLLOWING ALLOGENEIC STEM CELL TRANSPLANT: A NATIONAL SURVEY BY GITMO (GRUPPO ITALIANO TRAPIANTO MIDOLLO OSSEO)

Francesco Saraceni1, Eliana Degrandi2, Simona Piemontese3, Francesco Saglio4, Giorgia Battipaglia 5, Marta Lisa Battista6, Patrizia Chiusolo7, Jacopo Mariotti8, Stefania Bramanti8, Anna Grassi9, Fabio Benedetti10, Manuela Tumino11, Giorgia Saporiti12, Salvatore Leotta13, Sadia Falcioni14, Martina Chiarucci15, Gladis Bortoletto16, Elisabetta Terruzzi17, Annalisa Imovilli18, Carlo Borghero19, Eugenia Piras20, Francesco Zallio21, Vincenzo Federico22, Irene Maria Cavattoni23, Andrea Gilioli24, Alessandra Picardi25, Domenico Pastore26, Anna Paola Iori27, Valentina Giudice28, Carmen Di Grazia29, Luca Castagna30, Michele Malagola31, Attilio Olivieri1, Francesca Bonifazi32, Massimo Martino33

1Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy, 2GITMO Trial Office, Bologna, Italy, 3Ospedale S. Raffaele, Milano, Italy, 4CIttà della Salute e della Scienza, Torino, Italy, 5Università degli studi di Napoli Federico II, Napoli, Italy, 6Azienda sanitaria universitaria Friuli Centrale, Udine, Italy, 7Università Cattolica del Sacro Cuore, Roma, Italy, 8Istituto Humanitas, Rozzano - Milano, Milano, Italy, 9ASST Papa Giovanni XXIII, Bergamo, Italy, 10Ospedale Borgo Roma di Verona, Verona, Italy, 11Azienda Ospedaliera di Padova, Padova, Italy, 12Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico Milano, Milano, Italy, 13Azienda Ospedaliero - Universitaria Policlinico - Vittorio Emanuele, Catania, Italy, 14AOC Ascoli Piceno, Ascoli Piceno, Italy, 15Azienda Ospedaliera Marche Nord, Pesaro, Italy, 16IRCCS Istituto Oncologico Veneto, Mestre, Italy, 17Azienda Ospedaliera San Gerardo, Monza, Italy, 18AUSL Reggio Emilia, Reggio Emilia, Italy, 19Ospedale S. Bortolo Vicenza, Vicenza, Italy, 20Centro Trapianti Midollo Osseo, Università di Cagliari, Cagliari, Italy, 21Azienda Ospedaliera di Alessandria, Alessandria, Italy, 22ASL Lecce, Lecce, Italy, 23Ospedale Generale Regionale Bolzano, Bolzano, Italy, 24Azienda Ospedaliero Universitaria di Modena, Modena, Italy, 25Ospedale A. Cardarelli - Divisione di Ematologia, Napoli, Italy, 26ASL Brindisi, Brindisi, Italy, 27Università la Sapienza, Roma, Italy, 28Università degli studi di Salerno, Salerno, Italy, 29UO Ematologia e Terapie Cellulari. IRCCS Ospedale Policlinico San Martino, Genova, Italy, 30Azienda Ospedaliera Ospedali Riuniti Villa Sofia - Cervello, Palermo, Italy, 31Università degli Studi di Brescia, Brescia, Italy, 32IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, 33Azienda Ospedaliera “Bianchi-Melacrino-Morelli” di Reggio Calabria, Reggio Calabria, Italy

Background: Leukemia relapse remains the main cause of treatment failure in patients undergoing allogeneic stem cell transplant (allo-SCT) for acute myeloid leukemia (AML). There is currently no general agreement concerning management of patients at high risk of leukemia recurrence or with overt hematological relapse after allo-SCT.

Methods: We conducted the present survey with the aim to evaluate the current practice in Italy concerning prevention and treatment of AML relapse after allo-SCT. A questionnaire was sent to GITMO centers including specific questions on three main topic: maintenance, preemptive therapy and treatment of hematological relapse. Most of the agents discussed below currently represent off label prescriptions.

Results: In all, 34/60 (57%) of centers performing allo-SCT in Italy completed the questionnaire. 22/34 (65%) routinely employ maintenance strategies (in the absence of measurable residual disease (MRD) or mixed chimerism) after allo-SCT in patients considered at high risk of relapse basing on pre-transplant features (high risk according to ELN 2022 or presence of FLT3-ITD, positive MRD or active disease). Among institutions who deliver maintenance therapy after allo-SCT, in patients harbouring FLT3-ITD 86% administer FLT3 inhibitors (95% Sorafenib, 5% Gilteritinib). In the absence of FLT3-ITD 68% use hypometilating agents (87% azacitidine, 13% decitabine); 2 centers consider addition of venetoclax to hypometilating agents, while one institution prescribes venetoclax as single agent. Interestingly, 26% of institutions administer prophylactic DLI in high risk AML patients, in the absence of MRD relapse or mixed chimerism. With respect to preemptive therapy, the trigger to start treatment is represented by MRD relapse in all centers; 74% of institutions consider mixed chimerism as trigger to start preemptive therapy as well. In patient harbouring FLT3-ITD 82% of centers prescribe FLT3 inhibitors (48% sorafenib +/- azacitidine, 52% gilteritinib). In the absence of FLT3-ITD 62% of institutions use azacitidine (19% with the addition of venetoclax), one institution use venetoclax monotherapy, while 29% use DLI as single approach especially in case of mixed chimerism in the absence of MRD relapse. Nevertheless, 32/34 (94%) use DLI in addition to pharmacologic preemptive therapy.

In patients experiencing haematological relapse after transplant 76% of institutions perform NGS analysis at the time of relapse, while 50% look for HLA loss in case of relapse after haploidentical transplant. In patients harbouring FLT3-ITD 91% of centers treat AML relapse with gilteritinib. In fit patients, in the absence of FLT3-ITD, 44% employ conventional chemotherapy, while 56% consider as first option hypometilating agents (mostly azacitidine) in combination with venetoclax.

All institutions consider second allo-SCT after salvage treatment in patients who relapse after first transplant. 62% of centers perform second allo-SCT only in patients achieving CR2, while 38% regardless achievement of response to salvage. Finally, 97% of institutions prefer a different donor for second allo-SCT, if available.

Conclusions: Most Italian transplant institutions currently implement strategies to prevent AML relapse after allo-SCT, despite the lack of approved agents in this setting. In patients with haematological relapse a second allo-SCT remains a main option after salvage treatment with standard chemotherapy or hypometilating agents in combination with venetoclax.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P034 TRENDS IN THE USE OF HEMATOPOIETIC CELL TRANSPLANTATION FOR ADULTS WITH ACUTE LYMPHOBLASTIC LEUKEMIA: A REPORT FROM THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT

Sebastian Giebel 1, Myriam Labopin2, Frederic Baron3, Ali Bazarbachi4, Eolia Brissot5, Gesine Bug6, Jordi Esteve7, Norbert-Claude Gorin8, Francesco Lanza9, Arnon Nagler10, Annalisa Ruggeri11, Jaime Sanz12, Bipin Savani13, Christoph Schmid14, Roni Shouval15, Alexandros Spyridonidis16, Jurjen Versluis17, Zinaida Peric18, Mohamad Mohty5, Fabio Ciceri19

1Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland, 2EBMT Unit, Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), AP-HP, Saint Antoine Hospital, Paris, France, 3GIGA-I3, University and CHU of Liège, Liège, Belgium, 4American University of Beirut Medical Center, Beirut, Lebanon, 5Sorbonne Universités, UPMC University of Paris 06, INSERM, Centre de Recherche Saint-Antoine (CRSA), AP-HP, Saint Antoine Hospital, Paris, France, 6University Hospital, Goethe University Frankfurt, Frankfurt am Main, Germany, 7Hospital Clínic of Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain, 8European Society for Blood and Marrow Transplantation, Paris Office, Hopital Saint-Antoine, Paris, France, 9Romagna Transplant Network, Ravenna, Italy, 10Chaim Sheba Medical Center, Tel-Hashomer, Sackler School of Medicine, Tel Aviv University, Tel Aviv, Israel, 11IRCCS Bambino Gesù Children’s Hospital, Rome, Italy, 12Hospital Universitari i Politecnic La Fe. Instituto de Investigación Sanitaria La Fe, Valencia, Spain, 13Vanderbilt University Medical Center, Nashville, United States, 14Augsburg University Hospital, Augsburg, Germany, 15Memorial Sloan Kettering Cancer Center, New York, United States, 16University of Patras, Patras, Greece, 17Erasmus University Medical Center Cancer Institute, Rotterdam, Netherlands, 18University Hospital Center Rijeka and School of Medicine, University of Rijeka, Rijeka, Croatia, 19Ospedale San Raffaele s.r.l., Haematology and BMT, Milano, Italy

Background: Indications for hematopoietic cell transplantation (HCT) for adults with acute lymphoblastic leukemia (ALL) evolve over time and vary among countries. In our previous survey we reported continues increase of the number of allogeneic (allo)-HCT procedures between years 2001-2015 [Giebel S, et al., Ann Hematol 2019]. In recent years the role of HCT has been challenged by new protocols incorporating bispecific antibodies or immunotoxins in first-line treatment. The goal of this study was to assess general trends in the number of various types of HCTs performed between years 2010 and 2021 in Europe.

Methods: Data reported to the EBMT registry were used for this analysis. Altogether, 14389 first allogeneic (n = 13795) or autologous (auto-, n = 594) HCTs were performed in the period 2010-2021.

Results: The number of allo-HCTs analyzed in 2-year periods increased from 2074 for years 2010-2011 to 2428 between 2018-2019 (17% increase) and then dropped to 2282 for years 2020-2021 (6% decrease). Similar pattern was observed for the largest European countries (Germany, UK, France, Italy, Spain).

Comparing years 2020-2021 and 2010-2011, median recipient age increased from 37.3 to 41.1 (p<0.0001), the proportion of matched sibling donor transplants decreased from 39% to 24.2%, while the rate of unrelated donor and haploidentical donor transplants increased from 50.8% to 54.3% and 5.1% to 20.2%, respectively; the proportion of cord blood transplants decreased from 5.1% to 1.2% (p<0.0001) (Table). In respective periods the proportion of patients with Ph-negative B-ALL increased from 28.6% to 43.2%, for Ph-positive B-ALL it decreased from 38% to 32.7% while for T-ALL it dropped from 33.4% to 24.1% (p<0.0001). Most patients were treated in the first complete remission: 70.2% between years 2010-2011 and 70.9% between 2020-2021. The proportion of patients administered allo-HCT in active disease decreased from 10.5% to 4.7% in respective periods (p<0.0001). Peripheral blood was used as a source of stem cells for allo-HCT in 74.1% of patients between 2010-2011 and 90.3% of patients between 2020-2021. The use of post-transplant cyclophosphamide increased from 1.7% to 29.6% in respective periods (p<0.0001).

The total number of auto-HCTs was 130 between years 2010-2011 and 48 between 2020-2021 (63% decrease).

Year

2010-2011

2012-2013

2014-2015

2016-2017

2018-2019

2020-2021

Total allo-HCT

2074

2319

2309

2383

2428

2282

MSD-HCT

809

800

721

780

665

553

URD-HCT

1054

1242

1250

1269

1285

1240

Haplo-HCT

105

190

278

293

434

462

Cord blood

106

87

60

41

44

27

Auto-HCT

130

98

127

112

79

48

Conclusions: Results of our analysis indicate moderate increase of allo-HCT procedures for adults with ALL in the period 2010-2019 followed by a slight decrease between 2020-2021 that might be related to SARS-CoV-2 pandemic. The profile of allo-HCTs has changed over time with more frequent use of haploidentical donors, post-transplant cyclophosphamide – based immunosuppression and the use of peripheral blood as predominant source of stem cells. In recent years less patients have been treated with allo-HCT in active disease which may reflect higher efficacy of immunotherapy-based salvage regimens. Auto-HCT is rarely used for the treatment of adults with ALL.

Clinical Trial Registry: Not applicable

Disclosure: GB has received honoraria from Novartis, Jazz, BMS and Gilead and travel grants from Jazz, Gilead and Neovii

19: Acute Leukaemia

P035 ALLOGENEIC STEM CELL TRANSPLANTATION FOR ADULT ACUTE LYMPHOBLASTIC LEUKEMIA - A SINGLE CENTER 25-YEARS EXPERIENCE

Veronika Válková 1,2, Cyril Šálek1,2, Antonín Vítek1, Markéta Marková1,2, Ludmila Nováková1, Mariana Koubová1, Michal Kolář1, Barbora Čemusová1, Hana Bartáková1, Petr Cetkovský1,2, Jan Vydra1,2

1Institute of Hematology and Blood Transfusion, Prague 2, Czech Republic, 2Faculty of Medicine of Charles University, Prague 2, Czech Republic

Background: Allogeneic stem cell transplantation (allo-SCT) may be curative for patients with high risk acute lymphoblastic leukemia (ALL). The aim of our analysis was to compare the results of allo-SCT in ALL over more than two decades and to identify the parameters that influence these results.

Methods: All patients (pts) older than 18 years who underwent allo-SCT at our center from March 1989 to July 2023 were included in the analysis. In total, there were 183 pts, median age 38 years (18-65, only 17% over 55 years), 116 (63%) were male, B-ALL-Ph pos 37%, T-ALL 17%, donors were MSD (26%), MUD (42%), MMUD (23%) and haplo (8%). The majority of grafts were PBPC (82%). Myeloablative conditioning (TBI based) received 158 (86%) pts, 62% of whom received Vep/12Gy TBI. A more detailed analysis in terms of GVHD and MRD was performed on a cohort of patients from 2007 (n = 136). At allo-SCT 105 pts (77%) were in CR1, 23 (17%) in CR2, 6% with active disease. In terms of MRD, 60 patients were negative and 59 positive before allo-SCT.

Results: With a median follow-up of 6.5 years (living pts), 5-year OS was 56% and EFS of 49%. The cumulative incidence of relapse and NRM was 30% and 27% respectively at 5 years. Acute GVHD grade 2-4 occurred in 23%, moderate/severe chronic GVHD in 11%. GRFS at 5 years was 42%. Relapse occurred in 23% of patients, at a median of 8 months. OS after relapse was 22% at 5 years (26% vs 19% in the last decade vs earlier, p= ns). In univariate analysis, significantly better outcomes in terms of OS were observed in the last decade (5y-OS 68%), optimal conditioning seems to be Vep/TBI, worse outcomes were observed in patients over 55 years. Survival was significantly better in pts reaching CR before allo-SCT, and also significantly better in MRD neg versus MRD pos pts (no difference between MRD neg CR1 or CR2), both before allo-SCT and at 3 months after allo-SCT. A multivariate Cox regression analysis confirmed the survival benefit for pts who reached MRD negativity before allo-SCT (p = 0.047) and for transplants from MSD (p = 0.02) or MUD (p = 0.02) over the other donors.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (4)

Conclusions: The outcomes of allo-SCT in ALL have improved significantly in the last decade, in our setting mainly due to the reduction of NRM. The role of novel therapies has been observed more in terms of of the possibility of achieving more CR (and even more MRD neg CR) before allo-SCT, rather than in the treatment of relapses after allo-SCT. Our results confirm the essential role of MRD monitoring during ALL treatment and in particular achieving MRD negativity before allo-SCT.

Disclosure: nothing to declare

19: Acute Leukaemia

P036 STRUCTURAL ANALYSIS OF LEUKEMIA-ASSOCIATED PROTEIN PTPN21 REVEALS A DOMINANT-NEGATIVE EFFECT OF THE FERM DOMAIN ON ITS PHOSPHATASE ACTIVITY

Zijun Qian 1,2, Lu Chen1,2, Jie Zhang2, Yuyuan Zheng1, Chun Zhou1,2, Haowen Xiao1,2

1Zhejiang University, Hangzhou, China, 2Sir Run Run Shaw Hospital, Hangzhou, China

Background: High expression level of protein tyrosine phosphatase non-receptor type 21 (PTPN21) has been detected in B-cell non-Hodgkin’s lymphoma and its mutations are associated with disease relapse in B-cell acute lymphoblastic leukemia (B-ALL) patients who received allo-HSCT. Recently, PTPN21 has been shown to be pivotal in maintaining cell mechanical properties and helps retain hematopoietic stem cells (HSCs) in the bone marrow niche through dephosphorylating the cytoskeleton-associated protein Septin1. However, the molecular structure and functions of PTPN21 have been less understood compared to other tyrosine phosphatases like PTP1 B. PTPN21 is a relatively large protein with two structured domains, FERM domain and the catalytic domain (PTP), and a long intrinsically disordered region in the middle. The mechanisms of regulation of PTPN21 phosphatase activity and the effect of mutations on its functions need to be explored.

Methods: We used protein purification and crystallization to determine structures of FERMPTPN21, PTPPTPN21 and a complex structure of the PTP and FERM domains. GST pulldown and MST titration as well as phosphatase assays and site direct mutagenesis were used to determine the interaction of PTP and FERM domains.

Results: Through biochemical assays, we found that PTPN21 FERM domain can interact with PTP domain, resulting in negatively regulating its phosphatase activity in vitro and in vivo. We further explored the structures of FERMPTPN21, PTPPTPN21 as well as a complex structure of the combination of PTP and FERM domains. The FERM-PTP complex structure allowed us to identify critical interface residues and mutations of these residues led to enhanced phosphatase activity of PTPN21 and induced activation of downstream ERK. More importantly, these mutations only activated ERK in the context of wild-typed PTP but not the catalytic inactive C1108S, indicating PTPN21’s phosphatase activity is indispensable.

Conclusions: We’ve demonstrated that PTPN21 catalytic domain PTP is autoinhibited by the FERM domain. Disruption of FERM-PTP interaction results in enhanced PTPN21 activity and activation of downstream MAPK signal pathway. Our work provides novel insights into understanding the role of PTPN21 in various cellular processes by discovering the molecular structure of the FERM and PTP domains of PTPN21. These findings are likely extendable to other FERM-containing tyrosine phosphatases like PTPN14 and PTPN3, which opens up new avenues for further mechanistic studies.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P037 EARLY DETECTION OF RELAPSE AND PROMPT TREATMENT WITH DLI FACILITATED BY PROPHYLACTIC LYMPHOCYTE STORAGE PERI-TRANSPLANT RESULTS IN EFFECTIVE DISEASE CONTROL IN ACUTE LEUKAEMIA AND MDS

Daire Quinn 1, Anne Parker1, Grant McQuaker1, Anne-Louise Latif1, David Irvine1, Ailsa Holroyd1, Dimitris Galopoulos1, Andrew Clark1

1Queen Elizabeth University Hospital, Glasgow, United Kingdom

Background: Relapse following allogeneic stem cell transplant carries a poor prognosis. DLI has been used in this setting but is less effective if delayed or given in the context of haematological relapse.

Methods: Aliquots of DLI from the PBSC collection were stored prophylactically pre-transplant. The transplant dose was always protected and was never < 4x106 CD34 cells/Kg. No DLI were stored if stem cell dose was lower than this threshold. Patients with relapsed acute leukaemia (AL) or MDS were detected early and treated with an effective salvage regimen followed by escalated doses of DLI, starting at 3x105 CD3 cells/Kg (VUD) or 1x106 CD3 cells/Kg (MRD). First dose DLI was given at 6-8 weeks post commencement of salvage treatment. Subsequent DLI doses (3x106, 1x107, 3x107 CD3 cells/Kg) were delivered at 8-12 week intervals intercalated with salvage therapy. Maximum dose was 3x107 CD3 cells/Kg (max cumulative dose 4.63 x107 CD3 cells/Kg). Patients were excluded if they had rapidly progressive disease, BM blasts >50% or ECOG >2.

Results:

Acute Leukaemia

Myelodysplasia

Specific Disease

AML 15, ALL 4

12

Median Age

51 (21-66)

60 (51-66)

Male sex

11

10

Conditioning

RIC

11

12

MAC

8

Mode of Presentation

MRD 10, Haematological 9

Mixed myeloid chimerism + cytopenia 12

2 year DFS

53%

42%

Median FU

30 months

22 months

Between 2015 and 2022 thirty one patients were treated with DLI for relapsed AL (n = 19) or MDS (n = 12). Strikingly, only 4 of these interventions occurred before 2019. Before that time selecting patients with low disease burden and being able to acquire DLI in a rapid time frame was frequently not possible. After 2019 four factors facilitated effective DLI administration- Early disease relapse, detection using MRD combined with assessment of myeloid chimerism, newer agents to achieve remission responses and the ready availability of DLI.

In relapsed MDS all patients had recurrent cytopenias associated with: NGS abnormalities, increasing blast % or cytogenetic abnormalities. Myeloid chimerism ranged from 4-64% at diagnosis returning to 100% donor in all responding patients. In the AL group 10 were diagnosed by MRD methods and 9 had haematological relapse. Median Time to relapse was 10 months in AL and 19 months in MDS

In AL salvage treatment included FLAG, Dasatinib, Gemtuzumab, Ven/Aza, Gilteritininb, Blinatumomab, Inotuzumab, Azacitidine alone or no therapy while MDS patients all received Azacitidine for 4-12 cycles. Median number of DLI infusions was 3. Median total dose was 1.4x107 CD3 cells/Kg in both groups. In responders the median total dose was 4.4x107 CD3 cells/kg. Median FU post DLI was 20 and 30 months overall or for responders in AL and 19 and 22 months in MDS.

CR was achieved in 63% of acute leukaemia and 58% of MDS patients. Two leukaemic patients relapsed at 16 and 34 months. Responses were otherwise durable, 80% remained in CR at 3 years. Three additional MDS patients responded transiently. Acute GvHD occurred in 2 patients. Two patients have moderate chronic GvHD. One died of acute neurotoxicity. Two year DFS was 53% in AL and 42% in MDS.

Conclusions: Combining early detection of disease relapse, followed by treatment with novel therapies and consolidation with DLI provides effective salvage therapy. High levels of durable disease control are achieved and low rates of GvHD are seen resultant on DLI dose escalation. Prophylactic storage of DLI allows this approach and is cost effective.

Clinical Trial Registry: None

Disclosure: None

19: Acute Leukaemia

P038 THIOTEPA, BUSULFAN, AND FLUDARABINE CONDITIONING REGIMEN IN T-CELL REPLETE HLA-HAPLOIDENTICAL BONE MARROW TRANSPLANTATION FOR ADULTS WITH ACUTE MYELOBLASTIC LEUKEMIA IN SECOND COMPLETE REMISSION

Ahmad Ibrahim 1, Ahmad Khalil2, Kamal Al Zahran3, Mohamad Hachem4, Pamela Sfeir5, Amin Abyad6, Jad Ibrahim7, Hussein Abou Abbas8, Ali Youssef8, Charbel Khalil2, Tamima Jisr9

1Makassed University Hospital, Middle East Institute of Health, University of Balamand and Lebanese University, Beirut, Lebanon, 2Middle East Institute of Health, Beirut, Lebanon, 3Middle East Institute of Health and Lebanese University, Beirut, Lebanon, 4Makassed University Hospital and Lebanese University, Beirut, Lebanon, 5University of Balamand, Beirut, Lebanon, 6Burjeel Medical City, Abu Dhabi, United Arab Emirates, 7American University of Beirut, Beirut, Lebanon, 8Makassed University Hospital, Beirut, Lebanon, 9Makassed University Hospital and Beirut Arab University, Beirut, Lebanon

Background: Haploidentical stem cell transplantation (HaploSCT) with thiotepa/busulfan/fludarabine (TBF) conditioning and post-transplant cyclophosphamide (PT-Cy) can achieve comparable outcomes for relapsed acute myeloblastic leukemia (AML) patients (pts) in complete remission (CR) to those of HLA-matched sibling or unrelated donor transplantation. However, a higher incidence of acute and chronic GVHD was reported with peripheral blood stem cell source (PBSC) compared to bone marrow (BM) in adult acute leukemia. In this study, we report the outcomes of adult pts (>18yo) with AML in second CR who underwent HaploSCT with unmanipulated BM source using TBF conditioning and PT-Cy.

Methods: This study is a retrospective analysis conducted between 2/2018 and 2/2023. The TBF-MAC included thiotepa IV 5mg/kg/d on d-7 and -6, IV busulfan 3.2mg/kg/d on d-4, -3, -2, and IV fludarabine 30mg/kg/d from d-5 to d-1. The TBF-RIC, for pts >60yo, involved reduced doses of thiotepa (5mg/kg on d-6) and busulfan (3.2mg/kg/d on d-3 and -2), with identical fludarabine doses. BM was infused on d0. The PT-Cy dose was 50mg/kg/d d + 3 and +4, followed on d + 5 by IV cyclosporine 3mg/kg/d, then tapered over 6 weeks after d + 60 if no GVHD. MMF was given at a dose of 15mg/kg every 12 hours d + 5 to d + 35. GCSF was started d + 5 until neutrophil recovery. Prophylaxis for VOD was ensured by ursodeoxycholic acid. Anti-microbial prophylaxis was given. Pts were monitored by PCR for CMV, EBV, HHV6 until d + 100, and for BK virus if indicated. Azacitidine was given as maintenance therapy after Haplo SCT for all pts, and sorafenib was given in case of FLT3 mutations.

Results: Thirty-two pts were included. Median age was 36yo (19-71). There were 17 females and 15 males. Twenty-four pts (75%) received TBF-MAC, and 8 (25%) TBF-RIC. Median number of nucleated cells infused was 5.2×108 kg (4.1-6.7). All pts achieved neutrophil recovery within a median of 17d (14-31), and platelet recovery (>20 109/L) within a median of 19d (10-42). All pts had full donor chimerism before d + 90. Seven pts (22%) developed grade II-IV aGVHD, including 2 (6%) with grade III–IV. Three pts (9%) developed non-severe cGVHD. Acute GVHD was refractory to steroids in 2 pts and was fatal in one. Fatal VOD occurred in 1 pt (3%). Eight pts (25%) had bacteremia, and 2 (6%) developed invasive fungal infections. Two pts (6%) developed fatal COVID-19. CMV reactivation, successfully treated with ganciclovir, occurred in 14 pts (44%). Seven pts (22%) died of infection, including 2 of COVID-19 and 5 of sepsis. Five pts (16%) relapsed within a median of 9 months (6-25) after Haplo SCT and died from AML between 1 and 6 months after relapse. Within a median follow-up of 22 months (1-68), 18 pts (56%) are still alive in CR with a median of 33 months (11-68). The 3y-OS and-DFS were 57% and 52%, respectively.

Conclusions: HaploSCT with BM source offers a curative therapy for adult AML pts in 2nd CR with low risk of GVHD, and low incidence of relapse. However, infections remained the main cause of death.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P039 REDUCED INTENSITY CHEMOTHERAPY WITH TYROSINE KINASE INHIBITOR FOLLOWED BY ALLOGENEIC STEM CELL TRANSPLANTATION WAS EFFECTIVE IN PATIENTS WITH PHILADELPHIA CHROMOSOME POSITIVE ACUTE LYMPHOBLASTIC LEUKEMIA

Dong Won Baek 1, Jung Min Lee1, Hyukjin Choi1, Joon Ho Moon1, Sang Kyun Sohn1

1Kyungpook National University Hospital, Daegu, Korea, Republic of

Background: With the introduction of tyrosine kinase inhibitors (TKI), survival outcomes of adult patients with Philadelphia chromosome positive (Ph-positive) acute lymphoblastic leukemia (ALL) has improved remarkably. Intensive chemotherapy plus TKI is still the standard approach to induce hematologic complete remission (CR) before allogeneic stem cell transplantation (allo-SCT). However, intensive chemotherapy before transplantation may have negative impacts on allo-SCT outcomes, including treatment-related mortality, worsening performance at the time of transplantation. In the current study, we compared the survival outcomes of intensive chemotherapy plus TKI and reduced intensity chemotherapy plus TKI in patients with newly diagnosed Ph-positive ALL. In addition, the clinical role of allo-SCT in the treatment of adult Ph-positive ALL was also evaluated.

Methods: We retrospectively analyzed data from 97 patients with newly diagnosed Ph-positive ALL who received intensive chemotherapy plus TKIs or reduced intensive chemotherapy plus TKIs over the past 10 years. Patients aged ≥ 20 years were included in this study. Patients who were in suitable condition for transplantation and had HLA matched donor underwent allo-SCT. Relapse-free survival (RFS) was determined from the date of diagnosis to the date of disease relapse or death from any cause. Overall survival (OS) was determined from the date of diagnosis to the date of death or last follow-up.

Results: In total, 97 patients were analyzed. Intensive chemotherapy was administered to 69 patients, and median age was 45 (range, 20-73) years old. Reduced intensity chemotherapy was administered to 28 patients, and their median age was 68.5 (range, 27-80) years old. In patients with intensive chemotherapy plus TKI, 55 patients (79.7%) achieved molecular CR after induction chemotherapy, and four patients (5.8%) died during induction therapy. In patients with reduced intensity chemotherapy plus TKI, 20 patients (71.4%) achieved molecular CR, while no one has died during induction therapy. In the intensive chemotherapy group, 46 patients (66.7%) underwent allo-SCT, and 20 patients showed disease relapse and three patients died after transplantation. In the reduced intensity chemotherapy group, 12 patients (42.9%) received allo-SCT, and four patients had relapsed disease and one died. In an analysis targeting only transplanted patients, there was no statistically significant difference in RFS and OS between patients who received intensive chemotherapy and patients with reduced intensity chemotherapy.

Conclusions: Reduced intensity chemotherapy decreased induction mortality compared to intensive chemotherapy. In addition, reduced intensity chemotherapy plus TKI was able to sufficiently induce molecular CR, and subsequent allo-SCT could improve long-term survivals.

Disclosure: The authors declare no conflict of interest.

19: Acute Leukaemia

P040 IMPACT OF CO-MUTATIONS ON THE TRANSPLANT OUTCOMES IN AML PATIENTS WITH DNMT3A MUTATION

Feng-ming Tien1, Xavier Cheng-Hong Tsai 1, Min-Yen Lo1, Hsin-An Hou1, Hwei-Fang Tien1

1National Taiwan University Hospital, Taipei, Taiwan, Province of China

Background: Acute myeloid leukemia (AML) with DNMT3A mutations is classified within intermediate-risk category according to ELN-2022 criteria. However, the impact of co-mutations on the transplant outcomes in this particular group remains to be explored.

Methods: A total of 148 patients (16.7%) with DNMT3A mutations were enrolled from a retrospective cohort of 887 AML patients homogeneously treated with intensive chemotherapy. Gene mutations were examined via targeted NGS, using the TruSight myeloid panel. Notably, none of the patients received oral azacitidine as maintenance therapy. Survival analysis modified according to Simon-Makuch method were used to compare outcomes between allogeneic stem cell transplantation (allo-HSCT) and postremission chemotherapy (PR-CT) in the first complete remission (CR1).

Results: The most frequent co-mutations in DNMT3A mutated patients were NPM1 (52.7%), FLT3-ITD (31.1%), and IDH2 (26.4%). 111 out of 148 (75%) patients achieved CR1. 68 patients received allo-HSCT: 30 with myeloablative conditioning, 38 with reduced intensity conditioning; 15 with a haploidentical donor, 23 with a matched sibling donor, 30 with a matched unrelated donor. Specifically, 30 patients received allo-HSCT at CR1, 11 at CR2, 27 at other disease statuses, resulting in a 5-year overall survival (OS) of 46%, 29%, 15%, respectively. For those with PR-CT without further transplant, the 5-year OS was 13%. Allo-HSCT in CR1 was associated with a better OS than PR-CT (median, not reached (NR) vs 15.3 months, P<0.0001).

Next we delved into the specific genetic subgroup within the DNMT3A mutated patients. In the NPM1-/FLT3-ITD- subgroup, the most frequent co-mutations were IDH2 (47.3%), ASXL1 (18.2%), NRAS (16.4%), IDH1 (16.4%). 37 out of 55 patients attained CR1. The relapse rate was 75% in those receiving PR-CT, and 50% in those receiving allo-HSCT in CR1. The lower relapse rate translate into a significantly better OS (median, 75.1 vs. 19.6 months, P = 0.015) and event-free survival (EFS)(median, 75.1 vs. 8.9 months, P = 0.001) in the allo-HSCT group.

In the NPM1 + /FLT3-ITD- subgroup, currently an ELN-2022 favorable-risk category, 39 out of 47 patients attained CR1. The relapse rate was 36.8% in those receiving PR-CT, and 0% in those receiving allo-HSCT in CR1. Allo-HSCT in CR1 was associated with a significantly better OS (median, NR vs. 98.2 months, P = 0.042) and EFS (median, NR vs. 18.7 months, P = 0.006). Lastly, in the IDH2 mutated subgroup, allo-HSCT in CR1 was associated with a significantly better EFS, while the OS remained similar.

Conclusions: The results indicated that NPM1-/FLT3-ITD- and NPM1 + /FLT3-ITD- subgroups in DNMT3A mutated AML benefit from allo-HSCT in CR1. Larger cohorts are warranted to validate these findings.

Disclosure: No relevant conflict of interest to declare.

19: Acute Leukaemia

P041 SALVAGE TREATMENTS HAVE EXTREMELY POOR OUTCOMES FOR PATIENTS WITH RELAPSED T-ACUTE LYMPHOBLASTIC LEUKAEMIA POST TRANSPLANT

Alexandros Rampotas 1, Fotios Panitsas2, Maximillian Brodermann1, Sridhar Chaganti3, Styliani Bouziana4, Emma Nicholson5, Samantha Drummond6, Sharon Allen7, Andrew King7, Henry Crosland3, Anne-Louise Latif6, Daniele Avenoso4, Claire Roddie1

1University College London Hospital NHS Foundation Trust, London, United Kingdom, 2Laikon University Hospital, Athens, Greece, 3University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom, 4King’s College London Hospital NHS Foundation Trust, London, United Kingdom, 5The Royal Marsden NHS Foundation Trust, London, United Kingdom, 6Queen Elizabeth University Hospital, Glascow, United Kingdom, 7Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom

Background: Relapse following allogeneic hematopoietic stem cell transplantation (allo-HCT) in T-Acute Lymphoblastic Leukemia (T-ALL) poses a formidable clinical challenge. Diverse treatment modalities have been employed, showcasing variable efficacy. Notably, recent years have witnessed the emergence of novel therapeutic strategies for T-ALL such as Chimeric Antigen Receptor T-cells targeting CD7, CD5 and CXCR7 among others with a few phase I/II trials being underway. This study examines outcomes in relapsed T-ALL post allo-HCT, spanning the period from 2010 to 2022, across six major UK transplant centers. Our findings underscore the pressing need for innovative therapeutic interventions in this context, emphasizing the urgency for further research and the development of novel approaches to address the poor outcomes of relapsed T-ALL after transplantation.

Methods: Retrospectively, we compiled data from adult cases experiencing post-transplant relapse in T-ALL. Survival analysis, employing Kaplan-Meier methodology and log-rank tests, was conducted. Descriptive analysis was undertaken to assess baseline and disease characteristics. Approval for this project was obtained from the British Society of Blood and Marrow Transplantation. All data management adhered strictly to the principles outlined in the Declaration of Helsinki.

Results: Twenty-one patients with relapsed T-ALL post allo-HCT were identified, and 20 with sufficient data underwent analysis, with a median follow-up of 32.6 months. Baseline and disease characteristics are detailed in Table 1, Median age was 37 years, while high-risk features included a mediastinal mass in 4/20, early T-cell precursor leukemia in 7/19, high white cell count in 8/19, and CNS disease in 4/20. Relapse occurred at a median of 6 months post-transplant. Intensive chemotherapy, including FLAG-IDA (N = 1), Nelarabine (N = 2), Venetoclax and UKALL14 phase 2 (N = 1), Nelarabine and UKALL14 induction (N = 1), was employed in 5/20 patients, while the rest received non-intensive chemotherapy or palliative and supportive options. Donor Lymphocyte Infusions were not utilised for any patient. Out of 5 patients who received intensive chemotherapy, only 1 achieved CR and four patients went on to receive a second transplant. Among them, one succumbed to transplant complications, while three achieved CR. Two patients relapsed after 12 and 18 months, respectively, while one remains in remission at 26 months post-transplant. Median overall survival was 3.18 months (2-5).

Total cohort

N = 20

Sex

Male 15/20 (75%)

Female 5/20 (25%)

Age (years) (median, IQR)

37, 23-51

Mediastinal mass at diagnosis

Yes 4/20 (20%)

No 16/20 (80%)

Early T-cell precursor leukaemia

Yes 7/19 (37%)

No 12/19 (63%)

Not known 1/19

High White cell count at diagnosis (>100 x10^9/L)

Yes 8/19 (42%)

No 11/19 (58%)

CNS disease

Prior to transplant

Yes 4/20 (20%)

No 16/20 (80%)

At Relapse

Yes 5/20 (25%)

No 15/20 (75%)

Prior line of therapy

1 9/20 (45%)

2 9/20 (45%)

3 2/20 (10%)

Type of relapse post allo-HCT

MRD only 2/20

Extramedullary Relapse 2/20

Morphological Relapse 15/20

CNS only 1/20

Testicular relapse 0/20

Time to relapse post allo-HCT (months) (median, IQR)

6, 4-12

Type of salvage treatment used

Intensive chemotherapy

FLAG IDA 1/20 (5%)

Nelarabine 2/20 (10%)

Nelarabine and UKALL induction protocol 1/20 (5%)

Venetoclax and UKALL phase 2 1/20 (5%)

Non-intensive chemotherapy

Vincristine, steroids and Daratumumab 1/20 (5%)

VIncristine and steroids 4/20 (20%)

Vincristine 1/20 (5%)

Other treatments

Radiotherapy 1/20 (5%)

Palliative treatment 5/20 (25%)

Intrathecal Methotrexate 3/20 (15%)

Second transplant

Yes 4/20 (25%)

No 16/20 (75%)

Donor Lymphocyte Infusion

Yes 0/20 (0%)

No 20/20 (100%)

Median Overall survival (months)

3.18 (2-5)

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (5)

Conclusions: The prognosis for post allo-HCT relapsed T-ALL is notably grim, with limited success observed in second transplant cases. Even within this context, outcomes remain disheartening. The anticipation for improved strategies is fervent, emphasizing the crucial need for innovative treatments. Ongoing investigations into CAR-T cell therapies, specifically CD7 and CXCR7, offer promising avenues. It is imperative that these novel approaches are actively incorporated into clinical practice, and patients are encouraged to participate in relevant trials. The urgency to explore and adopt these advanced therapeutic modalities underscores the commitment to enhancing outcomes for individuals grappling with relapsed T-ALL post allo-HCT.

Disclosure: Alex Rampotas received conference fees by Gilead. Claire Roddie declares Honoraria: Gilead Sciences Consulting or Advisory Role: Autolus, Kite/Gilead, Bristol Myers Squibb/Celgene Speakers’ Bureau: Novartis Pharmaceuticals UK Ltd, Gilead Sciences Travel, Accommodations, Expenses: Gilead Sciences, Autolus. Anna Louise Latif declares honoraria from Kite and Novartis. All other authors declare no conflicts of interest.

19: Acute Leukaemia

P042 FEASIBILITY OF ALLOGENEIC STEM CELL TRANSPLANTATION (HSCT) IN PATIENTS WITH ACUTE MYELOID LEUKEMIA PREVIOUSLY TREATED WITH CPX-351: REPORT FROM A SINGLE CENTER

Sabrina Giammarco 1, Elisabetta Metafuni1, Maria Assunta Limongiello1, Federica Sorá2, Eugenio Galli1, Filippo Frioni2, John Marra2, Patrizia Chiusolo2, Simona Sica2

1Fondazione Policlinico Universitario A. Gemelli, Rome, Italy, 2Universitá Cattolica del Sacro Cuore, Roma, Italy

Background: CPX-351 (VYXEOS®), a dual-drug liposomal encapsulation of daunorubicin and cytarabine in a synergistic 1:5 molar ratio, was approved for the treatment of t-AML or AML-MRC, based on improved survival and remission and comparable safety versus 7 + 3 chemotherapy in a randomized trial in older adults.

The increased use of CPX 351, in association with reduced intensity conditioning regimen and new strategies of GvHD prophylaxis such as PTCY, allows to proceed with HSCT in older patients or previously heavily treated, improving outcomes.

Methods: To assess the feasibility of HSCT in patients with s-AML and t-AML treated with CPX-351, we collected clinical data on 27 consecutive patients submitted to HSCT at our center from February 2019 to October 2023. Then we compared data with 30 patients with AML submitted to HSCT, previously treated with 7 + 3 induction chemotherapy from January 2019 to April 2020.

Results: Of 49 patients treated with CPX-351 from February 2019, all 27 patients with indication to proceed with HSCT, underwent to transplant to consolidate the obtained response. There were 14 males and 13 female, with a median age of 62 yrs (range 43-69). They were all in complete remission except two. Median HCT-CI was 4 (range 0-7). Unrelated donors were the source in 18 patients, haploidentical donors in 8 pts, sibling donors in 1 patient. Stem cell source was peripheral blood in 23 pts, bone marrow in 3 pts and CBU in 1 patient. The median CD34+ cell dose was 4x106/kg (range 0.1-12.779). The main conditioning regimen was the association of Thiotepa, Busulfan and Fludarabine, modulating the busulfan dose according to patient’s comorbidities and performance status, defined by HCT-CI. GVHD prophylaxis regimen was performed with cyclosporine, mycophenolic acid, and cyclophosphamide at day +3 and +5 after HSCT. Engraftment was reached in all patients except one patient who received CBU. Sepsis were identified in 14 patients out 27.

Seven patients developed acute GvHD, all with skin involvement (Stage 2-3); two patients also with gut involvement (Stage 3)and one patient with pulmonary involvement (severe).

1 years OS was 73%. Comparing the two populations, there was no difference in term of gender, disease status at transplant, donor type, donor sex, stem cell source, conditioning regimen, GVHD prophylaxis, sepsis occurrence and GVHD incidence. Significantly statistical difference was found in terms of median age (p = 0.0016), HCT-CI (p = 0.002) as expected, and in terms of median CD34+ stem cell infused, which were significantly higher in the younger comparison population (p = 0.04). (Table 1).

CPX-351

7 + 3 induction

p>0.05

Patients

27

30

Gender male/female

14/12

18/12

p = 0.3

Median age (range)

62 (43-69)

50 (24-68)

p = 0.00016

Disease status at transplant

CR/Refractory

25/2

25/5

p = 0.2

Median HCT-CI (range)

4 (0-7)

1 (0-6)

p = 0.002

Donor type

MUD 8/8

11

10

p = 0.2

MUD 7/8

6

3

Sibling donor

1

6

Haploidentical donors

8

8

CBU

1

3

Donor gender male/female

19/8

23/7

p = 0.2

Stem cell source

PBSC

23

20

p = 0.2

BM

3

7

CBU

1

3

Conditioning regimen

p = 0.5

Myeloablative

26

27

Reduced intensity regimen

1

3

Median CD34 cell dose

x106/kg (range)

4 0.1-1277()

9.45 (0.09-8.9)

P = 0.04

GVHD prophylaxis

p = 0.6

PTCY

31

32

OTHER

3

1

GVHD Incidence n°(%)

7/27 (26%)

14/30 (46%)

p = 0.2

Sepsis n°(%)

14/27 (51%)

16/30 (53%)

p = 0.9

Relapse n°(%)

8/27 (29%)

7/30 (23%)

p = 0.8

Overall Survival n°(%)

19/27 (70%)

23/30 (76%)

p = 0.8

Conclusions: Our monocentric experience showed data in line with those reported in the literature. The growing use of CPX-351 appears to be a safe and effective bridge therapy for HSCT in patients with a poorer prognosis, reproducing outcomes similar to younger population.

Disclosure: No disclosure to declare

19: Acute Leukaemia

P043 COMPARISON OF VENETOCLAX-BASED NON-ANTHRACYCLINE INDUCTION THERAPY VERSUS 3 + 7 FOLLOWED BY HEMATOPOIETIC STEM CELL TRANSPLANTATION IN NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA PATIENTS

Tran-Der Tan 1, Lun-Wei Chiou1

1Koo Foundation Sun Yat-Sen Cancer Center, Taipei, Taiwan, Province of China

Background: The current standard treatment for fit newly diagnosed acute myeloid leukemia patients is idarubicin or daunorubicin plus cytarabine (3 + 7 regimen), followed by allogeneic hematopoietic stem cell transplantation. However, less than half of them could enter into allotransplant because of not remission or the occurrence of treatment-related morbidity or mortality. Venetoclax plus azacytidine or low-dose cytarabine could achieve a high CR rate in unfit or elderly patients in phase III trials.

Methods: We conducted a treatment protocol of venetoclax 100 mg and posaconazole 300 mg a day according to the pharmacokinetics plus a standard dose of cytarabine (100 mg/m2/d x 7 days) as the induction and consolidation therapy, followed by allogeneic hematopoietic stem cell transplantation for newly diagnosed transplant-eligible patients. And we compared the efficacy and outcome with our prior 3 + 7 induction followed by allo-transplant patients.

Results:

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (6)

Between Mar 2019 and Dec 2023, we had 22 consecutive newly diagnosed transplant-eligible AML patients, including 8 therapy-related and 14 de novo AML. Eighteen underwent venetoclax plus standard-dose cytarabine, four underwent venetoclax plus azacitidine induction therapy, and twenty out of twenty-two were followed by allogeneic hematopoietic stem cell transplantation. CRc rate was 94.4% in our cohort and 90.9% in the adverse group of patients. Twenty of twenty-two of our patients were transitioned to allo-transplant, as compared with 46.9% in our previous 3 + 7 induction era between 2009 and 2018. Three-year overall survival was improved from 23% to 53% with the transplant patients’ median age from 43 to 52.8. The duration of neutropenia is similar but there was no significant severe watery diarrhea or oral mucositis in our present cohort as compared to the previous 3 + 7 group of patients.

Conclusions: Venetocla-based non-anthracycline induction therapy is feasible and has more propensity to allogeneic transplant in newly diagnosed AML patients and the pre-transplant CR rate and overall survival were improved as compared with conventional 3 + 7 induction, without more safety concerns.

Disclosure: No disclosure of conflict of interest.

19: Acute Leukaemia

P044 MYELOFIBROSIS GRADE II-III IS AN INDEPENDENT RISK FACTOR FOR RELAPSE OF ACUTE MYELOID LEUKEMIA AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Haixiao Zhang1, Erlie Jiang 2

1Peking Union Medical school, Tianjin, China, 2Peking Union Medical School, Tianjin, China

Background: Acute myeloid leukemia (AML) is a heterogeneous and aggressive disease caused by leukemia blasts. Bone marrow microenvironment (BMM) contributes to AML cell development. Myelofibrosis (MF) is characterized by excessive deposition of bone marrow reticulin fibrosis and other extracellular matrix proteins. MF of unknown etiology in hematological malignancies is acknowledged to be a reactive response to leukemic BMM.

AML with MF (AML-MF) is not recognized as an AML entity by the World Health Organization (WHO) classification system [6]. However, MF was approximately 30-72% at the time of diagnosis of AML and myelodysplastic syndromes (MDS), and 15% was marked reticulin fibrosis. Previous studies have shown that moderate to severe MF is a more advanced risk factor in patients with MDS [8-10]. Data pertaining to the AML-MF treatment and prognosis are very limited [11,12]. As far the only therapy to cure AML has been allogeneic hematopoietic stem cell transplantation (allo-HSCT). MF’s prognostic effect, which can be assessed by regular evaluation at diagnosis, in transplant AML patients is remains unknown.

Methods: To evaluate the prognostic impact of myelofibrosis (MF) on allogeneic hematological stem cell transplantation (allo-HSCT) outcomes in patients with acute myeloid leukemia (AML), we analyzed newly diagnosed patients with AML who received allo-HSCT in the National Longitudinal Cohort of Hematological Disease (NICHE, NCT04645199) from January 2014 to April 2023. Among the 532 patients, 255 (47.93%) patients were classified into the MF-0 group, 209 (39.29%) into the MF-1 group, and 68 (12.78%) into the MF-2-3 group at initial diagnosis according to the European consensus on the grading of MF.

Results: The MF-2-3 group showed poor overall survival, disease-free survival and cumulative incidence of relapse (CIR). Multivariate analyses showed that MF-2-3 (hazard ratio [HR] =2.17, P = 0.023) was an independent risk factor for CIR post-transplantation. Furthermore, MF delayed neutrophil and platelet engraftment and delayed B cell recovery post-transplantation. The MF-2-3 group had a higher cumulative incidence of acute graft-versus-host disease and a lower cumulative incidence of EBV-emia and CMV-emia post-transplantation than the MF-0 and MF-1 groups, however, the difference was not statistically significant.

Conclusions: These findings demonstrate the importance of MF in transplant outcomes and has attracted more attention to AML-MF.

Disclosure: none

19: Acute Leukaemia

P045 INCIDENCE AND MANAGEMENT OF ADULT ACUTE LYMPHOBLASTIC LEUKAEMIA RELAPSE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN LIMITED RESOURCE COUNTRIES

Rihab Ouerghi1, Nour Ben Abdeljelil 1, Sabrine Mekni1, Insaf Ben Yaiche1, Ines Turki1, Rim Dachraoui1, Dorra Belloumi1, Lamia Torjemane1, Rimmel Kanoun1, Saloua Ladeb1, Tarek Ben Othman1

1Service d’Hématologie et de Greffe, Centre National de Greffe de Moelle Osseuse, Tunis, Tunisia

Background: Relapse remains the major cause of failure in adults with high-risk acute lymphoblastic leukemia (ALL) after allogeneic hematopoietic stem-cell transplantation (AHSCT). The management of relapsed patients is a clinical challenge, especially in limited resource countries. The aim of our study was to evaluate the cumulative incidence of relapse (CIR) post AHSCT, identify predictive factors of relapse and evaluate the outcome and management of relapsed ALL in adult patients.

Methods: A retrospective study was conducted in adult patients who underwent AHSCT from HLA-identical sibling donors between November 2003 and December 2022. Conditioning regimens consisted of fractionated TBI (F-TBI) (3fractions/9.9Gy) with etoposide or cyclophosphamide or chemotherapy-based regimen (intravenous busulfan). GVHD prophylaxis consisted of cyclosporine and short course of methotrexate. Stem cell sources were bone marrow (BM) or peripheral blood stem cell.

Results: One hundred and five patients (69 males and 36 females) were included. Median age is 30 years (18-50 years): 60 B-ALL (57%) and 45 T-ALL (43%). Cytogenetic analysis was available for 83 patients: 33% of patients had high-risk cytogenetic abnormalities (25% had t(9,22) or positive BCR-ABL transcript). Before AHSCT, 86% of patients were transplanted in first complete remission (CR1). MRD was performed in 66% patients (MFC and/or molecular biology) and was positive in 55% of patients. The median time from diagnosis to AHSCT was 6 months (2-137m). Conditioning regimens were TBI-based in 60% of patients and 51% of patients received peripheral blood stem cell. CMV reactivation was observed in 41% of patients. The CI of acute GVHD (≥grade 2) and chronic GVHD were 32% and 50%, respectively. The CI of NRM was 17.6%. The 2-year CIR was 30% (medullary and combined relapse n = 26, extramedullary relapse n = 6, molecular relapse n = 4). The median time of relapse was 9 months (2-46m). Twenty-two patients were managed with palliative cares and 14 received salvage therapy (chemotherapy +/- radiotherapy n = 9, donor lymphocyte infusion n = 1, ITK only n = 4). A second CR was obtained in 8 patients and 3 patients underwent a second AHSCT. At last follow-up, 7 patients are alive. After a median follow up of 29 months (1-207m), the 2-year OS and EFS for the entire cohort and relapsed patients were 60% vs 31% and 53% vs 14%, respectively. At last follow-up, 49 patients (47 %) died. The main cause of death was relapse (n = 26; 53%). In univariate analysis, chemotherapy-based conditioning regimen was the only significant factor of CIR (p = 0.01).

Conclusions: The prognosis of adult patients relapsing after AHSCT is dismal. To reduce the risk of relapse TBI based conditioning regimen and immunotherapy should be considered when possible.

Disclosure: Nothing to declare

19: Acute Leukaemia

P046 IMPACT OF BODY MASS INDEX ON OUTCOME IN ADULT PATIENTS WITH ACUTE MYELOID AND ACUTE LYMPHOBLASTIC LEUKEMIA UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Iveta Oravcova1,2, Zuzana Rusinakova1, Eva Mikuskova1, Miriam Ladicka1, Ludmila Demitrovicova1, Alica Slobodova1, Vanda Mikudova1, Jana Spanikova1, Radka Vasickova1, Denis Urban1, Kristina Bandurova1, Lubos Drgona1,2, Silvia Cingelova 1

1National Cancer Institute, Bratislava, Slovakia, 2Faculty of Medicine Comenius University, Bratislava, Slovakia

Background: Obesity has been recognized as risk factor for many types of cancer and at the same time it is considered as an adverse prognostic factor associated with cancer survival. This study assessed the effect of body mass index (BMI) on transplantation outcomes in allogeneic hematopoietic cell transplantation (HCT) in AML and ALL recipients.

Methods: We retrospectively analyzed 98 adult AML and ALL patients who were followed up after alloSCT in National Cancer Institute in Slovakia between March 2013 and October 2023.

According to the World Health Organization (WHO) international BMI classification, patients were classified as obese when BMI was ≥ 30kg/m2. We divided patients into 2-groups: BMI < 30kg/m2 (non-obese patients) and BMI ≥ 30kg/m2 (obese patients).

Results: At median follow up 22,3 (4,1-125,8) months, 98 patients entered the analysis (median age at alloSCT 49 years, 53 (54,5%) male). Median BMI value was 24,84 kg/m2. We identified 80 patients with BMI < 30kg/m2 (81,6%) and 18 (18,4%) with BMI ≥ 30kg/m2. There was no difference in the prevalence of obesity between the genders (p = 0,299).

1 and 3- year overall survival (OS) for the BMI < 30kg/m2 group was 68,6% (CI 95%; 58,1% - 79,0%) and 45,9% (CI 95%; 33,3% - 58,5%) with median OS 34,5 months (CI 95% 18,3-57,0); and for the obese group 32,5% (CI 95%; 9,4% - 55,6%) and 13,0% (CI 95%; 0,0% - 27,9%) with median OS 4,6 months (CI 95% 3,0-8,5). The difference between groups was statistically significant, HR 0,34 (95% CI 0,15-0,79); (p = 0,0002).

1 and 3- year relapse free survival (RFS) for the BMI < 30kg/m2 group was 58,8% (CI 95%; 47,8% - 69,8%) and 41,2% (CI 95%; 29,1% - 53,4%) with median RFS 30,1 months (CI 95% 8,5-42,9); and for the obese group 20,7% (CI 95%; 0,4% - 41,1%) and 13,8% (CI 95%; 0% - 31,3%) with median RFS 3,5 months (CI 95% 1,9-4,4). The difference between groups was statistically significant, HR 0,39 (95% CI 0,18-0,87); (p = 0,001).

Obese patients have higher 12-months cumulative incidence of relapse rates than non-obese patients: 53,7% (CI 95%; 29,1%-99,3%) vs 29,5% (CI 95%; 20,5%-42,3%). There was no difference in non-relapse mortality between obese and non-obese patients.

Conclusions: Patients with BMI ≥30kg/m2 had significantly lower one and three-year survival rates and higher cumulative incidence of relapses than those with BMI <30kg/m2.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P047 VENETOCLAX COMBINED WITH HMAS-BASED THERAPY AS PRE-EMPTIVE TREATMENT FOR ACUTE MYELOID LEUKEMIA PATIENTS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Mimi Xu 1,2,3, Yuqing Tu1,2,3, Mengqi Xiang1,2,3, Yi Fan1,2,3, Xiang Zhang1,2,3, Tiemei Song1,2,3, Lian Bai4, Xiaoli Li5, Yang Xu1,2,3, Yuejun Liu1,2,3, Jia Chen1,2,3, Depei Wu1,2,3

1National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China, 2Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China, 3Key Laboratory of Stem Cells and Biomedical, Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China, 4Canglang of Suzhou Hospital, Suzhou, China, 5Soochow Hopes Hematonosis Hospital, Suzhou, China

Background: Accumulating evidences indicated the effectiveness and importance of measurable residual disease (MRD) guided pre-emptive therapy in acute myeloid leukemia (AML) patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, limited data is available on the pre-emptive utility of venetoclax (VEN) for MRD positive in AML patients post-HSCT. This study was conducted to explore the efficiency and tolerability of VEN combined with hypomethylating agents (HMAs) based treatment for AML patients with MRD positive post-HSCT.

Methods: We collected retrospectively the data from 10 patients who underwent first allo-HSCT with high-risk AML between July 2020 and April 2022 and initiated VEN-based pre-emptive threapy for MRD positivity between March 2021 and July 2022 in our center for analysis. Patients who experienced haematological relapse before VEN-based pre-emptive treatment were excluded. The VEN combined with HMAs-based treatment regimen comprised 100 mg VEN daily on day 1 to day 21 and 15 mg/m2 decitabine on day 1 to day 3 or 75mg/m2 azacytidine on day 1 to day 5 (or day 7). FLT3-ITD positive patients also added FLT3 inhibitor sorafenib (400mg scheduled for days 1 to 21). The primary endpoints of this study were MRD response, overall survival (OS) and event free survival (EFS). The secondary endpoints included adverse events (AEs), cumulative incidence of relapse (CIR) and nonrelapse mortality (NRM).

Results: 10 patients were included. The median age of the patients at the time of transplantation was 49 (range, 16-57)y. The rate of MRD response was 70.0% (7/10) and 71.4% (5/7) of whom achieved MRD response after first circle of VEN-based combination threapy. The median time from initiating VEN-based pre-emptive threapy to MRD response was 64 (10-175) d. For MRD response patients, two of whom appeared hematologic relapse on day 271 and 336 after MRD response respectively. Three patients had not achieved MRD response, 2 of whom experienced hematological relapse and 1 died from severe GVHD and transplantation associated thrombotic microangiopathy. During the study, we found no tumor lysis syndrome and severe AE for all patients. The median follow-up was 440.5 (88-872) d. 5 patients still had EFS and were alive with MRD negative CR at the time of last follow-up. Overall, the 1-y OS and EFS after VEN-based pre-emptive threapy were (80.0±12.6)% and (60.0±15.5)%, respectively. The cumulative incidence of relapse (CIR) at one years was (30.0±2.4)%, and nonrelapse mortality was (10.0±1.0)%.

Table 1 Patient characteristics

Total patients

10

%

Median age at transplantation, y (range)

49(16-57)

Gender

Male/Female

5/5

MRD positive detection method before pre-emptive threapy

Flow cytometry

4

40

PCR

8

80

DNA sequence

2

20

Disease status at transplant

CR

9

90

MRD−

1

10

MRD+

8

80

NR

1

10

Donor type

Haploidentical

7

70

Identical sibling

1

10

Unrelated

2

20

Median days of neutrophils ≥0.5 × 109/L after transplant

11(10-14)

Median days of platelets ≥20 × 109/L after transplant

12.5(10-74)

Conclusions: We conclude that VEN combined with HMAs-based regimen as pre-emptive treatment is efficient for patients with AML post-HSCT and with acceptable side effects.

Disclosure: Nothing to declare

19: Acute Leukaemia

P048 VENETOCLAX, AZACITIDINE, COMBINED WITH LOW-DOSE CYTARABINE IMPROVE THE REMISSION RATE IN OLDER OR UNFIT PATIENTS WITH NEWLY DIAGNOSED ACUTE MYELOID LEUKEMIA

Xiao Han1, Qingxiao Song 1, Kai Wan1, Mengyun Zhang1, Xue Liu1, Hongju Yan1, Cheng Zhang1, Qin Wen1, Xi Zhang1

1Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing, China

Background: Older or unfit patients with acute myeloid leukemia (AML) have a dismal prognosis. The combination of venetoclax with azacitidine had promising efficacy, but the minimal residual disease (MRD) negative rate is only 30-40%. In most cases measurable residual disease (MRD) positivity predicts hematologic relapse potentially allowing early therapeutic intervention.

Methods: This is a prospective, one-arm, multicenter, open clinical trial, Adults age ≥18 years with newly diagnosed AML ineligible for intensive chemotherapy were enrolled. Patients received induction treatment with venetoclax 100mg d1, 200mg d2, 400mg d3-28, azacytidine 75mg/m2 d1-7, cytarabine 10mg/m2 q12h d1-7. The primary observation was the remission rate (CR/CRi) and the negative rate of MRD.

Results: The baseline characteristics of 46 older or unfit patients who can evaluate the efficacy are presented in Table 1. Following the first course of Ven+AZA + LDAC treatment, the outcomes were as follows: 27 patients (58.7%) achieved complete remission (CR) with negative minimal residual disease (MRD), 14 patients (30.4%) achieved CR with positive MRD, 1 patients (2.2%) experienced partial remission (PR), and 4 patient (8.7%) did not achieve remission (NR). The overall response rate (ORR) was 91.3%. Subgroup analysis revealed that out of the 23 older patients, 16 (69.6%) achieved CR with negative MRD, 6 (26.1%) achieved CR with positive MRD. Among the 23 initially diagnosed unfit patients, 11 (47.8%) reached CR, with 8 (34.8%) showing negative MRD, 1 (4.3%) experiencing PR, and 3 (13.0%) not achieving remission. The median follow-up time was 8.3 months (range: 3-24.3 months), and the estimate median overall survival (OS) was 20.34 months. Subgroup analysis revealed that older patients had better survival outcomes compared to unfit patients, although the difference was not statistically significant. Among them, 9 patients had underwent allogeneic hematopoietic stem cell transplantation (HSCT), and 8 of the 9 HSCT patients were alive at the time of manuscript writing, One patient died because of SCT related thrombotic microangiopathy (TMA). This observation might be attributed to the limited sample size, indicating the necessity of gathering more data for comprehensive analysis. Safety: All the 23 older patients had neutropenia, which controlled after therapy, without severe infection and bleeding complications. 23 patients with unfit had severe complications such as severe infection, respiratory failure, or intracranial hemorrhage before treatment, but no treatment-related deaths occurred during the induction period.

Characteristic

N = 46

Frequency

Median (range)

59.29(32-73)

Older (average age)

23 (49.7)

50%

unfit (average age)

23 (66.4)

50%

Male, n (%)

25

54.35%

Baseline neutropenia, n(%)

Grade 3

7

15.2%

Grade 4

15

32.6%

FAB classification, n (%)

AML-M1

7

15.2%

AML-M2

20

43.5%

AML-M4

6

13.1%

AML-M5

10

21.7%

others

3

6.5%

Cytogenetic risk category, n (%)

Favorable

9

19.6%

Intermediate

20

43.5%

Adverse

17

36.9%

Somatic mutations

DNMT3A

12

30.77%

NPM1

11

28.21%

FLT3-ITD

10

25.64%

IDH2

7

17.95%

CEBPA

6

15.38%

IDH1

5

12.82%

TP53

3

7.69%

Transfusion dependent at baseline, n (%)

Red blood cells

15

32.6%

Platelets

13

28.3%

Conclusions: The incidence of remission was higher among newly diagnosed older patients who received Ven+AZA + LDAC, especially CR with negative MRD. However, the remission rate of unfit AML patients has been improved compared to traditional chemotherapy, but the negative rate of MRD has not improved. Infection, chemotherapy-related mortality, and the tolerability were acceptable. At the same time, it would be contribute to provide transplant conditions for patients and prolong survival. However, it is necessary to further expand the sample size and extend the follow-up time to assess the long-term survival rate.

Clinical Trial Registry: ChiCTR2200063804

Disclosure: Nothing to declare.

19: Acute Leukaemia

P049 TARGET THERAPY IN ACUTE LEUKAEMIA RELAPSES AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Riccardo Boncompagni 1, Chiara Camerini1, Ilaria Cutini1, Mirella Giordano1, Antonella Gozzini1, Chiara Nozzoli1, Edoardo Simonetti1, Riccardo Saccardi1

1BMT Unit, Careggi University Hospital, Firenze, Italy

Background: New drugs targeting acute leukaemia antigens or cellular survival/proliferation pathways are granting promising results in patients with relapsed and/or refractory acute leukaemia leading more patients to achieve a control of disease and access to allogenic hematopoietic stem cell transplantation (HSCT), the only potentially curative treatment. Here we investigated if target-therapy can confer a real-life survival advantage also in patients who relapse following HSCT, a population characterized from a very poor prognosis.

Methods: We retrospectively analysed data from patients affected by acute myeloid, mixed-phenotype, lymphoblastic leukaemia (AML, MPAL, ALL respectively) and blast crisis of chronic myeloid leukaemia (CB-CML) who performed allogeneic HSCT at Careggi University Hospital of Florence in the last 10 years. Primary endpoint of the current research was to compare overall survival (OS) and progression free survival (PFS) between patients treated or not with target therapy. Target therapy was administered based on availability at our institution.

Results: We found 288 HSCT performed for 275 patients. Indication for HSCT was AML for 196 (68%), ALL for 74 [24%; 25 (34%) with BCR/ABL mutation, 12 (16%) of T-lineage], CB-CML for 11 (4%) and MPAL for 10 (3%). Median age was 50 (19 – 69) and median comorbidity index (HCT-CI) was high (3; 0 – 9); however reduced intensity conditioning regimen was used only in 93 (32%). A significant number of AML was in active disease status at the time of HSCT (74, 38%), and molecular or cytofluorimetric minimal residual disease was detectable in more than half of the others (86 out of 122, 70%). Median OS was not reached, with 57% of patients alive with a 69 months median follow-up. Relapse incidence after HSCT was 36% (n = 100) and median PFS was reached at 18 months (95% confidence interval [CI]: 7 – 29). In the group who received a target therapy (n = 63) we observed both an OS (45 vs 7 months; p < 0,001) and PFS advantage (26 vs 2 months; p 0<,001). Donor lymphocytes infusions (DLI) was administered in 38% (n = 24) of target therapy group and conferred a further OS advantage (11 vs 42 months; p = 0,022). Finally, 3 out of 13 (23%) patients who performed a second HSCT after savage therapy with target therapy were alive and in complete remission at the time of their last follow-up.

Conclusions: Despite the low potency of monocentre retrospective study, our results support the hypothesis that new therapies for acute leukaemia are effective for patients who relapse following HSCT, and the possibility of combination of DLI and of a subsequent second HSCT may grant a longer survival in this very high-risk population.

Disclosure: We declare no potential conflict of interest.

19: Acute Leukaemia

P050 ALLOGENEIC STEM CELL TRANSPLANTATION IN THE MANAGEMENT OF ADULT ACUTE LYMPHOBLASTIC LEUKAEMIA: A TEN YEAR EXPERIENCE IN IRELAND’S NATIONAL ADULT ALLOGENEIC TRANSPLANT CENTRE

Conor Browne 1, Micheal Brennan1, Mairead Ni Chonghaile1, Sarah Maher1, Greg Lee1, Deirdre Waldron1, Nicola Gardiner1, Michelle Regan1, Lorraine Brennan1, Carmel Waldron1, Catherine M. Flynn1, Eibhlin Conneally1, Patrick J. Hayden1, Robert Henderson1, Elisabeth Vandenberghe1, Christopher L. Bacon1

1St James Hospital, Dublin 8, Ireland

Background: Adult acute lymphoblastic leukaemia (ALL) historically carries a poor prognosis with cure rates of less than 40%. Despite advances in therapeutics, allogeneic stem cell transplantation (AlloSCT) remains pivotal in the management of patients with high risk adult ALL.

Methods: The records of consecutive ALL patients receiving AlloSCT between 01/01/2013 and 31/12/2022 were retrieved from the St. James’ Hospital transplant-database and their electronic medical records. Progression free survival (PFS) and overall survival (OS) were estimated via the Kaplan-Meier method.

Results: 89 patients with ALL received AlloSCT. Clinical details are shown in Table-1. 74 (83%) had B-ALL with a median white cell count (WCC) of 30x109/L (0.5-830) at diagnosis. 15 (17%) had T-ALL with a median WCC of 100x109/L (4.9-470) at diagnosis.

38 patients (43%) had high-risk cytogenetics defined as having either a Philadelphia chromosome t(9;22)(q34;q11), N = 17 (9%), KMT2A rearrangement t(4;11)(q21;q23) N = 12 (13%), complex karyotype N = 2 (2%), low-hypodiploid N = 4 (4%) or near-triploid haplotype N = 3 (3%). 64 patients (72%) achieved complete remission (CR) post induction therapy. 13 (15%) had primary refractory disease and 16 (18%) in remission relapsed prior to transplant requiring re-induction. 73 (82%) were transplanted in CR1, 14 (16%) in CR2, 1 in CR3 and CR4.

The median time from diagnosis to transplant was 6 months (2-85). The median CD34 cell count infused was 3.32x106/kg (0.67-5.7) with a median CD3 cell count of 3.08x107/kg (1.21-52). The median days to neutrophil engraftment was 21 (10-34) and to platelet engraftment was 18 (9-96).

66 patients (74%) developed graft-versus-host-disease (GVHD); 63 (71%) acute-GVHD and 30 (34%) chronic-GVHD. Acute-GVHD occurred at a median of 30 days (13-298) post-transplant. 16 (18%) experienced Grade I, while 47 (53%) developed Grade II-IV acute-GVHD. Of those with acute-GVHD, 15 cases (24%) were steroid refractory requiring additional therapies including ruxolitinib, mycophenolate-mofetil, extracorporeal-photopheresis and ATG.

30 patients (34%) developed viral reactivation involving CMV, EBV or adenovirus. 5 patients had associated EBV post-transplant lymphoproliferative disorder.

9 patients (10%) received donor-lymphocyte infusions (DLI) at a median time of 12 months (4-33) post-transplant. 8 received pre-emptive DLI for mixed chimerism, 1 received therapeutic-DLI for relapsed disease. 3 patients developed acute-GVHD post-DLI. 7 cases (80%) achieved complete donor chimerism post-DLI.

The median duration of follow-up was 36 months (1-129). 18 patients (20%) relapsed at a median time of 8.5 months (1-63) post-transplant. Treatment strategies at relapse included blinatumomab, inotuzumab, Hyper-CVAD, nelarabine, TKIs, palliation and CAR-T therapy.

27 patients (30%) died during follow-up at a median time of 9 months (1-71) post-transplant; relapse-related mortality N = 11 (12%), transplant-related mortality (TRM) N = 14 (16%), lost to follow-up N = 2 (2%). Causes of TRM included GVHD (N = 3), veno-occlusive disease (N = 1), infection (N = 7), EBV-PTLD (N = 1), neurological complication (N = 1) and secondary malignancy (N = 1). The 100-day TRM rate was 6%.

The 5-year OS was estimated at 69% with a 5-year PFS of 62% as shown in Graph-1. OS or PFS didn’t differ significantly between those with acute-GVHD and those without (p = 0.7).

Table 1. Clinical Details

N = 89

Transplant Details

N = 89

Stem Cell Source: no. (%)

Median Age at Transplant (Range)

36 (16-61)

Bone Marrow

58 (65)

Peripheral blood

31 (35)

Gender: no. (%)

Conditioning Intensity: no. (%)

Male

50 (56)

Myeloablative

68 (76)

Female

39 (44)

Reduced Intensity

21 (24)

Induction Regimen: no. (%)

UKALL 12/14

64 (72)

MAC Conditioning Regimen: no. (%)

UKALL 03/11

14 (15)

Cyclophosphamide/TBI

60 (67)

Hyper-CVAD

9 (10)

Cy/TBI + ATG

4 (4)

FLAG-Ida

1 (1)

Busulphan/Cyclophosphamide

2 (2)

N/A

1 (1)

Fludarabine/TBI+PTCy

2 (2)

Donor Status: no. (%)

RIC Conditioning Regimen: no. (%)

Related

35 (39)

Flu/Melphalan/Alemtuzumab

20 (22)

Unrelated

54 (61)

Cy/TBI/Alemtuzumab

1 (1)

Co-Morbidity Index: no. (%)

GVHD Prophylaxis: no. (%)

HCT-CI 0-1

49 (55)

Tacrolimus

49 (55)

HCT-CI >/ = 2

28 (32)

Ciclosporin

38 (43)

HCT-CI N/A

12 (13)

PTCy/MMF

2 (2)

Recipient Viral Status: no. (%)

HLA status: no. (%)

CMV IgG +ve

39 (44)

Matched 10/10

76 (85)

EBV IgG +ve

81 (91)

Mismatched (single allelic)

13 (15)

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (7)

Conclusions: AlloSCT in high-risk patients is an effective strategy for the management of adult ALL with a 5 year OS of 69% in this cohort.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P051 FLT3-MUTATED AML: MANAGEMENT AND LONG-TERM OUTCOME OVER 24 YEARS AT A SINGLE CENTER

Saveria Capria 1, Silvia Maria Trisolini1, Lorenzo Torrieri2, Elena Amabile2, Giovanni Marsili3, Alfonso Piciocchi3, Walter Barberi1, Daniela Diverio1, Daniela Carmini1, Massimo Breccia2, Maurizio Martelli2, Clara Minotti1

1AOU Policlinico Umberto I, Rome, Italy, 2Sapienza University, Rome, Italy, 3GIMEMA Foundation, Rome, Italy

Background: The introduction of tyrosine kinase inhibitors (TKIs) in the treatment of FLT3-mutated AML has been a therapeutic breakthrough. However, prognosis remains challenging.

Methods: We retrospectively analyzed all patients with FLT3-mutated AML who were eligible for intensive therapy and treated at our institution since 1999, focusing on changes in management over time and the impact of TKI treatment.

Results: The cohort comprised 140 patients (68M/72F) with a median age of 51 years (14-73), 28 (21%) had WBC ≥100,000/µl at diagnosis and 38 were older than 60 years. The ITD/TKD ratio was 120/20 and 73 patients (52%) had NPM1 co-mutation.

Until 2018, 101/140 patients received the DCE regimen without TKI, while 39/140 patients diagnosed since 2018 received 3 + 7+midostaurin.

The overall CR rate was 64% (90/140), and 73% (53/73) in NPM1-mutant patients, regardless of TKI treatment during induction.

The CR rate was similar in both younger and older patients and was positively influenced by co-existing NPM1 mutation (p: 0.032) and WBC count <100,000/µl (p = 0.013) in univariate analysis. TKI administration during induction did not have a significant effect.

In a multivariate model adjusting for sex and age, only WBC count was confirmed to have an independent prognostic impact on response (OR 3.04 – CI 1.23-7.71 – p = 0.017).

Allogeneic transplantation was performed in 41/90 patients who achieved CR1 (22/60 no-TKI and 19/30 TKI), the transplantation rate between no-TKI and TKI patients is not statistically significant. Autologous transplantation was performed in 18 patients. No transplantation was performed in 31 patients.

The median probability of EFS at 3 and 6 years is comparable between autografted and allografted patients (1.1 vs. 1.6 years respectively – p = 0.9).

The cumulative incidence of 1-year non-relapse mortality was 0.00% for autologous and 28% (CI: 15-42) for allogeneic transplantation (p = 0.007), while the cumulative incidence of 1-year and 3-year relapse was 39% (CI: 17-61) vs. 15% (CI: 6.1-28) and 57% (CI: 30-77) vs. 21% (CI: 9.5-35), respectively (p = 0.004).

There was no statistical difference in outcome based on TKI administration at induction, although these data may be affected by the smaller number of patients in the TKI group.

Forty-three patients (34 no-TKI and 9 TKI) were refractory to induction. Twenty-five received salvage treatment (10 CHT + TKI, 4 single-agent TKI, 11 standard CHT). Of the 14 patients who received a TKI-containing regimen, 11 underwent allogeneic transplantation while none of the patients in the standard CHT group underwent transplantation. The 5-year OS of transplanted patients after salvage treatment is 51%.

Overall, 70/140 patients received stem cell transplantation, 18 autologous and 52 allogeneic (41 CR1 and 11 refractory). In 8/52 cases, preemptive TKI treatment was administered post-transplant due to FLT3 mutation recurrence in morphologic CR; all patients are alive at a median of 65 months (range 16-98) post-transplant and TKI treatment was discontinued after 2 years of continuous CR.

Conclusions: Allogeneic transplantation remains crucial in FLT3-mutated AML. However, several unanswered questions remain, such as how to optimize the use of different inhibitors to achieve the best pre-BMT response, and the potential impact of preemptive post-transplant therapy.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P052 THE ROLE OF TOLEROGENIC DENDRITIC CELLS IN SUPPRESSING AND DCLEU IN INCREASING ANTILEUKEMIC CYTOTOXICITY

A.S. Hartz1, L. Li1, H. Aslan1, E. Pepeldjiyska1, E. Rackl1, T. Baudrexler1, P. Bojko2, J. Schmohl3, A. Rank4, C. Schmid4, H.M. Schmetzer 1

1Working-Group: Immune-Modulation, Klinikum Grosshadern, Ludwig-Maximilians-University, Munich, Germany, 2Rotkreuzklinikum Munich, Munich, Germany, 3Department of Hematology and Oncology, Stuttgart, Germany, 4University Hospital of Augsburg, Augsburg, Germany

Background: Novel immunotherapies that overcome immunescape of acute myeloid leukemia (AML) cells are urgently needed. AML establishes a tolerant, immune-inert microenvironment e.g. by accumulating tolerogenic dendritic cells (tDCs). TDCs are dendritic cells and can be generally characterized by a modified cytokine production, ultimately leading to T-cell anergy. In this study we focused on tDCs expressing ILT-3, CTLA4, PD-1, IL3RA.

Leukemia-derived dendritic cells (DCleu) can be generated ex vivo directly from leukemic whole blood (WB) by using (clinically approved) GM-CSF and PGE-1 (Kit-M), leading to antileukemic immune responses after mixed lymphocyte culture enriched with patients’ T-cells (MLC).

Aims: To evaluate Kit-M on frequencies of DCleu or tDCs and on DC mediated (antileukemic) effects and to correlate results with patients’ clinical characteristics.

Methods: WB samples from 18 AML patients and 5 healthy WB samples were treated with vs without Kit-M to generate DC/DCleu and to quantify tDCs. After MLC, leukemia specific/antileukemic effects were assessed through the degranulation-, the intracellular IFNγ production- and the cytotoxicity fluorolysis assay. Quantification of cell subtypes was performed via flow cytometry.

Results: Treating leukemic WB with Kit-M vs control increased significantly frequencies of (mature) DCleu without induction of blast proliferation and significantly decreased frequencies of tDCs (DCs co-expressing ILT-3, CTLA4, PD-1 or IL3RA). After T-cell enriched MLC of Kit-M treated vs. not pretreated WB, frequencies of immunoreactive cells (e.g. non-naïve T-cells, CIK cells) were (significantly) increased, of regulatory T-cells (e.g. CD152 + T-cells) were (significantly) decreased, of activated T-cells (e.g. CD154 + T-cells) were increased and degranulating (CD107a + ) and intracellular IFNγ positive (e.g. CD107a+ non-naïve, - central memory, - non-naïve) T-cells were (significantly) increased. A cytotoxicity fluorolysis assay showed an improved blast lysis in Kit-M treated WB compared to control. We found negative correlations between frequencies of tDCs and improved blastolytic functionality and confirmed positive correlations between frequencies of DC/DCleu and improved blastolytic functionality. In patients with favourable (vs adverse) ELN risk stratification and response to chemotherapy (vs no response) lower frequencies of tDCs were found.

Conclusions: Kit-M treatment of leukemic WB was shown to induce DC/DCleu, to reduce tDCs and to induce leukemia-specific antileukemic immune cells after mixed lymphocyte culture. Reduction of tDCs correlated negatively with improved blastolytic functionality. Kit-M in vivo treatment could lead to an improved immune response and overcoming the immunosuppressive tumor environment in vivo.

Disclosure: Modiblast Pharma GmbH (Oberhaching, Germany) holds the European Patent 15 801 987.7-1118 and US Patent 15-517627 ‘Use of immunomodulatory effective compositions for the immunotherapeutic treatment of patients suffering from myeloid leukemias’, with whom H.M.S. is involved with.

19: Acute Leukaemia

P053 TREATMENT EFFICACY OF BLINATUMOMAB FOR MAINTENANCE THERAPY OF PATIENTS WITH B-LINEAGE ACUTE LYMPHOBLASTIC LEUKEMIA RECEIVING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION

Jiayu Huang1, Luxiang Wang1, Chuanhe Jiang1, Zilu Zhang1, Zengkai Pan1, Ling Wang2, Jieling Jiang2, Jiong Hu2, Jun Zhu3, Lijing Shen4, Suning Chen5, Yang Cao6, Xiaoxia Hu 1

1State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 2Shanghai Institute of Hematology, Blood and Marrow Transplantation Center, Collaborative Innovation Center of Hematology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 3GoBroad Medical Institute of Hematology (Shanghai Center), Liquan Hospital, Shanghai, China, 4Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 5National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China, 6Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

Background: Allogeneic hematopoietic cell transplantation (allo-HSCT) is still a potentially curative therapy for high-risk B-lineage acute lymphoblastic leukemia (B-ALL). However, relapse is the major cause for transplant failure, especially within the first two years after allo-HSCT.

Methods: This multicenter, retrospective study was designed by the First Affiliated Hospital of Soochow University, Wuhan Tongji Hospital, Shanghai Renji Hospital, Shanghai Zhaxin Hospital and Shanghai Ruijin Hospital, to evaluate the efficacy and safety of blinatumomab as maintenance therapy after allo-HSCT in B-ALL patients. Consecutive patients diagnosed with B-ALL from November 2021 to April 2023 were screened, and the eligibility criteria were as followed: (1) aged ≥ 14 years; (2) patients were MRD-negative measured by flow cytometry with a sensitivity of 0.02%; (3) B-ALL with CD19 expression; (4) patients with complete medical information. The last follow-up was November 30, 2023. As pre-determined schedule, blinatumomab was administrated as a continuous infusion for 14-21 days.

Results: A total of 17 patients were retrospectively enrolled. The median age was 30-year old (range, 14-60). Before allo-HSCT, one patient was in relapse, four in second complete remission (CR) and the other patients (n = 12) in first CR. 76.5% patients (n = 13) were categorized as high-risk B-ALL according to NCCN 2023. 88.2% (n = 15) patients received graft from haploidentical donors. The median interval from transplant to the start of blinatumomab maintenance was 102 days (range, 42 to 227), with a median course of 1 (range, 1-6). Only four treatment courses were temporarily discontinued due to infection in the first cycle of blinatumomab maintenance, but the remaining thirty treatment courses were completed without interruptions. With a median follow-up of 269 days (range, 120-674), the 1-year overall survival (OS), event-free survival, relapse, and graft versus host disease (GVHD)-free and relapse-free survival for all patients were 69.1% (95% CI 44.2%-100%), 72.6% (95% CI 49.9%-100%),10.4% (95% CI 8.3-12.5%) and 72.9% (95% CI 50.3%-100%) respectively. One patient (P15) received one cycle of blinatumomab at 3-month after allo-HSCT, and relapsed at 8.5 months. Outcomes of patients based on different classification were shown in Table 1. Patients with negative MRD pre-HSCT had a superior OS as compared to those with positive MRD (not reached vs. 286 days P = 0.024). Two patients died of non-relapse causes related to blinatumomab, including transplant-associated thrombotic microangiopathy (n = 1) and blood stream infection (n = 1). The incidence of grade III-IV acute GVHD associated with blinatumomab was 2.9% (n = 1).

Table 1: Follow-up outcome of allo-HSCT patients based on MRD pre-allo-HSCT or R-DRI or NCCN 2023 risk

Classification

Total (n)

Outcome

Sustained remission (n,%)

Relapse (n,%)

NRM (n,%)

Pre-allo-HSCT

MFC-MRD-

14

13(92.9)

0(0)

1(7.1)

MFC-MRD+

3

1(33.3)

1(33.3)

1(33.3)

R-DRI

Intermediate risk

12

11(91.7)

0(0)

1(8.3)

High risk

5

3(60)

1(20)

1(20)

NCCN 2023

Standard risk

4

3(75)

0(0)

1(25)

High risk

13

11(84.6)

1(7.7)

1(7.7)

Conclusions: Blinatumomab maintenance after allo-HSCT is safe and feasible, however, its long-term effect on immune reconstitution should be concerned.

Clinical Trial Registry: Not applicable.

Disclosure: This work was supported by the National Natural Science Foundation (82170206 to X.H.) and all authors have no conflict of interest to declare.

19: Acute Leukaemia

P054 EFFICACY, SAFETY AND TOLERABILITY OF MITOXANTRONE HYDROCHLORIDE LIPOSOME CONTAINING REGIMEN IN TREATING RELAPSED ACUTE MYELOID LEUKEMIA AFTER ALLOGENIC STEM CELL TRANSPLANTATION

Li Liu 1, Yigeng Cao1, Ni Lu1, Erlie Jiang1

1Center of Hematopoietic Stem Cell Transplantation, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin, China

Background: Disease relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a prevalent cause of poor prognosis and decreased survival rate for patients (pts) diagnosed with acute myeloid leukemia (AML). Currently, chemothrapy is one of the common strategies, however its efficacy is still limited. Mitoxantrone hydrochloride liposome (Lipo-MIT) is an innovative drug which could potentially provide a new approach for relapsed AML after allo-HSCT.

Methods: From April 29, 2022 to April 19, 2023, 13 adults with relapsed AML after allo-HSCT were enrolled (patient’s characteristics in Table 1). All ofpatients received the Lipo-MIT containing regimen with the dose levels of Lipo-MIT between 20 mg/m2 to 24 mg/m2, followed with donor lymphocyte infusion (DLI). The median dose of Lipo-MIT administered in clinical practice was 19.90 (range 17.00-31.51) mg/m2 per cycle.

Results: The study included a total of 13 pts with a median age of 35 (range 20-53) years old. The risk classification at initial diagnosis, based on the 2022 European LeukemiaNet (ELN) criteria, was favorable (1/13), intermediate (8/13) and adverse (4/13). Prior to the last allo-HSCT, all patients achieved remission, with 9 pts in complete remission 1 (CR1), 1 pts in CR2, and 3 pts in CR3 or higher. The pre-HSCT minimal residual disease (MRD) status, assessed by flow cytometry, was negative in 9 pts and positive in 4 pts. Three patients had extramedullary infiltration but not central nervous system (CNS) leukemia. Before the enrollment, 9 pts experienced their first relapse, with 5 relapsing within 12 months and 4 after 12 months, as well as 4 ptshad relapsed more than one time.

After one cycle of treatment, 4 pts achieved CR with negative MRD, 1 patient achieved CR but MRD remained positive, and 4 pts achieved CR with incomplete hematologic recovery (CRi). Both overall response rate (ORR) and composite (CRc) was 69.23% (95% CI 38.57-90.91%). The most common treatment regimen among patients who achieved CR/CRi was a MAC-based regimen (Lipo-MIT, cytarabine and cyclophosphamide, with or without targeted drugs), used in 6 out of 9 pts.

The median follow-up period was 6.41 months (range 2.10-16.43). Disease progression occurred in 4 out of 13 pts. Seven patients died after treatment, however, no correlation with treatment was obeserved. The median progression-free survival (PFS) was 3.06 months (range 0.62-7.62), while the median overall survival (OS) was 9.49 months (range 2.10-16.43). The hematological grade 3/4 TRAEs were anemia (53.85%), thrombocytopenia (46.15%), leukopenia (38.46%), and neutropenia (23.08%). The most common non-hematological grade 3/4 TRAEs was infections (76.92%), but no fetal infections were occoured.

Conclusions: Lipo-MIT containing therapy, particularly the MAC-based regimen, appears to be an effective and tolerable treatment for relapsed AML after allo-HSCT. It is worthwhile to further explore safety and more effective combination therapy of Lipo-MIT.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P055 STUDY ON THE EFFICACY AND SAFETY OF BLINATUMOMAB MAINTENANCE THERAPY OF HIGH RISK PH NEGATIVE ALL AFTER ALLO-HSCT

Zhang Jianping1, Li Nannan1

1Hebei Yanda Ludaopei Hospital, Langfang, China

Background: Immunotherapy provides more opportunities for Allo-HSCT after complete remission for refractory/recurrent B-ALL. However, it is still difficult to achieve long-term survival after Allo-HSCT recurrence, especially recurrence in the early stage. Therefore, how to reduce the recurrence of high-risk PH-negative B-ALL through prevention is still an urgent problem to be solved.

Methods: Group entry criteria:

  1. 1.

    With high-risk cytogenetic and molecular biological changes in accordance with the definition of ELN.

  2. 2.

    Receiving an immunotherapy before transplantation, there is still MRD positive or MRD returning to positive in a short time after turning negative.

  3. 3.

    Extramedullary leukemia occurred before transplantation.

  4. 4.

    MRD 10-6 to 10-4 after transplantation. Treatment: two months after Allo-HSCT, if there’s no active GVHD or uncontrolled infection, we’ll give two weeks Blinatumomab continuous injection. If there’s no contraindication after 45 days we’ll give another cycle of Blinatumoma. During the course we observe the infusion reaction, hepatorenal toxicity, hematotoxicity, GVHD and other complications.

Results: There are ten patients accepted Blinatumomab after Allo-HSCT. 3 patients got fever, 1patient got hypertension. 1patient got TA-TMA. 1patient developed chromosome clone from the donor, which returned to normal after three months. 2patients developed limited cGVHD. All other patient survive disease-free.

Conclusions: It is safe and feasible to Blinatumomab maintenance therapy of high risk PH negative ALL after Allo-HSCT

Disclosure: no

19: Acute Leukaemia

P056 THE IMPACT OF PRE-TRANSPLANT MDS-ASSOCIATED SOMATIC MUTATIONS AND CO-MUTATIONS IN PATIENTS WITH AML ON POST-TRANSPLANT OUTCOMES

Khalid Halahleh 1, Ayat Taqash1, Isra Muradi2, Farah Alul1, Osama Alsmadi1, Mohamad Ma’koseh1

1King Hussein Cancer Center, Amman, Jordan, 2Ahliyya Amman University, Al-Salt, Jordan

Background: Acute myeloid leukemia (AML) with myelodysplasia-associated gene mutations (MDS + ) is a new disease entity introduced in the International Consensus Classification of Myeloid Neoplasms 2022(ICC 2022). MDS-associated somatic mutations (MDS +) include SRSF2, SF3B1, U2AF1, ZRSR2, ASXL1, EZH2, BCOR, and STAG2. The aim of this study is to assess the frequency of this disease entity of AML, and evaluate the impact of pre-transplant MDS-associated somatic mutations on post-transplant outcomes.

Methods: Using King Hussein Cancer Center Registry, out of 176 newly diagnosed AML patients, 162 had available genomic abnormalities using targeted next-generation sequencing gene panel at diagnosis between “2016 – 2023”. 117 patients achieved complete remission (CR) and included in this analysis. Leukemia free survival (LFS) and overall survival (OS) were calculated using Kaplan Meier methods using a log-rank test and a Cox proportional hazard model was used for unadjusted analyses of time-to-event endpoints. Cumulative incidence of relapse (CIR) and nonrelapse mortality (NRM) were calculated using the Gray method. Factors with P < 0.05 in univariate analyses were included in multivariate analyses. P < 0.05 was considered statistically significant.

Results: 100 patients (85.5%) had de novo AML. Median age was 46y (18-78y). 82 patients (70%) were males. Pathogenetic mutations were detected in 104 (89%), -VUS in eight (6.7%) and 5 patients (4.3%) had negative on targeted gene panel. We identified 23 patients (20%) with at least one of MDS-associated mutations (MDS + ) and 94(80%) were negative (MDS-). 72 patients (69%) were in CR1, 54 (46%) received allo-HSCT (17 in CR1). Patients-disease-transplant-related covariate were comparable between the two groups, except patients age was younger (P-0.001) and more secondary AML in MDS+ group (P-0.016).

The median follow-up for survivors (n-45) was 16 months (0.5-52 months). 3-year CIR, LFS and OS were 39% (95% CI: 23%-55%), 61% (95% CI: 49%-76%), and 26% (95% CI: 16%-37%) respectively. The MDS+ group had less relapses (3-year CIR, 5% vs 40%) (Hazard ratio [HR]:1.327, 95% confidence interval [CI]: 0.758-2.323; P-0.79), comparable LFS (75.5% vs 60%) (HR: 0.853, 95% CI: 0.249-2.928; P-0.8), and OS of 24% vs 26%) (HR: 1.327, 95%CI: 0.758-2.323; P-0.322). In univariable cox regression analysis for LFS and OS, and NRM, none of the factors analyzed including gender, age (< 45y vs ≥ 45y), ELN 2022 AML classification, and MDS+ vs MDS- groups, was statistically significant for OS or NRM (all P>0.05). Younger age was statistically significant for LFS (P-0.018). Among the 23 MDS+ patients who achieved CR, and received allogeneic HSCT (n -7) showed better OS (3-year OS, 86% (53% vs Zero) (HR: 0.061, 95% CI: 0.008-0.476, P-0.0076), a trend toward improvement in LFS (3-year,100% vs zero%) (HR: NA, 95% CI: NA vs NA, P-0.9972) than those who received consolidation chemotherapy considering allo HSCT as non-time dependent covariate. No patients relapsed in the allogeneic HSCT patients (3-year, zero vs 49%) (95% CI: 8%- 82%; P-0.063), and NRM was comparable between the two groups (P-0.998).

Conclusions: MDS-associated somatic gene mutations are an independent prognostic factor for adverse outcomes in AML that might be overcome by allotransplant.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P057 TRIALS IN PROGRESS: A PHASE 1B SINGLE-ARM, OPEN-LABEL STUDY OF EMAVUSERTIB IN COMBINATION WITH AZACITIDINE AND VENETOCLAX IN AML PATIENTS IN COMPLETE RESPONSE WITH MRd

Adolfo de la Fuente 1, Claudio Cerchione2, Sebastian Scholl3, Jan Moritz Middeke4, Gaurav S. Choudhary5, Dora Ferrari5, Reinhard von Roemeling5, Uwe Platzbecker6

1MD Anderson Cancer Center, Madrid, Spain, 2Istituto Romagnolo per lo Studio dei Tumori, Meldola, Italy, 3Universitätsklinikum Jena, Jena, Germany, 4Technische Universität Dresden, Dresden, Germany, 5Curis Inc, Lexington, United States, 6University Hospital Leipzig, Leipzig, Germany

Background: Emavusertib is a novel potent oral inhibitor of interleukin-1 receptor-associated kinase 4 (IRAK4) with additional inhibitory activity against FMS-like tyrosine kinase 3 (FLT3) and CDC-like kinases (CLK1/2/4). Inhibition of these onco-proteins may induce remission thereby addressing a critical unmet need for novel therapies in acute myeloid leukemia (AML). Clinical studies with emavusertib monotherapy have demonstrated a significant reduction in AML blasts with clinical and molecular responses, including patients with relapsed or refractory AML, previously treated with an HMA and/or FLT3 inhibitors (Metzeler 2022).

Azacitidine + venetoclax (aza+ven) has been approved in newly diagnosed, unfit patients with AML. In the VIALE-A study, composite complete response (CRc) (CR, CRh, or CRi) in the absence of measurable residual disease (MRD) of <1 residual blast/1000 leukocytes (MRD negative [MRD−]) resulted in longer duration of response (DOR), event-free survival, and overall survival (OS), and better HSCT outcome compared with patients who achieved CRc but were MRD+ (Pratz, 2022). In pre-clinical studies, emavusertib in combination with aza+ven demonstrated synergistic antileukemic effects in AML cell lines, including azacitidine- or venetoclax-resistant cell lines. Adding emavusertib to aza+ven in MRD+ patients at the time of CR may convert MRD status without adding significant toxicity and confirm that emavusertib can be safely added to aza+ven as a potential new regimen in front-line therapy.

Methods: This is a single-arm, open-label Phase 1b trial evaluating safety and tolerability, PK, and conversion of MRD status with emavusertib as an add-on agent to aza+ven in AML patients who achieved CR or CRh with MRD+ based on local testing (EU CT Number 2023-505828-58-00). The primary objective of the study is to determine a safe and tolerable dosing schedule for the triple combination. Secondary objectives include MRD conversion rate, DOR, OS, and pharmacokinetics. The study will enroll approximately 24 patients at 5 to 10 sites globally. Patients will have received azacitidine and venetoclax as first line therapy and achieved CR or CRh after 1-6 cycles of aza/ven. If MRD status remains positive, emavusertib will be added to the existing well tolerated aza+ven regimen. The starting emavusertib dose is 200 mg BID for 7 days per cycle of 28 days. If well tolerated, duration of emavusertib treatment will be extended to 14 and 21, respectively; no intra-patient change of emavusertib dosing duration is planned. The patients will continue triple treatment (emavusertib+aza+ven) until consent withdrawal, disease progression, intolerable toxicity, or not achieving MRD- within 6 cycles of triple treatment. In this Phase 1b trial, MRD can be evaluated by local testing of bone marrow. Key exclusion criteria include residual toxicities and significant comorbidities.

Results: Not applicable.

Conclusions: Not applicable.

Clinical Trial Registry: EU CT Number 2023-505828-58-00

Disclosure: Nothing to Declare.

19: Acute Leukaemia

P058 PREVALENCE OF ELIGIBILITY FOR GERMLINE MUTATION TESTING IN PATIENTS WITH MDS/AML UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT IN A US ACADEMIC MEDICAL CENTER

Tyler Fugere 1, Samuil Ivanovsky1, Gomathy Nageswaran1, Brad Fugere1, Urooj Hudda1, Sravani Gundarlapalli1, Lakshmi Yarlagadda1, Zhongning “Jim” Chen1, Cesar Gentille1

1University of Arkansas for Medical Sciences, Little Rock, United States

Background: Predisposition to acute myeloid leukemia (AML) is increasingly recognized and referral for genetic counseling, germline testing, and extension of these services to family members should be considered in select patients. The National Comprehensive Cancer Network (NCCN) and European LeukemiaNet (ELN) have outlined suggestive disease features to prompt germline testing. Recognition of these familial syndromes is important to support clinical decision-making and assess the risk for hematological malignancies for family members. Given their rarity, it is unclear how many patients are actually screened. Our goal was to assess the prevalence of patients that met criteria for testing and that were referred for further evaluation in an academic medical center in the US.

Methods: We identified 132 patients who received a hematopoietic cell transplant for myelodysplastic syndrome (MDS) or AML at University of Arkansas Medical Sciences from July 1, 2018 to October 5, 2023. We determined the prevalence of eligibility for testing by considering patients who were either <40 years old at diagnosis or had a mutation with VAF >30% and had either a personal or family history of cancer as being eligible based on NCCN guidelines. We collected diagnosis date, transplant date, relapse date, and date of last contact or death to determine progression free survival (PFS) and overall survival (OS) per Kaplan-Meier. Age, disease type, conditioning regimen and eligibility for testing were analyzed using Cox proportional hazard regression.

Results: Overall, 102 patients had AML and 30 had MDS; 36 received a myeloablative and 96 a reduced intensity transplant. Thirty-six patients (27.3%) met criteria for germline testing but only 4 were tested. Of patients who met criteria for testing, median age was 55 years. NPM1 was the most prevalent mutation (10.3%) followed by DNMT3A (8.9%), FLT3 (7.6%), RUNX1 (6.3%) and TP53 (6.3%); 24 of these patients had at least 1 mutation that has been reported to be potentially germline. Outcomes for PFS and OS were not different between patients eligible for testing and ones who were not per Kaplan-Meier or multivariate analysis; median PFS and OS were not reached for either group.

Conclusions: Prior studies have identified a prevalence of germline mutations in AML of 7-14%, however, the prevalence of patients meeting eligibility for testing or getting tested in routine clinical practice has not been described. In our cohort, more than 20% of transplanted patients with MDS/AML met criteria and less than 5% were tested despite most of them having at least 1 potential germline mutation. Even though there was no apparent effect on outcomes, our sample size and lack of germline confirmatory testing makes interpretation of these results challenging. We believe awareness of the features suggestive of a germline disease is paramount and needs to increase to capture as many patients as appropriate as it could affect conditioning and donor options for transplant as well as help prevention or early detection of other hematologic or solid cancers.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P059 REFINED DISEASE RISK INDEX APPLIED IN AN UPPER-MIDDLE INCOME COUNTRY AS A TOOL TO ESTIMATE OVERALL SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN HAEMATOLOGICAL MALIGNANCIES

Shirley Quintana-Truyenque1, Naty López-Córdova 1, Jessica Meza-Liviapoma1, Lourdes López-Chavez1, Cindy Alcarraz-Molina1, Victor Mallma-Soto1, Claudio Flores-Flores2, Jule Vasquez-Chavez1

1Instituto Nacional de Enfermedades Neoplásicas (INEN), Lima, Peru, 2Universidad Peruana San Juan Bautista, Lima, Peru

Background: Allogeneic haematopoietic stem cell transplantation (Allo-HSCT) is a potentially curative option in a greater number of haematological malignancies. There are some prognostic models developed to estimate risks and survival in patients who will have this treatment. The Refined Disease Risk Index (R-DRI) stratifies neoplasms based on the status of the disease and remission before transplantation, there are 4 risk groups: low (LR), intermediate (IR), high (HR) and very high (V-HR), which correlate with overall survival (OS) at 2 years and have demonstrated to not been affected by age, conditioning intensity, graft source or donor type. The aim of the study was to estimate the overall survival post Allo-HSCT in our institution in relation to the R-DRI with the objective of implement its use in the Peruvian patients.

Methods: We reviewed retrospectively the data of 172 adults who underwent an Allo-HSCT between 2012 and 2023 in the Bone Marrow Transplantation (BMT) unit of the National Institute of Neoplastic Diseases (INEN). The information was extracted from the unit registry and was completed using the electronic medical record. Patients over 15 years of age were included. All donors were related, and the source used was peripheral blood. Risk classification was performed according to the R-DRI. Survival was estimated using the Kaplan-Meier method and comparisons were made using the Log-Rank test.

Results: The median age at HSCT was 26 years (range: 15 - 63), 51% of patients were male. Most frequent diagnoses were acute lymphoblastic leukaemia (ALL), Acute Myeloid Leukaemia (AML), Mixed-Phenotype Acute Leukaemia (MPAL), and Chronic myeloid leukaemia (CML), with 85, 41, 13 and 11 cases, respectively. Other diagnoses were (Myelodysplastic syndrome (MDS), Acute lymphoblastic lymphoma [LBL], Hodgkin lymphoma, and aggressive T/NK cell leukaemia with 8, 6, 1 and 1 cases, respectively. According to the R-DRI classification (does not include MPAL and LBL), patients were stratified as LR, IR, HR, and V-HR in 5%, 38%, 35% and 22% of cases, respectively. The risk that predominated in each diagnostic group was as follows: for ALL, AML, CML, and MDS were IR (46%), HR (51.2%), LR (45.5%) and HR (75%), respectively. The median follow-up was 3.9 years. The median OS for the entire cohort was not reached, and the survival rate at 2 and 4 years according to R-DRI for LR, IR, HR, V-HR were 100%, 68.3% and 57.4%, 64.7 and 53.5%, 47.4 and 38.3%, respectively. The 4y-OS rate of the combined IR and HR group vs V-HR was 54.6% and 38.3%, respectively (p = 0.071).

Conclusions: OS in our patients according to the R-DRI was higher than the reported in the literature, with a greater difference between the intermediate and high versus very high-risk groups. The R-DRI is an important tool that allows to estimate the prognosis after allogeneic transplant in our population; however, it must be taken into consideration that this study is retrospective and there are patients who did not reach the median follow-up, so it is recommended to continue with prospective studies.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P060 PREEMPTIVE DONOR LYMPHOCYTE INFUSION AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN PATIENTS WITH ACUTE MYELOID LEUKEMIA – SINGLE CENTRE ANALYSIS OF EFFECTIVENESS AND SAFETY

Anna Łojko Dankowska 1, Ewa Bembnista1, Paula Matuszak1, Magdalena Matuszak1, Bartosz Małecki1, Andrzej Szczepaniak1, Anna Wache1, Dominik Dytfeld1, Lidia Gil1

1Poznan University of Medical Sciences, Poznań, Poland

Background: Acute myeloid leukemia (AML) is a disease with intermediate sensitivity to donor lymphocyte, therefore effectiveness in patients (pts) with relapse of disease after allogeneic stem cell transplantation (alloHCT) is limited. However, donor lymphocyte infusion (DLI) can be used earlier to prevent relapse as a prophylaxis in pts with complete remission or as a preemptive treatment in patients with mixed chimerism or with positive minimal residual disease (MRD + ) in flow cytometry.

Methods: We retrospectively analyzed records of twenty adult patients with AML treated in our center with alloHCT from HLA-identical sibling (n = 5) or matched unrelated (n = 6) or haploidentical (n = 9) donor, conditioned with myeloablative (n = 12) or reduce intensity conditioning, who received DLI between 2018 and 2023 as a preemptive treatment due to decrease chimerism (n = 8) or MRD+ (n = 12). Median age at the transplantation was 44 years (range 23-65), median time between transplantation and first DLI was 9 months (range 4-50). Three pts were after second alloHSCT. At the time of first infusion all pts were without immunosuppression and without any symptoms of graft versus host disease (GvHD).

Results: The median follow up among survivors since first DLI was 22 months (range 1-58). Median number of DLI received per patient was 4 (range 1-8), the median CD3+ lymphocyte count per one infusion was 2,3x10e6 per kilogram of body weight (/kg) (range 0,1-20). The cumulative lymphocytes count per one patient was 7,64x10e6 CD3+ cells/kg, range 0,1-54,88. The minimum time between two infusions was 4, the maximum 30 and the median 7 weeks. DLI was effective (increase chimerism above 95% or negative MRD) in 11 pts (55%), 9 (45%) of them remained alive in remission of disease to date. One patient died due to secondary malignancy and one due to infection. Relapse of disease occurred in 8 pts and 7 of them died due to relapse. Acute GvHD were observed in only 3 pts (G2 = 2, G4 = 1), all these pts responded to DLI, but one died due to infection complications. Median overall survival (OS) was 24 months for whole group, 2 years OS for responders was 80%. 12 pts (60%) currently alive.

Among pts who responded to DLI the median cumulative lymphocyte count per one patient was 16,21x10e6 CD3+ cells/kg, (range 1,27-53,54). Among pts who did not respond to DLI the median cumulative lymphocyte count per one patient was 4,09x10e6 CD3+ cells/kg (range 0,1-54,88). Pts with GvHD received the median cumulative lymphocyte count 51,19x10e6 CD3+ cells/ kg (range 16,21-53,54). The difference was not statistically significant p = 0,57.

Conclusions: The relapse of AML after alloHCT remains the major reason for failure and is connected with poor prognosis. DLI can be effective tool to prevent relapse, but the optimal application, timing and dosage, has not yet been established. Our results confirm that this method is safe and associated with low risk of death. The effectiveness of DLI in preventing relapse could be increased in AML pts by combining with other drugs, but perhaps also by increasing the lymphocyte dose.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P061 IN CHILDREN WITH RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA GIVEN ALLOGENEIC-HSCT EMPLOYING REDUCED-INTENSITY CONDITIONING, GVHD IS LESS FREQUENT IF THE PROCEDURE IS CONDUCTED ON AN OUTPATIENT BASIS

Edgar Jared Hernández-Flores1, Moisés Manuel Gallardo-Pérez2, Max Robles-Nasta2, Daniela Sánchez-Bonilla2, Merittzel Abigail Montes-Robles3, Guillermo Ocaña-Ramm1, Olivia Lira-Lara4, Juan Carlos Olivares-Gazca2, David Gómez-Almaguer5, Óscar González Llano5, Yajaira Valentine Jiménez-Antolinez5, Guillermo José Ruiz-Delgado 2,1, Guillermo José Ruiz-Arguelles2,1

1Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico, 2Centro de Hematología y Medicina Interna, Clínica Ruiz, Puebla, Mexico, 3Universidad Anáhuac Puebla, Puebla, Mexico, 4Universidad Veracruzana, Veracruz, Mexico, 5Servicio de Hematología del Hospital Universitario “Dr. José Eleuterio González”, Monterrey, Mexico

Background: Advance in transplant technology (donor matching, alternative donor source, conditioning regimens), and supportive care have significantly improved the post-hematopoietic stem cell transplantation (HSCT) survival. Allogeneic HSCT is employed in the treatment of relapsed/refractory acute lymphoblastic leukemia (RR-ALL) in children. Outpatient conduction of reduced intensity allografting is being employed more frequently.

Methods: Compare the long term results of allografting children with B cell RR-ALL employing reduced intensity conditioning, the procedure being conducted either as inpatients or outpatients.

Results: 37 patients were analyzed. 18 were allografted as inpatients (IP) and 19 as outpatients (OP). In the statistical analysis, the two comparative groups have no significant differences in terms of age, sex, platelets recovery and acute GVHD. The table shows the salient features of these two groups. The median follow up for outpatient group is 35 months (IQR: 9-59) meanwhile the median follow up for inpatient is 20 months (IQR: 11-33). The median OS for the OP was 33 months, whereas for IP was 50 months (p = 0.342). Time to recover >500 neutrophils in was shorter in OP (12 versus 14 days), whereas the prevalence of chronic GVHD was significantly lower in OP (10% versus 61%).

Table 1. Salient features of 37 patients given allo-HSCT employing reduced intensity conditioning as inpatient or outpatient. a= p value obtained by chi square. b= p value obtained by Mann-Whitney´s U. c = p value obtained by student’s T-test. d= p value obtained by log-rank chi square.

Outpatient (n = 19)

Inpatient (n = 18)

p

Female

10

6

0.130 a

Male

7

12

Mean Age

8.1 + 4.1

7.8 + 4.2

0.840 c

Relapsed/ refractory

10/9

16/2

Days to >500neutrophils

12 (IQR = 3)

14 (IQR = 4.25)

0.022 b

Days to >20,000 platelets

13 (IQR = 1)

14(IQR = 2.25)

0.090 b

Acute GVHD, II-IV

8

7

0.842 a

Chronic GVHD

2

11

0.001 a

Median OS

33

50

0.342 d

Median follow up (months)

35 (IQR = 50)

20 (IQR = 22)

0.620 b

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (8)

Conclusions: These results suggest that allo-HSCT employing a reduced-intensity conditioning can be conducted safely as outpatients in children with RR-ALL could indicate. Moreover, the prevalence of chronic GVHD is lower in patients allografted as outpatients outside that there is no difference in children survival given outcome or income Allo-HSCT.

Clinical Trial Registry: NCT05780554

Disclosure: Nothing to declare.

19: Acute Leukaemia

P062 IMPLEMENTATION OF NON-TBI CONDITIONING AND POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR PEDIATRIC ACUTE LYMPHOBLASTIC LEUKEMIA IN RESOURCE-LIMITED SETTINGS

Lusine Krmoyan 1,2, Inga Khalatiani1,2, Mane Gizhlaryan1, Karen Meliksetyan1

1YEOLYAN Hematology and Oncology Center, Yerevan, Armenia, 2Yerevan State Medical University, Yerevan, Armenia

Background: Resource limitations in countries such as Armenia often impede access to advanced medical interventions, including haploidentical hematopoietic stem cell transplantation (HSCT). Consequently, patients with acute leukemia requiring allogeneic HSCT encounter significant challenges in seeking treatment abroad. This study documents innovative strategies employed in a resource-limited setting to facilitate HSCT for three pediatric patients without the option of international treatment.

Methods: A comprehensive preparatory protocol was established for two patients necessitating haploidentical HSCT due to the unavailability of high-resolution typing, chimerism assessment, donor-specific antibodies (DSA) testing, total body irradiation (TBI), and T cell depletion. The protocol included pre-treatment with rituximab to reduce DSA, followed by a conditioning regimen of fludarabine, treosulfan, and thiotepa. Cyclosporine A (CsA) from day -2 and mycophenolate mofetil (MMF) from day -1 were supplemented for acute graft-versus-host disease (aGVHD) prophylaxis, and post-transplant cyclophosphamide (PTCy) was administered on days +3 and +4 for in vivo T cell depletion. Peripheral blood and bone marrow were used as stem cell sources for one patient each. In a separate case involving allogeneic HSCT from a fully matched sibling donor, a similar preparatory protocol was implemented, with bone marrow as the stem cell source. Fludarabine, treosulfan, and thiotepa constituted the conditioning regimen, supplemented by CsA from day -2 and methotrexate on days +1, +3, and +6 to prevent graft-versus-host disease (GVHD).

Results: Despite resource constraints and the absence of advanced testing and treatment options, the patients exhibited promising outcomes. Post-transplantation assessments on days +30, +60, and +90 revealed chimerism levels more than 99% and negative minimal residual disease (MRD). Noteworthy complications, including severe gastrointestinal tract acute GVHD (grade II-III), HHV6 reactivation, and severe CMV reactivation, were encountered and managed in one patient.

Conclusions: This study demonstrates the successful implementation of non-TBI-based conditioning regimens and emphasizes the role of PTCy in achieving favorable outcomes in a resource-limited setting. PTCy emerges as a crucial strategy in reducing GVHD risks and facilitating HSCT despite limited technological resources, offering hope for patients facing similar constraints worldwide.

Disclosure: Nothing to disclouse.

19: Acute Leukaemia

P063 RELAPSE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENT WITH ACUTE MYELOBLASTIC LEUKEMIA

Insaf Ben Yaiche1, Nour Ben Abdejelil 1, Rihab Ouerghi1, Ines Turki1, Sabrine Mekni1, Lamia Torjemane1, Dorra Belloumi1, Rimmel Kanoun1, Saloua Ladab1, Tarek Ben Othman1

1Centre National de Greffe de Moelle Osseuse, Tunis, Tunisia

Background: Allogeneic hematopoietic stem-cell transplantation (AHSCT) is the only curative treatment for high-risk acute myeloid leukemia (AML). However, the relapse remains a substantial risk of after AHSCT

Methods: A retrospective descriptive study, included all consecutive patients with AML, who underwent AHSCT from HLA-matched sibling donor, between January 2018 and December 2022. Conditioning consisted on busulfan-iv and cyclophosphamide (BU/CY) or fludarabine-based. GVHD prophylaxis included cyclosporine and short course of methotrexate +/- antithymocyte-Globulin. Stem cell source was bone marrow or peripheral blood stem cells (PBSC). The study aimed to estimate cumulative incidence of relapse (CIR) after AHSCT and describe management therapy of relapsed patients.

Results: Sixty-three patients (9 children, 54 adults) were included with a median age of 33 years (6-49y). The sex-ratio was 1.86. At diagnosis, 21% patients were classified as low-risk (n = 13), 49% intermediate risk (n = 31), 24% high-risk (n = 15), unclassified risk in 6% (n = 4: missing data).

The median time from diagnosis to AHSCT was 7 months (4-54 months). At transplant, 84% of patients were in first complete remission (CR1). Conditioning regimen was BU/CY in 83% of patients. Stem cell source was PBSC in 59% of patients.

After a median follow-up of 32 months (2-67 months), the CIR was 27% (early n = 6, late n = 9). Median time of relapse was 15 months (3-20 months). Relapses were medullary and combined (n = 11), extramedullary (n = 2), molecular (PML-RARA, n = 1) and Cytogenetic (11q23 n = 1).

Among the 15 relapsed patients, 11 (73%) received salvage therapy (chemotherapy n = 8, DLI +/- azacitidine n = 2, ATRA n = 1). Second CR was observed in 6 patients, among them, 5 underwent a second AHSCT. At last follow-up, 45 patients are alive, 18 patients (28%) died (relapse n = 10, NRM n = 8). CI of NRM was 13%.

The 3-year overall survival and event-free survival were 31% vs 83% (p = 0.001) and 7% vs 82% (p<10-3), in relapsed and non-relapsed patients, respectively.

Conclusions: The prognosis of patients with AML relapsing after AHSCT is poor. Adapted risk stratification treatment at diagnosis, before and after AHSCT may improve outcomes.

Disclosure: Nothing to declare.

19: Acute Leukaemia

P064 IAMP21 ACUTE LYMPHOBLASTIC LEUKEMIA WITH DELETION AROUND SUBTELOMERIC REGION OF CHROMOSOME 21: REPORT OF ONE CASE AND REVIEW OF LITERATURE

Jiantuo Liu 1, Hongrui Li1, Xiangjun Chen2, Yanli He2

1Cytogenetics Laboratory, Wuhan Kindstar Medical Laboratory Co., Ltd, Wuhan, China, 2Union Hospital Affiliated to Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China

Background: Herein, our report describes the case of a 4-year-old man who presented initially with fever and bilateral instep pain in july 2023.

Methods: we performed a retrospective study to analyze the clinical characteristics, laboratory examination, and treatment.

Results: Physical examination were as follows, T 37.8°C, B 23/min, HR 101/min, BP 88/60mmHg, he was mentally ill and pale. One lymph node with a diameter of 1.5cm is palpable in the right neck. One or two lymph nodes with a diameter of 0.5cm is palpable in the bilateral groin, with soft texture, easy movement and no tenderness. Superficial lymph nodes in the left neck and armpit were not touched. Bilateral tonsil degree I, no exudate attachment. Normal cardiopulmonary function, no liver and spleen enlargement, limbs without muscle tension, extremity warm. Blood routine examination showed that, white blood cell count (WBC) 2.62×109/L, red blood cell count (RBC) 3.25×1012/L, hemoglobin (Hb) 82g/L, neutrophil absolute value 0.48×109/L, lymphocyte absolute value 1.82×109/L, monocyte absolute value 0.14×109 /L. platelet count 165×109/L, C-reactive protein 9.16mg/L, primitive naive cells 8%. The bone marrow examination showed 68.4% of naive lymphocytes, and the immunotype was consistent with B-ALL diagnosis. The RNA-seq showed normal result in tumor cells. RT-PCR test were negative by screened 43 fusion genes of leukemia. The patient showed a normal karyotype. Interphase FISH were as follows, MLL(-), BCR/ABL(-), PBX1/TCF3(-), MEF2D(-), the ETV6/RUNX1 probe observed the cluster of green signals indicates the amplification of RUNX1. The Tel21q probe captured one green signal indicates the deletion around subtelomeric region of chromosome 21. Above these characteristics defined iAMP21. According to the Chinese Children’s Cancer Group study ALL 2020(CCCG-ALL-2020), the patient was divided into intermediate risk group. Induction regimens were typically based on a backbone that included a combination of vincristine, prednisone and pegaspargase with daunorubicin, the patient remained MRD + (0.06%,>0.01%)after induction therapy(day46). High-dose methotrexate (HD-MTX), 6-mercaptopurine (6-MP) were incorporated into consolidation regimens, the patient achieved MRD negative(<0.01%) over a duration of 3 months. Maintenance regimens were based on a backbone of daily 6-MP and weekly methotrexate with the addition of periodic vincristine and prednisone. To date, the patient remained MRD negative after a total of five months of treatment.

Conclusions: Intrachromosomal amplification of chromosome 21 (iAMP21) defines a distinct cytogenetic subgroup of B-cell acute lymphoblastic leukaemia (B-ALL). It has since been estimated that B-ALL with iAMP21 accounts for 2% of all pediatric B-ALLs. In this study, we have validated the existence of iAMP21 with deletion around subtelomeric region of chromosome 21. iAMP21 is associated with inferior outcome, who require more intensive therapy.

Disclosure: Nothing to declare.

20: Aplastic Anaemia

P065 RESULTS OF HSCT WITH TCRΑΒ AND CD19-DEPLETION FROM MATCHED RELATED DONORS AND INFUSIONS OF CD45RA DEPLETED DONOR LYMPHOCYTES IN PEDIATRIC SEVERE APLASTIC ANEMIA

Daria Shasheleva1, Larisa Shelikhova1, Anna Bogoyavlenskaya1, Rimma Khismatullina1, Svetlana Radygina1, Dmitriy Balashov1, Yakov Muzalevsky1, Tatiana Salimova1, Dina Baidildina1, Elena Kurnikova1, Dmitriy Pershin 1, Pavel Trakhtman1, Galina Novichkova1, Alexei Maschan1, Michael Maschan1

1Dmitry Rogachev Federal Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation

Background: HSCT from matched family donors results in most favorable outcomes among children with severe aplastic anemia (SAA). Despite overall success, morbidity, associated with acute and chronic graft-versus-host disease (GVHD) is not completely prevented with current standard of pharmacologic prophylaxis. Depletion of ab T cells from the graft prevents GVHD, and improves outcome of hematopoietic stem cell transplantation from haploidentical donors, while infusions of donor memory lymphocytes (mDLI) (CD45RA-depleted) are able to transfer pathogen-specific immunity without the risk of GVHD. We evaluated the outcomes of ab T cell depletion and add-back of intermediate-dose mDLI among the pediatric SAA recipients of matched related grafts.

Methods: A total of 24 children with SAA (11 female, 13 male, median age 9,8 y) underwent allogenic HSCT from matched family donors (MFD) between February 2015 and May 2023, for all pts it was the first allo HSCT. TCR αβ + /CD19+ depletion of HSCT with CliniMACS technology was implemented in all cases. The median dose of CD34+ cells was 7,1 x106/kg (range 2,76-13,2), αβ T cells - 23x103/kg (range 1-184). All pts received an additional injection of memory T-cell (CD45RA-depleted) on day 0 at 1 million T cells per kg.

All patients received cyclophosphamide at 100 mg/kg, fludarabine at 100 mg/m2, rituximab 100mg/m2 and serotherapy with either rabbit ATG at 5 mg/kg (n-1) or horse ATG at 100 mg/kg (n-23).

Post-transplant GVHD prophylaxis included calcineurin (CNI)-based regimen and abatacept 10mg/kg on days -1, +7, +14 and +28. 22 pts received a graft from a 10/10 HLA-matched sibling, 2 from 9/10. Median time of follow-up for survivors was 2,9 years (range: 0.8 – 9.8).

Results: Primary engraftment was achieved in all evaluable patients (100%) with donor chimerism, the median time to neutrophil and platelet recovery was 11 (6-18) and 13 (9-19) days, respectively. One patient had aGvHD grade II and there were no incidence of grade III-IV aGvHD and TRM. The CI of CMV viremia was 17% with no incidence of CMV disease. No cases of ADV and EBV viremia and EBV disease were recorded. Two pts had thrombotic microangiopathies (TMA) with good response to rituximab, one pt. had partial red cell aplasia. The median recovery of T cells was on day+60 was 0,26 (0,04-0,9). Event-free and overall survival were 100%.

Conclusions: ab T cell-depleted transplantation with intermediate dose memory T cell add-back definitively prevents GvHD and provides a platform for safe HSCT from matched family donors in patients with SAA.

Disclosure: M. Maschan - Miltenyi Biotec: Honoraria

D. Balashov - Octafarm: Other: Lecture fee

20: Aplastic Anaemia

P066 MITOCHONDRIAL METABOLISM IN FANCONI ANEMIA PATIENTS POST HEMATOPOIETIC STEM CELL TRANSPLANTATION

Filomena Pierri1, Enrico Cappelli1, Stefano Regis1, Silvia Ravera2, Federica Grilli1, Gianluca Dell’Orso1, Stefano Giardino1, Maura Faraci 1

1IRCCS Istituto Giannina Gaslini, Genova, Italy, 2University of Genoa, Genova, Italy

Background: Fanconi anaemia (FA) is a rare genetic condition characterized by congenital abnormalities, progressive bone marrow failure (BMF), chromosome fragility, and cancer susceptibility. In addition to a DNA repair defect, FA patients present a pro-inflammatory condition with increased cytokine production and sensitivity and defective mitochondrial metabolism associated with a poor cellular response to oxidative stress. These defects are involved in the pathogenesis of BMF, concerning not only hematopoietic stem cells (HSCs) but also bone marrow (BM) derived mesenchymal stromal cells (MSCs), which are responsible for organizing a functional HSC niche in vivo and secrete cytokines and matrix proteins that function in HSC self-maintenance and marrow retention.

Hematopoietic Stem Cell Transplantation (HSCT) is the only curative treatment option for FA haematological manifestations. However, this procedure does not result in gene correction of MSCs and other somatic cells, which lead to a persistence of some FA features in the BM microenvironment of transplanted patients that are not fully explored so far.

In this study, we analysed the peripheral blood (PB) mononuclear cells (MNCs) energy metabolism of transplanted FA patients, comparing to their healthy donors and to patients affected by idiopathic severe aplastic anaemia (SAA) to investigate the persistence of some FA features in BM microenvironment post-HSCT.

Methods: We studied, before HSCT and at different time points after HSCT, MNCs metabolism from 4 FA patients undergoing haploidentical α/β/CD19 depleted HSCT (one for BMF and 2 for cytogenetic abnormalities without myelodysplasia), and 4 patients with SAA, 2 transplanted from a Matched Unrelated Donor (MUD), 1 from a Matched Related Donor (MRD), 1 from a haploidentical donor (Post Transplant Cyclophosphamide platform). We also collected samples of the 4 healthy donors of FA patients at the same time points of the recipients.

Oximetric, spectrophotometric, and enzymatic analyses have been used to analyse mitochondria metabolism and antioxidant response in pre and post HSCT peripheral blood (PB) in FA patients compared to PB from their healthy donor and SAA patients.

Results: MNCs from transplanted FA patients showed impaired mitochondrial function and reduced ATP synthesis compared to healthy donors and transplanted SAA. However, the associated oxidative stress was counteracted by antioxidant defences, although a slight increase in lipid peroxidation (MDA) was observed. Interestingly, treatment of MNCs with cytokine inhibitors corrects the mitochondria impairment, suggesting the persistence of a pro-inflammatory state in the bone marrow niche that can alter the mitochondrial metabolism.

Conclusions: In transplanted FA patients, the molecular signals secreted in the microenvironment remain, at least in part, those before transplantation, as the niche in the bone marrow consists of the patient’s somatic cells. This condition justifies the persistence of a pro-inflammatory state even after transplantation, which could lead to mitochondrial dysfunction not observed in healthy donors and SAA.

Therefore, understanding the composition of the bone marrow microenvironment in FA patients after HSCT could lead to the development of new therapeutic approaches to improve the long-term outcomes of transplanted FA patients.

Disclosure: None conflict of interest

20: Aplastic Anaemia

P067 ATLG HAS BETTER SAFETY THAN RATG IN THE TREATMENT OF ADULT AA WITH ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Zhang Jianping 1, Lu Yue1, Zhao Yanli1, Xiong Min1, Liu Deyan1, Cao Xingyu1, Wei Zhijie1, Sun Ruijuan1, Zhou Jiarui1

1Hebei Yanda Ludaopei Hospital, Langfang, China

Background: rATG has stronger immunosuppressive effect than ATLG, therefore rATG is the main choice for AA to receive Allo-HSCT. ATLG is only used as an alternative choice for patients who have received ATG treatment. Some studies have shown that the application of ATLG in Allo-HSCT in patients with leukemia can achieve the same efficacy as rATG, and has better safety. Our study is to observe whether ATLG can achieve the same curative effect as rATG and has better safety when AA patients receive Allo-HSCT.

Methods: In this study, a retrospective analysis was made of adult AA patients who received Allo-HSCT in our hospital from March 1, 2012 to June 1,2023. through the analysis of OS, GFFS, GVHD, CMV/EBV activation we explore whether ATLG can achieve the same curative effect as rATG and has better safety.

Results: A total of 218 eligible patients, male 110cases, female 108cases, median age 29y(18-58). VSAA 41cases, SAA 79cases, AA-PNH 34cases, TD-NSAA 64cases. Conditioning with rATG149 cases, ATLG 58 cases. Donor: MSD 39cases, URD 76cases, HID 103cases; Stem cell source: PBSC 86cases, BMSC + PBSC 132 cases. Graft count: MNC8.3 (2.99-18.52)×108/kg, CD34 + 5.0 (1.14- 18.31)×106/kg, CD3 + 1.75 (0- 20.44)×108/kg. Of the 218 patients, there were 212 assessable by neutrophil engraftment, median time 13d(9-41). There were 200 assessable by platelet engraftment, median time 13d(4-78). Median follow-up 34m(1-136), 5y OS 82.72%(0.76-0.88). 5y GFFS 68.41%(0.61-0.75). 5y OS: rATG 85.8%, ATLG76%, P = 0.390; 5y GFFS: rATG69.3%, ATLG64.1%, P = 0.178. Primary engraftment failure 2cases(0.0091%), secondary engraftment failure 1case(0.0045%). aGVHD (II-IV) in 100day rATG 22.15% (16.39%-29.93%), ATLG 27.59%(18.18%-41.86%), P = 0.39240. cGVHD in 5 years rATG31.54%(24.35%-40.86%), ATLG 33.87%(22.89%-50.10%), p = 0.66537. CMV recurrence rate in 180d:rATG65.44% (58.20%-73.57%), ATLG 46.95%(35.65%-61.82%); P = 0.00957. Incidence of EBV in 360days rATG33.56%(26.77%-42.06%); ATLG: 8.77%(3.80%-20.26%); P<0.001.

Conclusions: In AA patients who received Allo-HSCT, compared with rATG, ATLG had equivalent OS and GFFS, and equivalent aGVHD and cGVHD, and the incidence of CMV and EBV decreased significantly. ATLG is safe and reliable for GVHD prophylactic in patients with AA undergoing Allo-HSCT.

Disclosure: none

20: Aplastic Anaemia

P068 TREATMENT WITH DARATUMUMAB IN 4 PATIENTS WITH IMMUNE ANEMIA AFTER HSCT WITH ABO INCOMPATIBILITY OR OTHER IMMUNE-ERYTHROCYTE DISCREPANCIES

Marina Aranguren Ostolaza1, Candela Ceballos Bolaños 1, Itziar Oiartzabal Ormategui1, Nerea Arratibel Zalacain1, Pamela Millacoy Austenrritt1, Mónica Fernández Pérez1, Maria Cruz Viguria Alegria1, Carlos Manuel Panizo Santos1, Irene Sánchez Prieto1, Monserrat Lozano Lobato1

1Donostia University Hospital, Donostia-San Sebastian, Spain

Background: ABO group incompatibility is observed in up to 50% of HSCT cases. This increases the recipient’s risk of experiencing acute or delayed hemolytic reactions, delayed engraftment of red blood cells, and pure red cell aplasia (PRCA), which can lead to anemia with clinical repercussions, increased transfusion requirements, and related consequences such as iron overload or increased alloimmunizations. Although there is no standardized therapy for this group of complications, recently described severe post-transplant immune anemia cases have responded favorably to Daratumumab treatment. Its effectiveness is believed to stem from the direct immunosuppressive effect on antibody-producing plasma cells. However, we propose the additional hypothesis that the presence of CD38 receptors on the recipient’s red blood cell membrane and the antigen-antibody binding at this level may confer protection against hemolysis.

Methods: We present a descriptive study of 4 patients undergoing HSCT at the Donostia University Hospital between 2019 and 2022. All 4 patients received peripheral blood stem cell infusion from HLA identical unrelated donors and a conditioning regimen. All showed mixed chimerism by day +30, with two achieving complete chimerism by day +100. All 4 patients, due to major AB0 group incompatibility or other immune-erythrocyte discrepancies, presented PRCA or peripheral hemolytic anemia with severe clinical and analytical impact. After failed transfusion support and Rituximab therapy, all 4 received Daratumumab treatment.

Results: Patients with PRCA received between 3 and 8 weekly Daratumumab cycles. The mean pre-transplant packed red cell transfusion was 15.3 (range 1-27), during transplantation was 38 (range 12-61), and after completing Daratumumab treatment until now was 0.33 (range 0-1). Therefore, we observed a 99.13% reduction in transfusion support. Hemoglobin levels before Daratumumab initiation were 3.4 g/dL, 6.8 g/dL, and 8 g/dL. After the last cycle, they increased to 4.8 g/dL, 7 g/dL, and 13.9 g/dL, respectively. Patients reached maximum levels of 17.3 g/dL, 14.2 g/dL, and 14.5 g/dL without transfusion support. The mean days to achieve the target Hb>9g/dL after the last cycle were 44.66 days, and to maintain Hb>12 g/dL were 66.33 days.

The patient diagnosed with hemolytic anemia is analyzed separately due to the uniqueness of their evolution and response to treatment. In this case, we observed an optimal response to Daratumumab but also dependency on treatment due to episodes of severe anemia after its withdrawal. The outcome was the administration of 18 cycles and ultimately a second HSCT.

Lastly, laboratory findings showed significantly reduced agglutination and hemolysis when exposing AB red blood cells from a patient on Daratumumab treatment to anti-A and anti-B antibodies compared to the control using AB red blood cells without Daratumumab against these antibodies.

Full size table
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (9)

Conclusions:

  • Daratumumab is effective in post-transplant immune anemia confirmed by the increase in hemoglobin levels from early treatment phases, with a mean of 44.66 days to reach Hb>9g/dL after the last cycle.

  • Patients treated with Daratumumab reduced packed red cell transfusion requirements.

  • The binding of anti-CD38 on red blood cell membranes might exert a protective function against hemolysis by preventing the binding between anti-A or anti-B antibodies in ABO incompatibility.

Disclosure: Nothing to declare

20: Aplastic Anaemia

P069 UPFRONT T-REPLETE HAPLO-IDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR CHILDREN WITH SEVERE APLASTIC ANEMIA: A SINGLE CENTER EXPERIENCE FROM JORDAN

Ayad Ahmed Hussein 1, Nour Awni Ghanem1, Bayan Majed Alaaraj1, Tariq Ahmed Abdelghani1, Dina Mohammad Abu Assab1, Hadeel Hassan Al-Zoubi1

1Bone Marrow and Stem Cell Transplantation Center, Istishari Hospital, Amman, Jordan

Background: Severe aplastic anemia (SAA) is a potentially fatal disease that can be cured with hematopoietic stem cell transplantation (HSCT). Matched sibling donors are not always readily available, leading to delayed referral for HSCT after immunosuppressive therapy, which has a poor prognosis.

Methods: This is a retrospective analysis of all pediatric patients with SAA who received upfront haplo-identical HSCT at Dr. Ayad Bone Marrow and Stem Cell Transplantation center, Istishari Hospital, Amman-Jordan from October 2016 till November 2021.

Results: Seven patients with a median age of 7 years (2-10) were included. Five were males. All patients received rabbit anti-thymocyte globulin (rATG) 0.5 mg/kg on day −9 and 2 mg/kg on days −8 and −7, fludarabine 30 mg/m2/day IV on days −6 to −2, cyclophosphamide 14.5 mg/kg/day IV on days −6 to −5, and 200 cGy TBI in a single fraction on day-1. The graft versus host disease (GVHD) prophylaxis was with post-transplant cyclophosphamide (PT/Cy) at 50 mg/kg/day IV on days +3 and +4 post-HSCT. Mycophenolate Mofetil and tacrolimus were started on day +5 and stopped on days +28 and +365 post-HSCT, respectively. The donor was one of the parents in all cases. Four donors were 5/10, two 7/10 and one 8/10 HLA-matched. Five donor-recipient pairs had major blood group incompatibilities. Five patients received peripheral blood stem cells (PBSC), and two received GCSF-primed bone marrow (BM) grafts. The median time for neutrophil and platelet engraftment was 14 (12–17) and 18 (16–22) days, respectively. One patient developed primary graft failure and was successfully engrafted after a second transplant. None of the patients required ICU admission. Grade I- II acute GVHD occurred in four patients. Three patients had CMV reactivation, one EBV reactivation, and one BK virus cystitis. Two patients had chronic GVHD, of which one was extensive. At a median follow-up time of 55 months (26–90), all patients are alive and transfusion-independent.

Conclusions: Matched related donor (MRD) HSCT is recommended for patients with SAA and proven to improve the long term survival. Upfront T-replete haplo-identical HSCT is an effective and safe option in patients who lack MRD. Our data indicates that this approach might be feasible in countries with limited resources. This approach might change treatment algorithm for patients with SAA in the future. More prospective studies with a larger sample size are required.

Disclosure: Nothing to declare.

20: Aplastic Anaemia

P070 PERIPHERAL BLOOD AS GRAFT SOURCE FOR APLASTIC ANEMIA TRANSPLANT: OUTCOMES OF YOUNG ADULTS AND ADULTS IN A MEXICAN PUBLIC HOSPITAL

Guillermo Sotomayor Duque 1, Severiano Baltazar Arellano1, Roxana Saldaña Vazquez1, Humberto Guerra Ramos1, Raul Ramos1, Alma Fabiola Alvarado Charles1, Karen Machuca Adame1, Luis Omar Gudiño Cobos1, Victor Valerio Bugarin1, Rosa Elva De leon Cantú1, Roberto Hernandez Valdez1, Jose Marcelino Chavez Garcia1

1IMSS UMAE 25 Monterrey, Monterrey, Mexico

Background: Aplastic anemia (AA) is a benign hematologic pathology characterized by an autoimmune T-cell mediated attack on marrow niche and its consequent failure. Allogeneic transplant is the curative treatment of choice for patients under 40 years of age with a matched related donor (MRD); for patients who do not have an identical donor, or who have failed immunosuppressive therapy; related haploidentical transplantation has become a feasible option in several centers around the world.

Methods: Retrospective study in IMSS UMAE 25 Monterrey, Mexico. Descriptive analysis was performed with measures of central tendency, medians, and frequencies. Survival analysis was performed with the Kaplan Mier method and comparative with log rank with a significant p value <0.05.

Results: A total of 16 patients were included in a period from January 2017 to January 2023; 9 male (56%) and 7 female (44%) with a median age of 32 years (15-58). Only 4 patients had a MRD, 12 underwent an haploidentical transplant. Median follow-up was 8 months (1-20). Conditioning was based on a non-myeloablative scheme (ATG-Flu-Cy) in 83% and RIC scheme (Bu-Flu-Cy) in 17%. GVHD prophylaxis, was based on methotrexate and cyclosporine for the MRD group and tacrolimus, mycophenylate mofetil and post transplantation cyclophosphamide for the haploidentical one. Median CD34+ was 12.74 x 106 (6.9-21.6); graft source was peripheral blood in all patients and the donor was male in 100% of cases. Blood group compatibility was 94%. Previous to transplantation 3 patients underwent desensitization for donor-specific anti-HLA antibodies using a protocol that included plasma exchange, IVIg, Rituximab and MMF. Median chimerism of 99% (2-100%) was reach by day 30. Primary graft failure occurred in 1 patient and secondary failure in two. CMV reactivation befall in 31%. The incidence of GVHD at 18 and 24 months was 28% and 52% respectively (G I/II), with no patient developing acute or extensive severe GVHD.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (10)

Median time the follow-up was 9 months (1-27). Overall survival (OS) was 80% at 24 months in all patients; 75% in the MRD group and 83% for the haploidentical (log rank 0.049, p = 0.82). Patients younger than 40 years had OS at 24 months of 100% and for those older than 40 years it was 54% (log rank 4.71, p = 0.029)Fig 1. The sustained hematological response was 75% for MRD and 52% for HAPLO at 2 years (log rank 0.25, p 0.63) Fig 2. Transplant related mortality (TRM) was 19% (one patient due to primary graft failure, one due to CMV infection and one patient due to cerebral hemorrhage).

Conclusions: In our small cohort peripheral blood as graft source proved no difference in OS and GVHD frequency in either group, with a TRM below 20%. Patients under 40 years of age benefits the most from transplantation in a timely and early manner.

Disclosure: No disclosure

20: Aplastic Anaemia

P071 UPFRONT HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) IN CLASSICAL PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH)

Lorie Gandhi 1, Col (Dr) Rajiv Kumar1, Brig(Dr) Rajan Kapoor1

1Army Hospital Research and Referral, New Delhi, India

Background: Paroxysmal Nocturnal Hemoglobinuria (PNH) is a rare acquired hematological disorder characterized by episodic intravascular hemolysis. While Eculizumab is the standard of care in the West, the drug is seldom used in India due to its prohibitive cost. Hematopoietic Stem Cell Transplantation (HSCT) remains the only curative therapy.

Methods: A retrospective, observational study was conducted comprising classical/hemolytic PNH patients who underwent HSCT at our center from the year 2009 to 2023.

Results: Nine patients (five male, four female) with classical PNH who underwent HSCT were identified. Indication for transplant was hemolysis and recurrent need for blood transfusion for all patients. The median age was 28 years (Range 17-48 years). Median time from diagnosis to transplant was 15 months (8 months- 7 years). 3 patients initially presented as non- severe aplastic anemia with a small PNH clone size and later progressed to classical PNH. Out of 9, 7 patients were previously treated with androgens and steroids, 1 patient with Cyclosporine and 1 with Cyclosporine and Eltrombopag. One patient had a pulmonary thromboembolic episode. All patients had hyper-cellular marrow before transplant and median PNH clone size was 76% (28%- 92%). All patients underwent matched sibling donor (MSD) transplant. All received Myeloablative conditioning regimen (MAC): 5 received FLU-BU-ATG(BU-Busulphan, FLU- Fludarabine, ATG- Antithymocyte Globulin) 3 BU-CY-ATG, (CY-Cyclophosphamide) 1 FLU-BU-CY-ATG and 1 BU-CY. Median CD34+ stem cell dose was 6.0 × 106/kg (range 4.38–12.93 × 106/kg). All received cyclosporine and methotrexate as GVHD prophylaxis. Stem cells were derived from peripheral blood for all patients. All patients showed successful engraftment. Clone size at 90 days was <1% for all patients. Acute Graft Versus Host Disease (GVHD) Grade I–II occurred in 3 patients. Grade I-II Chronic GVHD occurred in three patients. Out of 9, 5 patients had CMV reactivation. 3 patients (33.33%) died. One patient (28-year-old male, received BU-CY conditioning) succumbed to Grade IV gut GVHD, 161 days post- transplant. One patient (48-year-old female received FLU-BU-CY-ATG) succumbed to veno-occlusive disease (VOD) post 23 days of transplant and one patient (31-year-old-male, received FLU-BU-ATG conditioning, with a history of thrombosis and a time from diagnosis to transplant of 7 years) died at day 256 post-transplant due to fungal pneumonia. The 6 surviving individuals are transfusion independent. Their median follow up was 8.5 months (3 months- 10 years).

Conclusions: Findings in this study suggest that in the setting of non availability of complement inhibitors, allogenic transplant with matched sibling donor has a curative potential in classical hemolytic PNH. Myeloablative conditioning is preferred. We found 100% engraftment with PBSC in patients with a history of multiple blood transfusions. Addition of ATG may prevent grade IV GVHD. Use of two alkylating agents as a part of conditioning regimen may increase chance of VOD. Early tapering of immunosuppression may prevent life threatening fungal pneumonia in PNH patients who have prior long term steroid exposure.

Disclosure: Nothing to declare

20: Aplastic Anaemia

P072 A REAL-WORLD ANALYSIS OF TREATMENT ADHERENCE AND CLINICAL OUTCOMES FOR PATIENTS WITH PAROXYSMAL NOCTURNAL HAEMOGLOBINURIA (PNH) RECEIVING PEGCETACOPLAN

Koo Wilson 1, Carly Rich1, Zalmai Hakimi1, Regina Horneff1, Jesse Fishman2, Jennifer Mellor3, Lucy Earl3, Yasmin Taylor3, Alice Simons3

1Sobi Pharmaceuticals, Stockholm, Sweden, 2Apellis Pharmaceuticals, Massachusetts, United States, 3Adelphi Real World, Manchester, United Kingdom

Background: Paroxysmal nocturnal haemoglobinuria (PNH) is a rare, multi-systemic disease, which can be fatal if left unmanaged. It is characterised by thrombosis, impaired bone marrow function and complement mediated haemolysis. Currently available treatments include complement protein 5 inhibitor (C5i), which inhibit intravascular haemolysis (IVH), however patients can still experience anaemia and fatigue due to residual IVH and emerging extravascular haemolysis (EVH). Pegcetacoplan is the first approved complement protein 3 inhibitor (C3i), for adults with PNH. It proximally blocks complement activation resulting in broader control of both IVH and EVH. Currently, there is limited real-world data to understand how adherence and administration of pegcetacoplan may impact clinical outcomes for patients with PNH. The aim of this study was to describe the effectiveness and treatment adherence of patients with PNH receiving pegcetacoplan.

Methods: Data were drawn from the Adelphi PNH Disease Specific Programme™, a real-world cross-sectional survey of physicians, and their patients with PNH from January-November 2022 in the United States (US), France, Italy, Germany, and Spain. Patients were eligible for inclusion if prescribed PEG for ≥1 month. Physicians completed surveys utilising data from patients’ medical charts alongside their own clinical judgement regarding patient demographics, pegcetacoplan dosing and frequency of administration, treatment adherence, and clinical outcomes. Descriptive statistics were reported.

Results: Fourteen physicians completed record forms for 61 patients receiving pegcetacoplan. Mean ± SD age was 37.1 ± 11.3 years and 59.0% were male. Patients had been diagnosed with PNH for a mean of 3.7 ± 3.3 years and were receiving pegcetacoplan for 5.9 ± 4.0 months. All patients received 1080 mg of pegcetacoplan per dose, with 98.3% receiving it every 3-4 days and 68.9% self-administered their treatment. Physicians reported 95.1% of patients receiving pegcetacoplan were completely adherent to their treatment regimen. At data collection, physicians reported a mean haemoglobin score of 11.5 ± 1.6 g/dL which was improved from 9.0 ± 1.5 g/dL at pegcetacoplan initiation. Improvement in lactate dehydrogenase (LDH) was seen from PEG initiation to date of data collection (30.0% vs 57.4% reporting LDH <1.5 x ULN), and a higher proportion reported no fatigue (1.6% vs 31.1%, respectively). Physicians considered all patients to have ‘well or very well controlled’ disease.

Conclusions: This study demonstrates high adherence in a real world setting to pegcetacoplan, with almost all patients receiving this treatment in line with the label indication. Physicians reported improvement in patient haemoglobin, LDH levels and reported a high level of disease control for these patients. Further research is needed to understand the long-term adherence and utilisation of pegcetacoplan in a real-world setting.

Disclosure: Wilson, Koo; Rich, Carly; Hakimi Zalmai, and Horneff, Regina are employees of Sobi and may own stocks/shares of the company. Fishman, Jesse is an employee of Apellis Pharmaceuticals and owns stocks/shares of the company. Mellor, Jennifer; Earl, Lucy; Taylor, Yasmin; Simons, Alice are full-time employees of Adelphi Real World, which received funding for participating in this research.

20: Aplastic Anaemia

P073 EXPERIENCE OF SALVAGE HAPLO-HSCT WITH PTCY FOR ACQUIRED SEVERE APLASTIC ANEMIA

Yuliya Mareika1, Nina Minakovskaya1, Natalia Kirsanava 1, Dmitriy Prudnikov1, Mariya Naumovich1, Lubov Zherko1, Aliaksei Kakunin1, Aliaksei Lipnitski1, Volha Mishkova1, Katsiaryna Vashkevich1, Anzhalika Solntsava1

1Republican Scientific and Practical Center for Pediatric Oncology, Hematology and Immunology, Minsk, Belarus

Background: Hematopoietic stem-cell transplantation (HSCT) from the human leucocyte antigen (HLA)-matched sibling donor (MSD) is the first choice for the treatment of severe aplastic anemia (SAA). An improvement over last decade’s results of HSCT from HLA-match unrelated donor (MUD) has made HSCT from MUD the front-line option treatment with no prior failed immunosuppressive therapy (IST) for young. Unfortunately, half of the patients do not have an available HLA-identical donor. They are considered for IST or alternative donor HSCT in case of treatment failure.

Methods: Two patients, diagnosed with idiopathic SAA, underwent a haploidentical HSCT from HLA 6/10 mismatch-related donor (fathers) after the failure of IST with severe infection complications and repeated granulocyte transfusions for the life-saving treatments (table 1). Conditioning regimen consisting total lymphoid irradiation (TLI) 4 Gy, antithymocyte globulin (ATG) (Genzyme) 7,5 mg/kg, fludarabin 150 mg/m2, thiotepa 10 mg/kg, cyclophosphamide (Cy) 30mg/kg, rituximab 200 mg/m2, was followed by transfusion of unmanipulated hematopoietic stem cells. Post-transplant Cy (PtCy) (40 mg/kg on days +3 and +4) was given to reduce the incidence of acute graft-versus-host disease (aGVHD). Granulocyte colony-stimulating factor (G-CSF) 5 μg/kg (from day +5) has been used post-HSCT for earlier neutrophil engraftment. MSC infusion was performed (day +11) to neutrophil and platelet engraftment, a lower risk of aGVHD.

Results: Both patients are alive and well at six and three months. Recovery of neutrophils and platelets was prompt with 100% donor chimerism. None severe toxic or infectious complications were diagnosed after HSCT. Nobody developed acute GVHD or any other immune complications. Immune reconstitution (CD4 + >100 microL) was achieved early. (Table 1).

Conclusions:

Granulocyte transfusion is a reasonable option to control infections and prepare the patients for HSCT. Conditioning regimen TLI/ATG/fludarabin/thiotepa/Cy/rituximab with PtCy reduces the incidence of graft rejection and severe aGVHD. Haplo-HSCT with PtCy in patients with SAA who failed IST with life-threatening comorbidity is a feasible salvage treatment with stable engraftment and an acceptable complication profile.

Table 1

Patient

1

2

Age (year)/sex

2 y.o., female

18 y.o., male

Prior IST

CSA + ATG+steroids+G-CSF

CSA + ATG+steroids+G-CSF

Heavily transfused preHSCT

Yes

Yes

Complications preHSCT

Cardiomyopathy with heart failure, enterocolitis, bronchiolitis

acute kidney failure, granulomatosis with polyangiitis, pneumonia, sepsis, EBV, HHV-6

Granulocyte transfusions preHSCT (n)

7

9

Time to HSCT from 1st day IST (days)

205

85

Source of HSC

Bone marrow

Peripheral blood

CD 34+ (x106/kg)

5,52

5,32

CD 3+ (x106/kg)

56,9

357

aGvHD prophylaxis

CSA, MMF→medrol

Medrol, ruxolitinib (w/o CSA – high risk TMA)

Granulocyte recovery (day)

+25

+14

Thrombocyte ≥20 (day)

+24

+39

Chimerism +30 day (%)

100

100

Chimerism +60 day (%)

100

100

Acute GVHD

no

no

Complications

Local cellulitis, CMV

ВKV hemorrhagic cystitis and nephropathy, mucositis HSV, CMV

CD4 + > 100microL (day)

+30

+71

Follow up

Alive and well (6 mo post)

Alive and well (3 mo post)

  1. F – female, M – male, CSA - cyclosporine A, EBV - Epstein-Barr Virus, HHV-6 - human herpes virus 6, MMF - Mycophenolate mofetil, TMA - thrombotic microangiopathy, CMV – Cytomegalovirus, ВКV – BK-virus, HSV - herpes simplex virus

Disclosure: Nothing to declare

21: Autoimmune Diseases

P074 THYMIC SIZE LONGITUDINAL CHANGES ON CHEST HIGH-RESOLUTION COMPUTED TOMOGRAPHY AFTER AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR DIFFUSE CUTANEOUS SYSTEMIC SCLEROSIS

Gregory Pugnet 1, Samia Collot1, Antoine Petermann1, Pauline Lansiaux2, Gwenaëlle Lorillon3, Nassim Ait Abdallah2, Mathieu Resche-Rigon4, Cécile Borel1, Zora Marjanovic5, Dominique Farge2

1CHU Toulouse, Toulouse, France, 2Unité de Médecine Interne (UF 04), CRMR MATHEC, Maladies Auto-Immunes et Thérapie Cellulaire, Centre de Référence des Maladies Auto-Immunes Systémiques Rares d’Ile-de-France MATHEC, AP-HP, Hôpital St-Louis, Paris, France, 3Service de Pneumologie et Allergologie, AP-HP, Hôpital St-Louis, Paris, France, 4SBIM Hôpital St-Louis, AP- HP, Université de Paris, Paris, France, 5Hématologie Clinique et Thérapie Cellulaire- Hôpital Saint-Antoine, AP-HP, Paris, France

Background: The thymus is a central lymphatic organ responsible for T-cell differentiation and maturation throughout life. The thymus reaches its maximum absolute weight at puberty, and thymic involution then begins, with fatty infiltration of the gland. Incomplete thymus involution is observed in 12 to 15% of systemic scleroderma (SSc) patients and Meunier et al. reported an association between SSc-related interstitial lung disease and incomplete thymus involution. However, few data exist on thymic evolution after resetting the “immunologic clock” by autologous stem cell transplantation (aHSCT) in early diffuse cutaneous systemic scleroderma. The aim of this study was, therefore, to evaluate thymic involution frequency in SSc patients who underwent aHSCT and thymic size longitudinal changes on chest high-resolution computed tomography (HRCT) after transplant.

Methods: Chest HRCT evaluation was performed before aHSCT and afterwards during yearly routine clinical and paraclinical follow-up in 33 consecutive dcSSc-patients between January 2000 and January 2016. Two independent chest radiologist experts blindly assessed the thymus. Patients were retrospectively classified as clinical responders or clinical non-responders according to: at least 25% improvement in mRSS and/or 10% improvement in FVC or carbon monoxide diffusing lung capacity as compared with baseline, and without need for additional immunosuppression within the 24 months after transplant.

Results: Altogether we evaluated 33 SSc patients (20 female, 13 male) with a mean age at transplantation of 45.5 ±13.5 years and a disease duration of 28.0 ±17.7 months. 27.3% (n = 9) of the evaluated patients showed incomplete involution of the thymus at baseline. At 24 months after aHSCT, significant enlargement of thymus tissue were observed as compared with before aHSCT (p = 0.001). Among the 24 patients with complete involution of the thymus at baseline two of them grows after transplant.

Nineteen patients were clinical responders at 2 years (probability of response 0.68, 95% CI (0.49;0.82). All the patients with an increase in thymic size were in the clinical responders group. The mean absolute change from baseline in thymic size at month 24 was +23.6 mm2 in the clinical responders group and −38.1 mm2 in the clinical non-responders group (p = 0.002).

Table 1: Patients (n = 33) clinical and functional characteristics and Systemic Sclerosis organ involvement before treatment by autologous hematopoietic stem cell transplantation

Characteristics

n (%)

or mean ± SD

n

Age at aHSCT, years

45.5 ±13.5

33

Sex, female

20 (60.6)

33

Disease duration since SSc diagnosis, months

28.0 ±17.7

33

Body mass index (kg/m2)

24.0 ±3.7

33

Smoking status (ever vs never)

9 (27.3)

33

Skin involvement

Modified Rodnan Skin Score (0-51)

24.0 ±10.9

32

Lung involvement

33

Interstitial lung disease

29 (87.9)

33

Pulmonary Hypertension

3 (9.1)

33

Cardiac involvement

17 (51.5)

33

Gastrointestinal involvement

16 (48.5)

33

Immunological status and biological values

Antitopoisomerase-1 antibody positive

23 (69.7)

33

Thymic size (mm2)

88.8 ±173.7

33

Incomplete involution

9 (27.3)

33

  1. SD: standard deviation; BMI: body mass index; AHSCT: autologous hematopoietic stem cell transplantation

Conclusions: Real-world data show a significant thymic size increase within two years after aHSCT for early diffuse cutaneous systemic scleroderma which is associated with clinical response.

Disclosure: The authors declare non conflict of interest for this work.

21: Autoimmune Diseases

P075 LONG TERM PERSISTENCE OF T MEMORY STEM CELLS FOLLOWING AUTOLOGOUS HAEMATOPOIETIC STEM CELL TRANSPLANTATION FOR MULTIPLE SCLEROSIS

Melissa Khoo1,2, Carole Ford1, Jennifer Massey1,2,3, Kevin Hendrawan4, Malini Visweswaran1,2, John Zaunders1,2, Ian Sutton3, Barbara Withers3, David Ma1,2,3, John Moore 1,2,3

1St Vincent’s Centre for Applied Medical Research, Sydney, Australia, 2The University of New South Wales, Sydney, Australia, 3St Vincent’s Hospital Sydney, Sydney, Australia, 4The University of Queensland, Brisbane, Australia

Background: Autologous haematopoietic stem cell transplantation (AHSCT) is a high-efficacy therapy for severe autoimmune diseases, including Multiple Sclerosis (MS). While the mechanisms underlying the clinical benefits are still under investigation, the regeneration of a diverse T-cell repertoire is thought to play a critical role in the re-establishment of self-tolerance. T memory stem cells (Tscm) have been reported to be important novel players in post-transplant immune reconstitution in the haploidentical allogeneic setting. The role of Tscm in autoimmune AHSCT is however unknown. Here, we present the first study to examine Tscm in patients with MS undergoing AHSCT, and also in a non-autoimmune non-Hodgkin’s lymphoma (NHL) comparator group.

Methods: Multicolour flow immunophenotyping of T-cell subpopulations (Tscm (CD3+CCR7+CD45RO-CD45RA+CD27+CD95+), naïve (Tn), central memory (Tcm), transitional memory (Ttm), effector memory (Tem), and terminal effector (Tte) T-cells, recent thymic emigrants (RTE)) was performed on PBMCs (pre-AHSCT; d8, d14, 3m, 6m, 12m, 24m, 36m post-AHSCT) and leukapheresis product cryopreserved from MS and NHL patients undergoing AHSCT with BEAM conditioning (following written informed consent (HREC SVH 10/206), according to the Declaration of Helsinki; MS: n = 22, NHL: n = 5, Healthy Controls (HC): n = 4). Alterations in plasma levels of IL-7 and IL-15 were detected using Milliplex High-Sensitivity Bead-Based Assays. Statistical analysis was performed using: repeated measures ANOVA with Holm-Sidak Post-Hoc test (timepoint analysis), and 2-way ANOVA (mixed-effects) with Sidak Post-Hoc test (MS vs NHL), with logarithmic transformations if required after residual analysis (p<0.05); non-parametric Mann-Whitney U test (pre-AHSCT vs HC; p<0.05); using GraphPad Prism 8.

Immunophenotyping of longitudinal PBMC samples revealed significantly elevated levels of both CD4+ and CD8+ Tscm in MS patients post-AHSCT (p<0.03 and p<0.04 respectively), with this difference persisting long-term to at least 36m. In contrast, no changes in Tscm levels were detected in NHL patients post-AHSCT. While pre-AHSCT, significantly higher proportions of CD8+ Tscm were observed in both MS and NHL patients compared to HC (MS: 8-fold, p<0.0005; NHL: 13-fold, p<0.02). The kinetics of the remaining T-cell subpopulations (Tn, Tcm, Ttm, Tem, Tte, and RTE) were as expected post-AHSCT for both MS and NHL, with patterns of transient differences that returned to baseline by 12-36m. Early post-AHSCT we detected significantly increased levels (>4-fold) of plasma IL-15 in both MS (d8: p<0.001; d14: p<0.05) and NHL (d8: p<0.01), which subsequently returned to baseline in NHL, but decreased below baseline in MS. Conversely, significantly reduced plasma IL-7 was found at d8, 3m and 6m post-AHSCT for MS (p<0.01), whilst NHL displayed no differences.

Conclusions: To our knowledge, this is the first study to demonstrate Tscm expansion in an autoimmune HSCT setting. Elevated CD4+ and CD8+ Tscm post-AHSCT was specific to MS patients, with the absence of Tscm changes in NHL suggesting this is unique to MS patients and not only a consequence of post-conditioning lymphopenia. Interestingly, this persisted long-term in MS patients with Tscm remaining elevated out to 36m. Furthermore, the surge in IL-15 early post-AHSCT, in conjunction with persistently decreased IL-7 levels, suggests a shift in the IL-15/IL-7 balance, which may contribute to Tscm expansion in MS patients.

Disclosure: Authors: Nothing to declare

21: Autoimmune Diseases

P076 EFFECTS OF HIGH DOSE IMMUNOSUPPRESSIVE THERAPY WITH AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN MULTIPLE SCLEROSIS FROM PATIENT’S PERSPCTIVE: LONG-TERM QUALITY OF LIFE OUTCOMES

Denis Fedorenko 1, Vladimir Melnichenko1, Anatoly Rukavitsin1, Nikolai Vasilev1, Tatiana Nikitina2, Natalia Porfirieva3, Tatiana Ionova2

1Pirogov National Medical Surgical Center, Moscow, Russian Federation, 2Saint-Petersburg State University Hospital, Saint-Petersburg, Russian Federation, 3Multinational Center for Quality of Life Research, Saint-Petersburg, Russian Federation

Background: Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease of the central nervous system which can lead to severe disability and result in profound quality of life (QoL) impairment. Patient’s QoL is an important outcome of MS treatment. It is shown that high-dose immunosuppressive therapy (HDIT) with autologous hematopoietic stem cell transplantation (AHSCT) is a valuable option in MS which results in clinical and QoL improvement. The data about long-term QoL outcomes after HDIT + AHSCT are limited. We aimed to evaluate QoL changes in patients with MS at different time-points terms after HDIT + AHSCT.

Methods: Patients with different types of MS who underwent HDIT + AHSCT were enrolled in a longitudinal, prospective and single-center study. Two low-intensity regimens BEAM-like and Cyclophosphamide were applied. RAND SF-36 was used for QoL assessment before AHSCT, at 6 and 12 months after AHSCT, then every 6 months during 2 years after AHSCT and then every 12 months after 2 years of follow-up. Patients who could not answer the questionnaire themselves were excluded and incomplete questionnaires were eliminated. For comparisons t-test or Wilcoxon test as well as Generalized Estimating Equations were used.

Results: In total, 240 patients with MS were included in the analysis: 132 relapsing-remitting MS (RRMS) (55%), 75 secondary progressive MS (SPMS) (31.3%) and 33 primary progressive MS (PPMS) (13.7%). Median age – 40 years old [Q1; Q3 – 32; 49], 35.8% - males. Median baseline EDSS – 4 [Q1; Q3 – 2; 6]. BEAM-like was used in 85 patients (35.4%), Cyclophosphamide – in 155 patients (64.6%). Mean follow-up was 21 months (range: 6-97). We found a significant increase of all 8 SF-36 scales in 12 months post-transplant as compared with base-line in the entire group (p<0.05). In RRMS patients the values of all SF-36 scales significantly improved (p<0.01) In patients with progressive MS statistically significant improvement was registered for 4 out of 8 SF-36 scales (p<0.01); changes for role functioning scales, bodily pain and mental health scales were not statistically significant. During the entire period of follow-up in patients without progression or relapse QoL further improved. Significant positive changes by all SF-36 scales as compared to baseline were observed for the whole group (p≤0.001). For patients with both remitting and progressive MS positive changes for physical and mental health components of SF-36 were revealed during the follow-up post-transplant (p<0.001 for physical and mental health components in remitting MS and physical component of progressive MS; p = 0.012 for mental health component in progressive MS).

Conclusions: HDIT + AHSCT resulted in significant and sustained improvement of QoL in MS patients post-transplant. Meaningful QoL improvement was observed both in patients with remitting and progressive MS at long-term follow-up.

Clinical Trial Registry: No

Disclosure: Nothing to declare

21: Autoimmune Diseases

P077 THE MATHEC-SFGM-TC REGISTRY FOR CELL THERAPY IN AUTOIMMUNE DISEASES: A DEDICATED TOOL FOR REAL-WORLD DATA COLLECTION

Pauline Lansiaux1, Manuela Badoglio2, Grégory Pugnet3, Mathieu Puyade4, Emmanuel Chatelus5, Thierry Martin5, Louis Terriou6, Alexandre Maria7, Marc Ruivard8, Jacques-Olivier Bay8, Bertrand Dunogué9, Matthieu Allez1, Hélène Zéphir10, Arsène Mékinian11, Eric Deconinck12, Sabine Berthier13, Françoise Sarrot-Reynaud14, Frédéric Garban14, Nicolas Maubeuge4, Guillaume Mathey15, Cristina Castilla-Llorente16, Céline Labeyrie17, Régis Peffault de la Tour18, Marie Robin19, Zora Marjanovic11, Dominique Farge 1

1AP-HP, Hôpital Saint-Louis, Paris, France, 2EBMT Paris Study Office, Paris, France, 3CHU Toulouse, Toulouse, France, 4CHU Poitiers, Poitiers, France, 5CHU Strasbourg, Strasbourg, France, 6CHU Lille, Lille, France, 7CHU Montpellier, Montpellier, France, 8CHU Clermont-Ferrand, Clermont-Ferrand, France, 9AP-HP, Hôpital Cochin, Paris, France, 10CHRU Lille, Lille, France, 11AP-HP, Hôpital Saint-Antoine, Paris, France, 12CHRU Besançon, Besançon, France, 13CHU Dijon, Dijon, France, 14CHU Grenoble, Grenoble, France, 15CHU Nancy, Nancy, France, 16Institut Gustave Roussy, Villejuif, France, 17AP-HP, Hôpital Bicêtre, Le Kremlin-Bicêtre, France, 18AP-HP Hôpital Saint-Louis, Paris, France, 19SFGM-TC, Hôpital Saint-Louis, Paris, France

Background: The European Society for Blood and Marrow Transplantation (EBMT) and International Society for Cellular Therapy (ISCT) Joint Accreditation Committee (JACIE) guidelines underline the necessity to yearly report all consecutive hematopoietic stem cell transplantation (HSCT) and patient’s real world data (RWD) monitoring in the EBMT registry, which allows healthcare professionals to assess activity, practices and treatment outcomes and ultimately to improve patient care. A 2015 European Medicines Agency (EMA) initiative encouraged better use of existing registries or development of new, high-quality registries where no existing RWD is available. In orphan rare autoimmune diseases (AD), registries represent an important source of safety and RWD, specifically for patients receiving HSCT or other Cellular Therapies (CT), as Mesenchymal Stroma Cell (MSC) or CAR-T. The EBMT Registry minimum data-set for AD contains transplant data and short-term (Day 100) post-transplant complications with a need to develop AD specific and longer-term monitoring RWD acquisition both at national and European levels.

Methods: Based on a tight collaboration with EBMT under the auspices of the French-speaking Society for Marrow Transplantation and Cellular Therapy (SFGM-TC), the MATHEC-SFGM-TC registry www.mathec.com)) was developed to register and follow-up all AD patients treated by CT in France. It is hosted by Epiconcept certified Health Data Host and compliant with the French independent administrative authority (CNIL) controlling the use of personal data. The selection of variables was based on those collected for the EBMT Registry core dataset and those used for studies promoted by the AD working part. RWD to be collected, include: a) CT treatments (HSCT/ other CT type, donor, source and associated procedures, complications and survival status) plus b) AD specific diagnosis, clinical biological RWD at baseline and during routine follow-up evaluation after CT at 3, 6 and 12 months, then biannually until 5 years and annually at least until 10 years for long-term follow-up. The MATHEC-SFGMTC centers were asked to include all consecutive AD patients receiving HSCT or other CT, with repeated alerts to update the follow-up as recommended by EBMT and by SFGM-TC. Informed consent was obtained for all patients before inclusion. Data managers were adequately trained and supervised by relevant CT and AD specialists to optimize data quality. All registry data are reported here.

Results: On December 6th 2023, 248 AD patients were included in the MATHEC-SFGM-TC registry: 225 patients with 229 HSCT (228 autologous, 1 allogeneic) since 1997 were followed in 31 centers for a median duration of 48 (19-115) months. CT and AD-specific and long-term RWD were collected for HSCT in 152 rheumatologic (138 scleroderma, 5 polychondritis), 55 neurological (43 multiple sclerosis, 7 CIPD) and 11 Crohn disease patients after either standard cyclophosphamide (n = 122), BEAM (n = 12), low dose cyclophosphamide (n = 62), or other (n = 16) conditioning with ATG (n = 179) and/or Rituximab (n = 45). RDW for MSC (20 Scleroderma, 8 Lupus) and CAR-T (1 lupus) patient were collected.

Conclusions: Combining RWD from multiple centres at registry national level are essential to conduct robust studies in rare AD with a sufficient number of patients and disease specific data.

Disclosure: Nothing to declare

21: Autoimmune Diseases

P078 ATG AND OTHER SEROTHERAPY IN CONDITIONING REGIMENS FOR ASCT IN AUTOIMMUNE DISEASES: A SURVEY OF THE EBMT AUTOIMMUNE DISEASES WORKING PARTY (ADWP)

Azza Ismail1, Rosamaria Nitti 2, Basil Sharrack1, Manuela Badoglio3, Pascale Ambron3, Myriam Labopin3, Tobias Alexander4, John Snowden5, Greco Raffaella2

1Sheffield NIHR Translational Neuroscience BRC, Sheffield Teaching Hospitals NHS Foundation Trust & University of Sheffield, Sheffield, United Kingdom, 2Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy, 3EBMT Paris study office / CEREST-TC - Saint Antoine Hospital - INSERM UMR 938 - Université Pierre et Marie Curie, Paris, France, 4Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, Berlin, Germany, 5Sheffield Teaching Hospitals NHS Foundation Trust; Division of Clinical Medicine, School of Medicine and Population Health, The University of Sheffield, Sheffield, United Kingdom

Background: Serotherapy is a key component of conditioning regimens for autologous hematopoietic stem cell transplant (ASCT) in autoimmune diseases (AD), however, it may be delivered in various dosing combinations across different centers.

Methods: Between December 2022 and November 2023, a survey on the current use of serotherapy for ASCT in ADs, with a focus on anti-thymocyte globulin (ATG), was undertaken by the Autoimmune Diseases Working Party (ADWP) among European Society for Blood and Marrow Transplantation (EBMT) centers, having performed more than 5 ASCTs for AD in adult patients since 2015.

Results: Forty-six EBMT centers (66%) responded to the survey. Among responding centers, 23.9% perform ASCT for neurological AD indications, 15.2% for rheumatological ADs, 58.7% for multiple indications. All centers use ATG, 12 centers (26.1%) also use Rituximab (91.7% of them use it in addition to ATG administration), no center uses Alemtuzumab.

Many centers (58.7%) use the same conditioning regimen for all AD indications. Cyclophosphamide-ATG is the most frequently used regimen (88.8% of centers), followed by BEAM-ATG (22.3%), whereas RIC regimens like cyclophosphamide-fludarabine-ATG were used less frequently. Most centers (93.5%) use ATG for all AD indications. Only one center uses serotherapy-free regimen (cyclophosphamide only), for neurologic indications.

Thymoglobuline is the most commonly used ATG type (89.1% of centers). Four centers report using Grafalon and one center uses Atgam. Among centers using Thymoglobuline, 41.5% administer a total dose (TD) of ≥7.5 mg/kg, while 53.6% administer <7.5 mg/kg. Each center using Grafalon administers a different TD (range 7.5-90 mg/kg). ATG administration is always fractionated over multiple days, most frequently 3 (32.6%) or 5 days (37%); half of centers divide the TD equally for each day of administration.

Rituximab TD is 500 mg (25% of centers) or 1000 mg (75%), the 1000 mg TD is equally fractionated in two days by 6/9 centers. Most centers administer ATG during chemotherapy (63%), while Rituximab is administered before chemotherapy (25%) or before chemotherapy and after HSC infusion (50%).

Test dose is used by 34.8% of centers before ATG administration (ranges 0.28-25 mg or 0.5-1 mg/kg), no center uses rituximab test dose. ATG administration is slower (48.9% of centers administer it over 12 hours) than Rituximab administration (41.7% of centers administer it over 4 hours and 41.7% over 6 hours).

Premedication is used to prevent serotherapy-related adverse events, including antihistamines (100% of centers), paracetamol (91.1%) and steroids (98%), in particular 37.8% of centers administer methylprednisone 1 mg/kg or equivalent and 35.6% administer methylprednisone 2 mg/kg or equivalent. One third of centers repeats premedication at fixed times during a single infusion.

For ATG, 80.4% of centers successfully administer the planned TD to all patients, for Rituximab, 75% of centers administer the TD to all patients. Among HSCT performed for neurologic indications, about half of patients develop clinical relapse during serotherapy administration.

Conclusions: This survey reveals a high degree of consistency across EBMT centres regarding the use of serotherapy in ASCT for AD. However, a wide variability in doses suggests the need for consensus guidelines to best standardize practice.

Disclosure: None related to the manuscript. AI and RN contributed equally to the work.

21: Autoimmune Diseases

P079 ADOPTING A STANDARD OF CARE: AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR SCLERODERMA

Ernesto Ayala 1, Madiha Iqbal1, Hemant Murthy1, James Foran1, Vivek Roy1, Mohamed Kharfan-Dabaja1

1Mayo Clinic Florida, Jacksonville, United States

Background: Three prospective comparative trials have proven the superiority of autologous hematopoietic stem cell transplantation (aHSCT) over other treatments in patients with Scleroderma. A position statement from the American Society for Blood and Marrow Transplantation endorses its use as “standard of care”. However, it remains underutilized in the US.

Methods: This is an IRB approved, retrospective review of all patients with Scleroderma that have undergone aHSCT at Mayo Clinic Florida since April 2020, and completed at least 3 months of follow up. Scleroderma diagnosis was made/confirmed by our Rheumatology Department based on published criteria.

Results: Twenty four patients were included. Median age at transplant was 49 years (range 21-68). 70% of patients were female, 54% were Caucasian. Median time from diagnosis to transplant was 33 months (5-188). Skin was involved in 100% patients, lung (interstitial disease) in 79%, esophagus in 75%, heart in 12%. Right heart catheterization was done in 13 patients (when recommended by Cardiology) with mild pulmonary hypertension found in 5. Gastric antral vascular ectasia was found/treated in one patient. Median time from consultation to transplant was 2 months (1-24). Hematopoietic stem cell mobilization was induced with G-CSF (24 patients) with the addition of plerixafor (13 patients). Median number of peripheral blood stem cell harvests was 2 (1-3). Myeloablative conditioning was used in 3 patients: Cyclophosphamide + rabbit ATG + 800 cGy total body irradiation (2 patients) or melphalan + rabbit ATG (1 patient with synchronic multiple myeloma). Non-myeloablative conditioning was used in 21 patients: Cyclophosphamide + rabbit ATG (19 patients) or cyclophosphamide + fludarabine + rabbit ATG. Two patients had modifications of conditioning due to volume overload. Twenty three grafts were CD34+ selected. Median number of infused CD34+ cells was 5.98 x 10^6/kg. Granulocyte engraftment occurred at a median of 12 days (0-14) and platelet engraftment at a median of 12 days (0-15), as well. Median hospital stay was 20 days (2-46). Two patients died during the conditioning regimen, one of them of a spontaneous pneumothorax followed by cardiac arrest, the second one from acute bowel ischemia and ARDS. Both had advanced cardiopulmonary disease from scleroderma. One patient died of severe pneumonitis, attributed to myeloablative radiation therapy. One late mortality (day +60 was due to multilobar pneumonia in the setting of cirrhosis with massive ascites. Median follow up after transplant is 12.5 months. Median modified Rodnan skin score pre-transplant was 24 (4-44), post-transplant was 12 (0-28). Median forced vital capacity (FVC) pre-transplant was 80% (41-103), post-transplant 74% (46-108). Median corrected DLCO pre-transplant was 66%, post-transplant was 60% (47-70). Non-relapse mortality was 16%. Relapse was 14%. For transplanted patients, 1 year overall survival is 91% (CI 80-100) and relapse free survival is 78.5% (CI 61-100).

Conclusions: High dose immune ablation with a non-myeloablative conditioning regimen induces durable remissions in most patients with Scleroderma. Mobilization and harvest of hematopoietic stem cells can be accomplished successfully without chemotherapy, even if CD34 selection is planned. Patients with advanced cardiopulmonary disease have a high risk of morbidity and mortality and should be excluded of this approach.

Disclosure: Nothing to disclose.

21: Autoimmune Diseases

P080 FLUDARABINE AND CYCLOPHOSPHAMIDE AS A SAFE AND EFFECTIVE LYMPHOABLATIVE CONDITIONING REGIMEN FOR MULTIPLE SCLEROSIS (MS)

Denis Fedorenko 1, Anatoly Rukavitsin1, Nikolai Vasilev1, Vladimir Melnichenko1, Tatiana Ionova2

1Pirogov National Medical Surgical Center, Moscow, Russian Federation, 2Saint-Petersburg State University Hospital, Saint-Petersburg, Russian Federation

Background: At present, high-dose immunosuppressive therapy with autologous hematopoietic stem cell transplantation (AHSCT) has been used with increasing frequency as a therapeutic option for MS patients. BEAM and cyclophosphamide 200 mg/kg are mostly common used as conditioning regimens for MS patients, but toxicity of both is still high. Thus, a new approach to reduce toxicity for MS patients is needed. The study is aimed to analyze toxicity of new conditioning regimen based on Fludarabine, Cyclophosphamide and Rituximab (R-Flu/Cph) to compare with traditional Cyclophosphamide and Rituximab (R-Cph) in MS patients.

Methods: 232 MS patients were included in this study, median age - 40 years; men/women – 132/100; relapsing-remitting MS – 125 patients, primary progressive MS – 61 patients, secondary progressive MS - 46 patients. EDSS score was from 1.0 to 7.5 (median - 4.0). All patients received AHSCT. Conditioning regimens were: R-Cph (Cyclophosphamide 200 mg/kg + Rituximab 500 mg/m2) – 49 patients, R-Flu/Cph (Fludarabine 150 mg/m2 + Cyclophosphamide 100 mg/kg + Rituximab 500 mg/m2) – 183 patients.

Results: Hematologic toxicity of regimens: duration of neutropenia was from 7 to 12 days (median - 10 days) in R-Cph group and from 3 to 10 days (median - 6 days) - in R-Flu/Cph group. The median duration of thrombocytopenia in R-Cph regimen was 7 days, in R-Flu/Cph - 4 days. Platelet transfusion received 63% of patients in R-Cph group and only 8.2% of patients in R-Flu/Cph group. Anemia grade I-II was observed in 90% of patients in booth groups. Oral mucositis was observed in 26% patients in R- Cph+R group (Grade I - 61,5 %; Grade II - 38,5 %) and in 4% of patients in R-Flu/Cph group (Grade I - 100 %). Enteropathy was observed more in R-Cph group - 16 % (grade II) to compare with R-Flu/Cph group (11%, mainly grade I) Infections: the incidence of infectious complications was higher in R-Cph group – 26% of patients to compare with R-Flu/Cph group - 11.4% of patients. One patient died from sepsis in R-Cph group (TRM – 2.0%), TRM in R-Flu/Cph group was 0%. Hepatic toxicity: in R-Flu/Cph group 10% of patients had hepatic toxicity (grade I and II). In R-Cph group hepatic toxicity was observed in 18% of patients (grade II). Range of immunosuppression: the minimal count of lymphocytes was significant lower in R-Flu/Cph group 0,0001 – 0,01 х 109/l (median 0.0008 х 109/l), than in R-Cph group (minimal count of lymphocyte - 0,001 – 0,06 х109/l, median 0.009 х 109/l, p<0.05).

Conclusions: R-Flu/Cph is a very promising program with less toxicity profile, less incidence of infections and significantly deeper rate of lymphoablation. Also TRM was 0% in R-Flu/Cph group to compare with 2% in R-Cph group. Further studies are needed to assess long-term effectiveness and toxicity of new conditioning regimen.

Clinical Trial Registry: No

Disclosure: Nothing to declare

21: Autoimmune Diseases

P081 IMMUNE RESET AND GRAFT COMPOSITION IN AUTOLOGOUS TRANSPLANTATION FOR MULTIPLE SCLEROSIS: EARLY RECOVERY OF NATURAL KILLER CELLS MAY BE IMPORTANT IN EFFECTING DURABLE RESPONSES

Latoya Reid 1, Oliver Gittner2, Malia Begley3, Rowayda Peters3, Andy Drake3, Matthias Klammer3

1University Hospital of the West Indies, Kingston, Jamaica, 2Steve Mills Stem Cell and Immunotherapies Laboratory, NHS Blood and Transplant, Southampton, United Kingdom, 3St. George`s University Hospital, NHS Foundation Trust, London, United Kingdom

Background: Autologous Haematopoietic Stem Cell transplantation (ASCT) is an effective therapeutic modality in patients with relapsing/remitting multiple sclerosis (MS) through immune regeneration with a less auto-inflammatory response pattern, but the mechanism of achieving durable responses remains poorly understood.

In malignant disease, we have previously studied graft composition and revealed high variability of NK cell content in the graft, early NK cell recovery post HSCT and an association of absolute lymphocyte count >0.5 x 10e9/L at D + 15 with more durable remission.

Similarly, early NK reconstitution post ASCT in MS may be responsible for later shaping a less auto-inflammatory immune reconstitution.

Methods: We monitored lymphocyte subsets (CD4, CD8, NK and B cells) monthly for three months and at 6 and 12 months after ASCT in 16 patients transplanted for Multiple Sclerosis. Autologous stem cells were mobilized with Cyclophosphamide 1.5 mg/m2 and GCSF and patients underwent HSCT after conditioning with Cyclophosphamide and ATG. The non-stem cell component of the graft was analyzed by flow cytometric analysis for B, T, NK cells and neutrophils using defrosted cryovials.

Results: Patient and disease characteristics are summarized in Table 1. The median age at ASCT was 35.5 years with a median EDSS score of 4. Most patients received Alemtuzumab (38%) followed by Natalizumab (31%) as their last disease modifying therapy prior to transplant. The median lines of therapy were two with a median of 14 months duration between last highly effective immunosuppressive therapy and transplant.

In the post-transplant period, total T cell numbers did not return to the low normal range until day 240. The CD4/CD8 ratio was clearly inverted until beyond Day 180. In contrast, in the early post-transplant period (Day 30), we saw the highest absolute number of NK cells with a sharp fall to low normal number in subsequent time points. This is also reflected in a high, inverted CD 56: CD 4 ratio in the first month post ASCT, only reverting to a physiological range at Day 240.

Table 1: Demographics and Disease Characteristics

Number of Patients

16

Sex No. (%)

Female

11 (69%)

Male

5 (31%)

Age in years

Median

35.5

Range

24-49

Expanded Disability Status Scale (EDSS) score

Median

4

Range

1.5-6.5

Last immunosuppressant prior to transplant No. (%)

Alemtuzumab

6 (38%)

Natalizumab

5 (31%)

Ocrelizumab

4 (25%)

Pulsed Steroids

1(6%)

Lines of immunosuppressive therapy

Median

2

Duration between last highly effective immunosuppressive therapy and transplant (months)

Median

14

Conclusions: Early immune recovery post ASCT in patients with MS is characterized by a high, inverted NK cell/ CD4 ratio, a finding we previously reported in malignant disease (Multiple Myeloma). Recent studies suggest that NK cells have an immunoregulatory function on Th17 responses, a pro inflammatory mediator implicated in Multiple Sclerosis. Further studies into the early kinetics of immune reconstitution in patients with MS post ASCT, the impact of previous therapies and the non-stem cell component of the graft are required to understand if these shape the durable immune reset and it`s mechanism.

Disclosure: Nothing to declare

21: Autoimmune Diseases

P082 REDUCED PRO-INFLAMMATORY INTERLEUKINS IL-6 AND IL-8 SECRETED BY SKIN FIBROBLASTS IN SYSTEMIC SCLEROSIS PATIENTS AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION

Gunter Assmann 1,2, Jan Weghorn1, Michael Schmidt1, Joerg Henes3, Claudia Pfoehler4, Frank Neumann2

1RUB University Hospital JWK Minden, Minden, Germany, 2Jose Carreras Center of Immungenetics and Gene Therapy, University of Saarland, Homburg, Germany, 3University Hospital Tuebingen, Tuebingen, Germany, 4University Hospital of Saarland Medical School, Homburg, Germany

Background: Systemic sclerosis (SSc) is an autoimmune connective tissue disease defined by fibrosis of skin and internal organs as well as vascular impairment accompanied by a significantly higher mortality rate than in the general population. Early severe courses of SSc should be provided for intensified treatment such as hematopoietic stem cell transplantation (autoTx).

In addition to the fibrosis several features of SSc often show a phenotype of auto-inflammation which is thought to be mediated by the TH17 pathway in addition to other inflammatory factors.

Methods: Here we investigated cultures of skin fibroblasts derived from SSc (3 to 6 months) after successful autoTx as well as without autoTx to evaluate the different expression of pro-inflammatory cytokines (interleukin-1-beta, interleukin-6 (IL-6), interleukin-8 (IL-8)) after stimulation of interleukin-17 (IL-17) and combined with tumor growth factor-beta (TGF-beta), measured by concentrations (ng/ml, +/-standard error) in the cellular supernatant using ELISA tests.

SSc patients (n = 8, aged 43 to 65 years) and three of them after autoTx (n = 3, aged from 54 to 59 years) underwent a skin biopsy of the lateral abdominal wall or forearm.

The cells were stimulated by 25 ng/ml of IL-17 and/or 2.5 ng/ml of TGF-beta for 48h. The differences between SSc and SSc after autoTx were statistically analysed by t-test (with fisher`s correction).

The ethics committee of the Saarland Medical Association has approved the experimental study (EK120/23).

Results: IL-6 secreted by skin fibroblasts showed a significantly higher concentration in SSc patients without autoTx compared to SSc after autoTx. After in-vitro stimulation of the skin fibroblasts by IL-17 the IL-6 as well as IL-8 concentrations were significantly higher in SSc patients without autoTx to SSc without autoTx (figure 1). Furthermore, after IL-17 stimulation together with TGF-beta the concentration of IL-8 (but not IL-6) supernatant concentrations also resulted in significantly higher values for SSc fibroblasts in patients without autoTx compared to patients after autoTx (1.99 + /-0.23 vs. 0.63 + /-0.06 [p = 0.0008]). IL- 1beta concentrations did not show any differences between the subgroups (data not shown).

Conclusions: Skin fibroblasts derived from SSc patients showed significantly stronger IL-6 and IL-8 driven pro-inflammatory properties than skin fibroblasts derived 3 to 6 months after the hematopoietic stem cell transplantation. In what way (1) IL-6 and IL-8 may be a potential biomarker for disease activity of SSc and (2) IL-17 could be a predominant pathway for activity in SSc patients remains the subject of further research.

Disclosure: G. Assmann: advisory boards, research grants from: UCB, Astrazeneca, Boehringer-Inglheim, Novartis, BMS, Vifor Pharm; C. Pfoehler: advisory boards, research grants from UCB, Novartis, BMS, Astrazeneca, AbbVie; J. Henes: advisory boards, research grants from: AbbVie, BMS, Boehringer-Ingelheim, Chugai, GSK, Janssen, NEOVII, Novartis, Pfizer, UCB; remaining authors: no discloser

21: Autoimmune Diseases

P083 THE QUALITY OF LIFE OF PERSONS WITH MULTIPLE SCLEROSIS IMPROVES AFTER AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION

Olivia Lira-Lara1, Moisés Manuel Gallardo-Pérez2, Miranda Melgar-de-la-Paz3, Paola Negrete-Rodríguez4, Luis Enrique Hamilton-Avilés5, Guillermo Ocaña-Ramm5, Max Robles-Nasta2, Daniela Sánchez-Bonilla2, Juan Carlos Olivares-Gazca2, Guillermo José Ruiz-Delgado 2,5, Guillermo José Ruiz-Arguelles2,5

1Universidad Veracruzana, Veracruz, Mexico, 2Centro de Hematología y Medicina Interna, Clínica Ruiz, Puebla, Mexico, 3Universidad Anáhuac Puebla, Puebla, Mexico, 4Universidad de las Américas Puebla, Puebla, Mexico, 5Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico

Background: Multiple sclerosis (MS) is a complex inflammatory, demyelinating and neurodegenerative disease of the central nervous system that causes a whole spectrum of neurological disorders as well as patient’s perception of an abnormal physical, emotional or cognitive state, associated with a profound decrease in the quality of life of affected patients. Currently, autologous hematopoietic cell transplantation (ASCT) is a validated therapeutic approach and has been shown to be superior to the use of new immunomodulatory agents. However, the impact of ASCT on the quality of life of patients with MS remains largely unknown. The objective is to identify the impact of autologous HSCT in the quality of life in patients with MS in a single institution.

Methods: A quasi-experimental, longitudinal, prospective and single-center study was conducted in our institution. The quality of life was determined in patients with MS before and one year after ASCT; it was determined by applying the MS-QoL 54 instrument which is a validated instrument for this pathology. The variables related to the physical and mental components of the instrument as well as demographic characteristics were studied. Patients who could not answer the questionnaire themselves were excluded as well as incomplete questionnaires. ASCT was conducted as outpatients following the “Mexican method”, which employs high-dose both cyclophosphamide and rituximab.

Results: 38 patients answered the questionnaire before and one year after the ASCT. 21 (56%) were female. Median age was 47 years (SD ± 8.85). Of the selected patients, 22 (58%) had relapsing-remitting MS, 12 (32%) primary progressive MS, whereas 4 (11%) had secondary progressive MS. The physical and mental components were analyzed between the quality of life in the patients prior to the ASCT and the follow-up after one year. In the energy component the differences observed before and one year after the ASCT were statistically significant (40.73 vs 49.57, p = 0.04), as well as the differences in the health perceptions (47.1 vs 56.31, p = 0.011), health distress component (40.65 vs 55.26, p = 0.002) and chance in health (31.57 vs 63.81, p <0.0001). On the other hand, in the physical component the differences between before and after one year of ASCT were not statistically significant (p = 0.069) as well as the differences in the mental component either (p = 0.218).

Conclusions: The study suggests that ASCT improves some aspects of the quality of life of persons with MS.

Disclosure: Nothing to declare

21: Autoimmune Diseases

P084 AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN THE TREATMENT OF MULTI-REFRACTORY STIFF PERSON SYNDROME: A CASE STUDY

Tamim Alsuliman 1, Dimitri Psimaras2, Nicolas Stocker1, Simona Sestili1, Anne Banet1, Zoé Van de Wyngaert1, Agnès Bonnin1, Manuela Badoglio3, Mathieu Puyade4,5, Dominique Farge6,5, Mohamad Mohty1, Zora Marjanovic1,5

1Saint-Antoine Hospital, APHP Sorbonne University, Paris, France, 2Hôpital Universitaire Pitié-Salpêtrière, Paris, France, 3EBMT Paris Study Office, Paris, France, 4CHU de Poitiers, Poitiers, France, 5MATHEC, Paris, France, 6Saint-Louis Hospital, Paris, France

Background: Autologous hematopoietic stem cell transplantation (AHSCT) has been successfully used to treat several types of autoimmune diseases (AD) such as multiple sclerosis. (1,2)

The “stiff person syndrome” (SPS) is a rare neurological AD with a prevalence of 1 to 2 patients per million (3), predominantly observed in women (2-3 female/1 man). (3) It is characterized by progressive stiffness of skeletal muscles, episodic painful muscle spasms, and prevention of volitional movements and ambulation in severe cases. Lumbar para-spinal rigidity limits the range body mobility. Spasms due to increased muscle stiffness occur spontaneously or secondary to different external and internal stimuli.(3–5)

Methods: Informed consent was obtained from the patient for the publication of this report. AHSCT is indicated with a grade 1 level of evidence for the treatment of multi-refractory / recurrent SPS. The national committee of transplantation in AD (MATHEC) approved the AHSCT, as a “Clinical Option”, for this patient. The mobilization was performed using cyclophosphamide 1g/m2 D1 and D2 plus G-CSF 30 MUI/day from D6. The conditioning regimen was cyclophosphamide 200 mg/kg total dose, Anti-thymoglobulin 6 mg/kg and Rituximab 500 mg before the conditioning and 500 mg in post-AHSCT.

Results: A 53-years-old female was diagnosed, at the age of 48 years (2016), with Stiff Person Syndrome muscular stiffness. One year later, onset of dysarthria and cerebellar instability were noticed. Neurological cerebellar symptoms progressively increased, with pyramidal involvement over 8 years. Contractions associated with instability increased and later she could no longer run. No cognitive disorder was described.

The spinal cord and brain MRI, alongside EMG did not show damage to the peripheral motor neuron. The ocular movement test was in favor of a cerebellar syndrome with defected floculli. The lumbar puncture revealed the presence of high anti-GAD antibodies titer in the CSF and plasma. The PET CT/CT did not show any underlying neoplasia. Diagnostic criteria included lower back and limbs stiffness, stimulated by emotions, oculomotor disorders, very good response to Valium.

She received six courses of IVIG from September 2016 to May 2017, with the onset of dysarthria. Rituximab was administered three times within two years. Cyclophosphamide was also administered concomitantly for six months. Azathioprine showed no efficacy while fampridine was not tolerated.

At the end of 2019, the Scale for the Assessment and Rating of Ataxia (SARA) score was at 11. Ocular movements test showed worsening nystagmus. Gabapentin was consequently initiated, then stopped rapidly for inefficacy. The patient was referred to hematology department and treated by AHSCT without major complications in 2021.

Six months Post-AHSCT the patient had better flexibility and mobility, could walk for 2 hours with mild assistance. Dysarthria improved with a SARA at 7.5.

2-years Post-AHSCT speech comprehension improved. She walks alone> hour without assistance. Maintain good balance on uneven ground. SARA was at 6.

Table1: Neurological manifestations: before, 6 months, and 2-years post-AHSCT

Before AHSCT

Post-AHSCT period

2 years post-AHSCT

SARA

11

7.5

6

Speech comprehension

impacted

Good

Very good

Walking

Very hard, robot-like

Better

Very good

Aide for walking

Permanent

Only if uneven ground

None

Stiffness

Severe

Very mild

Almost no stiffness

Flexibility

No flexibility

Better

Very good

Dysarthria

Severe

Better

Very good

Conclusions: AHSCT, as an intensive immunablation followed by was well tolerated and a very effective treatment for this patient with of immunotherapy -refractory SPS. This experience supports its use for refractory stiff-person syndrome.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P085 BRIDGING THERAPY PRIOR TO TREATMENT WITH ANTI-CD19 CAR T CELLS FOR RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA IN CHILDREN AND YOUNG ADULTS - A MULTINATIONAL RETROSPECTIVE ANALYSIS

Maike Breidenbach1, Peter Bader2, Andishe Attarbaschi3, Claudia Rossig4, Roland Meisel5, Markus Metzler6, Marion Subklewe1, Fabian Mueller7, Paul-Gerhard Schlegel8, Irene Teichert von Lüttichau9, Jean-Pierre Bourquin10, Gabriele Escherich11, Gunnar Cario12, Peter Lang13, Ramona Krauss1, Arend von Stackelberg14, Semjon Willier15, Christina Peters3, Tobias Feuchtinger 16,1

1Ludwig Maximilians University Munich, Munich, Germany, 2Goethe University Frankfurt, University Hospital, Frankfurt, Germany, 3St. Anna Children’s Hospital, Vienna, Austria, 4University Children’s Hospital Muenster, Muenster, Germany, 5Medical Faculty, Heinrich-Heine-University, Duesseldorf, Germany, 6University Hospital Erlangen, Erlangen, Germany, 7Friedrich-Alexander University of Erlangen-Nuremberg, Erlangen, Germany, 8Pediatric Hematology and Oncology and Stem Cell Transplantation, University Children’s Hospital Wuerzburg, Wuerzburg, Germany, 9TUM School of Medicine, Children’s Hospital Munich Schwabing, Technical University of Munich, Munich, Germany, 10University Children’s Hospital Zurich, Zurich, Switzerland, 11Clinic of Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 12University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany, 13University Children’s Hospital Tübingen, Tübingen, Germany, 14Charité Universitaetsmedizin, Berlin, Germany, 15University Medical Center Freiburg, Freiburg, Germany, 16Pediatric Hematology, Oncology and Stem Cell Transplantation, University Medical Center Freiburg, Freiburg, Germany

Background: Chimeric antigen receptor (CAR) T cells targeting CD19 are a well-established treatment option for children and young adults suffering from relapsed and/or refractory B-lineage acute lymphoblastic leukemia (ALL). Bridging therapy is the treatment between eligibility and administration of CAR T cells. Bridging has been designed to achieve a low leukemia burden prior to CAR T cell infusion. However, systematic data of bridging therapy are still limited and the effect on outcome, side effects and response to CAR T cell therapy is still poorly understood. With this retrospective, multinational, large-scale study, we strive to understand the impact of low- and high-intensity bridging regimens on a variety of outcome parameters in order to improve the basis for clinical decision making in bridging therapy prior to CAR T cell administration.

Methods: Real-world data were collected from 82 patients receiving 87 CAR T cell therapies from twelve sites in Germany, Austria and Switzerland. Treatments were classified into categories 1) no systemic therapy, 2) low-intensity therapy and 3) high-intensity therapy. Bridging therapies were defined as high-intensity if at least one chemotherapeutic agent of the following was given: cyclophosphamide/ifosfamide, etoposide, anthracyclines or other agents with high toxicity potential (intravenous methotrexate, platinum-based antineoplastic drugs, thiotepa, high-dose cytarabine, fludarabine). Low-intensity bridging therapies comprised the administration of steroids, vincristine, low-dose cytarabine, PEG-asparaginase/Erwinia asparaginase and oral maintenance therapy (mercaptopurine, thioguanine, oral methotrexate, hydroxyurea). The administration of specific chemotherapeutic agents as well as immunotherapies and targeted therapies was assessed. CAR therapies comprised CD19 2nd generation CAR T cell products from commercial and academic providers.

Results: 38 of 87 treatments were classified as high-intensity and 34 as low-intensity bridging regimens. Prior to 14 CAR T cell administrations no systemic bridging therapy was given, 1 of 87 bridging regimens could not be stratified. Between eligibility and apheresis, mostly low-intensity therapy or no systemic therapy was given. Within the period between apheresis and CAR T cell infusion, treatment diversified due to the heterogeneity of the cohort. Patient characteristics are listed in Table 1. Patients receiving a high-intensity bridging therapy had a significantly higher tumor burden at time point of eligibility defined by blasts in bone marrow and by measurement of minimal residual disease (MRD) compared to patients treated with a low-intensity or no systemic bridging therapy. Tumor burden within the two groups converged over the time of bridging therapy. However, until time of lymphodepletion, patients in the high-intensity group had significantly more often bacterial adverse events and mucositis and showed a lower performance status (Karnofsky/Lansky) than patients in the low-intensity/no systemic therapy group. High-intensity bridging therapy did not improve overall or disease-free survival.

Conclusions: In this retrospective cohort data, a low-intensity bridging therapy was associated with equivalent outcome of CAR T cell therapy in terms of overall and disease-free survival, when compared to high-intensity regimens. High-intensity bridging therapy was associated with more mucositis, bacterial adverse events and worsened performance status. Our study suggests that a low-intensity bridging regimen may be preferred whenever tumor burden and disease kinetics allow this treatment strategy.

Disclosure: Bader: Medac: Consultancy, Patents & Royalties: medac, Research Funding; Novartis: Consultancy, Research Funding; Neovii: Research Funding; BMS: Research Funding. Attarbaschi: JazzPharma: Honoraria. Rossig: Amgen, BMS, Novartis, Pfizer, Roche, MSD: Honoraria. Meisel: medac: Consultancy, Research Funding, Speakers Bureau; Novartis: Consultancy, Research Funding, Speakers Bureau; CELGENE BMS: Consultancy, Research Funding, Speakers Bureau; Bluebird Bio: Consultancy, Speakers Bureau; CRISPR Therapeutics: Consultancy, Research Funding, Speakers Bureau; Vertex: Consultancy, Research Funding, Speakers Bureau; Miltenyi Biotech: Research Funding; Gilead/KITE: Research Funding. Subklewe: Incyte Biosciences: Consultancy, Honoraria; AvenCell: Consultancy, Honoraria; Ichnos Sciences: Consultancy, Honoraria; Pfizer: Consultancy, Honoraria, Other: Travel Support, Speakers Bureau; AstraZeneca: Speakers Bureau; Takeda: Consultancy, Honoraria, Research Funding; Seagen: Research Funding; Roche: Consultancy, Honoraria, Other: Travel Support, Research Funding, Speakers Bureau; Novartis: Consultancy, Honoraria, Research Funding, Speakers Bureau; Miltenyi Biotec: Consultancy, Honoraria, Research Funding; Janssen: Consultancy, Honoraria, Research Funding, Speakers Bureau; Gilead/Kite: Consultancy, Honoraria, Other: Travel Support, Research Funding, Speakers Bureau; BMS/Celgene: Consultancy, Honoraria, Research Funding, Speakers Bureau; Amgen: Consultancy, Honoraria, Research Funding; Molecular Partners: Consultancy, Honoraria, Research Funding; GSK: Speakers Bureau; LAWG: Speakers Bureau; Springer Healthcare: Speakers Bureau; AbbVie: Consultancy, Honoraria; Autolus: Consultancy, Honoraria; advesya (CanCell Therapeutics): Consultancy, Honoraria; Genmab US: Consultancy, Honoraria; Interius BioTherapeutics: Consultancy, Honoraria; Nektar Therapeutics: Consultancy, Honoraria; Orbital Therapeutics: Consultancy, Honoraria; Sanofi: Consultancy, Honoraria; Scare: Consultancy, Honoraria. Mueller: Miltenyi, BMS, Novartis, Gilead, Janssen, Incyte, AstraZeneca, Abbvie, Sobi, Beigene: Honoraria; BMS, AstraZeneca, Gilead: Research Funding; AstraZeneca, BMS, Gilead, Janssen, Miltenyi biomedicine, Novartis: Consultancy. Cario: JazzPharma: Speakers Bureau; Servier, Amgen: Research Funding. Peters: AMGEN, Neovii, Jazz: Research Funding; Novartis: Consultancy; Riemser, Medac: Honoraria; AOP Orphan Drugs, Jazz, Neovii: Other: Meeting/Travel grant; Novartis, AMGEN: Membership on an entity’s Board of Directors or advisory committees. Feuchtinger: Servier: Research Funding; Miltenyi Biotec: Research Funding.

4: CAR-based Cellular Therapy – Clinical

P086 SAFETY AND EFFICACY COMPARISON OF HUMANIZED CD19 CAR-T VERSUS BLINATUMOMAB THERAPY FOR RELAPSED/REFRACTORY B-ALL PATIENTS

Kexin Wang 1,2,3,4, Songfu Jiang5, Yongxian Hu1,2,3,4, He Huang1,2,3,4

1Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Zhejiang University Medical Center, Hangzhou, China, 3Institute of Hematology, Zhejiang University, Hangzhou, China, 4Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China, 5The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China

Background: Chimeric antigen receptor T-cells targeting CD19 (CART19) and blinatumomab are 2 major types of novel immunotherapy strategies for patients with relapsed/refractory (R/R) B-cell acute lymphoblastic leukemia (ALL). Choosing which types of immunotherapies to use for individual patients can be a challenge.

Methods: We performed a study to compare the efficacy and safety of blinatumomab versus CART19 for R/R B-ALL. Patient data in CART19 cohort were from the clinical trial (NCT04532268). Data in blinatumomab cohort were from the real-world data. The complete remission (CR) rate, overall survival (OS), leukemia-free survival (LFS), and toxicities were compared in the 2 cohorts.

Results: A total of 106 patients were enrolled including 61 patients in hCART19 cohort and 45 patients in blinatumomab cohort. 36.06% (22/61) patients in hCART19 cohort and 46.67% (21/45) patients in blinatumomab cohort bridged to allogenic hematopoietic stem cell transplantation (allo-HSCT) respectively (P>0.05). The age, gender, prior treatment lines, and extramedullary disease were no statistically different between two cohorts (P>0.05). The tumor burden in hCART19 cohort was higher than blinatumomab (44.60% vs. 2.29%, P<0.001). CR or CR with incomplete count recovery (CRi) rate by day 28 was 96.6% (56/61) in hCART19 cohort with 53 patients having minimal residual disease (MRD) negativity, and 77.8% (35/41) in blinatumomab cohort with 34 patients having MRD negativity, respectively (P = 0.008). With a median follow-up of 10 months, patients receiving allo-HSCT after infusion have similar OS and LFS in two cohorts. For patients not receiving allo-HSCT after infusion, the median LFS was 54 days in blinatumomab cohort and 180 days in hCART19 cohort, the median OS was 300 days in hCART19 cohort and not reached in blinatumomab cohort. In addition, subgroup analysis revealed that patients with higher tumor burden (MRD>20%) before infusion have better CR rate (70.6% vs. 33.3%, P = 0.007) in hCART19 cohort than in blinatumomab cohort, as well as better OS after 30 days from infusion (P = 0.023). For patients with lower tumor burden (MRD≤20%), CR rate and OS are similar in hCART19 and blinatumomab cohort (CR: 83.3% vs. 77.8%, P = 0.514). Patients with higher tumor burden in hCART19 cohort had a higher incidence of severe cytokine release syndrome than blinatumomab (53.7% vs. 31.2%, P = 0.011). The incidence of severe immune effector cell-associated neurotoxicity syndrome in two cohorts were similar (4.9% vs. 0%, P = 0.656). All toxicities were reversible.

Conclusions: Our study suggests that hCART19 has better complete remission efficacy and long-term survival than blinatumomab in R/R B-ALL patients with high tumor burden (MRD>20%). For patients with low tumor burden, blinatumomab has similar therapeutic efficacy with hCART19. Thus, for patients with low tumor burden, blinatumomab is a good choice while for high tumor burden, hCART19 is a good choice.

Disclosure: Nothing to declare.

4: CAR-based Cellular Therapy – Clinical

P087 PREDICTIVE VALUE OF PRE-TREATMENT CIRCULATING TUMOR DNA GENOMIC LANDSCAPE IN PATIENTS WITH R/RMM UNDERGOING ANTI-BCMA CAR-T THERAPY: INSIGHTS FROM TUMOR CELLS AND T CELLS

Rongrong Chen 1, Chunxiang Jin1, Tingting Yang1, Kai Liu1, Mengyu Zhao1, Mingming Zhang1, Pingnan Xiao1, Jingjing Feng1, Ruimin Hong1, Shan Fu1, Jiazhen Cui1, Simao Huang1, Guoqing Wei1, He Huang1, Yongxian Hu1

1The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China

Background: B-cell maturation antigen (BCMA)-directed chimeric antigen receptor T (CAR-T) therapy has demonstrated remarkable efficacy and safety in patients with refractory or relapsed multiple myeloma (R/RMM). Circulating tumor DNA (ctDNA) reportedly exhibits distinct advantages in addressing the challenges posed by tumor heterogeneity in distribution and genetic variations in R/RMM.

Methods: Herein, we performed a comprehensive ctDNA analysis of 108 patients with R/RMM to determine its predictive value for prognosis after CAR-T therapy.

Results: We observed that high ctDNA level (å 1430ng/ml) strongly associated with shorter progression-free survival (PFS)(P = 0.007). Moreover, it was also significantly related to higher percentages of multiple myeloma cells in the bone marrow that was measured using flow cytometry (P = 0.013), lower percentages of CAR-T cells in the peripheral blood at peak (P = 0.037), and higher percentages of CD8+ T cells in the peripheral blood (P = 0.034), which enabled the integration of tumor and T cell effector-mediated factors for assessing treatment failure. Alterations in several individual genes indicated poor outcomes, including IGLL5 (P = 0.004), which plays a significant role in the immune response; IRF4 (P = 0.024), which is involved in NF-κB signaling pathway; and CREBBP (P = 0.041), which participates in the epigenetic regulation of gene expression. Multivariate logistic regression analysis revealed that ERBB4 expression was significantly associated with resistance to CAR-T cell therapy (P = 0.04). Among the five patients with ERBB4 mutation, four failed to achieve a complete response, and all (5/5) of them exhibited progression within 6 months. In addition, patients with two multiple-site ctDNA mutations had poor outcomes, with PFS of less than 6 months (P <0.001).

Conclusions: Finally, we built a ctDNA-based risk model that serves as an adjunct non-invasive measure of substantial tumor burden and a prognostic genetic feature that can assist in predicting the response to anti-BCMA CAR-T therapy.

Clinical Trial Registry: The study was conducted in accordance with the tenets of the Declaration of Helsinki and registered in the Chinese Clinical Trial Registry (ChiCTR2100046474) and National Clinical Trial (NCT04670055, NCT05430945).

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P088 FINAL RESULTS OF PROSPECTIVE CLINICAL TRIAL EVALUATING OUTPATIENT ADMINISTRATION OF AXICABTAGENE CILOLEUCEL IN HIGH-GRADE B CELL LYMPHOMA

Bhagirathbhai Dholaria1, Shakthi Bhaskar1, Vivek Patel1, Eden Biltibo1, Salyka Sengsayadeth1, Andrew Jallouk1, James Jerkins1, Brittney Baer1, Nur Ali1, David Morgan1, Muhamed Baljevic1, Bipin Savani1, Adetola Kassim1, Olalekan Oluwole 1

1Vanderbilt University Medical Center, Nashville, United States

Background: Pivotal trials of Axicabtagene Ciloleucel (axi-cel) were conducted in the inpatient setting because of the high rate of severe Cytokine Release Syndrome (CRS) and Immune effector Cell-Associated Neurotoxicity Syndrome (ICANS).

Methods: We devised a single-center non-randomized prospective trial (NCT05108805) to evaluate the feasibility and safety of outpatient administration of axi-cel. The primary endpoint of the study was to explore the safety and feasibility of outpatient Axi-cel. Eligible pts with R/R DLBCL per axi-cel FDA package label and met ZUMA 1 criteria were included in the study. They were fitted with wearable devices to measure temp, BP, Pulse-ox and heart rate for continuous remote monitoring. Pts were seen in-person once daily and had remote video telehealth evaluation at 16:00 and 22:00 daily through day 14. Fever up to 102F without other symptoms was managed in outpatient clinic. Here we report the final outcomes of complete study cohort.

Results: Twenty consecutive patients (pts) were treated from January 2022 to October 2023. Data cut off was December 16, 2023. The median pt age was 68 (range: 35-81) yrs and stage III-IV disease in 18 (90%) pts. Before lymphodepletion, baseline LDH was 251 U/L. The median number of therapies was 2.5 (range: 1-4), 10 (50%) pts ≥ 3 lines of therapies. The estimated median follow-up was 178 (95% CI: 81-258) days. Prophylactic dexamethasone 10mg/day, days 0-2 was given to 18 (90%) pts. 18 (90%) pts remained outpatient at least 72 hours after axi-cel administration, and one (5%) pt remained outpatient through day 30. The median time from day 0 to hospitalization was 3 (range 1-6) days and the median hospital stay was 5.5 (range 0-21) days. The reason for hospitalization was CRS in all admitted pts. Overall CRS was reported in 19 (95%) pts (grade 1 = 11, grade 2 = 8), ICANS in 8 (40%) pts (grade 1 = 4; grade 3 = 4). Time onset from day 0 to CRS was 3 (range 1-6) days and ICANS was 5 (range: 4-10) days. No pt had grade ≥3 CRS or grade ≥4 ICANS or treatment-related death by last follow-up. The median duration of CRS and ICANS were 3 days, each (Table 1).

Two (10%) pts required ICU admission (COPD exacerbation, SVT due to CRS) through day 30. By day 30, 14 (70%) pts achieved CR. There were 3 (15%) pts with PD/refractory disease at the last follow-up with one death from relapsed disease. Median EFS or OS was not reached by last follow up. Six-month estimated OS was 91% (95% CI: 0.74-1.0) and EFS was 81% (95% CI: 0.62-1.0). Our results were favorable compared to ZUMA 1 which reported median hospitalization of 15 days, grade ≥ 3 CRS and ICANS in 13% and 28% of pts, respectively and 6-months PFS of 49%.

Table 1: Outcomes of patients

Outcomes

N = 20 (%)

Day 30 responses

CR

14 (70%)

SD

4 (20%)

PD

2 (10%)

Day 90 responses (N = 17)

CR

11 (65%)

PR

1 (6%)

SD

2 (12%)

PD

3 (18%)

Best response

CR

17 (85%)

SD

1 (5%)

PD

2 (10%)

Median Time to hospitalization from day 0 (N = 19), days

3 (1- 6)

Median inpatient days by D + 30, days

5.5 (0-21)

Pts with ICU admission by D + 30, N

2 (10%)

CRS

Grade 0

1 (5%)

Grade 1

11 (55%)

Grade 2

8 (40%)

Median time to CRS from day 0, days

3 (1-6)

Median duration of CRS, days

3 (1-5)

ICANS

Grade 0

12(60%)

Grade 1

4 (20%)

Grade 3

4 (20%)

Median time to ICANS from day 0, days

5 (4-10)

Median duration of ICANS, days

3 (1-9)

Tocilizumab given

17 (85%)

Median doses of tocilizumab

2 (0-3)

Prophylactic dexamethasone

18 (90%)

Systemic steroids in addition to prophylactic dexamethasone

8 (40%)

Conclusions: This prospective feasibility trial confirmed that axi-cel can be safely administered in the outpatient settings with prophylactic steroids and remote monitoring with wearable devices without compromising safety or efficacy of CAR T therapy. Longer follow-up data will be presented at the meeting.

Disclosure: Bhagirathbhai Dholaria: Institutional research funding: Janssen, Angiocrine, Pfizer, Poseida, MEI, Orcabio, Wugen, Allovir, NCI, Atara, Gilead, Molecular templates, BMS, AstraZeneca, Adicet. Consulting/Advisor: MJH, Janssen, Pluri Biotech, BOXER CAPITAL, Ellipsis pharma, Lumanity, Autolus, Acrotech, ADC therapeutics, Gilead

Olalekan Oluwole: Consultancy and advisory board for: Pfizer, Kite, Gilead, AbbVie, Janssen, TGR, ADC, Novartis, Caribou, Cargo, Epizyme, Nektar, Autolus, Allogene. Institution funding: Kite, Pfizer, Daichi Sankyo, Allogene. Honoraria: Pfizer, Gilead

Eden Biltibo: BeiGene (consulting)

Andrew Jallouk: Kite-Gilead (consulting)

Rest of the authors declare no relevant COI.

4: CAR-based Cellular Therapy – Clinical

P089 EUPLAGIA-1: SEVEN-DAY VEIN-TO-VEIN POINT-OF-CARE MANUFACTURED GLPG5201 ANTI-CD19 CAR-T CELLS DISPLAY EARLY PHENOTYPES IN RELAPSED/REFRACTORY CLL INCLUDING RT

Esmée P. Hoefsmit 1, Sandra Blum2, Claire Vennin1, Kirsten Van Hoorde3, Sergi Betriu4, Leticia Alserawan4, Julio Delgado4, Nadia Verbruggen5, Anna D.D. van Muyden1, Henriëtte Rozema1, Ruiz Astigarraga1, Margot J. Pont1

1Galapagos BV, Oegstgeest, Netherlands, 2Galapagos GmbH, Basel, Switzerland, 3Open Analytics NV, Antwerp, Belgium, 4Hospital Clínic de Barcelona, Barcelona, Spain, 5Galapagos NV, Mechelen, Belgium

Background: The efficacy of chimeric antigen receptor T-cell (CAR-T) therapies relies in part on the preservation of early T-cell phenotypes and robust expansion in patients. Our decentralized and automated point-of-care (PoC) manufacturing platform allows administration of fresh autologous GLPG5201 CAR-T cells with a vein-to-vein time of 7 days.

Methods: Euplagia-1 (CTIS: 2022-501686-47-00) is an ongoing Phase 1/2 dose escalation study of PoC manufactured GLPG5201, an anti-CD19 4-1BB/CD3z CAR-T cell product, in patients with relapsed/refractory chronic lymphocytic leukemia or small lymphocytic lymphoma (CLL/SLL), including Richter Transformation (RT). Feasibility of PoC manufacturing, T-cell phenotyping in apheresis starting material and in final product as well as correlation between T-cell phenotypes and in-patient pharmacokinetics were evaluated. Data cut-off was 06 September 2023. T-cell phenotyping was assessed using exploratory flow cytometry (n = 12 evaluable patients at data cut-off) and pharmacokinetics were determined by qPCR (n = 13 evaluable patients at data cut-off).

Results: All phase 1 GLPG5201 clinical batches (n = 15) were successfully manufactured at the PoC facility and infused as a fresh product at one clinical site. Encouraging clinical efficacy was observed, with a 93% best objective response rate and a 57% complete response rate in the intention-to-treat analysis set (clinical data submitted in a separate abstract). The median vein-to-vein time was 7 days (range 7–14), with 80% of patients (12/15) receiving GLPG5201 in 7 days. Characterization of GLPG5201 showed an increase in CD4+ and CD8+ CAR-T cells with early phenotypes (naïve, stem cell memory [TN/SCM] and central memory [TCM]) compared to apheresis starting material. The median increase in percentage of early phenotypes (i.e. TN/SCM + TCM) in CD4+ and CD8+ CAR-T cells was 24.0 (increased in 8/10 patients) for CD4+ T-cells and 52.1 (increased in 10/10 patients) for CD8+ T-cells. Robust in vivo expansion of GLPG5201 was detected in all patients treated with both dose levels (DL1: 35×106 CAR+ T-cells, n = 6; DL2: 100×106 CAR+ T-cells, n = 7). Median peak expansion (Cmax) was 4.4×105 copies/μg DNA (range 0.52×105 – 9.2×105), median time to peak expansion (Tmax) was 14 days (range 9 – 20) and median area under the curve from day 0–28 (AUCd0-28) was 6.1×106 copies/μg DNA x days (range 0.63×106 – 9.6×106). Higher exposure (AUCd0-28) was observed for patients infused with DL2 (median AUCd0-28 8.8×106 copies/μg DNA x days) compared to DL1 (median AUCd0-28 3.5×106 copies/μg DNA x days). Expansion and exposure were similar for patients with CLL and patients with CLL + RT. 8/10 patients had measurable GLPG5201 in peripheral blood at week 14 post-infusion. Persisting CAR-T cells were detected up to 15 months post-infusion. Moreover, abundance of both TN/SCM CD4+ and CD8+ CAR-T cells in the final product positively correlated with CAR-T cell exposure in patients (Spearman rank correlation (95% CI) for CD4+: 0.791 (0.27-0.99) and for CD8+: 0.791 (0.28-0.98); n = 11).

Conclusions: The Galapagos PoC manufacturing platform enables a 7-day vein-to-vein infusion. Early phenotype CAR-T cells were enriched in the final product compared to apheresis starting material. GLPG5201 demonstrated robust expansion and durable persistence in CLL and CLL + RT patients post-infusion.

Clinical Trial Registry: CTIS: 2022-501686-47-00

Disclosure: Esmée P. Hoefsmit: Employee of Galapagos BV, Netherlands

Sandra Blum: Employee of Galapagos GmbH, Switzerland

Claire Vennin: Employee of Galapagos BV, Netherlands

Kirsten Van Hoorde: Employee of Open Analytics NV, Belgium

Sergi Betriu: Nothing to declare

Leticia Alserawan: Nothing to declare

Julio Delgado: Nothing to declare

Nadia Verbruggen: Employee of, and shareholder in, Galapagos NV, Belgium

Anna D. D. van Muyden: Employee of Galapagos BV, Netherlands

Henriëtte Rozema: Employee of Galapagos BV, Netherlands

Ruiz Astigarraga: Employee of Galapagos BV, Netherlands

Margot J. Pont: Employee of Galapagos BV, Netherlands

The study was funded by Galapagos NV (Mechelen, Belgium).

4: CAR-based Cellular Therapy – Clinical

P090 INCIDENCES AND FACTORS ASSOCIATED WITH EARLY HEMATOTOXICITY AFTER CAR T-CELL THERAPY ASSESSED BY EHA/EBMT ICAHT CRITERIA

Emily Liang 1,2, Aya Albittar1, Andrew Portuguese1,2, Jennifer Huang1,2, Natalie Wuliji1,2, Qian Wu1, Joseph De Los Reyes1,2, Nikki Pin1, Aiko Torkelson1, Delaney Kirchmeier1, Abigail Chutnik1, Barbara Pender1, Joshua Hill1,2, Noam Kopmar1,2, Rahul Banerjee1,2, Andrew Cowan1,2, Damian Green1,2, Ajay Gopal1,2, Christina Poh1,2, Mazyar Shadman1,2, Alexandre Hirayama1,2, Brian Till1,2, Erik Kimble1,2, Lorenzo Iovino1,2, Aude Chapuis1,2, Folashade Otegbeye1,2, Ryan Cassaday1,2, Filippo Milano1,2, Cameron Turtle3, David Maloney1,2, Jordan Gauthier1,2

1Fred Hutchinson Cancer Center, Seattle, United States, 2University of Washington, Seattle, United States, 3University of Sydney, Sydney, Australia

Background: The observation that distinct patterns of hematologic toxicity are not adequately captured by CTCAE grading led to the recently developed EHA/EBMT immune effector cell-associated hematotoxicity (ICAHT) grading system (Rejeski et al, Blood, 2023). We applied the ICAHT grading system to patients undergoing CAR T-cell therapy at a large institution in the US and assess factors associated with grade ≥3 ICAHT.

Methods: Adults who underwent CAR T-cell therapy for hematologic malignancies with commercial or investigational products at our center between 2013 and 2023 were included (n = 454). Grading of early hematotoxicity (day-0-30 after CAR T-cell cell infusion) was automated using the heatwaveR package in R per ICAHT criteria detailed in Rejeski et al, Blood, 2023. Associations with 50 patient, disease-related, and laboratory factors, and ICAHT grade ≥3 were modeled using univariate and multivariable logistic regression.

Results: The most common disease types were aggressive non-Hodgkin lymphoma (n = 216; 48%), acute lymphoblastic leukemia (n = 82; 18%), and multiple myeloma / plasma cell leukemia (n = 61; 13%) (Table). The most common CAR T-cell products were investigational CD19 CAR T-cell products (n = 180; 40%), axicabtagene ciloleucel (n = 114; 25%), and lisocabtagene maraleucel (n = 58; 13%).

Incidences of early ICAHT grades 1, 2, 3, and 4 were 141 (31%), 177 (39%), 47 (10%), and 30 (7%), respectively. Patients with grades 3-4 ICAHT had worse overall survival compared to those with grade 0-2 ICAHT (median of 6 s. 18 months, p < 0.001).

Baseline patient factors associated with grade 3-4 ICAHT included ALL (reference: aggressive NHL, OR = 3.0, 95% CI, 1.7-5.6, p < 0.001) and age (OR = 0.97, 95% CI, 0.96-0.99, p = 0.003). Pre-lymphodepletion (LD) laboratory factors associated with grade 3-4 ICAHT included ANC (OR = 0.18 per log10, 95% CI, 0.10-0.30, p < 0.001), lactate dehydrogenase (OR = 7.4 per log10U/L, 95% CI, 3.3-17.4, p < 0.001), C-reactive protein (CRP; OR = 5.9 per log10mg/L, 95% CI, 2.8-14.0, p < 0.001), and ferritin (OR = 6.5 per log10mg/L, 95% CI, 3.0-16.0, p < 0.001). Peak CRP (OR = 12.8, 95% CI, 5.3-33.9, p < 0.001) and ferritin (OR = 5.6, 95% CI, 3.7-8.7, p < 0.001) after CAR T-cell infusion were strongly associated with grade 3-4 ICAHT. In a multivariable model including disease type, pre-LD ANC, pre-LD LDH, and peak CRP, pre-LD ANC (OR = 0.22, 95% CI, 0.11-0.42, p < 0.001), pre-LD LDH (OR = 5.0, 95% CI, 1.7-14.6, p = 0.003), and peak CRP (OR = 6.7, 95% CI, 2.7-18.4) remained independently associated with grade 3-4 ICAHT.

Table. Demographic, disease, and treatment characteristics (N = 454)

Age at infusion, median (range)

60 (50, 68)

Male sex

286 (63%)

Prior HCT

186 (41%)

Disease

Aggressive NHL

216 (48%)

ALL

82 (18%)

MM/PCL

61 (13%)

Indolent NHL

53 (12%)

CLL

42 (9%)

CAR T-cell product

Investigational CD19 CAR T-cell product

180 (40%)

Commercial CD19 CAR T-cell product with CD28 costimulatory domain (axi-cel, brexu-cel)

143 (31%)

Commercial CD19 CAR T-cell product with 4-1BB costimulatory domain (liso-cel, tisa-cel)

70 (15%)

Commercial BCMA CAR T-cell product (cilta-cel, ide-cel)

61 (13%)

Conclusions: We found that 17% of patients undergoing CAR T-cell therapy at our center developed grade 3-4 hematotoxicity by EHA/EBMT ICAHT criteria. Grade 3-4 ICAHT was associated with worse OS. Multivariable logistic regression of factors associated with grade 3-4 ICAHT identified pre-LD ANC, pre-LD LDH, and peak CRP as independent predictors of grade 3-4 ICAHT.

Disclosure: Joshua Hill: allovir: Consultancy, Research Funding; moderna: Consultancy; deverra: Research Funding.

Rahul Banerjee: BMS: Consultancy; Janssen: Consultancy; Genentech: Consultancy; SparkCures: Consultancy; Sanofi: Consultancy; Caribou: Consultancy; Pfizer: Consultancy; Pack Health: Research Funding.

Andrew Cowan: BMS, Adaptive: Consultancy; Adaptive Biotechnologies, Harpoon, Nektar, BMS, Janssen, Sanofi, Abbvie: Research Funding.

Damian Green: Cellectar Biosciences: Research Funding; SpringWorks Therapeutics: Research Funding; Celgene: Consultancy; GlaxoSmithKline: Membership on an entity’s Board of Directors or advisory committees; Sanofi: Research Funding; Janssen Biotech: Consultancy, Research Funding; Ensoma: Consultancy; Seattle Genetics: Consultancy, Research Funding; Juno Therapeutics A BMS Company: Patents & Royalties, Research Funding.

Ajay Gopal: Compliment Corporation: Current holder of stock options in a privately-held company; Incyte, Kite, Morphosys/Incyte, ADCT, Acrotech, Merck, Karyopharm, Servier, Beigene, Cellectar, Janssen, SeaGen, Epizyme, I-Mab bio, Gilead, Genentech, Lilly, Caribou, Fresenius-Kabi: Consultancy; Merck, I-Mab bio, IgM Bio, Takeda, Gilead, Astra-Zeneca, Agios, Janssen, BMS, SeaGen, Teva, Genmab: Research Funding.

Christina Poh: BeiGene: Consultancy; Seattle Genetics: Consultancy; Incyte: Research Funding; Acrotech: Consultancy.

Mazyar Shadman: Fate Therapeutics: Consultancy; Genmab: Consultancy, Research Funding; Vincerx: Research Funding; MorphoSys/Incyte: Consultancy, Research Funding; Eli Lilly: Consultancy; Bristol Myers Squibb: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Kite, a Gilead Company: Consultancy; AbbVie: Consultancy, Research Funding; Mustang Bio: Consultancy, Research Funding; ADC therapeutics: Consultancy; BeiGene: Consultancy, Research Funding; AstraZeneca: Consultancy, Research Funding; Pharmacyclics: Consultancy, Research Funding; Janssen: Consultancy; MEI Pharma: Consultancy; Regeneron: Consultancy; TG Therapeutics: Research Funding.

Alexandre Hirayama: Novartis: Honoraria; Bristol Myers Squibb: Honoraria, Research Funding; Nektar Therapeutics: Honoraria, Research Funding; Juno Therapeutics, a Bristol Myers Squibb Company: Research Funding.

Till: Mustang Bio: Consultancy, Patents & Royalties, Research Funding; BMS/Juno Therapeutics: Research Funding; Proteios Technology: Consultancy, Current holder of stock options in a privately-held company.

Kimble: Juno/BMS: Research Funding.

Lorenzo Iovino: Mustang Bio: Current equity holder in publicly-traded company.

Aude Chapuis: Juno Therapeutics: Research Funding.

Ryan Cassaday: Amgen: Consultancy, Honoraria, Research Funding; Merck: Research Funding; Incyte: Research Funding; Autolus: Membership on an entity’s Board of Directors or advisory committees; Pfizer: Consultancy, Honoraria, Research Funding; Servier: Research Funding; Vanda Pharmaceuticals: Research Funding; Jazz: Consultancy, Honoraria; Kite/Gilead: Consultancy, Honoraria, Research Funding; PeproMene Bio: Membership on an entity’s Board of Directors or advisory committees; Seagen: Other: Spouse was employed by and owned stock in Seagen within the last 24 months.

Filippo Milano: ExCellThera Inc.: Research Funding.

David Maloney: A2 Biotherapeutics: Consultancy, Current holder of stock options in a privately-held company, Honoraria, Other: Member of the Scientific Advisory Board; Juno Therapeutics: Consultancy, Honoraria, Patents & Royalties: Rights to royalties from Fred Hutch for patents licensed to Juno Therapeutics/BMS, Research Funding; Janssen: Consultancy, Honoraria; Legend Biotech: Consultancy, Honoraria, Research Funding; Mustang Bio: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Lyell Immunopharma: Other: Member, CAR T Steering Committee; Incyte: Consultancy, Honoraria; Gilead Sciences: Consultancy, Honoraria, Other: Member, Scientific Review Committee, Research Scholars Program in Hematologic Malignancies; Kite, a Gilead Sciences: Consultancy, Honoraria, Research Funding; Pharmacyclics: Consultancy, Honoraria; Umoja: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Other: Participation on a Data Safety Monitory Board, Research Funding; Genentech: Consultancy, Honoraria, Other: Chair and Member of the Lymphoma Steering Committee; MorphoSys: Consultancy, Honoraria; Bristol Myers Squibb: Consultancy, Honoraria, Other: Member of the JCAR017 EAP-001 Safety Review Committee and Member, CLL Strategic Council, Member of the JCAR017-BCM-03 Scientific Steering Committee under BMS, Research Funding; Amgen: Consultancy, Honoraria; Navan Technologies: Consultancy, Honoraria, Other: Member of the Scientific Advisory Board; Bioline Rx: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Other: Participation on a Data Safety Monitory Board; Fred Hutch: Other: rights to royalties for patents licensed to Juno; Navan Technologies: Current holder of stock options in a privately-held company; Chimeric Therapeutics: Other: Member of the Scientific Advisory Board; ImmPACT Bio: Other: Member, Clinical Advisory Board, CD19/CD20 bi-specific CAR-T Cell Therapy Program; Interius: Other: Member, Clinical Advisory Board.

Jordan Gauthier: Kite Pharma: Consultancy, Honoraria; MorphoSys: Consultancy, Research Funding; Angiocrine Bioscience: Research Funding; Century Therapeutics: Other: Independent data review committee; Celgene (a Bristol Myers Squibb company): Research Funding; Legend Biotech: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Juno Therapeutics (a Bristol Myers Squibb company): Research Funding; Sobi: Consultancy, Honoraria, Research Funding.

4: CAR-based Cellular Therapy – Clinical

P091 COMPARISON OF IMMUNOPHENOTYPIC AND FUNCTIONAL PROPERTIES OF ANTI-CD19 CAR-T CELL PRODUCTS MANUFACTURED USING CLINIMACS PRODIGY AND G-REX PLATFORMS

Ekaterina Malakhova1, Dmitriy Pershin 1, Viktoria Vedmedskaia1, Mariia Fadeeva1, Elena Kulakovskaya1, Oyuna Lodoeva1, Tatiana Sozonova1, Elvira Musaeva1, Yakov Muzalevskii1, Alexei Kazachenok1, Vladislav Belchikov1, Anastasia Melkova1, Larisa Shelikhova1, Olga Molostova1, Michael Maschan1

1Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Moscow, Russian Federation

Background: The use of anti-CD19 CAR-T cell therapy for treating B cell malignancies has shown impressive results in recent years. Today, there are several cell culture platforms that allow the manufacturing of cell products for subsequent clinical use; the issue of choosing a platform for the manufacturing of CAR-T remains relevant. This study compares the immunophenotypic and functional properties of final anti-CD19 cell products manufactured using the CliniMACS Prodigy and G-Rex platforms.

Methods: A total of 73 cell products were obtained, 49 of which were produced according to сGMP using the automated CliniMACS Prodigy® system (Miltenyi Biotec) and 24 using the G-Rex®10M-CS bioreactor (Wilson Wolf). Autologous and haploidentical cell sources were used for the manufacturing (no statistical differences in the cell composition in the start product of the processes). At the start of manufacturing with CliniMACS Prodigy® system, 100 million CD4+ and CD8 + T cells were selected at CliniMACS Prodigy, after which T lymphocytes were activated, transduced, and expanded in an automated closed system. For production in the G-Rex bioreactor, 5 million CD4+ and CD8 + T cells were selected by sorting using MACSQuant Tyto (Miltenyi Biotec). Lentiviral Vector aCD19 CAR was used for T cell transduction in both platforms (without using transduction enhancers and spinoculation). During the stages of manufacturing, the efficiency of transduction, composition, immunophenotypic and functional properties of cell products were determined using flow cytometry. Functional testing included TNFα secretion assay and degranulation assay (CD107a + ) upon incubation with the CD19+ cell line JeKo-1.

Results:

CliniMACS Prodigy

(final product)

G-Rex

(final product)

P value

CliniMACS Prodigy

(start product)

G-Rex

(start product)

P value

CD8+ Naïve

n/a

45.7% (0.7-91.9%), N = 46

38.1% (14.4-80.9%), N = 24

ns

CD4+ Naïve

n/a

37.7% (3.9-75.1%), N = 46

49.3% (16.7-78.7%), N = 24

ns

CD8+ Central memory

86.1% (6.4-99.1%), N = 49

69.0% (22.0-89.0%), N = 19

0.0006

5.5% (0.9-23.8%), N = 46

6.4% (0.6-27.5%), N = 24

ns

CD4+ Central memory

83.4% (3.9-98.8%), N=49

72.0% (39.0-88.0%), N=19

0.0093

32.6% (13.7-64.4%), N=46

31.7% (16.7-53.1%), N=24

ns

CD8+ Effector memory

12.0% (0.7-85.5%), N=49

31.0% (10.0-76.0%), N=19

0.0001

22.6% (0.7-85.3%), N=46

22.7% (4.8-56.1%), N=24

ns

CD4+ Effector memory

16.0% (1.2-89.9%), N=49

28.0% (12.0-58.0%), N=19

0.0032

22.5% (5.0-73.7%), N=46

10.7% (4.0-45.3%), N=24

ns

CD8 + TEMRA

n/a

18.9% (2.4-62.9%), N=46

18.1% (3.7-55.2%), N=24

ns

CD4 + TEMRA

n/a

0.7% (0.2-10.7%), N=46

1.0% (0.1-5.4%), N=24

ns

CD8 + CD279+

7.7% (0.5-20.9%), N=47

7.6% (0.9-30.9%), N=16

ns

24.9% (0.6-67.9%), N=45

23.9% (6.8-46.0%), N=21

ns

CD4 + CD279+

13.6% (1.5-43.6%), N=48

18.3% (0.5-40.7%), N=16

ns

29.2% (4.1-62.2%), N=45

20.0% (7.0-44.8%), N=21

0.0341

CD8 + TIGIT+

7.6% (1.0-33.4%), N=35

7.7% (2.0-20.9%), N=22

ns

22.9 (2.9-63.7%), N=34

32.3% (4.8-59.1%), N=23

ns

CD4 + TIGIT+

4.5% (0.7-16.5%), N=34

2.9% (1.0-9.4%), N=22

0.0348

17.7% (7.1-48.9%), N=35

14.6% (5.4-24.0%), N=23

0.0220

Degranulation assay for CD8 + CAR-T cells

39.4% (22.5-57.0%), N=17

9.6% (2.0-22.9%), N=24

<0.0001

n/a

n/a

n/a

Degranulation assay for CD4 + CAR-T cells

33.4% (16.8-51.5%), N=17

13.5% (2.0-30.8%), N=24

<0.0001

n/a

n/a

n/a

TNFα secretion assay for CD8 + CAR-T cells

33.6% (14.2-100%), N=35

3.1% (0.8-10.9%), N=24

<0.0001

n/a

n/a

n/a

TNFα secretion assay for CD4 + CAR-T cells

68.8% (25.1-87.4%), N=34

10.6% (4.6-24.3%), N=24

<0.0001

n/a

n/a

n/a

A comparison of the immunophenotypic and functional properties of final cell products produced using the CliniMACS Prodigy or G-Rex platforms is presented in Table. Using both platforms, we obtain a stable product with sufficient expansion and transduction of T cells for subsequent CAR-T therapy. There was a tendency for the Tcm population to predominate in the final cell product, as well as low expression of cellular exhaustion markers in products manufactured by both methods. However, transduction efficiency was statistically significantly higher in products manufactured using the CliniMACS Prodigy system (p < 0.0001), and there was a greater predominance of Tcm over Tem in these products compared to products manufactured in bioreactor G -Rex. All products showed functional activity against the target cell line, but in products obtained using CliniMACS Prodigy it was more pronounced.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (11)

Conclusions: Both CliniMACS Prodigy and G-Rex platforms can be used to produce a high quality CAR-T cell product. Despite lower transduction efficiency and T cell counts in the final cell products in the G-Rex bioreactor, this was sufficient for subsequent CAR-T therapy. Regarding the differences in effector function found as well as the composition of Tcm/Tem, further analysis of the correlation with clinical response and CRS is required.

Clinical Trial Registry: Clinical Trial (NCT03467256)

Disclosure: Nothing to declare.

4: CAR-based Cellular Therapy – Clinical

P092 ALLOGENEIC HSCT AFTER CAR T-CELL THERAPY HAD DELAYED PLATELET ENGRAFTMENT, HIGHER RISK OF CYTOMEGALOVIRUS VIRUS VIREMIA AND THROMBOTIC MICROANGIOPATHY COMPARED TO CHEMOTHERAPY

Luxin Yang 1, Xiaoyu Lai1, Lizhen Liu1, Jimin Shi1, Yanmin Zhao1, Jian Yu1, Huarui Fu1, Yongxian Hu1, Mingming Zhang1, He Huang1, Yi Luo1

1The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China

Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) after Chimeric antigen receptor-modified T-cell (CAR T-cell) may achieve durable remission and long-term survival for relapsed/refractory acute lymphoblastic leukemia. However, reliable comparison of treatment-related toxicity of allo-HSCT after CAR-T therapy and traditional chemotherapy lacked.

Methods: Consecutive patients who received CAR T-cells therapy then underwent allo-HSCT during June 1st, 2016 and January 1st, 2023 were included. Propensity score matching was conducted between the CAR T-cells and chemotherapy cohort. The matching process incorporated disease, patient age, minimal residual disease (MRD) before HSCT, time interval from diagnosis to HSCT, donor resource, donor age and sex with a 1:1 ratio.

Results: Fifity-five patients with B cell acute lymphoblastic leukemia (ALL) and four patients with T cell ALL were included in the CAR T cohort. Most patients (86.4%) received anti-CD19 CAR T, four patients received anti-CD19/CD22 CAR T and four patients received anti-CD7 CAR T. All the patients achieved complete remission before transplantation. The median time from CAR T infusion to allo-HSCT was 84 (43-512) days. Thirty-four (57.6%) patients experienced grade 1-2 cytokine release syndrome (CRS) and twenty-five (42.4%) patients experienced grade 3-4 CRS. The median age was 28 (12-67) and 17.1(16-60) years in the CAR T and chemotherapy cohort, respectively. The patient age, gender, disease, minimal residual disease at HSCT, donor resource, donor gender and age were quite comparable between the two cohorts. The median time of neutrophil engraftment were 13 (7-20) and 12 (8-19) days in the CAR T and chemotherapy cohort. The CAR-T cohort had longer platelet engraftment time than chemotherapy cohort (15 days versus 13 days, p=0.042). Patients in the CAR T cohort had higher cumulative incidence of hemorrhagic cystitis (37.3% versus 15.3%, p=0.005). The 100-days cumulative incidence of septicemia of two cohorts were low (3.4% versus 5.1%, p=0.47). The 100-days cumulative incidence of EB virus viremia was 50.8% for the CAR T cohort and 40.7% for chemotherapy cohort (p= 0.48). The cumulative incidence of CMV virus viremia was higher for patients who underwent allo-HSCT after previous CAR T than chemotherapy (76.3% versus 62.7%, p=0.01). In the CAR T cohort, transplant-associated thrombotic microangiopathy (TA-TMA) occurred in four patients. No one in the chemotherapy cohort underwent TA-TMA. The incidence of TA-TMA was higher in the CAR T cohort (76.3% versus 62.7%, p=0.01). There is no difference in the cumulative incidence of grades III-IV acute graft-versus-host disease (GVHD) between cohorts (10.2% versus 16.9%, p=0.29). The cumulative incidence of any grades chronic GVHD in the CAR T cohort was 38.2%, which was significantly higher than 15.3% in the chemotherapy cohort (p=0.034). The incidence of moderate to severe chronic GVHD was comparable between cohorts. There was no difference between two cohorts in 2-year overall survival, progression-free survival, incidence of relapse and non-relapse mortality (NRM).

Conclusions: Allo-HSCT after CAR-T therapy had higher risk of transplant related toxicity compared to traditional chemotherapy. The incidence of survival, NRM, acute GVHD and moderate to severe chronic GVHD was comparable.

Disclosure: The authors declare that they have no competing interests.

4: CAR-based Cellular Therapy – Clinical

P093 AUTOMATED GRADING OF IMMUNE EFFECTOR CELL-ASSOCIATED HEMATOTOXICITY

Emily Liang 1,2, Kai Rejeski3, Sandeep Raj3, Aya Albittar1, Jennifer Huang1,2, Andrew Portuguese1,2, Natalie Wuliji1,2, Qian Wu1, Aiko Torkelson1,2, Delaney Kirchmeier1,2, Abigail Chutnik1, Barbara Pender1, Joshua Hill1,2,2, Noam Kopmar1,2, Rahul Banerjee1,2, Andrew Cowan1,2, Christina Poh1,2, Mazyar Shadman1,2, Alexandre Hirayama1,2, Erik Kimble1,2, Lorenzo Iovino1,2, Roni Shouval3, Jordan Gauthier1,2

1Fred Hutchinson Cancer Center, Seattle, United States, 2University of Washington, Seattle, United States, 3Memorial Sloan Kettering Cancer Center, New York, United States

Background: Following CAR T-cell therapy, hematologic toxicity has emerged as a substantial limitation due to associated infections and increased morbidity. Recognizing unique patterns of hematologic toxicity beyond the scope of CTCAE criteria prompted the development of the EHA/EBMT immune effector cell-associated hematotoxicity (ICAHT) grading system (Rejeski et al, Blood, 2023). Manually assessing longitudinal data is prone to subjectivity and demands extensive labor. Here we use a computational approach to automate the process of ICAHT grading to improve efficacy and accuracy of grading CAR T-cell-associated hematotoxicity.

Methods: The heatwaveR package (Schlegel and Smit, Journal of Open Source Software, 2023; https://robwschlegel.github.io/heatwaveR/), implemented through the R programming language, was originally created to identify marine heatwaves and coldspells. Utilizing this package requires a data table with the following columns: subject IDs, dates (multiple rows per patient), the laboratory value of interest (in this case, ANC) for each date, and the threshold above or below which events should be detected (e.g., 500 or 100 cells/μL).

Using the detect_event() function in heatwaveR, we can expediently identify instances where ANC decreases below the threshold and the duration of each instance, defined as the number of consecutive days during which the ANC is below the threshold (neutropenia “streak”). We also can set the minimum duration of streaks and whether to join streaks across a pre-specified number of days. We set the minimum streak duration to 1 day and required streaks to be joined if ANC did not recover above 500 or 100 for ≥3 consecutive days between streaks, based on the CIBMTR criteria for neutrophil engraftment.

Results: We applied heatwaveR to our longitudinal ANC dataset of patients undergoing CAR T-cell therapy at our center (Table). In this dataset, the most common disease types were aggressive non-Hodgkin lymphoma (n= 216; 48%), acute lymphoblastic leukemia (n= 82; 18%), and multiple myeloma / plasma cell leukemia (n= 61; 13%). The most common CAR T-cell products were investigational CD19 CAR T-cell products (n= 180; 40%), axicabtagene ciloleucel (n= 114; 25%), and lisocabtagene maraleucel (n= 58; 13%). We set thresholds of 500 and 100 to represent the cutoffs for ICAHT grading. For each patient, we extracted the maximum “streak” where ANC ≤ 500 and ≤ 100 cells/μL, then applied the ICAHT guidelines for duration and depth of neutropenia.

Thirteen percent of patients did not meet ICAHT criteria. In the remaining patients, the majority displayed grade 1 (31%) or grade 2 (39%) ICAHT. We noted grade 3 and grade 4 ICAHT in 10% and 7% of cases, respectively.

Table. Demographic, disease, and treatment characteristics (N= 454)

Age at infusion, median (range)

60 (50, 68)

Male sex

286 (63%)

Prior HCT

186 (41%)

Disease

Aggressive NHL

216 (48%)

ALL

82 (18%)

MM/PCL

61 (13%)

Indolent NHL

53 (12%)

CLL

42 (9%)

CAR T-cell product

Investigational CD19 CAR T-cell product

180 (40%)

Commercial CD19 CAR T-cell product with CD28 costimulatory domain (axi-cel, brexu-cel)

143 (31%)

Commercial CD19 CAR T-cell product with 4-1BB costimulatory domain (liso-cel, tisa-cel)

70 (15%)

Commercial BCMA CAR T-cell product (cilta-cel, ide-cel)

61 (13%)

Conclusions: This is a novel application of the heatwaveR package to automate ICAHT grading, eliminating subjectivity and labor. This approach allows rapid and automated ICAHT grading in large datasets. External validation of heatwaveR in a separate dataset from Memorial Sloan Kettering Cancer Center is ongoing. We plan to develop an online tool to help clinicians easily apply this approach to grade the severity of hematotoxicity per ICAHT criteria.

Disclosure: Kai Rejeski: BMS/Celgene: Consultancy, Honoraria; Novartis: Honoraria; Kite/Gilead: Other: Travel Support, Research Funding; Pierre-Fabre: Other: Travel Support.

Joshua Hill: allovir: Consultancy, Research Funding; moderna: Consultancy; deverra: Research Funding.

Rahul Banerjee: BMS: Consultancy; Janssen: Consultancy; Genentech: Consultancy; SparkCures: Consultancy; Sanofi: Consultancy; Caribou: Consultancy; Pfizer: Consultancy; Pack Health: Research Funding.

Andrew Cowan: BMS, Adaptive: Consultancy; Adaptive Biotechnologies, Harpoon, Nektar, BMS, Janssen, Sanofi, Abbvie: Research Funding.

Damian Green: Cellectar Biosciences: Research Funding; SpringWorks Therapeutics: Research Funding; Celgene: Consultancy; GlaxoSmithKline: Membership on an entity’s Board of Directors or advisory committees; Sanofi: Research Funding; Janssen Biotech: Consultancy, Research Funding; Ensoma: Consultancy; Seattle Genetics: Consultancy, Research Funding; Juno Therapeutics A BMS Company: Patents & Royalties, Research Funding.

Ajay Gopal: Compliment Corporation: Current holder of stock options in a privately-held company; Incyte, Kite, Morphosys/Incyte, ADCT, Acrotech, Merck, Karyopharm, Servier, Beigene, Cellectar, Janssen, SeaGen, Epizyme, I-Mab bio, Gilead, Genentech, Lilly, Caribou, Fresenius-Kabi: Consultancy; Merck, I-Mab bio, IgM Bio, Takeda, Gilead, Astra-Zeneca, Agios, Janssen, BMS, SeaGen, Teva, Genmab: Research Funding.

Christina Poh: BeiGene: Consultancy; Seattle Genetics: Consultancy; Incyte: Research Funding; Acrotech: Consultancy.

Mazyar Shadman: Fate Therapeutics: Consultancy; Genmab: Consultancy, Research Funding; Vincerx: Research Funding; MorphoSys/Incyte: Consultancy, Research Funding; Eli Lilly: Consultancy; Bristol Myers Squibb: Consultancy, Research Funding; Genentech: Consultancy, Research Funding; Kite, a Gilead Company: Consultancy; AbbVie: Consultancy, Research Funding; Mustang Bio: Consultancy, Research Funding; ADC therapeutics: Consultancy; BeiGene: Consultancy, Research Funding; AstraZeneca: Consultancy, Research Funding; Pharmacyclics: Consultancy, Research Funding; Janssen: Consultancy; MEI Pharma: Consultancy; Regeneron: Consultancy; TG Therapeutics: Research Funding.

Alexandre Hirayama: Novartis: Honoraria; Bristol Myers Squibb: Honoraria, Research Funding; Nektar Therapeutics: Honoraria, Research Funding; Juno Therapeutics, a Bristol Myers Squibb Company: Research Funding.

Till: Mustang Bio: Consultancy, Patents & Royalties, Research Funding; BMS/Juno Therapeutics: Research Funding; Proteios Technology: Consultancy, Current holder of stock options in a privately-held company.

Kimble: Juno/BMS: Research Funding.

Lorenzo Iovino: Mustang Bio: Current equity holder in publicly-traded company.

Aude Chapuis: Juno Therapeutics: Research Funding.

Ryan Cassaday: Amgen: Consultancy, Honoraria, Research Funding; Merck: Research Funding; Incyte: Research Funding; Autolus: Membership on an entity’s Board of Directors or advisory committees; Pfizer: Consultancy, Honoraria, Research Funding; Servier: Research Funding; Vanda Pharmaceuticals: Research Funding; Jazz: Consultancy, Honoraria; Kite/Gilead: Consultancy, Honoraria, Research Funding; PeproMene Bio: Membership on an entity’s Board of Directors or advisory committees; Seagen: Other: Spouse was employed by and owned stock in Seagen within the last 24 months.

Filippo Milano: ExCellThera Inc.: Research Funding.

David Maloney: A2 Biotherapeutics: Consultancy, Current holder of stock options in a privately-held company, Honoraria, Other: Member of the Scientific Advisory Board; Juno Therapeutics: Consultancy, Honoraria, Patents & Royalties: Rights to royalties from Fred Hutch for patents licensed to Juno Therapeutics/BMS, Research Funding; Janssen: Consultancy, Honoraria; Legend Biotech: Consultancy, Honoraria, Research Funding; Mustang Bio: Consultancy, Honoraria; Novartis: Consultancy, Honoraria; Lyell Immunopharma: Other: Member, CAR T Steering Committee; Incyte: Consultancy, Honoraria; Gilead Sciences: Consultancy, Honoraria, Other: Member, Scientific Review Committee, Research Scholars Program in Hematologic Malignancies; Kite, a Gilead Sciences: Consultancy, Honoraria, Research Funding; Pharmacyclics: Consultancy, Honoraria; Umoja: Consultancy, Honoraria; Celgene: Consultancy, Honoraria, Membership on an entity’s Board of Directors or advisory committees, Other: Participation on a Data Safety Monitory Board, Research Funding; Genentech: Consultancy, Honoraria, Other: Chair and Member of the Lymphoma Steering Committee; MorphoSys: Consultancy, Honoraria; Bristol Myers Squibb: Consultancy, Honoraria, Other: Member of the JCAR017 EAP-001 Safety Review Committee and Member, CLL Strategic Council, Member of the JCAR017-BCM-03 Scientific Steering Committee under BMS, Research Funding; Amgen: Consultancy, Honoraria; Navan Technologies: Consultancy, Honoraria, Other: Member of the Scientific Advisory Board; Bioline Rx: Consultancy, Membership on an entity’s Board of Directors or advisory committees, Other: Participation on a Data Safety Monitory Board; Fred Hutch: Other: rights to royalties for patents licensed to Juno; Navan Technologies: Current holder of stock options in a privately-held company; Chimeric Therapeutics: Other: Member of the Scientific Advisory Board; ImmPACT Bio: Other: Member, Clinical Advisory Board, CD19/CD20 bi-specific CAR-T Cell Therapy Program; Interius: Other: Member, Clinical Advisory Board.

Jordan Gauthier: Kite Pharma: Consultancy, Honoraria; MorphoSys: Consultancy, Research Funding; Angiocrine Bioscience: Research Funding; Century Therapeutics: Other: Independent data review committee; Celgene (a Bristol Myers Squibb company): Research Funding; Legend Biotech: Consultancy, Honoraria; Janssen: Consultancy, Honoraria; Juno Therapeutics (a Bristol Myers Squibb company): Research Funding; Sobi: Consultancy, Honoraria, Research Funding.

4: CAR-based Cellular Therapy – Clinical

P094 RELEVANT STUDIES ON CYTOKINES LEVEL AND CAR-T EXPANSION ASSOCIATED TO CD7 CAR-T THERAPY

Dongchu Wang1, Hui Wang1, Man Chen 1

1Hebei Yanda Lu Daopei Hospital, Langfang, China

Background: In the past few years, CD7 became a highly reliable marker of CAR-T therapy treating T cell acute lymphocytic leukemia(T-ALL). In this case, better understanding of CD7 CAR-T therapy on cellular level became meaningful.

Methods: PB samples were collected from 70 patients treated with CD7 CAR-T from Dec. 2020 to Nov. 2022 in Hebei Yanda Ludaopei Hospital. There were 56 males and 14 females, 2-60 with median age 19. Based on criteria of CRS and assessment results, 70 patients were split in different groups. According to CRS level, two groups were set as low (CRS0-1, 56 of 70) group and high group (CRS2-4, 14 of 70). 55 of 70 were classified as CR/CRi group, 15 of 70 were Non-CR group.

IL-1β, IL-2, IL-4, IL-5, IL-6, IL-8, IL-10, IL-12p70, IL-17A, IL-17F, IL-22, TNFα, TNFβ, IFN-γ, IL-2RA/sCD25, MIP-1α, MCP-1, GM-CSF, IL-15, REG3a, Elafin, ST-2, Granzyme B, TNFRI were measure with microbeads with Calibur flow cytometer, CAR-T cells, target cells(CD7+ cells), T cells subsets(CD3 + , CD4 + , CD8 + , CD8 + TCM, CD8 + TEM, Treg) were tested with Canto flow cytometer. All the cellular variations were calculated by comparing value of peak-time to value of D0 which was used as baseline. Time points day0, 4, 7, 11, 14, 21, 30 were monitored after CAR-T infusion.

Results: Patients in all 4 groups had similar median age and transfection efficiency. Based on criteria of CRS level, patients were split into low group and high group. CAR-T peak time was appeared significantly later in high group. In high group, CD8 + TEM cells was amplified significantly more than that in low group at peak time point. Clearance rates of target cells in low group were significantly higher. The level of IL-4, IL-6, IL-8, IL-10, IL-22, IFN-γ, sCD25, MCP-1, IL-15, GranzymeB were significant higher in high group. All median peak time of those 10 cytokines in high group were calculated, from day 10 up to day 15 after CAR-T infusion. MCP-1 had the largest time gap, which means MCP-1 reached peak time 9 days earlier than that of CAR-T cells. The median peak-time gap between cytokines level with CAR-T cells expansion was 6 days.70 patients were classified as CR/CRi group and Non-CR group. CAR-T expansion, T cell subsets variation showed no statistical difference between two groups, and only target cells clearance rate was significantly higher in CR group. For all 24 cytokines measured in this study showed no significant differences between both groups.

Conclusions: 10 cytokines were correlated to severity of CRS after CD7 CAR-T therapy. 10 cytokines showed positive correlation to the level of CRS, and reached peak time earlier than that of CAR-T expansion, which means all these 10 cytokines could be used as indicators to predict occurrence and severity of CRS. Concurrence time of peak expansion of CD7 CAR-T cells, and amplification of CD8 + TEM cells were significantly related to CRS. For all the patients who reached complete remission or CRi, CD7+ clearance rates were higher with statistical significance. Our results proved potential indicators for CRS prediction and prognosis.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P095 AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION COMBINED WITH CAR-T CELL THERAPY SIGNIFICANTLY IMPROVES PROGRESSION-FREE SURVIVAL IN RELAPSED REFRACTORY CENTRAL NERVOUS SYSTEM LYMPHOMA

Rui Liu 1, Fan Yang1, Fei Xue1, Zhonghua Fu1, Yuelu Guo1, Shaomei Feng1, Peihao Zheng1, Lixia Ma1, Hui Shi1, Biping Deng1, Xiaoyan Ke1, Kai Hu1

1Bejing Gobroad boren hospital, Beijing, China

Background: Relapsed or refractory Central nervous system lymphoma(R/R CNSL) is challenging to manage clinically. CAR-T therapy has demonstrated efficacy in relapsed and refractory large B-cell lymphoma, with some studies indicating a favourable response rate for R/R CNSL. Nevertheless, disease-free survival and long-term outcomes are suboptimal. Our study aims to explore the efficacy of CAR-T and the factors affecting its long-term survival in R/R CNSL. Additionally, we analysed the safety and effectiveness of ASCT combined with CAR-T therapy.

Methods: This single-centre, retrospective study analysed patients with R/R CNSL who received CAR-T therapy at the Beijing GoBroad Boren Hospital between 2019/03 and 2023/08. 69 patients were included. 46% were male, the median age is 51 (range19-79); 33% (23/69) is PCNSL, 67% (46/69) is SCNSL. The common histology in SCNSL was DLBCL(n=32), followed by HGBL(n=4), PMBL(n=3), BL (n=2), MCL (n=2) and others. Median number of previous treatment lines is 4 (range2-8). 70% (48/69) were IPI≥3.74% (51/69) were resistant to HD-MTX. 52% (36/69) were resistant to BTK inhibitors. 17% (12/69) patients had failed previous autologous hematopoietic stem cell transplantation. 68%(47/69) were parenchymal involved, 17%(12/69) were CNS involved, 14%(10/69) were both. The patients underwent a median of 1 (range 0-5) cycle of bridging therapy. 65.2%(45/69) achieve objective response (CR=33;PR=12) after bridging therapy. According to the response to bridging treatment, patients were divided into two groups: Bridging Effective (BE) group, comprising patients with CR/PR, and Bridging Ineffective (BI) group, comprising patients with SD/PD. In the BE group, 53.3% (24/45) received ASCT combined with CAR-T therapy.

Results: The 3-month ORR was 70% (48/69), and CRR is 61% (42/69). Incidences of any-grade CRS and ICANS were 70% (48/69) and 12% (8/69), with severe (≥3) CRS and ICANS at 12.5% (6/48) and 62.5% (5/8), respectively. BE group exhibited significantly higher 3-month ORR (84.4% vs. 41.6%) and CRR (82.2% vs. 20.8%) compared to BI group. Median follow-up time was 22.65 months. Median Progression-Free Survival (PFS) for BE group was 43.07 months, significantly longer than BI group’s 6.08 months (P<0.0001). Median Overall Survival (OS) was not reached for BE group, while it was 12.79 months for BI group (P<0.0001). 1-year OS rates were 100% vs. 51.4% (P<0.0001), and 1-year PFS rates were 74.7% vs. 9.9% (P<0.0001) for BE and BI groups, respectively. Among the 45 BE group patients, ASCT combined with CAR-T therapy showed more patients with ECOG performance status ≥ 2, more prior therapy lines, and all were HD-MTX-resistant (Table 1). Median PFS in ASCT combined with CAR-T cohort was significantly prolonged (not reached vs. 14.76 months, p=0.0008). Median OS was not reached in this cohort, whereas in CAR-T cohort, it was 27.48 months. However, there was no statistically significant difference. No statistical difference was observed in CRS and ICANS between the two cohorts.

Variable

CAR-T (n=21)

ASCT + CAR-T(n=24)

p-value

age (>60)

8/21(38%)

2/24(8%)

0.018

sex(male)

6/21(29%)

12/24(50%)

0.148

IPI≥3

11/21(52%)

17/24(71%)

0.208

ECOG≥2

7/21(33%)

17/24(71%)

0.013

Histology

-PCNSL

9/21(43%)

11/24(46%)

0.843

-SCNSL

12/21(57%)

13/24(54%)

Previous HD-MTX_resistance

15/21(71%)

24/24(100%)

0.005

Previous BTK inhibitors_resistance

14/21(67%)

13/24(54%)

0.398

Previous auto-HCT

5/21(24%)

3/24(12.5%)

0.328

Previous_regimen_lines≥3

11/21(52%)

20/24(83%)

0.027

Conclusions: Effective bridging therapy can significantly improve the efficacy of CART in the treatment of R/R CNSL. After bridging therapy is effective, the treatment model of ASCT combined with CAR-T can significantly improve disease-free survival.

Clinical Trial Registry: ChiCTR2200058972/https://www.chictr.org.cn/index.html

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P096 SAFETY AND EFFICACY OF BENDAMUSTINE VERSUS FLUDARABINE-CYCLOPHOSPHAMIDE LYMPHODEPLETION PRIOR TO CAR-T CELL THERAPY WITH AXICABTAGENE CILOLEUCEL FOR NON-HODGKIN LYMPHOMA: A SINGLE-INSTITUTION STUDY

Anthony Stack 1, Yuliya Shestovska1, Rachel Thomas2, Matthew Hamby1, Asya Varshavsky-Yanovsky1, Peter Abdelmessieh1, Michael Styler1, Henry Fung1, Rashmi Khanal1

1Fox Chase Cancer Center/Temple University Health System, Philadelphia, United States, 2Temple University Hospital, Philadelphia, United States

Background: Previously, bendamustine (Benda) lymphodepletion (LD) was shown prospectively to yield similar efficacy, with lower rates of Cytokine Release Syndrome (CRS), neurotoxicity (ICANS) and hematologic toxicity compared to fludarabine-cyclophosphamide (FluCy) prior to tisagenlecleucel, a 4-1BB-costimulated CAR-T cell product; however, the safety and efficacy of bendamustine lymphodepletion prior to axicabtagene ciloleucel (axi-cel), a CD28-costimulated CAR-T cell product, is largely unknown.

Methods: We performed a retrospective analysis to compare safety and efficacy outcomes of patients treated for non-Hodgkin lymphoma with axi-cel at Fox Chase Cancer Center from 10/1/2018 to 7/14/2023. All patients received lymphodepletion with Benda (90 mg/m2/day x 2 days) or FluCy (Flu 30 mg/m2/day and Cy 500 mg/m2/day x 3 days) based on availability, physician preference and institutional practice. P values were calculated using Mann-Whitney test for continuous variables and Fisher’s exact test for categorical variables. Grading of hematologic toxicities is based on the Common Terminology Criteria for Adverse Events Version 5.0.

Results: In total, 20 patients received Benda and 17 patients received FluCy LD prior to axi-cel and were included in our analysis. The ratio of high-grade/low grade lymphoma in the Benda vs FluCy groups was 2.3 and 3.3, respectively. In both groups, ≥90% had Eastern Cooperative Oncology Group performance scores ≤1, an identical median age of 63 and similar rates of bridging therapy utilization (75% vs 76%, respectively). Of note, 95% of patients in the Benda group received CRS/ICANS prophylaxis (all dexamethasone), compared to only 12% in the FluCy group (1 dexamethasone and 1 tocilizumab).

Overall (ORR) and complete response rates (CR) were [ORR 90%, CR 75%] and [ORR 65%, CR 53%] in the Benda and FluCy groups, respectively, and were not statistically different (p>.05). Likewise, 3-month progression-free and overall survival did not differ between groups. CRS rate was identical in both arms, occurring in 70%, while ICANS occurred in 15% after Benda vs 47% after FluCy (p=.069).

In the first 30 days post-CAR-T infusion, patients receiving Benda vs FluCy experienced significantly lower rates of grade 3-4 anemia (10% vs 58%, p=.004), neutropenia (30% vs 100%, p<.001), and thrombocytopenia (25% vs 65%, p=.022), respectively. Additionally, Benda vs FluCy patients experienced significantly higher median nadirs in hemoglobin (10.5 vs 7.4 g/dL, p=.008), absolute neutrophils (ANC) (1.35 vs 0 K/mm3, p<.001) and platelets (117 vs 34 K/mm3, p=.003), required fewer transfusions of red blood cells (10% vs 41%, p=.052) and platelets (5% vs 35%, p=.034), and had numerically lower requirement for G-CSF (15% vs 47%, p=.069), respectively.

In the first 30 days, patients receiving Benda vs FluCy LD experienced fewer days with platelets <50 k/mm3 (mean 2.6 vs 9.4 days, p=.010), and with ANC <1000 K/mm3 (mean 1.8 vs 12 days, p<.001), respectively. Persistent grade ≥3 anemia, thrombocytopenia and neutropenia beyond 30 days were uncommon and did not significantly differ between groups.

Conclusions: In this retrospective study comparing Benda to FluCy lymphodepletion prior to axi-cel, Benda yielded similar efficacy with significantly less early hematologic toxicity. Although CRS and ICANS rates were similar, this is confounded by increased utilization of prophylaxis in the Benda group.

Disclosure: Henry Fung has received honoraria from Incyte, AstraZeneca, Janssen Oncology, and Kite. Asya Varshavsky-Yanovsky has participated on advisory boards and/or has provided consultancy for Pfizer, Janssen and BMS. Peter Abdelmessieh has participated on advisory boards for AbbVie and Sobi. Anthony Stack, Yuliya Shestovska, Rachel Thomas, Matthew Hamby, Michael Styler and Rashmi Khanal have no conflicts of interest to declare.

4: CAR-based Cellular Therapy – Clinical

P097 PSEUDOPROGRESSION: A FREQUENT AND CLINICALLY RELEVANT ADVERSE EVENT OF CHIMERIC ANTIGEN RECEPTOR T CELL THERAPY

Torsten Schroeder 1, Tjark Martens1, Guranda Chitadze1, Fatih Yalcin1, Heiko Trautmann1, Britta Kehden1, Monika Brüggemann1, Christine Heinen1, Philipp Nakov,1, Nicolas Spath1, Lukas Sprenger1, Anca-Maria Albici1, Laura-Jane Kramp1, Matthias Ritgen1, Dominique Wellnitz1, Sebastian Lippross1, Maciej Simon1, Ingram Iaccarino Idelson1, Nico Gagelmann2, Wolfram Klapper1, Lars Fransecky1, Thomas Valerius1, Natalie Schub1, Christiane Pott1, Katja Weisel2, Winfried Alsdorf2, Fabian Müller3, Claudia Baldus1, Friedrich Stölzel1

1University Hospital Schleswig-Holstein, Kiel, Germany, 2University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 3University Hospital of Erlangen, Erlangen, Germany

Background: CAR-T-cell therapy (CARTCT) expands the range of therapeutic options for patients with hematologic malignancies. While complications such as cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) are well known and consensus grading classifications for documentation and treatment of these adverse events are used, pseudoprogression (PP) is mainly described after checkpoint inhibition. Some reports on CARTCT induced PP (CARTiPP) exist distinguishing it from progressive disease, while only few reports focus on acute local complications. Considering the paucity of data about CARTiPP, we aimed to assess patients treated in our clinical routine. We defined CARTiPP as initial volume increase of malignant manifestations within ten days of CARTCT followed by tumor regression without another cause being more probable. Distinction from true progression of malignancy can be achieved either retrospectively or by histopathological findings. Another special CARTiPP presentation may be severe bone pain in patients suffering from malignant bone lesions which was repeatedly observed after anti-BCMA CAR-T cells.

Methods: Here, we characterize a retrospective multicentric patient cohort with the occurrence of CARTiPP. Evaluation of clinical, laboratory, radiological, and/or histopathological data was carried out. We rated all events in a proposed grading scale for CARTiPP severity.

Results: A total of n=22 patients were identified with CARTiPP throughout a treatment period of 57 months with n=176 patients receiving 180 CAR-T-cell therapies. Thus, we observed a total CARTiPP frequency of 12 %.

Patients with CARTiPP had different B-cell non-Hodgkin lymphoma subtypes (n=10/114) or multiple myeloma (n=12/56) and were treated with commercial anti-CD19 or anti-BCMA CAR-T-cells. The median age of patients was 59 years (range 39-76 years). Staging before CARTCT assessed n=5 patients in very good partial remission (VGPR), n=3 in partial remission, n=6 with stable disease (SD) and n=8 with progressive disease (PD). The rate of CARTiPP was notably higher in multiple myeloma than in B-cell lymphoma patients (21 vs. 9 %) - findings that need to be confirmed by additional studies.

We observed variable complications, e.g. superior vena cava syndrome, severe bone pain, soft tissue swelling with airway compression and postrenal kidney injury. Concomitant CRS occurred in n=21, ICANS in n=5 patients. Complication management ranged accordingly from intensified monitoring to treatment with glucocorticoids/tocilizumab to invasive ventilation and once limb amputation due to compartment syndrome. There was no CARTiPP-associated mortality. In four affected cases intensive care treatment was required.

Conclusions: CARTiPP may be still underreported and lacks classification and prospective reporting in clinical trials and registries investigating CARTCT. Frequency and sometimes severity underline the need for raising awareness of treating physicians. Laboratory analyses and imaging may supplement clinical assessment and should be further examined. Establishment of risk assessment, preventive measures and early treatment to reduce therapy-induced morbidity/mortality as well as a systematic documentation analogous to CRS and ICANS is required for CARTiPP. The increasing availability of CAR-T-cell therapies for a steadily expanding number of disease entities with variable manifestations and different disease stages makes an increasing frequency of CARTiPP plausible.

Disclosure: - Friedrich Stölzel:

Travel Support: Janssen, medac

Honoraria: medac, GWT, Abbvie, Servier, Pierre Fabre

Research funding: medac, Servier

- Claudia Baldus:

advisory and speakers honoraria for Astrazeneca, BMS, Janssen, Kite, Novartis, Jazz, Astellas, Amgen

- Winfried Alsdorf:

Honoraria: GSK, Janssen

Travel/accomodations/expenses: Biontech, Janssen, Immatics

Research funding (institution): Affimed, Biontech

- Fabian Müller:

Advisory and speakers honoraria for Astrazeneca, BMS, Jansen, Kite, kyverna, Miltenyi, Novartis,

Research support from Astrazeneca, Kite.

4: CAR-based Cellular Therapy – Clinical

P098 CAR-T CELL TREATMENT OF LARGE B-CELL LYMPHOMA (LBCL): DO PROGRAM DURATION AND TREATMENT LINE MATTER?

Maria-Luisa Schubert 1, Michael Schmitt1, Andrea Bondong1, Mandy Wegner1, Simon Renders1, Isabelle Krämer1, Carsten Müller-Tidow1, Peter Dreger1

1University Hospital Heidelberg, Heidelberg, Germany

Background: At our institution, CD19-directed CARTs for treatment of multiply relapsed/refractory LBCL were introduced in November 2018 and extended to 2nd line (2L) application in September 2022. The aim of this study was to analyze development of patient characteristics and outcomes over time.

Methods: Eligible for this retrospective analysis were all adult patients who received commercial or experimental CD19 CARTs at the University of Heidelberg. Patients were grouped into 3 cohorts: cohort A >2L 2018-2020; cohort B >2L 2021-2023; cohort C intent-to 2L treatment. Primary endpoint was relapse/progression-free survival (PFS). Descriptive statistics were applied.

Results: Altogether 93 patients (cohort A: n=43, cohort B: n=35, cohort C: n=15) met the eligibility criteria. Of note, introduction of 2L CARTs was associated with a 67% decrease of >2L indications in 2023. Axicabtagene ciloleucel (axicel), tisagenelecleucel (tisacel), lisocabtagen maraleucel (lisocel), and experimental CARTs were used in 73%, 18%, 6%, and 3% of patients, respectively. Cohorts A and B were comparable in terms of underlying diagnosis with DLBCL as the predominant indication, and in terms of the proportion of patients who had received a prior autoHCT. Compared to cohort A, patients from cohort B were significantly older (median age 57 (20-76) vs 67 (25-82) years; p 0.049), but contained fewer patients who received CARTs beyond the 3rd (49% vs 70%; p 0.067) and 4th line (6% vs 30%; p 0.0083), respectively. More patients in cohort B had received holding/bridging therapies (89% vs 70%; p 0.057), with a smaller proportion of patients with refractory disease at lymphodepletion (46% vs 81%; p 0.0058). In cohort C, median age was 63 (26-82) years, holding/bridging was administered to 87%, and 67% of the patients had active disease at lymphodepletion. Because of progression through bridging therapy, 27% of the patients intended to receive 2L CARTs actually underwent CART infusion as 3rd-line therapy.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (12)

With an overall response rate of 74%, 74%, and 78%, response was comparable across all cohorts. However, CR was more frequently reached in cohort C (57% vs 36% (A) and 40% (B)). Crude non-relapse mortality (NRM) was 12%, 6%, and 7% across the cohorts A, B, and C, respectively, with infections being the predominant cause of death. All NRM events except one occurred with axicel. 12-month PFS was 30%, 43%, and 56% across cohorts A, B, and C, respectively (Logrank for trend p=0.052; HR 1.72 (0.96-2.72) for comparison of cohort A with cohort B) (Figure).

Conclusions: Despite constant response rates, PFS and NRM of >2L CARTs seem to improve with longer CART program duration. This might be partly explained with better patient selection, earlier use of CARTs, and more effective bridging strategies. The advent of 2L CARTs associates with a strong reduction, but not a complete abrogation of >2L indications. Whether 2L CART applications are indeed associated with superior outcomes compared to delaying CART administration beyond 2L needs to be confirmed by further studies.

Disclosure: MLS: honoraria Kite/Gilead. MS: honoraria BMS, Kite/Gilead, Novartis. RS: honoraria Kite/Gilead. PD: honoraria Novartis, Kite/Gilead, BMS, Miltenyi.

4: CAR-based Cellular Therapy – Clinical

P099 REAL-WORLD EXPERIENCE OF CAR T-CELL THERAPY: A SINGLE CENTRE EXPERIENCE

Francys Lopez Llanos1, Inês Rocha2, Gilda Teixeira1, Isabelina Ferreira1, Pedro Sousa1, Maria João Gutierrez1, Susana Pereira 1, Ana Carolina Freitas1, Carla Espadinha1, Nuno Miranda1

1IPOFG, Lisboa, Portugal, 2Centro Hospitalar Universitário Lisboa Central, Lisboa, Portugal

Background: Chimeric antigen receptor (CAR) T-cell therapy has changed the landscape of treatment for relapsed/refractory B cell lymphomas. We present real world patient characteristics and outcomes from a Portuguese center.

Methods: This is a single-center, retrospective study of consecutive patients receiving CAR-T cell therapy between October 2019 and December 2023. Data were retrieved by reviewing individual electronic health records at the Instituto Português de Oncologia Francisco Gentil, Lisbon, Portugal.

Results: 50 patients were accepted for CAR-T therapy. Median age was 60 [26- 74] years, 94% (N=47) had ECOG PS1 and male-to-female ratio was 2:1. Regarding diagnosis, 78% (N=39) had histologically diffuse large B-cell lymphoma (DLBCL), 4% (N=2) follicular lymphoma and 18% (N=9) mantle cell lymphoma (MCL). 36% (N=18) had ≥ 3 lines of therapy prior CAR-T49 patients (98%) underwent leukapheresis and 42 (84%) received CAR-T therapy (3 DLBCL and 3 LCM did not undergo lymphodepletion to medical complications or rapidely progressive disease, 1 is awaiting infusion). 81% (N=34) received Axicabtagene, 14.3% (N=6) with Bretuxabtagene and 4.8% (N=2) with Tisagenlecleucel. Median delay (Md) between leukapheresis and infusion was 64 days [30 - 141]. CRS occurred in 35 patients (83.3%), with grade 3 in 11,9% (N=5). ICANS occurred in 10 patients (23.8%), with grade 3 in12% (N=5). Infections were documented in 10 and pancytopenia in 9. Two second neoplasms were diagnosed 1 AML and 1 MDS.

PET-CT was used to evaluate post-CAR-T response. At D + 100, 33 patients (78.6%) had completed evaluation, 63.6% (N=21) in complete response (CR), 15.2% (N=5) partial response (PR) and 21.2% (N=7) disease progression (DP). At D + 180, of 22 patients (66.7%) with complete evaluation, 63.6% (N=14) in CR, 31.8% (N=7) in DP and 4.6% (N= 1) in PR. There are patients who have not yet reached assessment time intervals.

Post-CAR-T DP occurred in 16 patients (38.0%), in whom Md time between infusion and DP was 3 months [1-16]. Death occurred in 14 patients (33.3%), mostly due to DP (11 patients, 78.6%); the Md time between infusion and death was 6 months [1-29]. Two fatalities were infection related: 1 HHV6 encephalitis and 1 COVID19.

With a median OS of infused patients was estimated at 29.7 months [95% CI: 23.3-36.1] and PFS was 9.0 months [95% CI: 1.9-16.0].

There was a trend to higher mortality in older than 65 vs younger (p=0.19).

There was no difference between PFS with a vein-to-vein time below or over median.

We observed a significant difference in PFS when making cohorts for year of infusion: before 2021 PFS=24,6 months vs 2022 11.4 vs 2023 5.4 p=0.041.

Conclusions: Relapsed and refractory NHL, in anti-CD20 era, constitute a large patient population in need of new solutions. CAR-T therapy provides an answer for some, with acceptable toxicity. Our real-world data shows that over the years, with broader selection criteria, inclusion of older patients, and those with higher disease burden, worse outcomes are observed. Accessment of disease burden prior to CAR-T may help selecting more adequate candidates,. Better patient selection is necessary also due to economic burden of this therapy.

Disclosure: The authors declare no conflit of interests

4: CAR-based Cellular Therapy – Clinical

P100 IMPACT OF ACUTE KIDNEY INJURY IN LYMPHOMA PATIENTS TREATED WITH AXICABTAGENE CILOLEUCEL

Cristina Soler1, Rafael Hernani 1, Ariadna Pérez1, Jose Luis Piñana1, Juan Carlos Hernández-Boluda1, Ana Benzaquen1, Rosa Goterris1, Montse Gómez1, Paula Amat1, Blanca Ferrer1, Francesc Moncho1, Isidro Torregrosa1, Jose Luis Górriz1, María José Terol1, Carlos Solano1

1Hospital Clínico Universitario, INCLIVA Research Institute, Valencia, Spain

Background: Chimeric antigen receptor T-cell (CAR-T) therapy has improved outcomes in patients with relapsed/refractory (R/R) hematological malignancies. However, there is limiting data regarding the impact of acute kidney injury (AKI) on outcomes following CAR-T infusion.

Methods: Adult patients with R/R large B-cell lymphoma (LBCL) receiving commercial axicabtagene ciloleucel were consecutively enrolled at Hospital Clínico Universitario de Valencia from December 2019 to August 2023. Data regarding patient and disease characteristics, treatment course, and clinical outcomes were prospectively collected. Cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS) were graded following CARTOX scale. Chronic kidney disease (CKD) was defined by Kidney Disease Improving Global Outcomes (KDIGO) staging using estimated glomerular filtration rate (eGRF). Acute kidney injury (AKI) was defined by Common Terminology Criteria for Adverse Events version 5. The renal function was monitored from the onset of lymphodepleting chemotherapy (LDC) until one month after infusion. Subsequently, monitoring continued until day 90, unless death or R/R, whichever occurred first. All patients received allopurinol or rasburicase as tumor lysis syndrome prophylaxis.

Results: A total of 48 adult patients with R/R LBCL were included in this study (diffuse large B-cell lymphoma, n=44; primary mediastinal large B-cell lymphoma, n=4). Briefly, the median age was 56 years (range, 22–75). Three (6%), 7 (15%) and 9 (19%) patients had prior history of CKD, diabetes, or high blood pressure, respectively. Baseline eGRF was graded as follows: 1 (n=28), 2 (n=17) and 3a (n=3). Most patients were primary refractory (53%) and had progressive disease (63%) at LDC. Myc was rearranged in 11 (23%) patients. Baseline EASIX score was 2.47 (range, 0.49–552).

Sixteen (33%) patients developed AKI (grade 1, n=10; grade 2, n=6) at a median time of 14 days (-4 – 57) after CAR-T infusion. Of them, five (31%), 6 (38%) and 5 (31%) patients developed AKI between LDC and CAR-T infusion, within the first month following CAR-T infusion or beyond, respectively. AKI showed no association with any baseline characteristics. Only age (>65 years) correlated with abnormal baseline eGFR in univariate analysis (71% vs 29%, P 0.018). Neither CKD, abnormal baseline eGFR (>= G2 KDIGO) or AKI had an impact on toxicity (CRS/ICANS), overall response rate (ORR), or survival (Table 1). Six (13%) patients required nephrologist support due to the following causes: AKI (n=4), electrolyte imbalances (n=1), adjustment of nephrotoxic medications owing to a prior kidney transplant (n=1). None of the patients required renal replacement therapy. Twenty-six (54%) patients remained alive with a median follow-up of 384 days (range, 63–1323).

Conclusions: In this study, we found no correlation between CKD, baseline eGFR, or AKI and outcomes post-CAR-T therapy for R/R LBCL. Notably, one-third of patients experienced mild AKI throughout three distinct time periods: i) from LDC and CAR-T infusion; ii) from CAR-T infusion to day 30; iii) from day 30 to day 90.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P101 CHARACTERISTICS OF CENTRAL NERVOUS SYSTEM RELAPSE AFTER CD19 CAR-T CELL THERAPY IN R/R NHL

Mengyu Zhao 1, Tingting Yang1, YeTian Dong1, Jiazhen Cui1, Delin Kong1, Wenfa Huang2, Lei Wang2, Lixin Wang2, He Huang1, Yongxian Hu1

1Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2lnternational Cancer Center, Shenzhen University General Hospital, Shenzhen, China

Background: CD19-targeted chimeric antigen receptor T (CAR-T) cell therapy has shown remarkable antitumor activity against relapsed/refractory (R/R) non-Hodgkin lymphoma (NHL). However, NHL with central nervous system (CNS) relapse carries a poor prognosis and high mortality. We reported an analysis of 80 patients with NHL who underwent CAR-T cell therapy, and described the characteristics including the international prognostic index (IPI) and other clinical variables to evaluate the outcomes of CNS relapse in NHL patients treated with CAR-T cell therapy.

Methods: A total of 80 patients with NHL treated with CD19 CAR-T cell therapy in our center between April 2017 and November 2022 was retrospectively analyzed. Patient characteristics, laboratory parameters, and clinical outcomes were collected. CNS relapse was diagnosed by cerebrospinal fluid cytology, MRI or PET-CT.

Results: Baseline characteristics of all 80 patients are summarized. The median age was 58.5 (range, 10 to 79) years, and 57.5% of patients were male. Overall, 67 (83.8%) patients achieved complete remission (CR) or partial remission (PR) after CD19 CAR-T therapy. Seven (8.43%) patients developed isolated CNS relapse after CAR-T cell therapy, all of which were heavily pre-treated with multiple lines of chemotherapy prior to CAR-T cell therapy, and one of them had received autologous stem cell transplant before infusion of CAR-T cells. Only 4 of 7 patients with isolated CNS relapse achieved CR (n=4), and the median time to isolated CNS relapse from CAR-T cell infusion was 6.3(range, 1.53 to 25.8) months. Thirty-seven patients (46.2%) developed systemic recurrence without CNS involvement, with the median time from CAR-T to relapse of 3.2 (range, 0.467-30.8) months. The other 36 patients remained in remission at the last follow-up. During the median follow-up of 13.5 months of the whole cohort, the number of recorded deaths in isolated CNS relapse, systemic only recurrence, and no relapse patients was 5 (71.4%), 22 (59.4%), 1(2.78%), respectively. The 1-year OS and 1-year PFS of patients with isolated CNS recurrence was significantly lower compared to the no relapse group (OS, 35.7% [95%CI 12.1–100] vs 97.22% [95%CI 92–100], P<0.001; PFS, 28.6% [95%CI 8.86–92.2] vs 100% [95%CI 100–100], P<0.001), but showed no difference compared to the systemic only recurrence group (OS: 53.3% [95%CI 39.3–72.4], P=0.79; PFS: 13.0% [95%CI 5.53-30.5], P=0.17).

Conclusions: In conclusion, patients with CNS relapse showed significant difference in 1-year outcomes with no relapse group but not systemic relapse group in our study, which suggested the importance of effective prophylaxis against CNS relapse. Further investigation is necessary to better define the characteristics of patients who will benefit from prophylaxis.

Clinical Trial Registry: ChiCTR1800015575; NCT03118180; NCT04532281; NCT04532268; NCT04213469

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P102 DAY7-CART COUNT ENHANCES PREDICTIVE POWER OF PET30 AND POSSIBLY ANTICIPATES CD19 LOSS IN RELAPSE AFTER CART

Fabian Müller 1,2, Viktoria Blumenberg3,2, Veit L. Bücklein3,2, Ralph A. Bundschuh4,2, Dennis Harrer5,2, Klaus Hirschbühl4,2, Johannes Jung6,2, Wolfgang Kunz3,2, Igor Yakushev7,2, Anna Lena Illert6,2, Simon Völkl1,2, Rainer Claus4,2, Leo Hansmann5,2, Judith Hecker6,2, Dirk Hellwig8,2, Andreas Mackensen1,2, Marion Subklewe3,2, Michael Beck1,2

1University Hospital of Erlangen, Erlangen, Germany, 2Bavarian Cancer Research Center, Resp. Site, Germany, 3LMU University Hospital, LMU Munich, Munich, Germany, 4University Hospital of Augsburg, Augsburg, Germany, 5Clinic III Internal Medicine, University Hospital Regensburg, Regensburg, Germany, 6Klinikum Rechts der Isar, Technische Universität München, München, Germany, 7Klinikum Rechts der Isar, Technische Universität München, Munich, Germany, 8University Hospital of Regensburg, Regensburg, Germany

Background: CD19. CAR T cell therapy (CAR-T) has revolutionized current treatment practice in relapsed/refractory diffuse-large B cell Lymphoma (DLBCL) and transformed follicular lymphoma (tFL) by achieving cure in previously palliative situation. Despite this progress, predicting patients benefitting from CAR-T remains challenging. Independently, higher cell numbers at day 7 of CAR-T cell expansion (CAR-peak, at least 20/µl), as well as a negative PET scan as early as day 30 (PET30) have shown to be individually of high predictive power for lasting responses.

Here, we aimed to combine PET30 and d7-CAR-T-count to test whether they support each other’s predictive power.

Methods: DLBCL patients were treated with Tisa-cel, Axi-cel, or Liso-cel at 5 university hospitals of the Bavarian center of cancer research (BZKF) where clinical data was collected. CAR T cell numbers were assessed by flow cytometry. PET-CT scans were analyzed in a standardized manner among the five individual sites. The more recently implemented PET30 and the PET90 were analyzed.

Results: Overall 67 patients with DLBCL-NOS and four with tFL, together with a median age of 64.4 years were treated with CAR-T. 64 patients received bridging therapy to which 33 patients responded (PR/CR) and 30 did not (SD/PD, 8 missing data). 5 patients had an ECOG of 2 + , and 36 an IPI of 3+ at lymphodepletion when mean of LDH was 352 U/L, of CRP was 8.8 mg/L, and of ferritin was 808.6 ng/mL. By ROC analyses, response to bridging and the metabolic-IPI before CAR-T as well as PET30 and CAR-peak after infusion strongly predicted durable response. For 30 of the 71 patients, both, PET30 and day7-CAR-T-count were available. Among those, best long-term outcome was observed in patients with >20 CAR-T cells/µl and negative PET30 (8 patients). Only one/eight patients relapsed at month 5 and was CD19-negative. In contrast, none of the patients with positive PET30 and low Day7-CAR-T-count showed long-term remission. Of the 12 patients with positive PET30 and high CARs, one converted to CR, two died of unknown cause (NRM), one died of CAR-toxicity, and 8 relapsed. 6 of those 8 relapses were biopsied of which 3 presented with CD19 loss. Lastly, of 4 patients with negative PET30 and low Day7-CAR-count, one is in lasting remission, the others relapsed CD19 positive, where assessed.

Conclusions: In this small, retrospective cohort, PET30 and Day7-CAR-T-count greatly support each other in predicting lasting remission vs. relapse. In addition, relapse after high-CAR peak was associated with CD19 loss in 4 of 7 cases, whereas relapse after low CAR-peak was not possibly suggesting distinct resistance mechanisms.

Clinical Trial Registry: N/A

Disclosure: This research was funded by the Bavarian Cancer Research Center.

FM received honoraria and served on advisory boards for BMS, Janssen, Kite, Kyverny, Miltenyi, and Novartis and receied research support from AstraZeneca and Kite.

VBlu received research funding from Bristol Myers Squibb (BMS)/Celgene, Kite/Gilead, Janssen, Novartis, Roche, and Takeda and research funding and honoraria/consultancy fee from Kite/Gilead, Janssen, and Novartis.

VBue received research funding and/or honoraria from Amgen, Celgene/BMS, Kite/Gilead, Novartis, and Pfizer.

AM received honorarai and served on advisory boards for BMS, Janssen, Kite, Kyverny, Miltenyi, and Novartis and receied research support from Kyverna and Miltenyi.

MS received research funding, speaker’s bureau fee, and/or consultancy fee from Amgen, AstraZeneca, Aven Cell, BMS/Celgene, CDR-Life, Kite/Gilead, GlaxoSmithKline, Ichnos Sciences, Incyte Biosciences, Janssen, Miltenyi Biotec, Molecular Partners, Novartis, Pfizer, Roche, Seattle Genetics, and Takeda.

RB, DH, KH, JJ, WK, IY, ALI, SV, RC, LH, DH, and MB have no relevant conflict of interest.

4: CAR-based Cellular Therapy – Clinical

P103 EXCELLENT OUTCOME OF CAR-T CELL THERAPY IN CHILDREN WITH PB-ALL RELAPSING POST HSCT, WHO HAD VERY LOW MRD AT LYMPHODEPLETING CHEMOTHERAPY – POLISH CENTRES’ EXPERIENCE

Karolina Liszka 1, Paweł Marschollek1, Iwona Dachowska - Kałwak1, Monika Mielcarek-Siedziuk1, Jowita Frączkiewicz1, Anna Panasiuk1, Igor Olejnik1, Natalia Haze1, Monika Richert-Przygońska2, Krzysztof Czyżewski2, Robert Dębski2, Jan Styczyński2, Krzysztof Kalwak1

1Wroclaw Medical University, Wrocław, Poland, 2Nicolaus Copernicus University in Torun, Ludwik Rydygier Collegium Medicum in Bydgoszcz, Bydgoszcz, Poland

Background: CAR-T cell therapy brought about a revolution in pB-ALL treatment, improving the survival chances of children in whom the conventional options of therapy failed. Although the outcome is impressive (the newest ELIANA trial update results show 3-year EFS of 44% and OS of 63%, respectively), there’s still a question how to use the whole potential of CAR-T cells. CAR-T cell therapy is effective in treatment of pB-ALL relapse post HSCT, however acc. to Bader et al., outcome in patients with very early relapse (<6 months post HSCT) is significantly worse. In our series it was aimed to achieve the lowest possible MRD before lymphodepleting chemotherapy (LDC) independently from interval between HSCT and relapse.

Methods: We included 18 patients (10 boys, 8 girls, aged 2-17 yrs) with pB-ALL who relapsed post HSCT and were treated with tisagenlecleucel in Polish centres in Wroclaw and Bydgoszcz in years 2020-2023. All patients were severely pretreated, 3 children underwent two HSCT procedures. In 8 patients it was the first relapse, in 7 patients the second, in 2 children the third relapse and 1 patient relapsed four times before CAR-T cells infusion. Median time range between HSCT and relapse prior to CAR-T cell therapy was 9.3 months (range 2-132 months). Negative MRD before LDC was achieved in 5 patients, in others it ranged from below 10-4 to 1x10-2. Different strategies were used as bridging therapy – conventional chemotherapeutics and/or immunotherapeutic drugs.

Results: In the analysed cohort, 16 out of 18 (88%) children remain alive and well with negative MRD. In this group, median time between HSCT and relapse was 10.3 months. Three patients relapsed earlier than 6 months post HSCT. Two CD19-negative relapses were observed – both children died. Median observation time in presented patients is 12 months, 2-year OS is 82.5% and 2-year EFS is 83.6%. No CRS/ICANS ≥ grade III were observed.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (13)

Conclusions: Very low disease burden before LDC, stated as CR with negative or low MRD, ensured excellent 2-year OS and EFS. CAR-T cell therapy increases the chance for cure in high-risk patients, in whom HSCT had not guaranteed long-lasting remission. To enhance the probability of optimal outcome, the goal should be to obtain as low disease burden before LDC as possible, ideally with negative MRD. Every patient requires an individualized approach to bridging therapy but our results highlight the need for maximal possible reduction in MRD before LDC in patients relapsing post HSCT, especially when they relapse early. CR before CAR-T cells infusion not only lowers the risk of relapse but also minimizes the occurrence of post infusion complications such as CRS, ICANS or HLH.

Disclosure: Karolina Liszka – no conflict of interest

Paweł Marschollek – no conflict of interest

Iwona Dachowska – Kałwak – no conflict of interest

Monika Mielcarek-Siedziuk – speaker’s bureau: Novartis, medac, Amgen

Jowita Frączkiewicz – speaker’s bureau: Gilead

Anna Panasiuk – no conflict of interest

Igor Olejnik – no conflict of interest

Natalia Haze – no conflict of interest

Monika Richert-Przygońska - Lecture fees: Novartis, Gilead. Conference participation: Gilead

Krysztof Czyżewski – Lecture fees: Novartis, Gilead. Conference participation: MSD.

Robert Dębski - Lecture fees: Novartis

Jan Styczyński – lecture fees: Novartis, Gilead, MSD, AstraZeneca. Advisory Board: MSD, AstraZeneca, Pfizer, Moderna. Conference participation: medac, Roche

Krzysztof Kałwak – speaker’s bureau: medac, Novartis, Pierre Fabre, Neovii. Advisory Board: Pfizer, MSD

4: CAR-based Cellular Therapy – Clinical

P104 THE DARK SIDE OF CORTICOSTEROIDS IN CD19. CAR-T CELL THERAPY

Marcello Roberto 1,2, Francesco Iannotta1, Francesco De Felice1,2, Beatrice Casadei1, Gianluca Storci1, Serena De Matteis1, Noemi Laprovitera1, Francesca Vaglio1, Barbara Sinigaglia1, Enrica Tomassini1, Enrico Maffini1, Mario Arpinati1, Francesco Barbato1,2, Margherita Ursi1,2, Salvatore Nicola Bertuccio1, Daria Messelodi1, Irene Salamon1, Maria Naddeo1, Elisa Dan1, Luca Zazzeroni1, Cinzia Pellegrini1, Matteo Carella1,2, Marianna Gentilini2,1, Pier Luigi Zinzani1,2, Massimiliano Bonafè1,2, Francesca Bonifazi1

1IRCCS-Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy, 2University of Bologna, Bologna, Italy

Background: Chimeric antigen receptors (CAR)-T cells represent an advanced therapy targeting the CD19 antigen on B cells, with consistent efficacy and relative safety in patients diagnosed with relapsed or refractory (r/r) B‐cell lymphomas. Corticosteroids are often used for the management of CAR-T cell toxicities. However, conflicting data are described about their harmful impact on CAR-T cell therapy and patient’s outcome. This study aimed to explore the role of corticosteroids after infusion in overall survival (OS) and progression-free survival (PFS).

Methods: The study cohort included seventy-five patients with r/r B-cell lymphomas admitted to the “advanced cell therapy unit of IRCCS_AOU of Bologna” for CD19. CAR-T cell therapy. The median age was 60 years (19-71). The cell product was axi-cel (n=45, 60%) or tisa-cel (n=30, 40%). After infusion, 63 (84%) developed cytokine release syndrome (CRS) of any grade: 40 (53%) developed grade 1; 18 (24%) grade 2; 5 (7%) grade ≥3. Twenty-two (29%) patients developed immune effector cell-associated neurotoxicity syndrome (ICANS) of any grade: 6 (8%) developed grade 1, 7 (9%) grade 2, 9 (12%) grade ≥3. Thirty-eight (51%) patients received corticosteroids within 30 days after infusion for CRS and/or ICANS management (not for any other indications). Cumulative corticosteroid doses were reported as methylprednisolone-equivalent (median 1120 mg, range 380-13035). Available blood samples after CAR-T cell infusion were analyzed by FACS for CAR+T cell tracking.

Results: As expected, we found a strong association between corticosteroid administration after infusion and severe toxicities (grade ≥2 CRS and/or any grade ICANS occurrence). Thus, in order to determine the contribution of these variables on the time-dependent outcomes (OS and PFS), their distributions were marginalized. At first, we selected a subgroup of patients without severe toxicities (n=45) experiencing at most persistent grade 1 CRS. When comparing patients who received steroid (n=12; median methylprednisolone-equivalent dose was 1002.5 mg) with those who did not receive it (n=33), no significant differences in OS (p=0.80) and PFS (p=0.08) were found. Next, to address the impact of severe toxicities we focused on patients receiving corticosteroid (n=38) verifying that only grade ≥2 CRS impacted on OS (p=0.04) but not on PFS (p=0.08). Noteworthy, in a multiple Cox regression model (also including age, disease histotype, stage and bulky, CAR-T cell product, bridging therapy response, pre-lymphodepletion LDH levels) grade ≥2 CRS did not retain its significativity, while cumulative corticosteroid dose was associated with OS (p=0.002) and PFS (p=0.018). Finally we found no correlation between cumulative corticosteroid dose and CAR+T cell peak and area under curve (AUC0-30) levels as measured by MFC.

Conclusions: This evidence suggests that a higher cumulative corticosteroid dose after CAR-T cell infusion may have a harmful effect in OS and PFS, without compromising CAR+T cell expansion. Further analyses in larger cohorts are needed to confirm this result and to investigate the detrimental effect of steroids on CAR-T cell activity rather than quantity.

Disclosure: FB: scientific advisory boards and speaker fees: NEOVII, NOVARTIS, KITE, GILEAD, PFIZER, CELGENE, MSD.

PLZ: scientific advisory boards: Secura Bio BIO, Celltrion, Gilead, Janssen-Cilag, BS, Servier, Sandoz, MSD, TG Therap., Takeda, Roche, EUSA Pharma, Kiowa Kirin, Novartis, ADC Therap., Incyte, Beigene; consultancy: EUSA Pharma, MSD, Novartis; speaker’s bureau: Celltrion, Gilead, Janssen-Cilag, BMS, Servier, MSD, TG Therap., Takeda, Roche, EUSA Pharma, Kiowa Kirin, Novartis, Incyte, Beigene.

The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

4: CAR-based Cellular Therapy – Clinical

P105 MATCHING-ADJUSTED INDIRECT COMPARISON OF EFFECTIVE CHARACTERISTICS AMONG DIFFERENT BCMA TARGETING CAR-T IN TREATMENT OF RELAPSED OR REFRACTORY MULTIPLE MYELOMAS

Chunrui Li 1, Lu-Gui Qiu2, Di Wang1, Yongping Song3, He Huang4, Jianyong Li5, Bing Chen6, Jing Liu7, Xi Zhang8, Yu-Jun Dong9, Kai Hu10, Peng Liu11, Jian-Qing Mi12, Kaiyang Ding13, Zhenyu Li14, Qie Dong15, Fuyuan Zhang15, Guang Hu15, Wen Wang15

1Institute of Hematology; Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China, 2Chinese Academy of Medical Sciences and Peking Union Medical College, State Key Laboratory of Experimental Hematology, National Clinical Research Center for Blood Diseases, Haihe Laboratory of Cell Ecosystem, Tianjin, China, 3The Affiliated Cancer Hospital of Zhengzhou University, Henan Cancer Hospital, Zhengzhou, China, 4The First Affiliated Hospital, Zhejiang University, Hangzhou, China, 5The First Affiliated Hospital of Nanjing Medical University, Nanjing, China, 6Nanjing Drum Tower Hospital, Affiliated Hospital of Nanjing University Medical School, Nanjing, China, 7Third Xiangya Hospital of Central South University, Changsha, China, 8Medical Center of Hematology, Xinqiao Hospital, State Key Laboratory of Trauma, Burn and Combined Injury, Army Medical University, Chongqing, China, 9Peking University First Hospital, Beijing, China, 10Beijing Boren Hospital, Beijing, China, 11Zhongshan Hospital, Fudan University, Shanghai, China, 12State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Shanghai Institute of Hematology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China, 13The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China, 14Affiliated Hospital of Xuzhou Medical University, Xuzhou, China, 15Nanjing IASO Biotechnology Co., Ltd., Nanjing, China

Background: Several studies on BCMA targeting CAR-T cell therapies have disclosed their results, each with different characteristics in efficacy. These BCMA CAR-T cell therapies have been used in different marketing and clinical settings. However, there are no controlled studies directly comparing different CAR-T cell therapies. This analysis used matching-adjusted indirect comparisons (MAIC) to evaluate the comparative efficacy of Equecabtagene Autoleucel (Eque-cel) versus Ciltacabtagene Autoleucel (Cilta-cel), Idecabtagene vicleucel (ide-cel), zevorcabtagene autoleucel (zevor-cel, CT053) in patients with relapsed or refractory multiple myeloma.

Methods: Primary data sources included individual patient data from the FUMANBA-1 study (NCT05066646; N=103 for efficacy set) for Eque-cel and summary-level data from the CARTITUDE-1 study (NCT03548207; N=97 for efficacy set) for cilta-cel; data from CARTIFAN-1 study (NCT03758417; N=48 for efficacy set) for cilta-cel; data from KARMMA study (NCT03361748; N=128 for efficacy set) for ide-cel; data from LUMMICAR STUDY (NCT03975907 N=102 for efficacy set) for zevor-cel. Inter-study differences in design, eligibility criteria, baseline characteristics, and outcomes were assessed and aligned to the extent feasible. Clinically relevant prognostic factors were adjusted in a stepwise fashion by ranked order.

Results: This analysis revealed many different characteristics of these BCMA CAR-T therapies in RRMM, with eque-cel showing a better trend in MAIC-weighted efficacy outcomes.

The MAIC of the response rates, including overall and complete response rates and MRD negativity rate, were compared by odds ratio (OR) and 95% confidence intervals (CI). In FUMANBA-1 study versus CARTIFAN study, eque-cel showed a better trend than cilta-cel for overall response rate. When FUMANBA-1 study was compared to LUMMICAR study, eque-cel showed a better trend than zevor-cel for overall response rate and significantly better for complete response rate. In FUMANBA-1 study versus CARTITUDE-1 study, due to very limited effective sample size resulting from different patient characteristics, no clear trend was detected. When FUMANBA-1 study was compared to KARMMA study, eque-cel showed a better trend than ide-cel in overall response rate, complete response rate, and particularly MRD negativity rate, even with a limited effective sample size. The specific data is shown in the following table.

Because the median PFS was not reached in FUMANBA-1 study, 12-month PFS rate and 12-month MRD negativity maintaining rate were calculated by MAIC-weighted methods. Eque-cel showed higher 12-month PFS rate and MRD negativity maintaining rate after such adjustments. The specific data is also shown in the following table.

Table Summary of ORR,≥CR rate, MRD negativity rate,12 month PFS rate, and 12 month MRD negativity maintaining rate

FUMANBA-1(eque-cel)

versus

CARTIFAN(cilta-cel)

FUMANBA-1(eque-cel)

versus

LUMMICAR (zevor-cel)

FUMANBA-1(eque-cel)

versus

KARMMA(ide-cel)

FUMANBA-1(eque-cel)

versus

CARTITUDE1(cilta-cel)

MAIC(ESS*=75)

MAIC(ESS=49.4)

MAIC(ESS=10.7)

MAIC(ESS=7.2)

OR (95%CI)

OR (95%CI)

OR (95%CI)

OR (95%CI)

ORR

5.65 (0.94,33.9)

5.17(0.69,38.82)

11.57 (0, inf)

0.19 (0, inf)

≥CR rate

1.15(0.49,2.71)

6.08 (2.88, 12.83)

3.20 (0, inf)

0.29 (0, inf)

MRD negativity rate

0.8(0.08,8.36)

/

28.59 (0, inf)

/

12-month PFS rate

82.3%

93.2%

75.8%

94.2%

12-month sustained MRD negativity

77.8%

76.4%

84%

100%

  1. *ESS: effective sample size

Conclusions: After matching and adjusting for clinically relevant prognostic factors, eque-cel demonstrated a better trend of short-term efficacy when compared to cilta-cel of CARTIFAN study, ide-cel of KARMMA study and zevor-cel of LUMMICAR study. Eque-cel also showed a favorable long-term efficacy in patients with heavily pretreated relapsed or refractory MM.

Disclosure: Nothing to declare. Professor Lu-Gui Qiu is the corresponding author of this study.

4: CAR-based Cellular Therapy – Clinical

P106 IMPACT OF EARLY CAR CD4+ T-LYMPHOCYTES RECOVERY FOLLOWING CAR-T CELL INFUSION FOR RELAPSED OR REFRACTORY B CELL LYMPHOMAS

Massimiliano Gambella 1,2, Simona Carlomagno2, Rosa Mangerini1, Nicoletta Colombo1, Alessia Parodi1, Chiara Ghiggi1, Livia Giannoni1, Elisa Coviello1, Chiara Setti2, Silvia Luchetti1, Alberto Serio1, Antonella Laudisi1, Monica Passannante1, Alessandra Bo1, Elisabetta Tedone1, Simona Sivori2, Emanuele Angelucci1, Anna Maria Raiola1

1IRCCS Ospedale Policlinico San Martino, Genova, Italy, 2University of Genoa, Genova, Italy

Background: CAR CD4+ T cell lymphopenia is an emerging issue following CAR-T infusion. We focused on potential determinants of CD4+ T cell recovery and investigated a possible association with survival. We also described the kinetics of CAR CD8+ T, NK, and NKT cell recovery following CAR-T therapy for aggressive B-cell lymphomas.

Methods: We investigated a cohort of patients affected by Diffuse Large B-cell Lymphoma or Mantle Cell Lymphoma, treated at IRCCS Ospedale Policlinico San Martino, Genova, with commercially available CAR-T products. Peripheral blood immune cell subpopulations were characterized before lymphodepletion and following CAR-T infusion through multiparametric-flow cytometry, primarily focusing on CAR lymphoid subpopulations. Infectious complications were analyzed according to CD4 T cell recovery.

Results: Thirty-one consecutive patients were selected for the analysis. Six-month cumulative incidence of CAR CD4+ T cell recovery (i.e., ≥ 200cells/μl) was 0.43 (95% CI: 0.28-0.65). Among possible determinants of CD4+ T cell recovery, we recognized infusion of a 4-1BB product (tisagenlecleucel, TSA) in comparison with CD28-based ones (axicabtagene/brexucabtagene, AXI/BRX) (hazard ratio [95% CI]: 5.79 [1.16-24.12] p= 0.016). Higher CD4+ T cell counts resulted with TSA at month-1, -2, and- 3 following infusion. Moderate to severe infectious complications were registered in patients with prolonged CD4+ T cell lymphopenia.

We identified an association between early, month-1 CD4+ T cell recovery and a worse clinical outcome in the DLBCL cohort, upheld in a multivariate regression model for Overall Survival (HR: 4.46 [95% CI: 1.12-17.71], p= 0.03). The association was identified within both products.

Conclusions: We conclude that a faster CAR CD4+ T cell recovery is associated with TSA as compared to AXI/BRX. Moreover, our data suggest that early, month-1 CAR CD4+ T cell subset recovery could represent a “red flag” for CAR-T cell therapy failure in DLBCL patients.

Disclosure: No potential conflict of interest to declare

4: CAR-based Cellular Therapy – Clinical

P107 ACUTE INFECTION AND BASELINE C-REACTIVE PROTEIN – KEY MARKERS OF PROGRESSION IN RELAPSED REFRACTORY DLBCL PATIENTS RECEIVING CAR-T CELL THERAPY

Jayachandran Perumal Kalaiyarasi 1, Khalil Begg1, Luminita Keating1, Louise Corbett1, Paul Boraks1, Brendan OSullivan1, Andrew King1, Charles Crawley1, Benjamin Uttenthal1, Ram Malladi1

1Addenbrookes Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom

Background: Chimeric antigen receptor -T cell (CAR-T) therapy products Axicabtagene ciloleucel (Axi-cel) and Tisagenlecleucel (Tisa-cel) are approved as 3rd line therapy in relapsed/refractory diffuse large B-cell lymphoma (RR-DLBCL) patients. Multiple factors including tumour volume, extranodal sites, advanced stage, inflammatory markers, and refractory disease have been described to adversely influence survival outcomes. We have analysed the outcomes of RR-DLBCL patients who underwent CAR-T therapy in our centre and have identified Acute Culture Positive Infections within Day 28 of CAR-T infusion (ACPI28) and C-Reactive protein (CRP) at commencement of lymphodepletion as key factors influencing the early disease progression.

Methods: The clinical details of 50 consecutive patients who underwent CAR-T therapy in 3rd line for RR-DLBCL from December 2020 to June 2023 were collected from records. Survival outcomes were analysed by Kaplan-Meier Method and associations between variables influencing survival were tested using the Log-Rank Test.

Results: Median age at CAR-T infusion was 65 years. 56% were male. 24% had had prior ASCT. Axi-cel was infused in 90% of the patients. Cytokine release syndrome (CRS) was seen in 100% (Grades 2/3/4 - 42%/10%/0%). Grade 3/4 Immune effector cell therapy-associated neurotoxicity (ICANS) was seen in 20%.

ACPI28 were seen in 5 patients (10%). Of these 5 patients, 4 were blood-stream infections (Pseudomonas–2 patients, Enterococcus–1 patient, and one patient had both) and one was a urinary tract infection (Enterococcus). Out of 5 ACPI28 patients, progressive/relapsed disease (PD) and early death were seen in 3 and 1 patients respectively.

At 1 month and 3 months post CAR-T, PET-CT showed complete metabolic response (CMR) in 56% and partial response (PR) in 24%. At 3 months, 56% had CMR, 4% had PR. At the time of data cut-off, 20 had PD. Overall non-relapse mortality was 8%. Median follow-up was 17.7 months. Median progression-free survival (PFS) was 12.4 months (1-year PFS-52.7%); median overall survival (OS) was not reached (1-year OS-68.6%).

In univariate analysis, the only factors adversely influencing PFS were ACPI28 (p=0.007), elevated CRP before lymphodepletion (p=0.01), and non-achievement of CMR/PR at 1 month and 3 months (p<0.001). The factors adversely influencing the OS were elevated CRP at the baseline (p=0.005), ACPI28 (p=0.04), and non-achievement of CMR/PR at 3 months (p<0.001) (Table 1). Elevated CRP did not influence risk of CRS or ICANS but adversely influenced PFS and OS. Grade of CRS/ ICANS, baseline lactate dehydrogenase, response before CAR-T therapy, and use of tocilizumab/dexamethasone did not influence survival outcomes.

Full size table

Conclusions: We have identified 5 culture-positive bacterial infections within 28 days following CAR-T infusion. This study provides evidence for an unexpected association between this ACPI28 and risk of early lymphoma progression. We speculate that this could be due to poor CAR-T expansion due to adverse cytokine profile precipitated by the immune response to severe infection. Our observed association between raised baseline CRP and disease progression may reflect a mechanism of immune-suppressive environment due to inflammation. Our statistical analysis is limited to univariate analysis due to small sample size. This observation, however, warrants further exploration in a larger cohort suitable for multivariate analysis.

Disclosure: Nothing to Declare

4: CAR-based Cellular Therapy – Clinical

P108 THIRD-LINE CAR-T IN PATIENTS WITH EARLY RELAPSE FOLLOWING FIRST-LINE TREATMENT FOR AGGRESSIVE B-CELL LYMPHOMA: STILL A VALUABLE OPTION

Massimiliano Gambella 1, Anna Maria Raiola1, Livia Giannoni1, Elisa Coviello1, Chiara Ghiggi1, Anna Ghiso1, Riccardo Varaldo1, Stefania Bregante1, Carmen Di Grazia1, Alida Dominietto1, Antonella Laudisi1, Monica Passannante1, Silvia Luchetti1, Alberto Serio1, Alessandra Bo1, Emanuele Angelucci1

1IRCCS Ospedale Policlinico San Martino, Genova, Italy

Background: Following ZUMA-7 and TRANSFORM trials, CAR-T is the first choice for high-risk aggressive B cell lymphoma, intended as refractory to or relapsing within 12 months from first-line chemo-immunotherapy. Despite crossover to third-line CAR-T in roughly 50% of patients from the comparative arm, second-line CAR-T showed an advantage both in EFS and OS. Whether third-line CAR-T in high-risk patients is still an option for some subgroups, has to be clarified.

Methods: Thirty-eight consecutive patients affected by aggressive B cell lymphoma, treated at our center from October 2020 to November 2023 with a third-line anti-CD19 CAR-T (tisagenlecleucel or axicabtagene ciloleucel) were analyzed. Patients previously refractory to or relapsing within 12 months after a first-line, anthracycline-based (i.e., R-CHOP or intensified regimens) chemo-immunotherapy (CIT) were considered. Primary mediastinal lymphomas were excluded. Patients were categorised according to first-line response: refractory (ref-group; residual/progressive disease) vs relapse (rel-group; relapse within 12 months from treatment, following a documented complete metabolic remission). Chi-squared test was used to confront categorical variables. Progression-free survival (PFS) was calculated from infusion with the Kaplan-Meier estimator and groups compared using two-sided log-rank test. Censored patients for PFS had at least a 3-month follow-up duration following CAR-T infusion. Cox regression model was used to provide estimated hazard ratios.

Results: Eighteen patients fulfilled the pre-specified criteria, 13 refractory and 5 relapses. First-line CIT consisted of R-CHOP in 9/18 (50%) patients. An intensified regimen (R-DA-EPOCH or R-CODOX/R-IVAC) was administered in 6/13 (46%) patients in the ref-group and in 3/5 (60%) in the rel-group. The whole cohort received platinum-based chemotherapy as salvage treatment (R-DHAOx, 17 patients; R-ICE, 1 patient); all but one (17/18, 95%) did not respond, the sole responding patient relapsed 6 months after autologous transplant.

Third-line CAR-T consisted of tisagenlecleucel in 10/13 (77%) patients in the ref-group and 3/5 (60%) in the rel-group. At lymphodepletion, the two groups were comparable for CAR-T detrimental variables: LDH≥2ULN (p=0.45), extranodal sites ≥3 (p=0.81), bridging therapy response (p=0.71). No patients died of complications following CAR-T.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (14)

At a median follow-up of 11 months (95%CI: 4.5-17.4), relapse was reported in 10/13 (77%) patients in the ref-group and in 1/5 (20%) patients in the rel-group. Progression-free survival analysis showed a significant deterioration in the ref-group (HR: 8.24 95%CI: 0.97-69.37). The estimated 12-month PFS was 23.1% (95%CI: 2-46) in the ref-group and 75% (95%CI: 34-NR) in the rel-group (p= 0.02) (figure).

Conclusions: We compared outcomes following third-line CAR-T according to failure dynamics after first-line CIT, in the context of high-risk disease (refractoriness or ≤ 12 months relapse). Refractory disease had a deleterious impact on CAR-T outcomes, representing the group that mostly needs CAR-T anticipation to second line. In contrast, third-line CAR-T seems a good option for patients relapsing within 12 months from first-line CIT. In a prospective view, an attempt to standard salvage CIT could still be considered in relapsing patients.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P109 EASIX SCORE AS A PROGNOSTIC FACTOR OF CYTOKINE RELEASE SYNDROME IN CAR-T PATIENTS - A RETROSPECTIVE ANALYSIS OF MM AND LBCL COHORTS

Jaromir Tomasik 1, Batia Avni2, Sigal Grisariu3, Shlomo Elias3, Eran Zimran3, Polina Stepensky3, Grzegorz Basak1

1Medical University of Warsaw, Warsaw, Poland, 2Hadassah Medical Center, Jarusalem, Israel, 3Hadassah Medical Center, Jerusalem, Israel

Background: Endothelial dysfunction has been associated with complications following stem cell transplantation and chimeric antigen receptor (CAR)-T therapy. Due to the lack of easily available endothelium-related markers in clinical practice, the Endothelial Activation and Stress Index (EASIX) has been developed and proposed as a prognostic factor of adverse events or survival in hematological malignancies.

Methods: The aim of this retrospective cohort study was to assess the utility of the EASIX score as a prognostic factor of cytokine release syndrome (CRS) grade ≥3 in multiple myeloma/light chain amyloidosis (MM/AL amyloidosis) and large B-cell lymphoma (LBCL) cohorts. The first cohort (N=69) consisted of patients treated with anti-BCMA CAR-T therapy against MM (N=64) or AL amyloidosis (N=5). The second cohort (N=65) included patients treated with anti-CD19 CAR-T therapy against LBCL, specifically tisagenlecleucel/tisa-cel (N=55) and axicabtagen ciloleucel/axi-cel (N=10) administered as standard-of-care treatment. Occurrence of CRS grade ≥3 was considered as the primary endpoint. For both cohorts, the EASIX [(lactate dehydrogenase × creatinine) / platelets)] and simplified EASIX [s-EASIX, (lactate dehydrogenase) / platelets] score was calculated at four different time points before CAR-T infusion to assess its prognostic value.

Results: In the LBCL cohort, EASIX and s-EASIX scores measured before lymphodepletion (EASIX-pre and s-EASIX-pre) showed a significant relationship with CRS grade ≥3 (OR=1.06, p=0.027 and OR=1.05, p=0.026, respectively). The cutoff value of 1.835 for EASIX-pre was associated with 4,59-fold increased OR of CRS grade ≥3 (95% CI: 1.13-21.84, p=0.033), whereas s-EASIX-pre cutoff equaled 2.134 and was associated with 4.13-fold increased OR of CRS grade ≥3 (95% CI: 1.01-17.93, p=0.01). However, after internal validation with bootstrapping, the significance was lost both for the EASIX-pre cutoff (95% CI: 0.98-21.4) and s-EASIX-pre cutoff (95% CI: 0.73-23.26).

In the MM/AL amyloidosis cohort, we found that neither EASIX nor s-EASIX scores calculated at any time point were associated with the occurrence of CRS grade ≥3. An increase in either EASIX or s-EASIX scores did not increase the odds of developing more severe CRS.

Conclusions: In this multicenter retrospective study, we demonstrate that EASIX and s-EASIX scores should be employed with caution when concerning their utility in CRS prediction. Along with other researchers, we found the significant predictive value of the EASIX and s-EASIX scores (before lymphodepletion) for CRS grade ≥ 3 in the LBCL CAR-T cohort. On the contrary, these scores seem to have no value in the prediction of CRS in MM/amyloidosis CAR-T patients.

Disclosure: The authors declare no conflicts of interests

4: CAR-based Cellular Therapy – Clinical

P110 HEMATOTOXICITY POST CAR-T CELL THERAPY: REAL-WORLD DATA ON RISK FACTORS AND OUTCOMES

Zoi Bousiou 1, Ioannis Kyriakou1, Anna Vardi1, Despoina Mallouri1, Eleni Gavriilaki1, Christos Demosthenous1, Nikolaos Spyridis1, Alkistis-Kyra Panteliadou1, Eleni Papchianou1, Georgios Karavalakis1, Evangelia Yannaki1, Ioannis Batsis1, Ioanna Sakellari1

1G.Papanikolaou Hospital, Thessaloniki, Greece

Background: CAR-T cells have a unique toxicity profile, with cytopenia being a frequent but underestimated one. Therefore, we aimed to investigate risk factors and outcomes of hematotoxicity.

Methods: We analyzed hematotoxicity rates in consecutive CAR-T cell recipients in our JACIE-accredited center. Evaluation started from lymphodepletion until disease progression needing treatment or last follow-up. CAR-HEMATOTOX and IACHT (immune effector cell-associated hematotoxicity) grading were retrospectively calculated, and possible association with disease- and cellular product-related risk factors and outcomes was studied.

Results: Forty-five patients (29 male, 16 female, median age 47 years) received Axi-cel (24), Tisa-cel (14) or Brexu-cel (7) for DLBCL (30), PMBCL (3), MCL (6) or ALL (6) using Fludarabine and Cyclophosphamide for lymphodepletion. Prior transplantation was reported in 7 (auto=4, allo=3), bone marrow infiltration in 9 patients. The majority was heavily pretreated (median 3 lines of treatment, 2-9). Baseline cytopenia was found in 71% (n=32), affecting one (28.9%), two (22.2%) or all three (24.4%) lines; CAR-HEMATOTOX score was low in 26 (57.8%). Before infusion, 53% developed grade 3 cytopenia. CRS presented in 41 patients (grade ≥3=6), and ICANS in 16 (grade≥3=3), managed with tocilizumab in 80%, in combination with steroids in 51%. Hematotoxicity gr≥3 occurred in 39 patients (84%), either as early IACHT (n=37), late IACHT (n=20), or both (n=17), affecting mainly neutrophils (87%) and platelets (40%), while only 22% had anemia. All patients with anemia or thrombocytopenia had also neutropenia.

CAR-HEMATOTOX score was associated with early (p=0.015) and late IACHT (p=0.034). The only independent predictors of both IACHT and IACHT gr≥3 were CAR-HEMATOTOX score (p=0.024 and p=0.01), number of cytopenias (p=0.028, p=0.006) and development of ICANS (p=0.003, p=0.003, respectively).

Presence of IACHT was not associated with infections (p=0.836), or febrile neutropenia (p=0.169), even in cases of late (p=0.734 and 0.343), or late grade ≥3 (p=0.969 and 0.482). G-CSF was commenced 10 days after infusion if neutropenia was present, with median duration of 46 days (1-360), until recovery or progression. Eltrombopag has been administered in one patient without a response yet. G-CSF use before day 14 was not associated with CRS or ICANS and had no impact on early or late IACHT. Neutrophil recovery was achieved in 65% at a median of 50.5 days (15-180). Anemia and thrombocytopenia have not improved only in in 1 and 3 patients respectively.

With a median follow-up of 9 months (1-42), 56% (25) achieved complete and 27% partial disease response (12). Hematotoxicity did not affect response or disease relapse even when duration of cytopenia was >1 month. One-year OS is 72.7% with treatment-related mortality 6.7%. Only 1 death has been attributed to bacterial infection and the remaining 2 deaths to CRS.

Conclusions: Although hematologic toxicity post cellular therapy is common, CAR-HEMATOTOX score can predict high-risk patients. IACHT does not predispose to infection or disease response. Additional studies are needed to elucidate the pathophysiology of cytopenia and facilitate early interventions to improve outcomes in these patients.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P111 DONOR-DERIVED CLL-1 CHIMERIC ANTIGEN RECEPTOR T-CELLS FOR RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA BRIDGING TO ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A CASE REPORT UPDATE

Xiaojuan Miao1, Yanrong Shuai1, Ying Han1, Yilan Liu1, Nan Zhang1, Hao Yao1, Xiao Wang1, Guangcui He 1, Dan Chen1, Fangyi Fan1, Alex H. Chang2, Yi Su1, Hai Yi1

1Peoples Liberation Army The General Hospital of Western Theater Command, Chengdu, China, 2Tongji University School of Medicine, Shanghai, China

Background: The prognosis of relapsed/refractory acute myeloid leukemia (R/R AML) is still poor and the treatment remains a challenging issue. To explore the efficacy and safety of donor-derived CLL-1 chimeric antigen receptor (CAR) T-cell therapy for R/R AML followed by allogeneic hematopoietic stem cell transplantation (allo-HSCT) bridging upon remission.

Methods: A young male patient (18 years old) was diagnosed with CEBPA double-mutated AML. After initial chemotherapy, he achieved minimal residual disease (MRD)-negative complete remission (CR), that was followed by auto-HSCT. His leukemia disease was relapsed 6 months later. The salvage therapy with azacitidine and venetoclax was not helpful and his disease was deteriorated with 61% blasts in bone marrow (BM) and 63.18% blasts detected by flow cytometry (FCM) with a phenotype positive for CD34, CD117, HLA-DR, CD13, CD33, CD38, CD123 and CLL-1 upon admission. CAR T-cells targeting CLL-1 were manufactured using peripheral blood from a haploidentical donor, he received a total of 0.5*106/kg CAR T-cells with informed consent. The vital signs, complete blood counts, cytokines, serum ferritin, peripheral blood (PB) CAR T-cell counts, BM aspirate and MRD by FCM were monitored. Allo-HSCT from the same donor was started immediately after CR upon BM evaluation. Blood counts, BM aspirate and MRD, graft-versus-host disease (GVHD), and chimerism status were routinely monitored.

Results: The patient developed fever at day 8 after CAR T-cells infusion, which peaked at 39.2°C. After supportive care, the temperature returned to normal range at day 10. There were no episodes of hypoxia, hypotension or immune effector cell-associated neurotoxicity syndrome (ICANS). The cytokine release syndrome (CRS) was assessed as grade 1. CAR T-cells expanded in PB at the peak of 9.4% in the whole T cells at day 8. The patient achieved CR with 0.29% MRD in BM by FCM at day 11. He was then bridged to allo-HSCT immediately. He achieved neutrophil engraftment at day 18 and platelet engraftment at day 26 after transplantation. Complete donor chimerism was observed at day 23. MRD negative CR in BM was achieved at day 23. The patient is currently 16 months after transplantation, with sustain MRD negative CR in BM, and no extramedullary disease or GVHD.

Conclusions: Donor-derived CLL-1 CAR T-cell is effective and safe for treating R/R AML. Bridging to allo-HSCT immediately following CR may improve prognosis in these patients’ population.

Clinical Trial Registry: (www.chictr.org.cn number, ChiCTR2200058607.)

Disclosure: Nothing to declare.

4: CAR-based Cellular Therapy – Clinical

P112 A RETROSPECTIVE REVIEW TO IDENTIFY OPTIMAL LYMPHODEPLETION PRIOR TO TISAGENLECLEUCIL IN PATIENTS WITH REFRACTORY OR RELAPSED LARGE B-CELL LYMPHOMA IN SCOTLAND’S NATIONAL CAR-T CENTRE

Daire Quinn 1, Jennifer McLellan1, Sinead Connolly1, Grant McQuaker1, Anne Parker1, Dimitris Galopoulos1, Ailsa Holroyd1, Anne-Louise Latif1, David Irvine1

1Queen Elizabeth University Hospital, Glasgow, United Kingdom

Background: The advent of CAR T cell therapy directed against CD19 has radically improved management of patients with relapsed or refractory aggressive B cell lymphomas however the majority of patients treated with CD19 CAR T cell therapy progress post treatment. Lymphodepleting (LD) chemotherapy prior to CAR T cell infusion is a modifiable factor in optimising CAR T therapy through improving the microenvironment facilitating T cell expansion and activity. Inclusion of Fludarabine in LD regimens has improved efficacy and the most commonly used LD regimen is Fludarabine and Cyclophosphamide. The current practice of calculating Fludarabine dosing by body surface area can lead to variability in area under the curve (AUC) exposure in validated models3. AUC exposure correlates with outcomes in patients treated with Tisagenlecleucil (Tisagen) for ALL and Axicabtagene for DLBCL. The recommended doses of LD chemotherapy for Tisagen are lower in comparison to other products. Given these factors we performed a risk assessment in March 2022 and opted to increase LD therapy for Tisagen (Fludarabine 25mg/m^2 to 30mg/m^2 and Cyclophosphamide 300mg/m^2 to 500mg/m^2 each for 3 days) in line with other products. Here we report a retrospective review of all patients receiving Tisagen at the National CAR T service in Scotland before and after this change.

Methods: Retrospective analysis of all (21) patients receiving Flu/Cy lymphodepletion prior to Tisagen for aggressive B cell lymphoma in Scotland from November 2019 – November 2023. Patient characteristics, toxicities and outcome measures were analysed and estimated AUC exposure to Fludarabine was calculated using a previously validated population pharmacokinetics model. PFS and OS were calculated using Kaplan-Meyer estimates.

Results: Ten patients received Tisagen following standard conditioning (cohort 1), 11 received Tisagen following increased conditioning (cohort 2). Median age was 67 in cohort 1 and 70 in cohort 2. The majority were heavily treated including 6 previously transplanted patients. The majority of patients received bridging therapy. Estimated Fludarabine exposure was higher in cohort 2. CAR-T toxicity was low and broadly comparable between cohorts with marginally more grade 3-4 CRS/ICANs but less ICU admissions in cohort 2. Response rates were comparatively high in both cohorts; at 1 month: 40% and 60% achieved CR in cohort 1 and 2 respectively and at 3 months: 60% vs 63% achieved CR in cohort 1 and 2 respectively. Progression free survival (PFS) at 1y was higher in the 2nd cohort compared with the 1st cohort (67% versus 38%) and overall survival was similar (73% vs 70%) with median follow up of 314 and 487 days respectively.

Standard LD therapy (cohort 1)

Increased LD therapy (cohort 2)

Total

Number of patients (n)

10

11

21

Age (median/range)

67 (52-77)

70 (61-74)

68 (52-77)

Stage 1-2 / 3-4

2 / 8

4 / 7

6 / 15

Subtype DLBCL / RT / TFL

8 / 1 / 1

9 / 1 / 1

17 / 2 / 2

Median lines treatment

2.5 (2-4)

2 (2-4)

2 (2-4)

Prior transplant

3

3

6

Elevated LDH

7

7

14

Bridging therapy

8 (80%)

11 (100%)

19 (90%)

Est. AUC Fludarabine/ mgh/L

20.8 (15.7 - 24.3)

24 (15.1-25.8)

21.6 (15.1-25.8)

CRS total / grade 3-4

10 (100%) / 1 (10%)

11 (100% / 2 (18%)

21 (100% / 3 (14%)

ICANs total / grade 3-4

1 (10%) / 0

1 (9%) / 1 (9%)

3 (14%) / 1 (4%)

ICU

3 (30%)

2 (18%)

5 (24%)

Tocilizumab / Steroids

7 (70%) / 3 (30%)

10 (90%) / 4 (36%)

17 (81%) / 7 (33%)

Response 1 month

n= 10

n= 11

n= 21

CR / PR / SD / PD

4 (40%) / 5 (50%) / 1 (10%) / 0

6 (54%) / 3 (27%) / 1 (10%) / 1 (10%)

10 (47%) / 8 (38%) / 2 (10%) / 1 (5%)

Response 3 month

n=10

n=8

n=18

CR / PR / SD / PD

6 (60%) / 1 (10%) / 0 / 3 (30%)

5 (63%) / 0 / 0 / 3 (38%)

11 (61%) / 1 (6%) / 0 / 6 (33%)

PFS at 1y / OS at 1y

38% / 70%

67% / 73%

49% / 70%

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (15)

Conclusions: In this small single centre cohort, Tisagen was well tolerated and resulted in relatively high response rates in comparison to existing outcome data. While small numbers and retrospective data limit interpretation, escalation in LD therapy was associated with a higher estimated exposure to Fludarabine and with an improved PFS. This may point to the potential to improve outcomes in patients undergoing CAR-T therapy by optimising prior LD therapy.

Disclosure: David Irvine: Honoraria from Kite

Annie Latif: Honoraria Kite, Jazz, Daiichi Sankyo, Novartis, Amgen, AbbVie, Astellas

4: CAR-based Cellular Therapy – Clinical

P113 METAGENOMIC NEXT-GENERATION SEQUENCING VISUALLY DISPLAYS A FALSE-POSITIVE DETECTION OF HIV NUCLEIC ACID RELATED TO CART TREATMENT: A CASE STUDY IN A CHILD

M. I. Zhou 1, Lili Huang1, Zhenjiang Bai1, Shuiyan Wu1, Jun Lu1, Shaoyan Hu1

1Children’s Hospital of Soochow University, Suzhou, China

Background: Lentivirus vectors are commonly used to engineer CAR-T cells, but sometimes this can lead to false-positive human immunodeficiency virus (HIV) testing results after CAR-T infusion. However, identifying this false positive result can be confusing.

Methods: We present the first case of false-positive HIV-1 detected by metagenomic next-generation sequencing (mNGS) following CAR-T treatment. We analyzed a case of a five-year-old boy diagnosed with B cell acute lymphoblastic leukemia. The child developed cytokine-release syndrome and CAR-T-cell-related encephalopathy syndrome on day 3 after CAR-T Infusion. Due to the difficulty in distinguishing between the presence of HIV and sepsis, blood and CSF samples were tested for mNGS. Both the CSF and blood mNGS were positive for HIV-1-DNA.

Results: The detected DNA sequences covered only short fragments of the HIV genome. Concurrently, no HIV sequence was detected by blood metatranscriptome sequencing (RNA-seq). More sensitive HIV-1 RNA detection assay and repeated immunoassays in his history using an HIV antibody confirmed the result. No anti-HIV treatment was initiated. We also utilized mNGS to extract HIV detection sequences for subsequent analysis.

Conclusions: Several studies have reported the occurrence of false-positive HIV results following CAR-T treatment. These studies utilized NAAT for testing, but due to commercial confidentiality, limited information is available on the specific vector types used in the CAR-T product. In comparison to NAAT, mNGS offers a graphical representation of pathogen genome coverage, providing a more intuitive and easily interpretable approach for clinical assessments. Particularly, in cases where a significant portion of the genome is not covered, mNGS can help discern between non-specific sequence detection and false positives. If required, bioinformatics can be employed to further analyze and compare the identified nucleotide sequences using BLAST. The ability of mNGS to provide a comprehensive view of the genome is a unique advantage over NAAT, which can be explored in future clinical practice.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P114 IMPACT OF SERUM INTERLEUKIN 1-BETA LEVELS DURING CAR-T CELLS THERAPY ON CLINICAL OUTCOME: A SINGLE-CENTER EXPERIENCE

Ilaria Cutini 1, Riccardo Boncompagni1, Chiara Camerini1, Mirella Giordano1, Antonella Gozzini1, Alessio Mazzoni1, Gaia Mirabella1, Chiara Nozzoli1, Edoardo Simonetti1, Riccardo Saccardi1

1Careggi University Hospital, Florence, Italy

Background: CAR-T therapy has emerged as a breakthrough for treating relapsed/refractory large B cell lymphomas, delivering remarkable remission rates and enduring disease control. Despite its effectiveness, about one-third of patients experience disease recurrence. Understanding and optimizing CAR-T therapy effectiveness hinge on cytokines, pivotal for CAR-T cell activation and proliferation in vivo, enhancing the anti-tumor response. Whilst their roles in CRS and ICANS are acknowledged, limited data exist on their impact on hematological response, overall survival (OS), and disease recurrence (PFS).

Methods: We retrospectively evaluated 28 consecutive patients who underwent CAR-T therapy for Large B-cell Lymphoma (LBCL) and Mantle Cell Lymphoma (MCL) from March 2020 to September 2023. Patients were categorized into two groups based on whether they achieved complete remission (CR) at 3 months post-infusion. Clinical characteristics, including diagnosis, CAR-T type, median age, bulky disease, IPI risk score >2, bone marrow infiltration, relapse after autologous SCT, >2 previous lines of treatment, hematological response at the time of infusion, incidence of both CRS and ICANS, and usage of tocilizumab and steroids, were compared between the two groups. Non-continuous variables were compared using the Chi-square test. Serum levels of IL-6, IL-8, IL-1β, IL-10, and TNF-α were assessed at various time points (pre-lymphodepletion, day of CAR-T administration (D0), every 10 days from D10 to D60, and monthly from D60 to D90). The Spearman test determined correlations between interleukins and response at 3 months. Receiver Operating Characteristic (ROC) curve analysis established optimal cut-off values and their impact on PFS and OS.

Results:

Variables, n (%)

CR, 17 (60,7)

Not-CR, 11 (39,3)

p

10 (90,9)

Gender male, n (%)

13 (76,5)

ns

Median age, yo (range)

65 (35-74)

60 (35-71)

ns

ECOG PS

0, n (%)

14 (82,4)

ns

1, n (%)

3 (17,6)

11 (100)

ns

Diagnosis

LBCL, n (%)

13 (76,5)

10 (90,9)

ns

MCL, n (%)

4 (23,4)

1 (9,1)

IPI

0-2, n (%)

8 (47,1)

5 (45,5)

ns

>2, n (%)

9 (52,9)

6 (54,5)

Bulky before LK: Y, n (%)

9 (52,9)

5 (45,5)

ns

Bone Marrow infiltration: Y, n (%)

4 (23,5)

2 (18,2)

ns

Extranodal sites, n (%)

8 (47,1)

6 (54,5)

ns

Status before LK

Relapsed, n (%)

6 (35,3)

2 (18,2)

ns

Refractory, n (%)

2 (11,8)

2 (18,2)

Primary refractory, n (%)

9 (52,9)

7 (63,6)

Relapsed after ABMT, n (%)

4 (23,5)

3 (27,3)

ns

Bridging therapy

16 (94,1)

9 (81,8)

ns

RT, n (%)

6 (35,3)

2 (18,2)

CHT + RT, n (%)

7 (4,1)

7 (63,6)

immunotherapy, n (%)

3 (17,6)

Response before CAR-T therapy

ns

SD/PR/RC, n (%)

13 (76,5)

7 (63,6)

PD, n (%)

4 (23,5)

4 (36,4)

CRS, n(%)

16 (94,1)

6 (54,5)

0,013

CRS>=3, n(%)

1 (5,9)

1 (9,1)

ns

Tocilizumab Y, n(%)

9 (52,9)

5 (45,5)

ns

Dexa, n (%)

5 (29,5)

4 (36,4)

ns

ICANS, n (%)

7 (41,2)

2 (18,2)

ns

ICANS>=3, n(%)

1 (5,9)

1 (9,1)

ns

Dexa, n (%)

5 (29,4)

2 (18,2)

ns

Patient characteristics of the groups are detailed in Table 1. The CR group exhibited a higher incidence of CRS of any grade (94.1% vs. 54.5%, p=0.013). No significant differences were observed between patients who achieved CR and those who did not (not-CR) regarding diagnosis, CAR-T type, median age, bulky disease, IPI risk score >2, bone marrow infiltration, relapse after autologous SCT, number of previous lines, response state after the last therapy, bridging therapy, incidence of severe CRS and ICANS, as well as tocilizumab and steroids administration (p=ns).

Despite a correlation between serum IL-10 levels at the time of lymphodepleting therapy and D0 with the not-CR group, no impact was observed on OS and PFS, (p=ns). Serum levels IL-1β at various time points (D0, D10, D20, D30, D40, and D50) were significantly higher in[cn1] the CR group (rs -0.414 p=0.028, rs -0.408 p=0.033, rs -0.535 p=0.007, rs -0.575 p=0.003, rs -0.333 p=0.048, rs -0.439 p=0.011).

The ROC analysis set a cut-off value of 0.5 pg/mL for IL-1β at each evaluated time point (AUC ranging from 0.735 to 0.900). Kaplan–Meier estimates demonstrated that patients with IL-1β levels ≥ 0.5 pg/mL at D30 after CAR-T exhibited a longer median PFS (median not reached at 40 months vs. 3 months, p=0.002) and median OS (36 months vs. 12 months, p=0.033).

Conclusions: The small sample size and the retrospective nature might prevent other differences on the issue; however, our findings suggest IL-1b level at 1 month might have a role as prognostic factor for disease response and survival

Disclosure: none

4: CAR-based Cellular Therapy – Clinical

P115 THE SCOTTISH EXPERIENCE WITH CAR-T CELL THERAPY: A FOCUS ON DLBCL

David Hopkins 1, Anne Parker1, Dimitris Galopoulos1, Grant McQuaker1, Andrew Clark1, Ailsa Holroyd1, Anne-Louise Latif1, David Irvine1

1NHS Greater Glasgow and Clyde, Glasgow, United Kingdom

Background: Chimeric antigen receptor T cell therapy has transformed treatment for patients with relapsed/refractory aggressive B cell lymphoma, however the majority progress despite treatment and therapy is associated with significant short and long term toxicity. Licenced CAR T therapy has been available for R/R DLBCL in Scotland since 2019. Eligibility and advisability are considered by an inclusive national MDT.

We present the Scottish experience with licensed CAR-T cell therapy from inception of the service in 2019 to the present day. We focus on response to therapy, survival and both early and late toxicity. We also conducted retrospective analysis of potential predictors of poor outcome in this real-world dataset.

Methods: Data was extracted from the Scottish national CAR-T cell database. Clinical details and follow up data were accessed through SciStore. Survival curves were obtained by Kaplan-Meier method. Multivariate analysis was performed by ANOVA.

Results: 65 CAR-T cell therapies for R/R DLBCL have been delivered: (Axicabtagene (Axicel) (n=41), Tisagenlecleucil (Tisacel) (n=21) + trial products (3). 51 patients received bridging therapy. Median admission duration was 28 days (IQR 23-38). Day +30 CT/PET was performed in 57 of the 62 patients. ORR was 73.7%. Most (51) had Day +30 PET, 47.1% achieved CMR (Deauville score </=3). Day +100 PET was performed in 39 patients with ORR of 69.2% and CMR of 53.8%.

Median duration of follow up was 254 days. Day +100 OS was 89.2%. 1 year OS was 66.2%. Median OS and PFS for Axicel treated patients was 527 days 373 days. Median OS and PFS for Tisacel treated patients was 534 days and 505 days respectively.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (16)

We performed multivariate analysis on age, ECOG status, stage, bulk, extranodal disease, LDH and IPI. No clear prognostic features were identified. Cytokine release syndrome (CRS) was seen in 57 patients (91.9%). 6 patients experienced grade 3 + CRS (9.7%). 49 patients (79.0%) received Tocilizumab. 28 (45.2%) patients experienced immune effector cell neurotoxicity (ICANS). 9 patients experienced grade 3 + ICANS (14.5%). Patients requiring steroid, Anakinra and Siltuximab for ICANS were 33.9%, 14.5% and 3.2% respectively. 12 patients (19.4%) required ITU admission and 6 (9.7%) patients required HDU admission.

Infection risk is a significant issue following CAR T therapy. 24 patients (38.7%) had subsequent admissions for infection. 8 (12.9%) were admitted twice, 4 (6.5%) were admitted three times and 1 (1.6%) was admitted four times. This totalled 37 admission episodes. 17 episodes (45.9%) were due to bacterial infection. 8 episodes (21.6%) were due to COVID. There were no positive cultures in 12 episodes (32.4%). 6 patients (9.7%) died due to infection. Overall TRM was 11.3%. Non-relapsed TRM was 6.5%.

CD3 + /CD4 + T-Cell monitoring was conducted. 19 patients (30.6%) demonstrated recovery of CD3 + /CD4 + T-Cells to >200, in a median of 105 days (IQR 66-184).

Conclusions: Outcome measures and toxicity are in keeping with larger published real world datasets. No difference in PFS or OS was noted between patients receiving Axciel or Tisacel. No specific factors or combination of factors were prognostic in our treated cohort. Post treatment immunocompromise and infection is a significant concern.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P116 DONOR-DERIVED LYMPHOID ANTIGEN-DIRECTED CAR-T CELLS FOR RELAPSED/REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA IN CHILDREN AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: REPORT OF 17 CASES

Olga Molostova1, Larisa Shelikhova1, Rimma Khismatullina1, Evelina Lyudovskikh1, Yuliya Abugova1, Dmitriy Pershin 1, Maria Klimentova1, Irina Shipitsina1, Ekaterina Malakhova1, Maria Fadeeva1, Alena Kulakovskaya1, Lili Khachatryan1, Viktoria Vedmedskaya1, Galina Novichkova1, Alexei Maschan1, Michael Maschan1

1Dmitry Rogachev Federal Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation

Background: Patients with precursor B-cell ALL/lymphoma who have relapsed after allogeneic hematopoietic stem cell transplantation (allo-HSCT) have a dismal prognosis. We hypothesized that donor-derived lymphoid antigen-directed CAR-T cell therapy may induce remission in children with post-HSCT ALL/lymphoma relapse.

Methods: Seventeen children with r/r BCP-ALL/lymphoma were enrolled in a compassionate-use program (m/f ratio=12:5, median age 9,0 years). 15 patients had BCPALL, 2 had B cell lymphoblastic Lymphoma with BM involvement, all relapsing after multiple lines of treatment, including previous HSCT(n=17), blinatumomab(n=14), inotuzumab(n=3) and CAR-T cell infusion(n=6). Allo-CAR-T were derived from previous HSCT donors (haplo n=13, MRD n=4). The CAR-T cells were targeting CD19 and CD22 (n=11), CD19 (n=5) or CD22 (n=1). At the time of allogeneic CAR-T application, the disease burden was high MRD (n=11), overt leukemia (n=7), and extramedullary lesions(n=6). The first CAR-T cell doses were 100х103/kg (n=15), 160х103/kg (n=1), 350х103/kg (n=1), 5 patients received 2nd dose 100x103/kg (n=2), 500x103/kg (n=1) and 900x103/kg(n=2). Lymphodepletion included fludarabine, cyclophosphamide +/- cytarabine, 8 pts received prophylactic tocilizumab at day-1, 10 abatacept at day -1, +7, +14, +28

Results: Cytokine release syndrome (CRS) occurred in 13 (76%) patients and was grade ≤3; all pts received tocilizumab with good effect. Four patients had neurologic events (ICANS grade 1 n=3, ICANS grade 5 n=1), one patient with ICANS grade 5[MM1]. 5 patients had the signs of aGVHD (skin grade 1, n=4, liver=gut grade 4, n=1). The median time to CAR-T cell peak expansion was 14 days. ORR was 82%, eleven patients achieved complete MRD-negative remission, three had MRD negative remission in BM with CT detectable extramedullary lesion, 3 patients had disease progression, one of them with CD19 loss. Six patients received consolidative HSCT after CAR-T cells, all from alternative donors, two patients relapsed, one with CD19 loss. Twelve (71%) patients are alive with a median follow-up of167 days (40-532), 9(75%) of them in CR, one with stable disease and 2 with disease progression, 5 patients died (2 due to ICAN and sepsis, 3 in PD). Median time of follow-up for survivors was 0,5 years (range, 0,2 – 3,9).

Conclusions: Our early experience suggests that donor-derived lymphoid antigen-directed CAR-T cells expand in vivo and may provide CR with manageable safety profile. Prospective testing and further research of the approach is warranted.

Disclosure: M. Maschan - Miltenyi Biotec - Honoraria

4: CAR-based Cellular Therapy – Clinical

P117 CARDIOVASCULAR COMPLICATIONS OF CHIMERIC ANTIGEN RECEPTOR (CAR)-T CELL THERAPY

Giorgia Mancini 1, Alice Frangione1, Edlira Rrapaj1, Ilaria Scortechini1, Francesco Saraceni1, Roberta Domizi1, Simona Pantanetti1, Abele Donati1, Antonio Dello Russo1, Federico Guerra1, Attilio Olivieri1

1AOU delle Marche, Marche University Hospital, Ancona, Italy

Background: Chimeric antigen receptor (CAR) T cells today represents a breakthrough treatment for some relapsed/refractory (R/R) hematological malignancies and are currently indicated for the treatment of R/R acute Lymphoblastic Leukemia and R/R NHL. However, this therapy can be potentially affected by serious adverse reactions such as cytokine release syndrome (CRS) and severe neurological syndromes (ICANS); moreover a number of serious unexpected cardiac events has been reported; nevertheless the mechanisms of cardiotoxicity during CART cell therapy are unclear; the main hypotheses revolve around IL-6 mediated myocardial depression during CRS, takotsubo cardiomyopathy, and/or direct toxicity on cardiomyocytes by CAR-T cells. Hereby we present our preliminary prospective study conducted in cooperation with our dedicated cardio-oncology team.

Methods: 14 consecutive patients candidates for CAR-T therapy were prospectively enrolled since the inception of the CAR-T program at our institution. All patients were refractory to at least two lines of treatment including anthracycline. After lymphodepletive treatment, all patients received autologous CAR-T cells (axicabtagene ciloleucel, Yescarta). Prior to CAR-T cell therapy, all patients underwent baseline cardiac evaluation, including a 12-lead ECG, a complete echocardiography scan, and cardiac biomarkers. The same battery of tests was performed before discharge and after six months.

Variable

Value

Age (years)

63.5 ± 10.8

Female gender_ n (%)

6 (43%)

Diagnosis _ n (%):

DLBCL

12

Primary mediastinal large B cell lymphoma

1

ALL

1

Disease Status _ n (%):

CR

1 (7%)

PR

5 (36)

SD

2 (14)

PD

6 (43%)

BMI (Kg/m2)

26.3 ± 6.3

Previous lines of treatment_ n (%):

2

5 (36)

3

6 (43)

4

3 (21)

Hypertension_ n (%):

9 (63%)

Dyslipidaemia_ n (%):

7 (50%)

Diabetes Mellitus_ n (%):

2 (14%)

Previous CV history _ n (%)::

Coronary heart disease

1 (7%)

Carotid vascular disease

2 (14%)

Prior stroke/TIA

1 (7%)

Atrial fibrillation

1 (7%)

Median Follow up: months (range)

7, 4 months (1,9-18,4)

Outcome _ n (%):

Alive without disease

8 (57)

Died for relapse

5 (36)

Died for CART toxicity

1 (7)

Results: After CAR-T cell infusion, all patients experienced CRS of variable degree after a median of 1,2 days (1-6); among them four patients (28%) showed clinical manifestations suggesting a some degree of direct or indirect cardiac toxicity after a median follow-up of 12 days (7-40). One patient asintomatic with mild laboratory signs of cardiotoxicity (Grade I) was managed with only supportive therapy while two patients developing Grade II toxicity received cardioprotective drugs (ACE-Is and beta-blockers) for transient left ventricular systolic dysfunction (mean lowest LVEF 44%). One patient experienced a Takotsubo syndrome with concomitant CRS grade IV and complicated by cardiogenic shock on day 4, requiring high-dose vasopressors and mechanical ventilation. All patients showed a progressive recovery of ventricular function and survived to discharge; three died due to disease recurrence after a median of 7.4 months (range 4.2-8.2 months), with recovered cardiac function; one patient is alive in complete remission at the 12-month assessment, without cardiac function alterations.

Interestingly all the patients experiencing a CRS had similar demographical characteristics and medical history but standard echocardiogram before CAR-T showed a lower baseline LVEF (53.6±5.9% vs 63.2±7.7%; p=.03) and lower baseline right longitudinal systolic function markers (TAPSE: 25±4 vs. 20±3 mm; p=.02 S’: 09±.01 vs.12±.02 m/s; p=.03. Cardiac biomarkers (high-sensitivity Troponin I and NT-proBNP) did not show any significant predictive power at baseline.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (17)

Conclusions: CAR-T therapy is associated with an increased risk of serious cardiotoxicity, with up to 28% of all patients requiring unscheduled cardio-oncology evaluation and management both during admission and after discharge. Cardiac adverse events range in degree of severity, but their risk could be stratified by echocardiographic work-up. Larger number of patients is required to clarify if there is a relationship between the degree of CRS and the risk of cardiax toxicity and to identify potential trigger of cardiotoxicity.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P118 CORRELATION OF IL-6 LEVELS WITH THE EXPANSION OF LYMPHOCYTIC POPULATIONS, SEVERITY OF CYTOKINE RELEASE SYNDROME AND NEUROTOXICITY FOLLOWING CAR-T CELL THERAPY

Maria Liga 1, Dimitris Tsokanas1, Memnon Lysandrou1, Dionysia Kefala1, Evangelia Triantafyllou1, Angeliki Georgopoulou1, Eleftheria Sagiadinou1, Nikolaos Savvopoulos1, Vassiliki Zacharioudaki1, Alexandros Spyridonidis1

1University Hospital of Patras, Patras, Greece

Background: Genetically modified T-lymphocytes with chimeric antigen receptors (CAR-T cells) are an approved treatment for patients with relapsed/refractory B-cell malignancies. The main complications affecting patients outcomes are cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS). Interleukin-6 (IL-6) appears to play a key role in the pathophysiology of CRS. In patients with CRS, significantly elevated levels of IL-6 are observed and seems to play a role in the fundamental symptoms of the syndrome. The purpose of this study is to examine whether IL-6 levels are related to the severity of CRS and ICANS.

Methods: A measurement of IL-6 was conducted on days 0, 3, 6, and 10 following the administration of CAR-T and it was performed using the electrochemiluminescence immunoassay (ECLIA) method. Quantification of anti-CD19-CAR+ and activated HLA-DR + T lymphocytes was carried out using flow cytometry.

Results: A total of 10 patients received approved CAR-T cell therapy (7 axicabtagene ciloleucel, 1 tisagenlecleucel, 2 brexucabtagene autoleucel), with a median age of 61 years (range 20-68), for diffuse large B-cell lymphoma (n=6), primary mediastinal B-cell lymphoma (n=1), B-cell acute lymphoblastic leukemia (n=2), and mantle cell lymphoma (n=1). All patients (n=10) exhibited CRS (Grade 1 n=4, Grade 2 n=4, Grade 3 n=2, Grade 4 n=0), and 5 developed ICANS (Grade 1 n=3, Grade 2 n=0, Grade 3 n=2, Grade 4 n=0). The kinetics of IL-6 levels were assessed at 4 time points during the first 10 days after CAR-T cell infusion (d0,+3, + 6, + 10) for all patients as well as for patients who developed high and low-grade CRS separately. Increased IL-6 concentration was observed on d + 3 (median 49.87 pg/ml, range 5.69-2303) and d + 6 (median 109.3 pg/ml, range 1.5-1267). Similarly, the expansion of anti-CD19-CAR+ and activated HLA-DR + T lymphocytes (cells/μl) was evaluated at 8 time points after CAR-T infusion (d1,10,14,30±5,60±5,100±5,180±10 and 300±10), revealing the peak expansion of these populations on d + 10. IL-6 levels on d + 6 showed a statistically significant positive correlation with the severity of CRS (n=9, r=0.82, p=0.006) and the peak expansion(day +10) of anti-CD19-CAR + (median 70.5 cells/μl, range 18-1450, n=7, r=0.91, p=0.004) and activated HLA-DR + T lymphocytes(median 432 cells/μl, range 234-1684, n=7, r=0.91, p=0.004). In contrast, IL-6 levels showed no correlation with the severity of ICANS at any of the studied time points.

Conclusions: This study suggests the potential use of IL-6 levels as a biomarker for CRS but not for ICANS. The application of IL-6 measurement is an easy, fast and effective method that can contribute to the selection of patients for early interventions aimed at preventing complications of CAR-T therapy.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P119 EFFICACY OF ANTI-CD19 C AR-T CELL TREATMENT IN MANTLE-CELL LYMPHOMA HARBORING P53 ALTERATION: A MONOCENTRIC RETROSPECTIVE STUDY

Daniele Mannina1, Chiara De Philippis1, Jacopo Mariotti 1, Barbara Sarina1, Daniela Taurino1, Stefania Bramanti1, Armando Santoro1

1IRCCS Humanitas, Rozzano, Italy

Background: CAR-T brexucabtagene autoleucel (brexu-cel) changed the treatment algorithm of mantle-cell lymphoma (MCL) relapsed or refractory to chemo-immunotherapy and BTKi.1,2 The alterations involving p53, chromosomal deletions or gene mutations, emerged as a key prognostic factor for MCL.3 The predictive role on brexu-cel efficacy for MCL is still under investigation, due to the lack of data about p53 status both in pivotal trials and real-world reports. Here we describe brexu-cel outcome in a group of MCL patients harboring p53 alterations.

1 Wang Y, Jain P, Jain MD, ET AL. J Clin Oncol. 2023

2 Wang M, Munoz J, Reagan PM, et al. J Clin Oncol. 2023

3 Lew TE, Minson A, Seymour JF, et al. Lancet Haematol. 2023

Methods: Since 2021, 14 MCL patients received brexu-cel at Humanitas Cancer-Center. Test for p53 alteration (chromosome-17 deletion, gene mutation) is available for 14/16 patients. Eight of the 14 tested patients harbored p53 alterations. Table 1 summarizes patients’ characteristics.

Patients n= 8

Diagnosis: Mantle-cell lymphoma

Age

Median

58

(range)

(44 - 71)

Sex

Male

100% (8)

Female

P53 status

P53 mutation

87,5% (7)

Chromosome 17 deletion

12,5% (1)

Diagnosis-to-CAR time

Median

14 months

(range)

(4 - 297 months)

Previous lines

2

62,5% (5)

3

12,5% (1)

4

12,5% (1)

5

12,5% (1)

Previous exposure to

Anthracycline

100% (8)

Citarabine

25% (2)

Bendamustine

25% (2)

Rituximab

100% (8)

Ibrutinib

100% (8)

Venetoclax

25% (2)

High dose CT + ASCT

37,5% (3)

Allo-SCT

12,5% (1)

Performance status ECOG

87,5% (7)

1

12,5% (1)

Stage

IV

100% (8)

Bridging therapy

Ibrutinib

37,5% (3)

Ibrutinib + venetoclax

12,5% (1)

Steroids

12,5% (1)

None

37,5% (3)

Disease status at CAR-T

CR

PR

37,5% (3)

SD

12,5% (1)

PD

50% (4)

Results: Among 8 patients with p53 alteration, 5 (62,5%) received a bridging regimen: 3 (37,5%) with partial remission, 2 (25%) unresponsive. Three patients (37,5%) were in PD without bridging. After brexu-cel infusion, 1 (12,5%) experienced grade 3 CRS, 7 grade 1-2. Neurotoxicity (ICANS) was documented in 7 cases (87,5%); 5 patients (62,5%) needed ICU admission for grade 3-4 ICANS. All patients had complete resolution of CRS and ICANS. All patients achieved complete remission (CR) at day 30. One case of sepsis in prolonged cytopenia resulted in non-relapse mortality (12,5%). After a median follow-up of 6,1 months (range 3-32), no disease relapse was recorded.

Conclusions: Despite the small patient number and the retrospective nature, this report show a good outcome of brexu-cel treatment for MCL patients harboring p53 alteration who failed both chemo-immunotherapy and BTKi. The results suggest that brexu-cel efficacy may overcome the negative impact of p53. An earlier shift from standard chemotherapy to CAR immunotherapy may be wishable in the setting of p53-mutated MCL.

Disclosure: No potential CoI

4: CAR-based Cellular Therapy – Clinical

P120 OUTCOMES OF COMMERCIAL AND IN-HOUSE ANTI CD19 CAR T-CELL THERAPY FOR RELAPSED/REFRACTORY B-CELL LYMPHOMA IN SINGAPORE

Shin Yeu Ong1, Cindy Krisnadi 1, Samuel Sherng Young Wang1, Yun Xin Chen1, Michaela Su-fern Seng2, Shui Yen Soh2, Esther Hian Li Chan3, William Ying Khee Hwang1, Aloysius Yew Leng Ho1, Francesca Lorraine Wei Inng Lim1

1Singapore General Hospital, Singapore, Singapore, 2KK Women’s and Children’s Hospital, Singapore, Singapore, 3National University Cancer Institute, Singapore, Singapore

Background: While CD19 chimeric antigen receptor (CAR) T-cells have changed the outcomes of patients with relapsed/refractory (r/r) aggressive B-cell lymphoma (aBCL), financial toxicity and manufacturing time represent barriers to widespread implementation. In 2020, we initiated a clinical trial and expanded compassionate access using an in-house produced CD19 CAR-T cell incorporating a 4-1BB co-stimulatory domain. The aim of this study is to retrospectively evaluate outcomes among patients with r/r aBCL who received commercial and in-house CAR T-cell therapy in two tertiary hospitals in Singapore.

Methods: Adult patients with r/r aBCL after 2 prior lines of treatment were eligible for CAR T-cell therapy. Autologous T cells were transduced with a lentiviral vector encoding an anti-CD19 binding motif and 4-1BB/CD3z intracellular signaling domain. GMP-compliant manufacture was done using CliniMACS Prodigy, in a 12-day culture. Lymphodepletion was with cyclophosphamide and fludarabine. Cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS) were graded according to the American Society for Transplantation and Cellular Therapy (ASTCT) consensus criteria. All other adverse events were graded by CTCAE v.5.0 criteria. Efficacy was measured by overall response rate (ORR) and complete response (CR) rate. ORR was defined as rate of patients who achieved CR or partial response (PR), as assessed using the Lugano 2014 criteria. Overall survival (OS) was defined as the time from CAR T cell infusion to death due to any cause. PFS was defined as the time from CAR T cell infusion to disease progression/relapse or death from any cause.

Results: Between March 2020 and November 2023, 20 patients (tisagenlecleucel [n=17], axicabtagene ciloleucel [n=3]) and 10 patients received commercial and in-house CAR T-cell products respectively. Baseline characteristics were similar between the two cohorts. The best ORR/CR rates were 80%/65% among commercial cohort and 70%/30% among in-house cohort. Twelve-month PFS were 60.0% (95% CI: 25.3%–82.7%) and 55.9% (95% CI: 30.7%–75.1%) in-house and commercial cohorts, while 12-month OS were 58.3%(95% CI: 23.0%–82.0%) and 76.7%(95% CI: 48.8%–90.7%), respectively. Relative to commercial CAR T-treated patients, in house CAR T-treated patients did not show an increase in hazards associated with experiencing progression/relapse/death (aHR: 0.55 [95%CI: 0.14-2.2], p=0.402) or death (aHR: 1.10 [95% CI: 0.20–6.02]; p = .92), after adjusting for baseline number of prior therapies and presence of refractory disease. Any grade/grade ≥3 CRS were observed in 100%/10% and 90%/10% among commercial and in-house cohorts, while any grade ICANS/grade ≥3 ICANS were observed in 25%/10% and 10%/10% respectively. While early (<30 days) and late (30-90 days) grade ≥3 neutropenia rates were comparable in the commercial (83%/47%) and inhouse (100%/66.7%) cohorts respectively, grade ≥3 infectious complications were more common among patients receiving commercial CAR T-cell products (47% vs 10%, p=.044).

Conclusions: We observed comparable efficacy and favorable toxicity among patients receiving in-house versus commercial CAR T-cell therapy. The real-world efficacy of Kymriah in our patients were equivalent to trial and other real-world reports. Local manufacture enables access of high-risk patients with aBCL to CAR T-products despite financial constraints and rapidly progressing disease.

Clinical Trial Registry: N.A.

Disclosure: The authors report no conflicts of interest

4: CAR-based Cellular Therapy – Clinical

P121 CHOICE OF COMMERCIALLY AVAILABLE CAR T-CELLS PRODUCT FOR AGGRESSIVE LYMPHOMA: A SURVEY OF BEHALF OF THE CELLULAR THERAPY & IMMUNOBIOLOGY WORKING PARTY (CTIWP) OF THE EBMT

Urban Novak1, Jarl E. Mooyaart2, Michael Daskalakis1, Christof Scheid3, Anne Sirvent4, Ibrahim Yakoub-Agha5, Ron Ram6, Edouard Forcade7, Lucía López Corral8, Emma Nicholson9, Eugenio Galli10, Friedrich Stölzel11, Wolfgang Bethge12, Eva Maria Wagner-Drouet13, Jorinde D. Hoogenboom14, Stephan Mielke15, Caroline Arber Barth16, Federico Simonetta17, Christian Chabannon18, Jürgen Kuball19, Annalisa Ruggeri20, Florent Malard 21

1University Hospital Bern, Bern, Switzerland, 2EBMT Statistical Unit, Leiden, Netherlands, 3University of Cologne, Cologne, Germany, 4CHU Lapeyronie, Montpellier, France, 5CHU de Lille, Lille, France, 6Tel Aviv Sourasky Medical Center, Tel Aviv, Israel, 7CHU Bordeaux, Hopital Haut-Leveque, Pessac, France, 8Hospital Clínico, Salamanca, Spain, 9Royal Marsden Hospital, London, United Kingdom, 10Universita Cattolica S. Cuore, Rome, Italy, 11University Medical Center Schleswig-Holstein, Campus Kiel, Dresden,, Germany, 12Universitaet Tuebingen, Tuebingen, Germany, 13University Medical Center Mainz, Mainz, Germany, 14EBMT Leiden Study Unit, Leiden, Netherlands, 15Karolinska University Hospital, Stockholm,, Sweden, 16CHUV Centre Hospitalier Universitaire Vaudois, Lausanne, Switzerland, 17Département d’Oncologie, Service d’Hématologie, Geneva,, Switzerland, 18Institut Paoli-Calmettes Comprehensive Cancer Centre and Module Biothérapies du Centre d’Investigations Cliniques de Marseille, INSERM-Aix-Marseille Université-AP-HM-IPC, CBT-1409, Marseille, France, 19University Medical Center Utrecht, Utrecht, Netherlands, 20San Raffaele Scientific Institute, Hematology and Bone Marrow Transplantation Unit, Milan, Italy, 21Sorbonne Université, Paris, France

Background: Since the summer of 2018, several CAR-T cells products have been approved by the EMA to treat aggressive B-cell lymphoma, tisagenlecleucel (tisa-cel), axicabtagene ciloleucel (axi-cel) in and lisocabtagene maraleucel (liso-cel). While the exact labellings of these products are different, there is some important overlaps in product attributes and indications; there is so far no recommendation regarding the choice of one CAR-T cells product over another.

Methods: The CTIWP conducted a survey across reporting EBMT centers to build an overview of the CAR-T cell practices in aggressive lymphoma and criteria used by centers to guide their choice of a product rather than others. CAR T cell products offered for aggressive lymphoma, process at site, clinical application, and considerations guiding the choice of CAR T cells were obtained. Descriptive statistics were utilized to analyze answers by centers participating, as well as distribution of responses according to the questionnaire.

Results: Responses were received from 67 centers in 20 countries, of which, the majority offered access to several CAR-T cells products for aggressive lymphoma (n=56, 84%), 9 offered only one CAR-T cell product (13%) and 2 did not offer CAR-T cells (3%). The main reason to offer several CAR-T cell products was flexibility for production and lymphapheresis (n=48, 86%), followed by product profile and approved indications in terms of line of treatment and lymphoma entities (n=41, 73%). All reporting centers assessed patients’ eligibility for CAR-T cell based on the product approval and the majority of centers, n=58 (89%) selected themself the CAR-T product for an individual patient. Overall, tisa-cel, was used in 55 centers (89%), axi-cel in 57 centers (93%), while within the reporting period until 10/2023 only 14 centers (23%) used liso-cel.

National restrictions of reimbursement and on the label of tisa-cel and axi-cel were comparable, 57% versus 65% and 71% versus 74% respectively. Regarding CAR-T labelling, 44% of center agreed that axi-cel had a broader label versus 20% for tisa-cel. 44% and 39% of centers agreed that lymphapheresis process (frozen versus fresh) and lymphaphereis work load is less resource-consuming for axi-cel, (36% vs 12%). Manufacturing slot availability and ”vein-to-vein delays” have been deemed as limiting factors for 18% and 47% of center for tisa-cel and 37% and 32% for axi-cel.

The majority of centers (77%) agreed that axi-cel is more effective compared to other CAR-T cells. In contrast, 85% of the centers agreed that tisa-cel was less toxic. Tisa-cel was therefore the preferred choice for treating elderly patient from 43% of centers, versus 9% for axi-cel. In contrast axi-cel was the preferred product for bulky disease (49% vs 2%) and young high-risk patients (74% vs 6%).

Conclusions: The majority of centers have access to more than one CAR-T cells, mostly tisa-cel and axi-cel. While there were no main differences between the 2 products regarding access and cost, the majority of centers consider axi-cel to be more effective, while tisa-cel is considered to have a better safety profile, making axicel the preferred product in young patients with aggressive diseases, and tisa-cel in elderly patients.

Disclosure: Friedrich Stölzel : Travel Support: Janssen, medac, Honoraria: medac, GWT, Abbvie, Servier, Pierre Fabre, Research funding: medac, Servier. Federico Simonetta: institutional consulting fees from BMS/Celgene, Incyte, Kite/Gilead; speaker fees from Kite/Gilead, Incyte; travel support from Kite/Gilead, Novartis, AstraZeneca, Neovii, Janssen; research funding from Kite/Gilead, Novartis, BMS/Celgen. Jurgen Kuball, research suppot Novartis, miltenyi Biotech, Gadeta. Florent Malard: honoraria Therakos/Mallinckrodt, BMS, Sanofi, Jazz Pharmaceuticals, Gilead, Novartis, Aztrazeneca and MSD. Others authors have nothing to disclose.

4: CAR-based Cellular Therapy – Clinical

P122 ASSESSMENT OF MALNUTRITION DURING PREHABILITATION FOR PATIENTS UNDERGOING CAR-BASED CELLULAR THERAPY

Juliet Morgan 1, Helen Long2

1Cardiff & Vale University Health Board, Cardiff, United Kingdom, 2Cardiff & Vale University Health Board, Caerdydd, United Kingdom

Background: There is limited evidence regarding the incidence of malnutrition during a CAR-based cellular therapy transplant. However, patients will have increased nutritional requirements and experience side effects of their underlying condition and treatments which affects nutritional intake and status.

Malnourished patients have poorer outcomes than well-nourished patients. One marker of malnutrition is 10% weight loss, which is regarded as a negative clinical prognostic factor.

Nutrition & Dietetics at the University Hospital Wales set up a service for CAR-T patients in May 2020. Initial outcomes demonstrated reductions in unplanned percentage weight loss of over 10% with dietetic prehabilitation.

Malnutrition has multiple diagnostic parameters. ESPEN 2015 explains malnutrition can be identified in patients with a BMI <18.5kg/m2. They also advise that a Fat Free Mass Index (FFMI) <15kg/m2 in women and <17kg/m2 in men is another criteria for the diagnosis of malnutrition

EWGSOP2 2019 defines sarcopenia with handgrip strength (HGS) of less than 27kgs for males and 16kgs females.

Phase Angle (PhA) can be measured with Bioelectrical Impedence Analysis (BIA) and has been suggested as a useful prognostic indicator of ill health such as cancer and other potentially serious diseases. The higher the Phase Angle the healthier the cells. PhA <4.5 is considered a prognostic factor associated with malnutrition and poorer outcomes (A de Souza et all 2019)

Within our clinical area we would also define patients at risk of malnutrition and needing dietetic intervention if they are meeting less than 75% of energy and protein requirements.

Methods: During April 2022-November 2023, the Dietitian assessed all CART patients in the prehabilitation setting for malnutrition/sarcopenia using WAASP (All Wales nutrition screening tool), percentage weight loss, BMI, percentage of energy/protein requirements, BIA (FFMI and PhA) and HGS. A patient reported experience measures (PREM)’s questionnaire of the dietetic prehabilitation intervention was also designed and analysed.

Results: A total of 24 patients were included in CART prehabilitation nutritional assessment.

50% (n=12) scored high on the WAASP

29% (n=7) had >10% unplanned weight loss

No patients had a BMI less than 18.5kg/m2

25% (n=6) patients had energy intakes less than 75% of their nutritional requirements

58% (n=14) patients had protein intakes less than 75% of their nutritional requirements

FFMI assessement demonstrated 2/14 (14%) were malnourished

PhA assessment shows 5/14 patients (36%) had a PhA less than 4.5.

HGS assessment demonstrated 4/16 (25%) patients were malnourished

100% (n=16) of patients who replied to the PREMS questionnaire reported they strongly agreed/agreed that the dietetic input was clear, concise, easy to understand and empowered them to make dietary changes, benefited from the dietetic service and patient information.

Conclusions: The results demonstrate that patients that are due to receive CART are likely to have malnutrition/be at high risk of malnutrition in the prehabilitation setting. Multiple assessments and diagnostic criteria’s of malnutrition and sarcopenia are appropriate, feasible and useful for this patient group.

Further work in now in progress to use these measurements criteria throughout the whole CART pathway to demonstrate issues with nutrition and the benefit the dietitian can have with providing nutrition support.

Disclosure: Kite Gilead funded the Bioelectrical Impedence Analysis Bodystat Multiscan 5000 machine.

4: CAR-based Cellular Therapy – Clinical

P123 LEUKAPHERESIS FOR AUTOLOGOUS CHIMERIC ANTIGEN RECEPTOR T CELL THERAPY IN CHILDREN: REPORT FROM A SINGLE PEDIATRIC CENTER

June Iriondo 1,2, Josune Zubicaray1,2, Guzmán López de Hontanar3,1,2, Elena Sebastián1,2, Josefa Navarrete1, Ana Torre1, Rocio Rico1, Ana Castillo1, Blanca Herrero1,2, Alba Rubio1,2, Susana Buendía1,2, Alejandro Sanz1,2, Ana de la Cruz1,2, Almudena Galán1,2, Jesus Gonzalez de Pablo2, Manuel Ramirez1,2, Julián Sevilla1,2

1Hospital Infantil Universitario Niño Jesús, Madrid, Spain, 2Fundación para la Investigación Biomédica del Hospital Niño Jesús, Madrid, Spain, 3Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain

Background: CAR-T cell therapy has been progressively incorporated in the therapeutic landscape of many conditions in the last years. In the pediatric setting, it is mostly used for patients with acute lymphoblastic leukemia (ALL), though trials are ongoing in other diseases as well. Children represent a potential challenge for the first step of the process, which is the autologus T-cell collection, given their variability on total blood volume (TBV) and venous accesses, among others. Herein, we report the outcomes of the leukapheresis procedures for CAR-T therapy performed in our institution.

Methods: We retrospectively analyzed all autologous leukapheresis for CAR-T therapy performed in pediatric patients (up to 18 years of age) in our institution between June 2020 and November 2023.

Apheresis was performed on a Spectra Optia device (Terumo BCT), using the continuous mononuclear cell collection program. Central venous catheters were placed in all children <12 years and in those with poor peripheral venous accesses. The anticoagulant used was ACD-A. The circuit was previously primed with red packed cells in patients weighing <20Kg. Adverse events were recorded by the nurse in charge during the process.

Results: A total of 26 patients and 29 procedures were included in the analysis. Median age was 9 years (interquartilic range -IQR- 5-13.5 years), median weight was 28Kg (IQR 20.30-40.75 Kg) and the median TBV was 1,921 mL (IQR 1,912-2,915 mL). The baseline diagnosis was relapsed or refractory B-ALL in all cases. The median blood volume processed was 5,971 mL (IQR 5,172-7,070 mL), and median number of processed TBVs was 3.1 (IQR 2.4-3.5). Furthermore, the median duration of the procedures was 220 minutes (IQR 188-279 minutes).

The median CD3+ cell count pre-apheresis was 0.91 x109/L (IQR 0.55–1.39), and there were no patients with <0.2 x109/L CD3+ cells pre-collection. Median T-cell collection efficiency was 50.35% (IQR 42.65-66.58%), and the median CD3+ cell yield per apheresis procedure was 2.8x109/L (IQR 2.04-4.02 x109/L). 3 patients (11.5%) required 2 apheresis procedures: one due to a manufacturing failure, and two patients due to insufficient collection after the first leukapheresis.

Adverse events were observed during thirteen procedures (44.8%), were mild in all cases and did not require to interrupt the apheresis procedure. The most frequent adverse event was tachycardia in 6 patients (20.7%), and nausea/vomiting in 3 cases (10.3%).

Conclusions: In accordance with other reports, we found that leukapheresis for autologous CAR-T therapy is well tolerated in most children, considering that these patients suffer from an active relapsed disease at the time of the procedure. Furthermore, sufficient CD3+ cells are obtained in a single apheresis in most cases, though more procedures might be required in small patients or in those with lower pre-collection CD3+ counts.

Disclosure: J. S. reports financial support outside the submitted work for educational lectures by Novartis, Miltenyi, Amgen and Agios. Advisory board member for Rocket Pharma, Novartis, Sobi, Amgen and Agops. The other authors declare no competing interests.

4: CAR-based Cellular Therapy – Clinical

P124 SEVERE HHV-6 RELATED ENCEPHALITIS FOLLOWING ANTI-CD19 CAR-T TREATMENT: A CASE STORY FROM A POINT OF CARE PRODUCTION FACILITY

Nina Marie Birk1, Katrine Kielsen 1,2, Özcan Met3, Inge Marie Svane3, Eva Haastrup4, Lisbeth Pernille Andersen4, Kristian Schønning5, Jonas Nielsen6, Søren Lykke Petersen7, Marianne Ifversen1

1Copenhagen University Hospital Rigshospitalet, København Ø, Denmark, 2Institute for Inflammation Research, Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark, 3National Center for Cancer Immune Therapy, Copenhagen University Hospital, Herlev, Denmark, 4Cell Therapy Facility, The Blood Bank, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark, 5Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark, 6Intensive Care Unit, Rigshospitalet, Copenhagen University Hospital, København Ø, Denmark, 7Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark

Background: Human herpes virus 6 (HHV-6) encephalitis has been reported following CAR-T therapy; however, the source of HHV-6 reactivation is not well established, and the clinical diagnosis is challenging due to overlapping manifestations with ICANS.

We present a case of HHV-6 encephalitis in a 58-years old patient treated with academical anti-CD19 CAR-T cells. We hypothesized, that our patient was auto-infected with latent HHV-6 virus from the infused CAR-T cells, which due to activation and proliferation during CAR-T cell production yielded a high viral load and caused encephalitis.

Methods: The patient had a history of depressions but was otherwise neuropsychological healthy. CAR-T treatment was given due to refractory transformed follicular lymphoma as part of a clinical trial, investigating an academical autologous 4-1BB anti-CD19 CAR-T product (ref: N. Kekre et al). The pre-apheresis therapy was administered according to front line protocols. Lymphodepleting therapy consisted of cyclophosphamide and fludarabin.

HHV-6 DNA was measured by real-time PCR in cryopreserved cells from the CD4/CD8 negative and CD4/CD8 positive fraction of the apheresis product and from the transduced and expanded CAR-T cells of the final product. Detection limit of the of the R-GENE® kit was 1000 copies/106 cells.

Results: The patient developed CRS on day +4 post-infusion (max grade II), which resolved completely on tocilizumab. On day +15, the patient developed complete loss of memory, fever relapse, and generalized seizures, which treatment for ICANS did not relieve. On day +17, HHV-6 DNA was detected in blood, and Ganciclovir treatment was initiated leading to decreasing viral load. MRI on day +22 was consistent with encephalitis including affection of the hippocampus bilaterally. Lumbar puncture was not feasible due to due to low platelet counts, thus confirmatory HHV-6 detection in CSV was not possible. Slow recovery was observed from day +25, however severe neurological sequelae (such as amnesia) persist after 1 year. Table 1.

Notably, circulating numbers of lymphocytes increased rapidly post infusion from 0.10 x109/L at day -1 to 5.6 x109/L on day +11.

Table 1 Day according to CD-19 CAR-T cell infusion

Day according to CD-19 CAR-T cell infusion

Overview of treatment and outcome.

Day -13

Apheresis

Day -12 to 0

Local production of an academical 4-IBB anti-CD 19 CAR-T cell product using a lentiviral vector on the CliniMACS Prodegy from the fresh apheresis product. All release criteria for the final product were met.

Day -7 to -4

Lymphodepletion consisted of Cyclophosphamide 2 doses (500 mg/m2) and Fludarabine 4 doses (30 mg/m2). Pre-lymphodepletion, the patient had multiple PET-positive lymph nodes on both sides of the diaphragm.

Day 0

Infusion of 2.6 x 106/kg CAR-T cells

Day + 4 to + 9

The patient presented with chills, desaturation, low blood pressure (systolic BP 100-110), tachycardia (100 bpm), and fever >40 °C. The patient was diagnosed with CRS grade II and was treated with Tocilizumab (4 doses administered from day +6 to +9). CRS symptoms were completely resolved on day +11.

Day + 15

Sudden onset of loss of memory, disorientation, fever relapse (day 16), and generalized seizures. The patient was transferred to the ICU with a GCS of 3. Symptoms were categorized as ICANS grade 2-4 nad tretment with steroids, levetiracetam, and Tocilizumab was commenced. Eeg showed an encephalopathic pattern. CT examination of the brain with angiography was normal. No lumbar puncture was performed due to the patients coagulation status.

Day + 17

HHV6 A/B DNA positive in blood with 4.500.000 copies/ml. Treatment with Ganciclovir was initiated and Valaciclovir paused. The number of HHV-6 copies was reduced to 9300 copies/ml on day +23 and 850 copies/ml on day +29.

Day + 22

MRI showed symmetrical changes in the hippocampus, ulcus, and corpus amygdala without necrotic tissue, suggesting non HSV-1 encephalitis.

Day + 29

The patient remained cerebrally severely affected, however progress was seen: the patient was more often awake, recognized hospital staff and tried to speak. She was able to move her upper extremities and nod her head.

Day + 365

The patient is in complete remission on PET-CT for her transformed follicular lymphoma. There is continuing C-cell aplasia. However, she has severe cognitive impairment due to loss of short-term memory, presumably as a consequence of HHV-6 related encephalitis.

Conclusions: In conclusion, we present a case of clinically severe HHV-6 related encephalitis following infusion of autologous anti-CD19 CAR-T treatment. Notably, in this case the initial number of cells in the apheresis product were very high, indicating a high proliferation capability of the harvested cells. Further, the patient had an unusual expansion of lymphocytes post infusion, however HHV-6 was not detectable in neither the pre-manufactured lymphocytes, nor in the pre-infusion CAR-T product. Whether HHV-6 indeed was present in very few cells below detection limit or the combined treatment increases susceptibility to HHV-6 infection remains to be shown.

Clinical Trial Registry: Nor relevant- case story

Disclosure: Nothing to declare.

4: CAR-based Cellular Therapy – Clinical

P125 UNLOCKING POSSIBILITIES: THE IMPACT OF CYTOGENETIC ANALYSIS ON PATIENTS UNDERGOING CAR-T CELL THERAPY

Christos Varelas 1, Ioannis Kyriakou1, Eleni Gavriilaki2, Zoi Bousiou1, Ioannis Batsis1, Nikolaos Spyridis1, Evangelia Yannaki1, Giorgos Papaioannou1, Maria Gkaitatzi1, Michail Iskas1, Ioanna Sakellari1, Anastasia Athanasiadou1

1General Hospital “George Papanikolaou”, Thessaloniki, Greece, 22nd Propaedeutic Internal Medicine Clinic, Aristotle University of Thessaloniki, Hippokration Hospital, Thessaloniki, Greece

Background: CAR-T cell therapy is a novel form of immunotherapy that can induce durable remissions in patients who have received multiple lines of therapy with relapsed/refractory B-acute lymphoblastic leukemia (B-ALL), B-cell lymphomas (B-NHL) and multiple myeloma (MM). Cytogenetic analysis provides valuable data on the genetic and chromosomal abnormalities of neoplasms. The aim of this study is to investigate the value of cytogenetic analysis in patients undergoing CAR-T therapy so as to both search for pathological clones in the context of pre-existing disease or a possible secondary hematological malignancy and to detect the presence of chromosomal instability.

Methods: Our study included 50 patients (20 women, 30 men) with a median age of 53 years (range: 21 – 75). Six patients had relapsed/refractory B-ALL, 5 had MM and 39 had B-NHL (DLBCL, mediastinal primary and mantle cell lymphoma). The median number of prior treatments was 4 (range 2–9). G-banding cytogenetic analysis was performed on bone marrow cells after 24 & 48 hours as well as 72 hours culture with PHA + IL2.

Results: Results from cytogenetic analysis were available in 38 patients prior to CAR-T therapy administration. Two patients had a complex karyotype with chromosomal abnormalities associated with their disease (one with MCL, and one with DLBCL), experienced disease progression, and died at 2.5 months. Eighteen patients with a normal karyotype had single cell abnormalities (SCAs) with a median number of 2 SCAs (range 1-6). At a median follow-up time of 10.5 months (range 1-42.5), the probability of overall survival (OS) at 42.5 months was 43.6%. Nine patients of this group had disease relapse or progression and four died.

Of the remaining 18 patients who had a normal karyotype and no SCAs, 8 relapsed or had refractory disease and 4 of them died. One patient in this group died due to TRM in the ICU. The median follow-up time was 8 months (range 1-41) and the probability of overall survival at 41 months was 62.4%.

From the analysis of the two subgroups, no statistically significant difference emerged in terms of the probability of overall survival. One of the 12 patients in whom cytogenetic analysis was not performed before CAR-T therapy developed myelodysplastic syndrome (R-IPSS: intermediate risk) with an abnormal clone 6 months after cellular therapy. Thirty-one months later the patient is alive having undergone allogeneic transplant.

Conclusions: CAR-T therapy offers increased survival in patients who have received multiple prior lines of therapy and suffer from relapsed/refractory B lymphomas, B-ALL and MM. Cytogenetic analysis before and after the administration of CAR-T therapy appears to provide valuable information and is useful to be performed. The detection of low-frequency clonal hematopoiesis is essential and possibly beneficiary for the follow up of these patients. A larger number of patients with a longer follow-up time is needed to determine the exact contribution of cytogenetic analysis in these patients, as well as in the evaluation of the toxicity of previous treatment.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P126 T-CELL LARGE GRANULAR LYMPHOCYTE POPULATION INVOLVING CHIMERIC ANTIGEN RECEPTOR-MODIFIED T (CAR T) CELLS IN PATIENTS WITH CYTOPENIA AFTER CD19-TARGETED CAR T-CELL THERAPY: CASE SERIES

Aya Albittar 1, Alireza Torabi2, Emily Liang1,2, Andrew Portuguese1,2, Jennifer Huang1,2, Cecilia Yeung1,2, Erik Kimble1,2, Delaney Kirchmeier1, Aiko Torkelson1, Abigail Chutnik1, Ryan Cassaday1,2, Aude Chapuis1,2, Hans-Peter Kiem1,2, Filippo Milano1,2, Folashade Otegbeye1,2, Mazyar Shadman1,2, Brian Till1,2, David Maloney1,2, Cameron Turtle1,2,3, Jordan Gauthier1,2, Alexandre Hirayama1,2

1Fred Hutchinson Cancer Center, Seattle, United States, 2University of Washington, Seattle, United States, 3The University of Sydney, Camperdown, Australia

Background: CD19-targeted chimeric antigen receptor T-cell (CAR-T) therapy has transformed the treatment of relapsed/refractory (R/R) large B-cell lymphoma (LBCL). Prolonged or recurrent cytopenias (PC) are a well-recognized and poorly understood complication which can occur weeks to months (M) after CAR-T cell infusion. We report 3 cases of T-cell large granular lymphocyte (T-LGL) clonal expansion involving the CAR-T population in patients (pts) with PC.

Methods: Day (D) 0 refers to D of CAR-T infusion. Cytokine release syndrome (CRS) and Neurotoxicity (NT) were graded using ASTCT criteria. Cytopenias were graded using CTCAE v5.0.

Results: Case 1. A 52-year-old (y/o) female (F) with R/R primary mediastinal B-cell lymphoma with central nervous system (CNS) involvement was treated with out-of-specification tisagenlecleucel. Course was complicated by grade (G) 1 CRS and progressive disease. She had persistent G1-2 anemia and thrombocytopenia after D28 and developed prolonged norovirus gastritis 30.8 M after infusion, followed by recurrence of G3 neutropenia. Flow cytometry (FC) of bone marrow (BM) and peripheral blood (PB) identified an expanded T-LGL population representing 26.7 and 28.6% of white blood cells (WBCs), respectively. Evaluation of T-cell receptor (TCR) Vβ isoforms suggested oligoclonality. STAT3 mutation was not detected (ND). Repeat BM 43.6 and 52.4 m after CAR-T infusion showed persistent T-LGL population comprising 9.1 and 7.1% of WBCs by FC, respectively.

Case 2. A 62 y/o F with R/R diffuse LBCL (DLBCL), NOS and chronic hepatitis B was treated with axicabtagene ciloleucel. Course was complicated by G1 CRS, achieving complete response (CR). She had persistent G1-3 anemia and thrombocytopenia and G2-4 neutropenia after D28. PB FC on 18.6 m after infusion showed an expanded T-LGL population representing 16% of WBCs. A BM 23.7 m after infusion confirmed a T-LGL population comprising 30% of T cells. Clonal TCR (TRG) gene rearrangement was detected by PCR. STAT3 mutation was ND. A PB FC demonstrated that 43.9% of the cells with the T-LGL phenotype expressed the CAR transgene.

Case 3. A 50 y/o F, with R/R DLBCL, NOS with CNS involvement and history of cold agglutinin disease was treated with lisocabtagene maraleucel. Course was complicated by G2 CRS and G2 NT, achieving CR. She developed G4 persistent neutropenia after D28. BM FC on D27 identified a T-LGL population representing ~42% of the WBCs. TCR Vβ isoforms suggested a dominant isoform. TRG gene rearrangement by PCR detected an oligoclonal T-cell population. STAT3 and STAT5B mutations were ND. PB FC demonstrated 65.0% of the T-LGL phenotype cells expressed the CAR transgene.

Conclusions: We report 3 cases of T-LGL clonal expansion diagnosed after CAR-T therapy in patients with PC. In two of the patients, a subset of the clonal population expressed the CAR transgene. This case series highlights T-LGL as a potential cause of PC after CAR-T therapy.

Disclosure: Hirayama:Novartis; Bristol Myers Squibb:Research Funding;Nektar Therapeutics:Research Funding;Juno Therapeutics, a Bristol Myers Squibb Company: Research Funding. Kimble:Juno/BMS: Research Funding. Shadman:Mustang Bio, BMS, Pharmacyclics, Genentech, Inc., AbbVie, TG Therapeutics, BeiGene, AstraZeneca, Genmab, MorphoSys/Incyte, Vincerx: Research Funding; AbbVie, Genentech, Inc., AstraZeneca, Pharmacyclics, BeiGene, BMS, MorphoSys/Incyte, Kite, Eli Lilly, Genmab, Mustang Bio, Regeneron, ADC therapeutics, Fate Therapeutics, Janssen, MEI Pharma: Consultancy; regeneron: Consultancy, Research Funding; pharmacyclics: Consultancy, Research Funding; beigene:Consultancy, Research Funding; Lilly: Consultancy; abbvie: Consultancy; genentech: Consultancy, Research Funding. Till:Mustang Bio: Consultancy, Patents & Royalties, Research Funding; Proteios Technology: Consultancy, Current holder of stock options in a privately-held company; BMS/Juno Therapeutics: Research Funding. Milano:ExCellThera Inc: Research Funding. Cassaday:Amgen: Consultancy, Research Funding; Kite/Gilead: Consultancy, Research Funding; Incyte: Research Funding; Merck: Research Funding; Pfizer: Consultancy, Research Funding;Servier: Research Funding; Vanda Pharmaceuticals: Research Funding; Jazz: Consultancy; Autolus: Membership on an entity’s Board of Directors or advisory committees;PeproMene Bio: Membership on an entity’s Board of Directors or advisory committees; Seagen: Other: Spouse was employed by and owned stock in Seagen within the last 24 months. Maloney:Kite, a Gilead Sciences: Consultancy, Research Funding; Bioline Rx: Consultancy; Bristol Myers Squibb: Consultancy, CLL Strategic Council, Member of the JCAR017-BCM-03 Scientific Steering Committee under BMS, Research Funding; Amgen: Consultancy; Celgene: Consultancy, Research Funding; Juno Therapeutics: Consultancy Patents & Royalties, Research Funding; Umoja: Consultancy; A2 Biotherapeutics: Consultancy, Current holder of stock options in a privately-held company; ImmPACT Bio: CD19/CD20 bi-specific CAR-T Cell Therapy Program; Pharmacyclics: Consultancy; Legend Biotech: Consultancy, Research Funding; MorphoSys: Consultancy; Mustang Bio:Consultancy; Navan Technologies: Consultancy; Janssen: Consultancy; Novartis: Consultancy; Gilead Sciences: Consultancy; Genentech: Consultancy;Incyte:Consultancy;Navan Technologies: Current holder of stock options in a privately-held company; Lyell Immunopharma. Gauthier:Century Therapeutics; Juno Therapeutics (a Bristol Myers Squibb company): Research Funding; Angiocrine Bioscience: Research Funding; MorphoSys: Consultancy, Research Funding; Kite Pharma: Consultancy; Janssen: Consultancy; Legend Biotech: Consultancy; Celgene (a Bristol Myers Squibb company): Research Funding; Sobi: Consultancy. Research Funding.

4: CAR-based Cellular Therapy – Clinical

P127 TREATMENT WITH TISAGENLECLEUCEL OF RELAPSE REFRACTORY DIFFUSE LARGE B CELL LYMPHOMA – SINGLE CENTER EXPERIENCE UHC ZAGREB

Barbara Dreta 1, Sandra Bašić-Kinda1, Dino Dujmović1, Neno Živković1, Josip Batinić1,2, Ida Hude Dragičević1, Klara Dubravčić1, Margareta Dobrenić1, Ivana Ilić1, Snježana Dotlić2,1, Lea Galunić-Bilić1, Ivana Franić-Šimić1, Igor Aurer1,2

1UHC Zagreb, Zagreb, Croatia, 2School of Medicine, University of Zagreb, Zagreb, Croatia

Background: Treatment with chimeric antigen receptor T-cell (CAR-T) therapy revolutionized the approach to patients with relapsed/refractory diffuse large B cell lymphoma (r/r DLBCL).

Since 2020. treatment with tisagenlecleucel is available in our center. We performed a retrospective data analysis of treatment effectiveness and safety in our patients. We also did analysis of factors that could influence response to the treatment and development of complications.

Methods: We collected data from medical documentation in our center on patients treated with tisagenlecleucel with r/r DLBCL until December 2023.

Results: Since April 2020. we treated 32 patients with DLBCL, 75% male, median age 62.4 years (31,6-77,6). 65.6% had primary refractory disease or relapsed within 12 months after first line of treatment. At the time of last relapse IPI was 0 in 3,1%, 1 in 18,7%, 2 in 18,7%, 3 in 40,6%, 4 in 15,6% and 5 in 3.1% of patients. After bridging therapy 12,5% of patients were in CR, 59,4% in PR, 21,9% in SD, and 3,1% in PD while for 1 patient it was not available. With median follow up of 10,8 months response to CAR T cell treatment was CR 50%, PR 21,87%, PD 25%, not done in 3,12%. Median OS was 17,57 months and median PFS 12,23 months. AT 12 months OS was 59,2% and PFS 53,3%.

CRS occurred in 21 patients (65,6%), grade 1 28,1%, grade 2 34,4% and grade 5 3,1% with median on day 2. 15 received tocilizumab with addition of dexamethasone in 7. ICANS had 6 (18,7%), grade 1 6,2%, grade 2 3,1%, grade 4 3,1% and grade 5 6,2%, median on day 4. They were treated with dexamethasone and 2 with anakinra. Neutropenia grade 3 had 37,5%, grade 4 53,1% and thrombocytopenia grade 1 40,6%, grade 2 3,1%, grade 3 18,75% and grade 4 18,75%. Hypogammaglobulinemia was present in 22 patients (68,7%).

Prolonged neutropenia had 9 patients, gr2 1, gr3 4 and gr4 in 4, while prolonged thrombocytopenia had 11, gr3 2 and gr4 4.

Infections after 30 days had 12 patients, 7 had COVID and 1 had HSV viremia. One patient developed HLH. Secondary primary malignancy had 2, one cholangiocarcinoma and one adenocarcinoma. 14 patients died, 6 from progression of disease, 3 from CRS/ICANS, 1 with development of HLH, 1 patient cholangiocarcinoma, 2 COVID infection and 1 Aspergillus caused pneumonia.

Survival analysis regarding value of percentage of eosinophil and lymphocytes number showed no statistically difference. LDH level prior to lymphodepletion also but with visible tendency while CRP levels had significant difference. Patients who were primary refractory had no different outcome. Analysis of number of lymphocytes, percentage of eosinophils, LDH, CRP values and IgG levels prior to lymphodepletion compared to rate of CRS had no statistically significant difference.

Conclusions: In real world setting treatment with tisagenlecleucel had similar efficacy and safety as in clinical trials. Analysis of initial laboratory values could be used in real world setting to predict response to the treatment and development of complications, but further research is needed.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P128 SERUM AMYLOID-A IN CAR-T CELL THERAPY: A NEW ACUTE PHASE REACTANT

Eugenio Galli 1, Ilaria Pansini2, Patrizia Chiusolo1,2, Stefan Hohaus1,2, Anna Modoni1, Luca Montini1, Nicola Piccirillo1,2, Federica Sorà1,2, Simona Sica1,2

1Policlinico Universitario Agostino Gemelli, Rome, Italy, 2Univeristà Cattolica del Sacro Cuore, Rome, Italy

Background: Serum Amyloid-A (SAA) proteins compose a family of apolipoproteins produced mainly by the liver, but also by macrophages, kidneys, lungs, adipocytes, and synovial cells. SAA is the most prominent acute phase reactant, with a typical rapid increase of more than 3 log during early acute stimulations (24 hours) and a rapid decline, showing an impressive feedback regulation. Much interest has been generated by this marker, especially in the study of chronic inflammatory states and in autoimmune diseases. The role of SAA in the context of CAR-T therapy remains unexplored in existing literature.

Our study aimed to elucidate the role of SAA in CAR-T patients, specifically investigating its sensitivity to predict acute toxicities such as cytokine-release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome (ICANS), and its relationship with more commonly used acute-phase proteins (APP).

Methods: We studied 15 consecutive patients affected by Diffuse Large B-Cell Lymphoma (DLBCL) (53.3%), transformed Follicular Lymphoma (tFL) (26.7%), and Mantle cell Lymphoma (20%), treated at our institution with axi-cel (60%), tisa-cel (20%), or brexu-cel (20%) between May and November 2023. The median age at CAR-T infusion was 59 years; 73.3% of patients were male, and 26.7% female. Five patients had received a previous hematopoietic stem cell transplant (four autologous and one allogeneic).

Samples from peripheral blood (PB) were collected at baseline before lymphodepletion therapy (day -5), at infusion (day 0), and after CAR-T (day+2, +4, +7, +9, +11, and +14). SAA, ferritin, interleukin 6 (IL-6), C-reactive protein (CRP), and procalcitonin were measured at each timepoint.

Results: SAA levels peaked very early after CAR-T infusion, mostly between day+2 and day+7. Overall, we have found a very strong correlation between SAA, CRP (p<0.001) and procalcitonin (p<0.001), but not with ferritin (p=0.129) or IL-6 (p=0.260). This may be possibly biased by iron overload and the use of tocilizumab, respectively. Notably, the association with procalcitonin is kept both in patients with and without documented infections.

SAA levels at the time of start of lymphodepletion and at infusion of CAR-T did not predict CRS. Nevertheless, SAA showed a remarkable association with CRS, with higher grades of CRS corresponding to higher levels of SAA. Namely, the median SAA was 8.6, 273, and 347 mg/L when ongoing CRS was graded 0, 1, or 2, respectively (p<0.001).

We did not observe an association between SAA and ICANS.

We then confirmed that in our cohort all CRP (p<0.001), IL-6 (p<0.001), and procalcitonin (p=0.027), but not ferritin, were associated with CRS.

We moved subsequently to assess if SAA had a non-inferior association with CRS when compared to the others APP. We therefore applied a ROC analysis and obtained that the best association with CRS among the APP was dealing at SAA (with the highest Youden index of 57t.4[SH1]) with a sensitivity and specificity of 76% and 81%, respectively.

Conclusions: In conclusion, SAA emerges as a valuable acute-phase protein in the setting of CAR-T cells, not only associating with CRP and procalcitonin but also outperforming other APPs in characterizing CRS.

Disclosure: Nothing to declare

4: CAR-based Cellular Therapy – Clinical

P129 CHALLENGES IN CHIMERIC ANTIGEN RECEPTOR-T CELL PRODUCT ADMINISTRATION IN A HIGH TUMOR-BURDEN ELDERLY PATIENT WITH MANTLE-CELL LYMPHOMA

Umberto Pizzano1,2, Francesca Patriarca1,2, Marta Lisa Battista 1, Giuseppe Petruzzellis1,2, Gabriele Facchin1, Giuseppe Di Renzo1,2, Renato Fanin1,2

1Hematology and Stem Cell Transplantation Unit, University Hospital ASUFC, Udine, Italy, 2Department of Medical Area (DAME), Udine, Italy

Background: Brexucabtagene-autoleucel (Brexu-cel) is a CAR-T cell product approved for adults with mantle-cell lymphoma after two or more previous treatments, including a BTK-inhibitor, without an age limit.

Methods: Here we describe a 77-year-old woman case of mantle-cell lymphoma diagnosed in 2013, treated with chemotherapy plus autologous stem-cell transplantation, relapsed in May 2022, and, due to non-response to Ibrutinib, candidated to receive Brexu-cel. The patient suffered from previous transient ischemic attacks. Between lymphocytoapheresis and Brexu-cel administration she was hospitalized due to femur fracture and SARS-CoV2 infection so the product administration was postponed. At the time of lymphodepletion start the lymphoma was in progression (nodal localizations at CT-PET and increasing of lymphocytosis). Other risk factors for CAR-T cells associated toxicities were: high levels of ferritin (2577 ng/ml), PCR (86.15 mg/L) and neurofilament light-chain (Nfl) (97.3 pg/ml) titers, at basal.

Results: On day 0 after CAR-T infusion the patient developed a grade II CRS, subsequently evolved in grade III in spite of Tocilizumab administration, so dexamethasone 10 mg every 6 hours was started. Despite steroid, patient needed intensive-therapy support with noradrenaline and non-invasive-ventilation with progressive clinical improvement and tapering of steroid. On day 24 she developed an ideo-motor slowdown, time disorientation, confusion, with no bleeding signs on TC imaging and an ICE score of 3/10; this clinical state was linked with a delayed circulating CAR-T peak (3000/mmc), so we considered it as a grade II ICANS rather than a hospitalization-associated delirium. Restart of dexamethasone was necessary, associated with IL-1 receptor antagonist Anakinra (100 mg every 12 hours). After an initial clinical improvement we reduced steroid and Anakinra doses, but the patient started to experience a swinging cognitive state; we performed an MRI examination showing a possible recent cerebellar ischemic alteration, not clearly correlated with the clinical state; so we continued to treat it as an ICANS. Furthermore, a restaging PET-TC examination showed a good partial response characterized by diffuse reduction in number, dimension and metabolic uptake of nodal localizations. Finally, on day 41, she developed a coma state (GCS 4/15). Due to patient age, comorbidities and a persistent transfusions-requiring pancytopenic state, in accordance to relatives, we decided for no further interventions.

Conclusions: This case shows that a good metabolic response could be achieved even in a high-tumor burden heavily pre-treated elderly patient, after administration of a CAR-T cell product, but, on the other hand, there is a very high risk of life threatening complications. In this older patient, it could be difficult differentiate neurological age-related symptoms from ICANS manifestations. Imaging, associated with monitoring of peripheral CAR-T cells, cytokine levels and other biomarkers, such as Nfl, could be helpful. A better knowledge of toxicities pathophysiology, and new treatment possibilities, could contribute to offer a better outcome to these poor-prognosis frail patients.

Disclosure: No disclosures to declare

4: CAR-based Cellular Therapy – Clinical

P130 REGIONAL REPRESENTATION OF THE OUTCOMES OF A COHORT OF PATIENTS WITH B-CELL ALL TREATED WITH TISAGENLECLEUCEL FROM A CENTRE IN THE MIDDLE EAST

Fahad Bahkali MD 1, Ali Alahmari MD1, Riad El Fakih1, Amr Hanbali1, Rashed Al Bakr1, Abdulwahab Albabtain1, Naeem Chaudhri1, Walid Rasheed1, Ayman Saad1, Saud Alhayli1, Ahmed Abdrabou1, Samar Alfaqaawi1, Yazeed Bajuaifer1, Ahmed Bin Salman1, Alfadil Haroon1, Amal Hejab1, Taimoor Hussain1, Heba Madien1, Mostafa Saleh1, Momen Nassani1, Marwa Nassar1, Sara Samarkandi1, Mohamed Sharif1, Reem Alasbali1, Emad Ghabashi1, Majed Altareb1, Shaykhah Alotaibi1, Shihab Ahmed1, Reena Ulahannan1, Alfadel Alshaibani1, Hanan Alkhaldi1, Abdullah Alamer1, Mansour Alfayez1, Feras Alfraih1, Ahmad Alotaibi1, Fahad Alsharif1, Marwan Shaheen1, Fahad Almohareb1, Hazza Alzahrani1, Mahmoud Aljurf1, Syed Ahmed1

1King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia

Background: CAR T-cell therapy with Tisagenlecleucel has shown excellent outcomes in the AYA population with relapse or refractory B-cell ALL. However, there is limited regional representation, especially in Southeast Asia, the Middle East, and Africa, and outcomes in these populations are not known. Our center has been treating patients with Kymriah since march 2021, and we sought to present survival and response outcomes in these patients.

Methods: We retrospectively analyzed the CAR T-cell database for patients aged 14 to 25 who underwent CAR T-cell therapy and looked at baseline characteristics, response, and survival, as well as the toxicity profile.

Results: Between March 2021 and August 2023, we treated nineteen patients; the median age was 19 years. 57% of patients had had a prior transplant. The median lines of chemotherapy were 3. All of the products were successfully produced within specification. 36.8% of patients had a prior history of CNS involvement. All patients received lymphodepletion treatment with fludarabine and cyclophosphamide as per the recommended guidelines. The median dose of Kymriah was 2.6*10^6/kg. At the time of CAR T-cell infusion, 52.6% of patients had active disease, and 47.4% were in remission. One patient (5.2%) developed grade III-IV CRS. Two patients (10.5%) developed grade III-IV ICANS. Seventeen patients were alive and evaluable at 28 days post-CAR T-cell infusion. Response rate as per the day 28 assessment showed that 94% of patients were in remission. Persistence was assessed by B-cell aplasia and immunoglobulin levels. Seven patients (35.2%) lost their B-cell aplasia at a median time of 5 months. Seven patients (35.2%) went to transplant after CAR T-cell therapy. Median relapse free survival was 19.7 months. Median overall survival was not reached. At a median follow up of 18.5 months, the one-year relapse free survival was 60.2% and one-year overall survival was 77.4%. Overall survival of patients who were in remission prior to lymphodepletion was superior to those who had refractory disease (100% vs 60%, p=0.048).

ALL (N=19)

Age at infusion, y

Median age (range)

19 (14-25)

Sex, n (%)

Male

14 (73.3)

Female

5 (26.3)

Disease status at time of cellular infusion, n (%)

Primary refractory/relapsed

10 (52.6)

CR

9 (47.4)

No. of prior therapies

Median (range)

3 (1-6)

Prior HCT, n (%)

Allogeneic

11 (57.8)

Autologous

0 (0)

Time of follow-up since infusion, mo

Median (range)

18.5 (2-31)

Other specific characteristics, n (%)

Prior Central nervus system involvement

7 (36.8)

Prior blinatumomab

13 (86.4)

Prior inotuzumab

7 (36.8)

Conclusions: We present the first report of Tisagenlecleucel in a cohort of patients from Saudi Arabia, demonstrating excellent outcomes with reasonable safety profile.

Disclosure: nothing to declare

4: CAR-based Cellular Therapy – Clinical

P131 DELAYED ICANS IN POST BCMA CAR-T CELL THERAPY CHALLENGES

Mohamed Abuhaleeqa 1, Yara Afifi1, Sheima Mahmoud Ali1, Fatema Al Kaabi1, Inas El-Najjar1, Jayakumar David Dennison1, Mansi Sachdev1, Nameer Al Saadawi1, Ruqqia Mir1, Lev Brylev1, Mohamed Mostafa1, Yendry Ventura1, Krina Patel2

1ADSCC, Abu Dhabi, United Arab Emirates, 2MDAnderson Cancer Center, Houston, United States

Background: Multiple Myeloma is a hematological malignancy that has been difficult to cure despite many avilable lines of therapies. New modalities like Chimeric antigen Receptor modified T (CAR-T) cell therapy has given chance for Relapsed Refractory Multiple myeloma to get stringent responses and possibilities of long term disease control. This modality is associated with specific toxicities like Cytokines Release Syndrome (CRS) and Immune effector Cell associated Neurological Syndrome ICANS) and long term side effects such as delayed cytopenia and neurological sequel. In this abstract we discuss a case of Multiple Myeloma who developed delayed ICANS.

Methods: 37-year male presented with lytic lesions in the spine and ribs in September 2017 and work up confirmed diagnosis of multiple myeloma low risk as per International Staging System ISS-1. He received Bortezomib lenalidomide Dexamethasone four cycles, followed by Autologous Stem cell transplantation in April 2018 and maintenance lenalidomide. In December 2019, he relapsed with an extra-axial soft tissue disease treated with additional lines of treatment including radiotherapy, daratumumab, pomalidomide, and carfilzomib in various combination with steroids. He ultimately had CAR-T cell therapy in April 2023.

Results: After CAR-T infusion he suffered adverse effects CRS grade 3 and ICANS grade 3, requiring ICU hospitalization and treatment with Tocilizumab and steroids he had long ICU hospitalization but discharged home on two antiepileptic agents (Phenobarbital and Levetiracetam).

Day 90 assessment displayed stringent complete remission (sCR) with no evidence of biochemical markers and a negative PET (positron emission tomography).

He remained cytopenic post CAR-T cell therapy and required transfusion and growth factor support, IVIG and prophylaxis antimicrobial.

He was able to return home to continue follow close at home in SEP 2023 and was weaned off all his seizure medication by that time and off steroids as well.

6 months post his CAR-T cell therapy he was admitted to ICU (intensive care unit) with episode of seizure and post ictal confusion required intubation due to difficult protecting airway.

Every investigation into viral markers was negative. Myeloma screening was negative. MRI of the brain indicated Pan-meningitis figure (1). No abnormal cells in the CSF, high protein level and normal glucose levels. Generalized periodic epileptiform activity were seen on the EEG. He was treated for delayed ICANS grade 4 with steroids and antiseizure medications (Phenytoin and Levetiracetam).

He clinically improved and discharged with a prescription for steroids, phenytoin, and Levetiracetam and followed up in clinic. Unfortunately, he suffered another seizure at home unwitnessed and admitted to ICU, he was found to have septic shock with streptococcal pneumonia and passed away from septic shock.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (18)

Conclusions: In conclusion CAR T cell therapy can put patients with Relapsed Refractory Myeloma into deep remission but is associated with both acute and late complications. Late complications like Movement and Neurocognitive Toxicities (MNT) and Delayed ICANS are part of this but also the cytopenia and infection is another. High index of suspicion and close follow up is important for better support and adequate management for such patients.

Clinical Trial Registry: NA

Disclosure: no disclosures

3: CAR-based Cellular Therapy – Preclinical

P132 CAR T CELLS TARGETING CD28 SHOW PRECLINICAL ACTIVITY AGAINST T-ALl

Semjon Willier1, Julian Fäber1, Theodora Ispyrlidou2, Sophia Nikolaides2, Jonas Wilhelm2, Paulina Ferrada-Ernst2, Franziska Blaeschke2, Fiona Becker-Dettling2, Maike Breidenbach2, Theresa Käuferle2, Annika Peters2, Dana Stenger2, Christoph Klein2, Tobias Feuchtinger 1

1Faculty of Medicine, University of Freiburg, Freiburg, Germany, 2Hauner Children’s Hospital, LMU Hospital, Munich, Germany

Background: T-cell acute lymphoblastic leukemia (T-ALL) accounts for 15% of pediatric leukemia. The most promising T-ALL CAR T-cell therapy against CD7 confers cure for only some patients in clinical trials and CD7- relapse is frequent. Activating mutations of CD28 were described in T-cell malignancies but surface expression of CD28 in pediatric T-ALL has not been studied, yet.

Methods: Patient characteristics

T-ALL patients

healthy BM donors

Patients, n

54

14

Age at initial Dx, mean (range)

9 (3-17)

10.3y (1.7-19.1)

Gender, n (%)

Male

47 (87)

7 (50)

Female

7 (13)

7 (50)

Therapy status, n (%)

Therapy naive

45 (76)

-

Treated

7 (13)

-

Unknown status

6 (11)

-

Future relapse, n (%)

11 (20)

-

Death, n (%)

12 (22)

-

Time to relapse, mean (range)

1,8 (0,3-8,5)

-

Monitoring period, mean (range)

5,4 (0,1-12,1)

-

We measured expression of CD28 and 24 other surface proteins involved in T-cell co-signaling on T-ALL blasts from diagnostic patient samples (n=54) and healthy bone marrow donors (n=14). We observed CD28 overexpression on T-ALL blasts after gating on CD45dim CD7+ events within bone marrow (BM) samples of T-ALL patients compared to healthy counterparts.

We cultured CCRF-CEM T-ALL cells with monocyte-derived CD80+/CD86+ dendritic cells (DC) and observed significantly increased T-ALL proliferation, indicating a putative functional role of CD28 via DC in T-ALL.

As T-cells express CD28 at high frequency, we performed CD28-KO by CRISPR/Cas9 in primary T-cells. CD28-KO rates > 90% were reached and stable over 14 days. CD28-KO T-cell proliferation was unchanged and cytotoxicity was impaired only against CD80+ leukemic cells.

Results: Next, we designed 20 CAR molecules against CD28 based on five publicly available scFvs and identified two lead molecules by in vitro testing termed 28CAR_2 (scFv: TGN1412) and 28CAR_14 (scFv: CD28.3). Both molecules conferred comparable cytotoxicity against CCRF-CEM in vitro as CD7 CAR T-cells and against two CD28+ multiple myeloma cell lines. CD28KO prior to 28CAR transduction improved proliferation and decreased LAG3 expression.

Finally, we tested both 28CAR molecules in vivo by injecting 2.5e6 CAR T-cells into SCID mice bearing CCRF-CEM leukemia. Here, 28CAR_2 T-cells conferred superior survival compared to 28CAR_14 T-cells and comparable to CD7 CAR T-cells.

Conclusions: Taken together, our data highlight CD28 CAR T-cells as a novel synthetic immunotherapy against T-ALL and beyond.

Disclosure: SW was funded by Else-Kröner-Fresenius-Stiftung, Bayerisches Krebsforschungszentrum and Bettina-Bräu-Stiftung.

3: CAR-based Cellular Therapy – Preclinical

P133 HAMBURG REAL-WORLD EXPERIENCE WITH AXICABTAGENE CILOLEUCEL IN AGGRESSIVE B-CELL LYMPHOMA: IN-VIVO EXPANSION CORRELATES WITH TREATMENT OUTCOME

Susanna Carolina Berger1, Anita Badbaran1, Evgeny Klyuchnikov1, Kristin Rathje1, Nico Gagelmann1, Maria Geffken1, Dominic Wichmann1, Christian Frenzel1, Dietlinde Janson1, Christine Wolschke1, Nicolaus Kröger1, Boris Fehse1, Francis Ayuk 1

1University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany

Background: Recent evidence suggests a correlation between axicabtagene ciloleucel (axi-cel) in-vivo expansion and patient outcome. In our prior work in 21 real-world patients, we identified a peak chimeric antigen receptor-T cell (CAR-T) count of 16.1/µL to allocate patients into a strong- or weak expander group, which significantly correlated with better progression-free-survival (PFS) in these patients (Ayuk FA et al, Blood Advances 2021:5(11):2523-2527). If associated with patient outcome, CAR-T levels at fixed time points may provide guidance for subsequent early intervention strategies. Here, we present data from an expanded cohort with CAR-T counts analyzed at fixed time points.

Methods: Using our dedicated digital-PCR (dPCR) assay, we prospectively quantified CAR-positive T cells in 51 consecutive axi-cel patients treated for aggressive B-cell lymphoma at our clinic. DPCR data was correlated with clinical outcome as assessed per institutional practice. 47 patients were evaluable for a day 100 response. Statistical analysis was performed using SPSS Software (IBM, Germany).

Results: Median day of peak CAR-T counts was day 10. To enable earlier decision making, we used day 10 as well as day 7 for cut-off. Median CAR-T count on day 7 and 10 was 22.1/µL and 28.1/µL respectively. Using a cut-off of 16.1/µL on day 7 (n= 46 evaluable patients), 25/28 (89%) strong expanders were in complete remission (CR) or partial remission (PR) on day 100, compared to 7/18 (39%) weak expanders, p= 0.001. Using the cut-off of 16.1/µL on day 10, 25/30 (83%) strong expanders compared to 8/17 (47%) weak expanders hat CR or PR on day 100. Using 22.1/µL as cut-off on day 7, 20/23 (87%) strong expanders and 12/23 (52%) weak expanders were in CR or PR on day 100, p= 0.023. Using 28.1/µL as cut-off on day 10, 21/24 (88%) strong expanders compared to 12/23 (52%) weak expanders were in CR/PR on day 100. Receiver Operator Characteristic (ROC) analysis revealed a cut-off of 10.5/µL on day 10 as strongest for prediction of response and PFS. With this cut-off, 30/35 (86%) strong expanders were in CR/PR on day 100 compared to 3/12 (25%) weak expanders. PFS was also significantly better for strong compared to weak expanders (p < 0.001). The results of an in-depth statistical analysis including tumor burden and other relevant prognostic factors as well as association between expansion and side effects will be presented at the meeting.

Conclusions: Our data in an expanded cohort confirmed the previously described peak cut-off. Our data suggests that a fixed day 10 cut-off of 10.5/µL may be most appropriate to segregate weak and strong expanders with respective outcomes. If validated in multicenter cohort, these findings may help guide early interventions to improve patient outcome.

Clinical Trial Registry: The study was approved by the local ethics committee (PV7091) and written informed consent obtained from all patients.

Disclosure: Disclosures for Nicolaus Kröger: Advisory Board Gilead/Kite

Disclosures for Francis Ayuk: Honoraria: Novartis, BMS/Celgene, Gilead/Kite, Miltenyi Biotec, Janssen, Medac, Takeda, Mallinckrodt/Therakos.

Research funding: Mallinckrodt/Therakos

The other authors have nothing to declare related to this project.

3: CAR-based Cellular Therapy – Preclinical

P134 CD45-DIRECTED CAR-T CELLS WITH CD45 KNOCKOUT EFFICIENTLY KILL MYELOID LEUKEMIA AND LYMPHOMA CELLS IN-VITRO EVEN AFTER EXTENDED CULTURE

Maraike Harfmann 1, Tanja Schröder1, Dawid Glow1, Maximilian Jung1, Almut Uhde1, Nicolaus Kröger1, Stefan Horn1, Kristoffer Riecken1, Boris Fehse1, Francis Ayuk1

1University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: CAR-T cell therapy has shown impressive results and is now part of standard of care in treatment of B-lineage malignancies, whereas treatment of myeloid diseases has been limited by the lack of suitable targets. CD45 is expressed on almost all types of blood cells including myeloid leukemia cells, but not on non-hematopoietic tissue, making it a potential target for CAR-directed therapy.

Because of its high expression on T and NK cells, fratricide is expected to hinder CD45CAR-mediated therapy. Due to its important role in effector cell activation, signal transduction and cytotoxicity, CD45 knockout aimed at preventing fratricide in T and NK cells has been expected to lead to considerable functional impairment.

Methods: CD45 knockout was established on T and NK cell lines using CRISPR/Cas9-RNPs and electroporation, and the successful protocol was transferred to primary T cells. A combined protocol was developed enabling CD45 knockout and retroviral transduction with a third-generation CAR targeting CD45 or CD19. Functionality of CD45ko effector cells, CD45ko/CD45CAR-T and CD45ko/CD19CAR-T cells was studied using proliferation as well as short and long-term cytotoxicity assays.

Results: As expected, introduction of a CD45-CAR into T cells results in potent fratricide that can be avoided by CD45 knockout. Unexpectedly, the latter has had no negative impact on T- and NK-cell proliferation in vitro. Moreover, CD45ko/CD45CAR-T cells showed potent cytotoxicity against CD45-expressing AML and lymphoma cell lines in short-term and long-term co-culture assays. Pronounced cytotoxicity of CD45ko/CD45CAR-T cells was maintained even after four weeks of culture. In a further setup, we confirmed conserved functionality of CD45ko cells using a CD19-CAR. Again, proliferation and cytotoxicity of CD45ko/CD19CAR-T cells showed no difference to those of their CD45-positive counterparts in vitro.

In a final experiment, CD19CAR T cells were sorted into CD45pos and CD45ko cells and their capacity to perform repeated killings analysed. Both populations were re-stimulated with either TransAct or lymphoma cells using an excess of lymphoma cells in the second stimulation step (E:T ratio of 1:5). Re-stimulation provided a strong proliferation signal to both CD45pos and CD45ko CD19CAR-T cells, which grew faster than TransAct re-stimulated cells. Despite the low starting E:T ratio, lymphoma cells were essentially completely eliminated within 72 hours of co-culture. Moreover, CD45ko CD19CAR-T did not show an increase in expression of exhaustion markers.

Conclusions: We report efficient production of highly and durably active CD45ko/CAR-T cells. CD45 knockout did not impair functionality of CAR-T cells in vitro, independently of the target antigen. A recent publication by Wellhausen et al. reported the inability of second-generation CD45ko (CD19-) CAR-T cells to maintain long-term antitumor efficacy in NOD-SCID-IL2rγ-/- (NSG) mice. If the activity of our third-generation CD45ko CAR-T cells can be confirmed in vivo, CD45ko/CD45CAR-T cells might, for example, be useful as part of conditioning regimens prior to stem cell transplantation.

Disclosure: MH, TS, DG, BF and FA are co-authors of patent applications related to production and use of CD45CAR effector cells. All other authors declare no conflict of interest associated with this work.

3: CAR-based Cellular Therapy – Preclinical

P135 COMPARISON OF CD123- AND CD33-CAR-NK CELL PREPARATIONS IN A XENOGRAFT MOUSE MODEL FOR TREATMENT OF ACUTE MYELOID LEUKEMIA

Fenja Gierschek 1,2, Sabine Müller3, Julia Kostyra3, Jan Kuska3, Tobias Bexte1,2,4,5, Katja Stein1,2, Lisa Marie Reindl1,2, Sophia Thul1,2, Alina Moter1,2, Philipp Wendel1,2,6,7, Hadeer Mohamed Rasheed8,9, Ningyu Li8, Juliane Schlüter1, Sabine Hünecke1, Claudia Cappel1, Olaf Penack8,10, Thomas Oellerich2,4,6, Jan-Henning Klusmann1,2, Nina Möker3, Congcong Zang3, Evelyn Ullrich1,2,4,6

1Goethe University, Frankfurt am Main, Germany, 2Frankfurt Cancer Institute, Frankfurt am Main, Germany, 3Miltenyi Biotec B.V. & Co. KG, Bergisch Gladbach, Germany, 4University Cancer Center (UCT), Frankfurt am Main, Germany, 5German Red Cross Blood Service BaWüHe, Frankfurt am Main, Germany, 6German Cancer Consortium (DKTK) partner site Frankfurt/Mainz, Frankfurt am Main, Germany, 7Institute for Organic Chemistry and Biochemistry, Darmstadt, Germany, 8Charité, University Berlin and Humboldt-University Berlin, Berlin, Germany, 9Clinical Pathology Department, Alexandria, Egypt, 10German Cancer Consortium (DKTK) partner site Berlin, Berlin, Germany

Background: Although CAR-T cells have been designed as a cellular therapeutical alternative for treatment of acute myeloid leukemia (AML), clinical application is limited due to severe side effects and the need for autologous application or matched donors. In this context, CAR-engineered Natural Killer (NK) cells can be administered independent of HLA mismatch, creating the opportunity for donor independent “off-the-shelf” products with overall reduced side-effects. Recently, we reported on the anti-leukemic efficacy of CD33-CAR-NK cells in a preclinical setting. However, other established AML targets, such as CD123 are also showing promising results.

Methods: In this study, we compared a CD123-CAR-NK cell product generated by an automated, current Good Manufacturing Practice–compliant process using CliniMACS Prodigy® with CD123-CAR- and CD33-CAR-NK cells prepared by the conventional manufacturing protocol (noted as small scale in this study). Following a detailed cell product characterization, including functional cytotoxicity assays against CD123-expressing AML cell lines and primary cells, in vivo evaluation has been performed using the OCI-AML2 (GFP/luc+) xenograft NSG mouse model. Furthermore, the anti-leukemic efficacy of CD123-CAR-NK cells was directly compared to CD33-CAR-NK cells by targeting AML cells expressing equal levels of CD123 and CD33. To this end, CAR-expressing cells were produced via lentiviral transduction of IL-15/IL-2 expanded primary NK cell preparations from the same donor and CAR expression was adjusted to 30% in both CD123- and CD33- targeting NK cells. CAR efficacy was determined by in vitro cytotoxicity assessment, by in vivo imaging and histology/flow cytometry of bone marrow.

Results: A high CAR expression was achieved for both CD123-CAR-NK cell products generated from CliniMACS Prodigy® and small scale. Imaging analyses revealed an overall reduced leukemic burden in the mice treated with either CD123- or CD33-CAR-NK cells, compared to non-transduced (NT) NK cells, independent of the manufacturing methods. This effect was more pronounced in female mice compared to male mice, which most likely results from differences in weight. The in vivo imaging data was mirrored by flow cytometry data of the in vitro cytotoxicity assessment with varying effector to target ratios (5:1, 1:1, 1:5). Remarkably, the flow cytometry analysis of the bone marrow, isolated 21 days after tumor injection, showed significantly reduced amounts of leukemic cells in all the CAR-NK cell-treated female mouse groups compared to the untreated, as well as NT-NK cell-treated groups. Notably, CD33 targeting by CAR-NK cells showed generally less pronounced effects, concerning the leukemic burden compared to CD123 targeting.

Conclusions: Overall, targeting CD123 with CAR-NK cells in AML was shown to be more efficient than targeting CD33 in a side-by-side comparison in the AML xenograft model. Furthermore, the generation of CAR-NK cells using the CliniMACS Prodigy® system led to an efficient and reliable genetically engineered NK cell preparation. Taken together, our results point out that the automatized manufacturing of CD123 CAR-NK cells with a high anti-leukemic efficacy, provides a promising cell therapeutic concept for treatment of AML.

Disclosure: SM, JKo, JKu, NM and CZ are employees of Miltenyi Biotec.

OP has received honoraria or travel support from Astellas, Gilead, Jazz, MSD, Neovii Biotech, Novartis, Pfizer, and Therakos. He has received research support from Gilead, Incyte, Jazz, Neovii Biotech, and Takeda and is member of advisory boards to Jazz, Gilead, MSD, Omeros, Priothera, Shionogi, and SOBI.

EU has a sponsored research project with Gilead and BMS, and acts as medical advisor at Phialogics. Remaining authors declare no conflict of interest.

J.H.K has advisory roles for Bluebird Bio, Novartis, Roche and Jazz Pharmaceuticals.

5: Cellular Therapies other than CARs

P136 HELP FOR UKRAINIAN HEMATOLOGY PATIENTS (HUP) – A GLOBAL INITIATIVE SUPPORTING THE ESTABLISHMENT OF HEMATOLOGY CENTERS AND HEMATOPOIETIC CELL TRANSPLANTATION PROGRAMS IN DIFFICULT SITUATIONS

Irina Korenkova 1, Sergiy Klymenko2, Oleksandr Lysytsia3, Menachem Bitan4, Olena Lukianets5, Jan M. Zaucha6, Joannis Mytilineos7, Andreas Hochhaus8, Mats Heyman9, Francois Guilhot10, Natacha Bolanos11, Roman Kuts12, Hildegard T. Greinix13, Isabel Sanchez-Ortega14, Mickey Boon Chai Koh15, Bohdan Medvediev1, Susan Landgraf16, Rüdiger Hehlmann17, Mahmoud Aljurf18, Katherina Filonenko19, Reguia Belkhedim18, Torsten Haferlach*20, Arnold Ganser*21, Anna Sureda Balari*22, Damiano Rondelli*23, Dietger Niederwieser*24,25,26

1Kyiv Bone Marrow Transplantation Center, Kyiv, Ukraine, 2Feofaniya Hospital 53, Kyiv, Ukraine, 3National Specialized Children’s Hospital “Ohmatdyt”, Kyiv, Ukraine, 4Meuhedet HMO and The Hebrew University of Jerusalem, Jerusalem, Israel, 5Clinical Center of Oncology, Hematology, Transplantology and Palliative Care of Cherkasy Oblast Council, Cherkasy, Ukraine, 6Medical University of Gdansk, Gdansk, Poland, 7Zentrales Knochenmarkspenderregister Deutschlands (ZKRD), Ulm, Germany, 8Klinik für Innere Medizin II Am Klinikum 1 Universitätsklinikum Jena, Jena, Germany, 9Karolinska University Hospital Astrid Lindgren Children’s Hospital, Stockholm, Sweden, 10University of Poitiers CHU de Poitiers, Poitiers, France, 11Lymphoma Coalition Suite 108, Madrid, Spain, 12Ukrainian Bone Marrow Donor Registry, Kyiv, Ukraine, 13Medical University of Graz, Graz, Austria, 14EBMT, Barcelona, Spain, 15St George’s University Hospitals, London, United Kingdom, 16St. Georg Hospital, Leipzig, Germany, 17ELN-Foundation, Med. Fakultät Mannheim, Heidelberg University Weinheim, Weinheim, Germany, 18King Faisal Specialist Hospital & Research Centre, Oncology (Section of Adult Haematolgy/BMT), Riyadh, Saudi Arabia, 19Medical University of Gdańsk University Clinical Center, Gdansk, Poland, 20MLL Munich Leukemia Laboratory, Munich, Germany, 21Hannover Medical School, Hannover, Germany, 22Institut Català d’Oncologia – Hospitalet, IDIBELL, University of Barcelona, Barcelona, Spain, 23University of Illinois at Chicago MC 734 Division of Hematology/Oncology, Chicago, United States, 24University of Leipzig, Leipzig, Germany, 25Aichi Medical University School of Medicine, Nagakute, Japan, Nagakute, Japan, 26KaunoKlinikos University of Health Sciences, Kaunas, Lithuania

Background: In 2021 most of the Ukrainian patients with hematological diseases and in need of allogeneic hematopoietic cell transplantation (HCT) were treated abroad with governmental funding or not treated. Only 147 autologous and only 6 allogeneic HCT were reported to the EBMT and Worldwide Network for Blood ad Marrow Transplantation (WBMT) in adults from a country of 44 million population in 2021. In the same year, the European Leukemia Net (ELN) and the WBMT/EBMT were approached to support or establish HCT programs for adults in the country.

Methods: After a site visit at different Ukrainian hospitals, daily supervisory telemedicine was initiated in January 2022. A few days after the war started in February, representatives from major hematology societies (ASH, ASCO, EHA, DGHO, ECO, Lymphoma Coalition), transplant organizations (WBMT, EBMT, ASTCT) and physicians from Ukrainian institutions formed the HUP (Help for Ukrainian Hematology Patients) network with participation of the Ministry of Health (MOH) and the World Health Organization (WHO). On weekly video conference meetings, the needs were discussed and provided.

Results: Patients were allocated according to their wishes in Ukrainian Hospitals or abroad. Concomitantly, daily grand rounds were continued to help Ukrainian physicians to treat hematological patients in the country. Medicines not registered in the Ukraine were donated from different sources including sister cities, governments and EU commission. In parallel, standard operating procedures were provided to the institutions. Despite bombing, autologous HCT activities continued and allogeneic HCT programs for adults initiated. The EBMT established a routine tumor board with global experts to discuss treatments in the country or, if not available (e.g. CAR-T cell, bispecific antibody treatment), organized abroad. The Ukrainian unrelated donor registry was supported by the ZKRD to identify unrelated donors. The cooperation of the Ukrainian institutions was outstanding and culminated in the establishment of a national transplant society for adult and pediatric HCT (UABMT). The activity of adult allogeneic HCT from related, unrelated and haploidentical donors rapidly increased from 6 to 45 patients at the BMT center in Kyiv, Regional Hospital in Cherkhasy, and Children Hospital “Ohmatdyt” in Kyiv in the same year 2022 and even more in 2023. Autologous HCT also increased in these and other hospitals in Lviv and Kyiv. It rapidly became apparent that non-HCT infrastructure had to be improved. Despite availability of basic hematological diagnostics, the most sophisticated diagnostics were provided with the help of ASH and ASTCT at the Munich Leukemia Lab (MLL). Samples from the whole country are shipped weekly for cytology, cytogenetics, immunophenotyping, and molecular analyses to MLL and results available free of charge within a few days. Up to now, 2344 analyses from 582 patients were provided.

Conclusions: Despite a hostile environment, international societies, politicians, private institutions, WHO with the personal involvement of local physicians and MOH provided an example on how to establish a sustainable healthcare system for hematological patients in a low resource country in extreme distress. This effort represents a model that by using solidarity and new technologies may greatly impact other countries affected by socio-economic, or conflict challenges.

Clinical Trial Registry: Not applicable.

Disclosure: * senior authors contributed equally to the work

Financial disclosures not relevant to this work

5: Cellular Therapies other than CARs

P137 A RETROSPECTIVE ANALYSIS COMPARING ORCA-T TO POST-TRANSPLANT CYCLOPHOSPHAMIDE BASED ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT IN PATIENTS WITH MATCHED UNRELATED DONORS RECEIVING MYELOABLATIVE CONDITIONING

Amandeep Salhotra 1, Alexandra Gomez Arteaga2, Caspian Oliai3, Sagar S. Patel4, Jeremy Pantin5, Arpita Gandhi6, Bhagirathbhai Dholaria7, Edmund K. Waller8, Samer A. Srour9, Anna Pavlova10, Irene Agodoa10, J. Scott McClellan10, Nathaniel B. Fernhoff10, Mehrdad Abedi11, Everett H. Meyer12

1City of Hope, Duarte, United States, 2Weill Cornell Medicine, New York, United States, 3UCLA Jonsson Comprehensive Cancer Center, Los Angeles, United States, 4University of Utah, Salt Lake City, United States, 5Sarah Cannon Center for Blood Cancer at TriStar Centennial Medical Center, Nashville, United States, 6Oregon Health and Science University, Portland, United States, 7Vanderbilt University, Nashville, United States, 8Emory University, Atlanta, United States, 9MD Anderson Cancer Center, Houston, United States, 10Orca Bio, Menlo Park, United States, 11University of California Davis, Sacramento, United States, 12Stanford University, Stanford, United States

Background: Identifying an allogeneic hematopoietic stem cell transplantation (HSCT) modality that can reduce graft versus host disease (GvHD), lower non-relapse mortality (NRM), and increase relapse free survival (RFS) is key to improving outcomes for patients with hematologic malignancies.

Recently, the utilization of post-transplant cyclophosphamide (PTCy) as prophylaxis against GvHD has increased; however, PTCy-based HSCTs after myeloablative conditioning (MAC) are associated with higher toxicities requiring prolonged hospitalization, NRM and relapse at 1-year. Orca-T is a high-precision cell therapy biologic currently under investigation that includes stem and immune cells, derived from allogeneic donors, and leverages highly purified, polyclonal donor regulatory T cells to control alloreactive immune responses. Orca-T is administered with single agent tacrolimus as GvHD prophylaxis. To evaluate the relative efficacy of these treatment regimens, we retrospectively compared the two treatments utilizing existing data from similar patient populations.

Methods: Publicly available data was obtained from CIBMTR (Gooptu et al, 2021) and included patients transplanted between 2011-2018 who met the following criteria: GvHD prophylaxis with PTCy and calcineurin inhibitor (CNI)/mycophenolate mofetil (MMF), ≥ 18, and AML or ALL in first or second CR, or MDS. Patients with matched unrelated donors (MUD) treated with myeloablative conditioning and a GCSF-mobilized PBSC allograft between 2015 – 2018 were included. For comparison, patients ≥ 18 who received Orca-T between 2019-2022 as part of a multicenter Phase 1b single-arm trial (NCT04013685) were included if they had the following diagnosis: AML, ALL, or MPAL in CR/CRi or MDS; had an 8/8 MUD; and received MAC consistent with the Orca-T Phase 3 study (busulfan/fludarabine/thiotepa (BFT), total body irradiation (TBI)/Cy, or TBI/Etoposide).

Results: Baseline characteristics were similar across both groups for age, gender, and disease distribution (Table). The Orca-T group had higher DRI scores and longer follow-up time. All-grade chronic GvHD-free survival was 73% vs. 54% with Orca-T and PTCy, respectively. 1-year NRM was 3% with Orca-T vs. 16% with PTCy. Relapse-free survival (RFS) and overall survival (OS) at 1 year were 83% vs. 62% and 94% vs. 77% for Orca-T and PTCy, respectively. Notably, RFS and OS results were sustained at 2 years for patients treated with Orca-T.

Conclusions: Based on this retrospective analysis, Orca-T could potentially provide improved outcomes as compared to PTCy based MAC PBSC HSCT in patients with acute leukemia and MDS. The positive impact on RFS, NRM and OS, in addition to reduced GvHD seen with Orca-T highlight the importance of identifying treatment approaches that may be beneficial across all key transplant outcomes. A randomized Phase 3 registrational trial evaluating Orca-T vs. standard alloHSCT is currently ongoing (NCT05316701).

Baseline Characteristics

T-cell replete graft + PTCy/CNI/MMF

Orca-T + single agent tacrolimus

N= 83

N= 37

Patient Age (min, max)

50 (18, 71)

53 (19, 67)

Median Follow-up

361 days

642 days

Female (%)

49%

54%

Primary Disease (%)

-Acute Myeloid Leukemia

49%

41%

-Acute Lymphoid Leukemia

27%

35%

-Myelodysplastic Syndrome

24%

22%

-Mixed Phenotype Leukemia

-

3%

DRI Low/Int

80%

54%

DRI High/Very high

20%

46%

Myeloablative Conditioning

100%

100%

Outcomes, 1 year

All Grade Chronic GvHD-Free Survival, [95% CI] (mild/moderate/severe cGvHD)

54% [44, 66]

73% [54, 84]

Relapse [95% CI]

24% [15, 36]

16% [8, 32]

Relapse Free Survival [95% CI]

62% [50, 71]

83% [68, 92]

Non-Relapse Mortality [95% CI]

16% [10, 27]

3% [2, 18]

Overall Survival [95% CI]

77% [66, 85]

94% [79, 97]

Outcomes, 2 years

Relapse Free Survival [95% CI]

n/a

83% [66, 90]

Overall Survival [95% CI]

n/a

94% [79, 99]

Clinical Trial Registry: NCT04013685 https://classic.clinicaltrials.gov/ct2/show/NCT04013685

Disclosure: Study was sponsored by Orca Bio.

5: Cellular Therapies other than CARs

P138 PHASE I/II STUDY ON INFUSION OF ALLOREACTIVE OR EX VIVO IL-15 STIMULATED NK CELLS AFTER HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS WITH ACUTE LEUKEMIA

Carmen Mestre1,2, Odelaisy León Triana1,2,3, Alfonso Navarro Zapata1,2, David Bueno4, Luisa Sisinni4, Isabel Badell Serra5, Marta González Vicent6, Cristina Beléndez7, Laura Clares Villa1,2, Yasmina Mozo6, Karima Al-Akioui1,2, Victor Galán4, Pilar Guerra1,4, Carlos Echecopar4, Halin Bareke1,2, Cristina Aguirre1,2, Antonio Pérez Martínez 1,2,4,8

1Translational Research in Pediatric Oncology, Hematopoietic Transplantation and Cell Therapy, Hospital La Paz Institute for Health Research-IdiPAZ, Madrid, Spain, 2Pediatric Oncohematology Clinical Research Unit, Spanish National Cancer Center (CNIO), Madrid, Spain, 3Universidad de Castilla-La Mancha, Ciudad Real, Spain, 4La Paz University Hospital, Madrid, Spain, 5Pediatric Hemato-Oncology, Hospital Santa Creu I Sant Pau, Barcelona, Spain, 6Pediatric Hemato-Oncology, Niño Jesús University Children’s Hospital, Madrid, Spain, 7Pediatric Hemato-Oncology, Hospital Gregorio Marañón, Madrid, Spain, 8Universidad Autónoma de Madrid, Madrid, Spain

Background: Alloreactive Natural Killer (allo-NK) cells play a potent role as inducers of the graft-versus-leukemia (GvL) effect in haploidentical stem cell transplantation (haplo-HSCT). In absence of KIR-HLA donor/recipient mismatch, ex vivo IL-15 stimulated NK cells (IL15-NK) may overcome inhibitory signals. Therefore, we proposed a phase I/II multicenter clinical trial to determine the maximum tolerated dose (MTD) of a single infusion of allo-NK or IL-15-NK at day 7 after haplo-HSCT in pediatric patients with acute high-risk leukemias.

Methods: Eighteen pediatric patients with high-risk leukemia, nine in each group allo-NK vs IL15-NK, were recruited between 2020-2023 in 5 hospitals in Spain. We designed a dose-escalation trial (3 + 3) starting with the cohort 1 (up to 2x107 NKcells/kg), then cohort 2 (2-5x107 NK cells/kg) and finally cohort 3 (5-10x107 NK cells/kg). We compared the following outcomes between the two groups: the 1-year leukemia-free survival (LFS), overall survival (OS), GVHD Relapse-Free Survival (GRFS) and prevalence of acute and chronic graft versus host disease (aGVHD, cGVHD), infections, endothelial complications, relapses, and transplant related mortality. We also report the T and NK cell immune reconstitution, the phenotypic profile and functional capacity of NK cells in both groups and the alloreactive NK cell population in the Allo-NK group up to 360 days after haplo-HSCT.

Results: Nine patients (4 in allo-NK and 5 in IL15-NK), 7 patients (4 and 3) and 2 patients (1 in each group) were recruited in cohort 1, 2 and 3, respectively. The baseline characteristics of the two groups were comparable except for a higher prevalence of B cell acute lymphoblastic leukemia (78% vs 11%, p=0.018) in the IL15-NK group. The NK cells from the IL15-NK group exhibited higher expression of CD25 and CD69 receptors and cytotoxicity than allo-NK group. Within the allo-NK group, the most prevalent KIR-HLA mismatch clone was KIR2DL1 (55.6%).

The 1-year LFS, OS, and GRFS, of the entire cohort were 75.4% (95% CI: 56.9-99.8), 76% (95% CI: 57.9-99.8), and 66.7% (48.1-92.4%). The prevalence of grade III-IV aGVHD and moderate-severe cGvHD was 22.2% and 12.5%. The prevalence of any infection, endothelial complications, relapse, and transplant related mortality were 72%, 33%, 11% and 5% respectively. We found no significant differences between cohorts or the 3 dose escalation cohorts.

We observed a trend towards higher numbers of NK cells (0.78·103cell/µl vs 0.36·103cell/µl), T cells (1.54·103cell/µl vs 0.48·103cell/µl) in the IL15-NK arm compared to the Allo-NK arm in the first month post-transplantation. In the NK phenotype, we observed a tendency to increase the expression of CD16 and CD57 receptors between 30 to 360 days after transplantation in both groups. We observed an optimal functional capacity of NK cells against the K562 cell line during follow-up in both arms, with no differences between them.

Conclusions: In our study, the clinical outcome, NK cell phenotype and cytotoxicity were similar in both arms and cohorts. We observed no association between toxicity and different cohort of NK cell doses. Unfortunately, we were unable to complete cohort 3. A larger randomized trial is needed to provide more robust conclusions.

Clinical Trial Registry: NCT05304754

Disclosure: Nothing to declare.

5: Cellular Therapies other than CARs

P139 HARNESSING CIK CELL INTERVENTION FOR ACUTE LEUKEMIA OR MDS RELAPSE POST ALLO-HSCT

Eva Rettinger 1, Marie Luedtke1, Emilia Salzmann-Manrique1, Sabine Huenecke1, Melanie Bremm1, Claudia Cappel1, Gesine Bug1, Johann Greil2, Roland Meisel3, Eva Maria Wagner-Drouet4, Jan-Henning Klusmann1, Halvard Bonig5, Peter Bader1

1Goethe University Frankfurt, University Hospital, Frankfurt am Main, Germany, 2University Hospital, Heidelberg, Germany, 3Heinrich-Heine-University Duesseldorf, Duesseldorf, Germany, 4Johannes Gutenberg-University Mainz, Mainz, Germany, 5Goethe University Medical Center, Institute of Transfusion Medicine and Immunohematology, and German Red Cross Blood Center Frankfurt, Frankfurt am Main, Germany

Background: Relapse is the most common cause of treatment failure after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Our previous data supported the implementation of routine disease monitoring to guide immune interventions in patients allografted for high-risk leukemia. Consequently, we developed a donor-derived, IL-15-activated cytokine-induced killer cell therapy (CIK) to complement the graft-versus-leukemia (GvL) activity of an allo-HSCT, that subsequently became an approved investigational advanced therapy medicinal product (§4b Abs. 3 AMG, license number: PEI.A.11630.01.1).

Methods: In the prospective, multicenter phase I/II study presented here, CIK is being investigated for its feasibility, safety, and efficacy in relapsed patients with acute leukemia or myelodysplastic syndrome (MDS) after allo-HSCT (EudraCT number: 2013-005446-11).

Results: From March 2016 to March 2020, 23 patients (children, N=13; adults, N=10) were included in the study. Patients diagnosed with AML (N=17), ALL (N=5), and MDS (N=1) received their first (N=20, 87%) or second (N=3, 13%) allo-HSCT either in complete remission (CR) (CR1-4, N=21, 91%) or not in remission (NR) (N=2, 9%) at a median age of 7 (range, 2-12), and 56 (range, 20-71) years (P< 0.001). Allografts comprised bone marrow (N=9, 39%) or peripheral blood stem cells (N=9, 61%) from human histocompatibility antigen (HLA)-matched (N=18, 78%), or HLA-haploidentical donors (N=5, 22%).

Mixed donor chimerism and/or minimal residual disease (MRD) guided CIK intervention in 21 cases, and cytogenetics in another 2 patients at a median of 105 (range, 29 – 440) and 199 (range, 99 – 1321) days post-transplant, which were significantly different among children and adults (P=0.050). CIK intervention applied thereafter within 20 days (range, 0-69) included a median number of 2 infusions, (range, 1-8) and a median cumulative T cell dose of 6 x106/kG (range, 1-280.7) per patient, irrespective of age and donor type.

A total of 67 CIK doses were applied without acute side effects. Five patients developed acute graft-versus-host disease (aGvHD) grade ≥ II (children, N=3, aGvHD grade II; adults, N=2, aGvHD grade II and III). Limited chronic GvHD was reported in 3 cases. CIK was not associated with non-relapse mortality (NRM), that was observed in one patient.

20 patients met the inclusion criteria and received escalating CIK doses according to the protocol. CIK intervention was discontinued in one child due to the decision of the treating physicians. Complete molecular response (CMR) assessed after 300 days was 73% in children and 50% in adult patients, with CI CMR rates of 100%, 75%, and 57% observed among MDS, ALL and AML patients. pOS and pEF rates at 50 months were 62 and 50% in children and 73% and 47% in adults, and were 71 and 57% in AML, 80 and 30% in ALL and 0% in MDS patients.

Conclusions: In summary, the median time to overt relapse from molecular disease allowed to safely immune intervene with CIK therapy in most of the patients, assigning CIK a role as direct killers and indirect modulators of innate and adaptive immune responses. Based on this, a subsequent multicenter clinical phase III trial is in preparation for patients with pediatric AML in Germany.

Clinical Trial Registry: EudraCT number: 2013-005446-11

Disclosure: Peter Bader received a research grant and payment for lectures from Novartis, royalties and consulting fees from Medac, payment for lectures from Medac, Neovii and Amgen and support for attending meetings from Neovii and Medac. Jan-Henning Klusmann has advisory roles for Bluebird Bio, Novartis, Roche and Jazz Pharmaceuticals.

5: Cellular Therapies other than CARs

P140 RISK FACTORS FOR THE DEVELOPMENT OF GRAFT-VERSUS-HOST-DISEASE FOLLOWING DONOR LYMPHOCYTE INFUSION AFTER ALLOGENEIC STEM CELL TRANSPLANTATION

Eva Koster1, Peter von dem Borne1, Peter van Balen1, Erik Marijt1, Jennifer Tjon1, Tjeerd Snijders2, Daniëlle van Lammeren3, Hendrik Veelken1, Frederik Falkenburg1, Liesbeth de Wreede1, Constantijn Halkes 1

1Leiden University Medical Center, Leiden, Netherlands, 2Medical Spectrum Twente, Enschede, Netherlands, 3HagaZiekenhuis, The Hague, Netherlands

Background: Unmodified donor lymphocyte infusions (DLI) after allogeneic stem cell transplantation (alloSCT) can boost the Graft-versus-Leukemia effect, but may induce severe Graft-versus-Host-Disease (GvHD). Timing, HLA matching and number of infused lymphocytes are established factors determining the GvHD risk after DLI. We analyzed which other factors influence this risk.

Methods: We investigated three potential risk factors: presence of patient-derived antigen-presenting cells (estimated by the percentage of bone marrow [BM] patient cells) and lymphopenia (measured by the absolute lymphocyte count) at time of DLI and presence of a viral infection/reactivation close to DLI. The cohort consisted of patients with acute leukemia or myelodysplastic syndrome who prophylactically or pre-emptively received DLI after alemtuzumab-based alloSCT from a 10/10 HLA-matched donor. Patients with a high anticipated relapse risk received 0.3x106 or 0.15x106 T cells/kg in case of a related donor (RD) or unrelated donor (UD), respectively, at 3 months after alloSCT (n=88; median age 59 years; 39 RD, 49 UD). All other patients received 3x106 or 1.5x106 T cells/kg, respectively, at 6 months (n=76; median age 58 years; 30 RD, 46 UD). Separate multi-state models were constructed for the 3- and 6-month DLI cohorts, both with the following events: first DLI, second DLI, systemic therapeutic immunosuppression (tIS) for GvHD, stop tIS, relapse, and death. For the transition ‘first DLI to tIS’, separate multivariable Cox proportional hazards regression models were fitted for the three factors of interest, each combined with a composite factor consisting of conditioning intensity (myeloablative [MA] vs nonmyeloablative [NMA]) and donor type. Viral infection was time-varying and only fitted for the 3-month DLI. For the transition ‘tIS to death’, univariable Cox proportional hazards regression models were fitted with viral infection (3-month DLI) or BM chimerism (6-month DLI).

Results: The 3-month cumulative incidence of GvHD requiring tIS was 28% (95%-CI 20-40) after the low-dose 3-month DLI and 30% (95%-CI 22-43) after the 6-month DLI. For both DLIs, NMA-conditioned patients with an UD had the highest risk of GvHD (see table). For the 3-month DLI, viral infection within 1 week before and 2 weeks after DLI was an additional significant risk factor (hazard ratio [HR] 3.66 compared to no viral infection) for GvHD. At six months after alloSCT, viral infections were rare and not associated with GvHD. In contrast, mixed BM chimerism (HR 3.63 for ≥5% mixed chimerism [MC] compared to full donor chimerism [FDC]) was an important risk factor for GvHD after the 6-month DLI, and lymphopenia showed a trend for association with GvHD. During tIS for GvHD, patients with a viral infection close to the 3-month DLI and patients with ≥5% MC at time of the 6-month DLI had a nonsignificant higher risk of dying (HR 1.8 [95%-CI 0.6-5.6] and HR 2.0 [compared to <5% MC; 95%-CI 0.6-6.4], respectively). The impact of these factors on the current GvHD-relapse-free survival is shown in the figure.

Model

Factor

3-month DLI

HR (95%-CI), p-value

6-month DLI

HR (95%-CI), p-value

including BM chimerism

Conditioning/donor:

- MA UD vs MA RD

0.42 (0.05 - 3.61), 0.426

0.57 (0.13 - 2.42), 0.446

- NMA RD vs MA RD

0.55 (0.10 - 2.93), 0.484

0.57 (0.15 - 2.18), 0.410

- NMA UD vs MA RD

3.20 (1.12 - 9.13), 0.029

1.90 (0.66 - 5.47), 0.234

BM chimerism:

- 1-4% MC vs FDC

0.92 (0.32 - 2.67), 0.883

2.05 (0.70 - 6.05), 0.192

- ≥5% MC vs FDC

1.16 (0.39 - 3.48), 0.787

3.63 (1.16 - 11.29), 0.026

including absolute lymphocyte count

Conditioning/donor:

- MA UD vs MA RD

0.35 (0.04 - 2.93), 0.333

0.66 (0.17 - 2.66), 0.563

- NMA RD vs MA RD

0.52 (0.10 - 2.59), 0.424

1.01 (0.25 - 4.03), 0.993

- NMA UD vs MA RD

3.05 (1.20 - 7.70), 0.019

4.09 (1.35 - 12.42), 0.013

Absolute lymphocyte count (x106\l):

- 500-999 vs ≥1000

1.07 (0.46 - 2.50), 0.867

-

- <500 vs ≥1000

0.78 (0.29-2.11), 0.630

-

- <1000 vs ≥1000

-

2.05 (0.94 - 4.45), 0.069

including viral infection

Conditioning/donor:

- MA UD vs MA RD

0.33 (0.04 - 2.77), 0.308

- NMA RD vs MA RD

0.50 (0.10 - 2.51), 0.403

- NMA UD vs MA RD

3.10 (1.23 - 7.83), 0.017

Viral infection:

- 1 week before to 2 weeks after DLI vs none

3.66 (1.71 - 7.87), <0.001

- 2-8 weeks after DLI vs none

0.64 (0.08 - 4.96), 0.668

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (19)

Conclusions: These data demonstrate that other factors than only timing, dosage and patient-donor relation determine the risk for GvHD after DLI and should be taken into account when administering prophylactic DLI.

Disclosure: Nothing to declare.

5: Cellular Therapies other than CARs

P141 RELAPSE AFTER HSCT: MAY DLI/CIK CELLS STILL PLAY A ROLE IN THE CELLULAR THERAPY ERA? DLI/CIK AS A RESCUE STRATEGY FOR CHILDREN WITH POST-TRANSPLANT MRD-POSITIVITY

Francesca Limido1,2, Alex Moretti2,3, Riccardo Carnevale1,2, Sara Napolitano2, Francesca Vendemini2, Sonia Bonanomi2, Andrea Biondi1,2,3, Giuseppe Gaipa3, Adriana Balduzzi 1,2

1University of Milano Bicocca, Monza, Italy, 2Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 3Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy

Background: Allogeneic HSCT is a standard treatment for high-risk hematologic malignancies, yet disease relapse remains a challenge and a primary contributor to treatment failure. Post-HSCT relapse has a very poor prognosis with limited therapeutic options mainly encompassing donor lymphocyte infusion (DLI) and a second transplant. Recognizing the urgent clinical need for novel therapies, we recently investigatean approach involving the combination of DLI and cytokine-induced killer (CIK) cells expressing CD3 and CD56 antigens. This approach aims to enhance the GvHD risk. CIK cells, generated through in vitro stimulation of donor peripheral blood mononuclear cell, exhibit dual functionality, retaining T cell specificity while acquiring NK cell-like, non-specific anti-tumoral cytotoxicity. The aim of our study is to investigate the potential role of DLI/CIK infusion in paediatric HSCT recipients, assessing its impact on relapse treatment and prevention.

Methods: We present the analyses of pediatric patients enrolled in a phase II study, who received sequential DLI/CIK infusion (EudraCT n: 2008-003185-26 and compassionate use) comparing outcomes and GvHD risk with a pediatric cohort received DLI alone in our center.

Results: Among the 25 patients (ages 1-16ys, 13 female) who received DLI or DLI/CIK after HSCT from June 2017 to October 2022, disease statuses before HSCT varied, including hematological disease (1/25), molecular minimal residual disease (MRD) (14/25), negative evaluations (8/25). In 2 cases the disease evaluation was not performed. 12 patients underwent HSCT from HLA-matched Unrelated Donor (URD), 8 from identical siblings, and 5 underwent haploidentical HSCT. 20 out of 25 patients received therapy for molecular relapse, 2/25 for mixed chimerism with MRD negative, 2/25 for morphological relapse, and 1 for high-risk of relapse (pre-emptive) as showed in table 1. DLI cohort included 13 patients (age 7 ys, median doses administered 8), while the DLI/CIK group included 12 patients (age 9 y) who received sequential infusion of DLI and CIK (median doses of DLI 3; median doses of CIK 3). Among DLI group, 10 patients (77%) did not manifest GvHD, while 3 (23%) had aGvHD grade I-II (skin and lower gut). Only one patient developed mild cGvHD (oral cavity). Among DLI/CIK patients, 8 (67%) did not showed any GvHD manifestations, while 4 patients developed aGvHD. Interestingly, 3/4 developed sign and symtoms of GvHD after DLI, while 1 had aGVHD post-CIK (grade III, involving lower gut, and liver). Following DLI therapy, 9/13 patients had negative molecular MRD, 2 relapsed, and 2 manteined a positive molecular MRD. After DLI/CIK therapy, 10/12 patients had negative molecular MRD, and 2 had relapsed. The progression free survival was 43 months after DLI infusion. The overall survival at 24 months was 67,7% for DLI and 81,8% for DLI/CIK.

Table 1

Disease

GvHD post-HSCT

Indication

ALL

19 (76%)

Yes

11 (44%)

Molecular MRD +

20 (80%)

AML

4 (16%)

No

14 (56%)

Morphological relapse

2 (8%)

HL

1 (4%)

Mixed chimerism

2 (8%)

JMML

1 (4%)

Pre-emptive

1 (4%)

Conclusions: Our findings support the efficacy of sequential DLI and CIK cell administration as a viable therapeutic option for molecular or cytogenetic relapse post-HSCT. Notably, CIK cell administration without preceding DLI demonstrated a reduced risk of GvHD, a fundamental aspect to be considered especially in a pre-emptive strategy.

Clinical Trial Registry: EudraCT n: 2008-003185-26

Disclosure: The authors disclose no potential conflict of interest

5: Cellular Therapies other than CARs

P142 PHASE I/II PRELIMINARY RESULTS OF HUMAN PRO-T-CELLS MANUFACTURING IN VITRO TO ACCELERATE IMMUNE RECONSTITUTION AFTER UMBILICAL CORD BLOOD TRANSPLANTATION IN ADULT PATIENT WITH HEMATOLOGIC MALIGNANCIES

Elisa Magrin1, Jean-Sébastien Diana 1, Clotilde Aussel1, Eden Schwartz1, Jinmi Baek1, Naim Bouazza2, Aurélie Gabrion1, Alice Girardot1, Axelle Maulet1, Brigitte Ternaux1, Aurore Touzart1, Caroline Tuchmann-Durand3, Ludovic Lhermitte1, Vahid Asnafi1, Edouard Forcade4, Huynh Anne5, Jean-Marc Treluyer1,2, Olivier Hermine1,6,3, Marina Cavazzana1,6,3

1Hôpital Necker-Enfants Malades, APHP, Paris, France, 2GHU Paris Centre Université Paris Cité, Paris, France, 3Institut Imagine, Paris, France, 4Centre Hospitalier Universitaire (CHU) de Bordeaux, Pessac, France, 5Centre Hospitalier Universitaire (CHU) de Toulouse, Toulouse, France, 6Université Paris Cité, Paris, France

Background: Allogeneic stem cells transplantation (AlloSCT) is the treatment of choice for high- risk acute myeloid leukemias in complete first remission after induction therapy. Umbilical cord blood (UCB) grafts were frequently used for patients lacking an HLA- matched family donor as well as in the absence of an appropriate unrelated donor. UCB transplantations are associated with risk of acute and chronic GVHD, post- transplant immunodeficiency with increased risk of infections and relapse. The risk of infection and non- relapse mortality (NRM) or transplant- related mortality (TRM) is significantly higher in UCB transplantations as compared to matched-sibling donor, 10/10 matched-unrelated donor, or even haplo-identical SCT. All of these risks have been linked to a significant delay in immune reconstitution including various immune cell populations like CD4 and CD8 T cells, Treg, NK, iNKT, pDC and others, and thus it is now less used.

We therefore hypothesized that if T-cell-mediated immunity was rapidly generated after a partially HLA-compatible UCB transplantation, the risk of infection and relapse will be reduced without increasing the risk of GVHD. Thus, we set up this protocol based on the use of in vitro- committed Human T Lymphoid Progenitor cells (HTLPs) that seed the thymus immediately after transplantation, accelerating the production of mature and polyclonal T cells.

Methods: The safety and efficacy of CD7+ Human T lymphoid Progenitors (HTLP) was evaluated in adult hematologic malignancy. The investigational product consists of the cell suspension obtained after 7 days of ex-vivo CD34+ cells culture from UCB on the fusion protein DL-4, Retronectin® and a combination of cytokines, to generate CD7+ pro-T-cells. After a conditioning regimen, without serotherapy, the HTLP cell suspension was injected intravenously at the time of unmanipulated UCB HSCT on D0. To assess the procedure’s safety, the dose-limiting toxicity (DLT) and the efficacy, we planned an escalation dose, starting from 0.5x106 CD7 + /kg, in order to evaluate the incidence of grade III-IV graft- versus- host disease (GvHD) and the presence of > 50/mm3 CD4 + T cells at two consecutive measures within 4 months post SCT.

Results: We assessed the safety of the drug product (DP) at starting dose of 0.5x106 CD7 + /kg in two patients. They underwent to UCB transplantation for anaplastic lymphoma and high risk acute myeloid with a median differentiation over 88% at a proliferation index of 18. No CD3+ cells were detectable on infused DP after differentiation. So far, no early or mid-term toxicity due to the administration of HTLP has been reported. Primary engraftment was confirmed for both patients, and no aGVHD occurred. More than 50/mm3 circulating CD4 + CD3 + TCRαβ + T cells before 2 months of follow-up were observed and rapidly expanded to 200/mm3 before 4 months.

Conclusions: First dosing of 0.5x106 CD7 + /kg in the first human clinical trials using UCB confirmed safety and reproducible GMP manufacturing. Thus, HLTP may offer an exciting perspective for improving immune reconstitution in alternative hematopoietic transplants. Efficacy will be tested in future patients with a higher dose. Next two patients have already been enrolled and will be treated by the end of January 2024.

Clinical Trial Registry: EudraCT Number: 2019-004883-23

Disclosure: Nothing to declare.

5: Cellular Therapies other than CARs

P143 INNATE DONOR EFFECTOR ALLOGENEIC LYMPHOCYTES AFTER ALLOGENEIC STEM CELL TRANSPLANTATION - THE IDEAL TRIAL

Lia Minculescu 1, Lone Smidstrup Friis1, Soeren Lykke Petersen1, Brian Thomas Kornblit1, Niels Smedegaard Andersen1, Ida Schjoedt1, Helle Lesley Andersen1, Eva Haastrup1, Lisbeth Pernille Andersen1, Henrik Sengeloev1

1Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark

Background: Some of the main obstacles after allogeneic stem cell transplantation (SCT) remain disease recurrence and graft-versus-host disease (GVHD). Studies have shown that high amounts of gamma delta T cells and natural killer (NK) cells in transplanted stem cell graft as well as during early immune reconstitution are associated with improved relapse-free survival (RFS) and less acute GVHD in patients transplanted for myeloid malignancies. The aim of this trial is to administer innate donor lymphocyte infusion (iDLI) containing purified NK and gamma delta T cells from the original stem cell donor early post-transplant in order to test the anti-leukemic and immune-regulating properties of these effector cells in a clinical setting.

Methods: This is an on-going, open-label, genetically randomized phase 2b/3 clinical trial including adult transplant patients with the diagnoses acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Patients with a full HLA-matched sibling transplanted with a peripheral blood stem cell graft are randomized for the intervention group. The control group consists of patients with 10/10 HLA-matched unrelated donors and are matched using a propensity score including age, diagnosis, conditioning regimen, disease stage and risk score. The intervention consists of a prophylactic iDLI from the original stem cell donor 14 days after transplantation. The donor lymphocytes are harvested using the Spectra Optia Apheresis System (Terumo BCT) and the cell product is TCR alpha beta/CD19 depleted using the CliniMACS Prodigy System (Miltenyi Biotec). This procedure results in a cell product depleted of conventional alpha beta T cells and B cells and thereby consisting primarily of NK cells and gamma delta T cells. Targeted cell doses are >10e7 NK cells/kg and >10e6 TCR gamma delta cells/kg. The product is infused freshly to the patient the following day. All immune suppression and supportive post-transplant treatment remains standard of care for all patients. The primary outcomes are 2-year relapse-free survival and acute GVHD. Immune phenotyping of the stem cell graft, the iDLI and patient immune reconstitution is performed.

Results: 11 of the intended 40 patients in each group have been included and treated. Results for the intervention group are presented here. Median patient age was 60 (36-73) years with 8 AML patients and 3 MDS patients included. Median number of CD34 positive stem cells at transplant was 7.1 (4.6-9.6) x 10e6/kg. Median transplanted cells with the iDLI were 22.5 (5.4-47) x 10e6/kg for NK cells and 1.8 (0.8-22.5) x 10e6/kg for gamma delta T cells. No adverse events or unexpected side effects were observed with the iDLI infusion. Ten patients are alive and in remission by the current median follow-up time of 399 (155-615) days. One AML patient died from CNS relapse 146 days post-transplant. Two patients experienced grade 2-4 acute GVHD.

Conclusions: One fourth of the intended patient number have been randomized and treated with iDLI. Production and infusion of the iDLI is feasible and no adverse side effects have been observed. Clinical results await full patient inclusion and appropriate follow-up time.

Clinical Trial Registry: ClinicalTrials.gov ID NCT05686538

Disclosure: Nothing to declare.

5: Cellular Therapies other than CARs

P144 CD45RA DEPLETION PRESERVES PROPORTIONS AND CLONAL BREATH OF VIRUS-SPECIFIC T CELLS

Amandine Pradier 1,2, Astrid Melotti1, Chiara Bernardi1,2, Simona Pagliuca3, Sisi Wang1, Sarah Morin2, Federica Giannotti2, Stavroula Marouridi-Levrat2, Yves Chalandon1,2, Federico Simonetta1,2, Anne-Claire Mamez2

1University of Geneva, Geneva, Switzerland, 2Geneva University Hospitals, Geneva, Switzerland, 3Nancy University Hospital, Vandœuvre-lès-Nancy, France

Background: Quantitative and qualitative abnormalities in T cell subsets are responsible of infection-related morbidity and mortality after allogeneic hematopoietic stem cell transplantation (HSCT). Donor lymphocytes infusions (DLI) have the potential to improve immune reconstitution thus contributing to the prevention and treatment of infections. However, DLI administration carries a significant risk of GvHD. Over the last decade, CD45RA depletion (RAdep) gained popularity as a strategy to reduce the alloreactive potential of DLI by depleting their naïve T cell compartment. However, CD45RA is also expressed on a significant fraction of antigen-experienced T cells and the impact of RAdep on the virus specific T cell compartment is currently unknown. In the present work, we analysed the impact of RAdep on the proportion, phenotype and T-cell receptor (TCR) repertoire of CMV- and EBV-specific T cells.

Methods: Peripheral blood mononuclear cells from healthy controls (n=4) were CD45RA depleted using the CD45RA depletion kit (Miltenyi Biotech) according to the manufacturer’s instructions. Major T cell subsets defined as Naïve (CD45RA + CD27 + ), Memory (Mem: CD45RA-CD27 + ), Effector Memory (EM: CD45RA-CD27-) and TEMRA (CD45RA + CD27-) before and after CD45RA depletion were analysed by flow cytometry. Virus-specific CD4 and CD8 T cells were identified in total and RAdep T cells by flow cytometry based on IFNg and/or TNFa intra-cytoplasmic expression after 6h in vitro stimulation with peptides derived from CMV and EBV. TCRβ sequencing was performed on genomic DNA using LymphoTrack TRB Assay (Invivoscribe) and analyzed using MiXCR and ImmunArch. Viral-specific T cell clones were identified based on previously reported sequences summarized in Pagliuca et al., JCI Insight 2021.

Results: As expected, naïve T cells were almost completely absent after RAdep. In addition, RAdep resulted in a significant reduction in TEMRA CD4 T cells mirrored by an increase in CD4 Mem and EM cells. TCRβ sequencing revealed no impact of the RAdep on the TCR repertoire in terms of number of unique clonotypes and diversity. Importantly, the proportions of CMV-specific and EBV-specific T cells as quantified by flow cytometry and TCRβ sequencing were not affected by RAdep. Conversely, RAdep significantly impacted the phenotype of viral specific T cells, with reduction of CD8 TEMRA cells (CMV p=0.029; EBV p=0.027) and enrichment of CD8 Mem for CMV and EM for EBV specific T cells. RAdep had no significant impact on the clonal breath of CMV and EBV specific TCR repertoire as revealed by a similar number of virus-specific unique clonotypes. Conversely, we observed a slight but significant increase in clonal restriction as revealed by a higher Simpson index in CMV-specific T cells (p=0.029) and similar trend in EBV-specific T cells (p=0.057) after RAdep.

Conclusions: CD45RA depletion preserves the proportions and the clonal breath of the TCR repertoire of anti-viral T cells, which display a less terminally differentiated phenotype. Our results support the use of CD45RA-depleted DLI as a source of virus specific T cells to restore immunity and prevent infections after HSCT.

Disclosure: Yves Chalandon has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution.

5: Cellular Therapies other than CARs

P145 LAYING THE FOUNDATIONS FOR AN IMMUNOPOTENCY TEST FOR ALLOGENEIC MESENCHYMAL STROMAL CELLS

María Esther Martínez-Muñoz 1,2, Trinidad Martín-Donaire2, Rocío Sánchez2, Elvira Ramil2, Rosalía Alonso-Trillo2, Nuria Panadero1, Rocío Zafra2, Enrique Andreu3, Gloria Carmona4, Ana María García-Hernández5, Miriam López-Parra6, Gustavo Melen7, Luciano Rodríguez-Gómez8, Rosa Yáñez9, Cristina Avendaño-Solá1,2, Jose María Moraleda5, María Eugenia Fernández10, Rafael Francisco Duarte1,2

1Hospital Universitario Puerta de Hierro Majadahonda, Madrid, Spain, 2Instituto de Investigación Sanitaria Puerta de Hierro - Segovia de Arana (IDIPHISA), Madrid, Spain, 3Clínica Universidad de Navarra. Instituto de Investigación Sanitaria de Navarra (IDISNA), Navarra, Spain, 4Red Andaluza de diseño y traslación de Terapias Avanzadas de la Fundación Progreso y Salud (RAdytTA), Andalucía, Spain, 5Hospital Universitario Virgen de la Arrixaca. Instituto Murciano de Investigación Biosanitaria IMIB-Arrixaca, Murcia, Spain, 6Hospital Clínico Universitario de Salamanca. Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain, 7Hospital Universitario Infantil Niño Jesús. Fundación para la Investigación Biomédica Hospital Niño Jesús (FIBHNJS), Madrid, Spain, 8Banc de Sang i Teixits (BST), Barcelona, Spain, 9Centro de Investigaciones Energéticas, Medioambientales y Tecnológicas (CIEMAT), Madrid, Spain, 10Hospital General Universitario Gregorio Marañón. Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain

Background: Mesenchymal stromal cells (MSC) are used in a wide range of diseases, for immunomodulatory purposes, as well as regenerative therapy in early to late-phase clinical trials and have received approval for the treatment of disorders like Chrohn’s disease, acute myocardial infarction or spinal cord injury in some countries. There is a broad consensus that, despite different tissue sourcing and varied culture expansion protocols, human MSC products likely share fundamental mechanisms of action, but no standard potency tests to evaluate their immunomodulatory activity have been developed thus far. Validation of such assays would satisfy both scientific research and regulatory requirements for medicinal products, whilst it would optimize the use of MSC in immune disorders. These tests should ideally offer reproducible results in a timely manner to guide batch release.

Several studies have identified biomarkers that may be associated with the immunoregulatory potency of MSC, but these have not been clinically validated or analyzed in MSC from sources other than bone marrow (BM).

The aim of this study is to analyze the expression of the array of biomarkers that have been described so far, in BM-derived MSC, and provide the basis for the development of an immunopotency test, through multicenter validation for MSC from various sources and in clinical trials within the framework of the RICORS-TERAV Spanish network.

Methods: Allogeneic BM-derived MSC compliant with the ISCT criteria [Dominici M., 2016] and stored in cryovials were thawed and cultured until ≥70% confluence was reached. The expression of different markers was analyzed, both by flow cytometry (IDO-1, PDL-1, TNF-R1 and VCAM-1) and by quantitative RT-PCR (genes IDO-1, PTGS2, TNFAIP6 and ILR1RN), at baseline, and following a 24-hour stimulation in vitro with increasing doses of IFN-γ and TNF-α (1, 3, and 10 ng/ml for both stimuli).

Results: Sixteen MSC samples from eleven donors were analyzed. Stimulated MSC with IFN-γ and TNF-α showed an increase in the expression of the intracellular marker IDO-1 and the cell surface markers PDL-1 and VCAM-1 by flow cytometry, compared to baseline, but no differences were observed for TNF-R1 (Figure, percentage of expression). In addition, this stimulation consistently led to an enhanced expression of the gene IDO-1, as well as PTGS2 (COX-2), TNFAIP6 (TSG-6) and ILR1RN (Figure, RQ log10). This increase in the expression of all biomarkers was dose-dependent. Donor-to-donor differences were observed in the expression of IDO-1 and VCAM upon stimulation, outlining two discernible donor profiles of high and low level of response.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (20)

Conclusions: BM-derived MSC express biomarkers that have been associated with their immunomodulatory activity (IDO-1, PDL-1, VCAM-1, PTGS2, TNFAIP6 and ILR1RN) in response to IFN-γ and TNF-α stimuli, in a dose-dependent manner. This preliminary analysis of biomarker performance provides the basis for developing an in vitro potency test that can offer fast and reproducible results. Multicenter validation of this assay with MSC from different sources will be carried out by the RICORS-TERAV network, with funding from the Spanish ISCiii (ICI23/00085) in collaboration with regulatory agencies, to support the development of MSC immunomodulatory therapy in multiple indications.

Disclosure: Nothing to declare.

5: Cellular Therapies other than CARs

P146 CHARACTERIZATION OF CYTOKINE RELEASE SYNDROME AND NEUROTOXICITY IN COMMERCIALLY AVAILABLE BISPECIFIC ANTIBODIES AND CHIMERIC ANTIGEN RECEPTOR T-CELLS IN MULTIPLE MYELOMA

Eden Biltibo1

1Vanderbilt University Medical Center, Nashville, United States

Background: Relapsed refractory multiple myeloma patients with four or more prior lines of therapy are now eligible to receive B-cell maturation antigen (BCMA) directed chimeric antigen receptor (CAR) T-cell therapies or BCMAxCD3 or GPRC5DXCD3 bispecific antibodies (BsAbs). But these life-saving medications are given only in few institutions due to their side effect profiles, including cytokine release syndrome (CRS) and immune effector cell associated neurotoxicity syndrome (ICANS), limiting accessibility. Better characterization of the frequency, timing, intensity, and management of their side effects will allow safe expansion of treatment administration sites.

Methods: A systematic literature search was conducted to identify clinical trials that investigated FDA approved BsAbs and CAR T-cell therapies in RRMM patients. Eleven abstracts and publications were included for this analysis. Fisher’s exact test and Wilcoxon rank sum test was used to compare variables. P <0.05 was considered statistically significant.

Results: Neurotoxicity included ICANS, headache, seizures, anosmia, dysgeusia, dizziness, altered sensation, tremor, cogwheel rigidity, depressed level of consciousness, paresthesia, altered state of consciousness, aphasia, encephalopathy, somnolence, confusion, delirium and disorientation. CRS and neurotoxicity was reported in a study population of 746 patients who received BsAbs across 6 studies and in 1310 patients who received CAR T-cell therapy across 6 studies. In this data, 87.7% (range: 80-95) in the CAR T group and 57.6% (range: 13-76.3) in BsAb group experienced CRS (p-value: 0.003). Grade 3 or higher CRS was present in 5.5% (range:3-10) of the CAR T-cell and 1.5 % (range: 0-4.9) of the BsAb groups (p-value: 0.006). Median time of CRS onset was 4.5 days in the CAR T-cell and 2 days in the BsAb groups (p-value: 0.503). Median duration of CRS was 4 days in CAR T-cell and 2 days in the BsAb groups (p-value: 0.003). Neurotoxicity was reported in 16.4% (range: 5.3-28) of CAR T-cell and in 8.36% (range: 3-14.5) of BsAb recipients (p-value: 0.022). Grade 3 or higher neurotoxicity was identified in 2.17% (range: 0-5) of CAR T-cell and 1.27 (range: 0-3) of BsAb recipients (p-value: 0.507). Median time of neurotoxicity onset was 5.5 days in the CAR T-cell and 3 days in the BsAb groups (p-value: 0.429). Median duration of neurotoxicity was 3.5 days in CAR T-cell and 2 days in the BsAb groups (p-value: 0.427). There was no statistically significant difference in the proportion of tocilizumab and steroid use for CRS and ICANS among the groups.

Full size table

Conclusions: BsAbs demonstrated lower rate and severity of CRS and decreased incidence of neurotoxicity. Majority of BsAb recipients experienced CRS within the first cycle, during the step-up doses or following the first full dose. These findings indicate that BsAbs can be safely administered in community cancer centers, in the outpatient setting, expanding its reach to rural and economically disadvantaged patient populations. Prospective randomized data is required to validate these findings.

Clinical Trial Registry: Not available

Disclosure: Eden Biltibo

COI: Consulting: BeiGene

Source of Funding: American Society of Hematology, Robert A Winn Career Development Award, Multiple Myeloma Research Foundation.

5: Cellular Therapies other than CARs

P147 TRIAL IN PROGRESS: OBSERVATIONAL, POST-AUTHORISATION SAFETY STUDY TO DESCRIBE SAFETY AND EFFECTIVENESS OF TABELECLEUCEL IN PATIENTS WITH EBV + PTLD IN A REAL-WORLD SETTING IN EUROPE

François Denjean1, Asmaa Zkik1, Constance Battin1, Sophie Blanchet1, Florence Carrere1, Mathias Domostoj1, Valérie Cassan2, Pavan Randhawa 3, Roberta Valenti1

1Pierre Fabre Laboratories, Boulogne Billancourt, France, 2Pierre Fabre Laboratories, Toulouse, France, 3Pierre Fabre Laboratories, Reading, United Kingdom

Background: Post-Transplant Lymphoproliferative Disease (PTLD) is an aggressive, potentially fatal, lymphoproliferative disease that occurs after hematopoetic stem cell (HCT) or solid organ transplantation (SOT) in the context of extrinsic immunosuppression.

Tabelecleucel (Tab-cel) is the first off-the-shelf, allogeneic EBV-specific T-cell immunotherapy approved in the European Union for relapsed or refractory EBV+ PTLD following hematopoetic stem cell (HCT) or solid organ transplantation (SOT).

The European marketing authorization was granted under exceptional circumstances, based on the preliminary results of the ALLELE study a global, multicentre, open-label phase 3 trial (NCT03394365), demonstrating product efficacy with an objective response rate (ORR) of 50.0% (95% CI 23.0–77.0) and 51.7% (32.5–70.6) in HCT and SOT, respectively, and a favorable safety profile.

Based on the data emerging from this pivotal trial and to comply with regulatory obligations linked to the approval of tabelecleucel under exceptional circumstances, a post-approval safety study (PASS) is under start-up in Europe to ensure that data generation for tabelecleucel in patients with EBV + PTLD will continue in the real-world setting.

Methods: This study is an observational, multicenter, multinational post-authorisation safety study. All patients in Europe who are assigned to receive tabelecleucel in the real-world setting will be eligible to participate in the study.

The primary objective is to describe the safety of tabelecleucel in patients with EBV + PTLD following HCT or SOT in a real-world setting.

Secondary objectives include description of the effectiveness, the patient population, and treatment patterns, including dosing and schedule of tabelecleucel.

The analysis will be performed on all patients enrolled who receive at least one dose of tabelecleucel and by predefined subgroups including paediatric and elderly patients.

The follow-up period is 3 years from first dose for each subject registered in the study.

Results: Germany will be the first country participating in the study and contact with sites was initiated. The first annual report is expected by the end of 2024.

Conclusions: This PASS protocol will characterize the long-term safety and effectiveness of tabelecleucel in the overall EBV + PTLD patient population, and in pediatric and elderly subpopulations, in a real-world setting.

Disclosure: François Denjean, Asmaa Zkik, Constance Battin, Sophie Blanchet, Florence Carrere, Mathias Domostoj, Valérie Cassan, Pavan Randhawa and Roberta Valenti are Pierre Fabre employees.

The study is funded by Pierre Fabre Laboratories.

5: Cellular Therapies other than CARs

P148 CD45RO CELLS ADDBACK IN TCR ΑΒ/CD45RA DEPLETED HAPLOIDENTICAL HSCT LEADS TO SUPERIOR OUTCOMES IN INFANTS WITH INBORN ERRORS OF IMMUNITY: EXPERIENCE FROM A DEVELOPING COUNTRY!

Vimal Kumar Gunasekaran 1, Rishab Bharadwaj1, Meena Sivasankaran2, Niranjan Hegde2, Deepti Sachan1, Deenadayalan Munirathnam1

1Dr Rela Institute and Medical Centre, Chennai, India, 2Kanchi Kamakoti Childs Trust Hospital, Chennai, India

Background: Infusion of memory (CD45RO) T-cells post TCR-αβ/CD45RA depleted haploidentical HSCT could potentially augment immune reconstitution without increasing the risk of acute graft versus host disease (aGVHD), which is mainly induced by naïve (CD45RA) T-cells. We present our experience of the clinical profile, course, and outcomes of children with IEI who underwent TCR αβ/CD45RA depleted haploidentical HSCT with CD45RO addback.

Methods: This was a prospective observational study conducted over the last 2 years (2022-2023) at two tertiary care multi-specialty hospitals in Chennai, India. Eight children with IEI who underwent TCR-αβ/CD45RA depleted haploidentical HSCT were included. Pre-transplant lymphocyte subset profiles wherever relevant, and infections, were recorded. Myeloablative conditioning regimen (Thiotepa/Treosulphan/Fludarabine/Rabbit-ATG/Rituximab) was employed in all children. TCR-αβ/CD45RA depleted HSCs were infused on day ‘0’. On day +1 and then at regular intervals, escalating doses of CD45RO memory T-cells, were infused, as tolerated. Children were monitored for viral reactivations with weekly viral PCRs and GVHD. Lymphocyte subsets and donor chimerism levels were monitored at monthly intervals. Lymphocyte subset recovery was defined as absolute counts ≥200/microL for CD3 + CD4 + , CD3 + CD8+ and CD19+ cells and ≥ 150/microL for CD16 + CD56 + NK cells. Data was analyzed and inferences were drawn.

Results: Our cohort included 8 infants (two with SCID, two with CGD, one each with WAS, Hyper-IgM syndrome, Primary HLH and LAD). Median age of HSCT was 11 months (range 5.75-17.25 months). Three (37.5%) children had disseminated CMV infection, one had influenza and one had disseminated BCGiosis prior to HSCT. Two children with SCID had near zero lymphocytes and one with CGD had low normal lymphocyte profile. TCR-αβ depleted stem cells were infused on day 0 (median 17 million/kg; range15-19.25). All children received CD45RA negative CD45RO rich T-cell add-back (median 1 million/kg; range1-1.2). Subsequently, four children (50%) received add-back on day +15, one on day +30 and one on day +60. NK-cell recovery was noted with median time of 112.5 days (range 41-180), CD4+ and CD8+ cell recovery with median 150 days (range 90-180); B-cell recovery with median 164 days (range 95-210). Five (62.5%) children attained 100% donor chimerism with no evidence of GVHD; two (25%) had donor chimerism of 98.27% to 99.6%. CMV viremia was noted in 2 children (25%). one child (12.5%) had norovirus gastroenteritis. NO EBV or adenoviral reactivations were noted. Immune reconstitution was noted to be quicker and incidence of viremia lower compared to historical controls. One (12.5%) child had stage-1 gut GVHD (grade-II). Seven out of the 8 children are alive with good donor chimerism, free of infections and GVHD. One (12.5%) succumbed within one month of HSCT due to bacterial sepsis. The median follow up was 6 months.

Conclusions: Advances in haploidentical HSCT like TCR-αβ depletion, though have led to improved outcomes in infants with inborn errors of immunity (IEI), present with challenges like delayed immune reconstitution, viral reactivations, graft failure and GVHD. This approach of CD45RA negative CD45RO memory-T-cell add-back is feasible in developing countries, leading to quicker immune reconstitution, enhancing anti-viral immunity, without increasing the incidence of aGVHD.

Disclosure: Nothing to declare.

5: Cellular Therapies other than CARs

P149 IMPACT OF MYELOFIBROSIS ON PATIENTS WITH MYELODYSPLASTIC SYNDROMES FOLLOWING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Panpan Zhu 1,2,3,4, Xiaoyu Lai4,2,3,4, Lizhen Liu1,2,3,4, Jimin Shi1,2,3,4, Jian Yu1,2,3,4, Yanmin Zhao1,2,3,4, Luxin Yang1,2,3,4, Weiyan Zheng1,2,3,4, Jie Sun1,2,3,4, Wenjun Wu1,2,3,4, He Huang1,2,3,4, Yi Luo1,2,3,4

1The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Hangzhou, China, 3Institute of Hematology, Zhejiang University, Hangzhou, China, 4Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China

Background: Myelofibrosis (MF) is always considered as an adverse factor for patients diagnosed with myelodysplastic syndromes (MDS), whose treatment strategies are always based on various prognosis scoring systems. However, MF is not involved in prognosis scoring systems to guide therapy decision, and prognostic significance of MF grade in patients undergoing an allogeneic hematopoietic stem cell transplantation (allo-HSCT) is still elusive.

Methods: We retrospectively analyzed data from 153 patients with MDS who underwent allo-HSCT between March 2016 and December 2022 at Bone Marrow Transplantation, the First Affiliated Hospital, Zhejiang University School of Medicine. Patients progressed to acute myeloid leukemia (AML) prior to transplantation were excluded. The enrolled patients were divided the patients into MF-0/1 (N= 119) and MF-2/3 (N= 34) cohorts to explore the impact of MF on outcomes of all-HSCT.

Results: The median follow up was 23.1 (range: 0.1 – 91.3) months in the entire group. There were 89 males and 64 females, with a median age of 47 (range: 19 – 66) years. A total of 121 patients were diagnosed with MDS with increased blasts (MDS-IB), consisting of 57 with MDS-IB1/2 and 64 with MDS-fibrosis (Table). The 2-year rates of relapse, NRM, OS, and PFS were 10.9% (95% CI: 5.9 – 17.7%), 16.3% (95% CI: 10.2 – 23.6%), 76.6% (95% CI: 69.0 – 85.1%), and 72.8% (95% CI: 65.0 – 81.5%) in MF-0/1 cohort, and 16.9% (95% CI: 5.8 – 32.9%), 14.7% (95% CI: 5.3 – 28.7%), 71.8% (95% CI: 57.6 – 89.6%), and 68.4% (95% CI: 53.6 – 87.2%) in MF-2/3 cohort, respectively. No significant difference in outcomes of allo-HSCT was observed between two cohorts. Both univariate and multivariate analysis confirmed that MF-2/3 in patients with MDS had no effect on prognosis of transplantation. In addition, major/bidirectional ABO blood type between donors and recipients was an independent risk factor for (HR: 2.55, 95% CI: 1.25 – 5.21; P= 0.010) and PFS (HR: 2.21, 95% CI: 1.10 – 4.42; P= 0.025) in multivariate analysis. In the subgroup of patients diagnosed with MDS with increased blasts (MDS-IB), it was consistently demonstrated that clinical outcomes of MF-2/3 cohort were comparable to those with MF-0/1. The risk factors for OS and PFS in patients with MDS-IB were non-complete remission at transplantation and major/bidirectional ABO blood type.

Variables

MF-0/1

N= 119 (77.8%)

MF-2/3

N= 34 (22.2%)

P

Patient age, years

48 (19 – 66)

46 (23 – 64)

0.637

Patient sex, male / female

67 / 52

22 / 12

0.381

MDS subentities

 MDS-LB/h

26

6

NA

 MDS-IB1/2

57

 MDS-f

36

28

IPSS-R

 Low / Intermediate

34 (28.6%)

9 (26.5%)

0.810

 High / Very high

85 (71.4%)

25 (73.5%)

 Donor age, years

32 (12 – 55)

30 (15 – 59)

0.799

 Donor sex, male / female

78 / 41

23 / 11

0.820

Donor type

 HID

80 (67.2%)

22 (64.7%)

0.725

 MSD

21 (17.6%)

5 (14.7%)

 URD

18 (15.1%)

7 (20.6%)

Conditioning regimen

 RIC

35 (29.4%)

11 (32.4%)

0.742

 MAC

84 (70.6%)

23 (67.6%)

Conclusions: In conclusion, MF grade had no significant effect on prognosis of allo-HSCT in patients diagnosed with MDS. Major/bidirectional ABO blood type should be carefully considered in the context of more than one available donor.

Disclosure: No author has a potential conflict of interest or funding source.

5: Cellular Therapies other than CARs

P150 MANAGEMENT AND TOXICITY OF DONOR LYMPHOCYTE INFUSIONS IN PEDIATRIC PATIENTS WITH ONCOLOGICAL DISEASES AFTER ALLOGENIC STEM CELL TRANSPLANTATION IN A SINGLE INSTITUTION

Luisa Paschke 1, Marie Sophie Knape1, Katrin Vollmer1, Katja Gall1, Angela Wawer1, Jan-Henning Klusmann2, Dirk Reinhardt3, Hans-Jochem Kolb4, Belinda Simoes5, Stefan EG Burdach1, Irene Teichert von Luettichau1, Julia Hauer1, Uwe Thiel1

1Technical University of Munich, Munich, Germany, 2Goethe University Frankfurt, Frankfurt, Germany, 3University Duisburg-Essen, Essen, Germany, 4Ludwig-Maximilians-University, Munich, Germany, 5Ribeirão Preto Medical School, São Paulo, Brazil

Background: We retrospectively examine feasibility and toxicity of prophylactic and preemptive donor lymphocyte infusions (DLI) in pediatric patients after allogeneic stem cell transplantation for oncological diseases.

Methods: One female and five male patients with AML (n=5) and ALCL (n=1) were transplanted due to high risk disease (n=6) or due to relapse after initial allo-SCT (n=3) and treated with escalating DLI doses for prophylactic (n=6) and for pre-emptive (n=2) purposes after allogeneic stem cell transplantation (allo-SCT) at different remission status. Three out of six patients received one allo-SCT, two received two allo-SCT and one patient received three allo-SCTs in total. Initially patients received MSD (n=1) or MUD (n=2) as first allo-SCT. In case of relapse after initial allo-SCT, haploidentical grafts were further used in all respective patients. DLI were employed in prophylactic (n=6) or in pre-emptive intention (n=2). DLI were applied either as mono-treatment (n=2) or in combination with intrathecal prophylaxis (n=3) or in combination with intrathecal prophylaxis together with targeted treatment (n=2) or in combination with immune-modulatory salvage therapy with acacitidine/panobinostat (n=1). Premedication consisted of dimetindene. All DLI were initially given in complete remission (CR) and in all cases in the absence of GvHD. Application started after complete weaning of immune-suppression in a 6 weeks interval, respectively. Doses ranged from 1-15 doses (median=5 doses). A median dose of 5x106 CD3+/kg BW were administered with maximum dosage of 1x107CD3+/kg BW. The Median age at first allo-SCT and DLI was 7,5 years. For detailed patient characteristics refer to table 1.

Results: Median follow-up after first DLI per allo-SCT was 5.5 months. All DLI were well tolerated without any GvHD signs at last follow-up. No other side effects were reported. Disease relapse occurred in 1/6 patients after 10 months DLI discontinuation and in 2/6 patients (1 morphological and 1 molecular relapse) after first and in 1/6 after second DLI (1 molecular relapse). In the latter patient DLI was associated with CR. At last follow-up 5/6 patients are in CR.

Conclusions: This retrospective analysis indicates low DLI toxicity within the observation period even in increased doses both in HLA matched and in the haplo-setting. Long term follow up and larger study patient numbers are necessary to confirm these findings in order to develop standardized guidelines in the pediatric setting. As previously described, DLI may induce CR in pediatric patients as a salvage approach prior to re-allo-SCT. Retrospective matched pair analyses and prospective analyses may shed further light into the therapeutic role of DLI in children and adolescents with cancer.

Disclosure: The authors declare no conflict of interest. Uwe Thiel was funded by the Wilhelm Sander-Foundation (2018/07/21) and the Cura Placida Children’s Cancer Research Foundation (2021/08/23). Uwe Thiel is currently funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) —Project number 501830041, the “Zukunft Gesundheit e.V.”, the Dr. Sepp and Hanne Sturm Memorial Foundation and the Robert Pfleger Foundation.

29: Chronic Leukaemia and Other Myeloproliferative Disorders

P151 IMPROVING OUTCOMES IN MDS/MPN: TREOSULFAN-BASED CONDITIONING FOR ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Alessandro Bruno1, Lorenzo Lazzari 1, Elisa Diral1, Sara Mastaglio1, Daniela Clerici1, Sarah Marktel1, Francesca Lunghi1, Simona Piemontese1, Daniele Sannipoli1, Camilla Gariazzo1, Gianluca Scorpio1, Gregorio Bergonzi1, Consuelo Corti1, Matteo Giovanni Carrabba1, Massimo Bernardi1, Luca Vago1, Maria Teresa Lupo-Stanghellini1, Fabio Ciceri1,2, Jacopo Peccatori1, Raffaella Greco1

1IRCCS Ospedale San Raffaele, Milan, Italy, 2Università Vita-Salute San Raffaele, Milan, Italy

Background: MDS/MPN is a rare group of diseases mostly affecting elderly patients. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is nowadays the only potentially curative treatment for MDS/MPN, unfortunately with contradictory and disappointing results in terms of relapse and treatment related mortality (TRM). Moreover, transplant strategies across EBMT centers are highly heterogeneous and the best conditioning strategy is unknown in this difficult setting.

Methods: We conducted a single-center analysis on consecutive adult patients with MDS/MPN who underwent a first allo-HSCT for this indication at our institution between March 2009 and November 2022 and received a treosulfan-based conditioning. We collected data of 21 patients (13 CMML, 4 aCML, 3 MDS/MPN-U, 1 MDS/MPN-RS-T, according to WHO-2016). Conditioning regimen included treosulfan 14 g/m2 from day -6 to -4 and fludarabine 30 mg/m2 from day -6 to -2; two patients received a reduced dose of treosulfan of 10 g/m2 because of age at transplant ≥70 years. Conditioning was intensified with alkylating agents (melphalan, thiotepa) or radiotherapy (TBI 2-4 Gy or splenic irradiation in case of splenomegaly) in 10 patients. GvHD prophylaxis consisted in post-transplant cyclophosphamide, sirolimus and mycophenolate (MMF) in 15 patients, as our institution standard policy, while others received a combination of ATG based regimens with sirolimus or cyclosporine. Twenty patients received PBSC. Median numbers of infused CD34+ cells/Kg and CD3+ cells/Kg were 6.83 x 106 (range 2.10-10.87) and 2.34 x 108 (range 0.28-3.80), respectively.

Results: Median age at allo-HSCT was 61 years (range 35-71 y). Seventeen patients receive allo-HSCT in presence of active disease (15 upfront, 2 after failed induction treatment), while four patients were in first complete remission (CR1) after induction with hypomethylating agents (HMAs) or intensive chemoterapy. Comorbidity index was evaluable for 18 patients, 10 had a score ≥ 3. Two patients underwent allo-HSCT from a matched-related donor, 15 patients from a matched-unrelated donor and 4 patients from a haploidentical donor. Twenty patients (95%) engrafted. Median time to neutrophil ≥0.5x109/L was 22 days (range 14-59), to platelet ≥20x109/L was 32 days (range 13-188). Median follow-up was 28 months (range 1-141). Overall survival (OS), progression free survival (PFS) and GvHD/relapse free survival (GRFS) were 80.4%, 80.7% and 60.5% at 2-years. Relapse incidence was 9.8% at 2-years. Transplant related mortality (TRM) was 9.5% at 100 days and at last follow-up. Notably, no cases of veno occlusive disease were observed. The 100-day Cumulative Incidence (CI) of aGvHD grade 2-4 and 3-4 was 19.4% and 4.8%. CI of all grades cGvHD and moderate/severe cGvHD at 2 years was 30.1% and 15.3%. We found no significant differences in OS and PFS stratifying by diagnosis, disease status at transplant, conditioning intensity and donor type.

Conclusions: Treosulfan-based conditioning seems to be effective and well-tolerated in patients with MDS/MPN undergoing allo-HSCT, for both low risk of relapse and low TRM. Further studies on larger cohorts are necessary to confirm these findings and to clarify the role of pre-transplant induction treatment with HMAs or intensive chemotherapy in this disease setting.

Disclosure: Nothing to declare.

29: Chronic Leukaemia and Other Myeloproliferative Disorders

P152 PREVIOUS RESPONSE TO TKIS AS A FAVORABLE PROGNOSTIC FACTOR FOR SURVIVAL IN HSCT FOR CML: LONG TERM RESULTS OF A SINGLE CENTER IN BRAZIL

Vaneuza Funke 1, Giuliana Rosendo1, Daniela Setubal1, Caroline Bonamin Sola1, Samir Nabhan1, Rafael Marchesine1, Glaucia Tagliari1, Isabela Menezes1, Ana Lucia Mion1, Ricardo Pasquini1

1Federal University of Parana, Curitiba, Brazil

Background: Tyrosine kinase inhibitors (TKIs) have revolutionized therapy for patients with Chronic Myeloid Leukemia (CML) over the last two decades. However, some patients still do not achieve an adequate response to those drugs, and hematopoietic stem cell transplantation (HSCT) is indicated in this scenario. Furthermore, considering regions with difficult access to third generation TKIs, HSCT can be a feasible and cost-effective option.

Methods: We analyzed 70 patients with the diagnosis of CML resistant to TKIs, transplanted at a single HSCT Center in Brazil, from January 2001 to May 2021. The study is retrospective, held from data record in either HSCT database or medical chart. The study primary endpoint is to determine overall survival for CML patients who received HSCT after TKI failure. As secondary objectives we evaluated risk factors for OS in this cohort and estimated relapse-free survival (RFS), graft-versus-host disease-free/relapse-free survival (GRFS) and the incidences of relapse (RI), non-relapse mortality (NRM), acute and chronic GVHD. Survival curves were performed using Kaplan Meier Method and Fisher exact test was used for categoric variables. Multivariate analysis was performed using Cox proportional hazards models. Endpoints with competing risks were evaluated by Gray’s test. EZR was used for statistical analysis.

Results: Median age was 38 years (10-60) and 66% were male. Twelve patients were transplanted in first chronic phase (CP1), 17 in CP >1, 38 in accelerated phase (AP) and 3 patients in blast crisis (BC). Nineteen patients (27 %) had mutations identified. Thirty-three patients received HSCT from unrelated and 37 from related donors. Marrow or cord blood were used as graft source for 37 patients and 33 remaining received peripheral blood. Median survival for the whole cohort was 11 years; being 1-year OS: 70% (57,8-79,3), 5-year OS: 57,7 % (45,1-68,5) and 10-year OS: 51,4% (38,3-63,1). Five years estimated overall survival was no different for CP1 (60%) versus for advanced phases (45%); p=0,6. In the multivariate analysis, OS was significantly dependent on degree of response prior to HSCT [HR 5.89 (1.19- 29.16); p= 0.03] for patients without hematological response compared to patients with cytogenetic or molecular response. Cumulative incidence of non-relapse mortality (NRM) at 100-days: 12,9% (6,3-21,9) and at 1-year: 21,4 % (12,7-31,7). Cumulative incidence of acute GVHD (aGVHD) at 100-days: 34,3% (23,4-45,5) and of cGVHD at 1-year: 34,3% (23,3-45,5). Cumulative incidence of cGVHD at 2-years: 35,7% (24,6-47,0). At one year, cumulative incidence of relapse was 27,1 % (17,3-38) and at two years 34,3 (23,4 - 45,5).

Conclusions: The study corroborates the potential of HSCT in the scenario of therapeutic failure or reduced access to drugs, with a 5 year OS of about 60%. Patients who received HSCT in cytogenetic or molecular response had significant better OS, suggesting that kind of response as a potential target to be achieved before transplantation.

Disclosure: No disclosures.

29: Chronic Leukaemia and Other Myeloproliferative Disorders

P153 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT FOR MYELOFIBROSIS: UPDATED RESULTS FROM BRAZILIAN REGISTRY

Vaneuza Funke 1, Alberto Cardoso Martins Lima1, Nelson Hamersclak2, Vergilio Colturato3, Afonso Vigorito4, Gustavo Machado Teixeira5, Vanderson Rocha6, Livia Mariano6, Decio Lerner7, George Mauricio Navarro Barros8, Alessandra Paz9, Celso Arrais10, Claudia Astigarra11, Fabio Pires12, Fernando Barroso Duarte13, Ricardo Pasquini1, Mary Flowers14

1Federal University of Parana, Curitiba, Brazil, 2Hospital Israelita Albert Einstein, Sao Paulo, Brazil, 3Hospital Amaral Carvalho, Jau, Brazil, 4Universidade de Campinas, Campinas, Brazil, 5Federal University of Minas Gerais, Belo Horizonte, Brazil, 6Universidade de Sao Paulo, Sao Paulo, Brazil, 7Instituto Nacional do Cancer (INCA), Rio de Janeiro, Brazil, 8Hospital de Amor, Barretos, Brazil, 9Federal University of Rio Grande do Sul, Porto ALegre, Brazil, 10Federal University of Sao Paulo (UNIFESP), Sao Paulo, Brazil, 11Hospital Moinhos de Vento, Porto ALegre, Brazil, 12Hospital Beneficiencia Portuguesa, Sao Paulo, Brazil, 13Federal University of Ceara, Fortaleza, Brazil, 14Fred Hutchinson Cancer Center, Seattle, United States

Background: Allogeneic hematopoietic cell transplantation remains the only curative treatment for Myelofibrosis. However, advanced age of patients and the significant TRM limit the applicability of this procedure in clinical practice. Considerable research has been conducted to identify baseline risk factors that might predict transplantation results. Patient age, performance status, comorbidities, disease risk index, donor type, and HLA compatibility are established risk factors for allo-HCT outcome. However, there is no data of HSCT for this disease in regions with limited access to resources as Latin America. These data can be helpful in clinical practice to assess the risk-benefit ratio of transplantation in potential candidates, as well as provide benchmark for future prospective studies also in these countries.

Objectives: Primary objective: evaluate overall survival of a Brazilian cohort of patients who received HSCT for the diagnosis of MP or secondary to TE and PV. Secondary objectives: estimate non relapse mortality, relapse or rejection, and cumulative incidence of acute and chronic GVHD and identifying risk factors for OS, and NRM.

Methods: We analyzed retrospectively charts and database from 158 patients with the diagnosis of MF who received allo-SCT at 15 centers in Brazil from 1997 to 2020. Statistical analysis by EZR software. Bivariate analysis and estimation of OS were performed using Kaplan Meier method and Log Rank test for curve comparison. Cox Survival Model was used for multivariate analysis of risk factors for Overall Survival and Multivariable Fine-Gray competing risks regression model was used for non-relapse mortality at one year. P level of significance was <0,05. Characteristics of patients are showed in table 1.

Results: Overall Survival (OS) was 64% at one year, 52% at 3 years and 49% at 5 years One- cumulative incidence of relapse or rejection was 12 %. Cumulative incidence of non-relapse mortality (competitive risk) at one year was 30%. Cumulative incidence of acute GVHD grade II or IV at 100 days was 16% and of chronic GVHD at 2 years was 32%. At multivariate analysis by Cox Model, CD 34 < 5 x 106 HR 1.94 (1.15- 3.28; p= 0.0013), relapse or rejection HR 4.19 (2.38-7.36; p= 0.00000064, DIPSS intermediate 2 or high-risk HR 2,17 (1,16 - 4.05; p= 0.0016) and grade III-IV acute GVHD HR 2,39 (1.45-3.94; p=0,00064) were significantly associated with inferior survival. At Multivariable Fine-Gray competing risks regression model for non-relapse mortality, only Donor type HR 2,02(1,1 - 3.71; p= 0.023) was identified as independent risk factor.

Conclusions: At multivariate analysis CD34 dose < 5 x 106, relapse or rejection, DIPSS High risk or Intermediate II and acute GVHD grade III-IV were risk factors for survival while Donor type was a risk factor for non-relapse mortality. Major limitation of this study is the retrospective nature which lead to missing data, however, it still can provide a benchmark for future prospective studies.

Disclosure: No disclosures

29: Chronic Leukaemia and Other Myeloproliferative Disorders

P154 RESULTS OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH CHRONIC MYELOID LEUKEMIA AFTER THERAPY WITH TYROSINE KINASE INHIBITORS

Larisa Kuzmina1, Vera Vasilyeva 1, Zoya Konova1, Marya Dovydenko1, Olga Koroleva1, Olga Pokrovskaya1, Darya Mironova1, Elena Parovichnikova1

1National Medical Research Center for Hematology, Moscow, Russian Federation

Background: Tyrosine kinase inhibitors (TKIs) are used as first-line therapy in patients with chronic myeloid leukemia (CML). Allogeneic hematopoietic cell transplantation (allo-HSCT) is treatment option of the for patients in the second chronic phase of the disease or acceleration phase, or who resistant to two or more lines of TKI.

Methods: We analyzed the results of treatment of 33 patients (22 males/ 11 females) with CML who underwent allo-HSCT from February 2012 to August 2023. Indications for allo-HSCT were: ineffectiveness (n=9) or intolerance to TKIs (n=6) in 15 patients, accelerated phase (AP) or blast crisis in anamnesis (BC) in 18 patients (onset in 8 patients, during the treatment in 10). The T315I mutation was detected in 7 of 33 patients. All patients with BC received TKIs in combination with cytostatic therapy, and the chronic phase (CP) was achieved. The median age of all patients was 41 years (20-59). The median of TKI lines before allo-HSCT was 2 (1-7), 3 or more TKI lines were received by 16 patients. The median time from the diagnosis to allo-HSCT was 21 months (4-131 months).

Results: When assessing the overall (OS) and disease-free survival (DFS) of patients with CML after allo-HSCT, similar results were obtained in both groups (Figure 1a, Figure 1b). However, there was a high mortality associated with HSCT 26.7% in the TKI ineffectiveness/intolerance group versus 5.5% in the AP/BC group. The most frequent reason of death in the AP/BC group was disease relapses 5 (27.7%) versus in TKI ineffectiveness/intolerance group no one died due to relapse (Table No. 1). We also checked a chimeric BCR/ABL transcript in the early stages after allo-HSCT, more than 50% of patient had it. But in the AP/BC group were detected more relapses of disease 8 (44,4%) versus ineffectiveness/intolerance group only one (6,7%).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (21)

Table No. 1. Events after allo-HSCT

TKI ineffectiveness/intolerance (n=15)

AP/BC (n=18)

р

Failure/rejection

4 (26,7%)

5 (27,7%)

1,0

Death after allo-HSCT

5 (33,3%)

7 (38,9%)

1,0

Transplant related mortality

4 (26,7%)

1 (5,5%)

0,152

Disease related mortality

5 (27,7%)

0,0488

Received TKIs after allo-HSCT

6 (40%)

17 (94,4%)

0,0015

aGVHD

3 (20%)

3 (16,7%)

1,0

chGVHD

6 (40%)

9 (50%)

0,729

Detection of a chimeric BCR/ABL transcript of more than 0.01% in the early stages after allo-HSCT

8 (53,3%)

10 (55,5%)

1,0

Conclusions: When assessing the effectiveness of allo-HSCT in the ineffectiveness/intolerance group, a decrease in OS and DFS is associated with high transplant related mortality (26.7%), and in the AP/BC group - due to the disease relapse. Detection of a chimeric BCR/ABL transcript of more than 0.01% in the early stages after allo-HSCT in the AP/BC group may be a predictor of the development of disease relapse.

Disclosure: no

29: Chronic Leukaemia and Other Myeloproliferative Disorders

P155 ALLOGENEIC STEM CELL TRANSPLANTATION IN CHRONIC MYELOID LEUKEMIA IN ERA OF TYROSINE KINASE INHIBITORS: A 13-YEAR SINGLE CENTRE EXPERIENCE FROM INDIA

Mehak Trikha1, Lingaraj Nayak 1, Sachin Punatar1, Anant Gokarn1, Akanksha Chichra1, Sumeet Mirgh1, Nishant Jindal1, P.G. Subramaniam1, Nikhil Patkar1, Prashant Tembhare1, Gourav Chatterjee1, Sweta Rajpal1, Navin Khattry1

1Tata Memorial Hospital, Mumbai, India

Background: Allogeneic transplant remains highly effective treatment option for individuals with tyrosine kinase inhibitor(TKI)-resistant or advanced chronic myeloid leukemia(CML), especially in resource limited settings where third generation TKI are not easily available. This retrospective analysis investigates outcomes of CML patients undergoing allo-transplantation in era of TKI from our center.

Methods: This study included all patients who received allogeneic transplant for CML between August 2010 and March 2023. Conditioning regimens used included Fludarabine-Melphalan, Busulphan-Cyclophosphamide, Fludarabine-4-8Gy TBI, or Fludarabine-Treosulfan-2Gy TBI. Graft-versus-host disease(GVHD) prophylaxis for matched sibling(MSD) or unrelated donor(MUD) transplants involved cyclosporine(CSA) with methotrexate or mycophenolate(MMF). For haploidentical transplant(HIT) patients, post-transplant cyclophosphamide(PTCy) along with CSA/tacrolimus and MMF was used. Peripheral-blood quantitative PCR for BCR-ABL and chimerism (VNTR for all patients, and FISH XX/XY for gender mismatched transplants) were done monthly for 1 year following transplant. Prophylactic TKIs were not used post-transplant except in patients with blast crisis. Primary endpoint was Relapse-free-survival(RFS) which was assessed from day of transplant to loss of Major Molecular Response(MMR) (RQPCR >0.1% BCR-ABL), death or last follow-up. Deep Molecular Response(DMR) was defined as RQPCR <0.01%. Molecular relapse was defined as loss of MMR in 2 consecutive peripheral-blood RQPCR at least 2 weeks apart. Overall survival(OS) was calculated as per standard definition. Possible prognostic factors analyzed were phase of CML at diagnosis, type of transplant, Disease Risk Index(DRI), HCT-CI, EBMT scores, duration of TKI pre-transplant, female to male transplant and acute/chronic GVHD. Univariate and multivariate analysis for RFS and OS were done using standard statistical methods.

Results: A total of 73 CML patients were transplanted during this period with median age of 31 years (range: 9-56), with 75% being male. Demographic details, disease parameters and transplant details are shown in table 1. TKI failure was the primary reason for transplant(75%) and Blast Crisis(BC) in 25%. T315I mutation was present in 19% of patients. Median time from diagnosis to transplant was 36 months. Median time to neutrophil and platelet engraftment were 16 days and 13 days respectively. Acute and chronic GVHD rates were 42% and 43%, respectively. Acute GVHD of grade 2, Grade 3 and Grade 4 were seen in 15(20.5%),5 (6.8%),1(1.4%) respectively. Ten patients received donor lymphocyte infusions(DLI), of whom 5 are long-term survivors. At Day 100, 59% achieved DMR, 12% -MMR and 15% less than MMR. At 1-year, 36% were in DMR, 19% -MMR, 15% had molecular relapse, 4% had undetermined status and 26% were deceased. Median follow-up was 84 months, and non-relapse-mortality(NRM) was 28% for the whole group with NRM in MSD being 23% and in HIT being50%. Seven-year OS and RFS were 55% and 47%, respectively. On multivariate analysis, use of TBI was associated with worse RFS (7year RFS 60% vs. 17%, non-TBI vs. TBI, p-0.043) and chronic GVHD (7year RFS of 55% vs 36%, p-0.005) was associated with better RFS.

Characteristics

All patients (N=73)

Gender

Male

54 (73%)

Female

19 (27%)

Median age at transplant(years)

31 years (range- 9-56 years)

Disease phase at diagnosis

CP

56 (76%)

AP

9 (12.3%)

BC

18 (24.6%)

Status at transplant

CP1

26 (35%)

CP2

26 (35%)

CP3

14 (19%)

Persistent disease

5 (6.8%)

TKI received

Imatinib

58 (80%)

Dasatinib

13 (17.85)

Bosutinib

1 (1.3%)

Nilotinib

1(1.3%)

Ponatinib

2(2.6%)

BCR-ABL KD mutation analysis

T315I

14 (19.1%)

Other than T315I

26 (35.6%)

Negative

18 (24.6%)

Not Done

15 (20.5%)

EBMT score

>3

30 (41%)

< 3

43 (59%)

DRI index

High

12 (16.4%)

Low

57 (78%)

intermediate

4 (5.4%)

Type of transplant

MSD

56(76.7%)

Haplo-transplant

15 (20.5%)

MUD

2 (2.7%)

Donor gender mismatch

Female to male

22 (30.1%)

Other combinations

51 (70%)

Source of stem cell

PBSC graft

72 (98.6%)

BM graft

1 (0.4%)

Conditioning regimen

Flu-Mel

42 (57.5%)

Bu-Cy

4 (5.4%)

Flu-TBI(7.2Gy and more)

8 (10.9%)

Flu-treo-TBI(2Gy)

5 (6.8%)

Flu-TBI(less than 7.2Gy)

13 (17.8%)

FTT

1 (1.3%)

GVHD prophylaxis

CsA+Methotrexate

44 (60.2%)

CsA+MMF

14 (19.1%)

CsA+MMF+PTCy

7 (9.5%)

TAc+MMF+PTCy

8 (11%)

Acute GVHD

Yes

41 (56%)

No

32 (44%)

Chronic GVHD

Yes

43 (59%)

No

30 (41%)

Slippage of chimerism

Yes

25 (34%)

No

48 (66%)

DLI given

Yes

10 (13.7%)

No

63 (86.3%)

Conclusions: Transplantation demonstrates promising long-term survival for TKI-resistant and advanced CML. Compared to TBI, Alkylating agent based conditioning regimen was associated with better relapse free and overall survival.

Disclosure: NOTHING TO DECLARE.

16: Conditioning Regimens

P156 ATG-TARGETED DOSING STRATEGY REDUCED CMV/EBV REACTIVATION AND IMPROVED SURVIVAL WITHOUT INCREASING GVHD AFTER URD-PBSCT: A SINGLE-CENTER, PROSPECTIVE, SINGLE-ARM, CLINICAL TRIAL

Sheng Chen 1, Jishan Du1, Haitao Wang2, Nan Wang1, Liping Dou2, Daihong Liu2

1Medical School of Chinese PLA, Beijing, China, 2The Fifth Medical Center of Chinese PLA General Hospital, Beijing, China

Background: Anti-thymocyte globulin (ATG) is widely used in allogeneic hematopoietic stem cell transplantation to prevent severe graft-versus-host disease (GVHD) and graft failure. However, overexposure to ATG may increase cytomegalovirus (CMV), Epstein-Barr virus (EBV) reactivation, non-relapse mortality, and disease recurrence. We established a targeted dosing strategy based on ATG concentration monitoring and conducted a phase 2 trial to evaluate the safety and efficacyof the doseing strategy in adult unmanipulated haplo-PBSCT, discovered the ATG-targeted dosing strategy reduced CMV/EBV reactivation andimproved survival without increasing GVHD after haplo-PBSCT. In this trial, we extended the ATG-targeted dosing strategy to adult unrelated donor allogeneic hematopoietic stem cell transplantation, a similar, encouraging result is attained.

Methods: ATG was administered for 4 days (-5 days to-2 days) during conditioning. The ATG doses on-3 days and -2 days were adjusted by our dosing strategy to achieve the optimal ATG exposure. The primary endpoint was CMV reactivation on +180 days. Between December 2020 and January 2023, 30 URD-PBSCT patients were enrolled and 38 URD-PBSCT patients who received the conventional fixed ATG dose (cumulative 10 mg/kg) comprised historical control. The transplantation protocols in historical cohort were the same as that in the trial including conditioning regimens, GVHD prophylaxis, and support therapy.

Results: Compared to the historical control cohort, the cumulative incidences of CMV reactivation and persistent CMV antigenemia on +180 days for the trial cohort were significantly decreased (CMV reactivation: 81% vs. 30%, p < .001) and No statistical significance was noted in the cumulative incidences of EBV reactivationcumulative (73%vs65, p=.46) and the incidences of grade II–IV (36%vs43%, p= .0.78) on +100 days. No significant difference was observed in the 1-year CIR 18.1% vs 20.6%, p= .96) between the two cohorts. Overall, the comparison of the 1-year DFS (72.3%vs22.1%, p=.034) between the two cohorts revealed a significant difference in survival.

Conclusions: ATG-targeted dosing strategy reduced CMV reactivation and improved survival without increasing GVHD after URD-PBSCT.

Clinical Trial Registry: The trial was registered at www.clinicaltrials.gov as NCT04778618.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P157 IMPACT OF FLUDARABINE DOSE ON OUTCOME AFTER ALLO-HCT WITH REDUCED INTENSITY CONDITIONING FOR OLDER PATIENTS WITH AML: A STUDY FROM THE ALWP OF THE EBMT

Guillaume Dachy 1, Myriam Labopin2, Gerard Socié3, Cristina Castilla-Llorente4, Edouard Forcade5, Igor Wolfgang Blau6, Patrice Ceballos7, Eric Deconinck8, David Burns9, Claude Eric Bulabois10, Radovan Vrhovac11, Anne Huynh12, Didier Blaise13, Johan Maertens14, Thomas Schroeder15, Jacques-Olivier Bay16, Bipin Savani17, Alexandros Spyridonidis18, Fabio Ciceri19, Mohamad Mohty20

1Uclouvain, Woluwe-Saint-Lambert, Belgium, 2Université Pierre et Marie Curie, Paris, France, Paris, France, 3Saint-Louis Hospital, Paris, France, 4Gustave Roussy Cancer Campus, Villejuif, France, 5CHU Bordeaux, Pessac, France, 6Charité Universitätsmedizin, Berlin, Germany, 7CHU Montpellier, Montpellier, France, 8Hopital Jean Minjoz, Besancon, France, 9Birmingham Centre for Cellular Therapy and Transplant, Birmingham, United Kingdom, 10CHU Grenoble Alpes, Grenoble, France, 11University Hospital Center Zagreb, Zagreb, Croatia, 12Institut Universitaire du Cancer de Toulouse – Oncopole, Toulouse, France, 13Institut Paoli Calmette, Marseille, France, 14University Hospital Gasthuisberg, Leuven, Belgium, 15University Hospital Essen, Essen, Germany, 16CHU ESTAING, Clermont, France, 17Vanderbilt University Medical Center, Nashville, United States, 18University Hospital of Patras, Patras, Greece, 19Ospedale San Raffaele, Milan, Italy, 20Hôpital Saint-Antoine, Paris, France

Background: Although conditioning regimens have been extensively studied over the past 15 years, rigorous evaluation of their pharmacokinetic profiles has only recently been investigated. Fludarabine exposure showed a wide variability among patients in most pharmacokinetic studies, with a significant impact on outcomes after allogeneic hematopoietic stem cell transplantation (allo-HCT). An optimal fludarabine exposure might be hypothesized in terms of non-relapse mortality (NRM), graft failure and disease control. This was confirmed by recent studies analyzing chimeric antigen receptor (CAR) T-cell therapy for acute leukemias, showing that optimal fludarabine exposure was associated with better leukemia-free survival (LFS), improved B cell reconstitution and higher expansion of the CAR T-cells. We therefore aimed to study the impact of fludarabine dosage on outcomes of a homogeneous acute myeloid leukemia (AML) cohort after allo-HCT.

Methods: We performed a retrospective study based on the European Society of Blood and Marrow Transplantation (EBMT) registry, including all patients aged over 50 years, with AML in first complete remission (CR1) receiving an allo-HCT between 2010 and 2022 with peripheral blood stem cells after reduced intensity conditioning based on fludarabine and busulfan (6.4mg/kg IV or 8mg/kg oral), from a matched sibling or unrelated donor. Graft-versus-host disease (GVHD) prophylaxis included anti-thymocyte globulin. Haploidentical donor grafts and post-transplant cyclophosphamide were excluded, as well as patients with chronic kidney disease.

Results: 1907 patients met the inclusion’s criteria, with a median age of 63 years old (interquartile range [IQR] 58.7-67.1) and a median follow-up of 32 months (IQR 29.6-35.8). Secondary AML was diagnosed in 21%. Most patients had a low comorbidity score (hematopoietic cell transformation-specific comorbidity index [HCT-CI] <3: 73.2%) and received a transplant from a matched unrelated donor (≥ 9/10: 63.2%). Univariate and multi-variate analyses stratified according to fludarabine dose, i.e., 110-130 mg/m², 140-150 mg/m², 150-160 mg/m² and 170-190 mg/m² showed association of all dose groups superior to 130 mg/m² with a lower LFS. We therefore stratified patients according to fludarabine dose (≤130 mg/m² or >130 mg/m2). Univariate analyses showed lower NRM (8.7% vs. 15.9%, p= 0.09) and higher LFS (62.9% vs. 53.2%, p= 0.004) resulting in better GVHD-free/relapse-free survival (GRFS) (58.6% vs. 43.4%, p= 0.001) and overall survival (66.7 vs. 60.6%, p=0.019) for patients exposed to lower doses of fludarabine (≤130 mg/m²). These observations were robust and confirmed in a multivariate analysis containing the following variables: fludarabine dose, patient age, year of allo-HCT, secondary AML, adverse cytogenetic risk, type of donor, performance index, comorbidity score (0, <3 or ≥3), female donor to male recipient combination, cytomegalovirus status. Fludarabine doses ≤ 130 mg/m² were associated with higher rates of LFS (hazard ratio [HR]: 1.46, p=0.011) and GRFS (HR: 1.5, p=0.006).

Conclusions: In an elderly population diagnosed with AML in CR1 and receiving a homogeneous conditioning regimen with fludarabine and busulfan before allo-HCT, our findings suggest an impact of fludarabine exposure on outcomes after allo-HCT. Lower doses (≤130 mg/m²) were associated with better LFS and GRFS, underscoring the importance of considering fludarabine dosing in the context of reduced intensity conditioning for allo-HCT in this patient population.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P158 TOXICITY COMPARISON OF TWO REDUCED INTENSITY CONDITIONING (RIC) TRANSPLANT REGIMENS FOR ADULT ALL IN CR1/2: RESULTS FROM THE RANDOMISED PHASE 2 MULTICENTRE ALL-RIC TRIAL

Anna Castleton 1, Rebekah Weston2, Aimee Jackson2, Matthew Beasley3, Rebecca Bishop2, Scott Chapman2, Patricia Diez4, Mohamed Elhaneid2, Adele Fielding5, Andrea Hodgkinson2, Stephanie Lane2, George Mikhaeel6, Nick Morley7, Eduardo Olavarria8, Eleni Tholouli9, Bela Patel10, Ronjon Chakraverty11, David Marks12

1The Christie NHS Foundation Trust, Manchester, United Kingdom, 2Cancer Research UK Clinical Trials Unit, University of Birmingham, Birmingham, United Kingdom, 3Molecular Radiotherapy Unit, Bristol Cancer Institute, Bristol, United Kingdom, 4Mount Vernon Cancer Centre, Northwood, London, United Kingdom, 5University of York, York, United Kingdom, 6St Thomas’ Hospital, London, United Kingdom, 7Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom, 8Imperial College Healthcare NHS Trust, London, United Kingdom, 9Manchester University NHS Foundation Trust, Manchester, United Kingdom, 10Barts Cancer Institute, Queen Mary University of London, London, United Kingdom, 11Oxford Cancer & Haematology Centre, Oxford, United Kingdom, 12Bristol Haematology and Oncology Centre, Bristol, United Kingdom

Background: ALL-RIC is a randomised trial comparing two RIC regimens for adults >40 years with acute lymphoblastic leukaemia (ALL), undergoing sibling or unrelated donor allogeneic stem cell transplant (alloSCT) in CR1/CR2. Standard RIC conditioning (fludarabine, melphalan and alemtuzumab - UKALL14) was compared to cyclophosphamide (50mg/kg x 2 days), total body irradiation (TBI; 8Gy in 4 fractions over 2 days) and alemtuzumab. We hypothesised that a low-dose TBI-based regimen would reduce relapse and improve EFS without significantly more toxicity or TRM. Here we compare toxicity and short-term outcomes in year 1 post-transplant.

Methods: One hundred and two patients were randomised, 89 were transplanted receiving the assigned conditioning regimen (control n=45, experimental n=44). Populations were compared for time to engraftment, total SAEs (including cardiac and pulmonary events), year 1 hospitalisations, rates of infection, and acute graft-versus-host-disease (aGvHD). In addition, we evaluated pre- and 12-month post-alloSCT pulmonary function tests (PFTs) - a 20% decline in FEV1 or TLCO (corrected for Hb) was considered clinically significant.

Results: Pre-alloSCT characteristics were comparable (table 1). Neutrophil and platelet engraftment was achieved in 84 out of 87 patients, with 3 out of 87 failing to engraft (n=2 control; n=1 experimental). No difference in time to neutrophil engraftment was evident between arms (p=0.433), whereas time to platelet engraftment was longer in controls (11 vs. 10 days, p=0.007).

Per patient, mean number of SAEs in control and experimental arms was comparable (1.87 vs 2.17, p=0.189), with no difference in grade 3+ AEs (n=32 control; n=25 experimental; p=0.330). Cardiac and pulmonary SAEs occurred in n=2 and n=9 patients respectively, with a total of 6 events (n=2 cardiac; n=4 pulmonary) in the control arm, 8 events (n=1 cardiac; n=7 pulmonary) in the experimental arm, and no difference in grade 3+ cardiac or pulmonary events (n=2 control; n=2 experimental).

Median total number of days hospitalisation were comparable between arms (control n=28 (18-401); experimental n=28 (20-87)), with equivalent transplant admission duration and hospital re-admission rates pre- and post-D100.

Infections were more common in the experimental arm (1.14 vs 1.38, p=0.008) but grade 5 infections did not differ (n=1 control; n=1 experimental). Rates of grade 1-4 aGvHD were equivalent, with minimal grade 3-4 aGvHD reported.

Sixty-six patients (74.2%) survived to 12 months; 27/66 (40.9%) had evaluable paired baseline and 12-month PFTs (11 control; 16 experimental). Four patients had clinically significant (≥20%) reductions in FEV1 or TLCOcorr; 3 were in the experimental arm.

Table 1:

Conditioning

Standard (n=45)

Experimental (n=44)

p-value

Median age, yrs (range)

54.3 (33.6-72.4)

55.6 (33.8-69.9)

Gender, male (%)

20 (44.4)

26 (59.1)

Remission status pre-transplant (%)

CR1

41 (91.1)

41 (93.2)

CR2

4(8.9)

3 (6.7)

Donor type (%)

Sibling

14 (31.1)

15 (34.1)

Unrelated

31 (68.9)

30 (66.7)

B-cell disease (%)

32 (71.1)

36 (81.8)

PFTs at baseline

FEV1<70%

1

5

TLCOcorr <70%

10

8

ANC >0.5x109/L not reached (%)

2 (4.4)

1 (2.3)

Median time to ANC >0.5x109/L, days

13

13

0.433

Platelets >20x109/L not reached (%)

1 (2.2)

0 (0)

Median time to platelets >20x109/L, days

11

10

0.007

Total SAEs (mean)

1.87

2.17

0.189

Infections (mean)

1.14

1.38

0.008

Median yr 1 hospitalisation days (range)

28 (18-401)

28 (20-87)

0.600

Acute GvHD*

Grade 1

5

12

Grade 2

2

4

Grade 3

1

0.297

  1. *2 patients in experimental arm had aGvHD with grade unknown

Conclusions: This is the first randomised trial comparing 2 RIC regimens for sibling or unrelated donor alloSCT in adult ALL. Cyclophosphamide/8Gy TBI conditioning is well tolerated in adults >40 years of age. It is deliverable with manageable increases in year 1 infectious toxicity, no increase in early cardiac or pulmonary toxicity, comparable rates of aGvHD, and no increase in time spent in hospital in the first 100 or 365 days. Additional PFT data from pre- and 1-year post-alloSCT are required to further evaluate clinically relevant differences between the two arms. Major outcome measures (EFS, OS, relapse) will be compared at 3-years median follow-up.

Clinical Trial Registry: EudraCT Number: 2017-004800-23.ISRCTN99927695

Disclosure: Nothing to declare.

16: Conditioning Regimens

P159 CXCR4 DIRECTED ENDORADIOTHERAPY WITH [177LU] PENTIXATHER ADDED TO TBI IS FEASIBLE AND SPARES THE HEMATOPOIETIC NICHE IN PATIENTS WITH RELAPSED/REFRACTORY ACUTE MYELOID LEUKEMIA

Krischan Braitsch 1, Martina Rudelius2, Maike Hefter1, Katrin Koch1, Katharina Nickel1, Katharina S. Götze1, Wolfgang Weber1, Florian Bassermann1, Matthias Eiber1, Mareike Verbeek1, Peter Herhaus1

1Technical University of Munich, Munich, Germany, 2Ludwig Maximilian University of Munich, Munich, Germany

Background: Therapy for acute myeloid leukemia (AML) has rapidly evolved but allogeneic hematopoietic stem cell transplantation (alloHSCT) remains standard for fit patients with high-risk disease. Myeloablative total body irradiation (TBI) based conditioning can be used in relapsed/refractory (r/r) AML patients considered refractory to chemotherapy, but the optimal conditioning regimen remains unclear. C-X-C-motif chemokine receptor 4 (CXCR4) is abundantly expressed in the hematopoietic system and regulates pivotal processes in hematopoiesis, immune response and stem cell homing. CXCR4 is commonly overexpressed in various malignancies, therefore radiolabeled peptides targeting CXCR4 like [68Ga] Pentixafor for diagnostic imaging and its therapeutic counterpart [177Lu] Pentixather were developed and proved efficacious in AML patients. Hematological toxicity is a major concern of CXCR4-directed ERT, as there is a significant “cross-fire” effect that targets the bone marrow (BM) niche. We have previously reported the feasibility of integrating CXCR4-directed ERT to TBI-based conditioning for r/r AML patients. Here, we report an update on seven patients treated with [177Lu]- Pentixather combined with TBI and chemotherapy prior to allo-HSCT with a focus on toxicity, engraftment, the impact on the BM niche and efficacy.

Methods: Seven patients with r/r AML were treated at our center between February 2019 and October 2023. In vivo CXCR4 expression on leukemic blasts was confirmed in all patients by [68Ga]-Pentixafor PET-imaging. ERT with [177Lu]-Pentixather was offered as compasionate use and all patients gave written informed consent prior to treatment. Pretherapeutic dosimetry was performed to obtain absorbed doses of the kidneys (organ at risk) for treatment tailoring. Conditioning consisted of [177Lu]-Pentixather ERT on d-15, TBI (8-10 Gy) on d-9 to d-7 and chemotherapy.

Results: Median patient age was 46 (42-57) years and they had previously received 4 (3-7) lines of intensive therapy, including allo-HSCT in four patients. Two patients were primary refractory and five had relapsed AML. At the start of conditioning, five had severe neutropenia with an ANC count of <500/µl. Baseline creatinine was 0.8 mg/dl (0.4-1.1) and no tumor lysis syndrome was observed. An increase in serum creatinine of ≥ 1.5 times occurred in three patients, which was attributed to infectious complications. Of those, two patients required hemodialysis during ICU stay. All patients developed fever and received broad- spectrum antibiotic and antifungal treatment during hospital stay. Four patients were transferred to the ICU of which 2 required invasive ventilation. Hepatotoxicity was common and occurred in all patients. Baseline bilirubin levels were normal in all patients (0.8 mg/dl (0.3-1.1)). Five patients developed I/II° and two developed III° hyperbilirubinemia. No long-term hepatic or renal toxicity was observed.

Time to leukocyte recovery in the responding patients was 23 (12-28) days and platelet engraftment occurred at day 35 (12-55) in median. The BM niche evaluated by histology and IHC showed regular structure and distribution of hematopoiesis after engraftment (see Figure 1 for representative images).

Response evaluation was available in six patients, one died before response assessment. Five achieved complete remission, one patient was refractory.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (22)

Conclusions: Conditioning with CXCR4-directed ERT plus TBI is feasible and, with promising response and engraftment rates. These results warrant further prospective studies.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P160 EVALUATION OF THE TRANSPLANT CONDITIONING INTENSITY (TCI) SCORE FOR SECOND ALLOGENEIC STEM CELL TRANSPLANTATION (ALLO-SCT2) - A STUDY OF THE ALWP OF THE EBMT

Giuliano Filippini Velázquez 1, Jacques-Emmanuel Galimard2, Alexandros Spyridonidis3, Jakob Passweg4, Didier Blaise5, Nikolaus Kröger6, Igor Wolfgang Blau7, Patrice Chevallier8, Tobias Gedde-Dahl9, Panagiotis Kottaridis10, Johan Maertens11, Emma Nicholson12, Arancha Bermudez Rodriguez13, Peter Dreger14, Gerald G. Wulf15, Mareike Verbeek16, Emanuele Angelucci17, Mathias Eder18, Uwe Platzbecker19, Bipin Savani20, Christoph Schmid1, Fabio Ciceri21, Mohamad Mohty2

1University Hospital and Medical Faculty Augsburg, Augsburg, Germany, 2EBMT Paris Study Unit, Sorbonne University, Saint-Antoine Hospital, Paris, France, 3University of Patras, Patras, Greece, 4University Hospital Basel, Basel, Switzerland, 5Aix-Marseille Université, Maseille, France, 6University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 7Charité Universitätsmedizin, Berlin, Germany, 8Centre Hospitalier Universitaire de Nantes, Nantes, France, 9Oslo University Hospital, Oslo, Norway, 10University College London Hospitals NHS, London, United Kingdom, 11University Hospital Gasthuisberg, Leuven, Belgium, 12Royal Marsden Hospital, London, United Kingdom, 13Hospital Universitario Marqués de Valdecilla, Santander, Spain, 14University of Heidelberg, Heidelberg, Germany, 15Universitätsklinikum Göttingen, Göttingen, Germany, 16Klinikum Rechts der Isar, Munich, Germany, 17IRCCS Ospedale Policlinico San Martino, Genova, Italy, 18Hannover Medical School, Hannover, Germany, 19University Hospital Leipzig, Leipzig, Germany, 20Vanderbilt University Medical Center, Nashville, United States, 21IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, Milan, Italy

Background: A second allogeneic stem cell transplantation (allo-SCT2) is the most intensive treatment for relapse of acute myeloid leukemia (AML) after first allo-SCT (allo-SCT1). Even more than for allo-SCT1, choosing the conditioning for allo-SCT2 requires a fair balance between toxicity and antileukemic efficacy. The Transplant Conditioning Intensity (TCI) score was developed in patients >45 years transplanted for AML in first complete remission (CR1) to redefine conditioning regimen intensities (Spyridonidis et al., BMT 2020/2023). The score performed better than the reduced intensity conditioning/myeloablative conditioning (RIC/MAC) classification for prediction of non-relapse mortality (NRM) relapse incidence (RI), overall-, and leukemia-free survival (OS/LFS). Using the EBMT registry, we evaluated the role of the TCI score in allo-SCT2.

Methods: To obtain a homogenous cohort, we selected patients with AML relapse after allo-SCT1 in CR1, who had received allo-SCT2 in CR2 between 2013 and 2022. Conditioning regimens were classified as low, intermediate, or high intensity, according to the TCI definition. The impact of TCI on NRM, RI, OS, and LFS after allo-SCT2 was tested.

Results: The study included 492 patients (median age: 46.9 years). Median interval from allo-SCT1 to relapse and from relapse to allo-SCT2 was 20.8 and 3.8 months, respectively. Donors for allo-SCT2 were matched sibling (14%), haploidentical (24%), and unrelated (62%), with 84% donor change. Karnofsky performance score was ≥90% in 71%. According to European LeukemiaNet 2017 recommendations, 30% had adverse cytogenetics. The TCI score for allo-SCT2 was low in 36% (n=178, median age: 52.8 years), intermediate in 47% (n=232, median age: 43.7 years), and high in 17% (n=82, median age: 42.2 years). Median follow-up after allo-SCT2 was 2.8 years. In the entire cohort, 2-year NRM was 18.4%; among TCI subgroups NRM was 19.8%/17.5%/18.4% for low/intermediate/high, respectively. Overall, 2-year RI was 41% (TCI low: 42.7%, intermediate: 42%, high: 34.7%). Two-year OS/LFS were 50.9%/40.5% without differences among TCI-based subgroups. No different role for TCI was observed in elderly patients.

In the multivariable analysis (Table 1), age >47 years (p<0.001), female donor for male recipient (p=0.006), haploidentical donor for allo-SCT2 (p=0.04), and cytomegalovirus serology (p=0.02) were predictive factors for NRM. Adverse cytogenetics (p=0.01) and interval from allo-SCT1 to relapse <18 months (p=0.03) were predictive for RI. Cytogenetics, shorter remission, age >47 years, and CMV status were also significant factors for OS and LFS.

There was a suggestion of a high TCI being predictive for a reduced RI (p=0.06) without significance for NRM, OS or LFS. In an unadjusted exploratory analysis, a higher TCI was associated with longer LFS among patients relapsing within one year from allo-SCT1 (p=0.04).

Full size table

Conclusions: In this cohort of patients undergoing allo-SCT2 in CR2, we did not observe an effect of TCI on NRM, nor on RI, OS, or LFS. A high TCI showed a trend towards reduced RI that was not counterbalanced by an increased NRM. These results do not support any intensity level as optimal conditioning for allo-SCT2 in this setting. A more intensive regimen might be of benefit in patients with remission <1 year after allo-SCT1.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P161 VENETOCLAX COMBINED WITH FLUDARABINE AND MELPHALAN CONDITIONING REGIMEN IN PATIENTS > 50 YEARS WITH MYELOID MALIGNANCIES: INTERIM ANALYSIS OF A PROSPECTIVE, MULTICENTER, PHASE II TRIAL

Panpan Zhu 1,2,3,4, Xiaoyu Lai1,2,3,4, Lizhen Liu1,2,3,4, Yibo Wu1,2,3,4, Jimin Shi1,2,3,4, Jian Yu1,2,3,4, Yanmin Zhao1,2,3,4, Yishan Ye1,2,3,4, Guifang Ouyang5, Yi Chen6, Weiyan Zheng1,2,3,4, Jie Sun1,2,3,4, He Huang1,2,3,4, Yi Luo1,2,3,4

1the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Hangzhou, China, 3Institute of Hematology, Zhejiang University, Hangzhou, China, 4Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China, 5Ningbo City First Hospital, Ningbo, China, 6The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China

Background: The use of a myeloablative conditioning (MAC) regimen prior to allogeneic hematopoietic stem cell transplantation (allo-HSCT) optimizes disease control but carry the risk of significant toxicity. Therefore, older patients are often unfit for MAC and relegated to reduced intensity conditioning regimens. The oral Bcl-2 inhibitor and BH3 mimetic venetoclax (VEN) would enhance the anti-leukemia effect of conditioning regimen. Here, we report a prospective, multicenter, phase II clinical trial (NCT05084027) to evaluate the safety and efficacy of adding VEN to fludarabine (Flu) and melphalan (Mel) conditioning regimen in patients aged > 50 years with myeloid malignancies.

Methods: Eligibility included: age > 50 years, acute myeloid leukemia (AML) in complete remission or myelodysplastic syndrome (MDS). Pts received VEN (400mg/d on days -8 to -2) in combination with Flu (30mg/m2/d on days -7 to -3) and Mel (140mg/m2/d on day-2 for patients < 60; 120 mg/m2/d on day-2 for patients ≥ 60), followed by peripheral blood stem cells infusion on Day 0. Graft versus host disease (GVHD) prophylaxis consisted of cyclosporin A, methotrexate, low-dose mycophenolate mofetil and rabbit antithymocyte globulin.

Endpoints included toxicity, overall survival (OS), disease-free survival (DFS), GVHD-free and relapse-free survival (GRFS), relapse, and non-relapse mortality (NRM).

Results: Fifty-one patients (pts) were enrolled between October 2021and July 2022, with a median follow up of 380 (125–771) days. The median patient age was 58 years (range, 51–67) and 37.3% were females (Table). There were 41 pts diagnosed with AML and 10 pts with MDS. Donor source was matched sibling donor (MSD) in 1 patient, unrelated donor (URD) in 7, and haploidentical donor (HID) in 43. A total of 25 pts were classified as high/very high risk based on refined disease risk index (rDRI).

Among these pts, grade 2 toxicities were stomatitis and gastrointestinal tract toxicity. Grade 3 stomatitis was observed in 14 pts (27.5%). No toxicity-related deaths were observed.

Cumulative incidence of Grade II-IV acute GVHD by day +100 was 9.8% (95% confidence interval (CI): 3.6–20%). Only 2 of 51 pts experienced Grade III acute GVHD. No patients experienced grade IV acute GVHD and there were no deaths due to GVHD. At 1 year, no patients experienced severe chronic GVHD, with the cumulative incidence of moderate chronic GVHD of 6.9% (95% CI: 1.7–17%). One-year cumulative incidences of relapse and NRM were 11.0% (95% CI: 4.0–22%) and 7.1% (95% CI: 1.8–18%), respectively. One-year probabilities of OS, DFS, and GRFS were 85.5% (95% CI: 75.3–97.0%), 81.7% (95% CI: 71.0 – 94.1%), and 75.5% (95% CI: 63.8–89.4%).

Table. Baseline characteristics in patients aged > 50 years with RIC regimens of VEN combined with Flu and Mel followed by allo-HSCT

Baseline characteristics

No. (%)

Median age (range, year)

58 (51 – 67)

< 60

35 (68.6)

≥ 60

16 (31.4)

Patient sex, female/male

19 (37.3) / 32 (62.7)

Diagnose

AML

41 (80.4)

MDS

10 (19.6)

HCT-CI ≥ 2

9 (17.6)

rDRI

Low

4 (7.8)

Intermediate

22 (43.1)

High/Very high

25 (49.0)

Transplantation type

MSD

1 (2.0)

URD

7 (13.7)

HID

43 (84.3)

Donor age

33 (21 – 50)

Donor sex, female/male

19 (37.3) / 32 (62.7)

Conclusions: Our analysis suggests the conditioning regimen comprising VEN combined with Flu and Mel is feasible and effective, and could achieve promising prognosis of allo-HSCT in patients aged > 50 years with myeloid malignancies.

Clinical Trial Registry: NCT05084027 (https://classic.clinicaltrials.gov/).

Disclosure: No author has a potential conflict of interest or funding source.

16: Conditioning Regimens

P162 FLUDARABINE/TREOSULFAN VS. FLUDARABINE/TBI CONDITIONING FOR ALLOSCT FOR AML AND MDS: COMPARABLE SURVIVAL RATES BUT DIVERGENT IMMUNE RECONSTITUTION

Lina Kolloch1, Philipp Berning1, Christian Reicherts1, Julian Ronnacker1, Simon Call1, Julia Marx1, Matthias Floeth1, Eva Esseling1, Jan-Henrik Mikesch1, Christoph Schliemann1, Georg Lenz1, Matthias Stelljes 1

1University Hospital Muenster, Muenster, Germany

Background: Allogeneic stem cell transplantation (alloSCT) is a standard treatment option for patients (pts) with acute myeloid leukemia (AML) or myelodysplastic neoplasms (MDS). Dose-reduced conditioning regimens, such as ‘FluTBI’ (fludarabine and 8 Gy fractionated total body irradiation) or ‘FluTreo’ (fludarabine and treosulfan) are employed for pts with AML in complete remission (CR) and MDS pts. FluTreo is preferred in older and/or comorbid patients, potentially offering lower toxicities compared to FluTBI.

Methods: In this retrospective study, we analyzed 215 pts with AML in CR and 96 MDS pts who received a first alloSCT between 2010 and 2022. Pts were conditioned with FluTreo (120-150 mg/m2 fludarabine, 30 g/m2 treosulfan) or FluTBI (120 mg/m2 fludarabine, 4x2Gy TBI) prior to alloSCT. Median follow-up of survivors was 34 months. Using propensity score matching, we identified 53 pair-matched pts from both groups (nearest matching for disease, sex, age at alloSCT and ECOG; caliper distance of 0.3 standard deviations).

Results: Following propensity score matching, differences in age at alloSCT (FluTreo 56 y vs. FluTBI 54 y), disease (AML: 83% vs. 81.1%), sex and ECOG were balanced. Both groups (FluTreo vs. FluTBI) were also equilibrated for ELN2017 risk categories for AML (adverse 40.9% vs. 27.9%) and IPSS-R risk categories for MDS (very high/high 66.7% vs. 70%). In terms of transplant characteristics, both groups demonstrated comparable rates of measurable residual disease (MRD) positivity before start of conditioning (59.1% vs. 51.2%) and similar distributions of donor types.

For the matched cohorts, we found comparable overall (OS) and relapse free survival (RFS) (3-yr OS: 80% vs. 69%; 3-yr RFS: 73% vs. 63%). Relapse incidences were comparable (3-yr RI: 23% vs 23%). Notably, a trend towards lower non-relapse mortality (NRM) was observed in the FluTreo cohort (3-yr NRM: 3.8% vs. 14%, p=0.08). The incidence of acute/chronic graft-versus-host disease (GvHD) was comparable between the matched groups (aGvHD grade III-IV) 100d: 7.5% vs. 5.7%; cGvHD (all grades) 3-yr: 29% vs. 32%). For pts with MRD prior to alloSCT RFS and OS did not significantly differ between both groups. Analysis of toxicities revealed higher frequency of mucositis in the FluTBI cohort (mucositis ≥CTC grade 2: 23% vs. 51%, p=0.005). Over the first three years after alloSCT, matched FluTreo pts displayed significant better ECOG performance status (PS) than FluTBI pts (median ECOG PS at 1-yr: 0.77 vs. 1.14, p=0.01). A subset of immune reconstitution represented by normalization of CD4 pos. lymphocytes (>300/µl) in the peripheral blood trended to be faster in the FluTreo group (100d: 19% vs. 2%, p=0.03), while normalization of CD8 pos. and B-lymphocytes did not display significant differences. Normalization of immunoglobulin G trended to be slightly faster in the FluTreo group, without reaching significance.

Conclusions: Our data revealed comparable and favorable survival rates for both conditioning regimens. FluTreo conditioning exhibited partially earlier immune reconstitution and less restriction regarding the ECOG performance status, emphasizing FluTreo as a compelling alternative to FluTBI. This warrants further evaluation in a comparative prospective trial.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P163 CONDITIONING FOR ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN PATIENTS WITH PERIPHERAL T-CELL LYMPHOMA (PTCL): INTERMEDIATE-DOSE TBI DOES THE JOB

Isabelle Krämer1, Thomas Luft 1, Ute Hegenbart1, Stefan Schönland1, Peter Stadtherr1, Linda Selberg1, Laila König1, Carsten Müller-Tidow1, Peter Dreger1

1University of Heidelberg, Heidelberg, Germany

Background: AlloHCT is an effective treatment for patients with relapsed/refractory PTCL. While there is sound evidence that graft-versus-lymphoma effects are the main driver of lymphoma eradication in this setting, the contribution of the conditioning regimen to successful alloHCT outcome in patients with PTCL is still unclear. In 2010, we adopted an intermediate-dose total body irradiation-based regimen (Bornhaeuser et al, Lancet Oncol 2012) as standard conditioning for alloHCT in r/r PTCL. Here we report our experience with this approach.

Methods: Eligible for this retrospective analysis were all adult patients who had undergone a first alloHCT for PTCL in our institution between 2005 and 2023 receiving conditioning with a TBI/fludarabine backbone. Specifically, TBI was given fractioned in 4 doses at 2Gy on 2 consecutive days along with fludarabine, and ATG in patients with unrelated donor, followed by standard CNI-based immunosuppression. Patients could receive only 3 fractions of TBI (=6Gy) in case of comorbidity or performance status-driven limitations. Primary endpoint was relapse/progression-free survival (PFS). For comparison, we also considered all patients who were conditioned without TBI during the same time corridor.

Results: Altogether 44 patients with a median age of 51 (20-66) years met the eligibility criteria. Diagnosis was AITL (30%), ALCL (27%), PTCL-NOS (18%), hepatosplenic TCL (HSTL) (11%), and other PTCL (14%). AlloHCT was administered as part of 2nd-line therapy in 68%, and in later lines in 30% of the patients. 39% of the patients had undergone a prior autologous or syngeneic HCT. Disease status was CR and PR in 27% of the patients each, and refractory in the remainder. Donors were MRD, MUD, and 9/10 MMUD in 43%, 39%, and 18% of the cases, respectively. The TBI target dose of 8Gy was met in 89% of the patients. 5 patients received only 6Gy, and in single patient with refractory enteropathy-associated TCL was escalated to 12Gy.

With a median follow-up of 6.4 (1-13.3) years, 27 patients are alive and well, and 17 died. Causes of death were PTCL progression (9), pneumonia (3), viral infections (2), and graft failure, cardiac infarction, other cancer in a single patient each. With two additional relapse events which could be successfully rescued, 5-year PFS, overall survival (OS), incidence of relapse/progression (RI) and non-relapse mortality (NRM) were 61%, 65%, 25%, and 14%, respectively. Survival outcomes were comparable across all PTCL subsets. Of note, although significantly worse compared to sensitive patients, 5-year OS of patients undergoing TBI-based alloHCT with refractory disease was promising with 43% (95%CI 20-66) (Figure).

21 patients with PTCL were allografted with TBI-free conditioning before 2010 or protocol-driven. This population was older (59 (21-74) years), had a lower proportion of refractory patients (33%), and contained 24% haplotransplants, but was otherwise comparable to the TBI cohort. With a median follow-up of 12.5 (1.5-17.3) years, 5-year PFS, OS, RI, and NRM were 45%, 43%, 24%, and 30%, respectively.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (23)

Conclusions: AlloHCT with intermediate-dose TBI/fludarabine conditioning is associated with low NRM and can provide durable survival in two thirds of patients with r/r PTCL including those with refractory disease status at transplant.

Disclosure: Isabelle Krämer reports training and travel grant from Janssen-Cilag, honoraria from AbbVie, congress participation fee from Lilly. Ute Hegenbart reports honorarium for talks from Janssen, Pfizer, Alnylam, Akcea, Prothena, Astra Zeneca; financial support of congress participation from Janssen, Prothena, Pfizer; advisory boards for Pfizer, Prothena, Janssen, Alexion and financial sponsoring of Amyloidosis Registry from Janssen. The remaining authors declare no competing financial interests..

16: Conditioning Regimens

P164 5-YEAR TRANSPLANT SUCCESS AFTER TREOSULFAN CONDITIONING

Rohtesh S. Mehta 1, Joachim Deeg1, Ted Gooley1, Stephanie J. Lee1, Laurel Thur1, Filippo Milano1

1Fred Hutch Cancer Center, Seattle, United States

Background: New regimens including Treosulfan (TREO) have been increasingly used in allogeneic hematopoietic cell transplantation (HCT), frequently in combination with fludarabine (FLU), where it has been effective in establishing engraftment with low toxicity and non-relapse mortality (NRM). Herein, we report long-term outcomes of patients treated with TREO-based regimens, which have not been described yet.

Methods: Retrospective analysis of all patients (n=345) who received TREO-based conditioning for HCT at the Fred Hutchinson Cancer Center (FHCC) between 2005-2019. Donors were matched related (RD), unrelated (URD) or cord blood (CB). Patients received TREO on days −6 to −4 at 10 or 14 g/m2/day (total 30 or 42 g/m2) and FLU IV on days −6 to −2 at 30 or 40 mg/m2/day (total 150-200 mg/m2). Most patients (n=255) also received 2Gy total body irradiation on day 0, followed by infusion of donor cells. Graft-versus-host disease (GVHD) prophylaxis included tacrolimus/methotrexate in the RD/URD cohort and cyclosporine/MMF for CB. The FHCC Long-Term Follow-Up questionnaire was used to analyze the return-to-work status information of patients who were alive 1 year post transplant (n=259).

Results: Median patient age at HCT was 50.2 years (range, 0.7-70.5), 50% were males, 199 (58%) received peripheral blood grafts, 120 (35%) received CB, and 26 bone marrow. Most had acute myeloid leukemia (54%) or myelodysplastic neoplasm (31%). The median follow-up among survivors was 65 months (range, 12-165.7) [Table]. Estimated overall survival (OS) at 5 years was 56% (95% confidence interval (CI) 51-61), relapse-free survival was 51% (46-56), relapse was 27% (23-32), NRM was 21% (17-26). The composite endpoint of immunosuppression-requiring chronic GVHD-free relapse-free survival (CRFS) was 42% (36-47) and GVHD-free relapse-free survival (GRFS: grade 3-4 acute GVHD, immunosuppression-requiring chronic GVHD, relapse, or death) was 38% (32-43). With respect to the return-to-work status, 196/259, 91/185 and 62/124 patients responded at 1-year, 3-years and 5-years post-HCT, respectively. The distribution at respective timepoints was: back to work (43%, 53%, 52%), school (11%, 7%, 6%), unemployed (3%, 3%, 6%), home (7%, 7%, 2%), limited by health (34%, 30%, 32%), and none of the above (3%, 1%, 2%).

BASELINE CHARACTERISTICS

N=345

%

Donor type (n, %)

HLA-matched unrelated

128

37

HLA-matched related

85

25

Cord blood

120

34

HLA-mismatched unrelated

12

3

Disease (n, %)

Acute myeloid leukemia

186

54

Myelodysplastic neoplasm

106

31

Acute lymphoblastic leukemia

36

10

Others

17

5

Cytomegalovirus (n, %)

Patient CMV +

225

67

Patient CMV -

113

33

Donor CMV +

90

35

Donor CMV -

166

65

Female-to-Male Donor-recipient sex (n, %)

62

27

Follow-up among survivors; median (range); in months

64.6 (12-165.7)

Conclusions: TREO-based regimens yield encouraging long-term outcomes. The return-to-work status in our population is consistent with reports from the registry studies [Bhatt et al. JTCT 2021;27(8):679.e1-679.e8], while GRFS and CRFS appear to be superior to what has been reported in large registry studies with other regimens [Mehta et al. JCO 2020;38(18):2062-2076, and Holtan et al. Blood 2015; 125(8): 1333–1338].

Clinical Trial Registry: N.A.

Disclosure: Medexus provided research funding for the analysis (F.M.).

16: Conditioning Regimens

P165 EFFICACY ANALYSIS OF HAPLOIDENTICAL TRANSPLANTATION USING BUSULFAN/CYCLOPHOSPHAMIDE AND MELPHALAN AS CONDITIONING REGIMEN FOR RELAPSED AND REFRACTORY ACUTE MYELOID LEUKEMIA

Fei Pan1, Guanlan Yue1, Kang Gao1, Kun Wang1, Huili Zhu1, Yujie Wang1, Zheren Wang1, Zhijie Wei 1

1Hebei Yanda Lu Daopei Hospital, Hebei, China

Background: The chemotherapy effectiveness for relapsed and refractory acute myeloid leukemia (R/R-AML) is extremely poor. Despite the emergence of new treatment methods such as CAR-T cell therapy and targeted drug treatments, the long-term survival rate remains low, at around 40%. Currently, only allogeneic hematopoietic stem cell transplantation offers a potential cure. This study compares the modified Busulfan/Cyclophosphamide and Melphalan (Bu/Cy+Mel) regimen with traditional regimen and analyzes the efficacy of haploidentical hematopoietic stem cell transplantation in 94 cases of R/R-AML at a single center.

Methods: From November 2012 to July 2023, a total of 94 patients with R/R-AML were consecutively treated at our hospital (Hebei Yanda Lu Daopei Hospital) (Table 1). Among them, 18 patients received the BU/Cy conditioning regimen, and 76 patients received the BU/Cy+MEL conditioning regimen. Transplant type: all were haploidentical. Conditioning regimen: Bu/Cy/Mel (Flud 30 mg/m2/d×5d; Ara-C 2g/m2/d×5d; Bu 3.2 mg/kg/d×4d, Cy 1.8g/m2/d×2d; Vp16 100mg/m2/d×2d; ATG 1.5mg/kg/d×5d, MEL 55mg/m2/d×2d). GVHD prevention: CsA+MMF+sMTX+ basiliximab ( + 1, +4), ATG total 7.5mg/kg divided over 5 days (-5 to -2, +7).

Results: The post-transplant survival status of patients in both groups is shown in Table 2. There was no statistically significant difference in the overall survival rate (OS) between the two groups. In remission, the OS of the Bu/Cy+Mel group was superior to that of the Bu/Cy group, with a statistically significant difference. In the NR (non-remission) state, there was no statistically significant difference in OS between the Bu/Cy+Mel group and the Bu/Cy group.

Conclusions: The application of the modified Bu/Cy+Mel conditioning regimen does not increase the transplant-related mortality in hematopoietic stem cell transplantation. Compared to traditional regimen, it can improve the efficacy of haploidentical hematopoietic stem cell transplantation in R/R-AML patients after achieving complete remission (CR), but the effect is not significant in NR patients, necessitating further improvement in the transplantation protocol. Post-transplant administration of four cycles of azacitidine (32mg/m2×7d) as a preventive treatment for relapse can improve the long-term survival rate of patients with relapsed and refractory acute myeloid leukemia. However, this requires long-term follow-up in multicenter clinical studies with large sample sizes.

Disclosure: No relevant conflicts of interest to declare.

16: Conditioning Regimens

P166 POST-TRANSPLANT CYCLOPHOSPHAMIDE RESULTS IN LOW INCIDENCE OF CHRONIC GRAFT-VERSUS-HOST DISEASE IN UNRELATED OR MATCHED-SIBLING DONOR STEM CELL TRANSPLANTATION WITHOUT COMPROMISING IMMUNE RECONSTITUTION AND GRAFT-VERSUS-LEUKEMIA EFFECT

Ioannis Tsonis 1, Ifigeneia Tzannou1, Tatiana Tzenou1, Natali El Gkotmi1, Maria Katsareli1, Ismini Darmani1, Vasiliki Bampali1, Evridiki Theodorou1, Maria-Eleni Karatza1, Maria Garofalaki1, Zoi Poulopoulou1, Marina Papageorgiou1, Soultana Tryfonidou1, Panagiotis Siamatas1, Eirini Tziotziou1, Eirini Grispou1, Stavros Gigantes1, Ioannis Baltadakis1, Aikaterini Souravla1

1Evangelismos General Hospital, Athens, Greece

Background: Post-Transplant Cyclophosphamide (PTCY) was first applied in the haploidentical stem cell transplantation (SCT) setting and it resulted in high percentages of engraftment and reduced frequency of acute and mainly chronic GvHD as well as non-relapsed mortality. It is increasingly being used in mismatched unrelated or fully matched donor transplants. We prospectively tested this approach in SCT from matched or mismatched unrelated (MUD/MMUD) and matched sibling donors (MSD) to reduce the incidence of acute and especially chronic graft-vs-host disease (GVHD).

Methods: Fifty-one consecutive patients who underwent allogeneic hematopoietic stem cell transplant from an unrelated donor were studied. PTCΥ was administered on days +3 and +4 at a dose of 50 mg/kg/day. Additional GvHD prophylaxis included tacrolimus and mycophenolate mofetil starting on day 0. In patients with no evidence of GvHD immunosuppression was gradually withdrawn by day +90. We evaluated the safety and efficacy of the procedure in terms of engraftment, non-relapse mortality (NRM), acute or chronic GvHD (aGVHD/cGVHD), relapse, survival, and immune reconstitution.

Results: From 01/2021 to 05/2023, 51 consecutive patients, aged 18-69 (median 52) years, received allogeneic SCT with PTCY for AML/MDS (n=36), NHL/CLL (n=2), primary myelofibrosis (n=2), CML (n=2), or ALL (n=9). Disease risk index (DRI) was intermediate in most patients (34/51), and high in 10. The conditioning regimen was myeloablative in 37 patients and reduced-intensity in 14. 36 patients received grafts from 8/8 MUD, 12 from MMUD (7/8), and 3 from MSD. Graft source was peripheral blood in all cases, with a median CD34+ cell dose of 6.97x10e6/kg (range, 3.27-15.82). Cumulative incidence (CI) of neutrophil engraftment (>500/ul) was 95.8%, and was achieved at a median of 16 (range 13-27) days. CI of grade ΙΙ-IV aGVHD was 46.7% (95%CI, 34.1-63.8), however grade ΙΙΙ-IV aGVHD occurred only in 3.9% (95%CI, 1.0-15.5). CI of cGVHD at 18 months was 13.4% (95%CI, 5.8-31.0), with only 1 case of extensive cGVHD. At a median follow-up of 14.5 (range 1-28) months, disease-free (DFS) and overall survival (OS) at 18 months was 76.8% (95%CI, 65.5-90.0) and 76.3% (95%CI, 64.9-89.8), respectively, and did not differ significantly between MUD and MMUD SCT. GvHD-free, relapse-free survival (GRFS) was 63.5% (95%CI, 50.3-80.1). CI of relapse and NRM was 4.3% and 15.5%, respectively. Infections accounted for NRM in the majority of cases. Median absolute CD8+ and CD4 + T cell counts were420/ul (range, 20-2225) and 179/ul (range, 13-594) respectively at 6 months versus 552/ul (range, 55-3002) and 364/ul (range, 60-787) respectively at 12 months. Viral infections occurred in 28 patients (respiratory:13, BKV cystitis:11, CMV infection/disease:7). In addition, 2 fungal and 1 atypical mycobacterial infection were documented.

Conclusions: In conclusion PTCY resulted in a remarkably low incidence of cGVHD in MUD and MMUD SCT. Relapse rate was low, immune reconstitution was robust for CD8+ but somewhat delayed for CD4 + T cells. However, opportunistic infections were not increased. Thus, PTCY may revert the negative impact of HLA mismatch in unrelated SCT and is also a beneficial option for MUD and MSD transplants.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P167 CONDITIONING INTENSITY IN STEM CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR ACUTE MYELOID LEUKEMIA: A STUDY FROM THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT

Jaime Sanz 1, Myriam Labopin2, Jurjen Versluis3, Didier Blaise4, Jacoppo Peccatori5, Juan Montoro1, Gwendolyn Van Gorkom6, Peter von dem Borne7, Hélène Labussière-Wallet8, Montserrat Rovira9, Péter Reményi10, Patrice Chevallier11, Mi Kwon12, Matthias Eder13, Jan Vydra14, Eolia Brissot15, Alexandros Spyridonidis16, Simona Piemontese5, Mohamad Mohty15, Fabio Ciceri5

1Hospital Universitari i Politècnic La Fe, Valencia, Spain, 2EBMT Paris Office, Hospital Saint Antoine, Paris, France, 3Erasmus MC Cancer Institute, Rotterdam, Netherlands, 4Programme de Transplantation & Therapie Cellulaire, Marseille, France, 5Hematology and Bone Marrow Transplant Unit, San Raffaele Scientific Institute IRCCS, Milano, Italy, 6University Hospital Maastricht, Maastricht, Netherlands, 7Leiden University Medical Center, Leiden, Netherlands, 8Centre Hospitalier Lyon Sud, Lyon, France, 9BMT Unit, Institute of Haematology and Oncology, IDIBAPS, Hospital clinic, University of Barcelona. Josep Carreras Leukaemia Research Foundation, Barcelona, Spain, 10Dél-Pesti Centrumkórház, Budapest, Hungary, 11CHU Nantes, Nantes, France, 12Hospital Gregorio Marañón, Madrid, Spain, 13Hannover Medical School, Hannover, Germany, 14Institute of Hematology and Blood Transfusion, Prague, Czech Republic, 15Hôpital Saint-Antoine, Sorbonne University, INSERM UMRs 938, Paris, France, 16Hematology, BMT and Institute of Cellular Therapy, University of Patras, Patras, Greece

Background: The impact of conditioning intensity in acute myeloid leukemia (AML) remains somewhat controversial. Moreover, specific studies on hematopoietic stem cell transplantation (HSCT) using post-transplant cyclophosphamide (PTCy) are lacking.

Methods: We retrospectively analyzed transplant outcomes in AML patients with intermediate or adverse-risk cytogenetics, in first complete remission (CR1) who a received a first allogeneic HSCT using PTCy between January 2010 and December 2022. The main objective was to analyze the impact of conditioning intensity on leukemia-free survival (LFS) according to cytogenetic/molecular risk.

Results: Overall, 1813 patients were included with a median age of 55 years (range, 18-75). The intermediate-risk cytogenetic group accounted for 1386 (76%) patients; 608 (34%) and 578 (32%) had mutated FLT3-ITD and NPM1, respectively. Myeloablative conditioning was used in 930 patients (51%), while 1130 (62%) received an intensified conditioning (score ≥ 2.5) based on the transplant condition intensity (TCI) score. Conditioning intensity using the myeloablative/reduced intensity stratification did not have any impact on transplant outcomes across the entire cohort. However, a high TCI score showed lower risk of relapse counterbalanced with higher risk of NRM, without effect on survival. When focussing on specific cytogenetic risk groups, conditionings with higher TCI score had no influence on outcomes in the adverse-risk group, while in the intermediate-risk group, its impact varied with FLT3-ITD status. Patients with wild type FLT3-ITD who received conditioning with a higher TCI score had increased NRM (HR 1.5; 95% CI 1.02-2.2) and lower GRFS (HR 1.33; 95% CI 1.05-1.69). In contrast, patients with mutated FLT3-ITD receiving intensified conditioning with higher TCI score exhibited reduced risk of relapse (HR 0.48; 95% CI 0.3-0.79) and improved LFS (HR 0.69; 95% CI 0.47-0.99).

Conclusions: For patients undergoing HSCT with PTCy for AML in CR1, it is crucial to consider cytogenetic risk and molecular status when selecting the the conditioning regimen. In intermediate-risk cytogenetics, intensive conditioning should be considered for mutated FLT3-ITD, but probably avoided for patients with wild type FLT3-ITD.

Disclosure: Authors declare no conflicts of interest.

16: Conditioning Regimens

P168 EVALUATION OF A BALTIMORE MODIFIED NON-TBI CONDITIONING BEFORE ALLOGENEIC TRANSPLANT FROM HAPLOIDENTICAL OR MISMATCHED UNRELATED DONORS, RESULTS FROM A MONOCENTRIC RETROSPECTIVE STUDY

Victoria Volpari 1, Claude Eric Bulabois1, Anne Thiebaut-Bertrand1, Sophie Park1, Martin Carre1, Mathieu Meunier1

1CHU Grenoble Alpes, Grenoble, France

Background: The Baltimore conditioning regimen is nowadays one of the most used reduced intensity conditioning (RIC) regimens in the field of haploidentical allogeneic stem cell transplantation (ASCT). It has been widely extended to mismatched unrelated donor (MMUD). However, total body irradiation (TBI) access is difficult in some centers and for practical reasons, chemo-based conditionings are preferred. For such a purpose, we replaced the 2 grays TBI with 2 doses of IV Busulfan. In this retrospective study, we aimed to evaluate the outcomes of old and/or frail patients transplanted after a non-TBI modified Baltimore RIC regimen consisting of fludarabine, cyclophosphamide and busulfan (FE2B2) for haploidentical or MMUD donor, and post-transplant high-dose cyclophosphamide as graft versus host disease (GVHD) prophylaxis.

Methods: We conducted a retrospective monocentric study. We included 65 adult patients who received the FE2B2 regimen between March 1st, 2016, and April 20th, 2022 at the Grenoble-Alpes University Hospital in France. FE2B2 consisted of Fludarabine 30 mg/m2 on days -6 to -2 (total dose 150 mg/m2), Cyclophosphamide 14.5 mg/kg/day on day -6 and -5 (total dose 29 mg/kg), and Busulfan 3.2 mg/kg/day on day -3 and -2 (total dose 6.4 mg/kg). On day 0, all patients received an unmanipulated PBSC allograft. GVHD prophylaxis consisted of high dose PTCy 50 mg/kg/day at day 3 and day 4, Ciclosporin 3 mg/kg/day or Tacrolimus 2 times a day and MMF beginning at day 5.

Results: Transplant was performed for AML in 38 patients (58.5%) with a status at transplant beyond CR1 for 16 patients, myelodysplastic syndrome in 15 patients (23%) and ALL in 6 patients (9.5%). Six patients had already received an allotransplant. Median age was 65 years. Thirty-seven patients (57%) received a transplant from a MMUD and 28 patients (43%) from an haploidentical donor.

Four patients experienced an early death before day 20 due to infection, cardiac insufficiency and hemorrhage. Engraftment occurred in 59/65 patients (91%) and in 96.7% of the patients were alive after day 20. Median follow-up was 291 days (IQR 125-1114). The 1-year probability of overall survival was 50% and was not significantly different depending on donor type (OS 0.913; p= 0.86). The cumulative incidences of grade II-IV and grade III-IV acute GVHD were 43% and 14.8% respectively. The 1-year cumulative incidence of chronic GVHD was 20%, mostly mild. The 1-year probability of DFS was 42.6%, 1-year NRM was 18%. One-year relapse rate was 38%. BK virus associated symptomatic cystitis occurred in 7 patients (11%).

Conclusions: The FE2B2 regimen appears feasible with an acceptable toxicity in this old and/or unfit category of patients with hematological malignancies and seems to be an alternative option to the classical Baltimore TBI-containing conditioning in centers who may have difficulties to TBI access. Nevertheless, high rate of relapse remains a matter of concern in this high-risk disease cohort where almost one-third of the patients were beyond CR1 at transplant. Post-transplant maintenance could be an option to mitigate this excess of relapse.

Disclosure: The authors declare no competing financial interests..

16: Conditioning Regimens

P169 EXPLORING THE IMPACT OF CONDITIONING REGIMEN INTENSITY ON ALLOGENEIC STEM CELL TRANSPLANT OUTCOMES: INSIGHTS FROM A SINGLE-CENTER STUDY

Vera Radici 1, Riccardo Marnoni1, Mirko Farina1, Eugenia Accorsi Buttini1, Sofia Terlizzi1, Lorenzo Masina1, Enrico Morello1, Gabriele Magliano1, Alessandro Leoni2, Federica Re2, Katia Bosio2, Simona Bernardi2, Domenico Russo1, Michele Malagola1

1Unit of Blood Diseases and Bone Marrow Transplantation, Cell Therapies and Hematology Research Program, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy, 2Research Center Ail (CREA), ASST Spedali Civili di Brescia, Brescia, Italy

Background: The Transplant Conditioning Intensity (TCI) index, proposed by EBMT, refines conditioning regiment intensity assessment compared to traditional MAC and RIC classifications. The intensity weight scores were assigned to commonly utilized drugs in the conditioning regimen. The sum of these scores was employed to calculate a discrete 3-category stratification TCI index (low, intermediate, high).

Methods: This retrospective study, conducted at the BMT center of the University Hospital of Brescia between March 2006 and September 2023, aimed to evaluate the predictive capacity of the TCI index in HSCT. Outcomes assessed included OS, NRM, GVHD-free relapse-free survival (GRFS) and the Cumulative Incidence of Relapse (CIR).

Results: In this study, 556 patients underwent HSCT for various hematologic disorders, with a median age of 53 years (range 16-75). Stem cell sources included peripheral blood stem cells (82%), bone marrow (16%), and umbilical cord blood (UCB, 2%). Donor categories included match-related (34%), haploidentical (18%), matched unrelated donor (46%), and UCB (2%). MAC and RIC regimens were distributed at 47% and 53%, respectively. MAC regimens were more used in younger patients with lower comorbidity, higher Karnofsky Performance Status (KPS>80), and diseases not in first remission. The TCI score distribution was 36% low, 37% intermediate and 22% high. Conditioning regimen intensity correlated significantly with patient age, comorbidities, and performance status. The TCI index demonstrated nuanced stratification, particularly in the age group 50-70, predicting OS and GRFS, whereas significance diminished below age 50. Median OS was 116.8 months versus 27.4 months in MAC and RIC groups (p<0.001). OS rates at 1 and 3 years were 71.5% versus 65.6% and 60.1% versus 44.8% in MAC and RIC (p<0.001). Stratification into Low, Intermediate, and High groups revealed median OS differences (p<0.001) and distinct OS rates at 1 and 3 years within each group (p<0.001). Statistical significance persisted in the 50-70 age group, with OS rates favoring MAC (p=0.005). GRFS rates at 1 and 3 years also exhibited significant differences between MAC and RIC (p<0.001) and within TCI categories (p=0.02).

In the multivariate analysis of overall survival, significant factors include the presence of disease at 1st CR (HR 0.69; 95% CI 0.52-0.90; p=0.006), an HCT-CI ≥3 (HR 1.58, 95% CI 1.22-2.06; p<0.001), a KPS≤80 (HR 1.69; 95% CI 1.11-2.57; p=0.01), and the patient’s positive CMV serology (HR 1.85; 95% CI 1.17-2.92; p=0.009).

Cumulative incidence of NRM showed no significant difference across TCI categories, differently from previous studies. NRM rates at day +100, +180, and 2 years varied but were not significant (p=0.23). CIR showed a significant relationship between MAC and RIC (p<0.001), but not within TCI categories. While no association was identified between cGVHD and TCI, a statistically significant correlation was observed for grade II-IV aGVHD, indicating a heightened risk in High-intensity regimens.

Conclusions: The TCI index emerged as a valuable tool for assessing the impact of conditioning regimens on transplant-related outcomes, especially in patients aged 50-70, demonstrating predictive capability for OS and GRFS, nuanced stratification within TCI categories and aGVHD.

Disclosure: No disclosures.

16: Conditioning Regimens

P170 IMPACT OF PRETRANSPLANT MRD IN PATIENTS ALLOGRAFTED FOR AML AFTER TREOSULFAN-BASED CONDITIONING

Jacob Pyka 1, Nils Leimkühler1, Artur Schneider1, Jennifer Kaivers1, Rudolf Trenschel1, Annemarie Mohring1, Christian Reinhardt1, Thomas Schroeder1, Christina Rautenberg1

1University Hospital Essen, Essen, Germany

Background: Allo-SCT represents the only curative treatment option for a majority of AML patients but was traditionally reserved for young and fit patients (pts) due to related toxicities. However, the introduction of Treosulfan (Treo) as “toxicity-reduced” conditioning expanded the access of elderly and/or patients with comorbidities to allo-SCT due to a relevant reduction of treatment-related mortality, while preserving sufficient anti-leukemic effect. However, even in pts allografted in complete remission relapse represents the main cause of treatment failure after allo-SCT. It is unclear so far whether a specific condition regimen or intensity may be able to modifiy the prognostic impact of pretransplant measurable residual disease on posttransplant outcome.

Methods: We conducted a retrospective single center analysis over a 12-year-period (2010-2022) in patients with AML in first or second complete remission (CR) who were allografted after Fludarabine/Treo conditioning. OS and RFS were estimated by Kaplan-Meier method and logrank tests were applied for univariable comparisons, while multiple Cox-regression models were used for multivariable analysis. MRD was evaluated within 4 weeks prior allo-SCT either by flow-cytometry, qRT-PCR or NGS to detect disease-specific markers. The study was approved by the ethics committee of the University Hospital Essen (approval number: 22-10708-BO) and all patients gave written informed consent for scientific use of their data.

Results: Overall 200 patients with a median age of 58 years (range, 21 to 77) were included and of these 154 (77%) were allografted in CR1, while 46 (23%) were in CR2. The median post-transplant follow-up was 42.5 months (range, 1-156). Overall, 103 pts (52%) had detectable MRD prior allo-SCT, while 97 pts (48%) were MRD-negative. Besides a higher amount of pts with adverse ELN genetic risk in the MRD-positive subgroup (MRD-pos: 44/59 (43%) vs. MRD-neg: 26/70 (27%), p=0.03) patient-, disease- and transplant related characteristics were evenly distributed between both groups. Posttransplant OS-, RFS-, CIR, and NRM-probability were 72%, 59%, 32% and 11% at 2 years for the entire cohort. In univariable analyses MRD-positivity prior to transplant, sAML compared to de novo AML and absence of cGVHD were associated with poor RFS and these parameters also retained their prognostic significance in multivariable analysis. Regarding OS sAML, absence of cGvHD and presence of cKT were identified as poor prognostic factors in univariable analysis, whereas MRD-positivity prior allo-SCT had no impact. Multivariable analysis revealed cKT and absence of cGvHD as independent parameters associated with poor OS.

Conclusions: Our analysis shows that Flu/Treo-based conditioning cannot eliminate the negative impact of pretransplant MRD positivity on RFS. However, MRD positivity did not have an impact on OS in our analysis, which may be explained in part by graft-derived alloreactivity implied by cGvHD.

Disclosure: H.C.R. received consulting and lecture fees from AbbVie, AstraZeneca, Roche, Janssen-Cilag, Novartis, Vertex and Merck. H.C.R. received research funding from Gilead and AstraZeneca. H.C.R. is a co-founder of CDL Therapeutics GmbH.

T.S. received consulting and lecture fees from BMS, Jazz Pharmaceuticals; T.S. received research funding from Jazz Pharmaceuticals; T.S. received travel support from Medac.

C.R. received consulting and lecture fees from BMS, Servier, Jazz Pharmaceuticals; C.R. received research funding from Jazz Pharmaceuticals; C.R. received travel support from Medac.

J.P., N.L., A.S., A.M., J.K. and R.T. have nothing to declare.

16: Conditioning Regimens

P171 FLUDARABINE TREOSULFAN REDUCED-INTENSITY CONDITIONING REGIMEN PRIOR HAPLOIDENTICAL HEMATOPOIETIC CELL TRANSPLANTATION WITH POST TRANSPLANTATION CYCLOPHOSPHAMIDE IN FRAIL/OLDER AML PATIENTS: PRELIMINARY RESULTS OF A SINGLE CENTER EXPERIENCE

Benjamin Bouchacourt1, Anne-Charlotte Le Floch1, Sabine Fürst1, Sylvain Garciaz1, Samia Harbi1, Yosr Hicheri1, Thomas Pagliardini1, Boris Calmels1, Faezeh Legrand1, Claude Lemarie1, Federico Pagnussat1, Christian Chabannon1, Pierre-Jean Weiller1, Marie-Anne Hospital1, Norbert Vey1, Didier Blaise1, Raynier Devillier 1

1Institut Paoli Calmettes, Marseille, France

Background: Haploidentical transplantation (haplo-SCT) after non-myeloablative conditioning (NMAC) regimen and post-transplantation cyclophosphamide (PT-Cy) dramatically extended the feasibility of allo-HSCT in frail and/or older patients with AML who do not have a HLA identical donor (PMID:26261255). However, in this patient population who usually carries high risk AML, incidence of relapse remains high, approximately 40%. Different reduced intensity conditioning (RIC) regimens have been develop as an attempt to reduce the risk of relapse as compared to NMAC regimens but they failed to improved survival in older patients due to a higher NRM that counterbalances the benefit in antileukemic effect (PMID:35752739). Treosulfan was recently approved in association with fludarabine (FT30) in RIC regimen based on a phase III study showing a benefit in OS compared to fludarabine busulfan due to a lower toxicity and NRM in the context of older AML or MDS patients undergoing HLA identical allo-HSCT (PMID:31606445). However, there is no reported data of the same FT30 RIC regimen in the context of haplo-SCT with PT-Cy. We thus started in 2022 a program with FT30 RIC regimen prior haplo-SCT in AML patients unfit for myeloablative regimen and report here the safety results of the first 20 patients.

Methods: We included 20 AML patients undergoing first peripheral blood haplo-SCT who were unfit for myeloablative regimen from March 2022 to September 2023. They received fludarabine (150mg/m²) treosulfan (30g/m²) and a standard GVHD prophylaxis based on PT-Cy, CSA and MMF.

Results: Median age was 60 years (range: 49-68). At diagnosis, ELN 2022 risk was favorable, intermediate and unfavorable for 3 (15%), 7 (35%) and 10 (50%) patients, respectively. All patients received intensive induction chemotherapy as initial treatment of AML. At the time of haplo-SCT, they were in CR1 except one in MLFS and one in CRi. 5/20 (25%) needed salvage therapy to reach CR1. MRD at the time of haplo-SCT was negative, positive and unknown for 12 (60%), 5 (25%) and 3 (15%) patients, respectively. HCT-CI was ≥ 3 in 11 patients (55%).

No hemorrhagic cystitis or hepatic SOS was observed. Grade 1 and 2 mucositis was observed in 15 (75%) 10 (50%) and 5 (25%) patients, without any grade 3 or 4. All patients achieved neutrophil (>0.5 G/L) and platelets (>20G/L) engraftment in a median time of 17 (range: 11-25) and 20 (range: 8-161) days, respectively. All patient achieved full donor blood T cell chimerism on day+30. 3 patients developed grade II acute GVHD, no grade III or IV was observed. With a median follow up of 385 days (range: 100-648), 6 (30%) patients developed moderate to severe chronic GVHD, 3 of them after prophylactic DLI.

One patients died from NRM on day+91 (acute pulmonary failure) and one patient relapse from AML on day+183 and subsequently died. Thus, both 1-year LFS and OS were 89%.

Conclusions: FT30 RIC regimen prior haplo-SCT with PT-Cy provides rapid full engraftment and low early toxicity in AML patients who are unfit for myeloablative regimens, with promising antileukemic effect that need to be evaluated in prospective comparison.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P172 PHARMACOKINETIC ANALYSIS OF TARGETED DOSING OF ANTITHYMOCYTE GLOBULIN IN ADULTS: A PROSPECTIVE ANALYSIS

Jishan Du 1, Haitao Wang1, Nan Wang1, Chao Ma1, Fei Li1, Lu Wang1, Liping Dou1, Daihong Liu1

1Chinese PLA General Hospital, Fifth Medical Center of PLA General Hospital, Beijing, China

Background: Anti-thymocyte globulin (ATG) is widely used in allogeneic hematopoietic stem cell transplantation to prevent severe graft-versus-host disease (GVHD) and graft failure. However, overexposure to ATG may increase the risk of viral reactivation and delayed immune reconstitution. The optimal dosing regimen of ATG is still uncertain. We identified the optimal total areas under the concentrationtime curve (AUC) range of active ATG(100 to 148.5 UE/mL/day) in haploidentical peripheral blood stem cell transplantation (haplo-PBSCT) and established a targeted dosing strategy based on ATG concentration monitoring that ensures engraftment, effectively prevents acute GVHD, and reduces the risk of virus reactivation. We will compare the prognosis of the optimal AUC group and non-optimal AUC groups after using the targeted dosing strategy to evaluate the effect of the ATG-targeted dosing strategy.

Methods: From November 2020 to March 2022, 80 patients with malignant hematological diseases who received haplo-PBSCT for the first time were enrolled. All patients received ATG-targeted dosing strategy that ATG was administered for 4 days (-5 days to -2 days) and the ATG doses on -3 days and -2 days were adjusted to achieve the optimal ATG exposure.

Results: Total AUC of active ATG of all patients were calculated. The AUC value of 60 patients(75%) fell into the optimal range, and 20 patients(25%) fell into the non-optimal range. The median time of neutrophil engraftment and platelet engraftment was similar in the optimal and non-optimal AUC groups: 12.5 versus 12.5(p= .762) and 13 versus 13(p= .604). Similar outcomes were observed for grade II to IV acute GVHD and moderate and severe cGVHD between the two groups: 51.7% (95%CI,39.8%-64.7%) versus 55.0% (95%CI, 35.3%-76.9%; p= .696) and 9.2% (95%CI, 3.9%-20.7%) versus 8.3% (95%CI, 1.2%-46.1%; p= .940). The cumulative incidence of cytomegalovirus (CMV) reactivation and EpsteinBarr virus (EBV) reactivation tended to decrease in the optimal AUC group: 30.0% (95%CI, 20.1%-43.3%) versus 50.0% (95%CI, 30.8%-72.9%; p= .199) and 58.5% (95%CI, 46.3%-71.0%) versus 70.0% (95%CI, 49.9%-87.7%; p= .436). Overall survival, relapse, non-relapse mortality, and disease-free survival demonstrated no significant differences between the two groups. A total of 102 haplo-PBSCT patients who received the conventional fixed ATG dose(cumulative 10 mg/kg) comprised historical control. The number of patients falling into the non-optimal range in targeted dosing group was less than that in historical control group: 25% versus 42.2%; p= .016, and the non-optimal AUC group in targeted dosing group showed a lower cumulative incidence of CMV than the non-optimal AUC group in historical control group. The cumulative incidence of EBV, overall survival, relapse, and disease-free survival tended to decrease in the non-optimal AUC group in targeted dosing group compared with the non-optimal AUC group in historical control group.

Conclusions: ATG-targeted dosing strategy makes more patients fall into the optimal total AUC range of active ATG, and patients in non-optimal AUC group in targeted dosing group can still benefit from the ATG-targeted dosing strategy. The cumulative incidence of CMV of patients in non-optimal AUC group in targeted dosing group is higher than that in the historical control group.

Clinical Trial Registry: The trial was registered at www.clinicaltrials.gov as #NCT04778618.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P173 THE COMBINATION OF FLUDARABINE, BUSULFAN AND ANTI-T LYMPHOCYTE GLOBULIN IS WELL TOLERATED AND OFFERS EFFECTIVE REDUCED INTENSITY CONDITIONING THERAPY FOR PATIENTS WITH MYELOID DISEASE

Chris Armstrong 1, Hayley Foy-Stones1, Nicola Gardiner1, Catherine Flynn1, Eibhlin Conneally1, Nina Orfali1

1St. James’ Hospital, Dublin, Ireland

Background: The optimal conditioning platform and dosing strategies for older and less fit patients with myeloid disease is unclear, and several options with varying alkylator and T cell depletion strategies are used internationally. The balance of toxicity and effect must be carefully considered in this heterogenous patient group, where harnessing the GVL effect is critical to success.

Methods: We retrospectively reviewed the cases of 175 patients who underwent RIC alloSCT, conditioned with fludarabine (180mg/m2), busulfan (8mg/kg) and ATLG (30-60mg/kg) from 2015-2022 in the National Adult Stem Cell Transplant Programme in Ireland.

Results: Patient characteristics are summarised in (Table 1). The cumulative incidence of NRM at 1 and 2 years was 10.4% and 11% respectively, with high-risk MF (HR:8.61, p=0.005), severe aGVHD (HR:5.29, p<0.001) and invasive fungal disease (HR:8.0, p<0.001) predicting a worse outcome (Figure). OS at 1 and 2 years was 80.34% and 72.35% respectively, with worse survival in CIBMTR-DRI high-risk disease (HR:1.74, p=0.03) and ABO mismatch (HR:2.34, p=0.003) and better survival after cGVHD (HR0.35, p=0.02). The cumulative incidence of relapse was 19.5% and 27% at 1 and 2 years respectively, with higher relapse risk in high-risk AML and MDS (HR:1.82/2.07, p=0.03), those who received a cryopreserved stem cell graft (HR:2.38, p=0.02) and those with higher graft T cell content (HR:2.16, p=0.002), with improved relapse risk in those who developed cGVHD (HR:0.40, p=0.02). RFS at 1 and 2 years was 69.95% and 61.73%. Relapse risk was significantly reduced in those with mixed donor chimerism who received a pre-emptive DLI (HR:0.33, p=0.01), and those who relapsed and received a therapeutic DLI experienced longer survival (HR:0.43, p=0.03).

In those aged 65-74 (n=27) there were no NRM events and 1 and 2 year OS was 92.5% and 75.9% as opposed to 78% and 71.4% in those <65. The cumulative incidence of relapse was higher in older patients at 34.5% at 2-years as compared to 25.5% in those <65. RFS however was superior at 81.3% and 67.34% at 1- and 2-years compared to 67.85% and 60.62% in those <65. All major forms of GVHD were less common in older patients. Grade III-IV acute GVHD occurred in 10% compared to 15% aged <65 and chronic GVHD in 11.5% compared to 17%. Late immune reconstitution at 6 and 12 months was less robust in older patients, with a 1 year median CD4 count of 127 x106/L in those >65 compared to 167 in those <65, and this effect was most prominent in those >70 years. Infectious morbidity was modest, with PTLD in 3.4% and CMV reactivation in 16.6% in the era of letermovir prophylaxis.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (24)

Conclusions: FluBu3 with ATLG demonstrates a robust safety profile, with low NRM rates, severe aGVHD and extensive chronic GVHD risk. With a 2-year RFS of over 60%, it similarly offers effective disease treatment for older patients with myeloid disease, particularly when combined with judicious use of DLI. The regimen is well suited to elderly patients over the age of 65, although improvements in relapse risk and immune reconstitution justify a trial of reduced ATLG dosing.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P174 COMPARING PTCY AND GIAC IN HAPLOIDENTICAL SETTINGS: UPDATED ANALYSIS FROM A NATIONWIDE TRANSPLANTATION REGISTRY

Yi-Wei Lee 1,2, Xavier Cheng-Hong Tsai2,3, Tzu-Ting Chen4, Tung-Liang Lin5, Yi-Chang Liu6, Chi-Cheng Li7, Ming Yao2,3, Bor-Sheng Ko2,3,8

1Taipei Medical University Hospital, Taipei, Taiwan, Province of China, 2National Taiwan University Hospital, Taipei, Taiwan, Province of China, 3National Taiwan University Cancer Center, Taipei, Taiwan, Province of China, 4China Medical University Hospital, Taichung, Taiwan, Province of China, 5Chang Gung Medical Foundation, Taoyuan, Taiwan, Province of China, 6Kaohsiung Medical University Hospital, Kaohsiung, Taiwan, Province of China, 7Buddhist Tzu-Chi General Hospital, Hualien, Taiwan, Province of China, 8National Taiwan University, Taipei, Taiwan, Province of China

Background: The rising popularity of haploidentical hematopoietic stem cell transplantation (haplo-HSCT) can be attributed to the development of practical and efficacious T-cell-replete haplo-HSCT protocols. Notably, the posttransplantation cyclophosphamide (PTCy) approach and the utilization of granulocyte colony-stimulating factor-primed bone marrow combined with peripheral blood stem cells (GIAC) have played pivotal roles in this evolving landscape. We have reported the comparison of PTCy and GIAC in 2019 and this time we aimed to update the analysis focusing on the patients with hematologic malignancies.

Methods: This retrospective, observational study utilized data from the Taiwan Blood and Marrow Transplantation Registry (TBMTR). The TBMTR oversees data collection from 17 collaborative transplantation centers across Taiwan, ensuring internal quality control for accuracy and consistency. Approval for data collection and analysis was obtained from the institutional review board of each participating hospital, aligning with the principles of the Declaration of Helsinki and requiring written informed consent. The study focused on patients who underwent haplo-HSCT between 2011 and 2022. The cohort included individuals who received the modified GIAC approach (mGIAC) or PTCy. Haplo-HSCT was defined as receiving stem cells from a parent, offspring, or sibling with at least two or more mismatched HLA loci.

Results: A total of 391 patients have been enrolled in this study, including mGIAC group (n= 188) and PTCy group (n= 203). The patients receiving mGIAC were younger than those receiving PTCy (median 44.1 vs. 51.2). All the grafts in the PTCy group were peripheral blood stem cells, while in the mGIAC group were from bone marrow and peripheral blood stem cells. The patients in the mGIAC group had more favorable platelet (median, 20 vs. 33 days, P < 0.001) and neutrophil engraftment kinetics (median, 12 vs. 17 days, P < 0.001). The patients within the PTCy group had a lower cumulative incidence rate of grade III–IV acute graft-versus-host-disease (GvHD) (15.6 vs. 47.6%, P < 0.001), as well as limited and extensive chronic GvHD (37.3% vs. 47.1%, P= 0.067) compared with those in the mGIAC group. With the median follow-up of 32 months, these two strategies conferred to similar outcomes in terms of 2-year overall survival (median 49.5 for mGIAC and 54.6% for PTCy, P= 0.163). Regarding the causes of death, the majority of patients died of infection (30% in the mGIAC group and 28% in the PTCy group), followed by disease relapse and GvHD in both group.

Conclusions: In this updated analysis, the patients within the mGIAC group exhibited more favorable engraftment kinetics. Meanwhile, patients undergoing PTCy demonstrated improved outcomes without compromising the incidence of acute or chronic GvHD, as compared to the previous analysis conducted in 2019. In conclusion, both haplo-HSCT approaches are feasible for patients with hematological malignancies in Taiwan.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P175 THE RESULTS OF ALLOGENEIC HSCT AFTER TREOSULFAN–BASED CONDITIONING REGIMEN IN CHILDREN WITH AML: MULTICENTER RETROSPECTIVE STUDY OF THE POLISH PEDIATRIC STUDY GROUP FOR HSCT

Agnieszka Sobkowiak-Sobierajska 1, Olga Zając-Spychała1, Maksymilian Deręgowski1, Monika Mielcarek-Siedziuk2, Krzysztof Kałwak2, Agnieszka Zaucha-Prażmo3, Katarzyna Drabko3, Robert Dębski4, Jan Styczyński4, Jolanta Goździk5, Jacek Wachowiak1

1Poznan University of Medical Sciences, Poznan, Poland, 2Wroclaw Medical University, Wrocław, Poland, 3Medical University of Lublin, Lublin, Poland, 4Collegium Medicum in Bydgoszcz, Nicolaus Copernicus University in Torun, Bydgoszcz, Poland, 5Collegium Medicum, Jagiellonian University, Kraków, Poland

Background: Allogenic HSCT is indicated in high risk pediatric AML patients in first CR and in all patients with relapsed disease. However, so far there was no study concerning outcomes of allo-HSCT after treosulfan-based preparative regimen exclusively in children suffering from AML.

Methods: We performed a retrospective analysis of 85 pediatric patients with AML who underwent first allogenic HSCT following treosulfan between July 2000 and December 2022 in Polish pediatric transplant centers. The majority of patients were transplanted in CR1 (n=64; 75,2%) and had Lansky or Karnofsky score >80% (n=76; 89,4%) at HSCT. Most children were transplanted from unrelated donor (n=62; 72,9%) – in this group peripheral blood stem cells (PBSC) were the main stem cell source (n=47; 75,8%). Patients transplanted from matched sibling donor (n=23; 24,2%) mostly received stem cells from the bone marrow (BM) (n=17; 73,9%). In the conditioning regimen treosulfan was combined with different cytostatics, mostly fludarabine (n=72; 84,7%) and cyclophosphamide (n=11; 12,9%).

Results: Engraftment rate was high, the incidence of complete donor-type chimerism was 93,5%. Early regimen related toxicity was low and mainly affected oral cavity and gastrointestinal tract. No SOS/VOD was diagnosed. Non-relapse mortality (NRM) was low at 5,9% and occurred mainly in patients with pre-HSCT risk factors of NRM. Five-year probability of overall (5y-pOS) and event-free survival (5y-pEFS) was 75,4%±5% and 71,3%±5,1%, respectively. No statistically significant difference between 5y-pOS and 5y-pEFS was found in patients who were transplanted in CR1 and CR2 as well as those who received HSCT from MSD and MUD. The cumulative incidence of relapse was 23,8% (n=20). Five-year probability of relapse-free survival was 76,9%±4,7% and did not correlate with remission status (CR1 vs CR2) and type of donor. There was no significant difference in outcome after treosulfan-fludarabine-thiotepa regimen versus other treosulfan-based regimens. Acute graft versus host disease (GvHD) grade II-IV incidence was 31,8%. Extensive chronic GvHD was diagnosed in 8,2% of patients. One patient developed secondary malignancy.

Conclusions: Treosulfan-based regimen seems to be safe and effective in pediatric AML patients and may be used as an alternative to busulfan-based protocols, especially in patients with pre-HSCT high risk factors of severe regimen-related toxicities.

Disclosure: We have no conflicts of interest to disclose.

16: Conditioning Regimens

P176 OUTCOME OF ALLOGENEIC STEM CELL TRANSPLANTATION IN AML PATIENTS AFTER CONDITIONING WITH FLUDARABINE/TOTAL BODY IRRADIATION(2GY) WITH OR WITHOUT ANTI-THYMOCYTE GLOBULINE OR POST-TRANSPLANT CYCLOPHOSPHAMIDE

Thomas Heinicke 1, Myriam Labopin2, Emmanuelle Polge3, Annoek EC Broers4, Gwendolyn van Gorkom5, Michel Schaap6, Jürgen Kuball7, Goda Choi8, Peter von dem Borne9, Gitte Olesen10, Henrik Sengeloev11, Uwe Platzbecker12, Giang Lam Vuong13, Alexandros Spyridonidis14, Bipin N. Savani15, Fabio Ciceri16, Mohamad Mohty17

1Otto-von-Guericke University, Magdeburg, Germany, 2EBMT Statistical Unit/Sorbonne University/Saint-Antoine Hospital, ‘Paris, France, 3EBMT Paris Study Unit, Paris, France, 4Erasmus MC Cancer Institute, Rotterdam, Netherlands, 5University Hospital Maastricht, Maastricht, Netherlands, 6Nijmegen Medical Centre, Nijmegen, Netherlands, 7University Medical Centre, Utrecht, Netherlands, 8University Medical Center Groningen, Groningen, Netherlands, 9Leiden University Medical Center, Leiden, Netherlands, 10Aarhus University Hospital, Aarhus, Denmark, 11Rigshospitalet, Copenhagen, Denmark, 12Medical Clinic and Policinic 1, Leipzig, Germany, 13Medizinische Klinik m. S. Hämatologie, Onkologie und Tumorimmunologie, Berlin, Germany, 14University of Patras, Patras, Greece, 15Vanderbilt University Medical Center, Nashville, United States, 16Ospedale San Raffaele s.r.l., Milano, Italy, 17Sorbonne University, Saint-Antoine Hospital, Paris, France

Background: Use of anti-thymocyte globuline (ATG) and post-transplant cyclophosphamide (PTCy) are two strategies for graft-versus host disease (GVHD) prophylaxis. On behalf of the Acute Leukemia Working Party of the EBMT the effectivenes of these aproaches in the context of the fludarabine/total-body irradiation 2Gy (FluTBI2Gy) regimen in AML patients in complete remission (CR) was compared.

Methods: We analyzed the outcome of adult AML patients (pt) in CR that had received their first allogneic stem cell transplantation between 2006 and 2022 after conditioning with either FluTBI2Gy or FluTBI2Gy with ATG (FluTBI2GyATG) or FluTBI2Gy (+/-Cy pre-transplant) with PTCy (Flu(Cy)TBI2GyPTCy) using peripheral blood stem cells from matched sibling or unrelated donors (UD). The primary endpoint was leukemia-free survival (LFS). Secondary endpoints were overall survival (OS), relapse incidence (RI), non-relapse mortality (NRM), acute (a)GVHD, chronic (c)GVHD, extensive cGVHD (ext.cGVHD) and refined graft-versus-host-disease-free, relapse-free survival (GRFS). Multivariate analysis was done using Cox regression. All endpoints were censored at 3 years(y).

Results: A total of 694 pt were identified (FluTBI2Gy, n=447, FluTBI2GyATG, n=102, Flu(Cy)TBI2Gy-PTCy, n=145). Median follow-up was 68.6 months, 63.5 months and 35.2 months for the FluTBI2Gy, FluTBI2GyATG and Flu(Cy)TBI2Gy-PTCy groups, respectively, p<.001. Median age was 61.8y, 64.9y and 62.2y, respectively, p<.001. Karnofsky performance score was below 90 in 30.9%, 20.6% and 39.3%, respectively, p=.007. Proportions of pt in CR1 were 81.0%, 73.5% and 90.3%, respectively, p=.002. Adverse cytogenetic risk was present in 14.8%, 27.5% and 22.8%, respectively, p=.003. FLT3-ITD and NPM1 mutations were found in 34.2%, 23.5% and 22.8%, respectively, p=.009, and 38.3%, 29.4% and 28.3%, respectively, p=.041. Donors were UD in 64.4%, 95.1% and 74.5% of cases, respectively, p<.001, of which 11.7%, 22.0% and 25.3% were mismatched (9/10), respectively. Pt CMV-serostatus was positive in 66.2%, 53.9% and 57.2%, respectively, p=.023. Engraftment ranged from 97% to 98.6% with no difference between groups. In univariate analysis 3y-LFS and OS were comparable for the FluTBI2Gy, FluTBI2GyATG and Flu(Cy)TBI2GyPTCy groups being 51.0%, 41.0% and 48.0%, respectively, p=.13, and 55.8%, 45.6% and 50.5%, respectively, p=.12. Similarly, RI, NRM and aGVHD were not significantly different between the three groups. In contrast, rates of 3y-cGVHD and ext.cGVHD were higher with FluTBI2Gy compared to FluTBI2GyATG and Flu(Cy)TBI2GyPTCy with 52.3%, 28.9% and 35.1%, respectively, p=.001, and 35.2%, 13.9% and 22.3%, respectively, p=.001. In multivariate analysis FluTBI2Gy was associated with a significantly increased risk for cGVHD compared to FluTBI2GyATG and Flu(Cy)TBI2GyPTCy with a hazard ratio (HR) and 95%-confidence interval [95%CI] of 0.5 [95%CI: 0.3-.83], p=.008, and 0.46 [95%CI: 0.31-.68], p<.001, respectively. In addition FluTBI2Gy was associated with an increased risk of ext.cGVHD compared to Flu(Cy)TBI2GyPTCy, HR=.44 [95%CI: 0.27-.72], p=.001. Furthermore, worse GRFS was associated with FluTBI2Gy compared to Flu(Cy)TBI2GyPTCy, HR=.7 [95%CI: 0.52-.94], p=.018. In contrast, no increased RI was associated with the use of either ATG or PTCy, HR=1.22 [.81-1.83], p=.34 and HR=1.03 [.7-1.52], p=.87, respectively.

Conclusions: In conclusion, in the present study compared to FluTBI2Gy improved outcome of alloSCT in AML patients in CR using FluTBI2GyATG and Flu(Cy)TBI2GyPTCy in terms of cGVHD, improved GRFS using Flu(Cy)TBI2GyPTCy but no differences in engraftment or RI were observed.

Disclosure: Thomas Heinicke received consultancy fee from Eurocept, honoraria from Eurocept and JAZZ and travel grants from Eurocept, JAZZ and Stemline. Lam Vuong received fees for advisory board from Gilead, speakers bureau from JAZZ and travel grants from Celgene, Gilead and Janssen.

16: Conditioning Regimens

P177 CONFIRMING BETTER OUTCOMES WITH TBI-BASED MYELOABLATIVE CONDITIONING FOR ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION EVEN IN SECOND COMPLETE REMISSION

Mario Delia 1, Vito Pier Gagliardi1, Corinne Contento2, Daniela Di Gennaro2, Olga Battisti2, Camilla Presicce2, Immacolata Attolico1, Paola Carluccio1, Francesco Albano1,2, Pellegrino Musto1,2

1Hematology and Stem Cell Transplantation Unit, Azienda Ospedaliero-Universitaria-Consorziale (AOUC) Policlinico, Bari, Italy, 2”Aldo Moro” University – School of Medicine, Bari, Italy

Background: The total body irradiation (TBI)-based myeloablative conditioning regimen (MCR) in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) is considered the standard backbone for conditioning in acute lymphoblastic leukaemia (ALL) patients. In particular, the TBI-driven overall survival (OS) benefit has been mostly demonstrated when allo-HSCT was performed in ALL-patients with diagnostic high-risk features and in complete remission (CR1-2) at allotransplant. Moreover, it’s also known that high dose TBI (12 Gy) might result in a significant improvement of relapse rates, although there are evidences in favour of low dose TBI as causing equivalent OS. Therefore, we retrospectively analysed data from high dose TBI-based allo-HSCT in order to investigate the TBI impact on ALL patients with CR≥2 (not CR1) if compared with CR1, being excluded a possible suboptimal TBI-dose. Moreover, to better explore the benefit by TBI in not CR1 patients we used an historical cohort of allotransplanted ALL-patients when TBI was not used.

Methods: Data from patients affected by ALL, who underwent all-HSCT from November 2009 to April 2019 were retrospectively collected, updating their follow-up on June 30th, 2023. All patients underwent MCR allo-HSCT in CR, which was defined by <0.1% blasts evaluated by cytometry at allotransplant. Fifty allotransplants were performed in ALL-patients (median age, 32 years, r 17-54); male sex in 60% (n=30), undergoing allotransplant in first CR (CR1) and TBI-based (12 Gy) MCR in 42% (n=21) and 44% (n=22), respectively. B, T and mixed lineage were described in 66% (n=33, PH-positive in 16 cases), 30% (n=15) and 4% (n=2), respectively. No differences were documented between patients who underwent TBI-based MCR and who did not with regard to sex, age, cell lineage, therapy before allotransplant, CR (CR1 vs not CR1) and type of donor.

Results: The median OS was not reached (NR) and of 194 days (d) in CR1- and not CR1-allotransplants (p=0.006), respectively. The 3-year OS in CR1-patients was of 57 and 55% (p=ns) with TBI- and not TBI-based MCR, respectively. Of note, the 3-year OS in not CR1-patients was of 41 and 16% with TBI- and not TBI-based MCR, respectively. The 3-year CI of relapse (CIR) with TBI- and not TBI-based MCR patients was of 34% and 70%, respectively. Multivariate analyses of factors (age at transplant, CR1 vs not CR1, donor, TBI, cell lineage, disease-risk) with impact on OS, relapse and TRM were performed. In particular, CR1 (HR=0.378; IC95%: 0.155-0.917, p= 0.032) and TBI-usage (HR=0.349; IC95%: 0.128-0.949, p=0.039) were the factors impacting on OS and relapse, respectively. No variables impacted TRM.

Conclusions: OS survival was mostly affected by disease status at allo-HSCT rather than by TBI. Of note, TBI-based conditioning was independently associated with relapse risk both in CR1 (12%) and in not CR1 (52%), thus confirming the importance of TBI role even in these patients’ setting, given the unacceptable risk of relapse in case of TBI-sparing approach in not CR1 allotransplants (80%).

Clinical Trial Registry: not applicable.

Disclosure: No conflicts of interest to declare.

16: Conditioning Regimens

P178 POST TRANSPLANT CYCLOPHOSPHAMIDE PLUS ABATACEPT LEADS TO DECREASED DAY + 30 CHIMERISM LEVELS

Peter Abdelmessieh 1, Henry Fung1, Yuliya Shestovska1, Michael Styler1, Khanal Rashmi1, Asya Varshavsky-Yanovsky1

1Fox Chase Cancer Center, Philadelphia, United States

Background: Allogeneic stem cell transplantation (alloSCT) is a curative treatment for patients with hematological malignancies.

Post-transplant CD3 chimerism represents a possible tool to predict disease recurrence. The predictive value of chimerism analysis, however, is controversial, with conflicting results on its utility.

GVHD prophylaxis with post-transplant cyclophosphamide (PTCY) has been established as an effective stagey. Our center adopted PTCY in 2014 as part of our standard graft vs. host disease (GVHD) prophylaxis for all alloSCT regardless of donor source.

With the FDA approval of the drug abatacept for GVHD prevention in late 2021 our center opted to add this therapy to PTCY starting in 2022. Along with this addition we also decreased to dose of PTCY from 50 mg/m2 on days +3/4 to 25 mg/m2 in the hopes of mitigating the infectious, cardiac and genitourinary complication seen with traditional PTCY.

PTCY has gained popularity by many centers over the past decade but few studies have looked at the effect of CD3 chimerism levels in relation to PFS and GVHD rates in patients receiving this method of T-cell depletion. No studies have reported the effect abatacept has on has early CD3 chimerism levels and its predictive value in regards to progression free survival and GVHD.

Methods: We retrospectively reviewed the charts of 172 consecutive patients who received alloSCT between 1/2017 and 5/2023 at the Fox Chase-Temple BMT Program. Patients either received PTCY at a dose of 50 mg/m2 on days 3/4, a PTCY dose of 25mg/m2 with abatacept 10mg/kg IV on day +5, +14 and +28, and a PTCY dose of 50mg/m2 with abatacept 10mg/kg IV on day +5, +14 and +28. Tacrolimus was started on day 5 on all patients and mycophenolate was started on patients that received unrelated donor grafts. We included patients that received both myeloablative and reduced intensity conditioning. Chimerism levels were measured in blood samples for CD3 on days +30.

Results: Seventeen percent (15/86) of patients PTCY at a dose of 50 mg/m2 on days 3/4 had a day 30 CD3 chimerism <90%. The abatacept and PTCY 25mg/m2 cohort showed 50% (13/26) of patients had a day 30 CD3 chimerism <90%, and 33% (4/12) patients in the PTCY 50mg/m2 cohort.

Patents getting the traditional PTCY dose and no abatacept resulted in similar PFS and GVHD rates regardless of day 30 CD3 chimerism levels with an overall 1 year PFS of 70% (68/99).

In patients getting abatacept and PTCY 25mg/m2 1 year PFS is 55% (11/22) and 75% (6/8) in PTCY 50mg/m2.

Conclusions: The addition of abatacept to PTCY resulted in a increased rate of low (<90%) early CD3 chimerism levels compared to standard PTCY and was seen both in the 25mg/m2 and the 50mg/m2 arm. This decrease in early CD3 chimerism has lead to a trend of inferior PFS however further statistical investigation is warranted as our patient number were too small to show a statistical difference.

Disclosure: Peter Abdelmessieh: Abbive, Sobi.

Aysa Varshavsky: pfizer, janseen, bms.

Henry Fung: astra zeneca, incyte, kite, janssen.

16: Conditioning Regimens

P179 HEMATOPOIETIC STEM CELL TRANSPLANT WITH REGULATORY AND CONVENTIONAL T CELL ADOPTIVE IMMUNOTHERAPY IN TP53-MUTATED/DELETED AML

Rebecca Sembenico1, Loredana Ruggeri1, Simonetta Saldi1, Sara Tricarico2, Antonella Mancusi1, Valerio Viglione1, Francesco Zorutti1, Tiziana Zei2, Roberta Iacucci Ostini2, Eleonora Cristofani1, Roberto Limongello1, Cristina Mecucci1, Andrea Velardi1, Massimo Fabrizio Martelli1, Cynthia Aristei1, Maria Paola Martelli1, Alessandra Carotti2, Antonio Pierini 1

1University of Perugia, Perugia, Italy, 2Perugia General Hospital, Perugia, Italy

Background: Hematopoietic stem cell transplantation is the only curative option for high-risk AML and AML/MDS. The prognosis of TP53-mutated/deleted AML and MDS/AML after HSCT is still unacceptably poor because of the high relapse rate (Middeke et al, 2016; Badar et al, 2023). These outcomes are even questioning the HSCT role in such an unfavorable setting. Irradiation-based T cell depleted HSCT with regulatory and conventional T cell adoptive immunotherapy and no post-transplant immunosuppression (Treg/Tcon allo-HSCT) reduced the relapse rate in high-risk AML patients (Pierini et al, 2021). This study aimed to evaluate the efficacy of Treg/Tcon allo-HSCT in TP53-mutated/deleted AML.

Methods: We retrospectively collected data of 75 patients affected by AML or MDS/AML (>10% leukemic blasts) with mutation or deletion of TP53 in the last twenty years at our Center. TP53 involvement was evaluated by Sanger or next generation sequencing (NGS) and/or FISH.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (25)

Results: Figure 1. Survival outcomes in patients treated with a curative intent. OS comparing patients who received Treg/Tcon allo-HSCT (green line) and who did not receive HSCT (blue line).

Thirty-eight/75 patients (median age at diagnosis: 64 years) were considered eligible for a treatment with curative intent (age<72 years, no severe comorbidities), potentially followed by a HSCT. Nine/38 patients received Treg/Tcon allo-HSCT and no pharmacologic GVHD prophylaxis after the induction therapy, while twenty-nine/38 patients were considered unfit for HSCT or died of progression of disease before a HSCT could be performed. Two patients were transplanted in first complete remission, while seven patients were transplanted with persistent disease (median blast count at transplant 10%, range 0-80%). At the time of analysis, 8 patients (4 received Treg/Tcon allo-HSCT, 4 received only standard therapy and no Treg/Tcon allo-HSCT) are alive with a median follow up of 30.5 months. At 5 years, in the Treg/Tcon allo-HSCT group, 1 patient died of relapse while 3 patients died of NRM. NGS analysis after transplant showed clearance of TP53 mutated clones in 3 surviving patients, while we transiently detected a newly acquired TP53 mutated clone that was not present at diagnosis in one patient, despite the patient remained in complete remission with full donor chimerism. Such event suggests a control of the mutated clone by donor immunity over time. Overall survival at 5 years in patients who received Treg/Tcon allo-HSCT and in those who received only standard therapy was 55.6% and 13.8% (p 0.017) respectively (See Figure).

Conclusions: Prognosis of TP53-mutated patients is very poor. Outcomes of patients who did not receive allo-HSCT were dismal. Indeed, relapse or progression of disease occurred almost always and were the major cause of death. On the other hand, Treg/Tcon allo-HSCT improved survival in a small cohort of patients despite most of them being transplanted with active disease suggesting such approach exerts a powerful antileukemic activity. Limitations of our analysis include the low number of patients and the monocentric experience. Future studies are needed to evaluate efficacy of Treg/Tcon allo-HSCT in TP53 mutated/deleted AML patients that are not in remission at transplant.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P180 LOMUSTINE, ETOPOSIDE, CYTARABINE, MELPHALAN (LEAM) PROTOCOL AS CONDITIONING REGIMEN BEFORE AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION (ASCT) IN PATIENTS WITH LYMPHOMAS: A SINGLE CENTER EXPERIENCE

Željko Prka 1, Anamarija Vrkljan Vuk1, David Čičić1, Ena Sorić1, Zdravko Mitrović1,2, Marko Lucijanić1,2, Marija Ivić Čikara1, Željko Jonjić1, Mario Piršić1, Vlatko Pejša1,2, Ozren Jakšić1,2

1Clinical Hospital Dubrava, Zagreb, Croatia, 2University of Zagreb School of Medicine, Zagreb, Croatia

Background: Intensive (myeloablative) therapy before transplantation of autologous hematopoetic stem cells is still important therapeutic option for patients with non-Hodgkin’s (NHL) and Hodgkin’s lymphoma (HL). The most used protocol is the combination of carmustine, etoposide, cytarabine and melphalan (BEAM). However, a few years ago, due to availability problems and a significant increase in price, in our center, we started conditioning according to the LEAM protocol, in which carmustine was replaced by lomustine.

Our objective is to present the results of conditioning with the LEAM protocol in transplanted patients with Non Hodgkin’s lymphoma and Hodgkins lymphoma and compare them with a cohort of patients conditioned with other used protocols.

Methods: 127 patients were retrospectively analyzed in the period from 06/2007 to 08/2023. Of those patients, 61 received BEAM as myeloablative regimen, 36 LEAM, 19 BeEAM, 8 TEAM and 3 patients LACE protocol.

In 58% of our patients, ASCT was conducted during the first remission (as frontline consolidation treatment after high dose therapy).

Median follow-up was 87 months. The median age of the three most frequently used protocols (BEAM/LEAM/BeEAM) was 51 (19-68), 57 (18-69) and 53 (21-64) years. Of the 127 patients, 111 had NHL, and 16 had HL. The most common histological diagnosis was diffuse large B-cell lymphoma (DLBCL) (48 patients).

Results: The median survival of the entire patient cohort was nearly 14 years. Overall 4 year survival for the three most used protocols was as follows: LEAM 86%, BEAM 75%, BeEAM 47%. There is no statistically significant difference between the BEAM and LEAM protocols, although the survival curve shows a trend in the favor of LEAM. Both LEAM and BEAM are significantly better than BeEAM (p<0.0001 for both protocols). The frequency of infectious complications, including those with a fatal outcome, was the highest with the BeEAM protocol. There was no difference in toxicity between the BEAM and LEAM protocols.

BEAM

LACE

BeEAM

TEAM

LEAM

ALL

AGE (median)

51 (19-68)

63 (30-63)

53 (21-64)

51.5 (42-65)

57 (18-69)

53 (19-69)

SEX F

44

33

47

50

31

41

DIAGNOSIS

HL

8

1

3

4

16

NHL

53

2

16

8

32

111

DLBCL

19

10

2

17

48

MCL

6

1

2

2

11

OTHER

23

4

10

37

CNS

6

6

NONSPEC

5

1

3

9

FOLLOW UP

139.1

126.5

77.9

4.6

20.3

87.3

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (26)

Conclusions: According to the experience and results of our center, LEAM is equivalent to BEAM in terms of efficiency and toxicity, and an equivalent option of myeloablation therapy before ASCT in patients with lymphomas.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P181 SAFETY AND EFFICACY OF BUSULFAN 9.6 BASED RIC (AUGMENTED RIC) IN OLDER PATIENTS WITH AML WHO ARE NOT SUITABLE FOR MAC

Kalina Abrol 1, Ram Vasudevan Nampoothiri1, Michael Kennah1, Natasha Kekre1, David Allan1, Harold Atkins1, Lothar Huebsch1, Trang Doan1, Carolina Cieniak1, Sheryl McDiarmid1, Tim Ramsay1, Christopher Bredeson1, Ashish Narayan Masurekar1

1The Ottawa Hospital, Ottawa, Canada

Background: AML patients undergoing allo-HCT, who are not suitable to receive MAC get augmented RIC at our center. Augmented RIC and MAC regimens contain intravenous Busulfan 9.6 mg/kg (BU9.6) and 12.8 mg/kg (BU12.8) respectively. Hypothesis: Replacing RIC with BU9.6 would reduce relapse without increasing toxicity in this poor risk patient group and lead to outcomes that are similar to BU12.8.

Methods: We retrospectively compared outcomes in AML patients given BU9.6 and BU12.8 between January 2019 and December 2022 at The Ottawa Hospital. Baseline continuous and categorical variables in were compared using Mann-Whitney U test and Chi-Square test respectively. Survival analysis was performed by Kaplan-Meier plots. Multivariate analysis (MVA) was performed using Cox proportional hazard model adjusting for baseline variables and those influencing outcome in univariate analysis. Cumulative incidence of relapse, NRM, d180-Grade III/IV aGVHD and cGVHD requiring systemic therapy were calculated using death as a competing risk and compared using Gray’s test.

Results: BU9.6 and BU12.8 groups had 31 (22%) and 108 (78%) patients respectively. Out of 139 patients, 137 (99%) received PBSC. Everyone except matched related donor allo-HCT in BU9.6 received low dose ATG 2.5 mg/kg. Ninety-eight percent of patients engrafted. Median follow up was 19 months. Median age at allo-HCT was 59 years (range:19-73). Table 1 shows baseline characteristics. Patients in BU9.6 were older compared to BU12.8, despite the latter group having 35/108 (33%) patients who were ≥ 60 years of age. Of the 139 patients, 128 (92%) had HCT-CI ≤ 3.

Two-year overall survival (OS) in BU9.6 was lower compared to BU12.8: 52% [95%CI: 33-70] vs 70% [95%CI: 61-80], p =0.021 whilst two-year RFS and two-year GRFS were comparable. Univariate analysis demonstrated OS was influenced by sex and disease risk-index (DRI), RFS by, sex and DRI and GRFS by sex, HCT-CI and KPS. Cox MVA showed BU12.8 was superior to BU9.6 for OS [BU9.6 HR = 3.1 (95%CI 1.4-6.8), p = 0.005] and RFS [BU9.6 HR = 2.4 (95%CI: 1.2-4.8), p = 0.016] but for GRFS, the difference between the two groups was not statistically different [BU9.6 HR 1.8 (95%CI: 0.95-3.20, p = 0.073]. DRI-HR independently affected OS [HR = 2.27 (95%CI:1.2-4.3), p = 0.01] and PFS [HR = 1.92(95%CI:1.1-3.4), p = 0.001.

Cumulative incidence of relapse, NRM, aGVHD III/IV and cGVHD requiring systemic therapy did not show difference between BU9.6 and B12.8 regimens. Two-Year NRM in BU9.6 was 20% [95%CI: 9-40]

Baseline Characteristics

BU9.6 (N=31)

BU12.8 (N=108)

Age – median (range)

67 (47-73)

56 (19-69)

P=0.0001

Sex – male (%)

22 (71.0)

55 (50.9)

P=0.0735

Co-morbidities

10 (32.3)

46 (42.6)

P=0.2065

1-2

17 (54.8)

58 (53.7)

≥3

4 (12.9)

7 (6.5)

KPS <=90 (%)

26 (83.9)

72 (66.7)

P=0.0957

HLA Match (%)

Matched Related Donor

4 (12.9)

19 (17.6)

P=0.9707

Matched Unrelated Donor

24 (77.4)

71 (65.7)

Mismatched Unrelated Donor

3 (9.7)

18 (16.7)

ABO Matching (%)

Match

17 (54.8)

64 (59.3)

P=0.8454

Major and/or Minor

14 (45.2)

44 (40.7)

Product was cryopreserved (%)

24 (77.4)

58 (53.7)

P=0.0277

ELN Risk Classification (%)

Favorable

4 (12.9)

21 (19.4)

P=0.9070

Intermediate

14 (45.2)

51 (47.2)

Adverse

13 (41.9)

36 (33.3)

Disease Risk Index (DRI) (%)

Low

1 (3.2)

6 (5.6)

P=0.2774

Intermediate

28 (90.3)

87 (80.6)

High

2 (6.5)

15 (13.9)

Disease Status at Transplant

CR1 or higher

31 (100.0)

107 (99.1)

p>0.99

Conclusions: Augmented RIC using BU9.6 was found to be safe and effective in older patients with AML who are not suitable candidates for MAC. BU9.6 was associated with a NRM of 20%. The GRFS and CI of relapse were comparable with BU12.8 in our study with a median follow up of 19 months. Longer follow up and comparison of BU9.6 against existing datasets of BU6.4 based RIC are needed to understand the value of augmented RIC regimens. We confirm the value of DRI as an independent marker for OS and RFS in AML patients.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P182 TOTAL MARROW AND LYMPHOID IRRADIATION (TMLI) IN COMBINATION WITH CYCLOPHOSPHAMIDE AND ETOPOSIDE IMPROVES THE OUTCOME OF PATIENTS WITH POOR-RISK ACUTE LEUKEMIA

Anthony Stein 1, Monzr Al Malki1, Yan Wang1, Joycelynne Palmer1, Ibrahim Aldoss1, Haris Ali1, Ahmed Aribi1, Andrew Artz1, Brian Ball1, Len Farol1,2, An Liu1, Guido Marcucci1, Ryotaro Nakamura1, Vinod Pullarkat1, Eric Radany1, Firoozeh Sahebi1,2, Amandeep Salhotra1, Karamjeet Sandhu1, Eileen Smith1, Ricardo Spielberger1,2, Susanta Hui1, Stephen Forman1, Jeffrey Wong1

1City of Hope, Duarte, United States, 2Southern California Permanente Medical Group, Los Angeles, United States

Background: Total marrow and lymphoid irradiation (TMLI) delivers targeted doses of radiation to the bone marrow while maintaining low doses to vital organs. We have conducted a phase II study in which we combined TMLI (2000cGy to targets, 1200cGy to liver/brain) with cyclophosphamide (Cy) and etoposide (VP16) as a conditioning regimen to evaluate its anti-leukemia activity and safety/tolerability in patients with relapsed/refractory acute leukemia. We report here on 74 subjects (AML n=56, ALL n=18) treated on study during 05/2014-09/2020.

Methods: TMLI was administered on days -9 to day -5, VP16 60mg/kg (adjusted body weight) on day -4, and Cy 100mg/kg (ideal body weight) on day -2. Bone marrow (n=7) or peripheral blood stem cells (n=67) were given on day 0. Graft versus host disease (GVHD) prophylaxis was tacrolimus and sirolimus. The primary endpoint was progression free survival (PFS), and secondary endpoints included overall survival (OS), non-relapse mortality (NRM), and toxicity.

Results: Eighty-eight percent (49/56) in AML and 94% (17/18) in ALL experienced ANC recovery at a median of 18 (range: 10-192) and 18 (11-42) days post-HCT. Engraftment occurred for all subjects who achieved CR/CRi at day +30. The d30 CR/CRi rate was 52/56 (93%) in AML and 16/18 (89%) in ALL.

With a median follow-up of 1.8 (range: 0.1-5.9) years, the two-year PFS and OS estimates were 34% (95%CI: 22-46%) and 48% (35-61%) in AML, and 22% (7-43%) and 39% (18-60%) in ALL. The cumulative incidence of disease relapse/progression at 1-year and 2-year were 46% (95%CI: 33-59%) and 59% (45-71%) in AML, and 33% (13-55%) and 50% (24-71%) in ALL. The cumulative incidence of NRM at d100 and year 1 were 4% (95%CI: 0.7-11%) and 4% (0.7-11%) in AML, and 6% (0.3-23%) and 22% (7-44%) in ALL. Disease relapse/progression occurred in 50 (68%) subjects post-transplantation. Thirteen subjects died in remission because of chronic GVHD and/or infection.

From conditioning start to d30, the Grade≥2 Bearman toxicities were stomatitis (Gr2 n=17, Gr3 n=5), gastrointestinal (Gr2 n=10, Gr3 n=4), renal (Gr2 n=1, Gr3 n=5), hepatic (Gr2 n=3, Gr3 n=2), central nervous system (Gr2 n=3, Gr3 n=1), bladder (Gr2 n=4), pulmonary (Gr2 n=1), and cardiac (Gr2 n=1). Fifty-one percent (37/72) who were evaluable for acute GVHD experienced aGVHD. The cumulative incidence of grade III/IV aGVHD at day+100 was 12% (95%CI: 6-21%). The incidence of cGVHD was 47% (32/68), including 21% (14/68) with extensive cGVHD.

As exploratory analyses, we evaluated the associations between PFS and 5 key baseline factors in AML. By fitting 5 univariate Cox proportional hazards models, we found that peripheral blasts ≥20% at baseline, every additional prior regimen, and venetoclax use at one line before transplant were associated with higher hazard of either disease relapse/progression or death at any given time post-transplant (Table). After adjusting for venetoclax use and number of prior regimens, peripheral blasts ≥20% at baseline was associated with higher hazard of disease relapse/progression or death.

Variables

Overall1, N= 74

AML1, n= 56

ALL1, n= 18

Age at transplantation (years)

40.1 (16.5 - 59.3)

41.9 (18.8 - 59.3)

31.5 (16.5 - 56.8)

Total number of all prior drug regimens

3 (1 - 9)

3 (1 - 8)

4 (2 - 9)

Induction failure prior to conditioning2

51 (69)

42 (75)

9 (50)

Bone marrow blasts at baseline (%)3

20 (0 - 95)

20 (1 - 95)

18 (0 - 95)

Peripheral blasts at baseline (%)

0 (0 - 86)

1 (0 - 86)

0 (0 - 49)

Cytogenetic risk at baseline

Adverse4/Unfavorable5; Intermediate

48 (65); 26 (35)

35 (62); 21 (38)

13 (72); 5 (28)

Donor type

Sibling; Matched unrelated; Mismatched unrelated

35 (47); 20 (27); 19 (26)

26 (46); 15 (27); 15 (27)

9 (50); 5 (28); 4 (22)

Venetoclax use at one line before transplantation

13 (18)

11 (20)

2 (11)

  1. 1. Median (Range) or N (%); 2. Other disease status prior to conditioning: 1st relapse, 2nd relapse, 3rd or higher relapse, and 3rd remission; 3. One subject had inadequate sample for bone marrow blasts evaluation; 4. AML Cytogenetic Risk (ELN 2017); 5. ALL Cytogenetic Risk (SWOG).

Variables

Group

N

# of events

Univariate Cox proportional hazards models among AML (n=56)

Multivariable Cox proportional hazards models among AML (n=56)

Crude Hazard Ratio (95% CI)

p-value

Adjusted Hazard Ratio (95% CI)

p-value

Cytogenetic Risk per ELN 2017

Intermediate

21

17

1 (Reference)

-

Adverse

35

30

1.0 (0.6, 1.9)

0.918

Peripheral blasts at baseline

<20%

41

33

1 (Ref.)

-

1 (Ref.)

-

≥20%

15

14

2.1 (1.1, 3.9)

0.026

2.4 (1.2, 4.8)

0.01

Bone marrow blasts at baseline

<25%

29

23

1 (Ref.)

-

≥25%

26

23

1.5 (0.8, 2.6)

0.205

Total number of prior regimens

Every additional prior regimen

1.2 (1.0, 1.5)

0.032

1.1 (0.9, 1.4)

0.246

Venetoclax use at one line before transplant

No

45

37

1 (Ref.)

-

1 (Ref.)

-

Yes

11

10

2.0 (1.0, 4.1)

0.058

2.1 (0.9, 5.0)

0.084

Conclusions: 1) TMLI can be safely delivered in combination with VP16 and Cy.

2) The regimen is effective for patients with relapsed/refractory acute leukemia.

Clinical Trial Registry: clinicaltrials.gov, NCT02094794.

Disclosure: Stein: Sanofi: Current Employment, Current holder of stock options.

16: Conditioning Regimens

P183 TREOSULFAN-FLUDARABINE CONDITIONING REGIMEN WITH POST-TRANSPLANT HIGH-DOSE CYCLOPHOSPHAMIDE: A RETROSPECTIVE ANALYSIS

Aura Arola 1, Lotta Tapana2, Maija Itälä-Remes1

1Turku University Hospital and University of Turku, Turku, Finland, 2Auria Clinical Informatics, Turku, Finland

Background: In allogeneic hematopoietic stem cell transplantations (alloHSCT), toxicity related to conditioning regimen is a significant factor in transplantation-related mortality (TRM). Reduced intensity conditioning (RIC), on the other side, may increase the risk of relapse. Treosulfan is an alkylating agent used in conditioning, with myeloablative, immunosuppressive and antileukemic effects but minor non-hematological toxicity. Graft-versus-host disease (GVHD) prophylaxis with post-transplant cyclophosphamide (PTCy) is widely accepted in the setting of haploidentical donors, but the usage has also been extended to other donor types.

Methods: In this single center study, we evaluated retrospectively the safety and efficacy of treosulfan-based RIC regimen combined with PTCy. All our patients received treosulfan 10 mg/m2 iv for three days and fludarabine 30 mg/m2 iv for five days followed by PTCy 50 mg/kg on days 3 and 4 after alloHSCT. In combination with PTCy, short courses of tacrolimus or everolimus and mycophenolate mofetil (MMF) were used as GVHD prophylaxis.

Results: Between April 2018 and July 2022, 97 patients received treosulfan-fludarabine conditioning regimen combined with PTCy. Median follow-up time was 27 (range 1-59) months. Patient, disease, and transplantation characteristics are presented in Table 1. Median age of the patients was 62 (range 17-75) years. MDS and AML were the most common diagnoses (28.9 % and 26.8 %, respectively). Almost half of the patients were not in complete remission (CR) at transplant, and the disease risk index (DRI) was high or very high in 52.6 % of patients. Stem cell source was peripheral blood in all patients. The 1-year and 2-year OS was 88 % and 80 %, respectively, and relapse-free survival (RFS) 78 % and 72 %, respectively, while the cumulative incidence of relapse was 22 % and 28 %, respectively. Transplantation-related mortality (TRM) at one and two years was 5.3 % and 6.6 %, respectively. Causes of death were relapse (n= 17), GVHD (n= 2), infection (n= 3) and organ failure/toxicity (n= 1). There was one primary graft failure and one early rejection, both occurring in patients with myelofibrosis, and both having been rescued by the second allograft.

Table 1. Patient, disease and transplantation characteristics

Characteristic

All patients (n= 97)

Median age, years (range)

62 (17-75)

Median time from diagnosis to alloHSCT, months (range)

8 (2-215)

Diagnosis, n (%)

MDS

28 (28.9)

AML

26 (26.8)

ALL

3 (3.1)

MDS/MPN

7 (7.2)

MF

7 (7.2)

Lymphoma

15 (15.5)

Other

11 (11.3)

Disease status before alloHSCT, n (%)

Active disease

45 (46.4)

CR

52 (53.6)

Minimal residual disease (MRD) status before alloHSCT, n (%)

Positive

53 (54.6)

Negative

19 (19.6)

Not applicable

25 (25.8)

Donor type, n (%)

MUD

25 (25.8)

MRD

7 (7.2)

MMUD

2 (2.1)

Haploidentical

63 (64.9)

Median follow-up time, months (range)

27 (1-59)

Conclusions: Treosulfan-fludarabine conditioning regimen combined with PTCy had low transplantation-related mortality and acceptable relapse rate, especially taking into account the relatively high age of the patients, high or very high DRI in more than half of the patients, and the high proportion of patients with active disease and/or positive MRD at transplant.

Disclosure: Aura Arola: Medac.

Lotta Tapana: Nothing to declare.

Maija Itälä-Remes: Abbvie, Amgen, Medac, Novartis, Pfizer.

16: Conditioning Regimens

P184 CONDITIONING WITH I.V. BUSULFAN, ONCE VERSUS FOUR TIMES DAILY IN ADULTS: IMPACT ON PHARMACOKINETICS, ORGAN TOXICITIES AND SURVIVAL AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Claire Seydoux1, Michael Medinger1, Joerg Halter1, Dominik Heim1, Katharina M. Rentsch1, Jakob R. Passweg 1

1University Hospital of Basel, Basel, Switzerland

Background: Therapeutic drug monitoring of busulfan (Bu) used as myeloablative conditioning regimen with fludarabine or cyclophosphamide for allogeneic hematopoietic stem cell transplantation (allo-HSCT) is recommended to limit toxicity as pharmacokinetics (PK) displays great variability. Since 2019, Bu is given once a day (Bu1) instead of four times a day (Bu4) in Basel, with same 24h dose compared to 4 times daily used in previous years. The international guidelines for an optimal area-under-the-curve (AUC) for Bu1 and Bu4 remain controversial and have been extrapolated from the pediatric population but treatment tolerability differs in adults and few studies analyzed long-term outcomes.

Methods: The aim of this retrospective study was to analyze the correlation between Bu administration (Bu1 vs Bu4), Bu-PK and clinical outcome in 259 adult patients receiving Bu for allo-HSCT.

Results: Patients receiving Bu4 had slightly lower BMI (median BMI 24 vs 26 kg/m2; p=0.03) and were younger (median age 50 vs 54 years-old; p=0.13) than Bu1; 83% of Bu1 received in addition cyclophosphamide and 17% fludarabine, whereas patients in the Bu4 cohort received fludarabine in 85% and cyclophosphamide in 15% (p<0.01). Bu4 showed lower 24h AUC values (median of 3808) than Bu1 (4197 micromol/l*min, p=0.04), they stayed longer in the hospital than Bu1 (34 vs 31 days, p<0.01), showed a higher incidence of bacterial infections (47% vs 34%, p=0.04) and a tendency of more fungal infections (20% vs 11%; p=0.06) and renal toxicity (20% vs 9%; p=0.06). Bu1 resulted in grade ≥ 3 mucositis in 60% vs 37% in Bu4 (p<0.01). As for long-term outcomes, Bu1 showed a tendency of lower survival (Figure 1, p=0.07) with same treatment-related (TRM), relapse mortality and GvHD cumulative incidence (slightly lower cumulative incidences for all, not significant). Regarding PK, 135/259 (52%) of all patients fell within the FDA target range on day 1, 93 (36%) were under target and 31 (12%) were above the target. Patients with BMI >30kg/m2 and age > 60 years had significantly higher Bu-PK values (p<0.01 for both). Higher AUC values showed higher prevalence of renal and pulmonary toxicity (p=0.03 and <0.01, respectively), more infectious complications and particularly viral infections (p<0.01), with overall higher TRM incidence in patients with AUC > 3600 micromol/l*min (p=0.01). The optimal 24h AUC cut-off rate associated with the highest overall survival (OS) was 3966 micromol/l*min in Bu4 and 4180 micromol/l*min in Bu1.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (27)

Conclusions: As a conclusion, Bu1 seems to benefit the patients as compared to Bu4 in term of organ toxicities and long-term outcomes. A lower target AUC threshold than the one described in the literature may benefit the adult patients at long-term, irrespective of Bu administration.

Disclosure: No conflict of interest.

16: Conditioning Regimens

P185 NON MYELOABLATIVE VERSUS REDUCED INTENSITY CONDITIONING IN ELDERLY PATIENTS WITH PTCY. A MULTICENTRE RETROSPECTIVE COHORT ANALYSIS FROM GATMO-TC

Mariano Berro 1, Jorge Arbelbide2, Ana Lisa Basquiera3, Adriana Vitriu4, Silvina Palmer5, Martin Saslavsky6, Patricio Duarte7, Sebastian Yantorno8, Martin Castro9, Ruben Burgos10, Gabriela Sturich3, Gonzalo Bentolila11, Sol Jarchum12, Juan Real13, Gonzalo Ferini2, Georgina Bendek2, Maria M. Rivas1, Gustavo Kusminsky1

1Hospital Universitario Austral, Pilar, Argentina, 2Hospital Italiano de Buenos Aires, Buenos Aires, Argentina, 3Hospital Privado de Cordoba, Cordoba, Argentina, 4Instituto Alexander Fleming, Buenos Aires, Argentina, 5Hospital Británico, Buenos Aires, Argentina, 6CETRAMOR, Rosario, Argentina, 7CEMIC, Buenos Aires, Argentina, 8Hospital Italiano La Plata, La Plata, Argentina, 9Sanatorio Anchorena, Buenos Aires, Argentina, 10Clinica Conciencia, Neuquen, Argentina, 11FUNDALEU, Buenos Aires, Argentina, 12Sanatorio Allende, Cordoba, Argentina, 13Hospital Aleman, Buenos Aires, Argentina

Background: Reduced Intensity Conditioning (RIC) is associated with higher mortality but lower relapse incidence (RI) compared to Non-Myeloablative (NMA) conditioning. Post transplant Cyclophosphamide (PTCy), although not myeloablative, confers a higher transplant toxicity. The optimal conditioning for patients not candidates for myeloablation receiving PTCy is not clear. We aim to compare NMA vs. RIC in elderly patients transplanted for haematological malignancies with PTCy. Main objectives were GVHD incidence and survival outcomes.

Methods: We performed a retrospective cohort analysis of elderly patients (≥60 years o ≥55 years with comorbidities) who received an AlloSCT from 01/2015 till 12/2023 for haematological malignancies in Argentina. Lymphoproliferative diseases were excluded. Statistical analysis were performed with Easy R 1.33. Non-relapse Mortality (NRM), RI, acute and chronic graft-vs.-host disease (GVHD) were calculated with Gray´s test; Overall Survival (OS) and GVHD-Relapse free survival (GRFS) with log-rank. Match pair analysis was performed in a 2:1 ratio (RIC:NMA) with age (≥60 years) and HCT-CI (high risk).

Results: A total of 120 patients were included, with a median follow up time of 1.7 years. Peripheral blood stem cell was the source in 97% of the patients, and GVHD prophylaxis was PTCy, Mycophenolate and calcineurin inhibitor in the full cohort. Main disease was AML/MDS in 81% (N=97), 77% (N=93) received an haploidentical donor, 1 match sibling and 22% Unrelated donor (N=26; 20 match, 6 mismatch). Thirty-two percent of the patients received a NMA conditioning (N=38) compared to 68% with a RIC (N=82, 47 Flu-Bu and 35 Flu-Mel). Acute GVHD GII-IV incidence was similar (6 months NMA 13.2% vs. RIC 26%, p=0.14), with a significant increase in aGVHD GIII-IV incidence with RIC (6 months 11% vs 0%, p=0.03) and similar chronic GVHD. A non-significant increase in NRM was observed with RIC (100 days and 2 years 15% and 34% vs 13% and 21%, p=0.14), with similar relapse incidence (2 years 25% vs 42%, p=0.21). No significant differences were observed in OS (2 year 43% vs. 43%, p=0.68) and GRFS (2 years 30% vs. 33%, p=0.32). After a match pair analysis, RIC (N=60) was associated with a trend towards a higher NRM (2 years 35% vs. 17%, p=0.07), a lower OS (2 years 41% vs. 55%, p=0.07) and a significantly lower GRFS (2 years 21% vs 40%, p=0.01) compared to NMA (N=30).

Conclusions: In patients receiving PTCy, severe acute GVHD was higher after RIC compared to NMA, with similar survival outcomes. In a match pair analysis, a possible association with worst survival was observed for RIC. We propose a Latin American retrospective cohort analysis to explore these conclusions.

Clinical Trial Registry: not registered.

Disclosure: no CI to disclose.

16: Conditioning Regimens

P186 IMPACT OF TRANSPLANT CONDITIONING INTENSITY ON BOTH ACUTE AND CHRONIC GVHD IN HAPLO-PTCY TRANSPLANT: TWO GITMO CENTRES’ EXPERIENCE

Umberto Pizzano1,2, Simona Piemontese3, Gabriele Facchin1, Raffaella Greco3, Marta Lisa Battista1, Jacopo Peccatori3, Renato Fanin1,2, Fabio Ciceri3,4, Francesca Patriarca1,2, Maria Teresa Lupo-Stanghellini 3

1Hematology and Stem Cell Transplantation Unit, University Hospital ASUFC, Udine, Italy, 2Department of Medical Area (DAME), Udine, Italy, 3Hematology and Bone Marrow Transplantation Unit, San Raffaele Scientific University Institute, Milan, Italy, 4Università Vita-Salute San Raffaele, Milan, Italy

Background: Conditioning is the preparative regimen administered to the patients undergoing HSCT, before the infusion of the stem-cell grafts; although full consensus has not been reached, conditioning regimens have usually been classified as high-dose (MAC), reduced-intensity (RIC), and non-myeloablative (NMA). The EBMT has recently proposed an updated classification, assigning intensity weight scores for frequently used conditioning components in relation to their prognostic value for non-relapse mortality (NRM); and using their sum to generate a transplant conditioning intensity (TCI) score.

Methods: We revised the outcomes of haplo-HSCT, according to the new TCI score, in two GITMO centres. Primary endpoint was the evaluation of TCI impact on OS; secondary endpoints were DFS, GRFS, NRM, relapse incidence (RI), cumulative incidence (CI) of grade 2-4 aGvHD, and CI of cGvHD.

Results: From January 2014 to December 2021 135 patients received a haplo-HSCT in center-1 (C1) and 81 in center-2 (C2). We performed our analysis on overall 216 patients (patients/donor and disease features are reported in table 1). All patients received a PTCy-based GvHD prophylaxis.

According to the EBMT TCI score, 65 patients received a low TCI conditioning (30.1%), 79 patients an intermediate TCI conditioning (36.6%) and 72 a high TCI conditioning (33.3%). TCI low conditioning was mainly performed in C2, while TCI high conditioning in C1.

In univariate analysis, a higher TCI score was associated with a higher aGvHD grade 3-4 CI (p= 0.000038): 28 + /-2% for high group, 9 + /-2% for intermediate and 2 + /-1% for low. It was also associated with a higher 2-year cGvHD moderate-severe CI: 29 + /-1% for high score, 23 + /-1% for intermediate and 8 + /-1% for low (p=0.014). Moreover, there was a better trend, but not significant (p=0.088), for higher TCI score patients 2-year DFS: 45 + /-6% for low, 49 + /-6% for intermediate and 65 + /-6% for high. Overall, 2-year OS, GRFS, NRM, RI and cGvHD CI didn’t differ significantly according to TCI distribution. In multivariate analysis, results confirmed the impact of higher TCI on CI of aGvHD grade 3-4 and of cGvHD moderate-severe (HR 0.054 and 0.4, respectively).

Patient age – median (range)

57 (18-76)

Median follow-up – months (range)

46 (4-112)

Donor age – median (range)

38 (18-70)

Female into male, n (%)

51 (23.6%)

TCI score

• Low, n (%)

65 (30.1%)

• Intermediate, n (%)

79 (36.6%)

• High, n (%)

72 (33.3%)

Diagnosis

• Acute Leukemia, n (%)

149 (68.9%)

• Other diseases, n (%)

67 (31.1%)

Disease status at transplant

• Acute Leukemia not in CR, n (%)

61 (28.2%)

• Others, n (%)

155 (71.8%)

Table 1 – Patient, donor and disease features. TCI=transplant conditioning intensity; CR=complete remission

Conclusions: TCI conditioning stratification is able to provide a deeper insight in deciphering the impact of conditioning on clinical outcomes, not only in an AML restricted population, and also in the haplo-PTCy setting. TCI high resulted to be associated with a better DFS and a higher incidence of both severe acute and chronic GvHD, but not to a worst OS or higher NRM. Of note, in our population, TCI stratification didn’t identify different occurrence of NRM as in the original Registry analysis; a possible explanation relies upon the improvement in GvHD therapy, considering the advent of new drugs such as Ruxolitinib; however, further extended analysis are needed to validate in a large scale TCI stratification.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P187 CHANGES IN PROGNOSTIC VARIABLES FOR ALLOGENEIC TRANSPLANTATION WITH REDUCED INTENSITY CONDITIONING IN ELDERLY PATIENTS OVER TIME

Seong Kyu Park 1, Se Hyung Kim1, Jina Yun1, Chan Kyu Kim1, Jong Ho Won2

1Soonchunhyang University Bucheon Hospital, Bucheon, Korea, Republic of, 2Soonchunhyang University Seoul Hospital, Seoul, Korea, Republic of

Background: Allogeneic stem cell transplantation (SCT) necessitates meticulous consideration of the patient’s age, comorbidities, and reduced functional reserves. Reduced intensity conditioning (RIC) has emerged as an alternative regimen for patients not eligible for standard myeloablative conditioning. However, transplantation of elderly patients with myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) remains challenging due to age-related non-hematologic comorbidities, leading to heightened vulnerability to treatment toxicities. This study explores how clinical variables impacting allogeneic SCT outcomes in elderly patients have evolved over time.

Methods: Data were gathered from a total of 119 patients with AML or MDS who underwent RIC before SCT who were aged ≥ 60 years, or ≥ 55 years of age and unfit for SCT between 2010 and 2022. Patient data, including comorbidities, disease status, and transplantation method, were collected and divided into two groups based on transplantation period (period A: 2010-2015 vs period B: 2016-2022).

Results: Over time, the proportion of elderly patients aged ≥ 65 years tripled (15.8% vs 45.7%, p= 0.002). The use of anti-thymocyte globulin significantly increased (47.4% vs 97.5%, p < 0.001). In transplant outcome analysis, cases with complete donor chimerism at 3 weeks post-transplantation increased (42.1% vs 84.0%, p < 0.001). There was no difference in GVHD or viral reactivation. Recurrence frequency decreased by almost half (26.3% vs 14.8%, p=0.132). The 3-year survival rate improved from 47.4% to 76.5% (p= 0.087). Subgroup analysis revealed improved survival outcomes at 2-year after SCT for patients with pre-transplant disease status excluding non-CR (74.6% vs 57.7%, p < 0.001), MDS patients (77.4% vs 33.3%, p= 0.001), mismatched or haplo-identical donors (54.5% vs 21.4%, p= 0.002), and cases with complete chimerism at 3 weeks after SCT (67.1% vs 56.3%, p= 0.006). There was also an improvement in survival outcomes in cases with grade 2 or higher acute GVHD (2-year survival rate: 52.4% vs 35.7%, p= 0.023). In the case of chronic GVHD, there was a borderline statistical difference during period A, but after the introduction of ruxolitinib, the transplant results were almost the same. Prognostic factors included busulfan dose of conditioning regimen, HCT-CI based high risk, donor type, and mixed chimerism at 3 weeks after transplantation. In cases of mixed chimerism 3 weeks after transplantation, the frequency of TRM and recurrence within 1 year tended to increase, which led to poor survival outcomes.

Conclusions: Despite the increase in elderly patients, transplant outcomes have improved over time, reflecting the recent positive trajectory in the field of allo-SCT. And this study underscores the need for ongoing refinement of therapeutic approaches and vigilant management of prognostic factors.

Clinical Trial Registry: None.

Disclosure: The authors have no potential conflicts of interest to disclose.

16: Conditioning Regimens

P188 COMPARISON OF FLUDARABINE-MELPHALAN AND FLUDARABINE-TREOSULFAN AS CONDITIONING REGIMENS PRIOR TO ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Maria Liga 1, Dimitris Tsokanas1, Eleftheria Sagiadinou1, Memnon Lysandrou1, Angeliki Georgopoulou1, Evangelia Triantafyllou1, Vassiliki Zacharioudaki1, Anastasia Christopoulou1, Alexandros Spyridonidis1

1University Hospital of Patras, Patras, Greece

Background: The optimal counterpart in fludarabine-based (Flu) reduced intesity conditioning regimens in allogeneic hematopoietic cell transplantation (allo-HCT) is not defined yet. The combination of fludarabin-melphalan (FluMel) has shown efficacy but also toxicity. More recently treosulfan (Treo) has been introduced as a drug with potent immunosuppressive and antileukemic effect and a low toxicity profile. Thus, herein we compared the outocomes of patients receiving FluMel or FluTreo.

Methods: In this sinlge center analysis, we retrospectively analysed outcomes of patients who were selected to receive a fludarabin-based regimen combined with Mel 110 – 140 mg/m2 (FluMel) or Treo 30-42 gr/m2 (FluTreo), eg reduced toxicity regimens with a low or intermediate TCI score, from October 2005. Comaprison was done by univariate analysis using χ2 (categorical parameters), Mann-Whitney (continuous parameters) or log-rank (for OS) setting the statistical significance at p< 0.05.

Results: In total, 36 patients have recieved FluMel and 23 patients FluTreo. FluTreo patients were treated more recently (since April 2021) and thus the median follow up differed between these two groups. Detailed characteristics are listed in Table 1. There were no differences between age, HCT-CI, disease, disease status at allo-HCT and type of donor between the two groups. No difference in time to leukocyte and platelet engraftment was seen between the two groups. Transplant realted mortality was higher in the FluMel group (p=0.042) while relapse rate was similar, thus resulting in a significant better OS for the FluTreo group (p=0.004). When regimens were compared according to their TCI scores (low vs intermediate) irrespective of chemotherapy agent included, OS differed significantly (p=0.02).

Table 1. Demographic characteristics of the patients

Patient Characteristics

FluMel

FluTreo

P value

(univariate analysis)

Number of patients

36

23

N/A

Median Age (range)

47 (20-72)

52 (18-72)

0.58

Sex – M/F

22 / 14

17/6

0.27

Disease

AML

22

16

0.35

ΜDS

8

5

NHL

4

Β-ALL

1

Other diseases

1

1

Disease Stage before allo-HCT

Refractory Disease

3

3

0.49

Partial Response

3

Complete Response

26

18

Untreated

4

1

Donor

Sibling

9

4

0.35

WMUD

15

14

MMUD

10

5

Haplo

2

TCI score

Low

17 (47%)

9 (39.1%)

0.61

Intermediate

19 (53%)

14 (60.7%)

HCT-CI median (range)

1 (0-6)

0.5 (1-6)

ns

Engraftment

WBC≥1000/mmc, day (range)

+15 (10-24)

+14 (10-23)

0.86

PLT≥ 20.000/mmc, day (range)

+16 (7-57)

+16.5 (8-29)

0.44

PLT≥ 50.000/mmc, day (range)

+17 (11-127)

+16.5 (13-33)

0.49

Graft Failure

2

1

0.052

Transplant Related Mortality (TRM)

Yes

11 (31%)

2 (8.7%)

0.042

No

25 (69%)

21 (91.3%)

Relapse

Yes

9 (25%)

4 (17.4%)

0.55

No

27 (75%)

19 (82.6%)

Overall Survival

Days (range)

292 (12-2054)

154 (6-896)

0.004

Alive/Deceased

16/20

18/5

Conclusions: With the limitation of using FluTreo in more recent years, our single center analysis indicates a better toxicity profile of FluTreo vs FluMel without affecting relapse incidence. These data need prospective confirmation but are capable of establishing FluTreo in our strategy as the appropriate reduced toxicity regimen in frail pts.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P189 OUTCOMES OF PATIENTS WITH HIGH-RISK ACUTE LYMPHOBLASTIC LEUKEMIA IN CR1 UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION THROUGH A FIXED TRANSPLANTATION POLICY INCLUDING A CHEMOTHERAPY-BASED CONDITIONING

Enrico Santinelli1, William Arcese1, Gottardo De Angelis2, Benedetta Mariotti2, Giulia Ciangola2, Giuseppe Avvisati1, Laura Cudillo3, Paolo De Fabritiis4, Andrea Mengarelli5, Agostino Tafuri6, Antonio Bruno2, Ilaria Mangione2, Gaspare Adorno2, Adriano Venditti2, Alessandra Picardi7, Raffaella Ceretti 2

1Fondazione Policlinico Universitario Campus Biomedico, Rome, Italy, 2Fondazione Policlinico Tor Vergata, Rome, Italy, 3Ospedale San Giovanni-Addolorata, Rome, Italy, 4Ospedale Sant’Eugenio, Rome, Italy, 5IRCCS Istituto Nazionale Tumori “Regina Elena”, Rome, Italy, 6Ospedale Universitario Sant’Andrea, Rome, Italy, 7Ospedale Antonio Cardarelli, Naples, Italy

Background: Despite the significant improvements achieved in the frontline treatment of Acute Lymphoblastic Leukemia (ALL), patients with high-risk ALL in first complete remission (CR1) must proceed to an allogeneic hematopoietic stem cell transplantation (HSCT). In this context, although mostly based on retrospective studies, a myeloablative conditioning including total body irradiation (TBI) is usually preferred.

Methods: From October 2008 to February 2021, 54 consecutive patients affected by high-risk ALL (defined by t(4;11)/MLL rearrangements at 11q23, t(8;14), trisomy 8, monosomy 7, low hypodiploid, near triploid or complex karyotype or pro-B phenotype for B-ALL, and by pro-T, early T-cell precursor, T-mature phenotypes and complex karyotype for T-ALL; refractoriness on incomplete response to first induction chemotherapy) in CR1 underwent HSCT in a single Center. As induction therapy, 13 (24%) patients received conventional chemotherapy, while 21 (39%) were treated with a BFM adapted protocol. Twenty (37%) patients with a Ph+ ALL were given TKI with or without chemotherapy. All patients were transplanted according to an identical policy in terms of strategy for donor choice, conditioning regimen, GvHD prophylaxis and supportive care. All patients were prepared using a chemotherapy-based conditioning consisting of the Thiotepa, Busulfan and Fludarabine (TBF) combination.

Results: With a median follow up of 3.7 (range 0.08-15) years, the 5-year Overall Survival (OS) and Disease Free Survival (DFS) were 59% (95% CI, 47-53) and 51% (95% CI, 39-66), respectively. Characteristics of patients transplanted < vs ≥ 2015 year were comparable, except for the more recent introduction of the BFM protocol as induction therapy prior to transplant (p<0.001, Table). Excluding Ph+ cases by building a two-by-two contingency table, BFM protocol confirmed its better efficacy in achieving MRD negativity (χ2 =5,23, p=0.022; OR=0.137, 95% CI 0.02-0.85).

In multivariate analysis, the HSCT date (< vs ≥ 2015 year) and MRD (pos. vs neg.) significantly affected OS (HR 5.22, CI 2.02-13.47: p=0.002; HR 3.37, CI 1.39-8.16: p=0.007), DFS (HR 3.50, CI 1.52-8.04: p=0.003; HR 3.68, CI 1.61-8.46; p=0.004), and relapse (HR 3.87, CI 1.28-11.67: p=0.016; HR 7.84, CI 2.33-26.38: p=0.001). Acute GvHD grade II-IV had a negative impact on OS (HR 3.41, CI 1.34-8.68: p=0.01) and TRM (HR 9.25, CI 2.49-34.36: p=0.001).

HSCT < 2015

HSCT ≥ 2015

Patients characteristics

N=21

N=33

p value

Diagnosis (B-ALL Ph + /B-ALL Ph-/T-ALL)

8 (38%)/9 (43%)/4 (19%)

12 (36%)/8 (24%)/13 (39%)

0.259

Type of last therapy before transplant

<0.001

Conventional

11 (52%)

2 (6%)

BFM

2 (10%)

19 (58%)

TKI ( + /- CHT)

8 (38%)

12 (36%)

MRD status at transplant (Negative/Positive)

12 (57%)/9 (43%)

22 (67%)/11 (33%)

0.480

N° previous therapies (One/More than one)

15 (71%)/6 (29%)

26 (79%)/7 (21%)

0.537

Recipient CMV Status (Negative/Positive)

5 (24%)/16 (76%)

6 (18%)/27 (82%)

0.617

Donor CMV Status (Negative/Positive)

6 (18%)/15 (82%)

13 (39%)/20 (61%)

0.417

Recipient Sex (Male/Female)

9 (43%)/12 (57%)

13 (39%)/20 (61%)

0.801

Donor Sex Male (Male/Female)

11 (52%)/10 (48%)

17 (52%)/16 (48%)

0.951

Donor Type (MSD/MUD/MMUD/Haploidentical)

4(19%)/9(43%)/3(14%)/5(24%)

8(24%)/9(27%)/9(27%)/7(21%)

0.551

Stem cell source (BM/PBSC)

7 (33%)/14 (67%)

16 (48%)/17 (52%)

0.272

Conditioning intensity (MAC/RIC)

17 (81%)/4 (19%)

26 (79%)/7 (21%)

0.847

aGvHD g II-IV

5 (21%)

11 (33%)

0.455

aGvHD g III-IV

0 (0%)

3 (9%)

0.274

cGvHD

9/18 (50%)

10/31 (32%)

0.219

Extensive cGvHD

4/18 (22%)

8/31 (26%)

0.779

.

Table 1: Comparison of characteristics between patients transplanted until and beyond 2014..

Conclusions: This unicentric analysis confirms the prognostic role of MRD on survival of patients with high-risk ALL undergoing HSCT, highlighting the relevance of a more intensive chemotherapy approach to achieve MRD negativity prior to transplant.

Finally, this study supports the hypothesis that, in patients affected by high-risk ALL in CR1 who achieve MRD negativity, a chemotherapy-based conditioning, such as the TBF regimen, can be as effective as a TBI-based conditioning.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P190 DESENSITISATION TREATMENT FOR HLA-DONOR SPECIFIC ANTIBODIES IN EX-VIVO T CELL DEPLETED HAPLOIDENTICAL ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANT – A MULTI-CENTRE RETROSPECTIVE STUDY IN SINGAPORE

Christopher S W Tham1, Yeh Ching Linn1, Liang Piu Koh2, Jeffrey KS Quek1, Aloysius Y L. Ho1, Francesca WI Lim1, William Y.K. Hwang 1, Yeow Tee Goh1, Jing Jing Lee1, Hein Than1

1Singapore General Hospital, Singapore, Singapore, 2National University Cancer Institute, National University Health System, Singapore, Singapore

Background: Haploidentical haematopoietic stem cell transplant (haplo-HCT), by definition, involves human leukocyte antigen (HLA) mismatch between donor and recipient. Recipients may have HLA-antibodies, of which some may be donor specific antibodies (DSA). DSA can negatively impact engraftment. DSA desensitisation methods are routinely used prior to HCT, however data on its efficacy is limited.

Methods: This was a multi-centre retrospective analysis assessing consecutive adult patients undergoing ex-vivo T-cell (TCRαβ and CD45RA + ) depleted haploidentical HCT (haplo-TCD) from 2017 to 2023 in Singapore. Prior to transplant, recipient peripheral blood samples were screened for HLA-antibodies by flow cytometric assay. Those with a positive screen subsequently underwent solid phase single antigen testing (Luminex) to further characterise HLA-antibodies and quantify antibody mean fluorescence intensity (MFI). Only patients with DSA were selected for analysis. Antibody MFI was assessed at baseline and after desensitisation. Clinical data was collected from medical records. Statistical analysis was performed using GraphPad Prism version 10.1.1; Wilcoxon signed rank test for paired data, Mann-Whitney test for unpaired data, all p-values were two-tailed.

Results: 65 patients underwent haplo-TCD during the study period. 11 patients with at least 1 DSA were identified (see table 1 for details) – all received desensitisation consisting of 3 sessions of plasma exchange (PE) (1.5 times total plasma volume), 1 dose of intravenous immunoglobulin (IVIg) (1g/kg) and 1 dose of intravenous rituximab (375mg/m2). 2 patients had additional treatment (table 1).

Patients had a median number of 24 HLA class I (interquartile range; IQR 13-37.5) and 8 class II (IQR 2-18) allele-specific antibodies. Median MFI at baseline was 6483 (IQR 2930-13880) compared to 2308 (IQR 0-6991) post-desensitisation (p<0.0001). MFI analysis was stratified by >5000 versus <5000, as MFI >5000 is more likely to be clinically significant. Desensitisation led to a significant MFI reduction in both groups. A significantly greater proportion of reduction was observed in those with a baseline MFI of <5000. With a baseline HLA-antibody MFI <5000, median change was 100% (IQR 59.3-100), i.e. to an undetectable MFI, compared to a median of 53.1% (IQR 35.7-66.1) (MFI baseline>5000).

Donor-specific HLA-antibody MFI at baseline was 5387 (IQR 2348-14656) compared to 1559 (IQR 0-9478) post-desensitisation (p<0.0001). Post-desensitisation, MFI remained above 5000 in 5 out of 11 patients. 1 of these patients went on to receive further desensitisation (table 1).

Time to engraftment was compared to patients without DSA who underwent haplo-TCD over the same period (n=49). Median time to both neutrophil and platelet engraftment was similar in both groups. For neutrophils, this was 11 (IQR 10-18) days (DSA group) and 13 (IQR 11-16) days (without-DSA group), (p= 0.284). For platelets, this was 12 (IQR 11-13) days (DSA group) and 11 days (IQR 10-13) days (without-DSA group), (p= 0.363).

Table 1

Number of patients with DSA

11*

DSA Desensitisation Regimen

Age, years, median (IQR)

55 (39 - 61)

3xPE, IVIg, Rituximab

9*

Gender, number of patients (Male, Female)

1, 10

4xPE, IVIg, Rtiuximab

1

Diagnosis, number of patients

6xPE, 2xIVIg, 2xRituximab, Buffy coat infusion

1^

AML/MDS

9

Cell dose (CD34+x106/kg), median (IQR)

7.52 (6.71 - 8.48)

Atypical CML

1

Time to neutrophil engraftment, days, median (IQR)

11 (10 -18)

NK/T Cell Lymphoma

1

Time to platelet engraftment, days, median (IQR)

12 (11 - 13)

Ph+ B-ALL

1

HLA-antibodies, number, median (IQR)

Disease status at transplant, number of patients

Class I

24 (13 - 37.5)

CR1

4

Class II

8 (2 - 18)

CR2

3

All HLA-antibodies, MFI, median (range)

Stable disease

3

Baseline

7516 (825 - 25768)

Relapse/Progressive disease*

1*

Post-Desensitisation

2113 (0 - 20673)

Donor source, number of patients

Donor specific HLA-antibodies

Parent

1

Number per patient, median (IQR)

2 (1 - 2)

Sibling

3

Baseline MFI, median (IQR)

5387 (2348 - 14656)

Child

6

Post-desensitisation MFI, median (IQR)

1559 (0 - 9478)

  1. ^1 patient underwent further desensitisation as MFI remained > 5000
  2. *1 patient underwent desensitisation but did not undergo haplo-TCD

Conclusions: Desensitisation with PE, IVIg and rituximab significantly reduced HLA-antibody MFI. After desensitisation, time to engraftment was comparable in those with and without DSA, suggesting that this is an effective method of overcoming the obstacle of DSA in patients undergoing haplo-TCD.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P191 PROMISING OUTCOMES IN HAPLOIDENTICAL TRANSPLANTATION USING ABATACEPT IN COMBINATION WITH POST TRANSPLANT CYCLOPHOSPHAMIDE IN TREOSULFAN BASED CONDITIONING REGIMEN

Mohammed Debes1, Gabe Toth1, Tom Seddon1, Shahid Iqbal1, Leah Credidio1, Hannah Williams2, Fotini Partheniou2, Sophie Hughes1, Priscilla Hetherington1, Thomas Seddon1, Muhammad Saif 1

1Clatterbridge Cancer Centre, Liverpool, United Kingdom, 2Liverpool Clinical Laboratories, Liverpool, United Kingdom

Background: Number of stem cell transplantation carried out using haplo-identical donor (Hap-SCT) is increasing worldwide. A lack of suitable matched donor, lower cost, easy donor availability and acceptable GVHD control with post transplant cyclophosphamide (PTCy) are main factors driving increased use of haploidentical donors. In some reported studies, survival data in Hap-SCT is now matching outcomes from matched donors. Optimizing GVHD control, preventing infections and mitigating risk of graft rejection remain some of the challenges following Hap-SCT. Abatacept was recently approved as GVHD prophylaxis in combination with calcineurin inhibitor (CNI) and Methotrexate post stem cell transplantation. Abatacept is a CTLA4 analog which blocks co-stimulatory signal and prevent T cell activation. Treosulfan is emerging as an effective component of reduced toxicity regimen. To our knowledge, outcome data of this combination is not reported in adult hap-SCT.

Methods: We present our single center experience of using Treosulfan based conditioning with Abatacept and PTCy as GVHD prophylaxis in 14 patients who underwent hap-SCT (institutional approval number 2223-94). Conditioning consisted of Fludarabine, Treosulfan and Low dose TBI (2Gy) followed by post transplant cyclophosphamide. Fludarabine dose was 30 mg/m2 given on days -6 to -2, Treosulfan dose was 10 or 14 gram/m2 given on days -6 to -4. PTCy was given at a dose of 50 mg/kg/day on days +3 and +4. Tacrolimus and Mycophenolate Mofetil (MMF) were commenced on day+5. All patients received 3 doses of Abatacept on days +5, + 14, + 28.

Results:

Gender

12 male/2 female

Performance score (Kernofsky)

80 (range 70-90)

Donor specific antibodies

Stem cell source

Peripheral blood stem cells in all patients

CD34 infused (Median)

5 x 106 /kg (range 3.25 – 6.11 x 106 /kg)

CMV match

6 matched/ 8 mismatched

Median age

44 years (range 17-65)

ABO match

5 (all minor ABO incompatibility), 9 ABO matched

HCT-CI (Median)

3

Indications for transplantation

3 ALL, 6 AML, 2 CML, 1 MDS and 2 SAA.

Our results show OS of 83.3% at 5 years with a median follow up of 630 days (range 47 – 1728 days). Out of 14 cases, 2 died at 7 and 14 months and 12 patients are alive. 1 patient died of AML relapse and the other died of sepsis. Relapse rate was 14.28%, 1 AML and 1 ALL. Incidence of acute GVHD (grade II-IV) was 21.4% and incidence of chronic GVHD was 41.6%. Within the chronic GVHD group, 20% had mild form while 80% had severe cGVHD as per NIH criteria. Median relapse free survival (RFS) was not reached in this cohort. Median GVHD free survival (GFS) was 372 days, and median of GVHD and relapse free survival (GRFS) was 210 days.

Median neutrophil engraftment was 19 days, range 14-29 days, while median platelet engraftment was 32 days, range 16-138 days. Post-transplant complications included CMV reactivation in 29%, urinary BK virus in 14%, neutropenic fever in 50%, pneumonia in 21% and CRS in 14%. Cumulative rate of other viral infections including EBV, Adenovirus, Herpes, respiratory viruses was 43% and Line infections were seen in 21%.

In addition, Haemophagocytic lymphohistiocytosis (HLH) occurred in 14%, 1 patient experienced transient ischemic attack (TIA), 2 developed diabetes and 1 developed adrenal insufficiency over course of follow up.

All patient engrafted and achieved 100% donor chimerism at 1 year (for both myeloid and CD3 chimerism).

Conclusions: Abatacept in combination with PTCy in hap-SCT using Treosulfan based conditioning appears to show promising outcome and acceptable toxicity. However, given the severity of chronic GVHD, larger prospective trials are required to evaluate this strategy.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P192 FOLLOW-UP OF TRANSPLANT-ELIGIBLE MULTIPLE MYELOMA RECEIVED BUSULFAN-BASED CONDITIONING VERSUS HIGH-DOSE MELPHALAN

Sungnam Lim1, Byeong-Sok Sohn2, Seonyang Park 3

1Inje University, Busan, Korea, Republic of, 2Inje University, Seoul, Korea, Republic of, 3Haeundae Paik Hospital, Busan, Korea, Republic of

Background: High-dose melphalan (HDMEL) represents the standard conditioning regimen before autologous stem cell transplant (ASCT) in multiple myeloma, but recent updates have suggested combination of busulfan with melphalan is also associated with favorable outcomes. We previously reported that there was no difference in PFS between the two groups receiving conditioning using busulfan-based and high-dose melphalan. These results showed that busulfan-based conditioning has high efficacy with manageable adverse events.

So, we followed up on the previously published data and conducted additional analyses focusing on biochemical and clinical relapse-related events.

This retrospective study was performed to investigate the effectiveness and safety of a busulfan-based conditioning regimen without melphalan.

Methods: A total of 35 patients who had been treated with remission induction therapy for bortezomib, thalidomide, and dexamethasone (VTD) were enrolled in this study between March 2016 and September 2021. The conditioning regimens were as follows, busulfan-based regimen was composed of busulfan 3.2 mg/kg from days -7 to days -5, etoposide 200 mg/m2 from days -5 to days -4, cyclophosphamide 50 mg/m2 from days -3 to days -2 and HEMEL conditioning regimen consisted of melphalan from 140 mg/m2 to 200 mg/m2.

Results: The median age of patients was 58 years old (range, 47-67 years old) in the HD-MEL group and 57 years old (range, 48-63 years old) in the BuCyE group, respectively. The revised international staging system (R-ISS) was shown as follows; stage I with 0% in BuCyE vs. 27.2% in HD-MEL, stage II with 84.6% vs. 68.1%, and stage III with 15.4% vs. 4.5%, respectively.

The overall response rate before ASCT was 100% in both groups, including 76.9% in BuCyE and 63.6% in HDMEL with more than very good partial response, and 30.8% in BuCyE versus 40.9% in HDMEL with complete response(CR), respectively. The median follow-up for survivors was 38 months. The median PFS was 37.9 months in HDMEL (95% CI, 23.2 to 52.7 months) and 37.9 months in BuCyE (95% CI, 29.1 to 46.8 months) (P=0.676). The 5-year PFS was 12.8% for HDMEL versus 30.8% for the BuCyE, respectively. PFS was analyzed by dividing it into biochemical relapse and clinical relapse. When disease progression is limited to clinical relapse, the median PFS was 56.8 months in HDMEL (95% CI, 26.1 to 87.4 months) and 38.3 months in BuCyE (95% CI, 27.9 to 48.6 months) (P=0.611). Five-year overall survival rate was 76.1% in HDMEL versus 100% in BuCyE (P=0.168).

The average time from ASCT to leukocyte recovery (≥ 1,000/mm3 of absolute neutrophil count) and platelet recovery (≥50,000/mm3 of platelet count) is as follows: 11 days in HDMEL versus 12 days in BuCyE (P=0.101) and 39 days in HDMEL versus 34 days in BuCyE (P=0.840), respectively.

Conclusions: Our results of the follow-up study showed that a busulfan-based conditioning regimen of BuCyE could be expected to prolong PFS and survival through secondary treatment compared to the high-dose melphalan conditioning regimen.

Disclosure: Nothing to declare..

16: Conditioning Regimens

P193 SEQUENTIAL CONDITIONING REGIMEN WITH THIOTEPA, CLOFARABINE AND BUSULFAN (TEC/CLOB2A2) AND POST-TRANSPLANT CYCLOPHOSPHAMIDE IN ADULTS WITH REFRACTORY AML: A RETROSPECTIVE MONOCENTRIC STUDY

Amandine Le Bourgeois1, Alice Garnier1, Pierre Peterlin1, Maxime Jullien1, Chloe Antier1, Thierry Guillaume1, Patrice Chevallier 1

1Nantes University Hospital, Nantes, France

Background: Sequential allogeneic stem cell transplantation (Seq allo-SCT) is proposed to fit and relatively young patients with refractory hematologic malignancies. A combination of thiotepa, etoposide, cyclophosphamide (TEC) followed by fludarabine/busulfan/ATG (FB2A2) using any type of donor for refractory hematologic malignancies have shown some promising results (Dulery, 2018). As clofarabine may have better antileukemic effect than fludarabine, we have investigated in our patients with refractory AML a new sequential approach combining TEC+clofarabine/busulfan/ATG (TEC/CloB2A2).

Methods: We have investigated in our Hematology Department a new sequential conditioning regimen replacing fludarabine by clofarabine as part of a TEC/FB2 regimen with the hope to obtain less relapse and better survival in a cohort of patients with refractory AML. The TEC/CloB2A2 regimen consisted of Thiotepa 5 mg/kg at day(d)-13, Cyclophosphamide 400 mg/m²/d from d-12 to d-9 and Etoposide phosphate 100 mg/m²/d from d-12 to d-9, followed after 3 days of rest, by Clofarabine 30 mg/m²/d from d-5 to d-1, Busulfan 3,2 mg/kg/d d-5 and d-4 and Thymoglobuline 2,5 mg/kg/d d-3 and d-2. GVHD prophylaxis consisted of high-dose of post-transplant Cyclophosphamide (PTCY) 50 mg/kg/d at d + 3 and d + 5 in combination with mycophenolate mofetyl and cyclosporine from d + 6. Any type of donor have been considered.

Results: Between February 2020 and August 2022, 12 patients have been treated. Median age was 58 y. The majority of patients had primary refractory AML (n=9). All but 2 donors were haploidentical. One patient with a matched donor did not receive PTCY after transplant.

Two patients died during the aplasia phase, one at d + 6 of sepsis and multiple organ failure and one at d + 30 of disseminated fungal infection. The median time of neutrophils (> 1 Giga/L) and platelets (> 50 Giga/L) recoveries were 22 days (range : 12-137) and 34 days (range : 16-241), respectively. There were no primary or secondary graft failures. Responses were evaluated at a median of 57 days (range : 26-70). Five complete remission (CR), 3 CR with incomplete platelet recovery and two failures were documented for an overall CR/CRp rate of 67%. Only one cutaneous grade 2 acute GVHD occured after transplant but no chronic GVHD. Six patients received maintenance treatment after transplant to prevent relapse using 5’azacytidine alone in 1, 5 azacytidine+ donor lymphocyte infusion (DLI) in 2, DLI alone in 1. The last two patients received maintenance as part of a trial. The three patients receiving DLI developped acute GVHD thereafter including 2 grade 2 and one grade 3.

Six of the patients achieving CR/CRp relapsed (75%) at a median of 160 days (range : 48-344). At last follow-up (July 2023), only one patient is alive in CR (8%) at 795 days from tranplant (SET/CLoB2A2 +PTCY and sibling donor, grade 3 acute GVHD after DLI). 1-y and 2y overall and leukemia survival were both at 25% (9.3-66.6) and 8.3% (1.2-54.4), respectively.

Conclusions: The results of this small cohort showed that a sequential TEC/CloB2A2 conditioning regimen do not provide better outcomes compared to a TEC/FB2A2 sequential conditioning regimen for refractory AML patients.

Clinical Trial Registry: none.

Disclosure: none.

16: Conditioning Regimens

P194 CLINICAL OUTCOMES OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION USING BUSULFAN/CYCLOPHOSPHAMIDE AND MELPHALAN OR THIOTEPA AS CONDITIONING REGIMEN IN 37 PATIENTS WITH ACUTE MEGAKARYOBLASTIC LEUKEMIA

Fei Pan 1, Guanlan Yue1, Kang Gao1, Kun Wang1, Huili Zhu1, Yujie Wang1, Zheren Wang1, Zhijie Wei1

1Hebei Yanda Lu Daopei Hospital, Hebei, China

Background: Acute megakaryoblastic leukemia (AMKL) is a rare and heterogeneous subtype of acute myeloid leukemia (AML), which accounts for approximately 10% of childhood AML and about 1% of adult AML. Patients with non-Down’s syndrome AMKL (non-DS-AMKL) are often associated with a poor prognosis with a 3-year overall survival (OS) rate of less than 40%. Currently, allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a potential curative therapy for AMKL. However, the long-term survival post-transplant needs to be improved. Here we conducted a single-center study to evaluate the long-term efficacy and safety of allo-HSCT in 37 non-DS-AMKL patients using an innovative conditioning regimen of Busulfan/Cyclophosphamide (Bu/Cy) and Melphalan (MEL) or Thiotepa (TT).

Methods: From August 2015 to July 2023, a total of 37 non-DS-AMKL patients (M/F 21:16) were treated consecutively in our hospital. The median age was 2 years (1-9 years). Before transplantation, in bone marrow (BM), 29 patients were in complete remission (CR), 3 were in partial remission (PR) and the rest 5 were in non-remission (NR). Thirty-three patients received the haploidentical transplantation (donors from parents n=31, siblings n=2), and 4 received matched-unrelated donor transplantation. The stem cell sources were from BM and peripheral blood stem cells (PBSCs) among the 33 related donors, and PBSCs for the 4 unrelated donors. Patients received a median cell number of total mononuclear cells 20.15×108/kg, CD34+ cells 12×106/kg and CD3+ cells 4.66×108/kg. The conditioning regimens administered were modified Bu/Cy (fludarabine 30 mg/m2/d×5d; cytarabine 2g/m2/d×5d; Bu 3.2 mg/kg/d×4d, Cy 1.8g/m2/d×2d; etoposide 100mg/m2/d×2d; antithymocyte globulin 1.5mg/kg/d×5d) in 2 patients, Bu/Cy+MEL (MEL 55mg/m2/d×2d) in 31 patients, and Bu/Cy+TT (TT 2.5mg/kg/d×2d) in the rest 4 patients.

Results: The median follow-up time was 24 months (2-84 months), BM was assessed one month after transplantation, with all grafts fully donor-type, with a median leukocyte engraftment of +13 days, and a median platelet engraftment of +9 days. The OS was 54.1%. There was a significantly higher OS in the CR group (OS 69%) than the PR or NR groups (P<0.001). Among the CR patients, no significant difference was observed in the 3 different condition regimen groups with modified BU/CY group OS of 50%, BU/CY + MEL group OS of 69.6% and BU/CY + TT group OS of 75% (P=0.869). After transplantation, 17 patients died including 11 from relapse, 4 from GVHD, 1 from gastrointestinal bleeding and 1from cerebral hemorrhage. Post-transplant, the incidence of acute GVHD (aGVHD) was 35.13% (Grade II-IV), and severe aGVHD (Grade III-IV) incidence was 18.91%. And 51.35% of patients developed chronic GVHD. Other complication incidences were cystitis at 16.21%, cytomegalovirus at 5.94%, Epstein–Barr viremia at 13.5%.

Conclusions: Our study indicates that the use of modified Bu/Cy +MEL or +TT conditioning regimens in allo-HSCT procedures presents a feasible and effective approach, demonstrating an improvement in overall survival for non-Down Syndrome AMKL patients. Crucially, it was observed that achieving CR prior to transplantation significantly contributes to better outcomes. Therefore, it is strongly recommended that allo-HSCT is performed in CR status.

Disclosure: No relevant conflicts of interest to declare.

16: Conditioning Regimens

P195 RETROSPECTIVE EVALUATION OF NUTRITIONAL STATUS PRE AND POST HEMATOPOIETIC STEM CELL TRANSPLANTATION: A SINGLE CENTRE EXPERIENCE

Yuva Vishalini Ravindran 1, Maria Losa Maroto2, Amrith Mathew2, Sunrit Majumder2, Lewis David Oxley2, Venkatesh Karthikeyan3, Nitin Ramanathan4, David Waldron2

1Royal Wolverhampton Trust, Wolverhampton, United Kingdom, 2The University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom, 3All Indian Institute of Medical Sciences, Patna, India, 4Ananthapuri Hospitals, Trivandrum, India

Background: The successful delivery of allogeneic stem cell transplantation (alloSCT) is still limited by its non-relapse mortality (NRM). Poor nutritional status has been recognised as relevant for NRM. In the allogeneic transplant setting, malnourishment arises from a combination of toxic, inflammatory, and immunological mechanisms, culminating in caloric deficits through anorexia and metabolic catabolism. This retrospective study aims to meticulously evaluate the nutritional status of patients undergoing alloSCT and analyse its impact on transplant outcome.

Methods: A retrospective single analysis of all patients who underwent allograft at Queen Elizabeth Hospital from 2021-2022 was performed. Data was obtained from the hospital’s electronic system and was analysed using SPSS software.

Results: Data from a total of 157 patients was included in the study. Of the conditioning regimes, 77.1% (121) underwent a reduced intensity conditioned transplant (RIC) and 22.9% (36) underwent a myeloablative conditioned transplant (MAC). Artificial nutrition support (NGT or TPN) was undertaken in 38.9% (14) of MAC patients and 7.4% (9) of RIC patients. The mean age of patients undergoing a MAC was 33.8 years and for patients undergoing a RIC, it was 54.5 years. Patients undergoing a MAC had longer hospital stay with a mean of 31.3 days, whereas mean hospital stay for patients undergoing RIC was 26.5 days. Within the MAC group, 100% (36) of the patients developed mucositis, 41.7% (15) suffered from grade III and 25% (9) from grade IV, as per WHO criteria. With regards to the RIC group, 71% (86) developed mucositis, but 94% (81) was grade I and II and only 5.8% (5) developed grade III. No patients in the RIC group developed grade IV mucositis. Average weight loss at discharge for patients undergoing MAC was 8.33%, compared to the RIC group, where average weight loss was only 3.03%. NRM at day 100 was 2.8% within the MAC group and 7.4% within the RIC group.

Conditioning regimens

Myelo-Ablative conditioning

Reduced-Intensity conditioning

Aspects

Number of patients

22.9% (36)

77.1%(121)

Age Distribution

Mean: 33.8 years

Mean: 54.5 years

Mucositis Development and grades (WHO grading)

• Grade 0: 0%(0)

• Grade I: 16.7% (6)

• Grade II: 16.7% (6)

• Grade III: 41.7% (15)

• Grade IV: 25% (9)

• Grade 0: 28.3% (34)

• Grade I: 57.5% (69)

• Grade II: 10% (12)

• Grade III: 4.2% (5)

• Grade IV: 0% (0)

Parenteral Nutrition Requirement - NGT/TPN

38.9% (14)

7.4% (9)

Hospital Stay-Duration

Mean: 31.3 days

Mean: 26.5 days

Weights on admission

Mean 82.8 kg

Mean 79.3 kg

Weights on discharge

Mean 75.9 kg

Mean 76.9 kg

Weight loss in kilograms

7 kgs

2.4 kgs

Weight Loss in percentage

Average: 8.33%

Average: 3.03%

Table 1 - Comparison of significant aspects between MAC and RIC regimens.

Conclusions: This study suggests that patients undergoing myeloablative conditioned allogeneic transplants experienced longer hospitalizations, higher severity of mucositis with compromised oral intake and poor nutrition, leading to a higher weight loss when compared to patients undergoing reduced conditioned allogeneic transplants. These factors did not impact NRM at day 100 and at 2 years in our study, although due to the small sample size, this should be interpreted with caution. A better understanding of nutrition in the pre, peri and post-transplant setting and its relevance for NRM is required to standardised assessment of transplant candidates, to better monitor patients throughout their transplant journey and to design interventions to improve the nutritional status and consequently achieve better transplant outcomes.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P196 BEAM/LEAM VERSUS LACE CONDITIONING FOR AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN LYMPHOMAS – A RETROSPECTIVE, SINGLE-CENTRE ANALYSIS OF CLINICAL OUTCOMES AND TOXICITY PROFILE

Germano Glauber de Medeiros Lima1, André Dias Américo1, Eurides Leite da Rosa2, Pedro Paulo Faust Machado1, Juliana Matos Pessoa1, Hegta Taina Rodrigues Figueiroa 1, Fauze Lutfe Ayoub1, Isabella Silva Pimentel Pittol1, José Ulysses Amigo Filho1, Phillip Scheinberg1, Fabio Rodrigues Kerbauy1

1Beneficência Portuguesa de São Paulo, São Paulo, Brazil, 2Hospital 9 de Julho, São Paulo, Brazil

Background: The most effective conditioning regimen for lymphomas in autologous hematopoietic stem cell transplantation (aHSCT) remains unclear, as it has not been conclusively determined by randomized studies. In our research, we assessed the outcomes of using the LACE regimen (Lomustine, Cytarabine, Cyclophosphamide, and Etoposide) and the BEAM/LEAM protocols (Carmustine/Lomustine, Etoposide, Cytarabine, and Melphalan). This paper presents our findings on the clinical efficacy of these regimens and their influence on the incidence of neutropenic colitis (NC).

Methods: We conducted a retrospective, observational, and exploratory single-center cohort study at Hospital A Beneficência Portuguesa in São Paulo, Brazil. Adult patients (aged 18 years or older) diagnosed with Hodgkin’s or Non-Hodgkin’s Lymphomas were included. These patients underwent aHSCT as a form of consolidation therapy and were treated with one of three conditioning regimens: BEAM, LEAM, or LACE.

Results: From April 19, 2016, to January 24, 2023, a total of 84 patients met the inclusion criteria. Out of these, 24 underwent the BEAM/LEAM regimens (6 with BEAM and 18 with LEAM), while 60 received the LACE regimen as high-dose conditioning. The median age for both groups was 52 years. Despite the study’s non-randomized nature, comparable characteristics were noted between the groups in terms of age, sex, Hematopoietic Cell Transplantation-Comorbidity Index (HCT-CI), diagnosis (either Hodgkin or Non-Hodgkin Lymphomas), and previous lines of therapy. Notably, only 8.3% of patients in each group had a high-risk HCT-CI score.

At the 100, 365, and 730-day post-transplantation markers, no significant difference was observed between the groups in overall survival (OS) [BEAM/LEAM vs. LACE (100 days: 96% vs. 92%; 365 days: 87% vs. 80%; 730 days: 76% vs. 75%), p= 0.8] and progression-free survival (PFS) [BEAM/LEAM vs. LACE, (100 days: 83% vs. 90%; 365 days: 64% vs. 79%; 730 days: 50% vs. 64%), p= 0.2]. However, a significant difference was noted in the occurrence of neutropenic colitis (NC), with an odds ratio of 0.06 favouring the LACE conditioning regimen [OR 0.06 (95% CI, 0.02-0.2), p < 0.001], indicating a notably lower incidence of NC with LACE.

In terms of other outcomes, such as cumulative incidences of hospital discharge rates (considering in-hospital death as a competing event), requirements for transfusion, and rates of neutrophil or platelet engraftment, no significant differences were found between the groups. This similarity persisted across various time points (days 10, 20, 30, and 40 post-aHSCT).

General characteristics

Variable

BEAM/LEAM, n= 24

LACE, n= 60

P

Age, in years1

52 (44-57)

52 (46-54)

0.9

Female sex, n (%)

10 (41.7)

26 (43.3)

0.9

HCT CI, n (%)

0.3

Low (0)

17 (70.8)

32 (53.3)

Intermediate (1-2)

5 (20.8)

23 (38.3)

High (3 or greater)

2 (8.3)

5 (8.3)

Diagnosis

0.4

Hodgkin’s Lymphoma

5 (20.8)

18 (30)

Other Lymphomas

19 (79.8)

42 (70)

Prior Lines of Therapy, n (%)

0.3

1 Line

7 (36.8%)

16 (38.1%)

2 Lines

10 (52.6%)

16 (38.1%)

3 Lines

2 (10.5%)

10 (23.3%)

Unknown

1 mean (confidence interval 95%)

5

18

Outcomes

Variable

BEAM/LEAM

LACE

P

Overall survival 2

0.8

100 days

96 (88-100)

92 (85-99)

365 days

87 (75-100)

80 (69-93)

730 days

76 (60-97)

75 (62-91)

Progression free survival 2

0.2

100 days

83 (69-100)

90 (82-98)

365 days

64 (47-88)

79 (68-92)

730 days

50 (31-88)

64 (48-85)

Cumulative incidence of relapse2

0.3

100 days

4.4 (0.2-19)

1.8 (0.14-8.5)

365 days

23 (8-43)

10 (3.1-23)

730 days

37 (15-60)

25 (9.9-44)

Hospital discharge alive 2

0.9

D10

D20

78 (54-91)

80 (67-88)

D30

96 (66-100)

88 (76-94)

D40

92 (80-97)

96 (66-100)

In-hospital death 2,3

0.9

D10

3.3 (0.1-10)

D20

5 (1.3-13)

D30

4.3 (0.1-21)

5 (1.3-13)

D40

4.3 (0.1-21)

6.7 (1.9-16)

Neutrophil engraftment 2

0.3

D10

46 (25-64)

55 (42-67)

D20

96 (59-100)

97 (83-99)

D30

96 (59-100)

97 (83-99)

D40

96 (59-100)

97 (83-99)

Platelet Engraftment2

0.3

D10

8.3 (1.4-24)

6.7 (2.21-15)

D20

71 (47-85)

90 (78-96)

D30

92 (66-98)

93 (82-98)

D40

92 (66-98)

93 (82-98)

Packed red blood cells1

2.26 (1.4-3.2)

2.7 (1.8-3.6)

0.7

Platelet apheresis1

2 % (confidence interval 95%)

4.4 (3.1-5.7)

4.7 (3-6.4)

0.3

Conclusions: Despite its retrospective nature, single-centre scope, and limited patient cohort, our study highlighted a significant association: the LACE conditioning regimen, compared to BEAM/LEAM, substantially reduced episodes of NC, a serious and potentially fatal complication of aHSCT. Additionally, we noted comparable outcomes in both short-term (neutrophil/platelet engraftment and hospital discharge rates) and long-term (OS and PFS) aspects between these regimens. These findings suggest that the LACE regimen may be less toxic and potentially as effective as Carmustine-based regimens. Notably, our research also raises concerns regarding the role of melphalan in the development of NC. To conclusively address these findings and explore further implications, randomized prospective studies are essential.

Disclosure: André Américo received speaker fees from Janssen, Knight Pharmaceutical and MSD.

Eurides Rosa received speaker fees from Aztra Zeneca and MSD.

Fauze Ayoub received speaker fees from Janssen.

Phillip Scheinberg has done scientific presentations for Novartis, Roche, Alexion, Janssen, AstraZeneca; Grants/Research Support: Alnylam, Pfizer; has received grants or research support from Alnylam, Pfizer; has received consultancy fees from Roche, Alexion, Pfizer, BioCryst, Novartis, Astellas; and has been a speaker for Novartis, Pfizer, Alexion.

Hegta Figueiroa, Fábio Kerbauy, Isabella Pittol, José Filho, Juliana Pessoa, Germano Lima and Pedro Machado. have no conflicts of interest to declare.

16: Conditioning Regimens

P197 REAL-WORLD, SINGLE-CENTRE OUTCOME DATA OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION UTILISING TREOSULFAN-BASED CONDITIONING REGIMENS

Mohamed Debes1, Yeong Lim1, Thomas Seddon1, James Clarke1, Clare Hawkins1, Anna Smielewska2, Alex Howard3, Julie Grant1, Sajid Pervaiz1, Shahid Iqbal1, Arpad Toth1, Muhammad Saif 1

1Clatterbridge Cancer Centre, Liverpool, United Kingdom, 2Liverpool Clinical Laboratories, Liverpool, United Kingdom, 3Liverpool University Hospital Foundation Trust, Liverpool, United Kingdom

Background: Combining fludarabine with treosulfan (FT) has emerged as an effective conditioning strategy for reduced-intensity (RIC) and reduced-toxicity myeloablative (RTC) allogeneic hematopoietic stem cell transplantation (allo-SCT). In this retrospective study, we present real-world outcomes of allo-SCT comparing two different FT-based conditioning regimens utilizing different GVHD prophylaxis.

Methods: We conducted a retrospective analysis of patients undergoing allo-SCT with FT-based conditioning at our center from 2019 to 2023. Cohort included 95 patients, 29 received Fludarabine, Treosulfan and ATG (FTA) while 66 received Fludarabine, Treosulfan, Low dose TBI with post transplant cyclophosphamide (FTT). Fludarabine dose was 30 mg/m2 given at days -6 to -2, Treosulfan dose was 10 or 14 gram/m2 (decided on basis of HCT CI and age) given on days -6 to -4.

ATG dose of 2.5 mg/kg/day was given on days -3 and -2 (total 5 mg/kg) and LDTBI dose was 2 Gy. Post Transplant Cyclophosphamide (PTCy) was given at a dose of 50 mg/kg/day on days +3 and +4. The primary outcome was overall survival (OS), with secondary outcomes including relapse-free survival (RFS), graft-versus-host disease (GvHD)-free survival (GFS), relapse, GvHD and cumulative incidence of infection within the first 100 days post-transplant.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (28)

Results: Median OS was not reached in both groups at the time of analysis. FTA group had shorter median follow up of 442 days (range 60 – 687 days) whilst FTT group had a median follow up of 840 days (range 21 – 1569 days). Two year probability of survival was 67.7% in FTA group compared to 73.3% for FTT (p= 0.17). GRFS was also similar HR 1.11, 95% CI (0.49 – 2.48), P=0.78. There was no difference in relapse incidence between two groups with a relapse rate of 13.79% in FTA and 21.2% in FTT (P=0.57). Whilst median time to neutrophil engraftment was also similar in both groups (FTA 17.8 days, FTT 17.38 days, P=0.61), platelet engraftment was significantly delayed in the FTT cohort (28 days) compared to FTA group (19.5 days, p=0.000). FTA had acute GVHD (grade II-IV) incidence of 13.79%, while the FTT group had aGVHD (II-IV) incidence of 5.71%, P=0.22.

In FTA group, chronic GVHD (all severities) incidence was 24%. In FTT group this incidence was 20% (p=0.7). However, all chronic GVHD cases in FTA cohort were mild while in FTT 61.5% had mild and 38.46% had moderate to severe chronic GVHD as per NIH criteria.

There was a higher incidence of infections in the FTA regimen in the first 100 days post-transplant. Within evaluable cases, rate of viral infections in FTA was 93.1% and 57.14% in FTT, P=0.0014. EBV was the most common viral infection in the FTA (68.9%). Also, incidence of bacteremia in FTA group was 48.28% whilst that in FTT was 22.8% (p=0.039).

Conclusions: Our findings are consistent with reported results from other retrospective and prospective studies, underscoring safety and efficacy of FT-based conditioning regimens. ATG effectively mitigates severity of chronic GVHD relative to PTCy but increases incidence of infection. PTCy was associated with slower platelet engraftment in this cohort.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P198 IMPACT OF CO-ADMINISTERED ACETAMINOPHEN ON BUSULFAN PHARMACOKINETICS

Rutvij Khanolkar 1, Shahbal Kangarloo1, Jan Storek1

1University of Calgary, Calgary, Canada

Background: Although the interaction between acetaminophen and busulfan has been theorized due to overlap in metabolic pathways, no supporting empirical evidence has been reported to date. Here we report on a secondary analysis of an open-label, phase II clinical trial (NCT#03456817) of high-dose antithymocyte globulin (ATG, Thymoglobulin, 10mg/kg starting on day -4) compared standard low-dose ATG controls (4.5mg/kg ATG starting on day -2). In the high-dose and control patients, other myeloablative conditioning was identical and consisted of busulfan given from days -5 to -2. Prophylaxis of ATG-induced fever was with acetaminophen, which overlapped with busulfan on days -4 to -2 for high-dose ATG patients only, but on only day -2 for controls. Therefore, we investigated whether the increased co-administration of acetaminophen and busulfan would result in busulfan accumulation in high-dose ATG patients.

Methods: The study included 56 allogeneic hematopoietic cell transplant (HCT) recipients. On day -8, a test dose of Bu was administered and pharmacokinetics (PK) determined, based on which the daily Bu dose for days -5 to -2 was set (targeting total busulfan AUC of 15,000 uM.min). Bu daily area under the curve (AUC) was determined on days -5 and -2. The primary hypothesis was that the increase from day -5 AUC to day -2 AUC (ΔAUC) would be higher in high-dose ATG patients compared to low-dose ATG controls. Statistical comparison of ΔAUC between groups was conducted using Mann-Whitney test.

Results: The median ΔAUC was significantly higher in high-dose ATG patients (n=9) compared to controls (n=32) (25% vs. 3%, P=0.001). Based on this finding, the trial protocol was amended to give ibuprofen for fever prophylaxis to avoid supratherapeutic busulfan dosing. This resulted in a significantly decrease in ΔAUC for subsequent high-dose ATG patients (Group C; n=15) when compared to the initial 9 high-dose ATG patients that received acetaminophen (12% vs. 25%, P=0.034).

Conclusions: Acetaminophen likely increases busulfan AUC. This should be taken into consideration when acetaminophen is given to patients undergoing conditioning with busulfan.

Disclosure: The phase II study of high-dose ATG is supported in part by Sanofi.

16: Conditioning Regimens

P199 TREOSULFAN-BASED CONDITIONING REGIMEN IN PEDIATRIC ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A CASE SERIES OF SINGLE CENTER EXPERIENCE

Ilgen Sasmaz 1,2, Ali Antmen1, Utku Ayguneş1, Barbaros Karagun1, Duygu Turksoy1

1Acibadem Adana Hospital, Adana, Turkey, 2Cukurova University, Adana, Turkey

Background: Treosulfan is being increasingly used as part of conditioning regimens in pediatric allogeneic hematopoietic stem cell transplantation (HSCT) for both malignant and nonmalignant diseases. It has a low-toxicity profile, with the most commonly reported acute toxicities being skin, including nappy rash; diarrhea; mucositis; and hepatic toxicity; however, these are generally mild, and importantly, veno occlusive disease (VOD) is very rare. There are few reports using od treosulfan for allogeneic hematopoietic cell transplantation in children. We report our experiences with treosulfan based conditioning regimens in pediatric patients at our center.

Methods: A total of 545 allogeneic hematopoietic stem cell transplantations were performed at Acıbadem Adana Hospital Pediatric Bone Marrow Transplantation Unit in Turkey from 2013 to 2023. Sixty two patients out of 545 patients were conditioned for allogeneic hematopoietic cell transplantation with a treosulfan. Medical records and treatment modalities were evaluated retrospectively.

Results: In this study, 62 patients were conditioned with treosulfan, age ranging from 9 months to 17 years with an average of 8.8 years. Twenty nine patients were males, 33 were female. There were 31 patients with thalassemia major, 11 patient with sickle cell anemia, 13 patients with severe immune deficiency, 4 patients with familial hemophagocytic lymphohistiocytosis, one patient with myelodysplastic syndrome, one patient with metachromatic leukodystrophy, one patient with Diamond Blackfan anemia. Twenty six patients had HSCT from an unrelated donor. One patient with Griscelli syndrome had a haploidentical transplantation from mother. Patients received intravenous treosulfan doses of 10-14 g/m2/day on days -6 to -4. The dose of treosulfan was changed according to blood level in 14 patients. The dose of treosulfan was increased in seven of 14 patients, and the dose of treosulfan was reduced in seven of them. There was no serious toxicity. Severe mucositis was seen in three patients. Nine patients had limited skin toxicity including pigment changes, and occasional peeling. Three patients showed skin erosions and exfoliation. Minimal liver toxicity occurred in two patients. VOD developed only in one patient with thalassemia majör class 3. Four patients died in the first 100 days due to severe GVHD and sepsis.

Conclusions: In this retrospective study, we show that treosulfan-based conditioning regimen is a safe and effective treatment option in pediatric HSCT recipients, even in those with high-risk pre-HSCT clinical features or those with no HLA-identical family donor.

Disclosure: No.

16: Conditioning Regimens

P200 IMPLEMENTATION OF MACHINE LEARNING TO MEASURE THE IMPACT OF OVER- AND UNDER-EXPOSURE, AND PREDICT BUSULFAN’S PHARMACOKINETICS, IN PATIENTS RECEIVING CONDITIONING PRIOR TO HSCT

Dorian Protzenko1, Laurent Bourguignon2, Benjamin Bouchacourt 3, Raynier Devillier3, Joseph Ciccolini1

1COMPO Research Laboratory, Marseille, France, 2CHU de Lyon, Lyon, France, 3Institut Paoli Calmette, Marseille, France

Background: Bayesian adjustment of high-dose Busulfan has been implemented in our hospital, in order to control the exposure of patients receiving HSCT. Two exposure targets were considered, according to patient’s clinical condition: 16000 and 20000 µmol/L*h. However, despite this pharmacological monitoring, 17% of our patients had an exposure deviating by more than 20% from the overall target, and 35% were under- or over-exposed on at least one administration (defined as +/- 20% from the infusion exposure target).

We aimed to measure the impact of these under or over-exposures on the 1-year survival probability, and to propose a new methodology for exposure prediction.

Methods: This work is a retrospective study of patients that received pharmacokinetics guided dosage of Busulfan between March 2014 and July 2022 (n= 72 patients for 165 blood samples). Clinical, biological, and pharmacokinetics variables were collected. Statistical analysis and predictive model development were performed on R (version 4.3). The impact of under and over-exposure was estimated by incorporating exposure targets (cumulative AUC, maximal concentrations) into a 1-year survival predictive model. A 2nd model was computed to predict Busulfan’s exposure.

For each model, the performances of different formulas and algorithms were compared using sensibility/specificity, AUC-ROC, and the relative root-mean-square error (RMSE) of the predictions, after a 10-times 5-folds repeated cross-validation. Variable importance and model’s interpretation were assessed using Shapley values. Performances of the Busulfan’s exposure predictive model were compared to the current methodology performance using a Wilcoxon signed-rank test.

Results: Using exposure targets, biological variables at baseline (YGT, albuminemia, GFR rate) and a gradient boosting machine algorithm, we were able to predict the 1-year survival with a 0.8 mean AUC-ROC, a 80-85% specificity, for a 75-80% sensibility. Based on Shapley values, cumulative Busulfan AUC and maximal concentrations were the features the most strongly associated with the 1-year survival prediction. This model also highlights the need for more fine-tuning exposure targets.

Prediction of Busulfan exposure was performed using a Random Forest model, variables highlighted from scientific literature (age, ferritinemia, BMI/BSA), and using an exposure variable (the AUC reached on the first infusion). AUC was predicted with a relative-RMSE of 18%, lower than the current methodology (27.5%, p<0.05). Ferritinemia was negatively correlated to Busulfan exposure, when BMI/BSA and age were positively correlated. Higher exposure during the first infusion was associated with higher exposure in late infusions.

Conclusions: Both cumulative AUC and maximal concentrations have an impact on the 1-year survival. Over exposure, even compensated using Bayesian dose adjustments, can lead to a lower 1-year survival probability. Our survival model demonstrated good discriminating ability.

Busulfan exposure can be predicted using machine learning, using both clinical and biological variables. Early over exposure may lead to higher over exposure in the late infusions. Our predictive exposure model outperformed the existing methodology, showcasing its potential for improved precision in dosage adjustments. This new approach will be improved with external data, then implemented in order to reduce under- and over-exposure during the first infusion, before adapting the dosage of subsequent doses by Bayesian estimation.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P201 MITOCHONDRIAL COPY NUMBER VARIATION AS A SURROGATE MARKER OF ACQUIRED RESISTANCE TO THE BIFUNCTIONAL ELECTROPHILIC CHEMOTHERAPEUTIC DRUG BUSULFAN

Vid Mlakar1, Simona Jurkovic Mlakar1, Yvonne Gloor1, Isabelle Dupanloup 1, Yoann Sarmiento1, Denis Marino1, Mary Boudal-Khosbheen1, Chakradhara Rao Satyanarayana Uppugunduri1, Marc Ansari2,1

1University of Geneva, Geneva, Switzerland, 2University Geneva Hospitals, Geneva, Switzerland

Background: Mitochondria are the main source of cellular reactive oxygen species (ROS) and are also involved in the activation of apoptosis. Thus, changes in the number and functioning of the activity of mitochondria may affect the sensitivity of cells to cytotoxic agents. Little is known about the role of mitochondria in the sensitivity of myeloid cell lines such as MOLM13 to an alkylating agent, busulfan (BU) that is used in conditioning regimens before hematopoietic stem cell transplantation. In this study, we investigated how mitochondrial copy number (mtDNA-CN) and gene expression change after prolonged treatment with ROS-generating compounds BU and also with the DNA synthesis inhibitor cytarabine (Cyt).

Methods: MOLM13 cells were exposed to five consecutive BU treatments to acquire BU resistance (5TBU). The 5TBU and control cell lines were then exposed to five consecutive Cyt treatments to obtain cells resistant to both BU and Cyt, or Cyt alone (5TBU‒5TCyt or 5TCyt, respectively). We estimated the 50% inhibitory concentration (IC50) of BU in these cells. We confirmed our results by comparing the IC50 of BU in lymphoblastoid cells (LCLs) and their mitochondria content. We quantified mtDNA-copy number (CN) by real-time quantitative PCR of the MT-ND1 mitochondrial gene. We compared the gene expression profiles of control and resistant cell lines to BU using bulk RNA-sequencing.

Results: 5TBU MOLM13 cells showed a 2.6-fold increase in BU-IC50 values and had a 1.42-fold increase in mtDNA-CNs compared to control cells (p < 0.05). No mtDNA-CN gain was observed following treatment with Cyt. Similarly, mtDNA-CNs and BU IC50 are associated in LCLs but not Cyt. Genes that were found to be differentially expressed between control and resistant cell lines to BU were enriched in pathways involved in lipid and cholesterol metabolism.

Conclusions: We showed that mtDNA-CNs were increased with cellular resistance to BU, but not to Cyt, in both MOLM13 cells and LCLs, suggesting that mitochondria might play a role in the sensitivity of cells to cytotoxic agents. Our results also suggest that lipid and cholesterol synthesis could be involved in cellular response to BU.

Disclosure: No conflict of interest.

16: Conditioning Regimens

P202 CLOFARABINE, MELPHALAN AND THIOTEPA REDUCED-INTENSITY CONDITIONING CHEMOTHERAPY ALONG WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE-BASED GRAFT VERSUS HOST DISEASE PROPHYLAXIS FOR ALLOGENEIC STEM CELL TRANSPLANT

Alejandro Del Rio Verduzco1, Aileen Go1, Murali Kodali1, Matthew Ulrickson1, Nicolas De Padova1, Eric Smith1, Ivan Komerdelj1, Hayley Heers1, Jennifer Brookes1, Haley Pebler1, Rachael Yasuda1, Rajneesh Nath 1

1Banner MD Anderson Cancer Center, Gilbert, United States

Background: The recent national shortage of fludarabine led to the investigation of clofarabine as an alternative purine antimetabolite therapy in conditioning chemotherapy regimens for patients receiving allogeneic stem cell transplant (allo-HSCT). The combination of thiotepa, melphalan, and clofarabine (TMC) has been described in previous studies, with clofarabine doses ranging between 50 mg/m2 x 4 days and 20 mg/m2 x 5 days (Lang, Blood, 11 no. 110 10.1182, Boulad, Blood, 21 no. 120, 10.1182). We herein present our experience of utilizing a clofarabine dose of 30 mg/m2 x 5 days in combination with melphalan and thiotepa.

Methods: This IRB approved retrospective analysis included patients over age 18 years who underwent their first allo-HSCT with at least 30 days of follow-up at our institution using TMC as conditioning chemotherapy. Patients were excluded if they received an allo-HSCT from umbilical cord donor source or received methotrexate-based graft vs. host disease (GVHD) prophylaxis.

Results: 15 patients underwent allo-HSCT since 2022 using TMC chemotherapy conditioning. Median age was 60 (20-77) years. 10 (67%) patients were male and 5 (33%) female. 10 patients had a diagnosis of acute leukemia, (AML 5, ALL 4, AUL 1), 4 had MDS, and 1 had CML. Median HCT-CI was 3 (1 – 6). Modified Disease Risk Index (DRI) was Low/Intermediate in 8 (53%) and High in 7 patients (47%). Donor sources were MUD in 8 patients (53%), MMUD in 2 patients (13%), haploidentical in 3 patients (20%) and MRD/MMRD in 2 patients (13%). Patients received reduced intensity conditioning (RIC) with clofarabine 30 mg/m2, melphalan (50-140 mg/m2) +/- Thiotepa (1-5 mg/kg). All patients received GVHD prophylaxis with post-transplant cyclophosphamide 50 mg/kg on days +3 and day + 4 post-HSCT. Additional GVHD prophylaxis starting day +5 included tacrolimus/sirolimus in 7 patients (50%), and tacrolimus/sirolimus/rabbit ATG in 7 patients (50%).

All patients engrafted neutrophils and platelets with a median time to engraftment of 19 days (19-21), and 17 days (16-21) days respectively. 13 (87.5%) patients are alive at a median follow-up of 159 days. Non-relapse mortality occurred in 2 (13%) patients (GI toxicity; CVA). All patients have 97% or higher reported donor chimerisms 30 days after allo-HSCT. Grade 2-4 GVHD was reported in 4 (27%) patients (grade 3-4; 1). Out of the 10 patients who were followed for more than 100 days, 4 (40%) developed limited stage chronic GVHD. 4 patients (27%) experienced grade 3 or higher diarrhea suspected to be due to conditioning chemotherapy. 6 (40%) patients reported nausea/vomiting, with 2 (13%) instances being grade 3 or higher. 4 (27%) patients developed mucositis, 3 (20%) were grade 3 or higher. No instances of either sinusoidal obstructive syndrome (SOS) or capillary leak syndrome (CLS) occurred.

Demographics and Clinical Characteristics, n (%)

N=15

Recipient’s age at time of SCT, Median Years (IQR)

60 (49, 73)

Recipient gender, n (%)

Male

10 (67%)

Recipient’s diagnosis, n (%)

Acute myeloid leukemia

5 (33%)

Acute lymphoblastic leukemia

4 (27%)

Other

6 (40%)

Prior lines of therapy, n (%)

1

12 (80%)

2

3 (20%)

HCT Comorbidity Index, n (%)

1

2 (13%)

2

2 (13%)

3+

11 (74%)

Disease status at time of SCT, n (%)

Complete remission

12 (80%)

Active disease

3 (20%)

MRD status at time of SCT, n (%)

Negative by flow cytometry and/or next generation sequencing

8 (67%)

  1. IQR interquartile range, SCT stem cell transplant, MRD minimal residual disease
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (29)

Conclusions: Allo-HSCT with TMC RIC results in successful neutrophil and platelet engraftment comparable to a historical cohort that received thiotepa, melphalan and fludarabine (Nath R, CLML, 2023 10.1016). Mortality, severe GVHD, and toxicity were also observed to be relatively low. Small sample size precludes any meaningful subset or multivariate analysis.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P203 THE EFFICACY OF MODIFIED MELPHALAN AND BUSULFAN-BASED CONDITIONING REGIMEN FOR ASCT IN LOW-RISK AND INTERMEDIATE-RISK AML PATIENTS

Shulian Chen1, Xiaoyu Zhang1, Yi He1, Sizhou Feng1, Mingzhe Han1, Xingli Zhao2, Jie Bai3, Lijuan Li4, Zhihua Zhang5, Ying Wang1, Jianxiang Wang1, Weihua Zhai 1, Erlie Jiang1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China, 2Tianjin People’s Hospital, Tianjin, China, 3The Second Hospital of Tianjin Medical University, Tianjin, China, 4Tianjin Medical University General Hospital, Tianjin, China, 5The Affiliated Hospital of Chengde Medical University, Chengde, China

Background: Autologous stem cell transplantation (ASCT) is an important treatment option for some acute myeloid leukemia (AML) patients. However, high relapse rate is still a problem that cannot be ignored. The selection of conditioning regimen was crucial to reduce relapse rate after transplantation. In this study, we made refinements in the conditioning regimen with two alkylating agents, namely MCBA (the combination of melphalan, cladribine, busulfan, and cytarabine). We aim to investigate the efficacy of MCBA conditioning regimen for ASCT in low-risk and intermediate-risk AML patients who achieved CR after one course of induction chemotherapy.

Methods: This prospective multi-center clinical trial enrolled low-risk and intermediate-risk AML patients (non-M3) who achieved CR after one course of induction chemotherapy, followed by 1-3 courses of high-dose cytarabine consolidation chemotherapy, and underwent ASCT from May 2021 to June 2023 (ChiCTR Registration ID: ChiCTR2200056167). The MCBA conditioning regimen consists of melphalan 70mg/m2/d, day -6~-5, cladribine 5mg/m2/d, day -4~-2, busulfan 3.2mg/kg/d, day -8~-7, cytarabine 2g/m2/d, day -4~-2.

Results: This study included 21 AML patients from 5 tertiary hospitals in China. 14 males, 7 females, with median age 41 years (ranged 20–56 years). According to the 2017 European LeukemiaNet (ELN) criteria, 8 low-risk and 13 intermediate-risk. Neutrophil and platelet engraftment were achieved in all patients, with a median time of 13 days (ranged 11-27 days) and 32 days (ranged 12-150 days) respectively. With a median follow-up of 410 days (ranged 163-918 days), the estimated 1-year overall survival (OS) and 1-year disease-free survival (DFS) were 88.7±7.6% and 70.9±11.2%, respectively. 5 patients relapsed within 1 year, and the median time from ASCT to relapse was 300 (101-340) days. The probability of relapsed rate was 29.1±11.2%. It is worth noting that 6 patients were reclassified as adverse-risk according to the 2022 ELN criteria, and 3 of them relapsed. We speculate that patients who benefit from ASCT could be better selected based on the 2022 ELN criteria. Mucositis was the main reported regimen-related toxicity, mostly were mild and well tolerated.

Conclusions: The preliminary data demonstrated the efficiency and well tolerance of MCBA conditioning regimen for ASCT in low-risk and intermediate-risk AML patients who achieved CR after one course of induction chemotherapy. According to the 2022 ELN criteria, we can better screen patients and improve the efficacy of ASCT.

Clinical Trial Registry: ChiCTR Registration ID: ChiCTR2200056167.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P204 SECOND ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH REDUCED-INTENSITY CONDITIONING AND DONOR CHANGES IN HEMATOLOGICAL NON-MALIGNANCIES AFTER THE FIRST TΑΒ + CELL DEPLETED HAPLO-HSCT

Jianyun Liao 1, Chaoke Bu1, Lan He1, Jujian He1, Weiwei Zhang1, Zongxin Feng1, Yanfeng Zhou1, Bo Lyu1, Wenqian Shi1, Jing Wang1, Yuelin He1, Chunfu Li1

1Nanfang-Chunfu Children’s Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China

Background: The purpose of this study was to assess the safety and efficacy of a second allogeneic hematopoietic stem cell transplantation (allo-HSCT) with reduced-intensity conditioning (RIC) in patients with hematological non-malignancies who had implant failure after the first Tαβ+ Cell Depleted haplo-HSCT (TDH).

Methods: Between January 2020 and February 2023, 18 patients with hematological non-malignancies suffered implant failure in our center were enrolled and retrospectively examined. Donors were replaced in all patients for the second allo-HSCT after the first TDH. The conditioning protocol was as follows: post-transplant Cyclophosphamide (PTCy) as a baseline preventive treatment with overall RIC, Busulfan (BU): 100 mg/m2/d qd×1d, Thiotepa (TT) 5mg/kg/d qd×1d, Fludarabine (Flu): 30mg/m2 qd×3d, ATG-F (Fresenius): 10mg/kg/d, qd×2d.

Results: The median interval between the first and second HSCT was 26 days (range: 19-63). All patients had long-term engraftment with median engraftment times of 19 days for ANC to reach 0.5x109/L, 21 days for HB to reach 80g/L, and 26 days for platelets to reach 20x109/L, respectively. The mean infused mononuclear cells were 20.4×108/kg and CD34+ cells were 17.77×106/kg. The grade II-IV GVHD and III-IV severe acute GVHD rates were both 5.6%. Chronic GVHD was found in 22.22% of limited patterns. CMV and HHV6 reactivation rates were 16.67% (n=3) and 44.44% (n=8), respectively. No EBV infections occurred. B19 and ADV reactivation rates were both 5.56% with one case separately. Hemorrhage cystitis occurred in 44.4% of cases (n=8), grade I or II. Pulmonary infection occurred in 10 patients, viral encephalitis in 1 patient, and purulent meningitis in 1 patient. The disease-free survival (DFS) and overall survival (OS) rates of all patients were both 100%, with a median follow-up of 31 (10-39) months.

Conclusions: With our RIC regimens, donor change, and post-transplant maintenance therapy, the second allo-HSCT in relapsed hematological non-malignancies after the first TDH is a safe and effective treatment with high OS and DFS. Donor replacement is recommended for the second allo-HSCT.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P205 FIRST ALLOGENIC STEM CELL TRANSPLANTATION IN A PATIENT WITH ROIFMAN SYNDROME AND EBV-ASSOCIATED PLASMABLASTIC LYMPHOMA RELAPSE USING AUC CONTROLLED BUSULFAN BASED MYELOABLATIVE CONDITIONING

Marie Sophie Knape1, Katja Gall1, Katrin Vollmer1, Florian Ressle1, Felicitas Ferrari von Klot1, Angela Wawer1, Michael H. Albert2, Chaim M. Roifman3, Britta Eiz-Vesper4, Birgit Burkhardt5, Wilhelm Woessmann6, Irene Teichert von Lüttichau1, Julia Hauer1, Uwe Thiel 1

1Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany, 2Dr von Hauner Children’s Hospital, University Hospital, Ludwig Maximilians Universität München, Munich, Germany, 3Canadian Centre for Primary Immunodeficiency, Immunogenomic Laboratory, Division of Immunology and Allergy, The Hospital for Sick Children and the University of Toronto, Toronto, Canada, 4Institute of Transfusion Medicine and Transplant Engineering, Hannover Medical School, Hannover, Germany, 5Non-Hodgkin’s Lymphoma-Berlin-Frankfurt-Münster (NHL-BFM) Study Center and Pediatric Hematology and Oncology, University Hospital Muenster, Muenster, Germany, 6NHL-BFM Study Center and Pediatric Hematology and Oncology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: We successfully allo-transplanted a child with Roifman syndrome and EBV-associated plasmablastic lymphoma relapse using a myeloablative busulfan based conditioning regimen.

Methods: We describe a currently 8 year old female patient with a plasmablastic lymphoma relapse and Roifman syndrome who received allogenic SCT using AUC controlled Busulfan based myeloablative conditioning. Born small for gestational age the patient developed retinopathy, severe eczema, nephrocalcinosis and immunodeficiency.

In 10/2021 she presented with a spinal manifestation of a plasmablastic lymphoma (EBV positive, CD 20 negative, CD30 positive) which left her paraplegic. She received emergency laminectomy and was then treated with 6 cycles of DA EPOCH and intrathecal therapy. She relapsed in 09/2022 and received salvage chemotherapy with ifosfamide, carboplatin and etoposide followed by brentuximab vedotin treatment. For consolidation she received allogenic SCT in 02/2023 (MUD 10/10, EBV positive, female) after myeloablative conditioning with alemtuzumab, fludarabin und busulfan (cumulative AUC target dose: 85mg*h/L). GvHD-prophylaxis consisted of mycophenolat-mofetil and ciclosporin A.

Results: Despite a mild acute skin GVHD grade I-II, which was successfully treated with steroids the patient had an otherwise uncomplicated conditioning and SCT course. Stable neutrophil engraftment was achieved at d + 16, platelet engraftment on d + 15. On day +30 the patient developed EBV-reactivation associated with early multifocal plasmablastic lymphoma relapse. Immunosuppression was discontinued immediately, and rituximab was administered without any permanent effect. As compassionate use salvage therapy she was then treated with daratumumab, a monoclonal CD38 antibody, which lead to a very good partial response until disease progression in 10/23. The current therapy includes palliative radiation combined with a switch to a different CD38 monoclonal antibody (isatuximab) and adoptive third party EBV-specific T cell transfer.

Conclusions: We describe the first patient with Roifman syndrome who received an allogenic SCT due to plasmablastic lymphoma relapse. AUC controlled busulfan administration was well tolerated and enabled early and durable engraftment. CD38 targeted antibody treatment showed promising results in the treatment of the plasmablastic lymphoma relapse in this patient and could be considered earlier in the course of the disease.

Disclosure: The authors declare no conflict of interest. UT was funded by the Wilhelm Sander-Foundation (2018.072.1. and 2021.007.1.) and the Cura Placida Children’s Cancer Research Foundation (20210823). UT is currently funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—Project number 501830041, the “Zukunft Gesundheit e.V.”, the Dr. Sepp and Hanne Sturm Memorial Foundation and the Robert Pfleger Foundation.

16: Conditioning Regimens

P206 HIGH GRAFT FAILURE WITH FLUDARABINE, TREOSULPHAN CONDITIONING REGIMEN IN MYELOID MALIGNANCIES

Taner Tan1, Sinem Civriz Bozdağ 1, Ümit Barbaros Üre1, Elif Birtaş Ateşoğlu2, Emre Osmanbaşoğlu1, Ahmet Burhan Ferhanoğlu1, Hakan Kalyon1, Meltem Olga Akay1

1Koç University Hospital, Istanbul, Turkey, 2Yeditepe University, Istanbul, Turkey

Background: Fludarabine, busulphan has been widely used as conditioning regimen in allogeneic stem cell transplantation(alloHSCT) of myeloid malignancies. Recently, combination of fludarabine with treosulphan has been introduced as less toxic but regimens with similar efficacy especially in elderly patients. In our study, we aimed to analyze the outcome of treosulphan in myeloid malignancies who had alloHSCT.

Methods: We had retrospectively included twenty eight myeloid malignancy patients; who had alloHSCT with fludarabine, treosulphan conditioning regimen in Koç University School of Medicine BMT Unit. GVHD prophylaxis has been performed with cyclosporine and mycophenolate mofetil. Posttransplant cyclophoshamide has been used in all unrelated stem cell transplantations except 2 and in all haploidentical stem cell transplantations.

Results: We have included 16 AML, 7 Myelofibrosis, 4 High risk MDS and 1 CMML-2 patient. Seven intermediate 2 or high risk myelofibrosis patients had reduced intensity fludarabine treosulphan conditioning regimen. Stem cell source was peripheral blood in all patients and donors were unrelated in 5 out of 7 patients. Five patients had massive splenomegaly although ruxolitinib or radiotherapy has been used to reduce spleen size before transplantation. Median follow up of the patients were 7 months. Median day of neutrophile and platelet engrafmentwas 16,5(14-28) and 28(21-113), respectively. Neutrophile engraftment could not be observed in 1 patient and platelet engraftment could not be observed in 2 patients. One patient had a second alloHSCT as a consequence of graft failure and had no response. 3 out of 7 patients died during the follow up 2 in posttransplant 100 days. Acute GVHD has been observed only in 1 patient.

Median age of the patients excluding myelofibrosis 52(26-71), median follow up was 6.9 (0.4-52) months. Two years overall survival was observed in 20% of the patients and 23% of the patients progresed in the follow up. Neutrophile engraftment was observed in 12 (10-14) days. Neutrophile engraftment could not be observed in 3 patients. Platelet engraftment was observed in 17 (10-40) days and could not be observed in 6 patients. Acute GVHD has been observed 33 % of the patients.

Conclusions: Engraftment has been observed as a major issue with treosulphan based regimens in myeloid malignancies. In myelofibrosis group most of the patients had massive splenomegaly and in acute leukemia group majority of the patients had advanced disease which can explain the high engraftment failure rates. Progress and GVHD was not different with busulphan based regimens which have been used in previous studies.

Disclosure: Nothing to declare.

16: Conditioning Regimens

P207 ALLOGENEIC TRANSPLANTATION USING MYELOABLATIVE CONDITIONING REGIMES IS SAFE AND EFFECTIVE IN ELDERLY PATIENTS OVER 60 YEARS WITH AML AND MDS

Yan-Li Zhao 1, Yue Lu1, Xing-Yu Cao1, Jian-Ping Zhang1, Jia-Rui Zhou1, Zhi-Jie Wei1, Min Xiong1, De-Yan Liu1, Rui-Juan Sun1

1Hebei Yanda Lu Daopei Hospital, Langfang, China

Background: The median age at diagnosis of acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) are 68 and 75 years respectively, and the prognosis worsens with increasing age. For many patients, allogeneic hematopoietic cell transplantation (allo-HCT) is the only chance for cure. While allo-HCT for elderly patients over 60 years is increasing in the Western world, it remains rare in China, comprising less than 5% of all transplants according to data from CBMTR. Reduced-intensity conditioning (RIC) regimens are commonly chosen for elderly patients due to their lower transplant-related mortality (TRM), but they come with a higher risk of relapse compared to myeloablative conditioning (MAC).

Methods: The retrospective study analyzed 80 patients over 60 years, diagnosed with AML (n=55) and MDS (n=25), who underwent their first transplant at our center between March 2012 and March 2023. The cutoff date was October 2023, with a median follow-up time of 18.5 months (range, 7-110 months) for survivors.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (30)

Results: More than half patients underwent transplants in the last 3 years. The median age was 63 (60-71) years, with 20 patients aged 70 years or older. Disease status of 55 AML cases was 32(58.2%) in the first complete remission (CR1), 9(16.4%) in the second complete remission (CR2) and 14(25.5%) in no remission (NR). MDS patients included 7 with MDS EB1 and 17 with EB2. Donors were predominantly haploidentical (59 cases), with the remainder being identical siblings (11 cases) or matched-unrelated (10 cases). The median donor age was 35.5 (15-58) years old. Myeloablative regimens were used in all patients, including 79 cases with busulfan-based (3.2 mg/kg/day×3.5~4), and 1 with total body irradiation-based (TBI, 2Gy×5). Among Bu-based conditioning regimens, BuFlu were applied in 62 cases and BuCy in 17 cases. Antithymocyte globulin was applied for graft-versus-host disease (GVHD) prophylaxis. The median total mononuclear cell count (MNC) and the infused CD34+ cell count was 8.82×108/kg (range: 3.71-16.6 ×108/kg) and 4.79 ×106/kg (range: 4.75-14.83×106/kg). Except for 3 very early deaths (1 died of infection, 1 malignant arrhythmia and 1 early relapse), all patients received neutrophil engraftment at a median 14 days (range: 9-34 days) post-transplant. Seventy-four patients achieved platelet engraftment at a median 13 days post-transplant (7-33 days). The non-relapse mortality within 100 days was 6.7% (95%CI3.7-9.7) and at 1-year was 25.2% (95%CI 20.2-30.2). The incidence of grade II-IV acute GVHD (aGVHD) and grade III-IV aGVHD was 21% (95%CI 16.3-25.7) and 12.2% (95%CI 8.4-16.0), respectively. Cumulative incidence of relapses (CIRs) at 2-year was 15.1% (95%CI 11.6-19.6). The 2-year leukemia-free survival (LFS) and overall survival (OS) was 58.3% (95%CI 52.3-64.3) and 57.9% (95%CI 50.5- 60.3), respectively. For AML patients in remission, the OS was significantly better than patients in NR, with the 2-year OS of 63.1% (95%CI 54.2-72) vs. 42.9% (95%CI 39.7-56.1%) (p<0.001).

Conclusions: Our study demonstrated that allo-HCT using MAC conditioning regimes was a promising therapy for elderly patients with AML and MDS with a low relapse rate. Safety is manageable and the incidence of GVHD is relatively low. Achieve complete remission before transplantation can significantly improve survival.

Disclosure: Nothing to declare..

16: Conditioning Regimens

P208 COMPARISON BETWEEN DIFFERENT REDUCED INTENSITY CONDITIONING REGIMENS IN AML OR MDS

José Emilio Salinas 1, Yorman Flores1, Patricio Rojas1, Catherine Gutierrez1, Marcela Vidal1, Verónica Jara1, Elizabeth Rivera1, James Campbell1, María José Garcia1, Vicente Sandoval1, Maximiliano Vergara1, Mauricio Ocqueteau1, Felipe Palacios1, Pablo Sandoval1, Mauricio Sarmiento1

1Red de Salud Christus UC- Pontificia Universidad Católica de Chile, Santiago, Chile

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for several hematological diseases. Prospective and retrospective studies associate myeloablative conditioning regimens (MAC) with increased non-relapse mortality and less intense regimens with disease relapse, leading to similar overall survival.

Methods: Retrospective analysis of the different reduced-intensity conditioning regimens (RIC) used from 2013 to 2022 in a hematopoietic stem cell transplant reference center.

Results: The 3-year OS was 52%, 51%, and 57% in the reduced-intensity conditioning regimen based on total body irradiation, Melphalan, and Busulfan respectively (p=0.89). The 3-year progression-free survival (PFS) was 53%, 57%, and 55% in the reduced-intensity conditioning regimen based on total body irradiation, Melphalan, and Busulfan respectively (p=0.9).

Conclusions: We can conclude based on our data, that no reduced-intensity conditioning regimen is superior in terms of overall survival, progression-free survival and complications.

Clinical Trial Registry: No applicable.

Disclosure: Nothing to declare.

6: Experimental Transplantation and Gene Therapy

P209 PHASE 1 TRIAL RESULTS FOR PATIENTS WITH ADVANCED HEMATOLOGIC MALIGNANCIES TREATED WITH ORCA-T CELL THERAPY WITH REDUCED INTENSITY CONDITIONING AND SINGLE AGENT TACROLIMUS

Alejandro Villar-Prados 1, Katherine Sutherland1, Robert Negrin1, Sally Arai1, Matthew Frank1, Laura Johnston1, Robert Lowsky1, David Miklos1, Lori Muffly1, Saurabh Dahiya1, Andrew Rezvani1, Surbhi Sidana1, Parveen Shiraz1, Judith Shizuru1, Wen-Kai Weng1, Melody Smith1, Sushma Bharadwaj1, John Tamaresis1, Anna Pavlova2, Scott McClellan2, Everett Meyer1

1Stanford University, Stanford, United States, 2OrcaBio, Menlo Park, United States

Background: Reduced intensity conditioning hematopoietic bone marrow stem cell transplantation (HSCT-RIC) is the only curative option for many hematological malignancies in older/medically unfit patients. Strategies to minimize complications such as graft-versus-host-disease (GVHD) include donor graft engineering, which has not been widely tested in RIC due to a potential trade-off between engraftment and reduced GVHD with T cell depletion.

Methods: This is a single-center open-label phase 1 feasibility and safety study for dose escalation of conventional T cell infusion. Primary objectives include the incidence of grade III-IV acute GVHD, time of engraftment and donor T cell chimerism at day +30. Secondary objectives include relapse-free survival (RFS), severity of chronic GVHD and incidence of serious infections. There are two major independent trial arms: HLA-matched (either un/related (Arm A1-2)) and HLA-mismatched (7/8 mismatch; either un/related (Arm C). Arm A1 (11 patients) were conditioned with fludarabine 40mg/m2, melphalan 50 mg/m2 and 4 Gy of total body irradiation (TBI). Arm A2 (7 patients) received thiotepa 10 mg/kg, replacing melphalan. Arms A1-2 patients received single agent tacrolimus. Arm C (2 patients) received the same conditioning regimen as Arm A2 and dual-agent GVHD prophylaxis with tacrolimus and mycophenolate (taper Day +10, decreasing by 500 mg/week).

Results: A total of 18 HLA-matched and 2 HLA-mismatched patients have been recruited thus far in this interim dose escalation analysis. Combining both arms, the median patient age was 68 years (range 61 to 72), with 70% being male (14/20). 55% had acute myeloid leukemia (11/20), 15% had myelodysplastic syndrome (3/20), 10% had acute lymphoblastic leukemia (2/20), 1 patient with mixed phenotype leukemia (5%) and 15% had myelofibrosis (3/20). All acute leukemia patients were in complete remission (CR) or CR with incomplete count recovery prior to infusion of Orca-T. Median follow up was 9.65 months (range 2-22.6 months). Median time for neutrophil engraftment was 14.5 days (range 9 to 39). Based on these findings, no dose escalation of Tcon infusion was needed. None of the patients had infusion reactions nor engraftment syndrome. Median donor chimerism for CD15 was 100% at Day +30 (19/20, 99%-100%) and 100% (15/20, 98%-100%) at day 90. Median CD3 chimerism at Day +30 and +90 of transplant were 99% (19/20, 40%-100%) and 99% (15/20, 78%-100%), respectively. No patients developed grade III-IV acute GVHD. The incidence of moderate to severe chronic GVHD was 5% (1/20). The 12-month RFS thus far is 77% (95% CI 49% to 91%). The cumulative incidence for grade ≥3 infections in the first 90 days post-HSCT was 20%. No surviving patient has had disease relapse.

Conclusions: These early results demonstrate that Orca-T is a safe and feasible alternative to conventional transplant in the RIC setting, with robust and early donor myeloid and eventual full donor T cell engraftment. The low incidence of acute and chronic GVHD and disease relapse, suggest retention of graft-versus-leukemia effect. Few studies of donor graft-engineered products have been successfully evaluated in the RIC setting and merits larger multicenter studies evaluating GVHD-free and RFS post Orca-T to validate these findings.

Clinical Trial Registry: clinicaltrials.gov identifier: NCT05088356.

https://classic.clinicaltrials.gov/ct2/show/study/NCT05088356?term=everett+meyer&draw=2&rank=7.

Disclosure: -Robert Negrin: OrcaBio (Research funding), UpToDate (Patent and royalties), Cellenkos (consultant), Regimmune Inc (consultant), Garuda therapeutics (holder of stock options, board member/advisory committees), Amgen (board member/advisory committees), Co-immune (holder of stock options, board member/advisory committees), Appia Bio (board member/advisory committees), (Biorasi holder of stock options, board member/advisory committee)

-Matthew Frank: Roche/Genentech (holder of stock options), Kite (research funding). Consultant for: Allogene, Adaptive Biotech, EcoR1, BRVLH, Cargo Therapeutics, Gilead

-Robert Lowsky: OrcaBio (research funding)

-David Miklos: Research funding: Kite, Allogene, Adicet, 2Seventy Bio, Miltenyi, Celgene. Consultant: Adaptive Biotechnologies, Janssen, Bristol-Myers Squibb, A2Biotherapeutics, Amgen, Celgene, Genetech, Gilead, Incyte, Juno Therapeutics, LegendBiotech, MorphoSys, Mustang Bio, Navan Technologies, Novartis, Pharmacyclics, Umoja. BiolineRx (board member)

-Lori Muffly: Research funding: OrcaBio, BMS, Jasper, Kite, Astellas. Consulting: Amgen, Pfizer, Autolus

-Saurabh Dahiya: Consulting: Kite (and research funding), Bristol-Myers Squibb, Incyte, Adaptives Biotechnologies.

-Andrew Rezvani: Pharmacyclics (research funding)

-Surbhi Sidana: Research funding and consulting: Magenta Therapeutics, BMS, Jansen. Research Funding from Novartis, Allogene. Consulting: Sanofi, Takeda, Oncopeptides, Pfizer

-Judith Shizuru: Jasper therapeutics (consultant and current equity holder in publicaly traded company)

-Melody Smith: Consultant at BMS. Member on an entity’s board of directors or advisory committee at A28

-Anna Pavlova: OrcaBio (current employment)

-Scott McClellan: OrcaBio (current employment)

-Everett Meyer: OrcaBio (research funding)

-Alejandro Villar-Prados, Katherine Sutherland, Sally Arai, Laura Johnston, Parveen Shiraz, Weng-Kai Wen, Sushma Bharadwaj, John Tamaresis: nothing to declare.

6: Experimental Transplantation and Gene Therapy

P210 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH CONDITIONING INCLUDING DONOR CAR-T CELLS FOR R/R B-CELL LYMPHOMA AND R/R MULTIPLE MYELOMA FAILING CAR T-CELL THERAPY

Fan Yang1, Rui Liu 1, Zhonghua Fu1, Teng Xu1, Hui Shi1, Lixia Ma1, Guoai Su1, Biping Deng2, Tong Wu1, Xiaoyan Ke1, Kai Hu1

1Beijing Gobroad Boren Hospital, Beijing, China, 2Cytology Laboratory, Beijing Gobroad Boren Hospital, Beijing, China

Background: The prognosis of refractory/relapsed aggressive B-cell lymphoma (R/R B-NHL) and multiple myeloma (R/R MM) who failed to autologous chimeric antigen receptor-T (CAR-T) cells is extremely poor.

Methods: From September 2020 to November 2023, 18 patients were enrolled. The median age was 41 (26-64) years old. The diagnosis included DLBCL(NOS)(n=5), High grade B-cell lymphoma (n=6), Burkitt’s lymphoma(n=3) and Multiple myeloma (n=4). Eleven cases were with TP53 mutations. After bridging therapy, 7 patients achieved CR, 7 PR and 4 PD before transplantation who did not respond to multi-line therapies including ASCT (n=7) and local radiotherapy (n=7). Before the trial, the expression of CD19 and CD22 antigen in R/R B-NHL and BCMA antigen in R/R MM patients was positive confirmed by pathology. There were matched sibling donor in 4 cases, matched unrelated donor in 1 case and haploidentical donor in 13 cases. Conditioning with busulfan, fludarabine-based regimen combined with donor CAR-T cells was applied. Calmodulin inhibitor, mycophenolate mofetil, a short-term methotrexate and/or Anti-human Thymocyte Immunoglobulin were used for graft-versus-host disease (GVHD) prophylaxis.

Results: The median donor CAR-T cells infused were 2(range,0.2-4.88) ×106/kg. Cytokine release syndrome (CRS) occurred in 16 cases with 4 cases in grade III. No immune effector cell-associated neurotoxicity syndrome (ICANS) was observed. Granulocyte colony-stimulating factor–mobilized peripheral blood stem cells were infused 7 days after donor CAR-T with the median CD34+ cells 6 (range,3-8.19) ×106/kg. The neutrophil and platelets engraftment was achieved in all cases on median days 15 (range,11-24) and 16 (range,13-65) respectively post-transplant 0.18 cases (100%) were all donor type by STR analysis on bone marrow at 28 days after transplantation.2 cases of grade III acute GVHD were seen. For donor CAR-T cell expansion, the peak time in vivo was on median 5(range,1-20) days after infusion. The median peak lever of CAR-T cell was 50.81% (range,0.091-116) by PCR. Levels of CAR-T cells were very low after the first 2 months, and the longest lasting time was 239 days post-transplant. With a median follow-up of 8.9 (range, 0.69-33.8) months, 17 cases (17/18, 94.4%) achieved a CR, and the longest duration of response (DOR) reached 34.5 months. In 6/17 (35.3%) patients, the disease recurred after remission. Four patients (4/6, 66.6%) were given donor CAR-T after relapse and three patients (3/4, 75%) achieved CR again. The 2-year PFS and OS were 30.2% (95%CI, 9.5-54.3) and 40.4% (95%CI, 14.7-65.2), respectively. In comparison, PFS and OS were significantly longer in the CR and PR cohorts than in the PD cohort before transplantation, resulting in a median PFS of 8.6 vs. 10.2 vs. 1.9 months (P=0.0006). Four patients from the pre-transplant PD cohort died from disease progression. Four patients (4/18, 22.2%) died from infection.

Conclusions: Allo-HSCT with conditioning including donor CAR-T cells is a safe and effective strategy for R/R B-NHL and MM failing CAR-T cell therapy. CRS is manageable and has no influence on hematopoiesis reconstitution. Acute GVHD and viral reactivation was mild. The Poor efficacy was associated with high tumor burden before transplantation.

Clinical Trial Registry: ChiCTR2000040665 https://www.chictr.org.cn/showproj.html?proj=65108.

ChiCTR2100043251 https://www.chictr.org.cn/showproj.html?proj=121159.

Disclosure: Nothing to declare.

6: Experimental Transplantation and Gene Therapy

P211 SMADCAM-1 IS DECREASED AFTER ALLOHCT, ALONG WITH GUT MICROBIOTA DYSBIOSIS, AND IS ASSOCIATED WITH HEMATOPOIETIC RECOVERY

Karen Fadel 1, Lama Siblany1, Razan Mohty1, Nicolas Stocker1, Eolia Brissot1, Rémy Dulery1, Anne Banet1, Simona Sestili1, Zoe van de Wyngaert1, Agnes Bonnin1, Ramdane Belhocine1, Tounes Ledraa1, Antoine Capes1, Mohamad Mohty1, Béatrice Gaugler1, Florent Malard1

1Sorbonne Université, Paris, France

Background: During allogeneic hematopoietic cell transplantation (alloHCT), the high dose chemotherapy used in the conditioning regimen, along with broad-spectrum antibiotics, leads to gut microbiota dysbiosis, with a detrimental effect on patients’ outcomes. Furthermore, while it has been suggested that microbiota composition may influence hematopoiesis, and it was shown that fecal microbiota transplantation after alloHCT is associated with a higher neutrophil level, no study has evaluated the impact of microbiota dysbiosis on hematopoietic recovery. Recognizing the crucial role of the MAdCAM-1–a4b7 axis in the migration of hematopoietic cells outside the bone marrow, and recent findings indicating that a low sMAdCAM-1 level is a hallmark of gut microbiota dysbiosis, we aimed to assess whether the sMAdCAM-1 level is associated with hematopoietic recovery after alloHCT.

Methods: We analyzed the sMadCAM-1 level in a cohort of 279 patients that underwent an alloHCT in Saint-Antoine Hospital, Paris, between 2012 and 2018. sMadCAM-1 levels were assessed by ELISA on serum samples collected before the start of the conditioning regimen, at day 0 and at day 20, around hematopoietic recovery. Antibiotic exposure data were collected from day -30 to day 30 after alloHCT, with a particular focus on antibiotics with a broad anti-anaerobic spectrum (piperacillin-tazobactam, imipenem, and meropenem).

Results: We found a significant decrease in sMadCAM-1 level between the preconditioning evaluation and day 0 (median 8815 pg/mL, versus 4234 pg/mL, p<0.0001) and a further decrease in sMadCAM-1 between day 0 and day 20 (4234 versus 3355, p<0.0001). In a subgroup of 60 patients with stool and serum sampling at the same time point (day 0), we found a correlation between Shannon diversity index and sMadCAM-1 (spearman r, 0.21, p=0.056), and the median sMadCAM-1 concentration was significantly lower in patients with a low versus high Shannon diversity index (4685 versus 6240, p=0.04). We then assessed the impact of antibiotic exposure on sMadCA-1 level and found that early use of antibiotics (before day 0), was associated with a significantly lower sMadCAM-1 level at day 0 (3980 versus 4891, p=0.01). We then assessed the impact of sMadCAM-1 level at day 20 on engraftment. Median absolute neutrophil count (ANC >0.5 G/L) and platelet recovery (>20 G/L) were significantly lower in patients with higher sMadCAM-1 levels at day 20, being 15 days and 10 days, respectively, versus 16 days (p<0.0001) and 12 days (p=0.004), respectively, in patients with lower sMadCAM-1 levels. The day 28 cumulative incidence of ANC >0.5 G/L and platelets >20 G/L was significantly higher in patients with a higher sMadCAM-1 level, being 99.0% and 91.6% versus 94.8% (p=0.002) and 83.3% (p=0.02), respectively, in patients with lower levels.

Conclusions: We found that alloHCT is associated with significant decrease in sMadCAM-1 levels. This deacrease correlates with a diminished diversity in gut microbiota and was more pronounced in patients receiving broad-spectrum antibiotics. Furthermore, we found that sMadCAM-, known to be involved in hematopoietic cell output from the bone marrow, was associated with neutrophil and platelet recovery after alloHCT. These findings suggests that microbiota dysbiosis mediates its detrimental effect on hematopoiesis through the MAdCAM-1–a4b7 axis.

Disclosure: Nothing to declare.

6: Experimental Transplantation and Gene Therapy

P212 PENTOSTATIN AND CYCLOPHOSPHAMIDE DECREASE GRAFT REJECTION BUT INCREASE TRANSPLANT-RELATED COMPLICATIONS IN HAPLOIDENTICAL HCT FOR SICKLE CELL DISEASES

Emily Limerick1, Matthew Hsieh 1, Julia Varga1, Mary Lacy Grecco1, Jennifer Brooks1, Wynona Coles1, Jackie Queen1, Neal Jeffries1, Courtney Fitzhugh1

1National Heart, Lung, and Blood Institute/National Institutes of Health, Bethesda, United States

Background: Sickle cell disease (SCD) increases morbidity and causes early mortality. Hematopoietic cell transplantation (HCT) is a potentially curative therapy. While nonmyeloablative HCT can be safe and effective in adults with severe organ damage, early haploidentical (haplo) HCTs were plagued with high rates of acute graft rejection. Pentostatin and cyclophosphamide prevent murine allograft rejection.

Methods: We analyzed adults with first haplo HCT for SCD at the National Institutes of Health (NIH) from November 2010 through April 2023 on either of two nonmyeloablative protocols (NCT00977691, NCT03077542). Both protocols included alemtuzumab and 400cGy total body irradiation; the second protocol added pentostatin plus oral cyclophosphamide (PC) preconditioning. Graft-vs-host disease (GVHD) prophylaxis included post-transplant cyclophosphamide (50-100mg/kg) and sirolimus.

Results: The cohort included 39 patients. The median age was 32 years: 62% men and 92% HbSS. There were 18 (46%) patients on the original haplo protocol (haplo-1) and 21 (54%) on the PC protocol (haplo-PC). Demographic characteristics were similar between the protocols.

One-year overall survival was 95%; in the first year, there was 1 death on each protocol. At one year post-HCT there was more acute rejection on the haplo-1 compared to the haplo-PC group (8 (44%) vs. 1 (5%) p= 0.004). The median follow-up was longer for the haplo-1 cohort than haplo-PC (9.7 vs. 4.3 years; p < 0.0001). Two patients on the haplo-1 protocol developed myeloid malignancies; no patients on haplo-PC have developed a myeloid malignancy.

Given the differential follow-up between the two protocols, we examined rates of full (≥95% donor lymphoid and myeloid) compared to mixed chimerism in patients free of SCD at 4-years post-HCT. On haplo-1, 1 of 8 (12.5%) patients had full donor chimerism while 4 of 8 (50%) haplo-PC patients had full donor chimerism (p= 0.14). Of the patients who were alive and free of SCD at last follow-up, 4 (67%) haplo-1 patients were off immunosuppression and 11 (73%) in the haplo-PC cohort. Two (5%) patients on the haplo-PC protocol developed grade II-IV acute GVHD. No patients on either protocol developed moderate to severe chronic GVHD.

There was more viral reactivation on the haplo-PC protocol than haplo-1: 62% and 28% (p= 0.05), but viral disease rates were similar: 11% and 19% respectively (p= 0.67). There were more autoimmune complications in the haplo-PC protocol than haplo-1: 19% versus 0%, respectively (p= 0.11). While this is not a statistically significant difference, this is indeed clinically relevant.

Conclusions: This study demonstrates that nonmyeloablative haplo HCT for SCD with PC preconditioning decreases acute graft rejection with minimal GVHD. Notable complications associated with haplo-PC include viral reactivation and autoimmune complications. The haplo-PC protocol recently closed to accrual. A new regimen is indicated that optimally balances efficacy and toxicity, particularly in SCD patients with severe organ damage who are excluded from other haplo HCT studies.

Clinical Trial Registry: NCT00977691, NCT03077542.

Disclosure: Nothing to declare.

6: Experimental Transplantation and Gene Therapy

P213 GRANULOCYTES TO INDUCE DONOR-DERIVED T CELL EXPANSION AFTER T REPLETE, MISMATCHED CB IN POST-TRANSPLANT RELAPSED AND REFRACTORY PAEDIATRIC ACUTE LEUKAEMIA: RESULTS OF GRANS TRIAL

Srividhya Senthil 1, Roisin Borrill1, Denise Bonney1, Ramya Hanasoge-Nataraj1, Madeleine Powys1, Omima Mustafa1, Robert Wynn1

1Royal Manchester Children’s Hospital, Manchester, United Kingdom

Background: Refractory and post-transplant relapsed Acute Leukaemia remain difficult to cure. We have reported previously that third-party pooled granulocytes induce massive, transient, donor-derived T-cell expansion after Cord Blood (CB) transplant. These expanded T-cells are cytotoxic, memory and mostly CD8, in contrast to the infused graft T-cells which are predominantly naïve and CD4, and appear a priori candidate anti-tumour T-cells. In our highly innovative prospective clinical trial, GRANS study (NCT05425043), we have given such granulocytes peri-transplant with T-replete, deliberately mismatched CB to enhance the graft-versus-leukaemia effect of the transplant, in those with the highest risk disease, namely primary refractory and relapsed/ refractory paediatric Acute Leukaemia. Here we report outcomes of all patients treated with this approach, including trial and off-trial patients.

Methods: Case records were analysed, and data collected about patients’ demographics, disease burden at the time of transplant, transplant and donor characteristics and the outcomes of transplant including engraftment, acute and chronic GVHD, relapse and transplant related mortality.

Results:

Demographics

No of Patients

Sex

Male

12

Female

10

Diagnosis

Secondary AML

3

Infant ALL with myeloid switch

2

Mixed phenotypic AML

4

Infant ALL with myeloid switch post CAR T

1

HR AML

12

Molecular and cytogenetics

KMT2A Rearrangement

6

Monosomy 7

3

NUP98/NSD1

2

CBFA2T3-GLIS2

2

MECOM rearrangement

2

PICALM-MLLT10

1

12p deletion

2

TP53 Mutation

1

FLT3 ITD mutation

1

t(16,21)

1

t(8,21)

1

NRAS mutation

1

Previous HSCT

Yes

15

No

7

Pre-transplant disease burden

M2, M3 marrow

6

MRD positive

10

MRD negative

3

Aplastic marrow

3

Conditioning

Busulphan Based

5

Treosulphan Based

17

This is a very high-risk cohort of 22 patients with a median age of 7.3 years (range: 1.2 to 17 years), having either or all, early post-transplant relapse, refractory disease, or high risk molecular or cytogenetics. These characteristics are summarised in Table 1. All patients were referred via a national MDT, and many were on palliative care pathways. All patients were given at least 7 daily doses of granulocytes starting from D-1 and they were well tolerated. Immune suppression was withdrawn early.

Patient outcomes are summarised as follows: 4 Patients experienced primary graft failure (GF), and 3 patients died, 2 with progressive leukaemia. One patient engrafted from a second CBT, achieved molecular remission, but experienced molecular relapse at 7 months, and remains alive receiving salvage chemotherapy. All other patients engrafted, and had early, significant, donor-derived, transient, predominantly CD8 T-cell expansion. Only one patient experienced TRM (4.5%), and died after engraftment, in remission with disseminated acyclovir resistant HSV. Of the remaining 17 patients, 16 entered MRD-negative remission. In 10, this remission is sustained at a median follow up of 31.6 (range: 3 – 84 months). The OS and EFS are 54.5% and 45.4% respectively. The commonest cause of mortality was relapse and 5 of 9 (55%) were late relapses. Only 13% (n=3) experienced severe acute GVHD of the skin and gut needing treatment. No patient experienced cGVHD.

Conclusions: This protocol, based on mismatched CBT, in a cohort of highest risk acute leukaemia has encouraging early results and continues to be taken forward in our programme. The rates of acute GVHD are tolerable, there is no cGVHD and the TRM is remarkably low, despite the mismatched graft. There is a higher graft failure rate than we usually see in CBT in malignant disease. In those that do not experience GF, the remission rates are high but some patients relapse. In our next protocol, we plan to use an expanded CB to reduce GF, and cord derived DLI after cessation of immune suppression to reduce late relapse. We plan to enrol adult patients besides children in our 2024 protocol.

Clinical Trial Registry: NCT05425043.

https://clinicaltrials.gov/study/NCT05425043.

Disclosure: The authors declare no conflict of interest..

6: Experimental Transplantation and Gene Therapy

P214 GENOME EDITING OF LONG-TERM REPOPULATING HEMATOPOIETIC STEM CELLS USING CCR5-UCO-HETTALEN

Valeriia Laushkina1, Alena Shakirova1, Olga Epifanovskaya1, Vladislav Sergeev1, Kirill Lepik1, Marina Popova1, Alexander Kulagin 1

1First Pavlov State Medical University of St. Petersburg, Saint Peterburg, Russian Federation

Background: Gene therapy effect duration after hematopoietic stem cells (HSCs) transplantation with modified genome is still not finally determined. It is considered that the durable effect can be achieved by high aptitude of long-term HSCs (LT-HSCs) to the gene editing (GE) procedure by programmable nucleases, because of the ability of lifelong hematopoiesis restoring from this CD34+ cells subpopulation. The insufficient GE efficacy in LT-HSCs arises as the main risk in this field.

The primary objective of the study was to test a standard CD34+ HSC’s GE protocol by CCR5-Uco-hetTALEN targeted to CCR5 in regards to the efficiency in LT-HSCs and conditions optimization for LT-HSC’s gene modification boost for HIV gene therapy.

Methods: PBSC products were obtained from healthy donors in accordance with the Declaration of Helsinki. CD34+ cell isolation was performed by MACS using CD34 MicroBead Kit (Miltenyi Biotec, Germany). The estimation of cell selection purity and LT-HSC’s percentage were performed by FACS using CD34 + /CD90 + /CD45RA- antibody panel. Selected cells were activated for 48h as described [Shakirova et al., 2020] or under prolonged up to 72h protocol. The presence or absence of 1mM SR1 (Stem Cell Technologies) during GE was also tested. Afterwards, cells were transfected with 25 μg/ml CCR5-Uco-hetTALEN mRNA using Gene Pulser Xcell System (Bio-Rad, USA). After the 5-day-long post-transfection incubation the CD90 + LT-HSCs were sorted. The CCR5 knockout efficiency was evaluated using digital droplet PCR (ddPCR) using DNA extracted from sorted CD34 + /CD90 + /CD45RA- LT-HSCs and bulk CD34+ HSCs according to the published protocol [Mock et al., 2015; Schwarze et al., 2021].

Results: The mean CD34+ selection efficiency and CD90+ cell sorting purity were 87,6±7,7% and 98,3±1,3%. respectively (n=3). The addition of SR1, the aryl-hydrocarbyl receptor antagonist, which contributes to the preservation of stem markers during ex vivo culturing of HSCs and maintains their multilinear potential, led to CD90+ events enrichment during the sorting procedure. The CCR5 knockout efficiency was numerically higher for bulk CD34+ compared to LT-HSCs (58,75±11,74% vs. 49,93±11,51%, p=0,22) in standard activation conditions (SR1-, 48h). The addition of SR1 during 48h activation led to a slight increase (p=0,29) of CCR5 knockout in bulk CD34+ (66,54±10,21%) and LT-HSC (56,27±10,49%). An increase of CD34+ activation time before transfection up to 72 h shifted the efficiency of CCR5 gene editing in LT-HSCs by 1.08 times in SR1+ conditions and 1.18 times without it.

Conclusions: The efficacy of the CCR5-Uco-hetTALEN was tested under various conditions to optimize GE in LT-HSCs. We have observed the minor increase of CCR5 knockout rate in both bulk CD34+ HSCs and sorted LT-HSCs with the addition of SR1 and the extension of activation time before transfection, however conclusions should be drawn cautiously due to the limited observations number. A balance between HSC activation to maximize GE and stemness preservation for engraftment potential should be assessed in vivo which is the perspective of the study.

Disclosure: Nothing to declare.

6: Experimental Transplantation and Gene Therapy

P215 ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION IMPROVES PROGNOSIS IN AML PATIENTS WITH CSF3R MUTATION AND CEBPA DOUBLE MUTATION

Jiaxin Cao1, Yawei Zheng 1, Aiming Pang1, Erlie Jiang1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China

Background: The incidence of CSF3R mutation in acute myeloid leukemia (AML) patients ranges from 1% to 3.6%. CSF3R mutation and CEBPA double mutations suggest a poor prognosis.

Methods: The study included a total of 28 AML patients with CSF3R mutation who had completed allogeneic haematopoietic stem cell transplantation at the Transplantation Centre of Blood Diseases Hospital, the Chinese Academy of Medical Sciences from September, 2016 to April, 2023 and analyzed the clinical data of these patients retrospectively.

Results: The 28 patients had a median age of 35.5 (5,63) years, 10 (35.7%) were male and 18 (64.3%) were female. 17 (60.7%) received haploidentical transplantation and 11 (39.3%) received allogeneic transplantation. Of the 28 patients with CSF3R mutation who received HSCT, the 3-year cumulative relapse rate was 14.6% (median follow-up time 574.5 (86,1933) days). The 3-year cumulative relapse rate after allogeneic HSCT was 10.00% in AML patients with CSF3R mutation and CEBPA dm and 21.89% in AML patients with CSF3R mutation only. There was no statistical significance between the two groups, P=0.68.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (31)

Conclusions: Overall, AML patients with CSF3R mutation who received HSCT had a lower relapse rate than previously reported. The rusults suggests that receiving allogeneic HSCT may ameliorate the high risk of relapse in AML patients with CSF3R mutation and CEBPA dm.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P216 CAST REGIMEN FOR GVHD PROPHYLAXIS YIELDS LOWER RELAPSE RATE AND IMPROVED DISEASE-FREE SURVIVAL: A CIBMTR PROPENSITY SCORE MATCHED CASE-CONTROL ANALYSIS

Samer Al-Homsi1, Todd DeFor2, Kelli Cole1, Frank Cirrone1, Maher Abdul-Hay1, Stephanie Wo1, Andres Suarez-Londono1, Sharon Gardner1, Jingmei Hsu1, George Yaghmour3, Caitrin Bupp2, Stephen Spellman 2

1New York University Grossman School of Medicine, New York, United States, 2Center for International Blood and Marrow Transplant Research and National Marrow Donor Program, Minneapolis, United States, 3University of Southern California, Keck School of Medicine, Los Angeles, United States

Background: To improve graft-versus-host disease (GvHD) prevention following peripheral blood haploidentical hematopoietic stem cell transplantation, we previously reported the outcomes of patients receiving a novel combination of post-transplant cyclophosphamide (PTCy), abatacept and short course of tacrolimus (CAST). Herein, we hypothesized that CAST is more effective than the commonly used combination of PTCy, tacrolimus and mycophenolate mofetil (MMF).

Methods: Patients enrolled in the CAST study were compared to a historical control cohort of patients from the Center for International Blood and Marrow Transplant Research (CIBMTR) database using nearest neighbor propensity score-matching. A propensity score for GvHD prophylaxis was developed using multivariable logistic regression. The final variables used to generate the propensity score were recipient age, conditioning regimen, disease risk index and comorbidities. Four cases of the original CAST trial were excluded (2 due to lack of consent, 1 due to lack of match at 1 decimal place and 1 case of primary myelofibrosis due to a disproportionate number in the control cohort). After matching, the balance between the 2 groups was assessed striving for a less than 0.1 standardized difference but allowing a maximum difference of 0.2 between cohorts. The primary end point was cumulative incidence of grades II-IV acute GvHD by day 120 while secondary endpoints included cumulative incidence of grades III-IV acute GvHD and moderate to severe chronic GvHD, relapse, non-relapse mortality (NRM), disease-free survival (DFS), overall survival (OS), and GvHD- and relapse-free survival (GRFS).

Results: A very good balance between cases and controls was obtained with propensity score matching as measured from the density distribution and standardized differences in covariates (median=0.09). The comparative outcomes are summarized in the table and graphs. In terms of the primary endpoint, there was a raw trend toward lower incidence of grades II-IV acute GvHD in favor of the CAST cohort. The lack of statistical significance may be due to the small number of events. There was also a trend toward improved GRFS. On the other hand, relapse rate was statistically significantly lower and DFS was higher in favor of the CAST cohort. The improved relapse rate and DFS could be related to the early discontinuation of tacrolimus and the known favorable effects of abatacept on natural killer cells.

CAST

Probability (95% Confidence Level)

Control

Probability (95% Confidence Level)

p

Value

Acute GvHD; grade II-IV (day + 120)

16.7% (7-29.5%)

28.6% (15.9-43.2%)

0.14

Acute GvHD; grade III-IV (day + 120)

4.8% (0.1-14.4%)

9.5% (3-20.7%)

0.39

Non-GvHD mortality (day + 120)

4.8% (0.1-11.0%)

7.1% (0.2-17.6%)

0.62

Chronic GvHD; moderate to severe (1 year)

16.7% (7-29.5%)

14.3% (5.4-26.5%

0.77

Treatment-related mortality (1 year)

4.8% (0.4-13.3%)

11.9% (3.9-23.5%)

0.24

Relapse rate (1 year)

9.5% (2.6-20.3%)

26.2% (14-40.6%)

0.05

Disease-free survival (1 year)

85.7% (73.7-94.5%)

61.9% (46.9-75.8%)

0.01

Overall survival (1 year)

88.1% (76.7-96%)

76.2% (62.3-87.7%)

0.16

GRFS (1 year)

66.7% (51.9-80%)

47.6% (32.9-62.6%)

0.07

Conclusions: Despite the limitations of this analysis due to the size of cohorts and concessions made for matching purposes including collapsing or limiting variables, the CAST regimen provides improved outcomes in comparison to the control regimen in term of relapse rate and DFS. These results provide a strong impetus to examine the CAST regimen in a large phase III trial against the commonly used regimen of PTCY, tacrolimus and MMF.

Clinical Trial Registry: Not Applicable.

Disclosure: Al-Homsi: BMS, Advisory Board.

Yaghmour: Speaker Bureau: Jazz, Incyte, Astellas, BMS, Secura, blueprint, SOBI, Karius, Kite/Gilead. Advisory board- Gilead, Alexion.

12: Graft-versus-host Disease – Clinical

P217 SEQUENTIAL INTRAVENOUS UMBILICAL CORD DERIVED MESENCHYMAL STROMAL CELLS (MSCS) FOR SALVAGE TREATMENT OF STEROID-REFRACTORY ACUTE GVHD (RS-AGVHD): A PHASE IB/II STUDY

Yanmin Zhao 1,2,3,4, Yi Luo1,2,3, Jimin Shi1,2,3,4, Jian Yu1,2,3,4, Lizhen Liu1,2,3,4, Xiaoyu Lai1,2,3,4, Huarui Fu1,2,3,4, Yishan Ye1,2,3,4, Nainong Li5,6,7, Erlie Jiang8, Qiong Xie9, Jundong Gu9, Zhibo Han9, Zhongchao Han9, He Huang1,2,3,4

1Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Institute of Hematology, Zhejiang University, Hangzhou, China, 3Zhejiang Province Engineering Laboratory for Stem Cell and Immunity Therapy, Zhejiang University, Hangzhou, China, 4Zhejiang Laboratory for Systems and Precision Medicine, Zhejiang University Medical Center, Hangzhou, China, 5Hemopoietic Stem Cell Transplantation Center, Fujian Medical University Union Hospital, Fuzhou, China, 6Fujian Provincial Key Laboratory on Hematology, Fujian Institute of Hematology, Fuzhou 350001, China, Fuzhou, China, 7Fujian Medical University Hematology Translational Center, Fuzhou 350001, China, Fuzhou, China, 8Hematopoietic Stem Cell Transplantation Center, Institute of Hematology and Blood Diseases Hospital, Peking Union Medical College and Chinese Academy of Medical Sciences, Tianjin, China, 9National Engineering Research Center of Cell Products, Tianjin AmCellGene Engineering Co., Ltd., Tianjin, China

Background: To investigate the safety and initial efficacy of MSCs for salvage treatment of patients with SR-aGVHD who had received previous steroid-containing immunosuppressive therapy.

Methods: A phase I b/II, multicenter, open-label clinical trial of a third-party, off-the-shelf preparation of human umbilical cord derived mesenchymal stromal cells (hUC-MSCs) (ChiCTR2200061603) enrolled grades II to IV steroid-refractory aGVHD patients who had been treated with previous steroid-containing immunosuppressive therapy was performed between June 2022 to October 2023. This trial consist of 2 parts: a phase Ib dose escalation part using a standard 3 + 3 design, which planed biweekly i.v. infusions of hUC-MSCs, at 0.5× 106per kg, 1.0× 106per kg and 2.0× 106 per kg for 3 weeks, to determine the maximum tolerated dose (MTD). Phase II of this work is an expansion cohort study of hUC-MSCs at RP2 D 1.0×106 per kg with 15 patients.

Results: A total of 25 patients were enrolled with the median follow up period of 202 (10,330) days. Of the included 25 cases,11 patients (44.0%) were grade III/IV and 14 patients (56.0%) were grade II aGVHD at enrollment. hUC-MSCs was well tolerated without dose-limiting toxicities in aforementioned three dosages. The reported adverse events (AEs) potential associated with hUC-MSCs were hypofibrinogenemia (4.0%), upper respiratory tract infection (4.0%), leukopenia (4.0%), hypogammaglobulinemia (4%) and anemia (8.0%) within follow-up period [median 202 (10,330) days]. Grade 3 or higher AEs were reported in 15 patients (15/25, 60%) with 109 events, all of them were supposed to be uncorrelated with hUC-MSC. Five patients unfinished the trial: 2 was due to withdrawn ICF; 3 was due to SAE, of which 1 died of SAE (transplant-associated thrombotic microangiopathy (TA-TMA) and 1 die of pulmonary infection, 1 withdrawn the trial due to leukemia recurrence. At Day 28, 20 patients (80%, 95% CI:59.3%–93.2%) had an overall response, including 10 (40%) of complete responses. And three patients who was evaluated partial response at day 28 achieved complete responses at day 56 (Table 1). Responses were observed across skin (19/19, 100%), gastrointestinal tract (9/13,69.2%), and liver (2/2, 100.0%) at day 28. Durable overall response at day 56 was 80% (20/25). The probability of 180-day overall survival (OS) and non-relapse mortality was 88.0% (95% CI, 68.8-97.5%) and 8.0% (95% CI, 1.0-26.0%), respectively. The 180-day cumulative incidence of relapse of primary hematologic disease was 8.0% (95%CI,1.0-26.0%).

Table 1. Overall Response at Day 28 and Day 56 of different dosage

Response

0.5×106cells/kg

1×106 cells/kg

2×106cells /kg

Total

(N=5)

(N=17)

(N=3)

(N=25)

Day 28

N (N, Miss)

5 (0)

17 (0)

3 (0)

25 (0)

CR [n, (%)]

3 (60.0)

5 (29.4)

2 (66.7)

10 (40.0)

PR [n, (%)]

1 (20.0)

8 (47.1)

1 (33.3)

10 (40.0)

PD [n, (%)]

0 (0.0)

0 (0.0)

0 (0.0)

0 (0.0)

NR [n, (%)]

1 (20.0)

4 (23.5)

0 (0.0)

5 (20.0)

ORR(95% CI)

80.0 (28.4, 99.5)

76.5 (50.1, 93.2)

100.0 (29.2, 100.0)

80.0 (59.3, 93.2)

Day 56±2

N (N, Miss)

5 (0)

17 (0)

3 (0)

25 (0)

CR [n, (%)]

4 (80.0)

7 (41.2)

2 (66.7)

13 (52.0)

PR [n, (%)]

0 (0.0)

7 (41.2)

0 (0.0)

7 (28.0)

PD [n, (%)]

0 (0.0)

0 (0.0)

0 (0.0)

0 (0.0)

NR [n, (%)]

1 (20.0)

3 (17.6)

1 (33.3)

5 (20.0)

ORR(95% CI)

80.0 (28.4, 99.5)

82.4 (56.6, 96.2)

66.7 (9.4, 99.2)

80.0 (59.3, 93.2)

Conclusions: Collectively, our results support hUC-MSCs as an effective salvage treatment for SR-aGVHD with safe profile.

Clinical Trial Registry: ChiCTR2200061603.

https://www.chictr.org.cn/showproj.html?proj=171558.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P218 NOVEL GVHD PROPHYLAXIS WITH POST-TRANSPLANT INTERMEDIATE DOSE CYTOXAN AND ABATACEPT – LOW RATES OF ACUTE GVHD AND TRANSPLANT-RELATED TOXICITY

Asya Varshavsky Yanovsky1, Yuliya Shestovska 1, Shalina Joshi1, Dzhirgala Mandzhieva1, Peter Abdelmessieh1, Rashmi Khanal1, Michael Styler1, Henry Fung1

1Fox Chase Cancer Center, Philadelphia, United States

Background: Abatacept was FDA approved for GVHD prophylaxis based on ABA2 study, that showed reduction in acute GVHD (aGVHD) rates with addition of Abatacept to standard Methotrexate/CNI based GVHD prophylaxis. Smaller retrospective studies showed feasibility of addition of Abatacept to post-transplant Cytoxan (PTCy) GVHD prophylaxis. Decreasing the dose of PTCy has being evaluated with the goal to decrease chemo related toxicities. Based on this rationale, we introduced a novel GVHD prophylaxis regimen combining intermediate dose PTCy 25mg/kg on days 3, 4 with Abatacept 10mg/kg on days 5, 14, 28 (PTCy25/Aba) for 10/10 matched unrelated (MUD) and matched related donors (MRD) in 1/2022.

Here we report our findings on efficacy and safety of the PTCy25/Aba regimen.

Methods: We retrospectively reviewed patients who received 10/10 MUD or MRD allo SCT at our program since 1/2022 with novel PTCy25/Aba GVHD prophylaxis and compared to a matched group of patients transplanted in 2020-21 with institutional standard PTCy 50mg/kg day 3, 4 (PTCy50) prophylaxis. All patients received tacrolimus and 10/10MUD patients also received MMF days 5-30.

Results: We identified 30 patients who received PTCy25/Aba and compared to a matched group of 28 patients who received PTCy50. Baseline patients’ characteristics are described in Table 1.

Table 1

PTCy25/Aba

PTCy50

N

30

28

Age (mean (range))

59(42-72)

56(20-73)

Sex M:F (%)

57:43

57:43

MA: RIC (%)

70:30

71:29

MUD:MRD (%)

70:30

64:36

Dx: AML: MDS: ALL (%)

37:50:13

79:19: 2

KPS (%)

90-100

40%

57%

70-80

57%

36%

60 and below

3%

7%

HCT CI (%)

0-2

40%

36%

3-4

30%

43%

5+

30%

21%

Day of Discharge (mean (range))

24.1 (14-35)

28.5(19-70)

P 0.02

ANC engraftment

14 (11-19)

17.7(14-30)

P 0.00

PLT Engraftment

Average (range)

20.1(12-41)

29.5(16-48)

P 0.00

CD3 Chimerism DD30-60

56: 44

88:12

P 0.01

70 + : <70 (%)

CD3 Chimerism 100-180

88:12

95:5

P 0.61

70 + : <70 (%)

100d aGVHD (%), Gr 0-1: Gr. 2+

69:31

52:48

P 0.27

6m aGVHD (%), Gr 0-1: Gr. 2+

68:32

50:50

P 0.26

CMV reactivation 100d

0%

14%

P 0.04

Hemorrhagic cystitis 100d

7%

18%

P 0.24

100d Survival

97%

89%

P 0.344

1y Survival

62%

(9pts excluded - less than 1y f/u)

67%

(1pt excluded-<1y f/u)

P 0.76

100d progression free

86%

88%

1y progression free

(pts who died in CR before 100d/1y assessment excluded from this analysis)

60%

61%

P 1

Safety and efficacy outcomes are presented in Table 1.

Patients treated with PTCy25/Aba had earlier hematological recovery post-transplant compared to PTCy50 (mean ANC engraftment: 14 vs 18 days, p 0.03; mean PLT engraftment: 19 vs 29 days, p 0.00) and resulting shorter duration of post-transplant hospitalization (24 vs 29 days, p 0.03). There were no cases of CMV reactivation in the cohort treated with PTCy25/Aba vs 14% with PTCy50. Incidence of hemorrhagic cystitis was numerically (though not statistically significantly) lower in the PTCy25/Aba group (7% vs 18%). No cases of SOS were observed in both groups. Acute GVHD incidence was similarly low in both groups, with only one case of grade 3-4 GVHD in each group (3%) and 68% vs 50% patients had none or grade 1 GVHD by 6 months. Early (day 30-60) CD3 donor chimerism was lower in the PTCy25/Aba group compared to PTCy50 group, but improved by day 100-180 assessment and the difference between the groups resolved.

There was no difference between OS and PFS at 100 days and 1 year between the groups (Table 1, of note 9 patients in the PTCy25/Aba group have not reached 1 year follow up by the time of this analysis).

Conclusions: Novel GVHD prophylaxis combining intermediate dose of PTCy and Abatacept resulted in similar low rates of acute GVHD compared to standard dose PTCy in 10/10 MUD, with earlier hematological recovery, decreased CMV reactivation and trend towards less hemorrhagic cystitis. Longer follow up is needed to assess effects on chronic GVHD, long terms survival and relapse.

Disclosure: Asya Varshavsky Yanovsky: Pfizer, Janssen, BMS- advisory.

Henry Fung: Astra Zeneca, Incyte, Kite, Janssen - advisory, honoraria.

Peter Abdelmessieh: AbbVie, Sobi - advisory.

12: Graft-versus-host Disease – Clinical

P219 REDUCING PTCY DOSE IN PATIENTS RECEIVING CAST FOR GVHD PREVENTION RESULTS IN ACCELERATED ENGRAFTMENT AT THE COST OF HIGHER INCIDENCE OF GVHD

J. Andres Suarez-Londono 1, Kelli Cole1, Frank Cirrone1, Stephanie Wo1, Maher Abdul-Hay1, Jingmei Hsu1, Sharon Gardner1, Mohammad Abu-Zaid1, Gloria Contreras Yametti1, Benjamin Levinson1, Judith Goldberg1, A. Samer Al-Homsi1

1New York University Grossman School of Medicine, New York, United States

Background: The combination of post-transplant cyclophosphamide (PTCy), abatacept (A) and a short course of tacrolimus (T) (CAST) is a promising novel regimen for prevention of graft-versus-host (GvHD) following peripheral blood (PB) haploidentical transplant. Mechanistic and early clinical data suggest that the dose of PTCy can be decreased to diminish its toxicity while maintaining its efficacy. We aimed to explore the feasibility and efficacy of reduced-dose PTCy in patients (pts) receiving CAST.

Methods: We initiated a phase II Simon 2-stage clinical trial for adult pts with hematological malignancies undergoing haploidentical PB transplants. Pts received PTCy 25 mg/kg days (d) +3 and +4, A 10 mg/kg d + 5, +14, +28 and +56 and T starting on d + 5 and tapered from d + 60 until d + 90 (re-CAST); the primary endpoint being the incidence of grades II-IV acute GvHD (aGvHD) by d + 120. Conditioning regimens with different intensities were permitted. Herein, we compare the early results of this ongoing trial to the results of the original CAST trial. Wilcoxon rank-sum tests were used to compare the median d of neutrophil (ANC) and platelet (plt) engraftment. Gray’s test was used to compare the cumulative incidence functions of acute and chronic GvHD (cGvHD). The studies were performed sequentially at the same institution and had the same inclusion and exclusion criteria.

Results: The characteristics of the 19 patients enrolled in the re-CAST study as of December 13, 2023 are summarized in the table. Median follow up was 187 d (range 43 – 407). Median time to ANC and plt engraftment were 13 and 18 d, significantly lower than in the CAST study (18 and 30 d, p<0.0001 for both). There was no graft failure. In the re-CAST study, 14 pts had a follow up >120 d or already developed aGvHD. Four pts developed grade II and 1 grade III aGvHD. Cumulative incidence of grade II-IV aGvHD was 29.4% (95% CI, 13.5%-64%) as opposed to 17.4% (95% CI, 9.2%-50.7%) in the CAST study (p=0.20). There was one case of grade III aGvHD (7.1%, 95% CI, 1%-36%). Median time of onset of aGvHD was 62 d. Eleven pts had a follow-up of >180 d or already diagnosed with moderate to severe (MS) cGvHD. Six pts developed MS cGvHD, mostly with mucocutaneous manifestations. Two pts died without developing cGvHD. Cumulative incidence of MS cGvHD was 67.4% (95% CI, 23.3%-100%) versus 15.9% (95% CI, 8%-31.7%) in the CAST study (p=0.003). There were no cases of steroid-refractory or fatal GvHD. The incidence of CMV and EBV reactivation were 42% and 10%. One pt died of PTLD and 1 of disease relapse. Overall, 2 pts relapsed.

Conclusions: Our results suggest reducing the total dose of PTCy to 50mg/Kg following PB haploidentical transplant results in accelerated engraftment at the expense of increased incidence of cGvHD and should be restricted to well-designed clinical trials. The re-CAST study was amended to increase the total dose of PTCy to 75 mg/Kg.

Clinical Trial Registry: ClinicalTrials.gov ID: NCT05621759.

Disclosure: M. Abdul-Hay: Advisory boards: Kite, Daiichi, Rigel and Incyte; Speaker Bureau: Jazz, Takeda and Servier A. Samer Al-Homsi: educational grant from BMS; Scientific Advisory Board member for Incyte.

12: Graft-versus-host Disease – Clinical

P220 RUXOLITINIB IN ACUTE AND CHRONIC GRAFT-VERSUS-RECEPTOR DISEASE: LONG-TERM REAL-LIFE EXPERIENCE IN A MULTI-CENTRE STUDY

Virginia Escamilla Gómez 1, Lucía López Corral2, Beatriz Astibia Mahillo3, Patricia Alcalde Mellado1, Francisco Manuel Martín Domínguez1, Marina Acera Gómez2, Pedro Asensi Cantó4, Juan Montoro Gómez4, Melissa Torres Ochando5, Asunción Borrego Borrego5, Leslie González Pinedo5, María Teresa Zudaire Ripa6, Marta González Vicent7, Ana Benzaquén Vallejos8, Isabel Izquierdo9, Irene García Cadenas10, Sara Redondo Velao10, Alberto Musseti11, Eloi Cañamero Giro12, Pilar Palomo Moraleda13, Guillermo Orti Pascual14, Pedro Antonio González Sierra15, Lucía García16, Valentín García Gutiérrez3, José Antonio Pérez-Simón1

1Hospital Universitario Virgen del Rocío, Sevilla, Spain, 2Hospital Clínico Universitario de Salamanca (CAUSA/IBSAL), Salamanca, Spain, 3Hospital Universitario Ramón y Cajal, Madrid, Spain, 4Hospital Universitario y Politécnico La Fe, Valencia, Spain, 5Hospital Universitario de Gran Canaria Doctor Negrín, Las Palmas, Spain, 6Hospital Universitario de Navarra, Navarra, Spain, 7Hospital Niño Jesús, Madrid, Spain, 8Hospital Clínico Universitario de Valencia, Valencia, Spain, 9Hospital Universitario Miguel Servet, Zaragoza, Spain, 10Hospital de la Santa Creu I Sant Pau, Barcelona, Spain, 11Hospital Duran i Reynals, Institut d\’Investigació Biomèdica de Bellvitge (IDIBELL), Barcelona, Spain, 12Hospital German Trias I Pujol, Barcelona, Spain, 13Hospital Universitario Central de Asturias, Asturias, Spain, 14Hospital Universitario Vall d´Hebron, Barcelona, Spain, 15Complejo Hospitalario Universitario de Granada, Granada, Spain, 16Hospital Son Espases, Mallorca, Spain

Background: The JAK1/2 inhibitor ruxolitinib has been approved for the treatment of adults with acute or chronic graft-versus-receptor disease (GvHD) based on the REACH-2 and 3 studies. However, real-life studies are needed to validate the results of clinical trials and clarify other aspects such as efficacy in specific organs, efficacy beyond first-line treatment or the possibility to reduce immunosuppression.

Methods: Analysis of 352 adult patients with acute or chronic GvHD refractory to corticosteroids through a retrospective, descriptive and multicenter study from 17 Spanish centers belonging to the Spanish Group for Transplantation and Cellular Therapy (GETH-TC). Data were collected between October/2015 and September/2022.

Results: The baseline characteristics and decrease in immunosuppression are shown in table 1.

Acute GvHD group. A total of 119 patients were included. The overall response (OR) was 59.7% (33.6% complete remissions, CR); median to best response was 4 weeks (range: 1-77). By organs, response in skin was 77.2% (CR: 67%), liver 63.1% (CR: 42.1%) and gastrointestinal tract 50% (CR: 25%). No significant differences were observed in terms of efficacy among patients treated in 2nd versus 3rd or successive lines. 73 patients died due to the following causes: GvHD progression 22/119 (18.5%), relapse 4/119 (3.4%), infection 37/119 (31.1%), others 10/119 (8.4%). 9 patients (7.6%) relapsed of their underlying disease.

Among responding patients, 52.9 % and 47.2% were able to withdraw corticosteroids and ruxolitinib, respectively. The 6-month overall survival (OS) between responders vs non-responders was 69.1% (95%CI:55.2-79.5) vs 19.6% (95%CI:9-32), p=0.00.

Chronic GvHD group. 233 patients were included in the analysis. The OR and CR were 65.7 % and 18.5%, respectively. The median time to observe the best response was 6 months (range: 0.25-48). By organs, responses were: gastrointestinal tract 75.9% (CR: 24%), scleroderma 69.8% (CR: 22.4%) and lung 37.8% (CR: 1.5%). No significant differences were observed in terms of efficacy between patients treated in 2nd versus 3rd or successive lines. 56 patients died due to the following causes: GVHD progression 15/233 (6.4%), relapse 4/233 (1.7%), infection 23/233 (9.9%), others 14/233 (6%). Relapse of the hematological disease was observed in 10 patients (4.3%).

Among responding patients, 40.1% and 34.8% were able to withdraw corticosteroids and ruxolitinib, respectively. The 2-year OS between responders vs non-responders was 86.8% (95% CI: 79.6-91.6) vs 67.8% (95% CI: 55.5-77.4), p=0.00.

Table 1. Patients characteristics

N (%)

Adults

(n= 352)

Gender

Male

203 (57.7)

Female

149 (52.3)

Age

Median (range)

52 (14-75)

Underlying disease

AML

119 (33.8)

ALL

56 (15.9)

MDS

71 (20.2)

MM

8 (2.3)

HL

12 (3.4)

NHL

54 (15.4)

Myelofibrosis

11 (3.1)

AA

1 (0.3)

Primary inmunodeficiencies

1 (0.3)

Other

19 (5.4)

Disease status previous HSCT

CR

236 (67)

PR

37 (10.5)

SD

51 (14.5)

Others

28 (7.9)

Type of trasplant

Related HLA identical donor

144 (40.9)

Haploidentical

56 (15.9)

Unrelated donor

152 (43.2)

Conditioning regimen

Myeloablative

175 (49.7)

Reduced-intensity

175 (49.7)

Source of progenitor cells

Peripheral blood

336 (95.4)

Bone marrow

15 (4.3)

Umbilical cord

1 (0.3)

Conclusions:

  • Ruxolitinib in the real world setting shows very similar results in terms of response to prospective clinical trials.

  • By organs, favorable results were observed in the skin, gastrointestinal tract, liver and lung. Of these, those that responded best in acute GvHD were the skin and in chronic GvHD were scleroderma and gastrointestinal tract.

  • These results seem not to be influenced by previous lines of treatment (<2 vs ≥2; <3 vs ≥3).

  • In the groups of responding patients, a reduction in the number of patients on immunosuppressants at the end of treatment was observed in both study groups.

.

Disclosure: *This project has been carried out through the GETH-TC (Spanish Group for Transplantation and Cellular Therapy - Grupo Español de Trasplante Hematopoyético y Terapia Celular) and funded by Novartis.

12: Graft-versus-host Disease – Clinical

P221 PHASE 1 TRIAL INVESTIGATING THE SAFETY AND TOLERABILITY OF LEFLUNOMIDE IN PATIENTS WITH STEROID-DEPENDENT CHRONIC GRAFT-VERSUS-HOST DISEASE (CGVHD) AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT (ALLOHCT)

Amandeep Salhotra 1, Dongyun Yang1, Sally Mokhtari1, Badri Modi1, Monzr Al-Malki1, Haris Ali1, Idoroenyi Amannam1, Nicole Karras1, Karamjeet Sandhu1, Ahmed Aribi1, Salman Otoukesh1, Andrew Artz1, Guido Marcucci1, Stephen Forman1, Eileen Smith1, De-Fu Zhang1, Steven Rosen1, Ryotaro Nakamura1

1City of Hope Comprehensive Cancer Center, Duarte, United States

Background: Current FDA approved agents for cGVHD are associated with low complete response rates and high cost. Leflunomide is an FDA-approved drug for treatment of rheumatoid arthritis (RA) and is in clinical trial development to treat multiple myeloma, solid tumors, acute GVHD, and COVID19. Based on its role in promoting lymphocyte apoptosis and its anti-inflammatory properties, we conducted a phase 1 trial (NCT04212416) to study leflunomide’s safety in patients with cGVHD (primary objective). The 6-month anti-cGVHD activity, defined by overall response rate (ORR per NIH consensus criteria), was the secondary objective.

Methods: Patients received a loading dose of leflunomide (100 mg X 3 days) followed by a daily maintenance dose of 20 mg, which is the approved dose in RA (Fig 1). The planned duration of therapy was six 28-day cycles, with responding patients being allowed to continue therapy. The CTCAE v5.0 was used to assess toxicity. Changes in cGVHD severity were assessed by physician reported cGVHD activity assessment form.

Results: The trial consisted of two phases; safety lead-in: n=6 followed by expansion phase : n=12 with a planned dose de-escalation to 10 mg dose, if >1 patient experienced dose limiting toxicities (DLT) in the safety lead (first 28 days). The median age was 53 years (range 18-72). Most patients received PBSC graft (89%). Donors were matched unrelated (55%) or related (45%). Conditioning was MAC (50%) or RIC (50%). The most common GVHD prophylaxis was tacrolimus-sirolimus (56%) and prior aGVHD was reported in 60% patients. Median prior lines of therapy were 3 (range 2-7). Moderate and severe cGVHD were seen in 9 and 9 patients, respectively. Most common organ involvements were oral (77%), skin (61%), ocular (66%), joint/fascia (44%), lung/BOS (33%) and GI (11%).

Adverse events (AE) were GI (diarrhea: 27%, nausea/vomiting: 5%, abdominal pain: 5%), cytopenias (anemia: 16%, leukopenia: 11%), hepatic failure (5%), lab abnormalities (AST/ALT increase: 11%, bilirubin increase: 5%), fatigue (11%), and headache (5%). The AEs (n=29) were mostly grade 1-2 (82%), and grade 3-4 AEs were seen in 18% including one DLT (sepsis). The best ORR at any time in the first 6 cycles was 55% [n=10; partial responses (PR)], 38% (n=6; stable disease) and 11% (n=2; progressed). The mean duration of therapy was 4.7 months, 12 (66%) patients completed 6 cycles of planned therapy. Overall response rates was 55% (n=10 had partial responses; PR), n=6 (33%) had stable cGVHD and n=2(11%) had progressive symptoms. Reasons for early discontinuation of leflunomide were plateauing of response (n=2), AEs (n=2), progressive cGVHD (n=1), and death (n=1; sepsis). There was a mean prednisone dose reduction of 41% from study start to end (9.4 vs 5.5 mg/day).

Conclusions: In conclusion, leflunomide is safe in patients with cGVHD, and is associated with a promising ORR of 55% in patients with multisystem cGVHD involvement. Correlative immunologic and molecular studies are underway to compare the impact of leflunomide on lymphocytes between responders and non-responders.

Clinical Trial Registry: NCT04212416.

Disclosure: Salhotra: Gilead: Research Funding; Jazz Pharma: Research Funding; Sobi: advisory board; Sanofi: Speakers Bureau; OrcaBio: Research Funding; Kura Oncology: Research Funding; BMS: Research Funding.

Al Malki: Tscan: Consultancy.

Aribi: Seagen: Consultancy; Kite, a Gilead Company: Consultancy.

Aldoss: Takeda: Consultancy; KiTE: Consultancy; Sobi: Consultancy; Amgen: Consultancy, Honoraria; Pfizer: Consultancy; Jazz: Consultancy.

Pullarkat: Pfizer: Consultancy, Speakers Bureau; Servier: Consultancy, Speakers Bureau; Novartis: Consultancy, Speakers Bureau; Amgen: Consultancy, Speakers Bureau; Jazz Pharmaceuticals: Consultancy, Speakers Bureau; Genentech: Consultancy, Speakers Bureau; AbbVie: Consultancy, Speakers Bureau.

Sandhu: Autolus Therapeutics: Consultancy;.

Artz: Astra Zeneca: Advisory Board; Magenta Therapeutics:: Advisory Board; Radiology Partner: Current equity holder in private company, Spouse equity interest; Abbvie: Consultancy.

Ali: Karyopharm: Consultancy; GSK: Consultancy; Pharmaessentia: Consultancy; Blueprints: Speakers Bureau; BMS: Speakers Bureau; Incyte: Research Funding.

Marcucci: Ostentus Therapeutics: Current equity holder in private company, Research Funding.

Nakamura: Napajen: Consultancy; Sanofi: Consultancy; Blue Bird: Consultancy; International Consortium: Other: consortium chair; Jazz Pharmaceuticals: Consultancy, Other: research collaboration; Miyarisan: Research Funding; NCCN: Other: guideline panel for HCT; Leukemia & Lymphoma Society: Mt. Sinai: Other: Acute GVHD; Omeros: Consultancy; BMT CTN Steering Committee: Membership on an entity’s Board of Directors or advisory committees; NCTN Lymphoma Steering Committee: Membership on an entity’s Board of Directors or advisory committees.

Stein: Sanofi: Current holder of stock options in a privately-held company.

Rosen: Exicure: Current holder of stock options in a privately-held company; Pepromene Bio, Inc: Current holder of stock options in a privately-held company; Pepromene Bio, Inc: Board of Directors or advisory committees; NeoGenomics: Board of Directors or advisory committees; Verastem, Inc: Consultancy; Trillium Therapeutics, Inc: Consultancy; PharmaGene, LLC: Consultancy; Apobiologix/Apotex Inc: Consultancy; Exicure: Consultancy; Pheromone Bio, Inc: Consultancy; January Biotech: Current holder of stock options in a privately-held company; Trillium Therapeutics: Current holder of stock options in a privately-held company; SLAM BIOTHERAPEUTICS, INC:.

12: Graft-versus-host Disease – Clinical

P222 COMPARISON OF EFFICACY AND TOLERABILITY OF RUXOLITINIB ALONE OR RUXOLITINIB WITH EXTRACORPOREAL PHOTOPHERESIS OR EXTRACORPOREAL PHOTOPHERESIS MONOTHERAPY FOR STEROID REFRACTORY AGVHD: A GITMO RETROSPECTIVE STUDY

Marta Lisa Battista 1, Francesca Patriarca1,2, Miriam Isola2, Gabriele Facchin1, Patrizia Chiusolo3,4, Domenico Russo5, Attilio Olivieri6, Bruna Puglisi7, Ilaria Scortechini7, Alessandro Rambaldi8, Irene Maria Cavattoni9, Albana Lico10, Riccardo Saccardi11, Angela Cuoghi12, Marco Ladetto13, Vincenzo Federico14, Maria Caterina Micò15, Antonella Geromin1, Anna Maria Gallina16, Elena Oldani8, Eliana Degrandi17, Renato Fanin1,2, Fabio Ciceri18, Massimo Martino15

1Hematology and SCT Unit - Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy, 2DAME University of Udine, Udine, Italy, 3Università Cattolica del Sacro Cuore, Roma, Italy, 4Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy, 5University of Brescia and Spedali Civili, Brescia, Italy, 6Hematology and SCT Unit - Università Politecnica delle Marche/A.O.U delle Marche, Ancona, Italy, 7Hematology and SCT Unit - A.O.U delle Marche, Ancona, Italy, 8Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy, 9Ematology and BMTU - Hospital of Bolzano (SABES -ASDAA), Bolzano, Italy, 10Hematology Division - San Bortolo Hospital, Vicenza, Italy, 11Azienda Ospedaliero Universitaria Careggi, Firenze, Italy, 12Azienda Ospedaliero Universitaria, Modena, Italy, 13Azienda Ospedaliera SS Antonio e Biagio e C. Arrigo, Alessandria, Italy, 14Vito Fazzi Hospital, Lecce, Italy, 15Grande Ospedale Metropolitano Bianchi-Melacrino-Morelli, Reggio Calabria, Italy, 16IBMDR - Registro Nazionale Donatori di Midollo Osseo - E.O. Ospedali Galliera, Genova, Italy, 17Trial Office GITMO Gruppo Italiano per il Trapianto di Midollo Osseo, Cellule Staminali Emopoietiche e Terapia Cellulare, Bologna, Italy, 18San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy

Background: The aim of this Italian Group for Bone Marrow Transplantation, Haematopoietic Stem Cells and Cell Therapy GITMO retrospective multicenter study was to compare 3 different salvage treatments for steroid refractory acute GvHD (SRaGvHD) - ruxolitinib alone, ruxolitinib in association with extracorporeal photopheresis (ECP), ECP monotherapy - in terms of response rate at day 28, outcome and adverse effects.

Methods: The study included 233 adult patients who underwent allo-SCT from 2015 to 2021 at 18 GITMO centers treated with ruxolitinib and/or ECP for SRaGvHD.

Clinical patient and transplant features are summarised in table 1.

Median time from transplant to aGvHD and to SRaGvHD onset were 23 and 56 days, respectively.

The second line therapies were: ECP monotherapy for group 1 (124 patients, 53%), ruxolitinib plus ECP for group 2 (53 patients, 23%) and ruxolitinib monotherapy for group 3 (56 patients, 24%).

Patients n=233

Median age at transplant, years (range)

54 (18-75)

Sex, n (%)

• Male

150 (64)

• Female

83 (36)

HCT-CI >2, n (%)

85 (37)

Diagnosis, n (%)

• AML

95 (41)

• MDS

49 (21)

• ALL

33 (14)

• Lymphomas

33 (14)

• other

23 (10)

Active disease at transplant, n (%)

62 (27)

Conditioning regimen, n (%)

• MAC

144 (62)

• RIC

89 (38)

Immunosuppressive prophylaxis, n (%)

• ATG/ALG regimen

110 (47)

• PTCy regimen

55 (24)

• non ATLG-PTCy regimens

68 (29)

Donor type and HLA match, n (%)

• Matched sibling

48 (21)

• Haploidentical related

55 (24)

• Matched unrelated

70 (30)

• Mismatched unrelated

60 (26)

SC source, n (%)

• PB

195 (84)

• BM

38 (16)

Median CD34/kg infused cells, x10^6/kg patient (range)

5.37 (0.7-15.2)

Median CD3/kg infused cells, x10^7/kg patient (range)

17.4 (0.3-67.5)

Results: Characteristics of patients and transplants were equally distributed among the 3 groups without significant differences, except for an increased median number of CD3+ cells infused at transplant in group 3 (p=.016). Among GVHD features, grade III-IV SRaGvHD was significantly less frequent in group 1 (28%) in comparison with group 2 (74%) (p<.00001) and group 3 (61%) (p=.00003). Moreover, ruxolitinib with ECP or alone was significantly more frequently administered to patients with multiorgan involvement [64% and 41% of patients of groups 2 and 3, respectively) in comparison with ECP monotherapy (36% of patients of group 1) (p=.00063 and p=.015, respectively). ECP monotherapy was more frequently used in patients with skin single-organ involvement [52% of patients of group 1 versus 21% of group 2 and 23% of group 3 (p=.00009 and p=.00025, respectively].

No significant differences were found in overall and complete responses rates at day 28 among the 3 groups (79% and 58%, in group 1; 66% and 45%, in group 2; 80% and 64%, in group 3, respectively).

Median treatment duration was 80 days (range 2-1295) for ECP in groups 1-2 and 111 days (range 7-1142) for ruxolitinib in groups 2-3, with no significant differences (p=.804 and p=.094 respectively).

Onset of moderate-severe cGVHD was observed in 65 patients (28%) and more frequently noticed in group 2 (p=.001).

Among the most commonly observed infectious events, bacteremias occurred in 28 patients (12%) and were more frequent in group 3 (p=0.025); clinically relevant CMV reactivations occurred in 53 patients (23%) with no significant difference among the 3 groups.

After a median follow-up from transplant of 14 months (range 0.39-90), 1-year overall survival after transplant and after 2L treatment start were 70% and 63% in group 1, 58% and 55% in group 2 and 68% and 55% in group), without any significant differences among the 3 groups.

Conclusions: In GITMO experience ECP monotherapy has been mostly used in patients with SRaGVHD and prevalent skin involvement. Although ruxolitinib alone or with ECP were administered to patients with significantly more severe and multiorgan involvement, efficacy and infectious complications were similar to patients treated with ECP, suggesting that ruxolitinib could overcome the worse prognosis of these difficult-to-treat patients.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P223 MINI-DOSE METHOTREXATE COMBINED WITH METHYLPREDNISOLONE FOR INITIAL TREATMENT OF MILD ACUTE GVHD: A MULTI-CENTER, RANDOMIZED TRIAL

Yu Wang 1, Qi-Fa Liu2, De-Pei Wu3, Zheng-Li Xu1, Ting-Ting Han1, Yu-Qian Sun1,4, Fen Huang2, Zhi-Ping Fan2, Na Xu2, Feng Chen3, Ye Zhao3, Yuan Kong1, Xiao-Dong Mo1, Lan-Ping Xu1, Xiao-Hui Zhang1, Kai-Yan Liu1, Xiao-Jun Huang1,5

1Peking University People’s Hospital, Beijing, China, 2Nanfang Hospital Affiliated to Southern Medical University, Guangzhou, China, 3The First Affiliated Hospital of Soochow University, Soochow, China, 4Beijing Ludaopei Hematology Hospital, Beijing, China, 5Peking-Tsinghua Center for Life Sciences, Beijing, China

Background: There is an urgent unmet need for effective initial treatment for acute graft-versus-host disease (aGVHD) adding to the standard first-line therapy with corticosteroids after allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Methods: We performed a multi-center, open-label, randomized, phase 3 study. Eligible patients (aged 15 years or older, had received allo-HSCT for a hematological malignancy, developed aGVHD, and received no previous therapies for aGVHD) were randomly assigned (1:1), to receive either 5mg/m2 methotrexate (MTX), days 1,3,8 and then weekly combined with corticosteroids or corticosteroids alone.

Results: The primary endpoint was overall response rate (ORR) at day 10. 157 patients were randomly assigned to receive either MTX plus corticosteroids (n=78; MTX group) or corticosteroids alone (n=79; control group). The day 10 ORR was 97% for MTX and 81% for control (p= .005). Among patients with mild aGVHD, day 10 ORR was 100% for MTX and 86% for control (p= .001). Median time to first response (at least reaching partial response) was 3 days both for MTX (range 1-11) and control (range 2-8); p < .001). The median time needed to achieve a maximal response (complete response or partial response) was 6 days both for MTX and control. Estimated 6-month response durability was 74% (64 to 84) for MTX and 61% (50 to 72) for control (p= .047). The 1-year estimate for failure-free survival (defined in the part of “outcomes”) for MTX was 69% (95% CI 59–79) and 41% (29–53) for control (p= .002). Post-hoc multivariable analysis of risk factors for FFS showed that MTX was the only protective factor (hazard ratio [HR] 0.49, 95%CI, 0.30 to 0.82; p= .007; Table). There were no differences in treatment-related adverse events between the two groups.

Full size table

.

Conclusions: In conclusion, mini-dose MTX combined with corticosteroids can significantly improve the ORR in patients with mild aGVHD and is well tolerated.

Clinical Trial Registry: The trial was registered with clinicaltrials.gov(NCT04960644).

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P224 IMPROVED LONG-TERM OUTCOME OF RUXOLITINIB PLUS ECP VS RUXOLITINIB ALONE IN STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST DISEASE (SR-AGVHD)

Iryna Lastovytska1, Evgeny Klyuchnikov1, Silke Heidenreich1, Normann Steiner1, Radwan Massoud1, Niko Gagelmann1, Johanna Richter1, Christian Niederwieser1, Kristin Rathje1, Tatiana Urbanowicz1, Ameya Kunte1, Janik Engelmann1, Christina Ihne1, Cecilia Lindhauer1, Franziska Elisabeth Marquard1, Mirjam Reichard1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schäfersküpper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Ina Rudolph1, Gabriele Zeck1, Rolf Krause1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger 1

1University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: Steroid-refractory acute graft-versus-host disease (SR-aGvHD) is a serious complication after allogeneic hematopoietic stem cell transplantation (allo-SCT) and is associated with high mortality. Ruxolitinib - a JAK1/2 inhibitor - has become the standard of care treatment for SR-aGvHD but treatment failure is still frequent and there is still an unmet clinical need for improvement. Recently, our group has reported encouraging outcome in combining ruxolitinib with extracorporal Photopheresis (ECP) in SR-aGvHD (Modemann et al BMT 2020). Still, a comparison to treatment with ruxolitinib alone is lacking so far.

Methods: In this retrospective study we compared the long-term outcomes of 78 patients with SR-aGVHD treated at the University Medical Center Hamburg/Germany between December 2015 and August 2022. Treatment included either ruxolitinib alone (ruxo, n=29) or ruxo with ECP (ruxo-ECP, n=49). Patients in earlier years received ruxolitinib alone while Ruxo-ECP was used in more recent years. Ruxolinitb was started with 2 x 10 mg and ECP was started twice weekly. Patients were well balanced between both arms except for aGvHD grade IV which was higher in the ruxo-ECP arm (45% vs 14%, p<0,001). In both arms, steroids were tapered rapidly, and median steroid treatment was 39 days in ruxo and 35 days in ruxo-ECP.

Results: The overall response rate (ORR) including complete remissions (CR) of aGvHD at day 28 was 89,6% and 31% for ruxo versus 86% and 0% (p<0,001, respectively) for ruxo-ECP. At day 56, the ORR and CR rate was 90 % and 72 % in ruxo versus 86% and 19% (p<0,001, respectively), suggesting no improvement by adding ECP to ruxo. However, at 6 months, 24% in the ruxo arm but only 7% in the ruxo-ECP arm lost response, while ORR and CR increased in ruxo-ECP to 61% and 50% vs. 50% and 40%, (p=0,018, respectively) in the ruxo alone. At 12 months, ORR and CR were only 22% and 16,6% in the ruxo alone arm but 72% and 64% (p<0,001) in the ruxo-ECP arm. Furthermore, at 1 year, the cumulative incidence of chronic GvHD was significantly higher after ruxo alone versus ruxo-ECP: 51% (95% CI:33-69%) vs. 25% (95% CI:15-38%, p= 0.01) leading to GvHD/Relapse-free survival (GRFS) at 1 year of 7% (95% CI: 2-24%) after ruxo alone and 28% (95% CI: 17-42%, p=0.034) after Ruxo-ECP. We compared the NRM and relapse incidence at 1 year between ruxo alone and ruxo-ECP groups. Relapse: ruxo alone, 21%. (10-40%) vs. ruxo-ECP 10% (5-21%, p=0,18). NRM: ruxo alone 38% (22-57%) vs. ruxo-ECP 47% (34-65%), p=0,96.

Conclusions: Despite the limitations of this retrospective single-center study, the data suggest a better long-term control of aGvHD and less chronic GvHD at 1 year by ruxolitinib plus ECP than ruxolitinib alone in SR-aGvHD.

Disclosure: No conflict of interest.

12: Graft-versus-host Disease – Clinical

P225 COMPARISON BETWEEN POSTTRANSPLANT CYCLOPHOSPAMIDE AND ATG BASED REGIMENS AS GVHD PROPYLAXIS IN ONE-ANTIGEN MISMATCH UNRELATED DONOR TRANSPLANTATION

Gabriele Facchin1, Marta Lisa Battista 1, Maria De Martino2, Miriam Isola2, Antonella Geromin1, Chiara Savignano3, Valentina Simeon1,2, Alessandra Soravia1,2, Giuseppe Di Renzo1,2, Umberto Pizzano1,2, Martina Pucillo1,2, Renato Fanin1,2, Francesca Patriarca1,2

1Division of Hematology and Stem Cell Transplantation, University Hospital ASUFC, Udine, Italy, 2University of Udine, Udine, Italy, 3University Hospital ASUFC, Udine, Italy

Background: Use of posttransplant cyclophosphamide (PTCy) as graft-versus-host disease (GVHD) prophylaxis was recently extended to the mismatched unrelated donor hematopoietic stem cell transplantation (MMUD-HSCT) but there are only few comparative studies with calcineurin inhibitor+methotrexate±ATG regimens in this setting.

Methods: In this retrospective study we analyzed 78 adult patients who underwent one-antigen (HLA A, B, C or DR) MMUD HSCT between January 2015 and December 2022 at our Center. We compare the incidence of acute GVHD, PFS and OS in recipients of prophylaxis based on PTCy+Mycophenolate-mofetil+cyclosporine (PTCy group, n=20) vs calcineurin inhibitor+methotrexate+ATG (ATG group, n= 58).

Results: Median age at transplant was 58 (21-73) and 20% of patients was >65-year-old. The male/female sex ratio was 42/36. The most common underling hematological disease was acute leukemia (47/78-61%). Most patients of ATG group underwent to transplant before 2020 while most patients of PTCy group were transplanted after 2020. The two groups were well balanced in terms of age at transplant, sex, HSCT indication, HCT-CI, locus mismatch, graft source, intensity of conditioning regimen, CD34 + /kg and CD3 + /kg. More patients were in complete response (CR) before transplant in PTCy group (15/20-75% vs 28/58-49%, p=0,020) and received letermovir prophylaxis (90% vs 24%, p<0,001). There was no difference in acute GVHD (aGVHD) grade 2-4 incidence but there is a tendency of incidence reduction of severe aGVHD (grade 3 and 4) in PTCy cohort (0 vs 7/58-12%, p=0,071). Steroid refractory acute GVHD was significant more frequent in ATG group (18/58-31% vs 1/20-5%, p=0,019). VOD, microangiopathy, liver toxicity incidence was similar between two groups. At a median follow up of 19 months, 1/20 (5%) of patients were dead due to NRM in PTCy group vs 24/58 (41%) in ATG group (p= 0,002). The most common causes of NRM were GVHD complications (PTCy: 1 of 20 [5%]; ATG: 9 of 58 [16%]), infections (0 of 20; 5 of 58 [9%]) or others (0 of 20; 10 of 58 [17%]). The 1-year PFS was 63% (95% CI 51%-72%) globally, 90% (95% CI 66%-97%) in the PTCy group vs 53% (95% CI 40%-65%) in the ATG group (p=0,009). The 1-year OS was 65% (95% CI 54%-75%) globally, 95% (95% CI 69%-99%) in the PTCy group vs 55% (95% CI 42%-67%) in the ATG group (p=0,012). Figure 1.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (32)

Figure.1 OS in PTCy vs ATG groups.

Conclusions: In our real-life experience, PTCy results in a significative reduction of NRM rate and steroid refractory aGVHD incidence in one antigen MMUD HSCT setting. In PTCy cohort there is also a superior 1-year PFS and OS even if data should be further analyzed considering unbalanced distribution of CR rate. Studies with larger number of cases and prospective trials are needed to better define the optimal strategy to prevent GVHD in MMUD setting.

Disclosure: no conflict of interest to declare.

12: Graft-versus-host Disease – Clinical

P226 PROSPECTIVE EVALUATION OF PLASMA INFLAMMATORY PROTEINS FOR PREDICTION OF SEVER GRAFT-VERSUS-HOST DISEASE AFTER TRANSPLANT

Mingyang Wang1, Erlie Jiang 1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China

Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is an important treatment of hematological diseases. Acute graft-versus-host disease (aGVHD) is one of the main complications of allo-HSCT. The incidence of grade 2-4 aGVHD is as high as 50%, which seriously leads to non-relapse mortality (NRM) after allo-HSCT and affects succuss of allo-HSCT. Therefore, establishing a prediction method for severe aGVHD, identifying and intervening patients at high risk of severe aGVHD in advance is a problem that needs to be solved urgently. We aimed to establish a predictive method for severe aGVHD by detecting the levels of peripheral blood inflammatory proteins in allo-HSCT patients 14 days after transplantation.

Methods: Peripheral blood plasma samples of patients at 14 days after allo-HSCT were prospectively collected. The samples for detection were selected according to the condition of aGVHD after transplantation. A total of 33 cases of severe aGVHD, 17 cases of grade 2 aGVHD and 38 cases of grade 0-1 aGVHD were selected. 92 inflammatory proteins were quantitatively detected in a total of 88 plasma samples. A method for predicting severe aGVHD after allo-HSCT was established by differential analysis and Logistic regression model. Whether inflammatory proteins could predict NRM was also analyzed.

Results: Among the 88 patients, 60 (68.2%) underwent haploidentical HSCT and 28 (31.8%) underwent matched sibling donor HSCT. The median time of aGVHD onset was 32 (IQR 25 - 45) days after transplantation. Compared with patients without severe aGVHD, the plasma levels of IL-17A, IL-18, IL-10, PD-L1, CXCL9, uPA, TNFB, CD244, CXCL10, ADA, and IFN-γ were significantly upregulated in patients with severe aGVHD, and FGF-19 levels were significantly downregulated. Among these differential proteins, IFN-γ and CXCL10, CXCL10 and CXCL9, IFN-γ and CXCL9, IL-10 and IL-17A had obvious positive correlations. A Logistic regression model was established using the five most significantly different proteins, namely IL-17A, IL-18, IL-10, PD-L1 and CXCL9. In 88 patients, the model predicted severe aGVHD with an area under the receiver operating characteristic curve (AUROC) of 0.78. Using the Bootstrap method to internally validate the model, the AUROC was 0.759 (95% CI, 0.757 - 0.761), and the precision was 0.717 (95% CI, 0.713 - 0.715). The calibration curve showed that the probability of severe aGVHD predicted by the model was similar to the actual probability of severe aGVHD. The results of clinical decision curve analysis showed that preemptive intervention for patients with high risk of severe aGVHD after transplantation would have high benefits, except in rare cases when the damage to the patient was much greater than the benefit of the intervention. In addition, the levels of the detected inflammatory proteins had no significant effect on the occurrence of NRM in patients with severe aGVHD.

Conclusions: The results of this study showed that the Logistic regression model established based on the levels of IL-17A, IL-18, IL-10, PD-L1 and CXCL9 in the peripheral blood 14 days after allo-HSCT could predict severe aGVHD. Through large-scale cohort research and full verification, it is expected to become an effective method applied clinically for predicting severe aGVHD.

Disclosure: The authors declare no confilct of interest.

12: Graft-versus-host Disease – Clinical

P227 LYMPHOPENIA BUT NOT ABSOLUTE LYMPHOCYTE COUNT BEFORE ANTI-T-LYMPHOCYTE GLOBULIN (ATLG) ADMINISTRATION IMPACTS TRANSPLANT OUTCOMES IN MATCHED UNRELATED PERIPHERAL BLOOD STEM CELL ALLOGENEIC STEM CELL TRANSPLANTATION

Radwan Massoud 1, Evgeny Klyuchnikov1, Normann Steiner1, Christian Niederwieser1, Ameya Shirinian Kunte1, Silke Heidenreich1, Kristin Rathje1, Nico Gagelmann1, Tetiana Perekhrestenko1, Gaby Zeck1, Rolf Krause1, Christine Wolschke1, Dietlinde Janson1, Francis Ayuk1, Nicolaus Kröger1

1University Medical Center Hamburg Eppendorf, Hamburg, Germany

Background: Data on optimal ATLG dosing in MUD-PBSC-allo-SCT is limited. Conventional weight-based dosing may deplete T-cells and lead to unfavorable outcomes. Absolute lymphocyte count (ALC)-based dosing on ATLG administration day has been proposed as an alternative. We conducted a retrospective study at University Medical Center Hamburg-Eppendorf (UKE) to investigate the impact of ALC and ATLG dose on transplant outcomes.

Methods: We included 396 patients who underwent allo-SCT (years 2018-2022) with PBSCs from MUD (HLA 10/10) and received ATLG at a dose of 30mg/kg (n=151) or 60mg/kg (n=49) between days -4 and -1. Patients were categorized into the “NM-ALC” group with non-measurable lymphocytes (ALC < 0.1Mrd/L) and the “M-ALC” group with measurable lymphocytes (ALC ≥ 0.1Mrd/L) at time of ATLG application, allowing for a comparison of transplant outcomes between these groups.

Results: Patients, mainly with AML (42%) or MDS(18%), were divided into NM-ALC (n=71) and M-ALC (n=325) groups. Median follow-up was 12 months (range,1-51). Engraftment was similar across groups. ALC analyzed as a continuous variable, had no impact on outcomes.

NM-ALC group had lower 1-year-OS than M-ALC (65% vs. 77%, p=0.006). This difference persisted in the ATLG-30mg/Kg subgroup (NM-ALC 60% vs 74% M-ALC, p=0.001). Within NM-ALC, ATLG-60mg/Kg improved 1-year-OS over 30mg/Kg (80% vs. 60%, p=0.027). On MVA a higher ECOG(≥2) reduced OS (HR 3.03, 95%CI 1.85-4.9, p<0.0001).

NM-ALC group had lower 1-year-PFS than M-ALC (56% vs. 67%, p=0.008). This difference persisted in the ATLG-30mg/Kg-subgroup (NM-ALC 49% vs 65% M-ALC, p=0.001). Within NM-ALC, ATLG-60mg/Kg improved 1-year-PFS over 30 mg/Kg (80% vs. 49%, p=0.024). On MVA a higher ECOG(≥2) reduced PFS (HR 2.37, 95%CI 1.74-3.8, p < 0.0001).

GRFS was comparable between groups. In NM-ALC, ATLG-60mg/Kg had higher 1-year-GRFS than 30mg/Kg (67% vs. 34%, p=0.038). In M-ALC, ATLG-60 mg/Kg showed a trend towards better 1-year-GRFS compared to 30 mg/Kg (47% vs 38%, p=0.08). On MVA Higher ECOG(≥2) decreased GRFS (HR 2.1, 95%CI 1.4-3.16, p<0.0001).

NM-ALC had higher 1-year-NRM than M-ALC (24% vs. 13%, p=0.02). This difference persisted in the ATLG-30mg/Kg-subgroup (NM-ALC 27% vs 15% M-ALC, p=0.013). In MVA, a higher ECOG (≥2) (HR 2.65, 95%CI 1.3-5.4, p < 0.0075) and patient age (HR 1.05, 95%CI 1.019-1.08, p=0.0015) increased NRM.

There were no differences in CIR.

NM-ALC group had a trend towards lower incidence of aGVHD-GII-IV at day 100 than M-ALC (7% vs. 15%, p=0.06). However it was significantly lower within the ATLG-30mg/Kg-subgroup (5% vs. 17%, p=0.037). On MVA we observed a trend for increased aGVHD II-IV for the M-ALC (HR 5.62, 95%CI 0.96-7.1, p=0.06).

NM-ALC group had a lower incidence of cGVHD-moderate/severe at 1yr than M-ALC (17% vs. 28%, p=0.049). On MVA a trend for increased cGVHD-moderate/severe for the M-ALC vs NM-ALC was observed (HR 1.72, 95%CI 0.9-3.21, p=0.099).

Conclusions: In conclusion, although ALC itself shows no significant effect on outcomes, NM-ALC is linked to a reduced GVHD incidence but detrimentally affects NRM, PFS, and OS. Notably, these adverse impacts persist regardless of lower ATLG dosages.

Disclosure: nothing to declare.

12: Graft-versus-host Disease – Clinical

P228 A PROSPECTIVE, MULTI-CENTER, RANDOMIZED, CONTROLLED CLINICAL TRIAL TO EXPLORE THE ROLE OF MESENCHYMAL STEM CELLS IN PREVENTING GRAFT VERSUS HOST DISEASE

Han Yao 1, Xiaoqi Wang1, Ruihao Huang1, Haixia Fu2, Ren Lin3, Yimei Feng1, Xiaojuan Deng1, Ting Chen1, Lidan Zhu1, Jia Liu1, Yuqing Liu1, Lu Zhao1, Lu Wang1, Shichun Gao1, Huanfeng Liu1, Cheng Zhang1, Peiyan Kong1, Li Gao1, Qifa Liu3, Xiaohui Zhang2, Xi Zhang1,4,5

1Army Medical University affiliated Xinqiao Hospital, Chongqing, China, 2Peking University People’s Hospital, Beijing, China, 3Nanfang Hospital, Southern Medical University, Guangzhou, China, 4Jinfeng Laboratory, Chongqing, China, 5State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China

Background: Although haploid hematopoietic stem cell transplantation (haploid-HSCT) is a highly beneficial therapy for acute leukemia, it is now acknowledged as a critical issue that needs to be resolved in associated with a higher risk of graft-versus-host disease (GVHD). MSCs are one option for prophylaxis against GVHD. Currently, the majority of researches on MSCs in GVHD prevention focuses on MSCs preventing aGVHD or cGVHD alone. Our aim is to lower the incidence of both aGVHD and cGVHD by using an effective and safe method through co-transplanting MSCs and HSC, as well as staged post-transplant administration of MSCs (“MSC 0-day regimen”).

Methods: Umbilical MSCs were infused into adult haploid-HSCT patients without undergoing genetic modification or gene editing. Using the complete randomization process, the groups were split into 1:1 ratios. MSCs should be administered on day 0, once per week for the first month post-HSCT, once every 2 weeks for the second month post-HSCT, and then once for the third month post-HSCT, with a dose of 1*106 per kilogram. The primary endpoint was the incidence of 2-year cGVHD, whereas the secondary endpoints were the incidence of aGVHD, 2-year overall survival (OS), and leukemia relapse-free survival. R version 4.3.0 have been utilized for the statistical analysis.

Results: Demographic features of recipients, disease information and donor characters concerned with age, mismatch of gender, HLA typing, ABO blood types, the percentage of collateral kin donors, and the nucleated cells of the graft showed no significant difference(p>0.05). The only exception was the amount of CD34+ cells, which was 5.328 (±2.348) in the MSC group and 6.153 (±2.166) in the control group (p=0.012).

The incidence of clinical cGVHD varied between the two groups (46 vs 27, p= 0.001). The incidence of 2-yearc GVHD was 0.389 (95% CI: 0.274-0.697) in the control group and 0.289 (95% CI: 0.189-0.575) in the MSC group (p=0.044). The incidence of mild and moderate severity was similar between the groups (p=0.746 and 0.344, respectively); the cumulative incidence of severe cGVHD was significantly different (p=0.043), with the control group was 0.109 (95% CI: 0.035-0.173) and the experimental group was 0.054 (95% CI: 0.017-0.125). There were variations in lung cGVHD, with 9 cases in the control group and none in the MSC group, but no appreciable changes were found in the incidence of cGVHD in skin, mouth, eyes, liver, gastrointestinal tract, or joints between the groups (p > 0.05).

The control group had an overall cumulative incidence of aGVHD of 0.534 (95% CI: 0.399- 0.650) while the MSC group had an incidence of 0.348 (95% CI: 0.199-0.501) (p=0.009). A comparison analysis between the control group and MSC revealed significant differences between the two groups’ 2-year GVHD and relapse free survival(GRFS), which were 0.370 (95% CI: 0.279-0.490) and 0.597 (95% CI: 0.487-0.732), respectively(p< 0.001). No discernible differences between the two groups in terms of OS, relapse, or adverse events.

Conclusions: The “MSC 0-day regimen” is helpful in preventing both cGVHD and aGVHD in haploid-HSCT, with no increased adverse consequences and post-transplant recurrence.

Clinical Trial Registry: ChiCTR1900022292;https://www.chictr.org.cn/showproj.html?proj=37370.

Disclosure: We declare that we have no conflict of interest.

This work was supported by National Key Development Program of China (2017YFA0105502). We are also grateful to the support, cooperation, and trust of the patients.

12: Graft-versus-host Disease – Clinical

P229 RUXOLITINIB AS SECOND-LINE TREATMENT FOR BOS AFTER HEMATOPOIETIC CELL TRANSPLANTATION

Feng Chen 1, Yuqing Tu1,2,3, Jia Chen1,2,3, Shushu Xu1,2,3, Yi Fan1,2,3, Mimi Xu1,2,3, Mengqi Xiang1,2,3, Tiemei Song1,2,3, Xiang Zhang1,2,3, Xiaoli Li4, Lian Bai5, Depei Wu1,2,3

1National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China, 2Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China, 3Key Laboratory of Stem Cells and Biomedical, Materials of Jiangsu Province and Chinese Ministry of Science and Technology, Suzhou, China, 4Soochow Hopes Hematonosis Hospital, Suzhou, China, 5Suzhou Canglang Hospital, Suzhou, China

Background: Bronchiolitis obliterans syndrome (BOS) is the manifestation of pulmonary chronic graft-versus-host disease (cGVHD) after hematopoietic cell transplantantion (HCT). Ruxolitinib has shown a significant response in cGVHD, but few studies have reported its efficacy in BOS.

Methods: All patients who developed BOS post-HCT between 2015 and 2022 and failed to the first-line treatment were retrospectively screened. First-line therapy including glucocorticoid and calcineurin inhibitors (FK506/CSA), with or without FAM (Fluticasone + Azithromycin + Montelukast). Enrolled patients were divided into three groups as follows: Group A (N=59) were exposed to ruxolitinib usually at a dose of 5 mg twice daily; Group B (N=39) were exposed to alternative second-line drugs at the direction of the physician in charge; Group C were patients continued existed therapy after first-line treatment evaluation (N=48). Lung symptom score was be performed using both the symptoms and FEV1 scores whenever possible.

Results: Three months after treatment, the overall response rate (ORR) in Group A was 83.05%, significantly higher than 56.41% in Group B (p= 0.004) and 62.50% in Group C (p= 0.016). For patients with Score 3, the ORR was 57.89% in Group A, which was also higher than that in Group B (ORR=23.53%, p= 0.037) or in Group C (ORR=10.00%, p= 0.013)(Table). Multivariate analysis revealed that lung symptom score (OR= 0.300, 95% CI 0.097–0.930, p= 0.037) and interval from BOS diagnosis to ruxolitinib initiation (OR= 0.584, 95% CI 0.344–0.994, p= 0.047) were independent risk factor in Group A. Adverse effects had no statistical difference among the three groups.

Table: Overall response rate in three groups.

ORR, n (%)

Group A

Group B

Group C

P1

P2

Total

49 (83.05)

22 (56.41)

30 (62.50)

0.004

0.016

Lung symptom score 0

4 (100)

3 (100)

8 (100)

/

/

Lung symptom score 1

30 (96.77)

13 (92.86)

18 (94.74)

0.555

0.721

Lung symptom score 2

4 (80.00)

2 (40.00)

3 (27.27)

0.197

0.049

Lung symptom score 3

11 (57.89)

4 (23.53)

1 (10.00)

0.037

0.013

P1, Group A vs. Group B; P2, Group A vs. Group C; ORR, overall response rate.

Conclusions: We found ruxolitinib was effective and safe as a second-line option for BOS post-HCT. The efficacy could be improved if used ruxolitinib earlier after diagnosis or when the lung symptom score is low.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P230 POST-TRANSPLANTATION CYCLOPHOSPHAMIDE WITH OR WITHOUT PROPHYLACTIC DONOR LYMPHOCYTE INFUSION AFTER HLA-MATCHED ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN MDS/AMl

Joost van der Hem 1, Jurjen Versluis2, Iris Erpelinck2, Chantal Mouws2, Peter Von dem Borne1, Annoek Broers2, Stijn Halkes1

1Leiden University Medical Centre, Leiden, Netherlands, 2Erasmus Medical Centre, Rotterdam, Netherlands

Background: Post-transplantation cyclophosphamide (PTCy) – based graft-versus-host disease (GvHD) prophylaxis is increasingly used for HLA-matched allogeneic hematopoietic stem cell transplantation (alloHSCT). While GvHD can be successfully prevented by PTCy-based alloHSCT, relapse incidence remains of concern due to a potential reduced donor-derived alloreactivity against the underlying the disease. This alloreactivity can be enhanced after alloHSCT by early tapering of prophylactic immunosuppression or administration of prophylactic donor lymphocyte infusion (proDLI). Currently, no optimal strategy for this post-transplantation immunomodulation has been defined. In this retrospective study, we report the outcomes of two immunomodulatory strategies after PTCy-based HLA-matched alloHSCT, comparing a short course of monotherapy ciclosporine A (CsA) with prolonged dual immune suppression with mycophenolate mofetil (MMF) and tacrolimus (TAC) in combination with subsequent proDLI.

Methods: Two university hospitals in the Netherlands participated in this study, which included patients that received a PTCy-based peripheral blood alloHSCT with an HLA-matched related or 10 out of 10 matched unrelated donor between 2019-2022 for high-risk myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) in complete remission after induction therapy. In the LUMC, Leiden, PTCy was combined with dual prophylactic immunosuppressive treatment (MMF until day +30 and TAC tapered until day +120-150 and proDLI was administered at 4-6 months post-transplant in the absence of GvHD and after tapering of immune suppression. In the ErasmusMC, Rotterdam, PTCy was combined with CsA until day +70 and DLI was administered pre-emptively (preDLI) in case of minimal residual disease (MRD) or persistent mixed chimerism. The primary endpoints were cumulative incidence (CI) of severe GvHD and relapse 1 year after alloHSCT. Severe GvHD was defined by the need of systemic immunosuppressive treatment for acute or chronic GvHD. For CI of GvHD, death and relapse were competing events. For relapse, only death was a competing event. The CI curves were compared with the Gray’s test.

Results: A total of 92 patients received alloHSCT with the MMF/TAC/proDLI strategy and 130 patients CsA/preDLI (table 1). Baseline characteristics were comparable, although MMF/TAC/proDLI patients had a lower HCT comorbidity index and received hypomethylating agents pre-transplant more frequently. Fifty-nine MMF/TAC/proDLI patients (64%) received DLI at a median time of 4.8 months after alloHSCT (IQR: 4.2-6.2) of whom 3 patients developed severe GvHD. Eight CsA/preDLI patients (6%) received DLI (median 6.5 months, IQR 6.2-7.8), of whom none developed severe GvHD. The CI of severe GvHD 1 year after alloHSCT was 10% (95% confidence interval: 4-18) in MMF/TAC/proDLI and 34% (26-42) in CsA/preDLI (p<0.001). The CI of relapse at 1 year was 13% (6-22) in MMF/TAC/proDLI and 11% (6-17) in the CsA/preDLI group (p=0.8). Overall survival at 1 year was comparable with 83% (74-93) for MMF/TAC/proDLI, and 78% (71-86) for CsA/preDLI.

MMF/TAC/proDLI (n=92)

CsA/preDLI (n=130)

Sex

Male

59 (64%)

83 (64%)

Age at alloSCT

Median (range)

63 (22-78)

61 (23-75)

HCT comorbidity index

45 (49%)

19 (15%)

1-2

30 (33%)

38 (29%)

>2

17 (19%)

73 (56%)

ELN risk score

Favorable

18 (20%)

28 (22%)

Intermediate

32 (35%)

31 (24%)

Adverse

36 (39%)

63 (49%)

Relapsed MDS/AML

6 (7%)

8 (6%)

Induction treatment

Intensive chemotherapy

64 (70%)

121 (93%)

Hypomethylating agents

28 (30%)

9 (7%)

MRD positivity before alloSCT

MRD- / no MRD

63 (69%)

82 (63%)

MRD+

29 (32%)

48 (37%)

Donor

Matched related donor

20 (22%)

27 (21%)

Matched unrelated donor

72 (78%)

103 (79%)

Conclusions: The combined strategy with dual immune suppression and proDLI after PTCy-based HLA-matched alloHSCT yielded low severe GVHD rates without compromising the anti-leukemic effect compared with a short course of CsA and preDLI strategy. Longer follow-up is needed to determine the long-term GVHD- and relapse free survival after MMF/TAC/proDLI.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P231 IMPROVED GRFS WHEN ATG/ATLG IS USED IN ALLO-PBSCT FROM MATCHED SIBLING DONORS - AN EBMT REGISTRY STUDY BY THE TRANSPLANT COMPLICATIONS WORKING PARTY

Agnieszka Piekarska 1, Mouad Abouqateb2, William Boreland2, Christophe Peczynski2, Jan Maciej Zaucha1, Nicolaus Kröger3, Robert Zeiser4, Fabio Ciceri5, Thomas Schroeder6, Thomas Luft7, Jakob Passweg8, Desiree Kunadt9, Matthias Stelljes10, Igor Wolfgang Blau11, Uwe Platzbecker12, Ibrahim Yakoub-Agha13, Didier Blaise14, Anna Maria Raiola15, Johanna Tischer16, Eva Maria Wagner-Drouet17, Julia Winkler18, Christoph Schmid19, Gerald Wulf20, Matthias Edinger21, Johan Maertens22, Friedrich Stölzel23, Jan Vydra24, Pavel Zak25, Ivan Moiseev26, Hélène Schoemans22, Olaf Penack11, Zinaida Perić27

1University Clinical Center, Medical University of Gdańsk, Gdańsk, Poland, 2EBMT Paris Study Office / CEREST-TC, INSERM UMR-S 938, Sorbonne University, Hôpital Saint Antoine, Paris, France, 3University Hospital Eppendorf, Hamburg, Germany, 4University of Freiburg, Freiburg, Germany, 5IRCCS Ospedale San Raffaele s.r.l, Milano, Italy, 6University Hospital Essen, Essen, Germany, 7University of Heidelberg, Heidelberg, Germany, 8University Hospital Basel, Basel, Switzerland, 9University Hospital TU Dresden, Dresden, Germany, 10University of Muenster, Muenster, Germany, 11Charité, Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany, 12Medical Clinic and Policinic 1, Leipzig, Germany, 13CHU de Lille, Lille, Germany, 14Programme de Transplantation & Therapie Cellulaire, Marseille, France, 15IRCCS Ospedale Policlinico San Martino, Genova, Italy, 16Klinikum Grosshadern, Munich, Germany, 17University Medical Center Mainz, Mainz, Germany, 18University Hospital Erlangen, Erlangen, Germany, 19Klinikum Augsburg, Augsburg, Germany, 20Universitaetsklinikum Goettingen, Goettingen, Germany, 21University Regensburg, Regensburg, Germany, 22University Hospital Gasthuisberg, Leuven, Belgium, 23University Hospital Schleswig-Holstein, Campus Kiel, Kiel University, Kiel, Germany, 24Institute of Hematology and Blood Transfusion, Prague, Czech Republic, 25Charles University Hospital, Hradec, Czech Republic, 26RM Gorbacheva Research Institute, Pavlov University, St Petersburg, Russian Federation, 27University Hospital Centre Rijeka and School of Medicine, University of Rijeka, Rijeka, Croatia

Background: According to the current EBMT recommendations, rabbit anti-thymocyte or anti-T-lymphocyte globulin (rATG/ATLG) should be used in the matched sibling donor (MSD) setting with a high risk of graft-versus-host disease (GVHD). However, discrepancies between recommendations and clinical practice were found in the EBMT survey. Therefore, we performed a real practice study to evaluate ATG/ATLG impact on post-transplantation outcomes.

Methods: This EBMT Registry-based study included adult patients with hematological malignancies after the first allogeneic peripheral blood stem cell transplantation (allo-PBSCT) performed from MSDs between 2014-2021 with or without rATG/ATLG as an adjunction to calcineurin inhibitor and antimetabolite-based GVHD prophylaxis. Patients transplanted with post-transplantation cyclophosphamide, ex vivo T-cell depletion, or alemtuzumab were excluded.

We conducted multivariate analyses using Cox cause-specific hazard models, adjusting for known risk factors and variables that exhibited significant differences between the two primary comparison groups: non-ATG and ATG.

Patients receiving rATG/ATLG were pooled for dose-dependent analyses to have received intermediate-ATG (Thymoglobulin 4.5-6 mg/kg or Grafalon 25-35 mg/kg), low-ATG (below intermediate-ATG), or high-ATG (above intermediate-ATG) categories according to the total dose reported.

Results: Overall, 11420 patients were included (non-ATG n=7680 and ATG n=3740). The patient-, donor- and transplant-related characteristics are presented in Table 1.

Table 1. Patient-, donor- and transplant-related characteristics by graft-versus-host disease prevention strategy.

Non- ATG (N=7680)

ATG

(N=3740)

Total

(N=11420)

p-value

Patient Sex, n (%)

0.691

Male

4473 (58.2%)

2193 (58.6%)

6666 (58.4%)

Female

3207 (41.8%)

1547 (41.4%)

4754 (41.6%)

Donor Sex, n (%)

< 0.011

Male

4298 (56.0%)

1966 (52.6%)

6264 (54.9%)

Female

3382 (44.0%)

1774 (47.4%)

5156 (45.1%)

Age at Transplant, years

< 0.012

median [Q1, Q3]

49.7 (37.2, 58.7)

53.9 (43.1, 60.8)

51.3 (39.1, 59.6)

[Min, Max]

18.0 - 77.5

18.1 - 78.9

18.0 - 78.9

Karnofsky, n (%)

< 0.011

< 90

1737 (23.9%)

1055 (29.3%)

2792 (25.7%)

>= 90

5520 (76.1%)

2551 (70.7%)

8071 (74.3%)

Missing count

423

134

557

Disease Risk Index, n (%)

< 0.011

Low

755 (9.8%)

287 (7.7%)

1042 (9.1%)

Intermediate

4946 (64.4%)

2455 (65.6%)

7401 (64.8%)

High

1600 (20.8%)

835 (22.3%)

2435 (21.3%)

Very high

379 (4.9%)

163 (4.4%)

542 (4.7%)

Hematological Malignancies, n (%)

AML

3473 (45.2%)

1973 (52.8%)

5446 (47.7%)

ALL

1403 (18.3%)

285 (7.6%)

1688 (14.8%)

MDS

764 (9.9%)

470 (12.6%)

1234 (10.8%)

NHL

754 (9.8%)

266 (7.1%)

1020 (8.9%)

MPN

339 (4.4%)

353 (9.4%)

692 (6.1%)

CML

262 (3.4%)

91 (2.4%)

353 (3.1%)

MM

180 (2.3%)

98 (2.6%)

278 (2.4%)

HL

218 (2.8%)

49 (1.3%)

267 (2.3%)

MDS & MPN

158 (2.1%)

106 (2.8%)

264 (2.3%)

CLL

128 (1.7%)

49 (1.3%)

177 (1.5%)

Other

1 (0.0%)

0 (0.0%)

1 (0.0%)

Donor to Patient

< 0.011

CMV serostatus, n (%)

Neg to Neg

1241 (16.7%)

759 (20.6%)

2000 (18.0%)

Neg to Pos

1029 (13.9%)

580 (15.7%)

1609 (14.5%)

Pos to Neg

618 (8.3%)

361 (9.8%)

979 (8.8%)

Pos to Pos

4541 (61.1%)

1989 (53.9%)

6530 (58.7%)

Missing count

251

51

302

Myeloablative Conditioning, n (%)

< 0.011

No

2621 (34.5%)

1681 (45.4%)

4302 (38.1%)

Yes

4973 (65.5%)

2023 (54.6%)

6996 (61.9%)

Missing count

86

36

122

TBI, n (%)

< 0.011

No

5506 (71.8%)

3227 (86.3%)

8733 (76.5%)

Yes

2167 (28.2%)

513 (13.7%)

2680 (23.5%)

Missing count

7

7

  1. 1. Pearson’s Chi-squared test
  2. 2. Wilcoxon rank sum test
  3. ATG, anti-thymocyte globulin; AML, acute myeloid leukemia; ALL, acute lymphoblastic leukemia; CLL, chronic lymphocytic leukemia; CML, chronic myelogenous leukemia; HL, Hodgkin lymphoma; GVHD, graft-versus-host disease; MDS, myelodysplastic syndrome; MPN, myeloproliferative neoplasm; MM, multiple myeloma; MMF, mycophenolate mofetil; MTX, methotrexate; NHL, non-Hodgkin lymphoma; TBI, total body irradiation

The incidences of aGVHD II-IV and III-IV at day +100 in the non-ATG and ATG groups were 27.6% vs. 21.6% and 12.2% vs. 8.2%, respectively. The median follow-up was 2.9 years. The cumulative incidence of chronic GVHD (cGVHD) and extensive cGVHD at 2 years was 48.9% vs. 30% and 25.5% vs. 13%. The 2-year overall survival (OS) was 62.9% vs. 63.3%, non-relapse mortality (NRM) 16% vs. 12.5%, relapse incidence (RI) 30.2% vs. 34.7%, progression-free survival (PFS) 53.9% vs. 52.8%, and GVHD-free relapse-free survival (GRFS) 32.2% vs. 40.7%.

In multivariate analysis, rATG/ATLG use lowered the risk of both aGVHD II-IV (HR 0.7, 95% CI 0.62 to 0.79; p<0.001) and cGVHD (HR 0.45, 95% CI 0.41 to 0.51; p<0.001), including extensive cGVHD (HR 0.39, 95% CI 0.34 to 0.46; p<0.001). The positive influence of rATG/ATLG was also observed for OS (HR 0.89, 95% CI 0.81 to 0.97; p=0.009), NRM (HR 0.63, 95% CI 0.54 to 0.74; p<0.001), and GRFS (HR 0.72, 95% CI 0.67 to 0.78; p<0.001). However, we noted increased RI risk in the ATG group (HR 1.22, 95% CI 1.11 to 1.33; p<0.001) without significant impact on PFS (HR 1.0, 95% CI 0.92 to 1.08; p=0.98).

The outcomes were optimal for the intermediate dose range in a subgroup analysis of patients who received rATG/ATLG. Low rATG/ATLG doses increased cGVHD incidence (HR 1.28, 95% CI 1.03 to 1.59; p=0.029). High rATG/ATLG doses had an unfavorable impact on OS (HR 1.29, 95% CI 1.05 to 1.59; p=0.017), NRM (HR 1.56, 95% CI 1.10 to 2.22; p=0.012), PFS (HR 1.29, 95% CI 1.07 to 1.56; p=0.007) with a trend towards worse GRFS (HR 1.18, 95% CI 0.99 to 1.41; p=0.061).

Conclusions: Our results support the EBMT guidelines to use rATG/ATLG in MSD alloPBSCT recipients with hematologic malignancies. The best outcomes, including GRFS are obtained with the recommended intermediate rATG/ATLG dose.

Disclosure: There is nothing to declare for this topic except:

AP has received honoraria or travel support from Alexion, AstraZeneca, Astellas, Gilead, MSD, Novartis, Pierre Fabre, Pfizer, Sanofi and SOBI.

FS has received travel support from Janssen and Medac, honoraria from Medac, GWT, Abbvie, Servier and Pierre Fabre and research funding from Medac and Servier.

HS reports having received personal fees from Incyte, Janssen, Novartis, Sanofi and from the Belgian Hematological Society (BHS), as well as research grants from Novartis and the BHS, all paid to her institution and not directly related to this work. She has also received non-financial support (travel grants) from Gilead, Pfizer, the EBMT (European Society for Blood and Marrow Transplantation) and the CIBMTR (Center for International Bone Marrow Transplantation Research).

OP has received honoraria or travel support from Gilead, Jazz, MSD, Neovii, Novartis, Pfizer and Therakos. He has received research support from Incyte and Priothera. He is a member of advisory boards to Equillium Bio, Jazz, Gilead, Novartis, MSD, Omeros, Priothera, Sanofi, Shionogi and SOBI.

RZ reports receiving personal fees from Incyte, MNK, Novartis, Sanofi, Medac and Neovii.

12: Graft-versus-host Disease – Clinical

P232 NAVIGATING GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS IN MATCHED SIBLING DONOR ALLOGENEIC TRANSPLANTS: A COMPARATIVE ANALYSIS OF POST-TRANSPLANT CYCLOPHOSPHAMIDE VERSUS DIVERSE REGIMENS – INSIGHTS FROM A SINGLE-CENTER EXPERIENCE

Majed Altareb1, Carol Chen1, Mats Remberger2, Armin Gerbitz1,3, Dennis Kim 1,3, Rajat Kumar1,3, Wilson Lam1,3, Ivan Pasic1,3, Fotios Michelis1,3, Igor Novitzky-Basso1,3, Auro Viswabandya1,3, Jonas Mattsson1,3, Arjun Datt Law1,3

1Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology, Princess Margaret Hospital, Toronto, Canada, 2KFUE, Uppsala University Hospital, Uppsala, Sweden, 3University of Toronto, Toronto, Canada

Background: Post-transplant cyclophosphamide (PTCy) is a promising strategy to mitigate Graft-versus-Host Disease (GVHD) in HLA-matched allogeneic stem cell transplant (allo-SCT). This study seeks to examine the impact of PTCy on clinical outcomes, GVHD incidence, GVHD- and relapse-free survival (GRFS) and overall survival (OS) in patients undergoing HLA-matched allo-SCT. We compared the outcomes of patients who underwent HLA-matched sibling donor allo-SCT for hematologic malignancies and received PTCy-based GVHD prophylaxis with those receiving other protocols.

Methods: We analyzed transplant outcomes among 413 patients who received HLA-matched sibling donor allo-SCT for hematological malignancies at Princess Margaret Hospital from January 2010-March 2023.

Results: Patient demographics and clinical characteristics are shown in Table 1. Four hundred- thirteen individuals received allo-SCT from HLA matched siblings for hematological malignancy: of these 217 recipients were males (52.5%), with a median recipient age of 56 (range 18-73). At the time of transplant, 240 (58.1%) patients were in first complete remission (CR1) and 57 (13.8) in CR2/3. All recipients received peripheral blood stem cells. The median infused CD34+ cell dose was 6.8 × 106/Kg recipient weight (range 1.2- 25.8). 198 (48%) patients received myeloablative conditioning (MAC), whereas 215 (52%) received reduced-intensity conditioning.

With a median follow up of 53.6 (4.9-163) months, OS at 3 years was 56.7% (95% CI: 51.5-61.5), Non-relapse mortality was 16.8% (95% CI: 13.3- 20.6) at 1 year and 24.6% (95% CI: 20.4-29.0) at 3 years. Cumulative incidence of relapse at 3 years was 20.8% (95% CI: 16.9-25.0). At day-100, the incidence of grade 2-4 and grade 3-4 acute GVHD was 32.0% (95% CI: 27.5-36.5) and 9.2% (95% CI: 6.7- 12.2), respectively. The incidence of moderate-severe chronic GVHD at 3 years was 44.1% (95% CI: 38.9- 49.1). GRFS at three years was 38.8% (95% CI: 25.0-43.7). Our findings revealed a significant reduction in both acute and chronic GVHD incidence among patients receiving PTCy-based prophylaxis compared to those receiving conventional regimens. In univariable analysis, use of PTCy had a significant impact on GVHD, with lower risk of grade 2-4 aGVHD (HR=0.52, P < 0.001), grade 3-4 aGVHD (HR=0.24, P < 0.001), and moderate-severe cGVHD (HR=0.35, P < 0.001). This reduction is of paramount importance given the considerable morbidity and mortality associated with GVHD, as patients in the PTCy group demonstrated superior GRFS (HR=0.37, P < 0.001).

Transplant-related complications, including CMV infections and organ toxicities were comparable between the PTCy and conventional prophylaxis groups. This suggests that while PTCy exerts its GVHD-protective effects, it does not compromise the overall safety profile of the transplant. It is noteworthy that OS at specified time points post-transplant was comparable between PTCy based regimens & other groups (67.15% (95% CI: 50.3- 81.8), P= 0.032), indicating that the integration of PTCy into GVHD prophylaxis regimens does not compromise long-term outcomes.

Variable

N (%) or Median (Range)

Age

56 (18-73)

≥65y

71 (17.2)

Gender:

Male

217

Female

196

Diagnosis:

AML

224 (54.2)

ALL

48 (11.6)

MDS

63 (15.3)

CML

19 (4.6)

MF

39 (9.4)

CMML

11 (2.7)

MPAL

9 (2.2)

Pre-transplant Status:

CR1

240 (58.1)

CR2-3

57 (13.8)

Stable disease

108 (26.2)

Not in CR

7 (1.7)

Donor age (Median)

54 (8-74)

CD34+ dose

6.8 (1.2-25.8)

Female donor to Male recipient

107 (25.9)

Graft Source:

Fresh graft:

212 (51.3)

Frozen graft

201 (48.7)

Conditioning:

MAC

198

RIC

215

GVHD prophylaxis:

CsA+MMF

163 (39.5)

CsA+MTX

47 (11.4)

PTCy+CsA+MMF

88 (21.3)

PTCy+ATG+CsA

79 (19.1)

CsA+MTX + ATG

33 (8.0)

PTCy+CsA

3 (0.7)

ATG dose:

No ATG

301 (72.9)

2 mg/kg

56 (13.6)

4.5 mg/kg

56 (13.6)

KPS <90

89 (21.5)

HCT-CI: ≥3

137 (33.2)

DRI:

Low

6 (1.5)

Intermediate

203 (49.2)

High

133 (32.2)

Very high

71 (17.2)

Follow up (months)

53.6 (4.9-163)

Conclusions: Our study provides compelling evidence that PTCy as a GVHD prophylaxis in HLA-matched sibling donor allo-SCT significantly reduces GVHD incidence without compromising other outcomes. The retrospective nature of this analysis warrants cautious interpretation and prospective studies with larger cohorts and extended follow-up periods are essential for validation.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P233 POST TRANSPLANT CYCLOPHOSPHAMIDE AS GVHD PROPHYLAXIS IN PATIENTS RECEIVING HLA MISMATCHED (7/8) UNRELATED DONOR FOR MYELOID MALIGNANCIES: RESULTS OF THE GITMO PROSPECTIVE PHYLOS TRIAL

Anna Maria Raiola 1, Benedetto Bruno2, Antonio Maria Risitano3, Federico Mosna4, Irene Maria Cavattoni4, Francesco Onida5, Giorgia Saporiti5, Francesca Patriarca6, Marta Lisa Battista6, Vincenzo Pavone7, Anna Mele7, Patrizia Chiusolo8, Simona Sica8, Barbara Loteta9, Carmen Di Grazia1, Angelo Michele Carella10, Dalila Salvatore10, Enrico Morello11, Alessandro Leoni11, Luisa Giaccone2, Paolo Bernasconi12, Elisabetta Terruzzi13, Nicola Mordini14, Carlo Borghero15, Francesco Zallio16, Franco Narni17, Anna Grassi18, Attilio Olivieri19, Adriana Vacca20, Nicoletta Sacchi21, Giovannino Ciccone22, Anna Castiglione22, Emanuele Angelucci1, Francesca Bonifazi23, Fabio Ciceri24, Eliana Degrandi25, Massimo Martino9

1IRCCS Ospedale Policlinico San Martino, Genoa, Italy, 2AOU Citta’ della Salute e della Scienza di Torino, University of Torino, Torino, Italy, 3Hematology and Bone Marrow Transplant Unit AORN S. Giuseppe Moscati Avellino, Avellino, Italy, 4Ematology and BMTU, Hospital of Bolzano (SABES – ASDAA), Teaching Hospital of Paracelsus Medical University (PMU), Bolzano, Italy, 5Hematology-BMT Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, University of Milan, Milano, Italy, 6Division of Hematology, S. Maria della Misericordia Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy, 7UOC Ematologia con Trapianto, AO Car Panico Tricase (LE), Tricase, Italy, 8Sezione di Ematologia, Università Cattolica del Sacro Cuore, Fondazione Policlinico A. Gemelli IRCCS, Roma, Italy, 9Stem Cell Transplantation and Cellular Therapies Unit, Grande Ospedale Metropolitano, “Bianchi-Melacrino-Morelli”, Reggio Calabria, Italy, 10Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo (FG), Italy, 11Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia; University of Brescia, Brescia, Italy, 12Fondazione IRCCS Policlinico San Matteo, Pavia, Italy, 13Haematology Unit, San Gerardo Hospital, Monza, Italy, 14Division of hematology, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy, 15“San Bortolo” Hospital, Vicenza, Italy, 16SS Antonio and Biagio and C. Arrigo Hospital, Alessandria, Italy, 17AOU di Modena, Modena, Italy, 18Hematology-BMT Unit ASST Papa Giovanni XXIII, Bergamo, Italy, 19Clinica di Ematologia, Azienda Ospedaliero Universitaria delle Marche, Ancona, Italy, 20Businco Hospital, University of Cagliari, Cagliari, Italy, 21Italian Bone Marrow Donor Registry, E.O. Galliera Hospitals, Genova, Italy, 22SSD Epidemiologia Clinica e Valutativa Clinical Trial Center e Clinical Trial Quality Team AOU Città della Salute e della Scienza di Torino (Molinette) e CPO - Piemonte, Torino, Italy, 23Istituto di Ricerca e Cura a Carattere Scientifico Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, 24Istituto di Ricerca e Cura a Carattere Scientifico San Raffaele Scientific Institute and Vita-Salute San Raffaele University, Milano, Italy, 25Trial Office GITMO Gruppo Italiano per il Trapianto di Midollo Osseo, Cellule Staminali Emopoietiche e Terapia Cellulare, Bologna, Italy

Background: Hematopoietic cell transplantation (HCT) is curative for hematologic diseases, but outcomes are inferior when using HLA-mismatched unrelated donors (MMUD). Single HLA mismatches at HLA-A, -B, -C, or -DRB1 locus (7/8 HLA-matched) were associated with higher non-relapse mortality (NRM), and higher incidence of acute GvHD (aGVHD). Post-transplant high-dose Cyclophosphamide (PTCy) is effective in overcoming the negative impact of HLA disparity in the haploidentical setting. In the light of these results, we investigated the efficacy and safety of PTCy, with a calcineurine inhibitor (CNI) and mycophenolate (MMF), in improving clinical outcomes of MMUD transplants in patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS) by reducing aGvHD rates.

Methods: A prospective single arm phase II study (Phylos – NCT03270748 EURODRACT 2017-003530-85) on HCT from MMUD with PTCy as GVHD prophylaxis for patients with myeloid malignancies in complete remission (CR) was conducted by GITMO. The primary objective was to test whether the cumulative incidence (CI) of grade II –IV aGVHD, estimated with the Gooley method (with not GVHD related deaths as competitive events), could be reduced from 50% to 35%, with one-side alpha error of 5%. All patients were prepared for transplant with busulfan (3,2 mg/kg/day total dose 12.8, mg/kg), combined with fludarabine (40 mg/m2/day), from day −6 through day −3, and received GVHD prophylaxis with PTCy, on days +3, +4, CNI and MMF starting on day +5. The ethical committees of the participating centers approved the study. All patients signed written informed consent.

Results: Seventy-seven consecutive patients (AML: 64, MDS:13) were enrolled at 26 Italian transplant centers (January 2020 - November 2022). Median age was 53 years (range 19 – 65y). The source of hematopoietic cells was PB for 72 (94%) and BM for 5 (6%) patients respectively. Eighteen/64 (29%) of AML patients had not achieved CR after 1° induction and 25/64 (39%) had positive minimal residual disease at transplant. The 100-day cumulative incidence (CI) of grade II-IV aGVHD was 18.2% (90%CI: 11.6 – 25.9) (14 patients) (fig.1) and 6.5% (95%CI: 2.9-12.1) for grade III-IV (5 patients). Four patients died without engraftment, and one patient had autologous recovery. Seventy-three patients (95%) had full-donor chimerism with complete neutrophil engraftment by day +30. The 1-year CI of chronic GVHD (cGVHD) and moderate/severe cGVHD were 13.4% (95%CI: 6.8 - 22.1) and 9.2% (95% CI: 4.1 - 17.2) (10 and 7 patients) respectively. The 1-year CI of non-relapse mortality was 9.2% (95%CI: 4.0 -17.0), and the relapse rate was 23.8% (95%CI: 14.9-33.9) (6 and 18 patients). The 1-year overall survival and graft-relapse free survival were 78.5% (95%CI: 67.4-86) and 56.4% (95%CI: 44.4-66.7) respectively. Causes of death were sepsis (n=3), pneumonia (n=2), multi organ failure (n=2), and relapse (n=9).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (33)

Figure 1. CI of aGVHD grade II-IV in patients undergoing HSCT from MMUD for myeloid malignancies.

Conclusions: Our prospective multicenter study, in a homogeneous patient cohort, supports the hypothesis that PTCY may substantially improve clinical outcomes of MMUD transplantation. The use of the PTCy platform as GVHD prophylaxis may expand access to MMUD HCT in patients who lack HLA identical donors.

Clinical Trial Registry: Phylos – NCT03270748 EURODRACT 2017-003530-85.

Disclosure: no conflict of interest.

12: Graft-versus-host Disease – Clinical

P234 EXTRACORPOREAL PHOTOPHERESIS AS SALVAGE TREATMENT IN PEDIATRIC PATIENTS WITH STEROID-REFRACTORY ACUTE GVHD: A MULTICENTER, RETROSPECTIVE STUDY

Daria Pagliara1, Valentina Sofia1, Claudia Del Fante2, Manuela Tumino3, Francesco Saglio4, Giovanna Leone1, Gianluca Viarengo2, Mattia Algeri 1, Giovanna Giorgiani2, Giovanna Del Principe1, Barbarella Lucarelli1, Pietro Merli1, Anna Colpo3, Cesare Perotti2, Marco Zecca2, Alessandra Biffi3, Franca Fagioli4, Franco Locatelli1,5

1Bambino Gesù Children Hospital IRCCS, Roma, Italy, 2Foundation IRCCS Polyclinic San Matteo, Pavia, Italy, 3Padua University Hospital, Padova, Italy, 4Regina Margherita Children’s Hospital, Torino, Italy, 5Catholic University of the Sacred Heart, Roma, Italy

Background: Extracorporeal photopheresis (ECP) is a treatment option for steroid-refractory acute graft-versus-host disease (SR-aGvHD); it has an immunomodulatory effect through the reinfusion of autologous mononuclear cells after exposure to 8-methoxypsoralen and ultraviolet A light irradiation.

Methods: We performed a retrospective, multicenter study recruiting pediatric patients in 4 Italian medical centers, aimed at investigating the efficacy of ECP for the treatment of SR-aGvHD. Subjects had undergone hematopoietic stem cell transplantation (HSCT) from any donor type and with the employment of any cell source. All consecutive patients treated with ECP for SR-aGvHD were included and data on indication to HSCT, disease status, conditioning, GvHD prophylaxis and treatment, GVHD severity, ECP administration, treatment response and outcome were collected and analyzed.

Results: Between 01/2013 and 06/2023, a total of 135 pediatric patients developed SR-aGvHD and were treated with ECP. Patients and transplant characteristics are reported in table 1.

At the time of ECP start, 66.7% of patients had aGvHD grade I-II, while 33.3% presented aGvHD grade III-IV. About 50% of patients had only skin involvement, while 46.7% had both skin and visceral involvement. Median mononuclear cell count (MNC) at ECP start was 1830/mmc. ECP was started after a median of 12 days (range 0-194) after the onset of GvHD. ECP treatment regimen was included 3 procedure/week for 88 out of 135 patients and 2 procedure/week for the remaining 47 patients. The procedure was safe, being performed also in 16 children below 3 years of age.

Best overall response (BOR) at any time, defined as the proportion of patients who achieved either complete response (CR) or partial response (PR), was 80%, including 71.1% CR, with no difference between skin-only (42.2%) and visceral involvement (37.8%) (p=ns). No association was noted between the number of previous lines of treatment after failure of steroids and response to ECP. Additionally, we did not observe any difference in response rate to ECP based on the treatment schedule (2/ week vs 3/week) (p=ns). Conversely, MNC at start of ECP was associated with response rate, with 71% of patients in the lower quartile (MNC <1200/mcl) achieving CR/PR as opposed to 83% of patients with higher MNC (p=0.01). Median time from start of ECP to BOR and to start of steroid tapering were 25 days (range 7-100) and 10 days (range 1-92), respectively. Three-year event-free survival for patients achieving CR/PR was higher than that of those not responding to ECP (71.4% vs 33.1%) (Figure 1).

Finally, 29% of patients achieving CR/PR with ECP subsequently developed chronic GvHD, as compared to 59% who did not respond to ECP (p<0.001).

Table 1 - Patients’ characteristics

Age at TCSE (median – range)

8.8

0.9-21.9

Gender

M (63.7%), F (36.3%)

Diagnosis

Malignant (80%)

ALL (45.9%)

AML (20.7%)

Other (13.4%)

Non-malignant (20%)

Beta-thalassemia (5.2%)

AA 1 (3.7%)

Fanconi anemia (2.2%)

Other (8.9%)

Donor type

MFD (11.8%)

MUD (54%)

haploidentical (34%)

Conditioning regimen

MAC (93.3%)/RIC (6.7%)

TBI-based (43.7%)/chemo-based (56.3%)

GvHD prophylaxis

CSA (6.7%)

CSA + MTX + /-ATG (51.8%)

ATG + T-cell depletion (29.5%)

Other (12%)

Time of GvHD onset after HSCT

31.5 days

Grade of aGvHD at onset

aGvHD grade I (20%)

aGvHD grade II (65.2%)

aGvHD grade III (11.9%)

aGvHD grade IV (2.2%)

Grade of aGvHD at time of ECP start

aGvHD grade I (9.6%)

aGvHD grade II (60.7%)

aGvHD grade III (26.7%)

aGvHD grade IV (3%)

Organs involved

skin (48.9%)

GI (2.9%)

liver (1.5%)

skin + visceral (46.7%)

Treatment line when ECP started

2 (35.5%)

3 (54.1%)

4 (7.4%)

5 (2.2%)

GvHD as cause of death

8.1%

.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (34)

Image 1.

Conclusions: This study, including the largest cohort of pediatric patients treated with ECP, showed good results in a poor-prognosis population together with a good safety profile. ECP is a valid treatment option in patients with SR-aGvHD in children and can increase the probability of survival, reduce exposure to corticosteroids and lower the risk of subsequent development of chronic GvHD.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P235 BELUMOSUDIL PHASE 1 DOSE-ESCALATION STUDY IN HEALTHY VOLUNTEERS AND PHASE 2 STUDY IN PATIENTS WITH CGVHD WHO FAILED ≥1 LINE OF SYSTEMIC THERAPY IN CHINA

Ying Wang 1, Depei Wu1, Xiang Zhang1, Yuhua Li2, Yanjie He2, Qifa Liu3, Li Xuan3, Zhenyu Li4, Kunming Qi4, Yuqian Sun5, Shunqing Wang6, Wenjian Mo6, Lei Gao7, Ye Hua8, Yu Wang8, Ying Zhang8, Nico Fu9

1The First Affiliated Hospital of Soochow University, Suzhou, China, 2Zhujiang Hospital, Southern Medical University, Guangzhou, China, 3Nanfang Hospital, Guangzhou, Guangdong, China, 4The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China, 5Peking University People’s Hospital, Beijing, China, 6Guangzhou First People’s Hospital, Guangzhou, China, 7Xinqiao Hospital of Army Medical University, Chongqing, China, 8BioNova Pharmaceuticals (Shanghai) Limited, Shanghai, China, 9Sanofi Medical Division, Shanghai, China

Background: Belumosudil is a first-in-class Rho-associated coiled-coil containing protein kinase 2 (ROCK2) inhibitor. ROCK2 is associated with chronic graft-versus-host disease (cGVHD)-mediated proinflammatory response and fibrosis. Belumosudil has been demonstrated to be an effective treatment for steroid refractory cGVHD, mainly in non-Asian patients (aged≥12 years) in 2 phase 2 studies. It is now crucial to perceive the safe dose of belumosudil and investigate its overall benefit–risk balance for cGVHD in Chinese population.

Methods: A phase 1, single-center, randomized (9:3), double-blind, placebo-controlled, dose-escalation study was designed to assess the tolerability, safety, and pharmacokinetics (PK) of single-dose belumosudil (200 mg/400 mg) in healthy volunteers (N=24) in China. PK parameters were calculated based on WinNonlin 6.4, and the other statistical analyses were performed based on SAS® 9.4. All treatment-emergent adverse events (TEAEs) and drug-related TEAEs were also evaluated. Based on this phase 1 study results, a multicenter, open-label phase 2 study (NCT04930562) was conducted to evaluate safety, efficacy, and PK of oral belumosudil 200 mg QD in cGVHD patients (N=30) who received ≥1 line of systemic therapy in China. Primary endpoint: overall response rate (ORR); secondary endpoints: duration of response (DOR), time-to-response (TTR), changes in Lee Symptom Scale (LSS) score, organ response rate, corticosteroid and CNI dose change, failure-free survival (FFS), time-to-next-treatment (TTNT), overall survival (OS), and safety.

Results: In the phase 1 study, belumosudil was absorbed rapidly and peaked at 1.5 hours (range, 0.5–2.0 hours). Median T1/2 for 200 mg and 400 mg dose cohorts was 10.35 hours and 8.92 hours, respectively. Mean AUC0-24 for 200 mg and 400 mg dose cohorts was 9040.44 ng*h/mL and 9984.00 ng*h/mL, respectively. Overall, belumosudil (single dose of 200 mg and 400 mg) was well tolerated, and no unexpected safety events were observed. A total of 17.4% of participants reported TEAEs; 2 participants in 400 mg dose group experienced drug related TEAEs (BMI decreased and blood triglycerides increased). In the phase 2 study, after first administration of 200 mg belumosudil, median Tmax was 2.5 hours (range, 1.4–5.8); mean T1/2 was 3.8 hours; mean AUC0-t was 22347.5 h*ng/mL; mean AUC0-inf was 22844.5 h*ng/mL. ORR of 73.3% (95% CI: 54.1–87.7) was reached in a median follow-up of 12.9 months. High ORR was maintained in all subgroups regardless of cGVHD severity, number of organs involved, prior number of lines of therapy, or prior ibrutinib and ruxolitinib treatment. Median DOR, FFS, OS and TTNT were not reached; TTR was 4.3 weeks (range, 3.9–48.1). Overall, 50.0% of patients achieved clinically meaningful response in terms of reduction in LSS score by ≥7 points from baseline. Corticosteroid and CNI dose reductions were reported in 56.7% and 35.0% of patients, respectively. Most TEAEs were mild to moderate in severity; 36.7% patients developed grade≥3 TEAEs and 13.3% experienced ≥1 grade≥3 drug related TEAE.

Conclusions: Single dose belumosudil is safe and well-tolerated in healthy Chinese population, with a PK profile similar to global studies. Belumosudil also demonstrated a favorable benefit–risk balance in treating cGVHD patients who received standard corticosteroid therapy in China.

Clinical Trial Registry: ClinicalTrials.gov NCT04930562.

Disclosure: Ying Wang: Nothing to declare. Depei Wu: Nothing to declare. Xiang Zhang: Nothing to declare. Yuhua Li: Nothing to declare. Yanjie He: Nothing to declare. Qifa Liu: Nothing to declare. Li Xuan: Nothing to declare. Zhenyu Li: Nothing to declare. Kunming Qi: Nothing to declare. Yuqian Sun: Nothing to declare. Shunqing Wang: Nothing to declare. Wenjian Mo: Nothing to declare. Lei Gao: Nothing to declare. Ye Hua: Grants or contracts: BioNova Pharmaceuticals Ltd; Stock or stock options: BioNova Pharmaceuticals Ltd. Yu Wang: Employee of BioNova Shanghai Ltd. which was authorized for clinical development and commercialization of Belumosudil in China. Ying Zhang: Nothing to declare. Nico Fu: Employee of Sanofi..

12: Graft-versus-host Disease – Clinical

P236 IMPACT OF RESPIRATORY FUNCTION TESTS IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Francisco Manuel Martin Dominguez 1, Rocío Parody-Porras1, Virginia Escamilla-Gómez1, Nancy Rodríguez-Torres1, Cristina Blázquez-Goñi1, Javier Delgado-Serrano1, Juan Luis Reguera-Ortega1, Ildefonso Espigado-Tocino1, José Antonio Rodríguez-Portal2, José Antonio Pérez-Simón1

1University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS / CISC), Seville, Spain, 2Hospital Virgen del Rocío, CIBER de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III, Seville, Spain

Background: Pre-transplant respiratory function tests (RFTs) are one of the most useful tools for determining the risk of transplant-related mortality being included in pretransplant score risk index. By contrast, the predictive value of the spirometric parameters after transplant on the occurrence of pulmonary complications and particularly GvHD has not been extensively explored.

Methods: Single-center retrospective study of adult patients (≥18 years), consecutively undergoing Allogeneic Hematopoietic Stem Cell Transplantation (HSCT) between January 2011 and December 2020.

Spirometric values were collected pre-transplant (FEV1, FEV1/FVC and DLCO), and at 3-, 6- and 12-months post-transplant.

The analyses were performed using R Core Team (2021), Kaplan Meier curves were performed for overall survival, as well as for cGVHD events and Complications, where the curves were compared using log rank or Gehan-Breslow for the different post-transplant spirometric parameters. Statistical significance has been established for p-value <0.05.

Results: A total of 471 patients were included, with a median age of 49 years. The most frequent diagnosis was AML/MDS (50.4%). The most common type of donor was matched unrelated donor (MUD) (33.5%). 71% of patients received reduced-intensity conditioning. The results were analyzed based on three variables:

  1. 1.

    Median overall survival (OS) was 58 weeks. DLCO ≤ 65% was an independent risk factor for mortality at any time (OS among patients with pre-transplant DLCO ≤ 65% 46.5% at 5 years vs 63.5% among those with DLCO > 65%; p=0.02) and at any of the three post-transplant measurement points (OS among patients with DLCO >/≤ 65% at 3 months postrasplant 83.5% vs 51.5% at 5 years, p=0.03; for those with DLCO >/≤ 65% at 6 months 62.5% vs 41.5%, p=0.01 and for those with DLCO >/≤ 65% at 12 months 54.5% vs 34.3%, p=0.001); FEV1 ≤ 65% and FEV1/FVC ≤ 65% were also associated with a significantly higher mortality both pre-transplant and at 12 months post-transplant.

  2. 2.

    Median onset of cGvHD was 45 weeks. Patients with a DLCO ≤ 65% at 6 months had a cumulative incidence of overall cGvHD of 40% as compared to 3.6% for those with DLCO > 65% (p=0.001); similarly, the incidence of overall cGvHD at 12 months was 5.6% vs 43.8% (p=0.001) for patients with DLCO >/≤ 65%.

  3. 3.

    At 48 weeks of follow-up, mortality due to to pulmonary-related (PRM) complications was 15%. Patients with FEV1 ≤/> 65% at 6 moths had a PRM of 33.5% vs 9.8% (p=0.02), those with FEV1/FVC ≤/> 65% at 6 moths had a PRM of 19.6% vs 1.8% (p=0.028) and those with DLCO ≤/> 65% 22.5% vs 2.3% (p=0.037).

Conclusions:

  1. 1.

    DLCO impairment is an independent risk factor for mortality at any time during transplantation (both pre-transplant and post-transplant); in addition, FEV1 and FEV1/FVC are associated with higher mortality both pre-transplant and 12-month post-transplant.

  2. 2.

    A DLCO ≤ 65% at 6 and 12 months identifies at patients with a significantly higher risk of cGVHD.

  3. 3.

    With 48 months follow-up, 15% of deaths were due to pulmonary related complications. Alteration of FEV1, FEV1/FVC, and DLCO ≤ 65% at six months post-transplant were associated with an increased risk of pulmonary related deaths.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P237 MANAGEMENT OF CORTICOSTEROIDS IN PATIENTS RECEIVING RUXOLITINIB TREATMENT FOR CHRONIC GRAFT-VERSUS-HOST DISEASE: A POST HOC ANALYSIS FROM THE RANDOMIZED PHASE 3 REACH3 STUDY

Emily Kintsch1, Zhenyi Xue1, John Galvin1, Franco Locatelli2,3, Robert Zeiser4, Valkal Bhatt 1

1Incyte Corporation, Wilmington, United States, 2IRCCS Ospedale “Bambino Gesù, Rome, Italy, 3Catholic University of the Sacred Heart, Rome, Italy, 4University Medical Center Freiburg, Freiburg, Germany

Background: Ruxolitinib (RUX) is an oral, selective Janus kinase (JAK)1/JAK2 inhibitor approved for steroid-refractory chronic graft-versus-host disease (cGVHD) in patients aged ≥12 years after failure of 1‒2 lines of systemic therapy. Management of corticosteroid use in patients treated in the REACH3 study warrants further discussion.

Methods: This post hoc analysis examined corticosteroid dosing in patients with steroid-dependent (STD; steroid dose at enrollment <1.0 mg/kg/day) and steroid-progressive (STP; steroid dose at enrollment ≥1.0 mg/kg/day) cGVHD who received RUX or best available therapy (BAT) in the randomized phase 3 REACH3 study. These definitions for STD and STP were chosen to describe and define steroid burden at enrollment. Responders were patients who achieved overall response (complete response [CR] + partial response [PR]) without the requirement of additional systemic therapies for an earlier progression, mixed response, or non-response. CR and PR were defined per the 2014 National Institutes of Health consensus criteria. Mean cumulative steroid dose was the sum of mean steroid dose for each time interval up to Day 180 (D180).

Results: Of the 311 patients included in this post hoc analysis, 159 received RUX (STD, n=131 [82.4%]; STP, n=28 [17.6%]), and 152 received BAT (STD, n=132 [86.8%]; STP, n=20 [13.2%]). Mean (SD) age was 47.5 (15.4) years for STD patients (47.0 [15.2] for RUX; 48.0 [15.6] for BAT) and 43.7 (18.2) years for STP patients (44.3 [17.1] for RUX; 43.0 [20.1] for BAT. Cumulative mean (IQR) steroid dose in RUX vs BAT at D180 from the original REACH3 study safety population was 46.7 mg/kg (13.8‒64.6) for RUX and 54.1 mg/kg (17.8‒79.1) for BAT (Table). Responders from the original study safety population (RUX, 49.7%; BAT, 26.6%) also had lower cumulative steroid dose (RUX, 41.5 mg/kg; BAT, 39.5 mg/kg) compared with non-responders (RUX, 53.4 mg/kg; BAT, 60.2 mg/kg; Table). In this post hoc analysis, RUX STD patients had lower cumulative steroid dose compared with BAT STD patients (41.4 mg/kg vs 48.2 mg/kg, respectively). Similarly, cumulative steroid doses in the STP subgroups were 72.0 mg/kg for RUX and 95.0 mg/kg for BAT (Table). As expected, STD patients receiving RUX or BAT had lower cumulative steroid doses compared with their STP counterparts (Table). Mean steroid dose decreased over time in STD patients who received RUX or BAT, with RUX and BAT responders having lower mean steroid dose than non-responders. Mean steroid dose was reduced over time in STP patients who received RUX or BAT, with RUX responders having the lowest dose by D169‒179.

Table 1. Cumulative Steroid Dose at Day 180

RUX

BAT

N=165*

N=158*

Cumulative steroid dose, mean (IQR), mg/kg

46.7 (13.8‒64.6)

54.1 (17.8‒79.1)

Responder

Non-responder

Responder

Non-responder

n=82*

n=83*

n=42*

n=116*

41.5 (14.4‒60.14)

53.4 (13.7‒70.4)

39.5 (15.2‒57.2)

60.2 (19.9‒87.8)

Steroid Dependent

Steroid Progressive

Steroid Dependent

Steroid Progressive

n=131

n=28

n=132

n=20

41.4 (13.1‒54.1)

72.0 (42.1‒93.1)

48.2 (16.8‒69.2)

95.0 (51.8‒121.8)

  1. * Patients from original REACH3 study safety population. Mean (IQR) of cumulative steroid dose at Day 180 is calculated as the sum of the mean (IQR) cumulative steroid dose at each time interval based on observed values. Patients from this post hoc analysis.

Conclusions: RUX treatment for cGVHD resulted in lower cumulative mean steroid doses compared with BAT at D180 in the REACH3 study. This was consistent in patients who were STD or STP. STD and STP RUX responders had lower mean steroid doses over time than non-responders. Among STP patients, mean steroid dose decreased more over time for RUX responders than for BAT responders. These data suggest that RUX treatment can help reduce long-term steroid exposure in patients with cGVHD.

Clinical Trial Registry: NCT03112603.

https://classic.clinicaltrials.gov/ct2/show/NCT03112603.

Disclosure: EK is a fellow at Incyte Corporation and is employed by Saint Joseph’s University.

ZX, JG, and VB, are employees and stockholders of Incyte.

FL received honoraria from Novartis, Amgen, and Miltenyi.

RZ received honoraria from Novartis, Incyte Corporation, Sanofi, and Mallinckrodt.

12: Graft-versus-host Disease – Clinical

P238 A PROGNOSTIC MODEL FOR NON-RELAPSE MORTALITY RISK BASED ON MACHINE LEARNING IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM-CELL TRANSPLANTATION

Jingjing Yang1, Xiawei Zhang 1, Liping Dou1, Daihong Liu1

1The Fifth Medical Centre, Chinese PLA General Hospital, Beijing, China

Background: Prognostic biomarkers are widely used to assess non-relapse mortality (NRM) in patients after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, the current models are based on the concentrations of biomarkers measured at fixed time points and cannot achieve dynamic monitoring of NRM risk. This limited their utility as a reference for treatment to guide individualized therapy.

Methods: The prospective, multicenter study was registered at ClinicalTrials.gov (NCT04284904) and comprised 621 patients who received allo-HSCT between August 2018 and February 2022 from 7 Chinese transplant centers. The concentrations of five biomarkers (ST2, REG3α, TNFR1, IL6, and IL8) were prospectively measured at day 7/14/28/60/90 post-HSCT. Patients with acute GVHD were monitored at onset and weekly for the concentration of biomarkers during the first month of treatment. A dynamical monitoring model was developed based on machine learning to predict the 6-month NRM after allo-HSCT and then validated in an independent multicenter cohort.

Results: The model had a high AUC for 6-month NRM (0.86 for the training set, 0.86 for the testing set, and 0.94 for the validation set). Based on model scores, the model identified 11.66% (54/463) of patients in the high-risk (HR) group, 22.03% (102/463) in the moderate-risk (MR) group, and 66.31% (307/463) in the low-risk (LR) group. In the validation cohort, there were 18.35% of patients with HR, 24.68% of patients with MR, and 66.31% of patients with LR. The model identified high-risk patients with the highest 6-month NRM (HR vs. MR vs. LR, 34.75% vs. 9.98% vs. 1.30%, P < 0.001). The algorithm performed equally well in the validation cohort (6-month NRM: HR vs. MR vs. LR, 41.38% vs.2.56% vs. 0.00%, P < 0.001). Acute GVHD severity and model score were significant predictors of NRM in multivariate analysis (model score, P < 0.0001; acute GVHD grade, P=0.0014). During dynamic monitoring, the model consistently identified patients with high-risk NRM across various post-HSCT time points. For acute GVHD patients with treatment, the model successfully separated patients into groups with different NRM.

Conclusions: We established a dynamical monitoring model for predicting NRM in the patients after allo-HSCT, which held the potential to identify high-risk individuals and improve individual treatment.

Clinical Trial Registry: We established a dynamical monitoring model for predicting NRM in the patients after allo-HSCT, which held the potential to identify high-risk individuals and improve individual treatment.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P239 A HIGH FREQUENCY OF TYPE 3 DENDRITIC CELLS IN CD1C + DC ON DAY + 28 AFTER HSCT COULD BE AN EFFECTIVE INDICATOR OF SEVERE GVHD IN PEDIATRIC PATIENTS

Xin Liu 1, Bohan Li1, Di Yao1, Minyuan Liu1, Peifang Xiao1, Jun Lu1, Jie Li1, Yuanyuan Tian1, Shaoyan Hu1

1Children’s Hospital of Soochow University, Suzhou, China

Background: Graft-versus-host disease (GVHD) resulted in a significant comorbidity and was a leading cause of death following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Although the direct damage to GVHD target organs is caused by T cells, dendritic cells effectively trigger T cell activation. These dendritic cells play a key role in the induction, regulation and maintenance of GVHD. Previous research has indicated that type 3 dendritic cells (DC3) constitute a distinct class of dendritic cells that notably promote inflammation and accumulate abnormally in autoimmune diseases such as Systemic Lupus Erythematosus (SLE). However, limited research hasn’t explored the association between DC3s and GVHD.

Methods: A total of 63 patients underwent allo-HSCT at the Children’s Hospital of Soochow University and were eligible for this study. Peripheral blood samples were collected from the children after allo-HSCT, and DCs subsets were analyzed by flow cytometry. We compared the proportion and number of DCs subsets in children with no GVHD (grade 0) or mild GVHD (grade I) to those with severe GVHD (grades II-IV).

Results: In the study, patients developing grade II-IV GVHD within 28 days after HSCT were found to have a lower frequency and number of pDCs (median frequency, 0.0069%; range, 6.33E-05~0.1793%, median number, 0.0914cell/μl, range, 0.0018~1.8492 cell/μl) than patients with grade 0-I GVHD (median frequency, 0.0626%, range, 0.0007%~0.6980%, P=0.0002; mediate number, 0.3101 cell/μl, range, 0.0138~3.4893 cell /μl, P=0.0049). Patients who developed grade II-IV aGVHD showed significant difference in the frequency (median, 0.0221%; range, 0.0023~0.2837%) of CD1c+DCs compared to patients with grade 0-I GVHD (median frequency, 0.1228%; range, 0.0018~0.7272%, P=0.0067). But there was no significant difference in the number between the two groups. Patients with grade II-IV GVHD showed a significantly higher frequency of DC3s in CD1c+DCs (median frequency, 26.8%; range, 2.33~70.7%) than patients with grade 0-I GVHD (median frequency, 13.3%, range, 1.17%~53.6%, p=0.0003). These data suggest that the frequency of DC3s in CD1c+DCs is linked to the occurrence of severe GVHD.

We used Receiver Operating Characteristic (ROC) curve analysis to explore thresholds of DC3s as a discriminator between grade II-IV GVHD patients and grade 0-I GVHD patients. The median DC3s level in all tested patients was 18.5% (range, 1.17~70.7% in CD1c+DCs) on day +28 after HSCT. We chose the threshold of DCs level at 24.50%(in CD1c+DCs) to evaluate the difference in patients with high (above the threshold) versus low (below the threshold) DC3 levels. Patients in the high DC3 group(>24.50% in CD1c+DCs) had a significantly greater risk of developing grade II-IV GVHD compared with patients in the low DC3 group (p<0.0001).

Conclusions: We found that a high frequency of DC3 in CD1c+DC on day +28 after HSCT was significantly associated with severe GVHD. A higher frequency of DC3 in CD1c+DC seems to be accompanied by more severe GVHD. Therefore, regular examinations of DCs, particularly DC3s, after transplantation are of great significance for predicting GVHD.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P240 HAPLOIDENTICAL HEMATOPOIETIC CELL TRANSPLANTATION WITH UNRELATED CORD BLOOD ENHANCES SURVIVAL AND REDUCES THE INCIDENCE OF AGVHD FOR PEDIATRIC ACUTE MYELOID LEUKEMIA

Fei Pan1, Jing Long2, Guanlan Yue1, Kang Gao1, Kun Wang1, Huili Zhu1, Yujie Wang1, Zheren Wang1, Yuan Feng1, Lili Shi1, Zhijie Wei 1, Kaiyan Liu2

1Hebei Yanda Lu Daopei Hospital, Hebei, China, 2Beijing Lu Daopei Hospital, Beijing, China

Background: Haploidentical hematopoietic cell transplantation (haplo-HCT) is a curative treatment for pediatric acute myeloid leukemia (AML). Haploidentical transplantation, potent in its strong graft-versus-leukemia (GVL) effect, however, still faces major failure due to the severe acute graft-versus-host disease (GVHD). At the same time, GVHD is also an important factor affecting the long-term prognosis and quality of life of patients. Therefore, how to reduce the incidence of GVHD and improve long-term survival after haplo-HCT are very important for pediatric AML. Addressing this, our study pivots on whether integrating unrelated cord blood into haplo-HCT can optimize pediatric AML patient survival and mitigate GVHD incidence.

Methods: Between April 2014 and December 2020, 42 pediatric (aged ≤14 years) AML patients at the first complete remission (CR1) who underwent haploidentical transplantation in our hospital were enrolled in our study. All received Bu / CY-based myeloablative conditioning regimens. CsA + MMF + sMTX + ATG were administrated for GVHD prophylaxis. For the haplo-HCT with unrelated core blood group (haplo-cord HCT), 24 patients were combined with 4 / 6 matched HLA unrelated core blood for transfusion, with the number of TNC infusion at 0.5 * 107/kg. For the control group, 18 cases received haplo-HCT without cord blood infusions. Baseline characteristics of the two groups are detailed in Table 1.

Results: As of November 20, 2023, the median follow-up time was 3.7 (0.3-9.7) years. There were no drug-toxicity-related deaths during the preconditioning regimens. After complete stem cell engraftment in all patients, the median days of neutrophil cell implantation was 14 (10-19) days in the control group versus 13 (10-22) days in the haplo-cord HCT group, and the median days of platelet implantation was 9 (6-16) days in the control group versus 9 (6-38) days in the haplo-cord HCT group. The haplo-cord HCT group exhibited a remarkably higher overall survival (OS) than the control group with the OS of 95.8% vs. 66.7% (P <0.05). No statistical differences were observed in relapse rate between the haplo-cord HCT and control groups (0% vs. 20%, P> 0.05). The haplo-cord HCT group experienced a significantly lower incidence of grade III-IV acute GVHD (aGVHD) compared to the control group (8.3% vs. 33.3%, P <0.05). But no significant difference was seen in the incidence of grade II-IV aGVHD (12.5% vs. 33.3%, P> 0.05).

Conclusions: Our study demonstrated that for pediatric AML patients, the haplo-HCT with unrelated cord blood could significantly improve the long-term survival, and also significantly reduce the incidence of severe aGVHD compared to haplo-HCT without unrelated cord blood. However, Future research, preferably through multi-center, randomized trials, is essential to solidify these findings and further refine this promising treatment pathway.

Disclosure: No relevant conflicts of interest to declare.

12: Graft-versus-host Disease – Clinical

P241 RUXOLITINIB AND ANTI-TNF-ALPHA THERAPY SUBSTANTIALLY IMPROVE SURVIVAL IN PEDIATRIC PATIENTS WITH SEVERE STEROID REFRACTORY GVHD

Jaspar Kloehn1, Manon Queudeville1, Johanna Schrum1, Ysabel Alessa Schwietzer1, Ingo Müller 1

1Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: Steroid refractory graft versus host disease (SR GvHD) remains a major cause of morbidity and mortality after HSCT. The REACH4 trial confirmed the promising results seen for ruxolitinib in adult SR aGvHD for pediatric patients, but the trial only included 12 patients (26.7%) with grade III and 4 patients (8.9%) with grade IV aGvHD. To further add evidence for ruxolitinib and anti-TNFalpha treatment in the management of SR GvHD, we report on our cohort of patients with severe SR GvHD during the last 10 years.

Methods: Retrospective single center analysis over 10.5 years (January 2013 to June 2023) at our institution. During the observation period, 370 allogeneic stem cell transplantations were performed in children. Twenty-nine patients (7.8%) developed SR severe GvHD according to the MAGIC (grades III or IV aGvHD) or modified NIH criteria (severe cGvHD), respectively. The median follow-up is 40 months (range 5 to 124 months).

Results: Median age at HSCT was 11 years (range 0 – 16 years). Underlying diseases consisted of hematological malignancies (31%), hemoglobinopathies (38%), inborn errors of metabolism (7%), inborn errors of immunity (21%) and bone marrow failure (3%). Fourteen patients (48%) received 10/10-HLA-matched unrelated grafts, nine patients (31%) received 9/10-matched unrelated grafts, five patients (17%) received HLA-identical related grafts and one patient (3%) received a haploidentical related graft. Twenty-nine patients (7.8%) developed severe SR GvHD: 23 patients developed acute GvHD (54%) and 20 patients had chronic GvHD (46%) (see table). Patients with SR aGvHD almost all suffered lower gastrointestinal involvement, among patients with SR cGvHD, skin was the commonly affected organ. All patients with SR GvHD received second line treatment with anti-TNFalpha antibodies (either infliximab or adalimumab), ruxolitinib or a combination treatment of anti-TNFalpha and JAK inhibition. The overall response rate (ORR) at day 28 of treatment was 65.5 %, with a complete response (CR) rate of 27.6 %. Best ORR at any given time point was as high as 89.7% with CR at any given time point at 69%. Importantly, despite the severity of SR GvHD, the majority of patients (58.6%) is disease-free and off treatment. Two patients (6.9%) are currently disease-free while still on treatment, five patients (17%) still suffer from GvHD, one patient died from leukemia relapse and four patients (13.7%) died due to treatment-related mortality (TRM). Causes of death were infections in two cases and GvHD and the other two.

GvHD classification

Number of patients (%)

SR aGvHD

16 (55.2)

Grade 3

7 (43.8)

Grade 4

9 (56.3)

SR aGvHD – organ involvement

Skin

9 (56.3)

Gastrointestinal tract

15 (93.8)

Liver

3 (18.8)

SR cGvHD

13 (44.8)

severe

13 (100)

SR cGvHD – organ involvement

Skin

11 (84.6)

Joints and fasciae

9 (69.2)

Gastrointestinal tract

6 (46.2)

Liver

4 (30.8)

Eyes

7 (53.8)

Lung

5 (38.5)

Conclusions: SR GvHD is a dreaded complication after HSCT with reported survival rates as low as 35% at 2 years and TRM-rates of above 50% in children. Treatment with anti-TNFalpha antibodies and ruxolitinib led to high ORR and CR rates and only four fatalities (13.7%) related to SR GvHD. Both, ruxolitinib and anti-TNFalpha treatment, as backbone in combination therapy, e. g. with steroids, calcineurin inhibitors and extracorporeal photopheresis, led to high response rates significantly reducing TRM. Results from prospective trials already demonstrated the efficacy of ruxolitinib in the setting of SR GvHD. The concomitant use of anti-TNFalpha treatment warrants further investigation.

Disclosure: Müller: Novartis (speaker honorary), Neovii (speaker honorary).

12: Graft-versus-host Disease – Clinical

P242 IMMUNOPHENOTYPE OF EXTRACELLULAR VESICLES AS A BIOMARKER OF CHRONIC GRAFT-VERSUS-HOST-DISEASE: A PILOT STUDY

Giuseppe Lia1,2, Federica Ferrando1,1, Michele Dicataldo1,2, Andrea Evangelista3, Alessia Cargnino1,2, Irene Dogliotti1,2, Stefania Bruno4, Maria Chiara Deregibus4, Aurora Martin1,2, Jessica Gill1,2, Chiara Maria Dellacasa5, Alessandro Busca5, Giovanni Camussi4, Benedetto Bruno1,2, Luisa Giaccone 1,2

1University of Torino, Torino, Italy, 2A.O.U. Città della Salute e della Scienza di Torino, Stem Cell Transplant Program, Torino, Italy, 3A.O.U. Città della Salute e della Scienza di Torino, Torino, Italy, 4University of Torino, Molecular Biotechnology Center, Torino, Italy, 5University Hospital A.O.U. “Città della Salute e della Scienza di Torino”, Division of Hematology, Torino, Italy

Background: Morbidity and mortality related to chronic Graft-vs.-Host Disease (cGVHD) still represent a major concern of allogeneic hematopoietic stem cell transplantation (SCT). Thus, new diagnostic and therapeutic biomarkers are needed to tailor the use of immunosuppressive drugs and to optimize cGVHD prevention and treatment. One potential attractive biomarker may be represented by Extracellular Vesicles (EVs): exosomes (EXs), microvesicles (MVs), and, in particular, EVs originating from T-Helper follicular or Dendritic/Monocytes cells (THDC), important players in cGVHD pathogenesis. In this study, we investigated the potential role of EV immunophenotype as a biomarker of cGVHD.

Methods: Twelve patients who underwent a SCT were selected for this study, 6 of them developed moderate (4) or severe (2) cGVHD, whereas 6 did not. None of them had previous acute GVHD. Serum samples were collected at the same time-points: (basal, before and after cGVHD onset or at similar time points in patients without). EVs were extracted using a protamine-based precipitation method. The expression of 37 EV surface biomarkers was characterized by a standardized flow-cytometry capture bead-based assay. Biomarkers expression was measured in total EXs and MVs, and in EVs from THDC. The risk of developing cGVHD and biomarker predictivity (AUROCs) was evaluated by logistic regression models. Odds Ratios (ORs) were estimated as absolute levels (abs) and as proportional changes (Δ) compared with basal levels. All statistical analyses were performed using STATA 15 and SPSS Statistics 25.

Results: Several of the 37 total EV surface and THDC derived EV biomarkers came out potentially associated to cGVHD developments, many of them are associated with adaptive and innate immunity dysregulation, which is known to be involved in the development of cGVHD. Among innate immune cells, alteration of scavenger receptor expression (such as CD204 and CD209) in Dendritic cells and Macrophages, and increased expression of CD326 in antigen-presenting cells have been linked to inflammation, tissue repair, and fibrosis in both acute and chronic GVHD. At basal timepoints, the fluorescence levels of CD142, CD209 and CD326 in EX from THDC showed the highest diagnostic performance (OR-p-AUROC: 23.5-0.027-0.944, >100-0.004-0.986, and 58.3-0.002-1, respectively). Furthermore, 3 EV biomarkers appeared to be higly predictive (AUROC >.8) before cGVHD onset: abs level of CD209 and reduced Δlevel of CD19 in EXs from THDC, and reduced Δlevel of CD146 in MVs from THDC (OR 16.91 p 0.048, OR 0.924 p 0.043, and OR 0.612 p 0.003, respectively). Interestingly, although with the limit of small sample size, the emerging biomarkers appeared to decrease at cGVHD resolution.

Conclusions: This pilot study, suggested some EV surface markers, which are warrant of further investigation in the setting of cGVHD A prospective study has been designed to further confirm these findings.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P243 T-CELL DEPLETED ALLOGENEIC STEM CELL TRANSPLANTATION WITH ALEMTUZUMAB IS A VIABLE ALTERNATIVE TO POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR PATIENTS WITH ACUTE MYELOID LEUKEMIA AND MYELODYSPLASTIC SYNDROME

Alexandra Rojek 1, Mylove Mortel1, Andrew Artz2, Richard Larson1, Hongtao Liu3, Mariam Nawas1, Adam Duvall1, Toyosi Odenike1, Gregory Roloff1, Wendy Stock1, Michael Bishop1, Satyajit Kosuri1

1University of Chicago, Chicago, United States, 2City of Hope National Medical Center, Duarte, United States, 3University of Wisconsin, Madison, Madison, United States

Background: Allogeneic stem cell transplantation (alloSCT) is often the only curative treatment option for patients with acute myeloid leukemia (AML) or myelodysplastic syndrome (MDS). Modifications to conditioning regimens and graft-versus-host disease (GVHD) prophylaxis are often needed to make alloSCT a safer and more tolerable procedure, even when a fully matched graft is available. Post-transplant cyclophosphamide (PTCy) has emerged as an appealing GVHD prophylaxis standard for patients with well-matched donors, but has not been compared to all available standard T-cell depletion strategies. We evaluated outcomes of propensity-score matched patients who received T-cell depleted alloSCT (TCD-SCT) with alemtuzumab at our center with patients receiving PTCy in a publicly available dataset from the Center for International Blood and Marrow Transplant Research (CIBMTR).

Methods: We retrospectively analyzed 58 patients with AML or MDS who received in vivo TCD-SCT with alemtuzumab at the University of Chicago from 2014-2022. All patients received a fully matched related graft (MRD) with 30mg of alemtuzumab or unrelated donor (MUD) graft with 60mg of alemtuzumab. Using 686 eligible cases from the CIBMTR CV20-01 dataset, we included patients who had a minimum of 12-month follow-up. We used an optimally matched-pair propensity score to compare the outcomes of patients receiving PTCy vs alemtuzumab in a 3:1 ratio, matching by variables in Table 1, and criteria were well balanced between cohorts after matching. Outcomes were evaluated by 1-year GVHD- and relapse-free survival (GRFS), overall survival (OS), and non-relapse mortality (NRM), and compared with log-rank tests of significance. Time to neutrophil and platelet engraftment between groups was compared with two-sided t-tests.

Results: Baseline patient characteristics are shown in Table 1, by CIBMTR-reported categories. We found that 1-year GRFS for those treated with alemtuzumab was 37% (95% CI: 27-52%) compared to 47% for PTCy (95% CI: 40-56%) (p=0.26). One-year OS for the cohort receiving alemtuzumab was 71% (95% CI: 61-84%) and 74% (95% CI: 68-80%) for the PTCy cohort (p=0.69). NRM at 1-year was 18% (95% CI: 7-28%) for patients receiving alemtuzumab compared to 12% (95% CI: 6-16%) for patients receiving PTCy (p=0.21). One-year cumulative relapse incidence was 26% for alemtuzumab, compared to 27% for PTCy. Among patients treated with alemtuzumab, 4 (7%) developed grade 3-4 acute GVHD, compared to 12 (7%) treated with PTCy (p= 0.94); 16 (27%) treated with alemtuzumab developed chronic GVHD requiring therapy, compared to 51 (29%) with PTCy (p=0.08). Average time to neutrophil and platelet recovery was 10 days for those receiving alemtuzumab compared to 18 days for PTCy (p<0.01 for both comparisons).

Table 1: Baseline characteristics of propensity-matched alemtuzumab and PTCy cohorts

Characteristic

Alemtuzumab: matched cohort (n= 58)

PTCy: matched cohort (n= 172)

Male gender, n (%)

36 (62)

113 (66)

Age, decade, n (%)

18-29

1 (2)

7 (4)

30-39

5 (9)

11 (6)

40-49

5 (9)

21 (12)

50-59

14 (24)

30 (18)

60-69

23 (39)

79 (46)

≥ 70

10 (17)

24 (14)

Race, n (%)

White

49 (85)

154 (90)

Black

2 (3)

9 (5)

Others

7 (12)

9 (5)

Karnofsky Performance Score, n (%)

≥ 90%

42 (72)

110 (64)

< 90%

16 (28)

62 (36)

Disease, n (%)

 AML

42 (72)

113 (66)

 MDS

16 (28)

59 (34)

Disease Risk Index (DRI), n (%)

 Low

34 (59)

25 (15)

 Intermediate

7 (12)

131 (76)

 High

17 (29)

16 (9)

HCT-CI, n (%)

14 (24)

46 (27)

 1-2

28 (48)

64 (37)

 ≥ 3

16 (28)

62 (36)

Graft type, n (%)

 MRD

27 (47)

68 (40)

 MUD

31 (53)

104 (60)

Conditioning intensity, n (%)

 Myeloablative

21 (36)

68 (40)

 Reduced intensity

37 (64)

104 (60)

Conclusions: In this comparative effectiveness study of patients receiving 30mg or 60mg of alemtuzumab with propensity-score matched patients treated with PTCy, survival outcomes and rates of serious GVHD are comparable. Neutrophil and platelet engraftment is significantly improved among patients receiving alemtuzumab. We acknowledge the retrospective nature and possibility of unmeasured confounding factors. Nevertheless, these results suggest that for selected patients in whom prolonged neutrophil and platelet engraftment may contribute to higher risk of early treatment-related mortality, alemtuzumab offers a viable prophylactic option for both acute and chronic GVHD.

Disclosure: Alexandra Rojek: Nothing to declare.

Mylove Mortel: Nothing to declare.

Andrew Artz: Stock/ownership in Radiology Partners; consulting/advisory role for Abbvie and Magenta Therapeutics.

Richard Larson: Consulting/advisory role for AbbVie, Actinium Pharmaceuticals, Curis, CVS Caremark, Epizyme, Immunogen, Jazz Pharmaceuticals, Kling Biotherapeutics, Novartis, Rigel, SERVIER, Takeda Science Foundation; Institutional research funding from Astellas Pharma, Celgene, Cellectis, Daiichi Sankyo, Gilead/Forty Seven, Novartis, Rafael Pharmaceuticals; Patents and royatlies from UpToDate.

Hongtao Liu: Consulting/adivsory role for Agios, BeiGene, CTI BioPharma Corp, NGM Biopharmaceuticals, Nkarta, Pfizer; Institutional research funding from Bristol-Myers Squibb and Karyopharm Therapeutics.

Adam Duvall: Nothing to declare.

Mariam Nawas: Nothing to declare.

Toyosi Odenike: Consulting/advisory role for Abbvie, Blueprint Medicines, BMS, Celgene, CTI, Impact Biomedicines, Kymera, Novartis, SERVIER, Taiho Pharmaceutical, Threadwell therapeutics; Institutional research funding from Abbvie, Agios, Aprea AB, Astex Pharmaceuticals, AstraZeneca, Bristol-Myers Squibb, Celgene, CTI BioPharma Corp, Daiichi Sankyo, Incyte, Janssen Oncology, Kartos Therapeutics, Loxo, Novartis, NS Pharma, OncoTherapy Science.

Gregory Roloff: Nothing to declare.

Wendy Stock: Honoraria from Abbvie and Pfizer. Consulting/advisory role for AstraZeneca, BEAm, Deciphera, GlaxoSmithKline, Jazz Pharmaceuticals, Kite, Kronos Bio, Kura Oncology, morphosys, Newave Pharmaceutical, Pfizer, Pluristem Therapeutics, SERVIER; Patents and royatlies from UpToDate. Travel and accommodations from Pfizer.

Michael Bishop: Honoraria from Bristol-Myers Squibb/Celgene/Juno, Incyte, and Sanofi/Aventis. Consulting/advisory role for Arcellx, Autolus, BMS, crispr therapeutics, Iovance Biotherapeutics, Kite, Novartis, Sana Biotechnology. Speakers’ bureau from Agios, Bristol-Myers Squibb, Incyte, Kite, Sanofi. Institutional research funding from Arcellx, crispr therapeutics, Iovance Biotherapeutics, Kite, Novartis. Expert testimony for Johnson & Johnson/Janssen. Travel and accommodations from BMS, Kite/Gilead, Novartis.

Satyajit Kosuri: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P244 EXTENDED ABATACEPT USE WITH SEROTHERAPY TO REDUCE ACUTE GVHD INCIDENCE IN PEDIATRIC PATIENTS TRANSPLANTED FROM HLA-MISMATCHED UNRELATED DONORS

Francesco Quagliarella1, Jochen Buechner2, Mattia Algeri1, Ingvild Heier2, Francesca Del Bufalo1, Phoi Phoi Diep2, Daria Pagliara1, Marco Becilli1, Barbarella Lucarelli1, Emilia Boccieri1, Federica Galaverna1, Ilaria Pili1, Roberto Carta1, Chiara Rosignoli1, Tiziana Corsetti1, Pietro Merli 1, Franco Locatelli1,3

1IRCCS Ospedale Pediatrico Bambino Gesù, Rome, Italy, 2Oslo University Hospital, Oslo, Norway, 3Catholic University of the Sacred Heart, Rome, Italy

Background: The use of mismatched unrelated donor (MMUD) for patients lacking a fully HLA-matched donor is associated with an increased risk of both acute and chronic GVHD, as well as of TRM.

Abatacept (ABA), a fusion protein that selectively inhibits T‐cell co-stimulation by binding to CD80 and CD86 on antigen‐presenting cells and thus blocking CD28-mediated signaling, has been successfully used in the MMUD HSCT setting to prevent acute GvHD as an alternative to serotherapy (Watkins, J Clin Oncol 2021).

Methods: Starting from February 2021, ABA was administered on an off-label basis in 2 centers to 54 consecutive pediatric/young adult patients receiving an unmanipulated graft from a MMUD with 1 or 2 mismatches for major HLA alleles (HLA-A, -B, -C, DRB1, DQB1). Initially, ABA was given at a dose of 10 mg/kg (max 750 mg/dose) on days -1, +5, +14, and +28 (16 patients, group 1). Thirty-eight patients (group 2) received two additional infusions every two weeks (day +42 and +56) due to the occurrence of 2 cases of late aGVHD in the first cohort. ABA was added to standard GvHD pharmacological prophylaxis (CsA, MTX and serotherapy) administered according to EBMT guidelines.

All patients, except 13 with congenital or acquired bone marrow failure, received either a TBI- or chemo-based myeloablative conditioning regimen. GVHD grading was evaluated according to Mount Sinai Acute GVHD International Consortium (MAGIC) criteria.

Results: Table 1 summarizes patient and HSCT characteristics. Median follow-up of surviving patients was 13 months (range, 3-28). All patients engrafted at day +30.

Cumulative incidence of grade II-IV and grade III-IV aGVHD for the entire cohort were 26.9% (95% CI 16.8-41.2) and 9.5% (95% CI 4.1-21.5), respectively. Notably, incidence of aGVHD was higher in patients who received four doses of ABA. In details, the cumulative incidence of grade II-IV aGVHD for patients of group 1 and group 2 were 52.8% (95% CI 30.8-78.4) and 16.4% (95% CI 7.6-32.8) (p=0.005), respectively, and for grade III-IV 20.0% (95% CI 9.6-50.0) and 5.3% (95% CI 1.3-19.7), respectively (p=0.1). In a multivariable model for grade II-IV aGvHD including also source of stem cells and type of mismatch (class I/II), the prolonged use of abatacept remained significantly protective (HR 0.95, 95% CI 0.92-0.99, p=0.01).

Overall (OS) and event-free survival (EFS) in the entire cohort were 86.4% (95% CI 71.6-93.8) and 81.0% (95% CI 65.8-89.9), respectively. Notably, the prolonged use of ABA resulted in higher survival probabilities (OS 92.0% vs 74.5%, p=0.04 and EFS 91.4% vs 60.2%, p=0.009).

Regarding safety profile, we recorded one infectious death due to invasive aspergillosis and several cases of EBV and CMV reactivation necessitating the use of rituximab or antiviral therapy, respectively. Three patients experienced a secondary graft failure.

Table 1

N

% - range

Age at HSCT (years, range)

7.8

1-20.0

Diagnosis

ALL

15

28

AML

12

22

Other malignancies

4

7

SAA/RCC

10

19

Other non-malignant diseases

13

24

Mismatch

8/10

3

6

9/10

51

94

Locus of MM

Class I

41

76

Class II

11

24

Source of Stem Cell infused

BM

36

67

PBSC

18

33

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (35)

Conclusions: Our data indicate that ABA, added to a standard pharmacological GVHD prophylaxis based on CNI + MTX+serotherapy, is a safe and effective approach for preventing aGvHD in pediatric patients undergoing MMUD HSCT for both malignant and non-malignant diseases, especially if administered up to day + 60, thus widening the potential donor pool for patients in need of an allograft.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P245 ENDOTHELIAL DYSFUNCTION AND RISK OF ACUTE GRAFT-VERSUS-HOST DISEASE AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: A PRELIMINARY ANALYSIS OF A SINGLE CENTRE STUDY

Francesco Romano1, Antonio Bianchessi1, Irene Defrancesco1, Giulia Losi 1, Beatrice Ferrari2, Virginia Ferretti2, Cristina Picone2, Erica Consensi2, Mara De Amici2, Giorgia Testa2, Daniela Caldera2, Caterina Zerbi2, Valentina Zoboli2, Luca Arcaini1,2, Nicola Polverelli2

1Università degli Studi di Pavia, Pavia, Italy, 2Fondazione IRCCS Policlinico San Matteo, Pavia, Italy

Background: Endothelial dysfunction has been increasingly correlated to the very early phases of acute graft-versus-host-disease (aGVHD) with endothelial cells acting both as a target of alloreactive donor T cells and as starting elements of the inflammatory process (“endothelial GVHD”).

Methods: We carried out a single centre prospective study to assess whether endothelial dysfunction at the time of allogeneic hematopoietic stem cell transplantation (HSCT) could be associated with the risk of aGVHD. We analyzed a cohort of 46 consecutive adult patients who underwent their first HSCT from any donor for hematological disease at the Division of Hematology of IRCCS Policlinico San Matteo in Pavia (Italy), from August 2021 to April 2023. Levels of circulating endothelial cells (CECs), circulating endothelial progenitors (CEPs) and some soluble biomarkers (plasminogen activator inhibitor-1 [PAI-1], vascular endothelial growth factor [VEGF], angiopoietin-2 [ANG-2], soluble VCAM [sVCAM]) were assessed on peripheral blood samples collected on the day of HSCT (T0) and at other fixed and dynamic time-points.

CECs and CEPs were identified by flow cytometry according to Lanuti et al. (Sci Rep. 2018;8(1)) and Mancuso et al. (J Thromb Haemost. 2020;18(10):2744-2750) protocol, respectively. CECs >30 cells/ml were considered elevated.1 For CEPs, normal reference has not been identified yet. As a proof-of-concept study, p values <0.2 were considered significant.

Results: Clinical and transplant characteristics of the patients are depicted in Table 1. Median follow-up was 9.2 months (IQR: 3.7-13.8 months). Day 100 cumulative incidence of aGVHD was 40% (95% CI: 26-54) with median time of onset of 36 days (IQR 27-72 days). Cumulative incidence of relapse at 6 months was 7% (95%CI: 1.8%-17.1%). Overall survival at 6 months was 86% (95% CI: 71-93%) and non-relapse mortality at 6 months was 8% (95% CI: 2.4-18.6%).

Median CECs and CEPs at T0 were 43/ml (IQR: 27-77) and 22/ml (IQR: 0-40), respectively. We found that CECs count >30 cells/ml at T0 was associated with higher risk of aGVHD of any grade (p=0.14, SHR 2.36 95% CI 0.7-7.3). In particular, day 100 cumulative incidence of aGVHD was 46.5% (95% CI 28-63) compared to 28.5% (95% CI 8-52) in patients with normal CECs count at T0.

As for CEPs, their values at T0 were divided in tertiles (1° tertile: ≤41 cells/ml; 2° tertile: 42-138 cells/ml; 3° tertile: ≥139 cells/ml). We observed that patients with CEPs ≥139 cells/ml experienced a better aGVHD-free survival and lower cumulative incidence of grade ≥II aGVHD compared to patients with CEPs count ≤41 cells/ml (p=0.093 and p=0.18, respectively).

Among soluble endothelial biomarkers, lower or within-the-range levels of ANG-2 at T0 showed correlation with aGVHD-free survival (p=0.10).

Table 1: Clinical and transplant characteristics of the study population

Clinical and Transplant Characteristics

Total, n (%)

aGVHD population**, n (%)

No-aGVHD population**, n (%)

Patients

46

21

25

Age, median (years) (IQR)

55 (42-62)

53 (32-59)

56 (43-65)

Sex M/F

18/28 (40/60)

10/11 (48/52)

8/17 (33/67)

Diagnosis

Acute leukemia

29 (63)

16 (76)

13 (52)

MDS

3 (6)

2 (10)

1 (4)

MPN

6 (13)

2 (10)

4 (16)

MDS/MPN

1 (3)

-

1 (4)

Lymphoma

4 (8)

-

4 (16)

Myeloid neoplasm with germ line predisposition

1 (3)

-

1 (4)

Severe Aplastic Anemia

2 (4)

1 (4)

1 (4)

Disease status at transplant (n)

Complete remission

29 (62)

14 (67)

15 (58)

Not in complete remission/active

8 (18)

4 (19)

4 (17)

Chronic phase

6 (13)

2 (10)

4 (17)

Onset

3 (7)

1 (5)

2 (8)

DRI-score*

Low

6 (14)

1 (5)

5 (21)

Intermediate

26 (58)

11 (58)

15 (58)

High

10 (23)

6 (32)

4 (17)

Very-high

2 (5)

1 (5)

1 (4)

Lines of therapy before HSCT

1

22 (53)

12 (60)

10 (45)

2

11 (28)

5 (25)

6 (30)

≥3

8 (19)

3 (15)

5 (25)

HCT-CI

Low risk

14 (31)

8 (38)

6 (25)

Intermediate risk

18 (40)

7 (33)

11 (46)

High risk

14 (29)

6 (26)

8 (29)

Donor type

Sibling HLA identical donor

4 (9)

2 (9)

2 (8)

Haploidentical donor

10 (22)

5 (24)

5 (21)

MUD HLA 8/8

18 (38)

7 (33)

11 (42)

MMUD HLA 7/8

14 (31)

7 (33)

7 (29)

Gender (donor/recipient)

F/F or M/M or M/F

43 (93)

19 (91)

23 (92)

F/M

3 (7)

2 (9)

2 (8)

Source of HSCs

PBSC

42 (91)

19 (91)

23 (92)

BMSC

4 (9)

2 (9)

2 (8)

Conditioning regimen

MAC

40 (87)

19 (90)

21 (83)

RIC

5 (11)

2 (10)

3 (13)

NMA

1 (2)

-

1 (4)

GVHD prophylaxis

CsA + MTX

27 (58)

12 (57)

15 (58)

CsA+MMF + PTCY

19 (42)

9 (43)

10 (42)

ATG use

24 (52)

9 (39)

15 (61)

  1. *DRI not applicable to patients with severe aplastic anaemia
  2. ** as for aGVHD population and no-aGHVD population we refer to patients who experienced acute graft versus host disease or not during follow up
  3. Abbreviations: aGVHD: acute graft versus host disease; ATG: anti-thymocyte globulin; BMSC: bone marrow stem cells; cGVHD: chronic graft versus host disease; CsA: cyclosporine; DRI: disease risk index; HCT-CI: Hematopoietic Cell Transplasntation-specific Comorbidity Index; HSCs: hematopoietic stem cells; HSCT: allogeneic hematopoietic stem cell transplantation; MAC: myeloablative conditioning; MDS: myelodysplastic syndromes; MDS/MPN: myelodysplastic syndromes/myeloproliferative neoplasms; MMF: micophenolate mofetil; MPN: myeloprilferative neoplasms; MTX: methotrexate; MUD: matched unrelated donor; MMUD: mismatched unrelated donor; NMA: non myeloablative conditioning; PBSC: peripheral blood stem cells; PTCY: post-transplant cyclophosphamide; RIC: reduced intensity conditioning

Conclusions: Overall, our findings suggest that early endothelial damage could contribute to the risk of aGVHD. Larger study cohorts are warranted in order to confirm our preliminary results.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P246 EXTENDED ABATACEPT DOSING MAY REDUCE RATES OF CHRONIC GRAFT VS HOST DISEASE IN PEDIATRIC PATIENTS UNDERGOING BONE MARROW TRANSPLANT FOR UNDERLYING HEMATOLOGICAL DISEASES

Eleanor Cook1, Pooja Khandelwal1, Ashley Teusink-Cross1, Michael Grimley 1

1Cincinnati Children’s Hospital Medical Center, Cincinnati, United States

Background: T-cell co-stimulation blockade with four doses of abatacept is FDA approved for acute GVHD prophyalxis in patients undergoing unrelated donor bone marrow transplant (BMT) for malignancies1 and under investigation for prophyalxis against cGVHD in the same population. Adoption of abatacept for extended GVHD prophyalxis has been limited by concerns about increase of graft failure/risk of relapse.

Hypothesis: Extending the duration of abatacept dosing beyond day+28 may reduce the incidence of cGVHD in pediatric patients undergoing BMT for non-malignant conditions without impacting engraftment or incidence of viral infections.

Methods: Patients with hemoglobinopathies who underwent allogeneic BMT and received ‘extended abatacept’ at a single, high-volume USA pediatric BMT center were retrospectively reviewed. Extended abatacept was defined as abatacept dosing with intent to prevent chronic GVHD, beyond (and in addition to) the routine four doses at D-1, +5, +14 and +28. Abatacept was dosed at 10mg/kg and administered monthly via the intravenous route, from as early as 60 days after BMT. Total duration of dosing was variable.

Results: Seventeen patients received extended abatacept (Table 1) (2018-2023). Nine were male (53%) and median age was 6.5yo (range 2-13yo). Fourteen patients underwent BMT for beta thalassemia (82%), three for sickle cell disease (18%). Nine patients received matched sibling or matched related graft (53%), 8 (47%) received an unrelated donor graft. 82% were 10/10 match, and all patients received marrow as graft source. The median number of extended abatacept doses received was 5 (range 2-13).

At a median follow up time of 3 years (range 4months-5.1years), there was no graft failure, graft rejection or mixed chimerism. All patients are alive at last follow up.

Chronic GVHD was diagnosed in two patients (11.7%), graded as mild (n=1) and severe (n=1). This is lower than published incidence of chronic GVHD in a similar patient population2. Our group has previously published outcomes for a cohort of beta thalassemia patients treated with the 4 dose abatacept regimen3: in this cohort, the incidence of cGVHD was 25%. On a sub-analysis of the current extended abatacept cohort, chronic GVHD incidence in the beta thalassemia patients (n=14) was 14%. Notably, this extended abatacept thalassemia cohort had a higher proportion of unrelated donors (50% vs 37%).

Clinically significant viral disease occurred in one patient with BK cystitis. Eleven patients had asymptomatic CMV viremia (65%). Of these, 10 received treatment, mostly with donor-dervied viral specific T cells (VSTs) (n=6) at a median of D + 43. Of note, only one patient required treatment for CMV beyond D + 60 (i.e. the time of starting extended abatacept regimen). Six patients had EBV reactivation (35%), 2 required intervention. One patient had adenoviral infection (concurrent with CMV and BK viremia- received VSTs). Two patients (12%) had BK viremia, 1 (with BK cystitis) required treatment with VST infusion.

All patients, n= 17

Sex, n (%)

Female

8 (47%)

Male

9 (53%)

Median age at HSCT, years (range)

6.53 (2-13)

Primary Diagnosis, n (%)

Beta thalassemia

14 (82%)

Sickle cell disease

3 (18%)

Donor Relation, n (%)

Unrelated donor (URD)

8 (47%)

Matched sibling donor (MSD)

7 (41%)

Matched related donor (MRD)

2 (12%)

HLA Match, n (%)

10/10

14 (82%)

9/10

3 (18%)

Donor Source, n (%)

Marrow alone

15 (88%)

Marrow + cord

2 (12%)

Preparative Regimen, n (%)

Busulfan/ Fludarabine/ Thiotepa

15 (88%)

Campath/ Fludarabine/ Melphalan/ Thiotepa

1 (6%)

Campath/ Fludarabine/ Melphalan

1 (6%)

GVH prophylaxis (in addition to abatacept), n (%)

Cyclosporin/ Mycophenolate

7 (41%)

Cyclosporin / Prednisone

7 (41%)

Cyclosporin / Methotrexate

2 (12%)

Cyclosporin alone

1 (6%)

Abatacept dosing

Median number of doses received (range)

5 (2-13)

.

Table 1. Patient and transplant demographics data.

Conclusions: Extended dosing of abatacept for patients undergoing BMT for non-malignant conditions reduced the rate of chronic GVHD without impact on engraftment and viral infections. Extended abatacept dosing appears to be safe and well tolerated in the era of VST availability.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P247 COMPARISON OF THE EFFICACY OF MYCOPHENOLATE MOFETIL AFTER ORAL OR INTRAVENOUS ADMINISTRATION IN THE PREVENTION OF ACUTE GRAFT-VERSUS-HOST DISEASE

Xinhui Zheng1, AiMing Pang1, Erlie Jiang 1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China

Background: MMF has been used extensively in allo-HSCT for GVHD prophylaxis, especially in patients with unrelated/haploidentical donors and other high-risk factors. However, the efficacy of oral dosage was limited by intestinal mucosal damage, the reduction of the intestinal flora involved in the metabolic pathways, and the intervention of CSA. The intravenous dosage was reported to be associated with higher C-max and AUC value, better concentration monitoring, and fewer gastrointestinal adverse effects, while clinical studies on the comparison between the two dosages were lacking.

Methods: Patients were eligible and taken into the analysis when they met the following criteria: (1) undergoing allo-HSCT from unrelated/ haploidentical donors at an age of 15–65 years between 2015 and 2021; (2) with a high risk of GVHD, such as advanced age of patients or donors (≥45 years old), and sex mismatch, defined as a female donor to a male recipient (F→M); (3) with a calcineurin inhibitor, MTX and MMF for GVHD prophylaxis; (4) MMF was administrated before transplantation and persisted for at least 30 days. Patients who have breaks from MMF administration for more than 5 days were not allowed. Patients who changed from IV dosages to oral dosages within 25 days after transplantation were not eligible.

Results: Between February 2015 and May 2021, 218 patients with various diseases, undergoing allogeneic SCT from unrelated/ haploidentical donors were enrolled, with a median age at transplantation of 35 (range 15–58) years. In competing risk models, the cumulative incidence of III-IV aGVHD at 100 days after HSCT was 0.21 (95%CI 0.13-0.32), and 0.09 (95%CI 0.05-0.14) in oral and IV groups (p=0.009), respectively. There was no statistical difference in I-IV, and II-IV aGVHD between the oral and IV group (I-IV aGVHD, 0.54(0.42-0.65) vs 0.45(0.37-0.53) p=0.18; II-IV aGVHD 0.33(0.22-0.44) vs 0.24(0.17-0.31) p=0.15). Among 67 patients suffering from I-IV aGVHD in the IV group, skin, gastrointestinal, and liver involvement occurred at the percentage of 67%, 36%, and 16.5%, respectively. And among 38 patients in the oral group, the ratio was 68%, 55%, and 26%. The cumulative incidence of cGVHD was higher in the IV group (2 years, 0.50(95%CI 0.41-0.58) vs 0.36(95%CI 0.25-0.47)), but the difference was not statistically significant. The median follow-up times were 1.5 (0.2-5.7) years for the whole cohort, 1.6 (0.2-5.7) years for the oral group, and 1.5 (0.2-3.7) years for the IV group. The overall 1.5-year OS was 74.9% (95%CI 0.69-0.81) and the IV dosage was associated with better OS (1.5-year OS, 80.6% (95%CI 0.74-0.89) vs 60.6% (95%CI 0.50-0.73), p<0.001).

Conclusions: In conclusion, among patients at a high risk of aGVHD, the IV dosage of MMF could decrease the incidence of III-IV aGVHD, and thus improve OS and NRM compared with the oral dosages, particularly in the case of tacrolimus combination. The IV dosage was also related to earlier neutrophil and platelet engraftment, but more researches are still needed in the future.

Disclosure: The authors declared that they have no conflicts of interest to this work.

12: Graft-versus-host Disease – Clinical

P248 NEAR ELIMINATION OF GVHD WITH AN ABBREVIATED CALCINEURON AND M-TOR INHIBITOR-FREE GVHD PREVENTION REGIMEN

A. Samer Al-Homsi 1, Kelli Cole1, Frank Cirrone1, Stephanie Wo1, J. Andres Suarez-Londono1, Jingmei Hsu1, Mohammad Abu-Zaid1, Maher Abdul-Hay1

1New York University Grossman School of Medicine, New York, United States

Background: Calcineurin and m-TOR inhibitor-free GvHD prevention regimens have many potential advantages. Post-transplant cyclophosphamide (PTCy) and bortezomib (Bor) is such a combination and produces results comparable to those obtained with the combination of methotrexate and tacrolimus (Al-Homsi et al. BBMT 2017). Abatacept (Aba) has been shown to decrease the incidence of acute GvHD in combination with methotrexate and tacrolimus in similar settings. We aimed to explore the safety and efficacy of adding a short course of Aba to CyBor (ABC).

Methods: Patients (pts) with hematological malignancies undergoing peripheral blood allogeneic transplant from matched sibling (MS), matched unrelated (MU) or mismatched unrelated (MMU) donors were enrolled in this phase I-II study. Patients were enrolled in 2 parallel strata for MS and MU donor and for MMU donor transplants. The primary end point was the incidence of grades II-IV acute GvHD by day (d) +120. With a 2-stage Simon design, a futility analysis was planned after enrollment of 22 and 19 patients in the 2 strata, respectively. Patients received fludarabine and busulfan (2 d) or fludarabine, busulfan (3 d) and thiotepa conditioning. The GvHD prevention regimen consisted of CyBor as previously described with Aba at 10mg/Kg on d + 5, +14, and +28. Patients receiving grafts from MU or MMU donors also received rATG at total dose of 5mg/kg fractionated on d -4 to -2.

Results: Pt numbers and characteristics are summarized in Table 1. As of December 7, 2023, the median follow-up for the entire cohort was 211 d. Median time to ANC engraftment was 17 d. One pt did not achieve platelet engraftment; for the remaining pts the median time to platelet engraftment was 25.5 d. There was no graft failure. Four pts met criteria of poor graft function; 3 received CD34 boost and recovered. Nineteen pts have >120 d of follow-up and are evaluable for the primary endpoint. Fifteen pts have >180 d of follow-up. No pt developed grade II-IV acute GvHD by d + 120. Two pts developed possible acute GvHD following CD34 boost. One pt developed moderate chronic GvHD limited to mucocutaneous involvement. Two pts relapsed; 1 with cytogenetic relapse only. Nine (36%) pts had CMV reactivation requiring preemptive therapy; 1 pt developed possible CMV gastrointestinal disease. Three pts developed EBV reactivation requiring therapy; 1 with PTLPD. All 3 recovered with rituximab. There was one treatment-related mortality due to ischemic bowel. None of the safety or futility stoppage rules were triggered.

Conclusions: This ongoing trial suggests that the entirely calcineurin and m-TOR inhibitor-free abbreviated combination of ABC is feasible and highly efficacious in preventing GvHD.

Clinical Trial Registry: NCT05289167.

Disclosure: M. Abdul-Hay: Advisory boards: Kite, Daiichi, Rigel and Incyte; Speaker Bureau: Jazz, Takeda and Servier.

A. Samer Al-Homsi: educational grant from BMS; Scientific Advisory Board member for Incyte.

12: Graft-versus-host Disease – Clinical

P249 EFFICACY AND SAFETY OF REDUCED-DOSES OF POST-TRANSPLANT CYCLOPHOSPHAMIDE IN PERIPHERAL BLOOD STEM CELL TRANSPLANTATION OUTSIDE THE HAPLOIDENTICAL SETTING

Sara Redondo 1, Anna Arrufat1, Albert Esquirol1, Silvana Saavedra1, Miguel Arguello-Tomas1, Ana Carolina Caballero1, Guadalupe Oñate1, Ana Garrido1, Jordi Lopez1, Sara Miqueleiz1, J.M. Portos1, Eva Iranzo1, M.E. Moreno-Martinez2, Mireia Riba2, Jorge Sierra1, Javier Briones1, Irene García-Cadenas1, Rodrigo Martino1

1Hospital de la Sant Creu i Sant Pau. IIB-Sant Pau and José Carreras Leukemia Research Institutes. Universitat Autónoma de Barcelona, Barcelona, Spain, 2Hospital de la Sant Creu i Sant Pau, Barcelona, Spain

Background: PTCy, a GvHD prophylactic strategy, has demonstrated significant efficacy in reducing the incidence of severe GvHD. However, the standard dose of 50 mg/kg/day on days +3 and +4 certainly adds toxicity, including potential cardiac events, hemorrhagic cystitis, and delayed engraftment and immune recovery.

Aiming to mitigate PTCy-related toxicity, in 2018, we introduced a new transplantation strategy based on reduced doses of PTCy (from 50 to 30 mg/kg/day on days +3 and +4) and tacrolimus as GvHD prophylaxis for adult patients undergoing an allo-HSCT from a matched sibling donor (MSD), matched unrelated donor (MUD), or a single-allele mismatched MUD (mMUD) who were considered at high risk of graft failure and/or early NRM. The new platform was used in case of significant cardiac comorbidities, ≤4 x 106/kg infused CD34+ cells, and/or myelofibrosis (MF) as baseline disease.

Methods: Retrospective, non-randomized single-center study, carried out in a tertiary university hospital.

Results: Between February 2018 and March 2023, 31 adult patients were included, with a median age of 59 years (range, 41-72), and MF (n=17, 54.8%) being the most prevalent underlying disease. Twelve patients (38.7%) had a HCT-CI ≥3, 4 (9.7%) had cardiac comorbidities, and 7 (22.6%) received ≤4 x 106/kg CD34 + . Most (77.4%; n =24) underwent a reduced-intensity conditioning regimen, and 6 (19.3%) had a mMUD (Table 1).

All patients had successful donor engraftment, except 3 who died during the neutropenic phase. Median time for neutrophil and platelet recovery was 20 (range, 16-31) and 21 days (range,17-35). Post-transplant growth factors were not used.

After PTCy administration, there were 2 (6.4%) cardiac events (1 left ventricular systolic dysfunction resulting in the patient’s death, despite no prior history of cardiac disease, and 1 acute pulmonary edema that resolved), 2 (6.4%) BK virus-related hemorrhagic cystitis, and 10 (32.3%) CMV infection (letermovir prophylaxis was not available). No cases of CRS, thrombotic microangiopathy, or veno-occlusive disease were identified. No delayed cardiac complications at last follow-up.

The day+ 120 cumulative incidence of grade II-IV and III-IV aGvHD was 43% (95% CI: 22.4-58.4) and 16.7% (95% CI: 2.2-29). The cumulative incidence of global and moderate to severe cGvHD at 20 months was 25.7% (95% CI: 5- 41.8) and 6.5% (95% CI: 2.7-15.6). The risk factor which had a significant impact on aGvHD in the univariable analysis was mMUD [HR 9 (95% CI: 1-86.9); p =0.05].

At data cutoff-November 2023, the median follow-up for survivors was 20 months (range, 10-45). The 20-month probability of OS and PFS were 75.9% (95% CI: 61.7-93) and 70.5% (95% CI: 54.8-90.7). The incidence of NRM was 6.7% (95% CI: 0-15) at 100 days, and 10% (95% CI: 0-20) at 12 months. GVHD-free/relapse-free survival and without systemic immunosuppression at 12 months was 63.6% (95% CI: 46-87.9).

Patient characteristics, N=31 (% in parentheses).

Age, median [range

59 [41-72]

Sex, Male

18 (58.1%)

Underlying disease

• Myelofibrosis

17 (54.8%)

• Acute Leukemia

10 (35.5%)

• Others

3 (9.7%)

Disease status prior HSCT

• Complete response

10 (32.3%)

• Partial response

7 (22.6%)

• Stable Disease

14 (45.1%)

EBMT risk score ≥ 5

16 (51.6%)

HCT-CI ≥ 3

12 (38.7%)

Disease risk index (rDRI)High-Very high

8 (25.8%)

Cardiac event prior to HSCT

4 (12.9%)

Donor type

• Matched sibling

10 (32.3%)

• Matched unrelated

15 (48.4%)

• Single-allele mismatched unrelated

6 (19.3%)

Reduced intensity Conditioning regimen

24 (77.4%)

Patient male-donor female

8 (25.8%)

Source, Peripheral blood

29 (93.5%)

CD34+ cells infused (x 106/kg)

5.14 [0.42-6.7]

CMV-/-

6 (19.4%)

Conclusions: Our study confirms the safety and efficacy of reduced doses of PTCy in adult patients with MF, prior cardiopathy, or those receiving lower CD34+ cells, undergoing allo-HSCT. These promising outcomes have led us to consider extending this dose of PTCy to all recipients outside the haploidentical setting, with the exception of mMUD.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P250 A LOW TRAJECTORY OF PLATELETS IN THE EARLY STAGES AFTER TRANSPLANTATION IS LINKED TO THE DEVELOPMENT OF SEVERE ACUTE GRAFT-VERSUS-HOST DISEASE (AGVHD)

Dan Feng1, Erlie Jiang 1

1Hematological Hospital, Chinese Academy of Medical Sciences, Tianjin, China

Background: Haploidentical Hematopoietic Stem Cell Transplantation (HID-HSCT) is a crucial approach for curing Acute Myeloid Leukemia (AML). Acute graft versus host Disease (aGVHD) is the most common complication in patients after transplantation, significantly impacting the prognosis. Previous studies have indicated that patients with aGVHD often experience a decrease in platelet count. Additionally, research has found a higher tendency for aGVHD occurrence in patients with poor platelet recovery within the first 100 days post-transplantation. However, the relationship between platelet trajectories, especially in the early post-transplant period, particularly within the first 10 days after stem cell infusion, and the occurrence of aGVHD remains unclear. This study aims to explore the relationship between early post-transplant (day 0 to day 10) platelet trajectories and the occurrence of aGVHD.

Methods: The research involved a total of 216 AML patients who underwent HID-HSCT (hematopoietic stem cell transplantation) at the Hematology Hospital of the Chinese Academy of Medical Sciences between January 2017 and March 2023. All these patients had achieved complete remission before the transplantation and were then divided into two groups: a training cohort comprising 165 cases and a validation cohort comprising 111 cases. Group-based trajectory modeling (GBTM) was used to categorize the platelet trajectories during the early post-transplantation period (0-10 days). The study’s primary endpoint was aGVHD (Acute Graft-Versus-Host Disease), with the secondary endpoint being the cumulative incidence rate of aGVHD within 100 days, graded according to the Glucksberg criteria.

Results: Three distinct platelet trajectories were identified in the training cohort using the GBTM model: low, medium, and high. The low platelet trajectory group had a significantly higher proportion of severe aGVHD cases compared to the high platelet trajectory group (28.6% vs. 4.9%, P < 0.001). Multivariate analysis showed that a low platelet trajectory was an independent risk factor for severe aGVHD (HR [95% CI]: 0.383 [0.212-0.692], P= 0.001). Furthermore, the cumulative competing risk analysis of severe aGVHD demonstrated that the low platelet trajectory group had a significantly higher 100-day cumulative incidence rate of severe aGVHD than the medium and high platelet trajectory groups (46.2% vs. 25.0% vs. 7.7%, P= 0.003).

In the validation cohort, after grouping patients based on their platelet trajectories using GBTM, it was still observed that those in the low platelet trajectory group had a higher 100-day cumulative incidence rate of severe aGVHD (37.5% vs. 12.5% vs. 8.3%, P= 0.030). Moreover, the early platelet transfusion demand group (≤5 days) had a higher 100-day cumulative incidence rate of severe aGVHD, consistent with platelet dynamic trajectories (35.7% vs. 15.2%, P= 0.035).

Conclusions: The study found that the trajectory of platelet count after stem cell transplantation can predict the occurrence of severe aGVHD, and patients with an early need for platelet transfusion are more prone to developing severe aGVHD. In previous studies, the absolute count of platelets has been considered a crucial indicator. For the first time, this study establishes a connection between the changes in platelet trajectories and the prognosis of the disease, which may hold significant implications.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P251 GVHD PROPHYLAXIS WITH THREE VERSUS FIVE DRUGS COMBINATION IN AML PATIENTS UNDERGOING ALLOGENEIC TRANSPLANT FROM HLA MISMATCHED UNRELATED OR HAPLOIDENTICAL RELATED DONOR

Alessandra Picardi 1,2, Stella Santarone3, Massimo Martino4, Gottardo De Angelis2, Mariangela Pedata1, Stefania Leone1, Annalisa Natale3, Benedetta Mariotti2, Barbara Loteta4, Serena Marotta1, Doriana Vaddinelli3, Maria Celentano1, Giulia Ciangola2, Maria Caterina Micò4, William Arcese5,2, Raffaella Cerretti2

1AORN Cardarelli, Naples, Italy, 2Fondazione Policlinico Tor Vergata, Rome, Italy, 3Pescara Hospital, Pescara, Italy, 4Grande Ospedale Metropolitano “BMM”, Reggio Calabria, Italy, 5Università Campus Biomedico, Rome, Italy

Background: Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) is an essential treatment for hematological malignancies. Nevertheless, the incidence of acute and chronic GVHD remains the major obstacle to the full benefit of the transplant. Regimens based on post-transplant cyclophosphamide (PTCy) or anti-thymocyte globulin (ATG) were both used for GVHD prophylaxis in the setting of mismatched donors, but no consensus on the more effective protocol has been reached. However, there are no data of comparison between Cyclosporine (CSA), methotrexate, mycophenolate-mofetil (MMF), ATG, and Basiliximab combination (CMMAB, 5 drugs combination) and the Baltimore like protocol including CSA, MMF and PT-CY (CMCy, 3 drugs combination). Aim of this retrospective study was to evaluate the impact of these 2 different GVHD prophylaxis strategies on the engraftment and GVHD incidence in AML patients who received an Allo-HSCT from mismatched unrelated or haploidentical related donor.

Methods:

Group A

Group B

p

5 drugs combination as GVHD proph

3 drugs combination as GVHD proph

N=51

N=37

Gender: M/F

24/27

20/17

0.52

Patient’s Age: median (range)

55 (20-70)

53 (21-73)

0,92

Disease status: (%): 1 CR

86

89

2CR

14

11

0.68

Conditioning regimen (%): MAC

57

54

0.79

RIC

43

46

Stem cell source (%): PB

8

70

<0.001

BM

92

30

donor Type (N): Haplo

51

34

mmMUD

-

3

Blood group mismatching %: minor

25

13

0.38

major

8

11

CMV risk (%): Low

3

5

Intermediate

94

77

0.11

High

3

18

In order to avoid the bias due to the effect of letermovir introduction in the clinical practice, data were collected from January 2019 to date at Transplant Program of AORN Cardarelli in Naples, Tor Vergata University in Rome, Ospedale Civile in Pescara, Ospedale Metropolitano in Reggio Calabria. Inclusion Criteria were: adult age, diagnosis of AML at high risk, Complete Morphological Remission at time of transplant, conditioning regimen according to Thiotepa, Busilvex and Fludarabine (TBF) schedule, 7/8 HLA mismatched from unrelated or haploidentical related donor, GVHD prophylaxis according to 5 (group A) or 3 drugs (group B) combination. Overall 88 patients were included in the analysis: 51 and 37 received 5 (group A) and 3 (group B) drugs as GVHD prophylaxis, respectively. The two patient series were matched for the variables described in table 1 and no significant differences were observed, but the stem cell source that was G-CSF priming BM in 93% of Group A and PB in 70% of Group B (p<0.001).

Results: The cumulative incidence (CI) of neutrophil engraftment was 97% with a median of 19 days (group A=13-31; group B=13-46) in both series (p= 0.1). The acute and chronic GVHD occurrence were 37% and 27% in the Group A and 13% and 13% in the Group B (respectively, p=0.025 and P=0.08). With a median follow up of 28.47 months (0.7-57) in the group A and 14.43 (0.5-46.46) in the group B, the 100days and 1-year Non Relapse Mortality (NRM) were 8% and 19.8% versus 5% and 8.1% (respectively, p= 0.69 and p=0.18). OS and Relapse Incidence were 65% and 20% in the group A and 76% and 16% in the group B (respectively p=0.5 and p=0.68).

Conclusions: In summary, these preliminary data on homogenous AML patients transplanted from HLA mismatched donor over a limited study period (2019-2023) and receiving TBF as conditioning regimen show that both GVHD prophylaxis schedules are effective in terms of cumulative incidence and timing of PMN engraftment. Although, comparing the two cohorts no significative impact on 100days, 1year-NRM and OS was observed, the 3 drugs combination seems more protective against the aGVHD occurrence (41% versus 13%; p= 0.01). However, an in depth-extension of the analysis is in progress.

Disclosure: nothing to declare.

12: Graft-versus-host Disease – Clinical

P252 CYTOKINE SECRETION DURING REDUCED INTENSITY CONDITIONING HAEMATOPOIETIC STEM CELL TRANSPLANT FOR MYELOID DISEASE, AN OPPORTUNITY FOR OUTCOME PREDICTION

Chris Armstrong 1, Hayley Foy-Stones1, Gardiner Nicola1, Doherty Derek2, Anthony McElligott2, Fiona O’Connell2, Eibhlin Conneally1, Catherine Flynn1, Nina Orfali1

1St. James’ Hospital, Dublin, Ireland, 2Trinity College, Dublin, Ireland

Background: AlloSCT is an inherently inflammatory process, with a range of pleiotropic cytokines mediating the amalgamation of recipient and donor immune systems. Individualised risk-assessments for transplant complications, including quantifiable biomarkers, are needed to improve outcomes for older patients in the RIC setting. Serial cytokine secretion monitoring may identify patients at increased risk of GVHD, relapse and survival and allow risk optimisation.

Methods: We measured 10 cytokines using multiplex electrochemiluminescence at 8 sequential time-points in 32 patients conditioned with fludarabine, busulfan (8mg/kg) and ATG (FluBu3+ATG) for myeloid diseases at the National Adult Stem Cell Transplant Programme in Ireland. The panel included IFN-γ, IL-1β, IL-2, IL-4, IL-6, IL-10, IL-12p70, IL-17A, IL-33 and TNF-α, measured at pre-transplant, day 0, 7, 14, 28, 60, 100 and 180.

Results: Patient characteristics are summarised in (Table 1). Cytokine secretion varied significantly through transplant and by cytokine. Bimodal peaks in IFN-γ were seen pre-transplant and at day 28. Early peaks at day 0 were seen in IL-2, IL-10 and IL-17A and mid-transplant peaks in IL-6 and IL-4 at days 14 and 28 respectively. IL-1β, TNF-α showed less variability over time and IL-12p70 increased sequentially. IL-33 secretion was low at all time-points.

Increased mortality risk was associated with heightened expression of IFN-γ pre-transplant (p=0.001), IL-2 at day 28 (p=0.03) and 60 (p=0.02), IL-6 pre-transplant (p=0.001), IL-12p70 at day 100 (p=0.04) and IL-17A at day 7 (p=0.04). Relapse was associated with an increased level of IL-1β pre-transplant (p=0.03), IL-12p70 at day 100 (p=0.03) and IL-17A at day 100 also (p=0.03). IFN-γ demonstrated a protective effect against grade II-IV acute GVHD at the day 14 time-point (p=0.004) and IL-6 at day 60 was associated with high-grade acute GVHD (p=0.04).

In those who developed acute GVHD there was evidence of organ tropism in several cytokines. Early IFN-γ excess at day 0 was associated with subsequent gastrointestinal GVHD (p=0.03), which was also seen with IL-1β at day 0 (p=0.05) and 7 (p=0.04) and a strong association with IL-17A at day 0 (p=0.005). Higher levels of early IL-2 were seen in those with subsequent liver GVHD at day 0 (p=0.01), whereas IL-4 showed a protective effect, with lower levels seen at day 60 (p=0.01).

Table 1 Characteristic

Age – year

Median: 60 (range: 47 – 74)

Sex

20(M), 12 (F)

Disease subtype AML

17

MDS

6

MPN

9

CIBMTR- DRI

2 (low) | 19 (intermediate) | 11 (high)

Age-adjusted HCT-CI

Median: 2 (range 1 - 6)

Donor source

MSD: 16 | MUD: 16

Donor age

Median: 38 (range: 20 – 67)

Engraftment

Neutrophils: Day 18 | Platelets: Day 19

Chimerism 3 months

Total: 25/30 | CD3+ fraction: 11/26

CD4 recovery 3/6 months

Day 100 median: 58 x 106/kg | Day 180: 95 x 106/kg

Follow-up

Median 502 days

OS – 6/12/18 months

6mo.: 93.75% | 12mo.:87.25% | 18mo.: 87.25%

RFS – 6/12/18 months

6mo.: 87.5% | 12mo.: 76.67% | 18mo.: 76.67%

Acute GVHD grade II-IV

15 (46.88%)

Acute GVHD grade III-IV

3 (9.38%)

Chronic GVHD

4 (12.9%)

Conclusions: Serial monitoring of pro- and anti-inflammatory cytokines during RIC alloSCT offers insights into the complexities of cell signalling from peri-conditioning inflammation, through the early alloreactive interface of engraftment and GVHD and ultimately immune tolerance and reconstitution. Several cytokines show promise in predicting clinical outcomes before overt manifestation, including detrimental RFS signals in those with early excessive IL-1β, IL-6 and IFN-γ and later followed by IL-17A and IL-12p70. A role for predictive modelling is further extended in the GVHD domain, where clear patterns of expression herald the risk of future organ-specific acute disease. The results of this study endorse validation in a larger cohort of patients using a refined panel of cytokines including IFN-γ, IL-1β, IL-6, IL-12p70 and IL-17A, employed at early, middle, and late time-points during RIC alloSCT using this technology which is adaptable to routine clinical laboratory practice.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P253 INTERFERON GAMMA ACTIVITY IN IMPAIRED HAEMATOPOIETIC STEM CELL (HSC) PROLIFERATION AND GRAFT-VERSUS-HOST DISEASE

Régis Peffault de Latour 1,2, Pietro Merli3, Emmanuel Monnet4, Malin Löfqvist4, Franco Locatelli3

1French Reference Center for Aplastic Anemia and PNH Hematology-Bone Marrow Transplantation, Hôpital Saint-Louis AP-HP, Paris, France, 2Université Paris Diderot, Paris, France, 3IRCCS, Bambino Gesù Children’s Hospital, Catholic University of the Sacred Heart, Rome, Italy, 4Swedish Orphan Biovitrum (Sobi), Basel, Switzerland

Background: Interferon-gamma (IFNγ) activity can be assessed in vivo by measuring circulating levels of its downstream target chemokine C-X-C motif ligand 9 (CXCL9). We investigated the association between IFNγ activity and diseases or conditions characterised by impaired HSC proliferation, including aplastic anaemia and graft failure (GF) following HSC transplant (HSCT), as well as in graft versus host disease (GvHD).

Methods: The role of IFNγ activity in patients at high risk of GF undergoing allogeneic HSCT and in patients with impaired HSC proliferation (e.g., severe aplastic anaemia) was prospectively investigated in a non-interventional, observational study (NCT04494061). All outcomes investigated were exploratory. For patients undergoing HSCT, outcome data from hospital records and blood for measuring CXCL9 levels were collected 7 days prior to HSCT, at HSCT, at days 1, 3, 5, 9, 13, 17, 21, 28, 31 and 37 post-HSCT, and at the time of suspected GF or GvHD up to day 100. For patients with impaired HSC proliferation, a single blood sample was taken at the time of diagnosis. An additional cohort of patients receiving allogeneic HSCT followed prospectively for GvHD was obtained from the French transplant network Cryostem. Blood samples were collected at the time of diagnosis in patients presenting with different forms and severity of GvHD or in patients without GvHD, at matching timepoints post HSCT. In both studies, serum CXCL9 levels were analysed using a validated immunoassay antigen test.

Results: Overall, 86 patients who underwent HSCT were enrolled in NCT04494061, and 15 patients with impaired HSC proliferation (primarily aplastic anaemia [14/15; 93.3%]), before the study was prematurely terminated for non-safety reasons. GF occurred in 5/86 (5.8%) patients who underwent HSCT. GvHD occurred in 33/75 (44.0%) patients (all acute GvHD). Mean maximum serum CXCL9 levels were higher in patients with GF versus without GF (2115 versus 896 ng/L, respectively), and with GvHD versus without GvHD (515 versus 280 ng/L, respectively). Mean serum CXCL9 levels were also higher in patients with impaired HSC proliferation versus healthy controls (632 versus 105 ng/L, respectively). However, not all patients with GvHD or impaired HSC proliferation had elevated CXCL9 levels (Table). Similarly, in the Cryostem cohort (N=52), mean serum CXCL9 levels were elevated in patients with GvHD versus those without GvHD (1073 versus 374 ng/L; p=0.0051), but demonstrated high interindividual variation, with not all patients with GvHD having elevated CXCL9 levels (Table). Patients with multiple organ involvement in GvHD tended to have the highest CXCL9 levels (mean, 1481 ng/L). Higher CXCL9 levels were also associated with increased severity of GvHD.

NCT04494061

CXCL9 levels, ng/L

Healthy volunteers

Impaired HSC proliferation

No GF

GF

Non-GvHDa

GvHD

N

65

15

78

5

42

33

Mean (SD)

105 (59)

632 (1174)

896 (1213)

2115 (2064)

280 (341)

515 (528)

Range

40–339

59–3842

40–6628

734–5753

47–1970

40–2642

Cryostem cohortb

CXCL9 levels, ng/L

Non-GvHD

Cutaneous aGvHD

Digestive aGvHD

Cutaneous and digestive aGvHDc

Pulmonary aGvHD

Overall aGvHD

N

10

20

11

9

2

42

Mean (SD)

374 (359)

1072 (700)**

861 (857)

1481 (1545)*

408 (355)

1073 (972)**

Range

40–1203

223–2365

40–2909

160–5333

156–659

40–5333

  1. aExcludes patients with graft failure.
  2. bMore information can be found at https://www.cryostem.org/en/.
  3. cIncludes patients with simultaneous cutaneous and digestive aGvHD.
  4. *p<0.05; **p<0.01 versus non-GvHD using 2-sided Wilcoxon rank sum test.
  5. aGvHD, acute graft versus host disease; CXCL9, chemokine C-X-C motif ligand 9; GF, graft failure; GvHD, graft versus host disease; HSC, haematopoietic stem cell; SD, standard deviation.

Conclusions: Elevated IFNγ activity, as measured by serum CXCL9 levels, was observed in all patients with GF, and commonly observed in patients with GvHD and in patients with impaired HSC proliferation. Interestingly, the highest mean levels of CXCL9 were observed in patients with GF, more severe GvHD and GvHD with multiorgan involvement. These data suggest a role for IFNγ in these conditions and support the investigation of potential treatment or preventive strategies targeting IFNγ in patients undergoing HSCT.

Clinical Trial Registry: ClinicalTrials.gov NCT04494061.

Disclosure: Régis Peffault de Latour is a consultant to Alexion, Amgen, MSD, Novartis and Pfizer, and has received research grants from Alexion, Amgen, Novartis and Pfizer. Pietro Merli is a consultant for Sobi and Jazz Pharmaceutical. Emmanuel Monnet and Malin Löfqvist are employees and shareholders of Swedish Orphan Biovitrum (Sobi). Franco Locatelli discloses honoraria from Amgen, Novartis and Sobi, and consulting fees from Amgen. This study was funded by Sobi.

12: Graft-versus-host Disease – Clinical

P254 DELETIONS OF COMPLEMENT FACTOR H-RELATED GENES CFHR-1 AND CFHR-3 AND ACUTE GRAFT-VERSUS-HOST DISEASE AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION

Lars Klingen Gjærde 1, Maja Milojevic1, Niels Smedegaard Andersen1, Lone Smidstrup Friis1, Brian Kornblit1, Marietta Nygaard1, Søren Lykke Petersen1, Ida Schjødt1, Jens D. Lundgren1,2, Daniel D. Murray1, Henrik Sengeløv1,2

1Rigshospitalet, Copenhagen, Denmark, 2University of Copenhagen, Copenhagen, Denmark

Background: Complement factor H-related (CFHR) proteins are important regulators of the alternative complement pathway, which is involved in the development of acute graft-versus-host disease (GvHD) after allogeneic hematopoietic cell transplantation (HCT). Around 20% of the population carry deletions spanning the CFHR-1 and CFHR-3 genes (CFHR1/3) due to non-allelic homologous recombination, resulting in decreased levels of the CFHR-1 and CFHR-3 proteins in plasma. We aimed to investigate the association between CFHR1/3 deletions and acute GvHD.

Methods: We included 305 adult patients who underwent an allogeneic HCT at Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark between 2015 and 2018. All patients were genotyped using an Affymetrix Axiom SNP array (N= 287) or an Illumina Infinium Global Screening array (N= 18). CFHR1/3 deletions were inferred from the total number of predictive alleles across 16 single-nucleotide polymorphisms shown to tag the deletion. Patients with total predictive allele frequencies at 0%, 50%, and 100% and were defined as having two, one, or no CFHR1/3 deletion, respectively. Multivariable cause-specific Cox regression models were used to estimate associations between having CFHR1/3 deletions and grade II–IV acute GvHD and other outcomes, adjusted for recipient age, conditioning intensity, donor type, stem cell source and use of anti-thymocyte globulin.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (36)

Results: The median age of patients was 57 years (Q1, Q3: 45, 65 years). Patients were mostly transplanted for acute myeloid leukemia (33%) using peripheral blood stem cells (84%) from an HLA-matched unrelated donor (69%). Non-myeloablative conditioning (mainly fludarabine and 2 Gy TBI) was used in 53% of patients. Heterozygous deletions of CFHR1/3 were identified in 95 (31%) patients, and homozygous deletions of CFHR1/3 were found in 16 (5%) patients. Patients with homozygous deletions had an increased risk of grade II–IV acute GvHD (44% at day +110, 95% confidence interval [CI]: 18%, 69%) compared to patients with heterozygous deletions (22% at day +110, 95% CI: 14%, 30%) and patients with no deletions (26% at day +110, 95% CI: 20%, 32%), but this difference was not statistically significant (p= 0.22, Figure). In the multivariable Cox regression model, patients with homozygous deletions, compared with those with no deletions, had a hazard ratio of grade II–IV acute GvHD of 2.10 (95% CI: 0.95, 4.69; p= 0.07). For patients with who were heterozygous for the deletion, the hazard ratio was 1.00 (95% CI: 0.60, 1.68; p= 1.00). No associations were found for overall survival (p= 0.42 for heterozygotes; p= 0.24 for homozygotes), non-relapse mortality (p= 0.95 for heterozygotes; p= 0.33 for homozygotes), or relapse (p= 0.56 for heterozygotes; p= 0.16 for homozygotes).

Conclusions: Heterozygous CFHR1/3 deletions were not associated with an increased risk of acute GvHD or other outcomes after allogeneic HCT. Patients homozygous for the CFHR1/3 deletion were observed to have a higher risk of acute GvHD, but due to the rare occurrence of homozygous deletions, this signal needs further investigation in a larger cohort.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P255 EXTERNAL VALIDATION OF SAINT LOUIS SCORE TO PREDICT ACUTE GRAFT-VERSUS-HOST DISEASE SEVERITY

Alessandro Criscimanna 1,2, Annalisa Ruggeri1, Elisa Diral1, Raffaella Greco1, Alessandro Bruno1, Luca Canziani1,2, Sarah Marktel1, Francesca Farina1, Sara Mastaglio1, Daniela Clerici1, Simona Piemontese1, Andrea Acerbis1,2, Chiara Secco1, Magda Marcatti1, Matteo Giovanni Carrabba1, Massimo Bernardi1, Consuelo Corti1, Jacopo Peccatori1, Fabio Ciceri1,2, Maria Teresa Lupo-Stanghellini1

1IRCCS San Raffaele Scientific Institute, Milan, Italy, 2Vita-Salute San Raffaele University, Milano, Italy

Background: Acute Graft-versus-Host disease (aGvHD) still represents a major cause of morbidity and mortality after allogeneic stem cell transplantation (HCT). Identifying as early as possible each patient’s disease trajectory may foster a prompt and tailored intervention. Mount Sinai aGvHD international consortium proposed an algorithm probability score based upon two biomarkers – ST2 and REG3alpha - to predict patient outcomes. This showed promising results, especially when complemented with clinical criteria. Hence, bed-side clinical parameters remain pivotal in aGvHD risk stratification. A new simplified clinical model was developed by Hospital St. Louis (HSL) group considering as high-risk those patients with initial grade III-IV aGvHD, liver involvement or age>50 yo.

Methods: We conducted a retrospective single-center study, involving all consecutive HCT performed in our center between January 2016 and December 2022.

Overall, 477 individuals were recorded, with the most common diagnosis being acute myeloid leukemia, and the median age at HCT being 55 years. HCT was carried-out from an HLA identical, unrelated, or haploidentical donor in 17.8%, 56%, and 26.2% of cases respectively. Transplant conditioning intensity was low in 30 cases, intermediate in 198 and high in 248 cases. GvHD prevention was mostly PTCy-based except for 18 patients. Additionally, 21 patients received ATG as well.

Results: With a median follow up of 31 months, 2 years overall survival (OS) for the entire cohort was 69 + 2%. aGvHD was diagnosed in 209 patients and this population was used for the validation of the HSL score. aGVHD at presentation had involvement of skin in 176, liver in 24 and gut in 75 patients. 71 patients developed grade III-IV aGVHD. Steroid refractoriness occurred in 56 patients. Individuals with Minnesota high-risk score were 56.

Given the difference in the median age at HCT among our pts cohort and the original HSL cohort (55 y vs 45 y) we analyzed age as a continuous variable and then identified the best threshold using the maximization of the logrank test (58 years for our cohort).

Age as continuous variable, as well as binary variable of 58 years, liver involvement and grade III-IV aGVHD were predictive of disease severity and showed an impact in overall survival.

OS was 83 + 5% versus 55 + 5%, p<0.001, in patients with low risk and high-risk HSL score, respectively. In adjusted multivariate analysis, increase in patient age (HR 1.02, 95%CI 1.001-1.04, p=0.03), liver involvement (HR 3.39, 95%CI 1.91-6.02, p<0.001) and gut involvement (HR 2.5, 95%CI 1.53-4.12, p<0.001) were independently associated with lower OS.

Conclusions: We confirm that the HSL score risk is a valid clinical score to assess high-risk patients for severe complication and mortality. Reliable clinical algorithms for GvHD severity stratification are particularly relevant where access to biomarkers is not yet fully available. In recent years, with the advent of reduced-toxicity conditioning regimens as well as the improvement in post-transplant strategies, HCT has become a treatment option even for older patients. Treating age as a continuous variable provide a more detailed and nuanced representation of the data with a higher level of precision.

Clinical Trial Registry: n.a.

Disclosure: Authors have no conflicts of interest to declare with respect to the topic of the abstract.

12: Graft-versus-host Disease – Clinical

P256 EVALUATION OF RISK FOR BRONCHIOLITIS OBLITERANS SYNDROME AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION WITH MYELOABLATIVE CONDITIONING REGIMENS

Kristina Maas-Bauer 1, Paraschiva Rassner1, Kristin Walther1, Claudia Wehr1, Reinhard Marks1, Ralph Wäsch1, Robert Zeiser1, Jürgen Finke1, Jesus Duque Afonso1

1University of Freiburg, Faculty of Medicine, Freiburg, Germany

Background: Lung function deterioration contributes to treatment-related morbidity and mortality in patients after allogeneic hematopoietic cell transplantation (allo-HCT). Better understanding of impaired lung function including bronchiolitis obliterans syndrome (BOS) as chronic manifestation of graft-versus-host disease (GVHD) might improve outcomes of patients after allo-HCT. In the present study, we evaluated the risk of developing BOS in patients after receiving myeloablative conditioning (MAC) regimens.

Methods: In this retrospective analysis, patients with acute myeloid leukemia (AML), myelodysplastic syndrome (MDS) and myeloproliferative syndromes (MPN) undergoing allo-HCT at our center between 1998 and 2019 were included. We focused on (1) adult (aged > 18 years) patients who received MAC conditioning with Busulfan/Cyclophosphamid (BuCy), Busulfan/ Fludarabin (FluBu4) or Thiotepa/Busulfan/Fludarabine (TBF MAC), (2) first allo-HCT from a matched sibling donor (MSD) or matched unrelated donor (MUD 10/10) or 9/10 mismatched unrelated donors (MMUD), (3) with an unmanipulated peripheral blood stem cell graft. Cyclosporine-based GVHD prophylaxis was combined with methotrexate or mycophenolate mofetil. In vivo T-cell depletion was performed with antithymocyte globulin or alemtuzumab. All clinical data were prospectively collected and retrospectively analyzed.

Results: Indication for allo-HCT were AML in 93.1% and MDS/MPN in 6.9% in the BuCy cohort and AML in all patients in the FluBu4 and TBF MAC cohort. Prior to allo-HCT, 175 patients received a conditioning with BuCy, 29 patients with FluBu4 and 37 patients with TBF MAC. The median age of patients at allo-HCT was 43 years with BuCy, 46 years with FluBu4 and 39 years with TBF MAC. Patients receiving BuCy had more frequently lung diseases prior allo-HCT (42%) compared to patients receiving FluBu4 (7%) and TBF MAC (19%). Patients treated with BuCy prior allo-HCT had the highest prevalence of current or previous smokers (30%) compared to patients in the other two groups (FluBu4, 10%; TBF MAC 5%). Due to the retrospective character of the study we also observed significant differences in the characteristics of the treatment cohorts including GvHD prophylaxis, HCT-CI score, EBMT risk score and GvHD incidence that warrant further investigations.

The cumulative incidence of BOS using the NIH diagnosis consensus criteria at 2 years after allo-HCT was lower in patients that received FluBu4 (8%) compared to patients that received either BuCy (23%) or TBF MAC (19%) as conditioning regimen, although these differences were not statistically significant (p=0.07). In multivariate analysis, risk factors for the development of BOS as forced expiratory volume during the first second (FEV1) < median (99)% of predicted (HR=2.39, p=0.004) prior to allo-HCT and CMV positivity of the patient (HR=2.11, p=0.014) were identified. In contrast, total lung capacity (TLC) <80% predicted (HR=0.12, p=0.02) and in vivo T-cell depletion (HR=0.29, p=0.001) were protective.

Conclusions: In summary, we found a trend for lower incidence of BOS in patients conditioned with FluBu4 compared to conditioning regimen with BuCy or TBF MAC and identified FEV1<99% of predicted prior allo-HCT and CMV patient positivity as risk factors for developing BOS and TLC<80% of predicted and in vivo T-cell depletion as protective factors in this cohort of patients.

Disclosure: Conflict of interest.

RZ received speaker fees from Novartis, Incyte, Sanofi, MNK, medac, Neovii.

12: Graft-versus-host Disease – Clinical

P257 THE MACROPHAGE ACTIVATION MARKER SCD163 AND ACUTE AND CHRONIC GRAFT-VERSUS-HOST DISEASE IN ALLOGENEIC HSCT

Maja Munk Kristoffersen 1, Elisa Peen1, Henning Grønbæk2, Holger Jon Møller2, Henrik Sengeløv1, Klaus Müller1, Lars Klingen Gjærde1, Katrine Kielsen1

1Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark, 2Aarhus University Hospital, Aarhus, Denmark

Background: Graft-versus-Host disease (GvHD) is a major cause of non-relapse mortality and long-term morbidity after allogeneic hematopoietic stem cell transplantation (HSCT), why it is of great interest to find biomarkers for the prediction of patients at risk of GvHD, which may lead to better prophylaxis and improved outcomes of allogeneic HSCT.

CD163 is a scavenger receptor for hemoglobin and haptoglobin, which is expressed by macrophages and subsets of dendritic cells and is shed in a soluble form (sCD163) upon cellular activation. sCD163 has previously been associated with acute and chronic GvHD after pediatric HSCT (Hergel et al., BMT, 2023). In this study, we aimed to study this association in a cohort of adult patients undergoing HSCT.

Methods: We included 153 adults who underwent allogeneic HSCT at Rigshospitalet, Copenhagen University Hospital between 2010-2018. Median age was 45.8 years (range 16.5-70.9). Diagnoses included AML (n=54), ALL (n=31), MDS (n=40), other malignancies (n=13) and benign disorders (n=15). Donors were MSD (n=39), MUD (n=102) or MMUD (n=12). Stem cell source was PBSC (n=91) or BM (n=62). Conditioning was either TBI-Cy/VP16 (n= 74), FluTreo (n=59) or other chemotherapy (n=20).

Plasma levels of sCD163 were measured by ELISA prior to conditioning (n=152), at day of transplantation (n=37) and at days +7 (n=37), +14 (n=37), +21 (n=32), +28 (n=87), and +90 (n=29) post-HSCT. 30 healthy adults with a median age of 27.1 years (range 20.9-39.5) were included as controls.

Results: Plasma levels of sCD163 were significantly higher in patients before conditioning compared to healthy controls (median 2.3 mg/L vs. 1.7 mg/L, p<0.0001). After HSCT, sCD163 levels rose and peaked at day +21 (3.3 mg/L, p<0.0016) and then gradually declined compared to pre-conditioning. Post-transplant sCD163 levels were not associated with TBI-based conditioning, graft type or donor type (all p>0.05).

Acute GvHD occurred in 48.4% of patients (n=74), 60 of these patients developed aGvHD grade II-IV. sCD163 levels were significantly elevated in aGvHD grade II at day +28 compared to grade 0-I (3.4 mg/L vs. 2.5 mg/L p<0.05), but not statistically different from grade III-IV. Furthermore, patients with steroid-refractory aGvHD (n=22) had significantly higher sCD163 levels at day +28 compared with patients with steroid-responsive aGvHD (3.8 vs. 2.9 mg/L, p=0.032).

Seventy-five patients (49%) developed cGvHD with median onset at day +255 (range 73-1830 days). 55 of these had extensive cGvHD, and 39 patients had aGvHD prior to cGvHD. sCD163 at day +28 were significantly elevated in patients with extensive cGvHD compared to limited cGvHD (3.2 mg/L vs. 2.2 mg/L, p=0.027).

Conclusions: Our findings suggest that elevated levels of the macrophage activation marker sCD163 early after allogeneic HSCT are associated with later severe alloreactivity such as steroid-refractory aGvHD and extensive cGvHD. Further analyses are ongoing to investigate whether monitoring of sCD163 can identify patients in high risk of extensive chronic GvHD and thus guide the immunosuppressive treatment given as GvHD prophylaxis.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P258 ETIOLOGY OF DIARRHEA IN PATIENTS UNDERGOING COLONOSCOPIC BIOPSIES POST ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT FOR SUSPECTED ACUTE GUT GRAFT VERSUS HOST DISEASE

Niket Mantri 1, Sachin Punatar1, Mukta Ramadwar1, Prachi Patil1, Shaesta Mehta1, Aditya Kale1, Shridhar Epari1, Sridhar Sundaram1, Koustubh Shekar1, Anant Gokarn1, Sumeet Mirgh1, Akanksha Chichra1, Nishant Jindal1, Lingaraj Nayak1, Libin Mathew1, Shesheer Kumar2, Rachana Tripathi2, Bhausaheb Bagal1, Navin Khattry1

1Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Mumbai, India, 2Huwel Labs, Hyderabad, India

Background: We routinely perform colonoscopy in patients with suspected lower-gut acute graft-versus-host disease (aGVHD) to distinguish GVHD from its mimics (viz. CMV colitis). We conducted this retrospective analysis to evaluate etiologies of diarrhoea by colonoscopy and biopsy for patients with suspected lower-gut GVHD post allogeneic hematopoietic cell transplant (AHCT).

Methods: This is a single-centre retrospective analysis of patients who underwent colonoscopy for suspected lower-gut aGVHD between May 2016-June 2023. Prior to colonoscopy, stool infective workup was done to rule out C.difficile, norovirus, rotavirus, astrovirus & adenovirus (AdV). Steroids could have been started for clinical suspicion of aGVHD prior to colonoscopy. At each colonoscopy, targeted biopsies were taken if abnormal findings were present. Additionally, random biopsies were taken from macroscopically normal looking mucosa for histopathology and tissue viral PCRs for CMV, EBV and AdV from different sites of colon (caecum to rectum). For tissue PCRs - lower limit of detection of CMV, EBV and AdV were 65, 100 and 250 IU/ml respectively. The initial treatment of histologically proven lower-gut aGVHD was 1-2 mg/kg methylprednisolone with oral budesonide. If biopsy showed concomitant CMV inclusion bodies (proven CMV colitis) or PCR copies >103IU/ml (possible CMV colitis), then ganciclovir 5mg/kg BD was started at physician’s discretion. If AdV-PCR copies were ≥104IU/ml, cidofovir 5mg/kg weekly for 2 weeks followed by 2 doses every 2 weeks was planned. No patient was treated for EBV-PCR positivity. If biopsy did not show GVHD, then steroids were rapidly tapered and stopped. Our primary objective was to identify the proportion of patients with both histologically confirmed gut GVHD and concomitant proven/possible viral colitis. We also analysed the correlation of colonoscopic findings with histology.

Results: Eighty-five colonoscopies were performed in 62 patients. The demographic parameters are shown in Table 1. Median time to colonoscopy from onset of diarrhoea was 4 days. Systemic steroids were started prior to colonoscopy in 51 episodes (60%). Colonoscopic findings, histology & results of tissue PCRs for CMV, EBV and AdV are shown in Table 1. Overall, 54 (63%) colonoscopic biopsies had histologically confirmed GVHD. Among these 54, 8 biopsies (15%) had proven CMV colitis. With respect to colonoscopic appearance (n=83), aGVHD was confirmed histologically in 58%(n=32/55) biopsies with macroscopically normal mucosa, 79% (11/14) with erythema alone, 90% (9/10) with erythema and ulceration and 25%(n=1/4) with ulcerations alone. Amongst remaining 30 biopsies without GVHD, 50%(n=15) had possible CMV colitis and 16%(n=5) adenoviral colitis. Amongst these patients who received additional antivirals, two-third patients responded.

Baseline characteristics and transplant details

Patient demographics

N=62, (%)

Patient age, median (range)

30 (3 – 57) years

Gender

Males

45 (72%)

Females

17 (27%)

Donor age, median (range)

31 (4 - 60) years

Donor gender

Males

39 (63%)

Females

23 (37%)

Female donor to male recipients

16 (26%)

Diagnosis

AML / MDS

15 (24%)

ALL / MPAL

22 (35%)

CML and other CMPD

20 (32%)

Lymphomas

5 (8%)

Type of transplant

MRD

39 (63%)

MUD

7 (11%)

Haplo-identical transplants

16 (26%)

Conditioning regimen intensity

Full intensity

7 (11%)

RIC

55 (89%)

GVHD prophylaxis

CNI + MTX / MMF + /- ATG

46 (74%)

PTCy based

16 (26%)

Number of colonoscopies

1

45 (73%)

2

13 (21%)

3 or more

4 (6%)

Findings of colonoscopy, histology and PCRs

Details

N=85 (%)

Colonoscopic findings

Normal findings

55 (65%)

Erythema (patchy or diffuse) alone

14 (17%)

Ulcerations alone

4 (5%)

Erythema + ulceration

10 (12%)

Missing data

2 (2%)

Histology findings

GVHD alone

46 (54%)

Non-specific colitis

16 (19%)

Inclusion bodies (alone)

1 (1.2%)

GVHD + inclusion bodies

8 (9%)

Other findings (mucosal atrophy)

1 (1.2%)

Normal histology

13 (15%)

Mucosal viral infection (by PCR) (n=84)

CMV

40 (47%).

Median 1.7 x 104 IU/ml

Adv

12 (14%)

Median 4.4 x 104 IU/ml

EBV

22 (26%)

Median 1.9 x 103 IU/ml

  1. Abbreviations – AML- acute myeloid leukemia, MDS- myelodysplastic syndrome, ALL acute lymphoblastic leukemia,
  2. MPAL-mixed phenotype acute leukemia, CML-chronic myeloid leukemia, CMPD-chronic myeloproliferative disorder, MRD- match related donor, MUD-match unrelated donor, RIC- reduced intensity conditioning, CNI-calcineurin inhibitor, MTX- methotrexate, MMF-mycophenolate mofetil, ATG- antithymocyte globulin, PTCy-post transplant cyclophosphamide. GVHD graft versus host disease, PCR-polymerase chain reaction, CMV-cytomegalo virus, Adv-adenovirus, EBV-epstein-barr virus

Conclusions: In suspected gut GVHD, more than one-third biopsies did not have histological evidence of aGVHD. This reiterates the need for colonoscopy which would thereby prevent unnecessary steroids. Half of the patients with macroscopically normal mucosa had proven aGVHD, thereby justifying the role of random biopsies. Concomitant proven CMV colitis was present in 15% of those with biopsy proven aGVHD. Even in those with normal histology, possible viral colitis is present in half of the biopsies. Therefore, it may be worthwhile doing tissue viral PCRs for all patients undergoing colonoscopic biopsies.

Disclosure: None.

12: Graft-versus-host Disease – Clinical

P259 ABATACEPT ADDED TO POST TRANSPLANT CYCLOPHOSPHAMIDE-BASED GVHD PROPHYLAXYS IN ALLOGENEIC HEMATOPOYETIC STEM CELL TRANSPLANTATION: ONE SINGLE CENTER EXPERIENCE

Paula Fernández- Caldas González 1,2, Rebeca Bailén1,2, Pilar Lancho1, Ignacio Gómez-Centurión1,2, Lucía Castilla1, Cristina Muñoz1, Santiago Sabell1, Javier Anguita1,2,3, Mi Kwon1,2,3

1Hospital General Universitario Gregorio Marañón, Madrid, Spain, 2Instituto de Investigación Sanitaria Gregorio Marañón, Madridspain, Spain, 3Universidad Complutense de Madrid, Madrid, Spain

Background: Graft-versus-host disease (GVHD) is still one of the main causes of morbidity and mortality after an allogeneic stem cell transplant (allo-SCT). Abatacept (Aba) is a human cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4) antibody that attenuates T-cell activation. Aba has recently been approved by the FDA for GVHD prophylaxis, in association with methotrexate (MTX) and a calcineurin inhibitor (CNI). However, few experiences have reported its results associated to high dose post-transplant cyclophosphamide (PTCy) with additional immunosuppression. Since February 2020, we incorporated Aba as additional prophylaxis in all patients who underwent haploidentical HSCT with low/intermediate DRI score and at least one of the following criteria: HCT-CI score ≥3 and/or age ≥55 years. Additionally, we considered individually its use in matched donor HSCT in patients with very high risk of GVHD. The purpose of our study was to compare the results of Aba as additional prophylaxis to PTCy based prophylaxis.

Methods: We performed a retrospective observational study including two cohorts. Cohort 1 (Aba-PTCy) included patients who underwent an allo-SCT between February 2020 and November 2022 treated with Aba added to PTCy-based prophylaxis. Cohort 2 (PTCy) included patients with matched characteristics who underwent an allo-SCT between March 2015 and October 2019 treated with PTCy-based prophylaxis without Aba.

Results: Twenty three patients were included in Cohort 1 (Aba-PTCy) and 24 in cohort 2 (PTCy). Baseline characteristics were balanced between cohorts (Table 1). All patients underwent a reduced intensity conditioning (RIC), received peripheral blood as source, and CNI and MMF as additional immunosuppression. In Cohort 1, Aba was administered in days +5, +14, +28. No adverse events were observed associated to Aba infusion. Cumulative incidence (CI) of grade II-IV aGVHD showed a trend towards a lower rate in the Aba-PTCy cohort compared to the PTCy cohort (9% vs. 45% at day 100, and 34% vs. 50% at day 180, p=0.2). Grade III-IV aGVHD at day 100 was significantly lower in the Aba cohort (0% vs. 16%) but similar at day 180 (16% vs. 13%, p=0.6). CI of cGVHD at days +180 and +365 were higher in the Aba group (18% vs. 4% and 44% vs. 12% (p=0.06)). CI of cGVHD moderate-severe at 1 year was 30% vs. 8% (p=0.2). With a median follow-up of 12 months for Aba-PTCy and 39 months for PTCy cohorts, estimated 1-year OS and EFS were 82% vs. 87% (p=0.7) and 78% vs. 87% (p=0.5), respectively. NRM was 8%. Both rates of CMV and EBV reactivation were similar between cohorts.

Aba-PTCy (n=23)

PTCy (n=24)

p

Sex (female, %)

12 (52)

12 (50)

0.8

Age (median, IQR)

63 (47-71)

62 (53-69)

0.15

Diagnosis (n, %):

AML/MDS

10 (42.4)

13 (54.1)

0.9

Others

13 (57.6)

11 (45.9)

Disease risk index high and very high (n, %)

1 (4.3)

7 (29.2)

0.13

HCT-CI score ≥3 (n, %)

19 (82.7)

16 (77.7)

0.1

Prior HSCT (autologous, n, %)

3 (13)

5 (20,8)

0.7

Prior HSCT (allogenic, n, %)

1 (4,3)

1 (4,2)

0.7

Donor (n, %):

HLA identical

4 (17.4)

1 (4.2)

0.06

Haploidentical

19 (82.6)

23 (95.8)

Donor sex (female, n,%)

8 (34.8)

9 (37.5)

0.8

HLA disparity GVHD direction n(%)

9 (45%)

11 (47.8)

Disease status previous HSCT (n, %)

CR

11 (47.8)

14 (58.3)

0.5

PR

3 (15)

5 (20.8)

SD/PD

9 (37.2)

5 (20.9)

IS discontinuation at day + 100(No, n,%)

18 (85.7)

21 (87.5)

0.8

Conclusions: In our experience, the addition of Aba to PTCy-based prophylaxis, using a RIC regimen and PBSC as stem cell source, was safe and provided low rates of acute GVHD at day 100 as compared to PTCy-based prophylaxis alone. However, incidence of chronic GVHD was not reduced. Further studies should be performed to address whether additional doses of Aba could reduce late onset aGVHD and cGVHD.

Disclosure: No conflict of interest.

12: Graft-versus-host Disease – Clinical

P260 SAFETY AND EFFICACY OF THE ROCK2-INHIBITOR BELUMOSUDIL IN CGVHD TREATMENT – A RETROSPECTIVE, MULTICENTER REAL-WORLD DATA ANALYSIS

Silke Heidenreich 1, Katharina Egger-Heidrich2, Jörg P. Halter3, Friedrich Stölzel4, Matthias Edinger5, Wolfgang Herr5, Nicolaus Kröger1, Daniel Wolff5, Francis Ayuk1, Matthias A. Fante5

1University Hospital Hamburg-Eppendorf, Hamburg, Germany, 2University Hospital Carl Gustav Carus, Dresden, Germany, 3Division of Hematology, University Hospital Basel, Basel, Switzerland, Basel, Switzerland, 4Division of Stem Cell Transplantation and Cellular Immunotherapies, University Hospital Schleswig-Holstein, Kiel, Germany, 5University Hospital Regensburg, Regensburg, Germany

Background: Chronic GvHD (cGvHD) is the most significant risk factor for late morbidity and mortality after allogeneic stem cell transplantation (alloHSCT). Treatment options beyond first-line corticosteroids and second-line ruxolitinib are limited. However, the selective ROCK-2 inhibitor belumosudil (KD025), a novel FDA-approved agent, has shown efficacy in two recent phase-2 trials. In this study, we analyzed the clinical benefits of belumosudil in a heavily pretreated patient cohort.

Methods: In this retrospective, multicenter study, safety and efficacy of belumosudil was analyzed in patients treated for refractory cGvHD at the University Medical Centers in Regensburg, Hamburg, Dresden, Kiel and Basel. Organ scoring and response assessment were performed according to the NIH-consensus criteria before start, and at month 1, 3, 6, 9, and 12 thereafter. Adverse events are reported according to the CTCAE criteria (version 5.0). The last-follow-up was on November 30th, 2023.

Results: In total, 21 patients (male, 67%) with a median age of 58 years (range, 26 – 76) were included into the analysis. The most common underlying diseases were acute leukemia (48%) and myeloproliferative neoplasm (29%). cGvHD onset was quiescent and progressive each in 38%, and de novo in 24% of patients. The cohort had undergone a median of 5 prior treatment lines (range, 3 – 13), all patients were refractory to the preceding line, and 18 patients had a history of ruxolitinib treatment. At start of belumosudil, all but one patient had severe cGvHD with a median of 3.5 NIH-defined organ manifestations (range, 1 – 6), and 29% (n=6) had additional atypical cGvHD manifestations. In median, belumosudil (200mg/d in 95% of patients) was started 42 months after alloHSCT (range, 16 – 319) with a treatment duration of 199 days (range, 35 to 619). The best objective response was a partial remission in 10 patients (48%) and stable disease in 11 patients (52%). Response was observed in all organ systems except for the liver. Most patients (8/10 patients) responded within three months after treatment initiation. Steroid dose reduction was achieved in 9 of 14 patients. Four patients with stable disease experienced a reduction in cGvHD symptom burden based on self-assessment and/or medical examination (3 patients with skin softening, one patient with closure of chronic skin ulcers), yet below response according to the NIH-criteria. Treatment-emergent adverse reactions CTCAE grade III-IV occurred in 4 cases and included pneumonia (n=2), retinal detachment, and congestive heart failure. During treatment with belumosudil, one patient with severe pulmonary cGvHD died due to a respiratory infection.

Conclusions: In summary, belumosudil was well tolerated and induced a remarkable response in a heavily pretreated cohort of cGvHD patients within a real-world setting. Further prospective trials are needed to determine the duration of response and the impact on GvHD-related mortality.

Disclosure: The authors declare to have no conflict of interest.

12: Graft-versus-host Disease – Clinical

P261 EFFICACY AND SAFETY OF IBRUTINIB FOR CHRONIC GRAFT-VERSUS-HOST DISEASE: A SYSTEMATIC REVIEW

Damai Santosa1, Daniel Rizky 1, Kevin Tandarto1, Ika Kartiyani1, Vina Yunarvika1, Desta Nur Ewika Ardini1, Budi Setiawan1, Eko Adhi Pangarsa1, Catharina Suharti1

1Diponegoro University/Dr. Kariadi General Hospital, Semarang, Indonesia

Background: Allogeneic hematopoietic cell transplantation (allo-HCT) can be a curative treatment for several types of blood-related conditions, but it may be limited by chronic graft-versus-host disease (cGVHD). The clinical symptoms of cGVHD are caused by a complex immune response involving both B and T cells. Ibrutinib is a medication that inhibits the Bruton’s tyrosine kinase (BTK) pathway, which is activated by the B-cell receptor and controls B-cell survival. By blocking both BTK and inhibitor of interleukin-2 inducible T-cell kinase (ITK), ibrutinib has shown promise as a treatment for cGVHD. The aim of this systematic review is to determine the efficacy and safety of ibrutinib for cGVHD patients.

Methods: We incorporated search engines from PubMed, Embase, Cochrane Library, Scopus, Web of Science, and ClinicalTrials.gov. The study was performed following the guidelines of the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) 2020 and AMSTAR (Assessing The Methodological Quality of Systematic Review). We used Risk of Bias- 2 (RoB-2) tool for assess the risk of bias in randomized controlled studies (RCTs) and Newcastle Ottawa Scale (NOS) for observational and open-label studies.

Results: A total of 7 studies were included in this study consisted of four open-label studies, two retrospective cohort studies, and one RCT study. These studies compared Ibrutinitib with standard therapies. Two studies investigated the pediatric population, and five studies investigated the adult population. Overall, these studies reported the overall response rate (ORR) of Ibrutinib for cGVHD were 54%-78%. The results showed that in pediatric patients, the ORR were 54-78%. The results also showed that in adult patients, the ORR were 67%-76%. The most common adverse effects observed across the seven studies included pyrexia, diarrhea, abdominal pain, cough, nausea, stomatitis, vomiting, headache, bleeding and bruising, infection, muscle aches, fatigue, oral bleeding, elevated transaminases, lower gastrointestinal bleeding, persistent dizziness, sepsis, pneumonia, reduced platelet count, exhaustion, sleeplessness, peripheral edema, and fatigue.

Conclusions: The majority of studies have indicated that Ibrutinib exhibits a high ORR and provides long-lasting responses, while also having manageable side effects.

Disclosure: No conflict of interest in this study.

12: Graft-versus-host Disease – Clinical

P262 WHEN SHOULD RUXOLITINIB BE CEASED? A SINGLE CENTRE, RETROSPECTIVE, REAL-WORLD ANALYSIS OF RUXOLITINIB IN GRAFT VS HOST DISEASE POST ALLOGENEIC STEM CELL TRANSPLANT

Harshita Rajasekariah 1,2, Barbara Withers1,2, Keith Fay1,3, Sam Milliken1,2, Nada Hamad1,2, John Moore1,2, Jacinta Perram1,2

1St Vincent’s Hospital, Sydney, Australia, 2University of New South Wales, Sydney, Australia, 3Royal North Shore Hospital, Sydney, Australia

Background: Ruxolitinib is an effective therapy for graft versus host disease (GVHD), with overall response rates (ORR) of 62% at 28 days in acute GVHD (aGVHD) and 49.7% at 6 months in chronic GVHD (cGVHD) as demonstrated in the pivotal REACH2 and REACH3 trials respectively. Efficacy of ruxolitinib outside of the clinical trial setting has only recently been reported in a few small international studies, with variable response rates. We reviewed our centre’s data and report our real-world outcomes of ruxolitinib use in GVHD.

Methods: We retrospectively analysed patients at St Vincent’s Hospital Sydney, Australia with GVHD managed with ruxolitinib between 2019 and February 2023. The following outcomes were assessed: ORR, time to clinical response, treatment duration, immunosuppression tapering, concurrent infection, disease relapse and overall survival.

Results: 41 patients received ruxolitinib for GVHD. Baseline characteristics are summarised in Table 1. An ORR of 93% was seen in our cohort. All patients with aGVHD achieved a response, with median time to response of 1 month (range 7 days- 20 months). 88% of patients with cGVHD achieved at least a partial response, with a median time to response of 7 months (range 1 - 28 months).

Median duration of ruxolitinib therapy was 11 months (range 3-36 months) in patients with aGVHD and 16 months (range 1-51 months) in cGVHD. Ruxolitinib was weaned in 37% of patients following improvement in GVHD, and concurrent immunosuppressive agents were weaned in 17% of patients. Of patients with aGVHD, 21% discontinued therapy following a complete response (CR), compared to 8% of patients with cGVHD. Of these patients, 2 patients developed recurrent cutaneous cGVHD following cessation. Ruxolitinib was also ceased due to infection (2%), cytopenias (10%), disease relapse (10%), intolerance (2%) and drug interactions (2%). 63% of patients continued ruxolitinib at last review, with 48% of these patients maintaining CR.

Infection occurred in 39% of patients, including respiratory infections (30%, predominantly RSV or rhinovirus), shingles (12%) and CMV reactivation (10%). Diarrhoeal illness with campylobacter and astrovirus occurred in 5% and 1 patient developed recurrent Escherichia Coli bacteraemia.

Disease relapse occurred in 20% of patients, with 75% taking ruxolitinib at time of relapse. 80% were alive at a median follow up of 16 months (range 1-48 months). Of those who died, 57% were taking ruxolitinib at the time of death. Cause of death included infection (7%), progressive disease (5%), secondary malignancies (5%) and bowel obstruction secondary to GVHD (2%).

Table 1 – Baseline Characteristics

Characteristic

Subgroup

Number of patients n=41 (%)

Age

59 years (range 33-81 years)

Gender

Male

26 (63%)

Female

16 (39%)

Transplant indication

Acute leukaemia

25 (61%)

- Acute myeloid leukaemia

16 (39%)

- Acute lymphoblastic leukaemia

8 (20%)

- Undifferentiated

1 (2%)

Myelofibrosis

3 (7%)

Myelodysplastic syndromes (MDS)

4 (10%)

MDS

2 (5%)

MDS/MPN

2 (5%)

Non-Hodgkin Lymphoma (NHL)

7 (17%)

Hodgkin lymphoma

2 (5%)

Conditioning

Reduced intensity

30 (73%)

Myeloablative

9 (22%)

Unknown

2 (5%)

Donor source

Matched sibling

12 (29%)

Matched unrelated donor (MUD)

23 (56%)

- Full MUD

8 (20%)

- Permissive mismatch

9 (22%)

- Mismatched MUD

6 (15%)

Haploidentical

5 (12%)

GVHD prophylaxis

Ciclosporin

29 (71%)

- With concurrent ATG

14 (34%)

- With concurrent methotrexate

21 (51%)

Tacrolimus with mycophenolate

6 (15%)

- With PTCy

4 (10%)

Unknown

6 (15%)

GVHD

Acute

15 (37%)

- Cutaneous

11 (27%)

- Gastrointestinal

2 (5%)

- Hepatic

1 (2%)

Chronic

26 (63%)

- Cutaneous

13 (32%)

- Gastrointestinal

3 (7%)

- Hepatic

3 (7%)

- Sicca syndrome

4 (10%)

- Lung

3 (7%)

- Joints

1 (2%)

Previous treatments

Systemic steroids

37 (90%)

Ciclosporin

22 (54%)

Tacrolimus

9 (22%)

Mycophenolate

20 (49%)

Other

- ECP

2 (4%)

- PUVA

1 (2%)

- Etanercept

3 (7%)

- Thalidomide

1 (2%)

Prior lines of therapy

1

4 (10%)

2

20 (48%)

3

11 (27%)

4

2 (5%)

5

2 (5%)

6

1 (2%)

Nil recent lines

1 (2%)

Concurrent immunosuppression used with ruxolitinib

Nil

2 (5%)

1 agent

9 (22%)

2 agents

25 (61%)

3 agents

5 (12%)

Conclusions: Our analysis demonstrates excellent clinical efficacy of ruxolitinib for GVHD, supporting the REACH2 and REACH3 outcomes in a real-world setting. A large number of patients had cutaneous GVHD, which may explain the superior outcomes reported. A large proportion of patients remained on ruxolitinib despite CR, reflecting the lack of evidence guiding withdrawal of therapy. Protracted therapy may increase the risk of infection, relapse and secondary malignancy, highlighting the need for further evidence to guide clinical practice around withdrawal of ruxolitinib for GVHD in CR.

Disclosure: Nil disclosures.

12: Graft-versus-host Disease – Clinical

P263 CLINICAL RESPONSE AND IMMUNOLOGICAL RECOVERY IN PATIENTS WITH STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST DISEASE (AGVHD) TREATED WITH THE ASSOCIATION OF EXTRACORPOREAL PHOTOPHERESIS (ECP) AND RUXOLITINIB

Chiara Marcon1,2, Chiara Savignano1, Eleonora Toffoletti 1,2, Angela Michelutti1, Margherita Cavallin1, Giovanni Barillari1, Renato Fanin1,2, Francesca Patriarca2,1

1ASUFC - Azienda Sanitaria Universitaria del Friuli Centrale, Udine, Italy, 2DAME, University of Udine, Udine, Italy

Background: The association of Ruxolitinib and ECP has been reported to be a feasible salvage treatment for steroid-refractory aGVHD; however, the potential benefit over Ruxolitinib alone and the effects on immunological recovery have not been fully investigated.

Methods: We retrospectively analysed steroid-refractory aGVHD patients treated with ECP or with the ECP and Ruxolitinib association in our center from January 2018 to July 2023. We compared treatment efficacy and recovery of lymphocyte subpopulations by flow-cytometry in the 2 treatment groups. Finally, we evaluated induction of apoptosis by 7-AAD flow-cytometry in the apheresis product at baseline before ECP irradiation and 48 hours after irradiation.

Results: A total of 25 steroid-refractory aGVHD patients were analysed: 13 in the ECP group and 12 in the Ruxolitinib plus ECP group. Grade III-IV aGVHD was present in 5 of 12 patients (42%) in the ECP plus Ruxolitinib group and in 4 of 9 patients (44%) in the ECP group (p=0.57). Liver and/or gut were involved in 9 of 12 patients (75%) in the ECP plus Ruxolitinib group and in 8 of 13 patients (61%) in the ECP group (p=0.47). Overall complete and partial responses were achieved in 9 (36%) and 10 patients (40%), respectively, without significant differences between the 2 treatment groups. Steroids were tapered in all responsive patients of the both groups.

We evaluated the recovery of the lymphocyte subpopulations during treatment in 12 patients belonging to the ECP group (6 patients evaluated, 14 tests performed) and to the Ruxolitinib plus ECP group (6 patients examined, 10 tests performed). Peripheral blood absolute white blood cell count, as well as absolute lymphocyte and CD3+ counts were significantly lower in the Ruxolitinib plus ECP group (p=0.022, 0.006 and 0.030, respectively). However, CD4+ cells percentage was higher in the Ruxolitinib plus ECP group compared to the ECP group (median 28.2% vs 18.3% respectively, p=0.039), while CD8+ cells percentage did not significantly differ in the two treatment groups (median 23.3% vs 36.5% respectively, p=0.082). Furthermore, ratio of T-regulatory lymphocytes to absolute lymphocyte count and to CD8+ lymphocyte count were significantly higher in the Ruxolitinib plus ECP group (p=0.030 and 0.037, respectively). No differences in the absolute counts of both RA+ and RA- T-regulatory subpopulations were reported. The results of immunological recovery were reported in Table 1.

Lastly, we evaluated induction of apoptosis in 14 out of 25 patients: patients who achieved complete response (CR) of both treatment groups showed an higher apoptotic CD3+ cells percentage in the apheresis product at baseline (median 0.13% in CR patients, 0.05% in patients not achieving CR; p=0.012).

Feature

Ruxolitinib plus ECP group

ECP group

P-Value

WBC (absolute count)

2600.00 (850.00-17620.00)

4843.00 (2100.00-10210.00)

0.022

Lymphocyte (absolute count)

196.00 (49.00-1305.00)

870.00 (201.00-4432.00)

0.006

CD3+ (absolute count)

76.16 (30.38-1174.50)

435.00 (35.34-4095.17)

0.030

CD3+ (%)

62.00 (27.20-94.00)

77.00 (15.50-93.00)

0.505

CD4+ (%)

28.20 (6.70-59.00)

18.30 (2.80-45.00)

0.039

CD8+ (%)

23.30 (4.10-80.00)

36.50 (11.90-86.50)

0.082

TregRA+ (absolute count)

0.60 (0.00-2.90)

0.50 (0.00-10.80)

0.954

TregRA + /CD3+

0.00 (0.00-0.10)

0.00 (0.00-0.01)

0.182

TregRA- (absolute count)

2.60 (0.10-12.40)

3.80 (0.00-11.10)

0.750

TregRA-/CD3+

0.04 (0.00-0.16)

0.01 (0.00-0.03)

0.063

TotalTreg (absolute count)

3.70 (0.20-13.50)

5.00 (0.50-11.10)

0.459

Treg/Lymphocyte

0.03 (0.00-0.11)

0.01 (0.00-0.03)

0.030

Treg/CD3+

0.04 (0.00-0.18)

0.01 (0.00-0.04)

0.055

Treg/CD4+

0.09 (0.01-0.24)

0.06 (0.02-0.10)

0.269

Treg/CD8+

0.12 (0.00-1.12)

0.03 (0.00-0.09)

0.037

Table 1: distribution of lymphocytes subpopulation in aGVHD patients treated with Ruxolitinib/ECP or ECP as second line.

Conclusions: In our cohort the association of Ruxolitinib and ECP did not result in higher response rate or greater steroid sparing. Evaluation of immunological recovery during treatment suggests an increment in CD4+ and T-regulatory lymphocyte responses in Ruxolitinib plus ECP group. Apoptotic CD3+ at baseline were increased in aGVHD achieving CR.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P264 GVHD PROPHYLAXIS WITH POST-TRANSPLANT HIGH DOSE CYTOXAN OVERCOMING THE BARRIER OF ALLOGENEIC HCT IN FUNCTIONAL HIGH-RISK PATIENTS

Asya Varshavsky Yanovsky1, Yuliya Shestovska 1, Matthew Hamby1, Dzhirgala Mandzhieva1, Shalina Joshi1, Peter Abdelmessieh1, Rashmi Khanal1, Michael Styler1, Henry Fung1

1Fox Chase Cancer Center, Philadelphia, United States

Background: Post-transplant Cytoxan (PTCy) was originally established as safe and effective GVHD prophylaxis in haploidentical transplants with BM grafts. Subsequently the use of PTCy was extended to matched donor and peripheral blood stem cell (PBSC) grafts. While PBSC grafts are traditionally associated with higher rates of GVHD compared to BM graft with MTX/ CNI prophylaxis, prospective BMT-CTN 1703 study showed improved 1-year GVHD-free, relapse free survival with PTCy compared to MTX based ppx in reduced intensity conditioning (RIC) PBSCT. At Fox Chase Cancer Center we adopted routine use of PTCy for all donor/graft types and both myeloablative (MAC) and RIC conditioning since 2014. We recently reported lower acute and chronic GVHD, lower non-relapse mortality (NRM) and a trend towards improved OS in PBSC transplant with PTCy compared to non-PTCy GVHD prophylaxis in an institutional retrospective analysis. Older patients, patients with high HCT-CI and low KPS represent unique challenges in transplant due to increased risk of GVHD and NRM, as well as increased cumulative chemotherapy toxicities. Here we analyze acute GVHD (aGVHD) and NRM outcomes in the subgroup of high functional risk patients, defined as age 65 + , HCT comorbidity index (HCT-CI) of 3+ and Karnofsky performance status (KPS) of 70 or less, with PTCy prophylaxis.

Methods: We retrospectively reviewed patients who received first PBSCT at our program between 2007-2021 from a 10/10 matched unrelated (MUD) or matched related (MRD) donor and had high functional risk (HFR) defined as at least one of the following: age 65 + , HCT-CI 3+ and KPS 70 or less. We analyzed aGVHD and survival/mortality outcomes using Fisher exact test. NRM was defined as mortality in CR.

Results: We identified 119 HFR patients who received MRD or MUD PBSCT between 2007-2021, 85 with PTCy and 34 with non-PTCy GVHD prophylaxis. Baseline patients’ characteristics are shown in Table 1.

1-year NRM and severe (grade 3-4) aGVHD were extremely low with PTCy in this high-risk population (1-year NRM: 6%, severe aGVHD: 3%), that was significantly lower compared to non-PTCy GVHD prophylaxis group (21% and 33% respectively) (Table 1). Even in the subgroup of patients who received myeloablative conditioning, 1-year NRM was favorable in the patients who received PTCy vs non-PTCy (9% vs 20%, not reaching statistical significance). 1 year OS was numerically favorable in the PTCy group (74% vs 62%, not reaching statistical significance).

Table 1 Baseline patients’ characteristics

Non PTCy

PTCy

n

34

85

Sex: M:F (%)

47:52

44:56

Age: Mean (range) (y.o)

54.5 (33-70)

56.8 (21-75)

Dx (%):

ALL

14

8

AML

17

39

CML

14

0.5

MDS

6

24

MPN

14

2.5

NHL/HL

20

19

Other dx

15

7

Intensity: MA: RIC/NMA (%)

59:41

38:62

Donor type: MRD : MUD (%)

62:38

35:65

Duration of follow up, Mean (range) (mon)

46.1(1-168)

35.6(1-96)

Results

Non PTCy

PTCy

P value

aGVHD

67:33

97:3

0.0004

gr.0-2 : 3-4 (%)

patients who died <1 year without aGVHD excluded from this analysis.

NRM 1y

21%

6%

0.036

NRM 1y for MAC

20%

9%

0.4076

OS 1y

62%

74%

0.267

OS 1y for MAC

60%

69%

0.5605

Conclusions: Our retrospective analysis demonstrates that in functionally high-risk population including older patients, patients with high comorbidity index and low KPS, PTCy GVHD prophylaxis results in extremely low incidence of aGVHD and lower 1-year NRM compared to non-PTCy prophylaxis. Lower 1-year NRM was likely related to the decrease in GVHD related mortality, and the trend towards lower NRM remained even in high functional risk patients who received PTCy GVHD prophylaxis with myeloablative conditioning. Overall, PTCy represents a reasonably safe option for patients who are at high risk for high transplant related morbidity and mortality.

Disclosure: Asya Varshavsky Yanovsky: Pfizer, Janssen, BMS- advisory.

Henry Fung: Astra Zeneca, Incyte, Kite, Janssen - advisory, honoraria.

Peter Abdelmessieh: AbbVie, Sobi - advisory.

12: Graft-versus-host Disease – Clinical

P265 MESENCHYMAL STROMAL CELLS IN PATIENTS WITH III-IV GRADE STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST-DISEASE: DAY 14 RESPONSE IS PREDICTIVE OF SURVIVAL

Adomas Bukauskas 1, Renata Jucaitienė1, Artūras Slobinas1,2, Inga Šlepikienė1, Andrius Žučenka1,2, Linas Davainis1, Valdas Pečeliūnas2, Igoris Trociukas1, Laimonas Griškevičius1,2

1Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania, 2Vilnius University, Vilnius, Lithuania

Background: Bone marrow-derived mesenchymal stromal cells (MSCs) is a potential therapeutic option for steroid-refractory acute graft-versus-host disease (SR-aGVHD).

Methods: All patients had steroid-refractory and biopsy-proven grade III-IV aGVHD after allogeneic hematopoietic stem cell transplantation (HSCT) or donor lymphocyte infusion. SR-aGVHD was managed with bone marrow-derived MSCs manufactured in a hospital setting at Vilnius University Hospital Santaros Klinikos. The target dose of MSCs was 1×10e6 cells/kg body weight and was administered intravenously once weekly. The MSC3 and MSC6 groups were scheduled to receive up to 3 and 6 infusions, respectively. The treatment response was evaluated on days 7, 14, and 28. Patients were evaluated for overall survival. Data were collected prospectively. The results were considered statistically significant at the 5% level (p<0.05).

Results: Fifty-three adult SR-aGVHD patients (Grade III, 48 and Grade IV, 5) received MSCs (MSC3, 17; MSC6, 36) as salvage treatment. The median age of the patients was 54 (19-71) years. 39 patients (74%) had been transplanted for acute leukemia. Involvement of the gastrointestinal tract, skin, and liver was observed in 91%, 45%, and 25% of patients, respectively. One organ was affected in 22 (42%) patients (gastrointestinal involvement in 91% of the cases), two organs in 27 (51%), and three organs in 4 (7%). The median time from HSCT/DLI to GVHD was 56 (18-221) days and from GVHD diagnosis to MSCs treatment was 11 (1-217) days. The median dose was 1x10e6/kg (0.68-1.33). 20 (38%) patients received ≤3 MSC and 33 (62%) >3 doses. Baseline characteristics were comparable between the MSC3 and MSC6 groups (Table 1). The median observation time was 2 (0-118) months with surviving patients having a median observation time of 62 (1-118) months. D14 and D28 complete/partial responders had a significantly better overall survival than non-responders (stable/progressive disease), 104 months vs. 1 month (p=0.000), respectively (Table 1).

The next line of treatment was initiated in 34 (64%) patients who failed to respond to MSCs: R-ECP-17, Tocilizumab-10, Vedolizumab-3, Ruxolitinib-3, Methylprednisolone re-initiation-1. Regardless of additional therapy, 26 (76%) patients died, all due to infectious complications alone or with concomitant GVHD, except for one patient whose cause of death was attributed to relapse.

At the last follow-up, 13 of the 53 patients were alive. The causes of death were infectious complications alone or with concomitant GVHD in 30 (75%), primary disease relapse in 9 (22.5%), and asystole in 1 (2.5%) patient. Out of 16 patients surviving for more than 6 months, limited and extensive cGVHD developed in 3 (19%) and 3 (19%), respectively.

No adverse events that could be clearly attributed to MSCs infusion were observed.

Table 1. III-IV grade SR-aGVHD patients’ baseline characteristics, response rate, and overall survival.

SR-aGVHD patients

MSC3

(N=17)

MSC6

(N=36)

MSC3 + MSC6

(N=53)

MSC3 vs MSC6

p value

Baseline characteristics

Patient age, years, median (range)

41 (20-60)

55 (19-71)

54 (19-71)

0.054

Patient gender, male, n (%)

7 (40%)

25 (69%)

32 (60%)

0.051

Primary disease, n (%)

0.097

-Acute leukemia

10 (59%)

29 (81%)

39 (74%)

•Acute myeloid leukemia

6 (60%)

20 (68%)

26 (67%)

-Other*

7 (41%)

7 (19%)

14 (26%)

GVHD grade:

0.697

-GVHD grade III

15 (88%)

33 (92%)

48 (91%)

-GVHD grade IV

2 (12%)

3 (8%)

5 (9%)

Organ involvement :

-Skin

5 (29%)

19 (53%)

24 (45%)

0.354

-Gastrointestinal

16 (94%)

32 (89%)

42 (91%)

0.439

-Liver

5 (29%)

8 (22%)

13 (25%)

0.844

Organ number:

0.294

-One

9 (53%)

13 (36%)

22 (42%)

•Single organ GI

8 (89%)

12 (92%)

20 (91%)

-Two

7 (41%)

20 (56%)

27 (51%)

-Three

1 (6%)

3 (8%)

4 (7%)

MSC treatment line:

0.552

- 2

16 (94%)

32 (89%)

48 (91%)

- >2

1 (6%)

4 (11%)

5 (9%)

MSC dose, x106/kg at day 1, median (range)

1.07 (0.85-1.33)

0.97 (0.68-1.31)

1.00 (0.68 -1.33)

0.091

No. of MSC doses

0.000

- One

1 (6%)

0 (0%)

1 (2%)

- Two

9 (53%)

1 (3%)

10 (19%)

- Three

7 (41%)

2 (6%)

9 (17%)

- Four

0 (0%)

5 (14%)

5 (9%)

- Five

0 (0%)

7 (19%)

7 (13%)

- Six

0 (0%)

20 (55%)

20 (38%)

- Seven

0 (0%)

1 (3%)

1 (2%)

Response rate and overall survival

Day 7 response rate

0.410

CR-0 (0%)

CR-0 (0%)

CR-0 (0%)

PR-3 (17%)

PR-6 (17%)

PR-9 (17%)

SD-12 (71%)

SD-30 (83%)

SD-42 (79%)

PD-2 (12%)

PD-0 (0%)

PD-2 (4%)

Day 14 response rate

0.741

CR-3 (17%)

CR-4 (11%)

CR-7 (13%)

PR-4 (24%)

PR-10 (28%)

PR-14 (26%)

SD-8 (47%)

SD-21 (58%)

SD-29 (55%)

PD-0 (0%)

PD-0 (0%)

PD-0 (0%)

Dead-2 (12%)

Dead-1 (3%)

Dead-3 (6%)

Day 28 response rate

0.864

CR-6 (35%)

CR-8 (22%)

CR-14 (26%)

PR-1 (6%)

PR-7 (19%)

PR-8 (15%)

SD-4 (24%)

SD-13 (36%)

SD-17 (32%)

PD–0 (0%)

PD–2 (6%)

PD–2 (4%)

Dead–6 (35%)

Dead–6 (17%)

Dead–12 (23%)

Estimated OS (95% CI)

6 months – 35.3% (12.6-58.0)

6 months – 28.6% (13.3-43.9)

6 months – 31.0% (18.3-43.7)

0.737

1 year – 29.4% (7.6-51.2)

1 year – 25.5 % (10.8-40.2)

1 year – 26.8% (14.6-38.9)

5 year – 29.4% (7.6-51.2)

5 year – 22.3 % (8.2-36.4)

5 years – 24.8% (12.8-36.8)

OS according to Day 7 CR/PR vs SD/PD

2 months vs 2 months, p=0.716

(N=17)

3 months vs 2 months, p=0.341

(N=36)

3 months vs 2 months, p=0.313

(N=53)

0.843

OS according to Day 14 CR/PR vs SD/PD OS

104 months vs 1 month, p=0.031

(N=15)

Not reached vs 1 months, p=0.001

(N=35)

104 vs 1 month, p=0.000

(N=50)

0.914

OS according to Day 28 CR/PR vs SD/PD OS

104 months vs 1 month, p=0.180

(N=12)

Not reached vs 2 months, p=0.000

(N=30)

104 vs 1 month, p=0.000

(N=42)

0.555

  1. OS – overall survival, CR-complete response, PR-partial response, SD-stable disease, PD-progressive disease, GI – gastrointestinal, *Other - AA>PNH, CML, CLL, IMF, MDS, NHL, MM

Conclusions: Among MSC treated III/IV grade SR-aGVHD patients, the day 14 non-responders have a dismal prognosis and should be considered for alternative treatment. More than 3 MSC doses do not seem to improve clinical outcomes.

Clinical Trial Registry: Study is registered in clinical trial registry ISRCTN18091201.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P266 JAK-2 INHIBITOR RUXOLITINIB FOR THERAPY OF ACUTE STEROID-REFRACTORY GRAFT-VERSUS-HOST DISEASE AFTER ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH HEMATOLOGICAL MALIGNANT DISEASES

Nikita Levkovsky1, Olesya Paina1, Anna Zvyagintseva1, Anna Osipova1, Alexandra Laberko1, Ivan Moiseev1, Tatyana Bykova 1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1

1R.M. Gorbacheva Memorial Institute of Hematology, Oncology and Transplantation, Pavlov University, Saint Petersburg, Russian Federation

Background: Allogeneic hematopoietic stem cell transplantation (alloHSCT) is a standard therapy option for high-risk malignant disease in children, but is associated with high risk of acute graft-versus-host disease (aGVHD), especially when haploidentical donor is used. About 50% of the patients have no response to first line steroids, and steroid-refractory GVHD (srGVHD) is one of the main causes of death. Based on the results of Phase III study, ruxolitinib is standard of care for srGVHD in adults, however accumulating data in children has not transplanted into clinical recommendations yet.

Methods: The single-center retrospective study was conducted from 2012 to 2022 and involved 26 patients younger than 18 years with hematological malignant diseases after alloHSCT who experienced acute srGVHD. ALL had 16 (61%) patients, AML - 7 (27%), MDS - 2 (8%) and JMML - 1 (4%). Six (23%) patients had relapsed/refractory disease status prior to alloHSCT. Median age at HSCT was 8 years (1-17), 20 (77%) were male, 6 (23%) - female. In 24 (92%) cases was used haploidentical donor, 1 (4%) and 1 (4%) unrelated and related donors respectively. SCT source was bone marrow in 25 (96%), peripheral blood stem cells in 1 (4%). Myeloablative conditioning was used in 13 (50%) cases, reduced intensity conditioning in 13 (50%). For aGVHD prophylaxis post-transplant cyclophosphamide was used at days +3, +4 in 23 (88%) patients, antithymocyte globulin in 2 (8%) and combination in 1 (4%). Ruxolitinib was administrated in srGVHD with a median dose of 0.3 (0.1 - 0.6) mg/kg. Response to therapy was assessed at days +28 and +56 after ruxolitinib initiation only in alive patients. Complete response (CR) is defined as a resolution of all manifestations in each site, partial response (PR) as an improvement in at least 1 site without progression in any other site.

Results: Median follow-up time after alloHSCT was 13 months (1.5 – 62.7). Acute GVHD II, III, IV grade was diagnosed in 11 (42%), 9(35%) and 6 (23%) patients respectively. Skin involvement was documented in 25 (96%), GI in 13 (50%) and liver in 6 (23%). Response at days +28 and +56 was assessed in 24 patients, 2 patients died before day +28 due to aGVHD progression, infection complication and thrombotic microangiopathy in one case. CR at day +28 was documented in 7 (29%) patients, PR in 9 (37.5%). On day +56 CR was achieved in 17 (71%) patients, PR in 4 (17%). Median time to PR was 11 days (2 – 53), median time to CR was 33 days (2 – 122). CR of skin was assessed in 19 (76%) patients, GI in 8 (61%), liver in 2 (33%). By CTCAE, 14 patients (54%) had grade 3-4 neutropenia, 7 (27%) grade 3 anemia, 8 (13%) grade 3-4 thrombocytopenia. In 2 (8%) patients ruxolinitib was stopped due to toxicity. 1-year OS was 69% (0.67 – 0.97). 1-year NRM was 20% (0.07-0.38). 1-year malignancy relapse incidence was 23,5% (0.095-0.41).

Conclusions: JAK2-inhibitor – ruxolitinib is effective therapy of acute srGVHD in children with low rate of adverse effects.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P267 THE IMPACT OF MEASURED RENAL FUNCTION BY IOTHALAMATE CLEARANCE ON ALLOGENEIC STEM CELL TRANSPLANT OUTCOMES IN PATIENTS RECEIVING POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR GVHD PROPHYLAXIS

Jade Kutzke 1, Christopher Cahoon2, Kristin Mara1, Jodi Taraba1, Anmol Baranwal1, Mark Litzow1, William Hogan1, Abhishek Mangaonkar1, Mehrdad Hefazi Torghabeh1, Aasiya Matin1, Mithun Shah1, Urshila Durani1, Saad Kenderian1, Robert Wolf1, Gabriel Bartoo1, Hassan Alkhateeb1

1Mayo Clinic, Rochester, United States, 2University of Utah, Salt Lake City, United States

Background: Post-transplant cyclophosphamide (PT-Cy) is the new standard of care for graft-versus-host disease (GVHD) prophylaxis in allogeneic stem cell transplantation (alloSCT). The impact of renal impairment in patients receiving PT-Cy for GVHD prophylaxis is currently unknown.

Methods: We evaluated adult patients who underwent alloSCT and received PT-Cy for GVHD prophylaxis between January 1, 2018 and December 31, 2022. Renal function was evaluated via three methods: Iothalamate-clearance based measured glomerular filtration rate (mGFR), Modification of Diet in Renal Disease (MDRD) estimated GFR (eGFR), and Cockcroft and Gault (CG) estimated creatinine clearance (eCrCl). Competing risk analysis was used to determine the impact of renal function, as predicted by the mGFR, eGFR and eCrCl, on non-relapse mortality (NRM), with relapse being considered as competing risk. Kaplan-Meier and log-rank method were used to compare survival outcomes. We used stepwise regression analysis to arrive at the final multivariate model.

Results: A total of 79 patients were evaluated. The median age at alloSCT was 58.4 years (IQR 42.6 - 64.2 years). The most common disease for alloSCT was acute myeloid leukemia (N= 30, 38%), followed by MDS (N= 15, 19%), and acute lymphoblastic leukemia (N= 14, 18%) (Table 1). Fifty (63%) patients received a haploidentical alloSCT. Competing risk analysis showed that an increasing mGFR was associated with a decreasing 1-year NRM (HR 0.97, 95% CI 0.96 – 0.99, P= 0.008). Neither increasing eGFR, increasing eCrCl nor decreasing serum creatinine was associated with a decreasing 1-year NRM. We found that mGFR cut-off of 92 mL/min/1.73 m2 was the optimal cut-off point for 1-year NRM (HR 7.84, 95% CI 1.01 – 60.7, P= 0.049). One-year NRM was significantly higher among patients with mGFR below 92 mL/min/1.73 m2 (22% vs. 3.03%, P= 0.02). The rounded mGFR of 90 mL/min/1.73 m2 was used to determine the impact on survival. C-index for the model for NRM was 0.64. Patients with mGFR ≤ 90 mL/min/1.73 m2 were older in age, had delayed neutrophil engraftment [median 20 days (IQR 18 – 25 days) vs. 19 days (IQR 15 – 21 days), P= 0.036] and had increased risk of bacterial infections post-transplant (HR 3.91, 95% CI 1.47 – 11.21, P= 0.008) (Table 1). Patients with mGFR > 90 mL/min/1.73 m2 had a superior 1-year post-alloSCT survival compared to those with an mGFR ≤ 90 mL/min/1.73 m2 (1-year OS rate: 88.7% vs. 67%, P= 0.03). There was no significant impact of mGFR on platelet engraftment, nor an effect on incidence of viral, fungal infections post-transplant. Upon adjusting for age, multivariate analysis showed that mGFR ≤ 90 mL/min/1.73 m2 showed a trend towards inferior survival at 1-year post-alloSCT (HR 3.05, 95% CI 0.97 – 9.55, P= 0.056).

Iothalamate clearance

> 90 ml/min/1.73m2

(N=36)

Iothalamate clearance

≤ 90 ml/min/1.73m2

(N=43)

P

Age at alloSCT

Mean (SD)

48.1 (16.3)

56.6 (12.9)

0.014

Median [Min, Max]

54.9 [19.6, 66.0]

59.2 [24.7, 74.4]

Gender

Male

25 (69.4%)

29 (67.4%)

1

Female

11 (30.6%)

14 (32.6%)

Diagnosis

AML

15 (41.7%)

15 (34.9%)

0.86

ALL

7 (19.4%)

7 (16.3%)

CML

2 (5.6%)

4 (9.3%)

MDS/MPN

7 (19.4%)

8 (18.6%)

Others

5 (13.9%)

9 (20.9%)

Graft Type

Bone marrow

4 (11.1%)

11 (25.6%)

0.18

Peripheral blood

32 (889%)

32 (74.4%)

HCT-CI score

Low

17 (47.2%)

22 (51.2%)

0.9

High (≥3)

19 (52.8%)

21 (48.8%)

Corrected Iothalamate

Clearance

Median [Min, Max]

106 [91.0, 130]

71.0 [14.0, 90.0]

<0.001

Estimate GFR

Median [Min, Max]

89.6 [57.6, 124]

69.2 [31.4, 172]

<0.001

Missing

0 (0%)

1 (2.3%)

CG CrCl

Median [Min, Max]

114 [56.6, 183]

92.9 [47.1, 198]

0.003

Missing

0 (0%)

1 (2.3%)

Melphalan

No

17 (47.2%)

21 (48.8%)

1

Yes

19 (52.8%)

22 (51.2%)

Cyclophosphamide

No

31 (86.1%)

27 (62.8%)

0.037

Yes

5 (13.9%)

16 (37.2%)

Fludarabine

No

1 (2.8%)

5 (11.6%)

0.29

Yes

35 (97.2%)

38 (88.4%)

Myeloablative Conditioning

No

24 (66.7%)

35 (81.4%)

0.22

Yes

12 (33.3%)

8 (18.6%)

Haploidentical transplant

No

14 (38.9%)

15 (34.9%)

0.89

Yes

22 (61.1%)

28 (65.1%)

Time to neutrophil engraftment

Median [Min, Max]

19.0 [14.0, 29.0]

20.0 [15.0, 47.0]

0.036

Missing

3 (8.3%)

1 (2.3%)

Time to platelet engraftment

Median [Min, Max]

28.0 [11.0, 209]

29.0 [14.0, 166]

0.31

Missing

6 (16.7%)

5 (11.6%)

Infection

Bacterial

No

17 (47.2%)

8 (18.6%)

0.013

Yes

19 (52.8%)

35 (81.4%)

Viral

No

24 (66.7%)

26 (60.5%)

0.74

Yes

12 (33.3%)

17 (39.5%)

Fungal

No

34 (94.4%)

40 (93.0%)

1

Yes

2 (5.6%)

3 (7.0%)

Grade II-IV Acute

No

29 (80.6%)

35 (81.3%)

1

Yes

7 (19.4%)

8 (18.7%)

Grade III-IV Acute

No

33 (91.7%)

41 (95.3%)

0.84

Yes

3 (8.3%)

2 (4.7%)

Chronic GVHD

No

32 (88.9%)

35 (81.4%)

0.54

Yes

4 (11.1%)

8 (18.6%)

Conclusions: We found that impaired renal function measured by iothalamate clearance was associated with a higher 1-year post-transplant NRM predominantly driven by increased risk of bacterial infections. It is imperative to ensure renal function is accurately measured prior to proceeding with alloSCT.

Clinical Trial Registry: N/A.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P268 RUXOLITINIB TREATMENT OUTCOMES IN ACUTE GRAFT-VERSUS-HOST DISEASE (AGVHD) IN REAL-WORLD SETTING IN FINLAND

Eeva Martelin 1, Arttu Kuikka2, Hanna Rajala1, Tuomas Ruohonen2, Hannu Mönkkönen3, Johanna Vikkula4, Kristiina Uusi-Rauva4, Urpu Salmennniemi1, Maija Itälä-Remes2

1Helsinki University Hospital, Helsinki, Finland, 2Turku University Hospital, Turku, Finland, 3Novartis Finland Oy, Espoo, Finland, 4Medaffcon Oy, Espoo, Finland

Background: Ruxolitinib is the first EMA-approved treatment for the patients with steroid-refractory (SR) acute or chronic graft-versus-host disease (aGvHD/cGvHD). The aim of this retrospective, non-interventional cohort study was to evaluate real-world efficacy of ruxolitinib in the treatment of aGvHD in Finland.

Methods: Patient population included adult aGvHD patients transplanted at Turku and Helsinki University Hospitals, the two centers which perform all allogeneic stem cell transplantations for adults in Finland. Medical records of patients with aGvHD treated with ruxolitinib between Jan 2019 to Aug 2021 were reviewed. The follow-up of each patient lasted from the initiation of ruxolitinib to death or end of study (EOS), 31 Aug 2022. Primary endpoint was best overall response rate to ruxolitinib (ORR; complete or partial response, CR or PR). Secondary endpoints were time to response, response duration and overall survival (OS). Timely events were estimated with Kaplan-Meier method. Time to best response was defined as time from onset of treatment to best response (event) or death/ruxolitinib discontinuation (censoring events). Response duration was defined as time from best response to aGvHD progression (event), cGvHD (competing risk), or non-GvHD death/EOS (censoring event).

Results: The baseline characteristics of 56 patients with aGvHD are summarized in Table 1. Two thirds of the patients (77 %; n= 43) were transplanted for acute myeloid/monocytic (n= 17) or lymphoblastic leukemia (n= 7) or high-risk myelodysplastic syndrome (n= 19). The median follow-up time was 21.8 months (interquartile range IQR: 11.8, 32.5). Of the 56 patients, 63 % (n= 35) were steroid-refractory or -dependent. The ORR was 91 % (95% confidence interval, CI: 83.5–98.5) with CR achieved by 39 patients (70 %) and PR by 12 patients (21%). The ORR remained the same regardless of the concomitant extracorporeal photopheresis. The median time to best response was 28.0 days (95 % CI: 21.0–38.0). aGvHD progression rate was the highest during the first three months reaching a plateau at six months, with 16.0 % (95% CI: 8.5–30.2) of the patients estimated to exhibit aGvHD progression. The median event-free survival was 10.6 months (95 % CI: 3.3–not reached). The median ruxolitinib dose was 10 mg at the onset of treatment (IQR: 10.0,10.0) and one month later (IQR: 10.0, 20.0). The most common cause of ruxolitinib discontinuation was achievement of response (64 %). Of the 51 patients treated with systemic corticosteroid (CS), 67 % (n= 34) stopped CS before the end of follow-up, while the remaining 33 % (n= 17) were on low dose at death/EOS. Eighteen patients died, nine from GvHD, during the follow-up. The estimated three-year OS was 64.1 % (95 % CI 48.2-76.3).

Table 1. Patient characteristics

Variable

Estimate/level

Overall (n=56)

Missing

Age at index, years

Median [IQR]

56.5 [41.2, 63.4]

0.0

Sex, N (%)

Female

28 (50.0)

0.0

Male

28 (50.0)

Type of donor, N (%)

Haploidentical relative

23–26*

0.0

HLA-identical sibling

<5*

Unrelated matched donor

29 (51.8)

Graft source, N (%)

Bone marrow transplant

<5*

0.0

Peripheral blood stem cell transplant

52–55*

Pretransplantation conditioning, N (%)

MAC

28 (50.0)

0.0

RIC

28 (50.0)

GvHD prophylaxis, N (%)

MTX-based

20 (35.7)

0.0

PTCy-based

36 (64.3)

aGvHD grade at ruxolitinib initiation, N (%)

Grade 1

15 (27.3)

1.8

Grade 2

23 (41.8)

Grade 3-4

17 (30.9)

Type of ruxolitinib treatment, N (%)

Any combination, with ECP

21–24*

0.0

Any combination, without ECP

31 (55.4)

Ruxolitinib in monotherapy

<5

  1. * Due to the anonymity requirement of the data authority, results with low subject numbers were masked and it was ensured that the masked numbers cannot be deduced based on other available numbers, thus other numbers additionally provided as a range, if needed. Abbreviations: CS, corticosteroid; ECP, extracorporeal photopheresis; HLA, human leukocyte antigen; IQR, interquartile range; MAC, myeloablative conditioning; MTX-based, methotrexate-based [methotrexate + calcineurin inhibitor (CNI)/mammalian target of rapamycin (mTOR) ± anti-thymocyte globulins (ATG)/anti-T lymphocyte globulins (ATLG) ± corticosteroid (CS) ± mycophenolate mofetil (MMF)]; PTCy-based, post-transplant cyclophosphamide-based [high-dose cyclophosphamide (HDCy) + CNI/mTOR ± MMF)]; RIC, reduced-intensity conditioning.

Conclusions: This study provides novel real-world data on the efficacy and safety of ruxolitinib in the treatment of aGvHD. The presented data is in agreement with the outcome data of clinical trials. Ruxolitinib appears as effective and safe in the real-world setting of severe or steroid-refractory aGvHD.

Disclosure: The study was funded by Novartis (Espoo, Finland). Eeva Martelin, Arttu Kuikka, Hanna Rajala, and Tuomas Ruohonen have nothing to declare. Hannu Mönkkönen is employed by Novartis. Johanna Vikkula and Kristiina Uusi-Rauva are employed by Medaffcon Oy (Espoo, Finland), which received payments from Novartis for conducting the study. Urpu Salmenniemi has been a member of advisory boards for Takeda, AstraZeneca, and Immedica, and provided consultancy to Viatris. Maija Itälä-Remes has been a member of advisory board for Novartis and provided consultancy to Incyte.

12: Graft-versus-host Disease – Clinical

P269 VON WILLEBRAND FACTOR AS A DIAGNOSTIC AND ACTIVITY INDICATOR OF CHRONIC GRAFT-VERSUS-HOST DISEASE

Antonela Lelas1, Lana Desnica 1, Ivan Sabol2, Desiree Coen Herak1,3, Marija Milos1,4, Ana Zelic Kerep1, Ante Vulic1, Nadira Durakovic1,3, Ranka Serventi Seiwerth1, Zinaida Peric5,6, Radovan Vrhovac1,3, Steven Zivko Pavletic7, Drazen Pulanic1,3

1University Hospital Centre Zagreb, Zagreb, Croatia, 2Rudjer Boskovic Institute, Zagreb, Croatia, 3University of Zagreb, Zagreb, Croatia, 4University of Mostar, Mostar, Bosnia and Herzegovina, 5University Hospital Centre Rijeka, Rijeka, Croatia, 6University of Rijeka, Rijeka, Croatia, 7National Cancer Institute, Bethesda, United States

Background: Endothelial injury plays an important role in development of chronic Graft-versus-Host Disease (cGvHD). Von Willebrand factor (VWF) is a relatively inexpensive and readily available biomarker of endothelial injury.

Methods: Von Willebrand factor antigen (VWF:Ag) and activity (VWF:Ac) levels were measured longitudinally in 83 cGvHD patients and correlated to corresponding NIH 2004 scores afterwards. Control group were 39 non-cGvHD post allo-HSCT patients who were evaluated one time only. Patients with systemic infections were not included in the analysis. P<0.05 was considered statistically significant.

Results: Median time between allo-HSCT and cGvHD diagnosis was 9 (2-202) months. Forty of eighty-three cGvHD patients (48.2%) were evaluated in the early phase of disease, defined as within 30 days from diagnosis. Patients that received peripheral blood stem cells (PBSC) higher incidence of cGVHD (p=0.014) and aGVHD (p<0.001) than control group, while age, sex, main diagnosis, and donor type were similarly distributed in both groups. Most of cGvHD patients had classic type (86.7%) of disease. At first evaluation, most cGvHD patients had active form of disease (59.8%), moderate or severe NIH cGvHD score (94.0%) and Karnofsky score >80% (84.1%). Organs most affected with cGvHD were mouth, skin, and eyes (59.0%, 54.2% and 54.2%), respectively. Even though VWF:Ag and VWF:Ac were statistically higher than the upper reference limit in both cGvHD patients and controls (p<0.0001), median levels of both markers were significantly higher in cGvHD patients than in controls (VWF:Ag 268.2% vs 191.9%; VWF:Ac 261.8% vs 178.2%; p= 0.003). Univariate analysis showing significant correlations between factors, demography, and NIH scores for cGvHD patients is shown in Table below.

cGvHD patients’ characteristics (n=83)

VWF:Ag (%)

VWF:Ac (%)

Age

τb*

0.262

0.272

P

0.001

<0.001

Time from alloHSCT to first evaluation

τb

-0.189

-0.219

P

0.011

0.003

PBSC

τb

0.272

0.267

P

0.003

0.003

cGvHD type of onset

τb

-0.199

-0.159

P

0.021

0.065

Activity of disease assesed by clinician

τb

-0.266

-0.200

P

0.004

0.029

Karnofsky score

τb

0.348

0.314

P

<0.001

<0.001

Number of organs affected with cGvHD

τb

0.261

0.283

P

0.001

0.001

NIH mouth score

τb

0.228

0.259

P

0.008

0.003

NIH liver score

τb

0.311

0.264

P

<0.001

0.002

NIH GIT score

τb

0.170

0.174

P

0.056

0.050

  1. *Kendall rank correlation coefficient tau-b (τb)

Multivariate analysis identified age, cGvHD NIH liver score and Karnofsky score as significant predictors of VWF:Ag levels (R2=31.2%, p<0.0001) while age, cGvHD NIH liver score and number of organs affected with cGvHD were predictive of VWF:Ac levels (R2=33.3%, p<0.0001). Longitudinal assessment of VWF during 4 year (0-7 years) median follow-up showed persistent elevation among cGvHD patients with active disease (defined as active disease assessed by clinician and/or on systemic therapy). Further analysis, among other, revealed statistically significant difference of VWF levels (p<0.0001) between patients in early phase of cGvHD and controls and between patients with active and inactive cGvHD (defined as inactive disease assessed by clinician and off systemic therapy).

Dynamics of VWF antigen and activity through follow-up time.

W30D – cGvHD evaluated within 30 days; postGvHD – after cGvHD resolution (inactive disease with no ongoing therapy).

Receiver operating curve (ROC) analysis showed good diagnostic potential of VWF:Ag>246.3% (AUC=0.733) and VWF:Ac>271.4% (AUC=0.728) for differentiating patients at early phase of cGvHD from non-cGvHD post-alloHSCT controls. VWF levels at early phase of cGvHD were not predictive for later development of sclerotic, inflammatory, or mixed cGvHD phenotype.

Conclusions: Results of our study suggested that von Willebrand factor can serve as potential cGvHD biomarker which can enable earlier diagnosis and more effective disease activity monitoring, especially regarding liver involvement. Further investigation through prospective studies on larger number of patients is prompted.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P270 A PROSPECTIVE PILOT STUDY OF GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS WITH POSTBIOTICS IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Jong-Ho Won 1, Seug Yun Yoon1, Sun Young Jeong1, Min-Young Lee1, Kyoung Ha Kim1, Namsu Lee1

1Soonchunhyang University Seoul Hospital, Seoul, Korea, Republic of

Background: Conditioning chemotherapy, antibiotics, and diet changes reduce intestinal microbiome diversity during allogeneic hematopoietic stem cell transplantation (allo-HSCT). As the diversity of the microbiome more decreases during the engraftment period in the transplantation process, acute graft-versus-host disease (aGVHD) incidence and mortality rate increase, according to numerous reports. Therefore, we suggested that rapid restoration of microbiome diversity might help in the prevention of GVHD. Postbiotics are metabolites generated by probiotic microorganisms that exhibit biological activity within the host. When the intestinal microbiome is compromised, we suggested that administering postbiotics directly would be beneficial. As a postbiotics, butyrate, one of the short-chain fatty acids, was administered. It was utilized despite the absence of any noteworthy adverse effects reported in previous human studies; it is currently available for purchase. Previous studies have mostly been based on 16S ribosomal RNA gene sequencing, which is limited in taxonomic resolution at the genus level and inferred functionality. In this study, we employed whole genome shotgun (WGS) sequencing metagenomics to acquire a more accurate representation of the dysbiosis of the intestinal microbiome in allo-HSCT.

Methods: A prospective study was conducted to evaluate the clinical effects and microbial changes in postbiotics intake in allo-HSCT recipients at Soonchunhyang University Seoul hospitals. Twelve patients who underwent allo-HSCT from September 2022 through August 2023 were enrolled in the study. The patients ingested butyrate (sodium butyrate 600 mg x 2capsules/day, BODYBIO) from the engraftment period (D + 14~D + 21) until day 90~100 after the transplantation. In addition, twenty three patients who underwent allo-HSCT from March 2021 through August 2022 were enrolled in the historical control group. Fecal samples were obtained from both the historical control group and the postbiotics group at three time points: before transplantation, at engraftment, and 100 days after transplantation. By matching their age and gender, eight patients were chosen from each group. Clinical data of the historical control group were collected from the medical records. WGS sequencing was performed on collected stool samples. (NCT#05808985).

Results: Fifty-one samples of stool were obtained from sixteen patients (the postbiotics group, n=8; the historical control group, n=8). The cumulative incidence of all grades of aGVHD was similar in the postbiotics group (n=5) compared to the historical control group (n=5). The incidence of grade (Gr) 3 to 4 aGVHD was lower in the postbiotics group (n=1) compared to the historical control group (n=3). In particular, the historical control group exhibits three cases of lower gastrointestinal aGVHD (Gr2, n=2; Gr4, n=1), whereas the postbiotics group reported lesser severe two cases (Gr1, n=1; Gr2, n=2). Patients self-reported that the median intake of postbiotics was 84%. The only discernible occurrence during administration was the unpleasant smells emitted by butyrate. Furthermore, there were no anticipated adverse effects associated with butyrate administration. Analysis of the microbiome is in progress; details may be revealed at the EBMT 2024 meeting.

Conclusions: In conclusion, the administration of postbiotics lacking of specific adverse effects is anticipated to prevent GVHD. Long-term follow-up of a larger cohort of patients is needed to define the role of butyrate in allo-HSCT.

Clinical Trial Registry: This study was registered on the ClinicalTrials.gov as #NCT05808985.

(https://clinicaltrials.gov/study/NCT05808985?cond=NCT05808985&rank=1).

Disclosure: The authors declare no conflicts of interest.

12: Graft-versus-host Disease – Clinical

P271 POST-TRANSPLANT CYCLOPHOSPHAMIDE WITH ANTI-THYMOCYTE GLOBULIN LOWERED SERUM IL-6 LEVEL COMPARED TO POST-TRANSPLANT CYCLOPHOSPHAMIDE ALONE AFTER HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION

Jeong Suk Koh1, Wonhyoung Seo1, Sora Kang1, Chul Hee Kim1, Myung-Won Lee1, Hyewon Ryu1, Hyo-Jin Lee1, Hwan-Jung Yun1, Deog-Yeon Jo1, Ik-Chan Song 1

1Chungnam National University Hospital, Daejeon, Korea, Republic of

Background: Post-transplant cyclophosphamide (PTCy) and anti-thymocyte globulin (ATG) are both common prophylaxis strategies of graft versus host disease (GVHD) in haploidentical-hematopoietic stem cell transplantation (haplo-HSCT). Recently, there have been reports that cytokine release syndrome (CRS) after haplo-HSCT affects the severity of acute GVHD. And IL-6 is a surrogate marker for CRS. However, there is lack of data about CRS between PTCy with ATG and PTCy alone in hematologic malignancy patients undergoing haplo-HSCT. This study aimed to compare the clinical outcomes and complications of haplo-HSCT according to levels of serum IL-6 between two groups.

Methods: This study included patients who underwent haplo-HSCT at Chungnam National University Hospital (CNUH) from January 2019 to February 2023. ATG was administered at a dose of 1.5 mg/kg on days -3 to -1 prior to transplantation. All patients received GVHD prophylaxis with PTCy at 50 mg/kg on days +3 and +4, followed by mycophenolate mofetil (MMF) and tacrolimus. The values of serum IL-6 levels were measured at day+3 after infusion of peripheral blood stem cell (PBSC). CRS was graded according to ASTCT scales. Manifestations occurring within 5 days after PTCy were included.

Results: A total of 40 patients who underwent haplo-HSCT were analyzed. Fever between day 0 and day +6 occurred in 22 of 40 patients (55%). Elevated levels of IL-6 were observed in PTCy with ATG patients compared to that in the PTCy patients. There was significance between the PTCy with ATG group and the PTCy group as regards IL-6 level 7.47±10.55 vs. 117.65±127.67 with p value= 0.003. The total number of patients with CRS grade 0 was higher than in PTCy with ATG compared PTCy group (p < 0.001). Serum IL-6 levels are associated to grade II-IV acute GVHD (r=0.57, p=0.000). The cumulative incidence (CI) of grade II-IV acute GVHD was significantly higher in the PTCy group (42.9% vs. 4.8%; p= 0.006). There was no significant difference in the CI of chronic GVHD between the PTCy with ATG and PTCy groups (72.1% vs. 82.0%; p=0.730). The CI of 1-year NRM was significantly higher in the PTCy group compared with the PTCy with ATG group (42.2% vs. 15.9%; p=0.022), where there was no significant difference in 1 year relapse mortality between the two groups (PTCy with ATG, 30.3% vs. PTCy; 28.5%; p=0.550). The 1-year Overall survival (OS) was significantly better in the PTCy with ATG group (PTCy with ATG, 75.9% vs. 35.3%; p=0.011).

Conclusions: Serum IL-6 levels were higher in PTCy alone group than in PTCy with ATG group. Addition of ATG before stem cell infusion affects IL-6 levels and reduces the incidence of CRS, grade II–IV acute GVHD in haplo-HSCT. In conclusion, this study suggests that PTCy with ATG strategy as GVHD prophylaxis in haplo-HSCT is beneficial for the clinical outcome and complication of HSCT.

Disclosure: The authors declare no potential conflicts of interest.

12: Graft-versus-host Disease – Clinical

P272 USE OF RUXOLITINIB IN STEROID DEPENDENT/REFRACTORY ACUTE AND CHRONIC GRAFT VERSUS HOST DISEASE: A SINGLE CENTRE EXPERIENCE

Dimitris Galopoulos 1, David Irvine1, Anne Parker1, Grant McQuaker1, Anne-Louise Latif1, Ailsa Holroyd1, Lorna Welsh1, Andrew Clark1

1Queen Elizabeth University Hospital, Glasgow, United Kingdom

Background: Graft versus Host Disease (GvHD) remains a major complication in allogeneic stem cell transplant. First line treatment consists of systemic corticosteroids, but in almost half of the cases GvHD will be steroid dependent/refractory (SD/SR GvHD). Unfortunately, no specific treatment in second line has proven to be superior to others. Recent experience in our centre showed that ruxolitinib can be effective in the management of SD/SR GvHD.

Methods: We retrospectively analyzed 54 patients who received ruxolinitib for treating either acute GvHD (aGvHD) or chronic GvHD (cGvHD). They all had steroid SD/SR GvHD. GvHD assessment and response were documented using the MAGIC consortium scoring system for aGvHD, and NIH criteria for cGvHD. Primary endpoints to this analysis were response to ruxolitinib: time to first response and to best overall response in patients who achieved complete (CR) or partial (PR) GvHD response, and secondary endpoint was the 5-year overall survival (OS) using the Kaplan-Meier method and log rank test comparing survival in cases who had complete versus any other GvHD response. We also documented discontinuation of steroids and other types of treatment for GvHD, as well as organ specific response, and discontinuation of ruxolitinib in cases who achieved CR.

Results: Patients’ characteristics are shown in table 1. 8 patients received ruxolitinib for aGVHD (all had Grade III/IV) and 47 for cGvHD (20% had mild, 26.7% had moderate and 53.3% had severe cGvHD). Median line of treatment for ruxolitinib was 3 (range: 2-9). It was used either as a single agent (2 patients), or in combination with other treatments (CNI, MMF, sirolimus, steroids, ECP, or topical-organ specific treatment). Median time to first response was 31 (12-94) days for all cases (CR 4 and PR 19 cases; first response rate 42.6%), 60 (31-94) days for aGvHD, and 29 (12-92) days for cGvHD. Median time to overall response was 97 (26-925) days for all cases (CR 22 and PR 9 cases; overall response rate 57.4%), 77(31-153) days for aGvHD, 106 (26-925) days for cGvHD. Organ specific response was 56.25% (27/48) for skin, 67.6% (23/34) for gastrointestinal tract, 57.5% (15/25) for liver, 60% (6/10) for oral, 38% (7/18) for eyes, 14.3% (1/7) for lungs, 33.3% (2/6) for genital tract, and 41.6% (5/12) for joints and fascia. Among responders, 56.8% (21/47) discontinued steroids and 51.9% (27/52) discontinued other types of GvHD treatment. 8 patients who showed complete response successfully discontinued ruxolitinib and GvHD has remained quiescent to date. Median 5-year OS (95% confidence interval, CI) for cases with GvHD in CR was 89.5% (95% CI 76.6-100), and 43.2% (95% CI 28.3-66) for all other cases (logrank p value=0.007).

Table 1: Patient baseline characteristics

Frequency

Percentage %

Gender

Male/Female

33/21

38.9/61.1

Age at transplant

48 years (range:15-68)

Follow-up time

1111 days (range: 60-8341)

Diagnosis

ALL

18

33.3

AML

17

31.5

MDS/MPN/CML

8

14.8

MF

6

11.1

NHL

4

7.4

AA

1

1.9

Positive CMV status (either donor or recipient or both)

21

39.6

Female donors to male recipients

7

13.2

ABO mismatch

Minor/Major/No

7/17/28

13.5/32.7/53.8

Donor type

Sibling/VUD/Haplo

22/32/0

40.7/59.3/0

HLA match 10/10

48

88.9

Conditioning regime type

RIC/MAB/NMAB

25/28/1

46.2/51.9/1.9

T cell depletion

No/ATG/Alemtuzumab

28/9/17

51.9/16.7/31.5

PTCy

Conclusions: Ruxolitinib shows great efficacy in treating SD/SR GvHD. Patients with lung, genital tract and ocular GvHD seem to respond less well, but assessment is challenging in chronic GvHD, as it is sometimes difficult to distinguish active GvHD from irreversible scarring; remnants of previously active disease that has now burnt out. A complete response of GvHD shows a favorable survival outcome.

Disclosure: Dr David Irvine: Kite (Honoraria and conference attendance), Janssen (conference attendance).

12: Graft-versus-host Disease – Clinical

P273 ALPHA-1-ANTITRYPSIN TREATMENT OF STEROID-RESISTANT ACUTE AND CHRONIC GRAFT-VERSUS-HOST DISEASE, SINGLE CENTER EXPERIENCE

Şebnem İzmir Güner1

1İstanbul Gelişim University Memorial Şişli Hospital, İstanbul, Turkey

Background: Acute and chronic graft-versus-host disease (GvHD) is a major complication that prevents successful outcomes after allogeneic bone marrow transplantation (BMT), an effective therapy for hematological malignancies. Several studies demonstrate that donor T cells and host antigen-presenting cells along with several proinflammatory cytokines are required for the induction of GvHD and contribute to its severity. Despite routine prophylaxis, clinically significant acute and chronic graftversus-host disease requiring systemic corticosteroids occurs in; 40% of patients undergoing HLA-matched allogeneic hematopoietic cell transplantation (HCT). Roughly half of the patients requiring high-dose corticosteroids will not respond, a condition termed steroid-resistant GVHD (SR-GVHD), which is associated with poor overall survival (OS). Alpha1-Antitrypsin (AAT), a naturally abundant serine protease inhibitor, is capable of suppressing experimental GVHD by downmodulating inflammation and increasing ratios of regulatory (Treg) to effector T cells (Teffs).

In this retrospective single center clinical study, we sought to determine the safety and response rate of AAT administration in SR-GVHD.

Methods: From 2021 through 2022, investigators at Memorial Sisli Hospital enrolled patients age over 18 years. All patients provided written informed consent approved by the institutional review boards. SR-GVHD was defined as: clinical evidence of GVHD in 1 or more than 1 target organs (skin, liver, gut) per consensus grading and lack of clinical response or progression despite receiving 1 mg/kg per day of prednisone or equivalent.

All patients had biopsy documenting histologic evidence of GVHD.

Patients characteristics are shown at table 1.

3 patients received AAT treatment before 100 days posttransplant.(min.26-max.52).

4 patients received AAT treatment after 100 days posttransplant. (min.160-max.390).

Eligible patients received AAT IV at a dose of 60 mg/kg per day for up to 4 consecutive weeks on days 1, 4, 8, 12, 16, 20, 24, and 28 (maximum, 8 doses) as therapy for SR-GVHD. AAT dose min. 3000-max.4000 performed. Administered doses min.4-max.8.

GVHD assessments were graded and recorded weekly.

Table 1. Patient characteristics

Characteristic

N

%

Total enrolled and treated

7

Median age (range), y

36,71 (19-65)

Male sex

6

85,7

Unrelated donor

3

Related donör

4 (1 patients underwent haploidentical allogeneic bone marrow transplantation from his mother)

57,1

Hematologic malignancy

Acute myeloid leukemia

3

43,2

T-cell Acute lymphoid leukemia

1

14,2

B-cell Acute lymphoid leukemia

1

14,2

Mantle cell Lymphoma

1

14,2

Paroxismal nocturnal hemoglobinuria

1

14,2

Conditioning

Myeloablative intensity

5

71,6

Reduced-Intensity Conditioning

1

14,2

Antithymocyte globulin containing

1

14,2

Median Karnofsky performance status (range), %

60 (20-100)

Target organ and severity grade Skin

1

2

3

4

Liver (+/- skin +/-gut)

1

14,2

3

4

3

43,2

Gut (+/- skin)

2

28,4

3

4

1

14,2

SR-GVHD definition

No response to steroids Progression with steroids

2

28,4

Steroid+fotophorezis

2

28,4

3

43,2

AAT treatment Response/Death

Yes

5

71,6

No

2 (These one patient died from GIS GVHD, the outher patient died from skin +GIS GVHD)

28,4

The most common infection

Bacteremia related to central venous catheters

3

43,2

Cytomegalovirus (CMV) reactivation (viremia) no cases of CMV-related organ disease.

4

57,1

Results: Among evaluable patients (n 5), the median time to PR after starting AAT was 14 days (range, 8-28 days) and the median time to CR was 28 days (range, 8-40 days), which is equivalent to 4 and 8 AAT doses, respectively. All patients had completed all 8 planned doses of AAT by day 28.

AAT administration was well tolerated, with no infusion reactions or drug-related grade 3 to 4 toxicity. 3 deaths occurred. Among these,1 occurred within 30 days of the last administered dose of AAT; because of progression of skin SR-GVHD. One patient died as a result of sepsis related to gram-negative septicemia 120 days after completing treatment. One patient died due to a relaps of his leukemia 180 days after completing treatment.

Conclusions: We demonstrate that twice weekly AAT infusion is safe and feasible and results in a significant proportion of clinical responses, without high rates of infection. In our study, although our number of cases was low, we tested that AAT treatment was effective and seems safe, with low rates of infection, demonstrates initial evidence for efficacy in SR-GVHD, and is a suitable candidate for future controlled trials in GVHD.

Disclosure: Conflict-of-interest disclosure: The author declare no competing financial and no conflict of interest.

12: Graft-versus-host Disease – Clinical

P274 EFFICACY AND SAFETY OF INFLIXIMAB THERAPY IN THE SETTING OF STEROID-REFRACTORY ACUTE GRAFT-VERSUS-HOST DISEASE

Khalid Halahleh 1, Jana Al Shurman2, Ayat Taqash1, Mohammad Ma’koseh1, Ahmad Abu Khader1, Isra Muradi3, Waleed Da’na1, Rawad Rihani1

1King Hussein Cancer Center, Amman, Jordan, 2King Hussien Cancer Center, Amman, Jordan, 3University of Tripoli, Tripoli, Libya

Background: Acute graft-versus-host disease (aGvHD) is the main cause of mortality and morbidity after allogenic heamatopoietic cell transplantation. Steroid therapy is the first-line treatment; however, less than 50% of patients achieve complete remission (CR). Reports suggest that the release of a cytokine TNF-α from the bone marrow during conditioning is involved in the pathogenesis of GvHD.

Methods: Using King Hussein Cancer Center (KHCC) Blood and Marrow Transplantation (BMT) Registry database, 18 subjects, who received infliximab for the treatment of grade II-IV acute graft-versus-host-disease (aGvHD) after the first allogeneic hematopoietic stem cell transplantation (HSCT) for malignant and nonmalignant conditions, were identified, from May 2015 to June 2020. We evaluated the outcome of anti-TNF-α therapy with infliximab in patients with steroid refractory (SR-aGvHD). Infliximab was administered intravenously at 10 mg/kg for a median of three doses (range, 1-4) on a weekly basis.

Results: Subjects,-transplant-related characteristics were detailed in table 1. Median age was 45.5y (12-63y). 12 subjects (66.7%) were males.16 subjects (89%) had malignant conditions. 15 subjects (83.0%) received matched related donor (MRD) transplants, and 13(72%) received reduced intensity conditioning. The graft was peripheral blood grafts in 16 subjects (89%). The overall objective response rate was 55% (22% CR, 33% PR) at 4 weeks; 33% (17.5% CR, 17.5% PR) at 8 weeks, and 45% (CR 28%, PR 17%) at 12 weeks. Response evaluation according to organ involved at 12 weeks, 7 subjects achieved complete response in the gut and 3 partial response; 8 achieved CR in the skin and 1 PR, and 1 achieved CR and 1 PR in the liver.

Median follow up of survivors (n-5) was 61months (52-152 months). Overall survival (OS) at 6, 12 and 60 months from the start of infliximab therapy was 44.5%, 39%, and 33% respectively. The main causes of death was gut aGvHD (n-7/13), relapse (n-3/13), and infections (n-2/18). Overall survival was significantly improved in infliximab responders versus non-responders at all-time points (5-year OS: 86% (53 vs NR; P-0.0003). 14 subjects (83%) developed infectious complications within 12 weeks of initiation of infliximab. These infections include10 bacterial infections and 13 viral reactivations (4 colitis, 1 pneumonitis). 2 patients died due to infections.

Table 1: Subjects,-transplant-related characteristics

Gender

Male

77%

Female

33%

Disease Entity

AML + MDS

44.5%

ALL+ Lymphoma

44.55

Bone Marrow failure

11%

Disease status at transplant

Active disease

22%

Complete remission

72%

Partial remission

6%

Conditioning intensity

Reduced intensity

72%

Myeloablative

28%

Stem cell source

Peripheral blood

89%

Bone marrow

11%

Donor type

MRD

83%

MMRD

17%

GvHD prophylaxis

CSA or TAC/MMF

72%

CSA or TAC/PTCy

6%

CSA or TAC/Methotrexate

22%

Objective response at 4 weeks

Complete response

22%

Partial response

33%

Infections

Viral

72%

Bacterial

55.5%

Patient status at last encounter

Alive

28%

Dead

72%

Conclusions: The use of infliximab in subjects with SR-aGvHD is associated with modest durable responses, however at the cost of increased infectious complications. The approval of less toxic novel therapies and combination approaches are desirable in this patient’s population.

Clinical Trial Registry: NA.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P275 THE EFFICACY AND SAFETY OF EXTRACORPOREAL PHOTOPHERESIS IN STEROID-DEPENDENT-REFRACTORY/ GVHD: COHORT STUDY FROM COMPREHENSIVE ADULT BONE MARROW TRANSPLANTATION PROGRAM IN JORDAN

Khalid Halahleh 1, Raneem Al zubi1, Mohammad Bssol1, Husam Abu-Jazar1, Mohammad Ma’koseh1, Ehab Omari1, Isra Muradi2, Rawad Rihani1

1King Hussein Cancer Center, Amman, Jordan, 2Isra Muradi, Al-Salt, Jordan

Background: GvHD is the leading cause of morbidity and mortality after allo-HCT. Extracorporeal photopheresis (ECP) has been introduced as an alternative treatment in steroid-dependent (SD-GvHD) and steroid-refractory GvHD (SR-GvHD).

Methods: Using King Hussein Cancer Center (KHCC) Blood and Marrow Transplantation (BMT) Registry database, we retrospectively reviewed medical records of patients who received ECP for acute and chronic GvHD between august 2013 and June 2023. We studied the efficacy and safety of ECP.

Results: 42 patients were included in the analysis. 19 patients with grade II-IV aGvHD and 23 with moderate to severe cGvHD received ECP. There were no major adverse events related to ECP. Thirteen patients had SD-GvHD, and 27 had SR-GvHD. 30 patients (71%) were males. Median age at ECP initiation was 39.5 year (range: 15-63 year). 34 donors (81%) were HLA-identical siblings and eight (19%) HLA-mismatched relatives. 19 patients (45%) received myeloablative conditioning. The majority received conventional GvHD prophylaxis; two patients received PTCy, and two ATG. Peripheral blood was the stem cell source in 40 patients (95%). 1-year cumulative incidence of aGvHD was 36%; and cGvHD was 13%.

The median number of ECP cessions were 27 (2-72) for aGvHD and 30 (2-72) for cGvHD. The objective response rate (ORR) for patients with aGvHD was 33% (10 CR 10; 4 PR), leading to 3-year overall survival (OS) of 49%. OS was significantly improved in patients with gut aGvHD compared with other sites (83% vs 40%, P-0.028). For patients with cGvHD, the ORR was 43% (11 CR 11; 7 PR), leading to 3-year OS of 70%. OS was significantly improved in patients with cGvHD compared to patients with no cGvHD (P-0.0001). Steroid dose could be reduced by 50% in 19 patients (45%) and discontinued in 36(86%) at a median of 95 days (15-461day). 1- and 3-year cumulative incidence of transplant-related mortality (TRM) was 24% and 43% respectively. TRM was associated with the lack of response to ECP (P-0.0004) and steroid refractoriness (P-0.0001). After a median follow-up of 63 months (11-180 month) for survivors, 3-year OS was 56%, which is significantly better in complete responders vs non-responders (77% vs11%; P-0.0006), and improved in patients who had immunosuppression discontinuation (86% vs 40%, P-0.0049).

Conclusions: Our study suggests that ECP is safe, effective for GvHD and might be introduced early in the course of GvHD treatment.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P276 COMPARING THE OUTCOMES OF MATCHED AND MISMATCHED UNRELATED ALLOHCT WITH DIFFERENT ANTI-THYMOCYTE GLOBULIN FORMULATIONS. A RETROSPECTIVE ANALYSIS ON BEHALF OF POLISH ADULT LEUKEMIA GROUP

Ugo Giordano1, Monika Mordak2, Małgorzata Sobczyk-Kruszelnicka3, Sebastian Giebel3, Lidia Gil4, Justyna Pilch1, Jarosław Dybko 5

1Wroclaw Medical University, Wroclaw, Poland, 2Lower Silesian Center of Oncology, Pulmonology and Hematology, Wroclaw, Poland, 3Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland, 4Poznan University of Medical Sciences, Poznań, Poland, 5Wroclaw University of Science and Technology, Wroclaw, Poland

Background: In spite of some essential immunotherapy progress in recent years, allogeneic hematopoietic stem cell transplantation (allo-HCT) still stands as a potentially curative treatment modality. The efficacy of rabbit anti-thymocyte globulin (r-ATG) in the prevention of graft-versus-host disease (GvHD) has been addressed by numerous studies. Nevertheless, merely few of them sought to carry out a direct comparison of two of the most frequently applied r-ATG formulations, that is Thymoglobuline (ATG-T, formerly Genzyme) and Grafalon (ATG-G, formerly Fresenius).

Methods: A total of 87 patients were included in the analysis, of whom 40 were administered ATG-T and 47 ATG-G. 100% of the individuals in the ATG-T group received methotrexate (MTX) with cyclosporine A (CsA) or tacrolimus (Tac) as GvHD prophylaxis compared to 85% in the ATG-G one. The ratios of the applied conditioning regimens (MAC/RIC/NMA) were 82.5%/10%/7.5% for ATG-T, and 53.2%/31.9%/14.9% for ATG-G (p=0.014). The median ages of both cohorts were 45 and 56 (p=0.055) with female/male ratios of 17/23 and 20/27 (p=0.832), respectively. The median times of follow-up were 27 months (7-42) for the ATG-T group, and 16 months (8-44) for the ATG-G one.

Results: No differences in the diagnosis, median CD34+ count, patients’ age, donors’ CMV status and age were found between the two groups. Also, no statistically significant discrepancies were observed in the occurrence of acute GvHD (aGvHD) comparing ATG-T and ATG-G. However, there was a tendency for ATG-T to cause more frequently aGvHD, aGvHD grades II-IV with a lower median time of onset of aGvHD in contrast to ATG-G (65% vs 44.7%, p=0.084; 42.5% vs 31.9%, p=0.308; 24 vs 29 days, p=0.366, respectively). Chronic GvHD (cGvHD) was less frequent in the ATG-T group compared to ATG-G (7.5% vs 38.3%, p=0.001). Patients treated with ATG-T experienced a higher incidence of CMV reactivations (70% vs 31.9%, p<0.001), with a shorter time between transplant and CMV (<61 days, 77.8% vs 33.3%, p=0.008) and a higher median CMV copy number (1000 vs 0, p=0.004). The number of patients who received letermovir as CMV prophylaxis was similar between the ATG-T and ATG-G groups (7.5% vs 10.9%). Interestingly, despite a higher number of CMV reactivations in the ATG-T cohort, there were more asymptomatic patients than in those who were administered ATG-G (85.7% vs 43.8%, p=0.005). The 5-year overall survival (OS) was comparable between the two cohorts (57.8% vs 59.0%, p=0.559) with, however, a higher 3-year relapse-free survival (RFS) in the ATG-T cohort (55.2% vs 37.4%, p=0.036).

Conclusions: We are aware of the relatively small sample size in our study. Nevertheless, the results suggest that ATG-T could be an efficacious formulation for preventing GvHD, guaranteeing comparable outcomes in aGvHD and a lower incidence of cGvHD in contrast to ATG-G. Of note, a higher 3-year RFS and similar 5-year OS resulted from administering ATG-T. A downside could be the higher number of CMV reactivations with ATG-T, however most of them were asymptomatic.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P277 RITUXIMAB FOR THE TREATMENT OF STEROID-REFRACTORY ORAL CHRONIC GRAFT-VERSUS-HOST DISEASE AFTER ALLO-HEMATOPOIETIC STEM CELL TRANSPLANTATION

Wei Ma 1, Xing-Yu Cao1, Song Xue2, Lei Dong1

1Hebei Yanda Ludao Pei Hospital, Langfang, China, 2Beijing Ludao Pei Hospital, Beijing, China

Background: Oral chroic graft-versus-host disease (cGVHD) is a common complication of allo-hematopoietic stem cell transplantation (allo-HSCT). Oral cGVHD may have serious impact on patients’ quality of life, indirectly affect the teeth and the oral flora, developing the risk of infections and even the oral cancer. There is no standard treatment for steroid-refractory cGVHD (SR-cGVHD). B cells play an important role in the pathogenesis of cGVHD. So B cells could potentially be a target for the treatment of oral cGVHD. In this study, we evaluated the efficacy and long term outcome of rituximab in treating oral SR-cGVHD.

Methods: A retrospective analysis was conducted on six patients who underwent allo-HSCT at our centre and developed steroid-refractory oral cGVHD between November 2011 and November 2023. After steroid-refractory all these patients were treated with rituximab. Rituximab was administered intravenously at a dose of 100mg once a week, with an average of three doses (rang: 1 to 6). The deadline for follow-up is November 2023. The median follow-up time is 9 months.

Results: The median age of the patients was 39.5 years old. There were 3 male and 3 female patients, all diagnosed with malignant disorders, Acute myelogenous leukemia (n=3), Acute B lymphoblastic leukemia(n=1), Peripheral T-cell lymphomas(n=1), Chronic myeloid leukemia-Blast phase(n=1) respectively. One patient underwent matched identical sibling donr transplantation and 5 patients underwent haplo-identical transplantation. The median time of neutrophil engraftment was 12 days(range:11-15), and the median time of platelet engraftment was 15 days(range: 11-29). GVHD prophylaxis consisted of ATG, CSA, MMF and MTX. None of the patients experienced grade III-IV aGVHD. At a median of 4.3(range: 2.5-6) months after transplantation, all patients developed cGVHD, with 5 patients experiencing moderate to severe cGVHD. Presence of other GVHD sites involved at enrollment:skin(n=3), eyes(n=3), mouth(n=6), liver(n=2), gastrointestinal tract(n=2), lung(n=2). Prior to receiving rituximab, patients had received median 4(rang: 3-6) lines of treatment for cGVHD. At a median time of 5months(range:3.5-11) after the onset of cGVHD, patients were initiated on rituximab therapy. After the use of rituximab, oral pain was reduced in 6 patients and oral ulcers healed in 3 patients. Steroid were tapered in all patients. No infections occurred in any of the six patients after the application of rituximab. All patients had decreased IgG levels and lower absolute peripheral blood B-cell counts. Oral GVHD re-exacerbated in only one patient after rituximab discontinuation The estimated 2-year overall survival rate was 66.7%. Two patients died after secondary transplantation due to relapse.

Conclusions: The study demonstrates that rituximab is effective in treating steroid-refractory oral cGVHD. Therefore, rituximab could be a preferred option for treating steroid-refractory oral cGVHD.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P278 EXTRACORPOREAL PHOTOPHERESIS FOR THE TREATMENT OF GVHD IN CHILDREN IN THE ERA OF TRANSPLANT FROM PARTIALLY MATCHED DONORS

Marianna Maffeis 1, Elena Soncini1, Giulia Baresi1, Stefano Rossi1, Chiara Gorio1, Emilio Ferrari2, Arnalda Lanfranchi3, Fulvio Porta1

1Pediatric Oncohematology Unit and Pediatric Bone Marrow Transplant Center, Children Hospital Brescia, Brescia, Italy, 2ASST Spedali Civili, Brescia, Italy, 3Stem Cell Laboratory, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy

Background: Graft-versus-host disease (GVHD) is a common complication after HSCT that can be observed especially in the setting of partially matched donor transplants and that can impact on overall survival and quality of life of children. The increased use of many immunomodulating and immunosoppressive drugs have recently been introduced for the treatment of GVHD. A treatment option for acute and chronic GVHD is extracorporeal photopheresis (ECP). Many studies about the use of ECP has been published, most of all in small cohorts of patients with both acute and chronic GVHD. In skin and liver steroid-refractory acute GVHD, the reported response rate is more than 60%, especially in patients with less severe GVHD. In chronic GVHD, overall response rates above 50% have been reported in patients with skin, oral, eye, liver, gastrointestinal and lung involvement. One of the advantages of ECP is its low toxicity, especially in fragile patients and those with uncontrolled infections, for whom systemic immunosuppressive therapy may be harmful.

Methods: We retrospectively analyzed our cohort of pediatric patients who received ECP for GVHD since 2010, when ECP became available at our center.

Results: We collected data on 30 pediatric patients. Twenty-four patients presented with acute GVHD and six with chronic GVHD. Most patients had high grade GVDH (24/30 grade III and 6/30 grade IV). Median age at the first ECP was 6.4 years. All patients presented with skin involvement, 15 patients had gut disease and 9 had liver involvement. Median number of ECP sessions was 16.9. 70% of patients achieved complete remission by using ECP (21/30). The best responses were observed in skin and gut GVHD. A total of 9 patients (30%) died after ECP due to infectious complications or serious clinical conditions already existing before ECP. All surviving patients continue to receive at least one immunosuppressive therapy for chronic GVHD.

Overall, ECP displays a substantial response rate and in particular steroid-sparing activity in skin GVHD. However, most patients continue to require some chronic therapy and cGVHD-related morbidity and mortality remain high.

Conclusions: In our experience, ECP was well tolerated and no fatal toxicities were observed. Our data show that ECP is feasible even in very young children weighing less than 10 kg (eg PID patients). Our results support previous reports of objective responses to ECP in cutaneous and visceral GVHD. The main limitation of ECP is the need for adequate central venous access, especially in young children. ECP is a gold standard in the treatment of GVHD, allowing control of this complication without exposing the patient to the potential drug toxicity observed with immunomodulatory or conventional immunosuppressive drugs.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P279 POST-TRANSPLANTATION CYCLOPHOSPHAMIDE IN HAPLOIDENTICAL TRANSPLANT: A REPORT IN LOW AND MIDDLE INCOME COUNTRY

Araceli Leal-Alanis1, Juan Luis Ontiveros-Austria1, Maria Alejandra Nuñez-Atahualpa1, Anghela Milenka Mendoza-Sanchez1, Bogar Pineda-Terreros1, Rubén Solis Armenta 1, Brenda Lizeth Acosta-Maldonado1, Manuel Valero Saldaña1

1Instituto Nacional de Cancerología, Mexico City, Mexico

Background: Hematopoietic stem cell transplantation (HSCT) feasibility has increased in the last decades due to availability of alternative donors. Haploidentical transplant (haplo-HSCT) with post-transplantation cyclophosphamide (ptCy) has emerged as a safe and efficacious strategy for graft-versus-host disease (GVHD) prophylaxis.

Methods: We analyzed all adult patients who received ptCy at the dose of 50 mg/kg per day +3 and +4 in haplo-HSCT between 2016 and 2022. Retrospective study that included patients ≥ 18 years of age that received treatment and follow up at Instituto Nacional de Cancerología in Mexico City.

Results: A total of 35 patients were included. The median age at transplant was 31.6 years (range, 21-42) and 51.4% were female. The most common diagnoses were acute lymphoblastic leukemia (n=19, 54.3%), acute myeloid leukemia (n=12, 34.3%), and others (n=4, 11.4%). At diagnosis 94.3% (n=33) were high risk patients and 62.9% (n=22) had received 2 prior lines of treatment. Cytomegalovirus (CMV) risk was intermediate in 82.9% (n=29) and high in 14.3% (n=5). Conditioning regimen was mostly myeloablative (n=25, 71.4%). Nine patients (25.9%) had positive anti-HLA antibodies, and only six (66.6%) required desensitization regimen. The mean dose of CD34+ cells infused was 6.43 x106/kg (±SD, 0.73). Cytokine release syndrome (CRS) was present in 80% (n=28) of our patients and only two were grade 3 CRS. All patients received an immunosuppression regimen with cyclosporine and mycophenolate mofetil (MMF). The median time to engraftment was 16.5 days (range, 13-48). Primary graft failure was observed in three patients (8.6%) and poor graft function in seven (20%). Fourteen patients had acute GVHD (40%), 6 were grade II-III, none grade IV. Chronic GVHD was documented in 25.7% (n=9) of cases. Twenty patients (57.1%) had CMV reactivation; in bivariate analysis, this was associated with acute GVHD (OR 1.58, p=0.044 IC 1.04-22.8). With a median follow-up of 29 months (IQR, 7-41), the two year overall survival (OS) and relapse free survival (RFS) were 70.6% and 85.2%, respectively. Incidence of relapse during follow up was 20% (n=7).

Conclusions: Haplo-HSCT with ptCy is a feasible and effective strategy that shows encouraging survival with acceptable acute and chronic GVHD incidences, without increasing relapse risk. Despite being a small retrospective study, it is interesting that our results are similar to larger analysis; besides, severe acute GVHD was not observed in our population.

Disclosure: none.

12: Graft-versus-host Disease – Clinical

P280 EFFICACY AND SAFETY OF RUXOLITINIB FOR PEDIATRIC ACUTE AND CHRONIC GRAFT-VERSUS-HOST DISEASE - A SINGLE CENTER EXPERIENCE

Shu-Wei Chou 1, Meng-Yao Lu1, Hsiu-Hao Chang1, Yung-Li Yang1, Shiann-Tarng Jou1

1National Taiwan University Children’s Hospital, Taipei, Taiwan, Province of China

Background: Acute and chronic graft-versus-host diseases (GVHD) are common complications for patients who received allogeneic hematopoietic stem cell transplantation (Allo-HSCT). Corticosteroids with or without calcineurin inhibitor remains the standard first-line therapy. However, there was no uniform second line treatments for steroid-refractory or steroid-dependent acute and/or chronic GVHD. Ruxolitinib, a Janus kinase (JAK) inhibitor, has been approved for steroid-refractory acute GVHD and chronic GVHD after failure of at least one lines of systemic therapy in adults and pediatric patients 12 years and older. However, the efficacy and safety has not been well-studied in pediatric population, especially in patients less than 12-year-old.

Methods: Patients less than 18-year-old who received ruxolitinib for either acute or chronic GVHD after Allo-HSCT were identified. The types of Allo-HSCT, severity of GVHD, treatments before ruxolitinib, response to ruxolitinb, and adverse events were obtained retrospectively by reviewing medical records.

Results: Seven patients received ruxolitinib for acute GVHD with median age of 6.3 years old. Grade II, III and IV acute GVHD happened in four, one and two patients respectively. Overall response rate(ORR) on day 28 of ruxolitinib was 86% with complete response (CR) rate 71%. All patients were able to taper at least 75% of initial steroid dose at day 56 of ruxolitinib. Three of five CR patients were able to taper off steroid before day 56 of ruxolitinib. Nine patients received ruxolitinib for chronic GVHD with median age of 9.0 years old. Three of them had moderate chronic GVHD and six of them had severe chronic GVHD. Three patients achieved CR, four patients had partial response and two patients remained unchanged for their best overall response for chronic GVHD. In six patients with steroid use while starting ruxolitinib, median steroid dose reduction up to week 24 after ruxolitinib initiation was 95%. In this 16-patient cohort, hematological adverse event was the most common side effects with grade 3/4 neutropenia in 7 patients and grade 3/4 thrombocytopenia in 6 patients. Besides, 9 of 16 patients (56%) suffered from at least one episode of viral reactivation.

Conclusions: Ruxolitinib use is associated with high ORR for GVHD after Allo-HSCT in our cohort, especially for acute GVHD. Besides, the dose of steroid could be reduced significantly after ruxolitinib use. However, blood counts and CMV/EBV status should be closely monitored due to high rate of severe cytopenia and viral reactivation.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P281 SYSTEMATIC LITERATURE REVIEW ON THE SAFETY, EFFICACY, AND EFFECTIVENESS OF EATG VERSUS COMPARATORS FOR GVHD

Erin Sheffels1, Kevin Kallmes1, Keith Kallmes1, John Pederson1, Barbara Możejko-Pastewka 2, Raj Gokani2, Judith Hey-Hadavi2, Andres Quintero2

1Nested Knowledge, St Paul, United States, 2Pfizer, Inc., New York City, United States

Background: Evidence on aplastic anemia and anti-thymocyte globulin (ATG) demonstrates that equine ATG (eATG) and rabbit ATG (rATG) have different effects on the immune system. We performed a systematic literature review to compare clinical outcomes for eATG and rATG for the prevention and treatment of graft vs. host disease (GVHD).

Methods: Using the PubMed database, we performed a PRISMA-compliant systematic review of prospective comparative trials on the use of ATG for the prevention and treatment of GVHD. We included RCTs and prospective clinical studies reporting eATG, without restrictions on publication date.

Patient data collected included age, sex, and indication for hematologic graft. Outcomes included GHVD severity, incidence of acute and chronic GHVD, graft vs. leukemia, engraftment, response to treatment, infection, thrombotic microangiopathy hospitalization, mortality, and event-free survival. Due to study heterogeneity, inferential statistics were not performed.

Results: From 1,553 records identified, we analyzed 32 studies (4,004 patients), including 27 on prophylaxis and 5 on the treatment of GVHD.

Among studies evaluating prophylaxis, eight reported eATG with comparators that included rATG, combinations of methotrexate, cyclosporine A (CsA), steroids, tacrolimus, and mycophenolate mofetil. Among studies evaluating the non-prophylactic management of aGVHD, two reported using eATG. Comparators included combinations of steroids, methotrexate, CsA, and monoclonal antibodies.

For prophylaxis, the incidence of acute GVHD (aGVHD), chronic GVHD (cGVHD), infection, and mortality were not significantly different between eATG and comparators. Among RCTs comparing eATG to methotrexate, one demonstrated significantly lower incidence of aGVHD and another demonstrated significantly lower incidence of infection, both favoring eATG. Among three non-randomized studies with non-eATG comparators, one demonstrated that adding eATG was less effective at reducing the risk of cGVHD and mortality, and two demonstrated a significantly higher incidence of aGVHD with eATG. In two other non-randomized prospective studies that directly compared eATG and rATG, there was a significantly higher incidence of aGVHD among patients managed with eATG. For the latter two studies, however, there were overlapping patients, and their ability to support meaningful inferences was limited by small sample sizes, with three patients receiving eATG and 25 patients receiving rATG.

Across studies on the treatment of GVHD, none compared eATG and rATG directly, and there were no significant differences between treatment arms for complete or partial response to treatment or mortality. In one study, relative to a combination of CsA and steroids, eATG significantly increased the incidence of CMV infection and pneumonitis, but not gram-negative or fungal infections. Across other studies, differences in any type of infection were not significant.

Conclusions: For GVHD propylaxis, eATG had lower rates of aGVHD than methotrexate in older studies, and relative to more current immunsuppressive cocktails, eATG may be less effective. For aGVHD, differences in outcomes between eATG and comparators were not significantly different. This review is limited by old data in RCTs, non-randomized protocols, and within- and between-study heterogeneity in prophylaxis for studies on treatment of GVHD. Further analyses data are needed to confirm our findings.

Disclosure: Barbara Możejko-Pastewka, Raj Gokani, Judith Hey-Hadavi, and Andres Quintero are employees of Pfizer Inc. Kevin Kallmes is employed by and holds equity in Nested Knowledge, Inc; he holds equity in and serves on the board of Superior Medical Experts, Inc., and Piraeus Medical, Inc.

12: Graft-versus-host Disease – Clinical

P282 A SERIES OF STEROID-REFRACTORY, SEVERE CHRONIC GRAFT- VERSUS- HOST DISEASE CASES, TREATED WITH RUXOLITINIB- A SINGLE-CENTER EXPERIENCES

Ewa Karakulska-Prystupiuk 1, Agnieszka Tomaszewska1, Piotr Boguradzki1, Piotr Kacprzyk1, Grzegorz Władysław Basak1

1Medical University of Warsaw, Warsaw, Poland

Background: Ruxolitinib (RUXO)-a JAK kinase inhibitor has been recently approved for SR-cGvHD.

Methods: A retrospective analysis of patients after allo-HSCT under the care of a single-center. The diagnosis of SR- cGvHD and the assessment of the severity of the disease were made based on the NIH2020 criteria, and treatment toxicity- on the CTCAE5.0 criteria. Patients gave written consent to treatment.

Results: Nine patients with SR-cGvHD treated with RUXO were identified. The patients underwent allo-HSCT for hematological malignancies, after myeloablative conditioning, (median-7 years before RUXO). All patients were diagnosed with severe SR-cGvHD (histopathologically confirmed in 5/9) despite at least 3 lines of prior treatment.

RUXO was administered at doses of 10-20 mg/day, depending on the choice of antifungal prophylaxis, the course of the disease or overlapping infections. RUXO was used for 3-24 months(median-10). All patients, except one, have been concurrently receiving other immunosuppressive drugs (5-tacrolimus, 1-sirolimus, 1-MMF, 2-MTX, 1-belumosudil and 5-corticosteroids).

Patient#1(30yo) cGvHDgrading: eyes-3, lung-2, joints-2, and the second episode of severe steroid- resistance nephrotic syndrome. Within 6 weeks of RUXO, a significant reduction in edema (by 20 kg), a decrease in proteinuria (from 13.5 to 5.5 g/day) and an improvement in eGFR were observed. The treatment was complicated by severe normocytic anemia and RUXO has been discontinued.

Patient#2(21yo) cGvHDgrading: skin-1, lung-3(bronchiolitis obliterans), treated chronically for 15 months with belumosudil with clinical improvement but without improvement in respiratory parameters. RUXO was introduced 9 months later. Tolerance of this complex treatment was good, but we did not observe any improvement in spirometry during 6 months of follow-up.

Patient#3(29yo) cGvHDgrading: skin-3, mouth-2, eyes-1, GI-3, lung-1, joints-3. He suffered from repeated intestinal obstructions caused by a critical stricture of the small intestine. He underwent laparotomy with the excision of this segment. Two years later, an MRI revealed a new intestinal obstruction. The patient started with RUXO, which was continued for 24 months. After treatment, clinical improvement was achieved, and recurrent symptoms of intestinal obstruction disappeared, but the radiological picture has not improved significantly.

Patient#4(41yo) cGvHDgrading: skin-1, GI-3, repeated diarrhea, anorexia, malabsorption, thickening of the intestinal wall, and intestinal stenosis on MRI. Clinical improvement was achieved after 12months of RUXO, but radiological results showed no significant improvement.

Patient#5(35yo)cGvHDgrading: skin-3 (deep sclerotic features) mouth-1, joints-2, history of constrictive pericarditis. During 24 months of RUXO, skin lesions decreased (from 3 to 2), joint mobility improved (from 2 to 1) with a 7-point reduction in the Lee cGVHD symptom scale.

Patient#6(50yo)cGvHDgrading: skin-3 (multiple morphea-like eruptions and lichen sclerotic lesion), lung-1. After 11- months of RUXO only stabilization of changes was achieved.

Patients:#7(66yo),#8(47yo),9#(45yo)cGvhD with a predominant skin involvement-grade 3 (lichen planus, ichthyosis, erythema) and GI–grade 1. After 7-10 months of RUXO, GI symptoms disappeared, and skin lesions have been decreasing (from 3 to 2).

Conclusions: RUXO may be effective in the treatment of classic SR-cGvHD with skin involvement and in nephrotic syndrome. Marked clinical improvement in the symptoms from the GI tract was observed, without impact on radiological features. Combined treatment of bronchiolitis obliterans with RUXO and belumosudil did not succeed.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P283 BELUMOSUDIL FOR SEVERE, REFRACTORY CHRONIC GVHD IN PEDIATRIC PATIENT: CASE REPORT

Agnieszka Sobkowiak-Sobierajska 1, Maksymilian Deręgowski1, Sandra Rutkowska1, Jacek Wachowiak1

1Poznan University of Medical Sciences, Poznan, Poland

Background: Steroid resistant chronic graft versus host disease (cGvHD) is a serious and potentially life-threatening complication of allogenic hematopoetic stem cell transplantation (allo-HSCT).

Methods: Here we report a 16-year-old boy with multidrug resistant cGvHD who was successfully treated with belumosudil.

Results: Patient underwent allo-HSCT from matched unrelated donor due to myelodysplastic syndrome. On day +44 skin aGvHD was diagnosed and it was successfully treated with steroids. On day +98 the boy presented with gastrointestinal tract aGvHD. There was no response after intial methylprednisolon and oral budesonid therapy and on day +105 we started extracorporeal photopheresis (ECP). After 8 procedures gradual and slow resolution of aGvHD symptoms was observed. He continued ECP for 14 months. Eight months after allo-HSCT new skin lesions and signs of liver involment occurred – exacerbation of GvHD was diagnosed and ruxolitinib 20 mg/day was added. Hepatic GvHD resolved and maculopapular skin lesions gradually decreased over 5 months, but in the meantime multiple sclerotic plaques occurred all over his skin. Joint contractures started around 13 months after allo-HSCT. At that moment patient was given rituximab with no clinical improvement. PFT revealed obturative changes (FEV1=32%), on chest CT scan there were discrete ground-glass opacities. The boy became extremely cachectic. Lung GvHD was diagnosed and he started with methotrexate and steroid therapy – intially methylprednisolone 1 mg/kg/day, then steroid pulses. Ruxolitinib doses were increased. The therapy was complicated with serious infections (CMV, Clostridium difficile, Klebsiella pneumoniae sepsis), follow-up CT scan and PFT revealed cGvHD progression in lungs, sclerodermatous skin changes worsened. The treatment was discontinued and imatinib with MMF was started. After 6 weeks MMF was stopped due to gastrointestinal bleeding, methylprednisolone dose was increased. Chest CT scan and PFT done 3 months after the start of imatinib showed stabilized lung function. Two months later (25 months after allo-HSCT) sudden deterioration of respiratory function occurred – dyspnea and desaturation on walking were observed. Methylprednisolone dose was increased and the decision of belumosudil therapy (200 mg/day) was done. Due to administrative procedures belumosudil was started 2 months later. At that moment the patient presented with severe chronic GvHD (lung score 3, skin score 3, joints and fascia score 3, eyes score 1, mouth score 1). Five weeks after the start of belumosudil significant improvement in respiratory capacity was observed – the boy was able to walk a short distance. Over next months gradual improvement in skin and joints was observed. At the follow-up visit 12 months after the start of belumosudil partial response to the therapy was found: lung score was 3 (but with improved physical activity), skin score 2 (with mostly superficial sclerotic features), joints and fascia score 1-2, eyes score 1, mouth score 0. No drug toxicity was observed during the whole treatment period. Patient remains in complete remission of myelodysplastic syndrome.

Conclusions: Belumosudil may effectively treat pediatric patients with severe cGvHD, especially the sclerotic subtype, also those who did not respond to several prior therapies along with its acceptable toxicity profile.

Disclosure: All authors declare that they have no conflicts of interest.

12: Graft-versus-host Disease – Clinical

P284 CLINICAL EVOLUTION OF PATIENTS WITH GRAFT-VERSUS-HOST DISEASE - THE EXPERIENCE OF A SINGLE CENTER

Ana Salselas 1, Pedro Leite-Silva2,1, Sara Lopes1, Lucia Vieira1, Susana Roncon1

1Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal, 2Group of Epidemiology, Results, Economy and Management in Oncology, IPO Porto Research Center (CI-IPOP), Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal

Background: Graft-versus-host disease (GvHD) continues to be the main cause of morbidity and mortality after allogeneic transplantation, significantly impacting the patient’s quality of life.

Methods: We carried out a retrospective analysis (2016 to 2023) of patients undergoing extracorporeal photopheresis (ECP) treatments due to refractoriness to previous corticosteroid therapy, in a total of 17 patients. To identify predisposing factors to the occurrence of GvHD, we conducted a descriptive, observational cohort study, analyzing data from the respective donor (age, HLA compatibility, sex, and degree of kinship), patient characteristics (presence of measurable disease) and graft (namely, number of CD3+ cells infused). This analysis was carried out from the records of the patient´s clinical records.

Results: Of the 17 patients with GvHD, 11 have died and 6 are still alive. The living patients showed heterogeneity in the donor’s characteristics and the clinical evolution to the chronicity of GvHD. After a period of ECP treatments, patients are currently on Ruxolitinib therapy and all patients remained free of the primary disease. Contrary to what is described in the literature, the patient with the lowest amount of CD3+ infused (1.3 x 10^8/Kg) developed GvHD earlier (15 days); the patient whose donor was the youngest (27 years) developed GvHD within 120 days; the patient whose donor was HLA 9/10 (A-disparity) developed GvHD 3390 days (10 years) after D0; the patient with measurable disease [molecular positive (0.2868%)] did not have relapse of primary disease (acute lymphoblastic leukemia – B) and had a good response to ECP. It is not possible to conclude the donor’s gender preference, as cases varied between unrelated male donors (3) and related female donors (3). The main cause of suspension of ECP was the difficulty in puncture of peripheral venous accesses. In patients who underwent ECP via a central venous catheter (CVC), there was difficulty in maintaining the CVC, with consequent contamination, infection, and sepsis.

About half of the patients improved with ECP and all patients maintained or improved staging with Ruxolitinib.

Full size table

.

Conclusions: Despite the small sample size, there are relevant results that should be the subject of a more in-depth study in collaboration with other clinical teams and research centers that address this issue. The knowledge acquired can contribute to the improvement of our Centre’s performance in providing care during the acute and chronic phases of GvHD.

The issue of peripheral venous access puncture requires a new approach, highlighting the need for research and development of new techniques for venous accesses that allow ECP treatment, considering the efficacy demonstrated in most patients.

It is also proposed to evaluate the ECP associated with Ruxolitinib as a treatment with more effective results in the involvement of organs in GvHD and the consequent improvement of the patient’s quality of life.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P285 USE OF MYCOPHENOLATE MOFETIL FOR GRAFT VERSUS HOST DISEASE PROPHYLAXIS IN PATIENTS UNDERGOING HEMATOPOIETIC STEM CELLS TRANSPLANTATION FROM UNRELATED DONORS

Davide Pio Abagnale1, Andrea Cacace1, Giuseppe Gaeta1, Maria Luisa Giannattasio1, Linda Piccolo1, Francesco Grimaldi1, Simona Avilia1, Mara Memoli1, Lucia Ammirati2, Valentina Maglione2, Alessandro Severino1, Catello Califano2, Fabrizio Pane1, Giorgia Battipaglia 1

1Federico II University of Naples, Naples, Italy, 2A. Tortora Hospital, Pagani, Italy

Background: Graft versus host disease (GVHD) prophylaxis in unrelated donor transplantation (UD-HSCT) mainly relies on the use of a calcineurin inhibitor in association to either methotrexate (MTX) or mycophenolate mofetil (MMF) when a myeloablative (MAC) or reduced-intensity (RIC) conditioning regimen is used, respectively. Concerns about the higher risk of GVHD with MMF compared to MTX exist. However, the implementation of GVHD prophylaxis through the addition of antithymocyte globulin (ATG) or post-transplant cyclophosphamide (PTCy) may reduce this complication.

Methods: We report transplantation outcomes in patients undergoing 9/ or 10/10 UD-HSCT and receiving cyclosporine A (CsA), MMF with the addition of PTCy or ATG as GVHD prophylaxis. MMF was administered at a total dose of 3 grams per day from day -3 when it was associated with CsA and ATG, and from day +6 when it was associated with CsA and PTCy. The drug was discontinued at day +35.

Results: We report 17 consecutive patients (6 males, 11 females) who underwent UD-HSCT (10/10, n=12; 9/10, n=5) in our center from July 2020 to November 2023. Median age at transplant was 51 (range 26-66) years. Main diagnosis were AML (n=13), ALL (n=3), MDS/MPN (n=1). All but 2 patients with active disease, were transplanted in CR (CR1, n=12; CR2, n=3). Sorror score was >2 in one patient. Stem cell source was peripheral blood in all but one patient. ATG was added in most cases (10/10, n=12; 9/10, n=1) to CsA and MMF, while PTCy was used in the remaining 4. Conditioning regimen was myeloablative in 12, reduced-intensity in 3, sequential in 2 patients. Thirteen patients (76%) experienced mucositis, including 5 grade 3. Neutrophil and platelet engraftment occurred in all patients with a median time of 14 (range 9-23) and 17 (range 8-44) days, respectively. Acute GVHD (aGVHD) of all grades occurred in 5 patients, including 1 grade 3-4. Chronic GVHD occurred in 2 patients, one being extensive. Relapse occurred in 3 patients, with one undergoing a second allo-HSCT after CR2 obtention. Four patients died, 2 due to GVHD, one due to disease relapse and one due to unknown causes. With a median follow-up of 17 (range 4-40) months, 1-year PFS, OS and GRFS were 64±14%, 81±10% and 56±14%, respectively.

Conclusions: When adding ATG or PTCy to CsA, addition of MMF was feasible and resulted in no major toxicities and in acceptable GVHD incidence in UD-HSCT with both MAC and RIC regimens.

Disclosure: Nothing to declare.

12: Graft-versus-host Disease – Clinical

P286 RETROSPECTIVE ANALYSIS OF RUXOLITINIB TREATMENT FOR THE MANAGEMENT OF STEROID REFRACTORY GRAFT VERSUS HOST DISEASE IN ADULTS WITH HEMATOLOGIC MALIGNANCIES

Bogar Pineda Terreros1, Araceli Leal Alanis1, Brenda Lizeth Acosta Maldonado1, Luis Manuel Valero Saldaña1, Rubén Solís Armenta 1, María Alejandra Núñez Atahualpa1, Anghela Milenka Mendoza Sánchez1, Liliana Mey Len Rivera Fong1

1Instituto Nacional de Cancerología, Mexico City, Mexico

Background: Allogenic hematopoietic stem cell tansplantation (HSCT) improves survival in patients with hematologic malignancies. However, acute Graft Versus Host Disease (aGVHD) and chronic Graft Versus Host Disease (cGVHD) represent major causes of morbidity and mortality. Up to 60% of patients with aGVHD and 50% of patients with cGVHD may be refractory to steroids. Ruxolitinib has demonstrated efficacy in the management of this conditions as second line therapy.

Methods: Retrospective and descriptive analysis of Ruxolitinib treatment for steroid refractory aGVHD and cGVHD in a single center.

Results: A total of 16 patients with steroid refractory GVHD were analyzed. Median follow up was 18.6 months (2 - 52). Median age at HSCT was 34 years old. Indications for HSCT were high risk acute lymphoblastic leukemia (50%), high risk acute myeloblastic leukemia (37.5%), Relapsed Hodgkin lymphoma (6.3%) and high risk myelodysplastic neoplasia (6.3%). Eight patients (50%) received matched sibling donor HSCT and 8 (50%) haploidentical HSCT. Mieloablative conditioning and reduced intensity conditioning were used in 50% of patients each. Ten patients (62.5%) developed both aGVHD and cGVHD. aGVHD presented in 13 patients (81.4%), with Grade I in 31%, Grade II in 38%, Grade III in 23% and Grade IV in 8%; the most common affected organ in aGVHD was skin. Of the patients who developed aGVHD, 6 had involvement of 1 organ (46%) and 7 of 2 organs (54%). cGVHD developed in 13 patients (81.3%), with mild cGVHD in 31%, moderate cGVHD in 61% and severe cGVHD in 8%; the most common affected organ was liver. Of the patients who developed cGVHD, 5 had involvement of 2 or less organs and 8 involvement of 3 or more organs.

Ruxolitinib was indicated for steroid refractory aGVHD in 4 patients (25%), for steroid refractory cGVHD in 11 patients (68.8%) and 1 patient (6.3%) for both aGVHD and cGVHD.

Overall response was observed in 87.5%. Of the 5 patients with Ruxolitinib treatment for steroid refractory aGVHD, 3 patients (60%) achieved complete response, 1 patient (20%) partial response and 1 patient (20%) progressed. Of the 12 patients with Ruxolitinib treatment for steroid refractory cGVHD, 5 patients (41.5%) achieved complete response, 6 patients achieved partial response (49.8%) and 1 patient (8.3%) progressed. In steroid refractory aGVHD, median time to achieve partial response was 10.75 days (3 – 20), and median time to complete response was 38 days (3 – 104). In steroid refractory cGVHD, median time to partial response was 33 days (3 – 117), and median time to complete response was 253 days (42 – 905).

Improved survival was observed when complete response with Ruxolitinib was reached. One patient had post-transplant relapse and 2 died while on Ruxolitinib treatment.

Half of patients developed infections after Ruxolitinib therapy, most of them grade 3 (43.8%).

Conclusions: Ruxolitinib is an effective and safe treatment for steroid refractory aGVHD and cGVHD.

Overall responses were more than 80%, with a half of patients achieving complete response.

Most infections related to this treatment were grade 3 or less. It is important to consider the risk of infections when Ruxolitinib is indicated.

Disclosure: Authors have nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P287 DR3 AND GITR AGONISTS BOOST ENDOGENOUS TREG POOL TO PREVENT ACUTE GVHD

Juan Fernando Gamboa Vargas 1, Isabell Lang2, Svetlana Stepanzow2, Olena Zaitseva2, Estibaliz Arellano Viera1, Carolin Graf1, Haroon Shaikh1, Muhammad Daud1, Hermann Einsele3, Harald Wajant2, Andreas Beilhack1

1University Clinic Würzburg Interdisciplinary Center for Clinical Research, Würzburg, Germany, 2University Clinic Würzburg Division of Molecular Internal Medicine, Würzburg, Germany, 3University Clinic Würzburg, Wüzrburg, Germany

Background: The tumor necrosis factor superfamily (TNFRSF) receptors (TNFRs) play a pivotal role in orchestrating the immune system, tissue repair and homeostasis. Recent studies with tumor necrosis factor receptor 2 (TNFR2, CD120b, TNFRSF1B) agonists identified this TNFR as a promising therapeutic target on regulatory T cells (Tregs) to prevent acute graft-versus-host disease (aGvHD) in preclinical mouse models of allogeneic hematopoietic cell transplantation (allo-HCT). Here we investigated in mouse and human systems the usefulness of two other TNFRs as targets on T cell subsets to regulate acute graft-versus-host disease (aGvHD).

Methods: To investigate the function of DR3 and GITR, we used fusion proteins of the DR3 ligand TL1A and of the GITR ligand GITRL along with a ligand-based TNFR2 agonist as benchmark. Employing human and mouse in vitro assays, and in vivo MHC-mismatched allo-HCT model (9 Gy myeloablative conditioning, H2-Kq→H2-Kb, 6x105 T cells plus 5x106 bone marrow cells), we conducted comprehensive analyses, including flow cytometry, gene expression profiling, cytokine measurements, immunofluorescence microscopy, and histopathologic evaluation.

Results: First, we observed elevated expression levels of DR3, GITR and TNFR2 in mouse splenic Treg cells that significantly exceeded those of conventional CD4 T cells. Second, we treated mouse and human T cell cultures with DR3-, GITR-, and TNFR2-agonists. Agonist treatment significantly expanded Tregs (human: DR3 agonist 1.5-fold, human GITR agonist 1.3-fold, TNFR2 agonist 1.2-fold, P ≤ 0.001; murine: DR3 agonist 1.6-fold, GITR agonist 2.09-fold, TNFR2 agonist 1.6-fold, P ≤ 0.05). Third, we treated mice with equal amounts of agonistic Fc(DANA) or IgG1(N297A) fusion proteins of DR3-, GITR- and TNFR2-specific ligand molecules, or an irrelevant-isotype control antibody. Notably, these treatments significantly expanded Tregs 3- to 4-fold in comparison to controls. Next, we treated mice with these agonists 4 days before allo-HCT (n=8/group). Intriguingly, in all the three agonist treated groups expanded Tregs persisted after transplantation and the treated mice displayed reduced aGvHD symptoms and, importantly, reduced mortality. Histopathological examination revealed particularly in the TNFR2-agonist treated mice reduced tissue damage, which was less pronounced in DR3- and GITR-treated recipients. Remarkably, DR3 agonist treated allo-HCT recipients displayed 100% survival while none of the controls survived 40 days after allo-HCT. Also, TNFR2 agonist treatment (80% survival) and GITR agonist treatment (40% survival) improved survival significantly. However, the latter group developed significantly more skin-GvHD symptoms than DR3- and TNFR2 agonist treated mice.

Conclusions: These findings highlight the potential for next-generation TNFR agonists for expanding and activating Tregs to avert aGvHD. Particularly, agonists for DR3 and TNFR2 proved powerful to prevent aGvHD in our preclinical models. Targeting the endogenous Treg pool emerges as a potent strategy for restoring tissue homeostasis and immunotolerance in patients undergoing allo-HCT.

Disclosure: The University of Würzburg has filed several patents and patent applications for TNFR-agonists with H.W., I.L. and A.B. as co-inventors. H.W. and A.B. are consultants for and have received research funding from Dualyx NV. H.W. and A.B. are founders of TrimmunoTec GmbH.

11: Graft-versus-host Disease – Preclinical and Animal Models

P288 ROLE OF BCL2 IN GRAFT VERSUS HOST DISEASE

José Antonio Bejarano García1,2, Melanie Nuffer1,2, Mª José Palacios Barea1,2, Luzalba Sanoja Flores1,2, Mª Reyes Jiménez León1,2, Javier Delgado Serrano2, Teresa Caballero Velázquez2,1,3, Alfonso Rodríguez Gil1,2,3, José Antonio Pérez Simón 2,1,3

1Instituto de Biomedicina de Sevilla (IBiS), Sevilla, Spain, 2Hospital Universitario Virgen del Rocío (HUVR), Sevilla, Spain, 3Universidad de Sevilla, Sevilla, Spain

Background: One of the fundamental mechanisms to control homeostasis in the immune system is the control of apoptosis, where Bcl2 family proteins play a fundamental role. These are differentially expressed in immune populations and are under a complex regulation system depending on the state of lymphocyte maturation and activation. The role that these proteins may play in the pathophysiology of GVHD is unexplored, and they might potential therapeutic targets.

Methods: Murine models of acute GVHD have been used using animals inbreds that overexpress Bcl2 in hematopoietic cells as donors. In addition, the effect of Venetoclax (Bcl2 inhibitor) has been evaluated. In vitro, activated or resting T lymphocytes have been treated with Venetoclax, and viability, proliferation and expression of Bcl2 proteins have been measured by cytometry and western blot (WB). Finally, the expression of Bcl2 in lymphocytes from 37 patients undergoing allo-HSCT and in 11 healthy controls was quantified by flow cytometry.

Results: Using murine models we demonstrate that the use of Venetoclax post-transplant leads to greater mortality related to GVHD. Cytometry, WB and coimmunoprecipitation assays show that resting T lymphocytes are exclusively dependent on Bcl2 for their survival, which is confirmed in in vitro assays that show a high sensitivity to Venetoclax; On the contrary, under activation, T cells are dependent on Mcl1 and Bcl-xL, being resistant to Venetoclax. Using a Bcl2 overexpression model, we observed that Bcl2 overexpression reduces the levels of ROS and cytotoxicity markers such as CD107a in T lymphocytes. Moreover, overexpression of Bcl2 in donor lymphocytes decreased the incidence and severity of GVHD in a dose-dependent manner. This finding is confirmed ex vivo in samples from 37 patients undergoing transplantation in which a greater expression of Bcl2 in T lymphocytes related to a lower risk of GVHD.

Conclusions: Higher expression of Bcl2 in donor T lymphocytes is related to a lower cytotoxic effect and less post-transplant GVHD, so the use of post-transplant Bcl-2 inhibitors should be carried out with caution in patients undergoing allogeneic transplant.

Disclosure: The work was funded by AbbVie Pharmaceuticals.

11: Graft-versus-host Disease – Preclinical and Animal Models

P289 DIMETHYL FUMARATE INHIBITS TFH DIFFERENTIATION AND CGVHD VIA NRF2

Huanle Gong 1, Fulian Lv1, Yang Xu1, Depei Wu1

1The First Affiliated Hospital of Soochow University, Suzhou, China

Background: We have previously reported that Dimethyl fumarate (DMF) effectively inhibits acute GVHD (aGVHD) and preserves graft-versus-leukemia (GVL) response, while the role of DMF in chronic GVHD (cGVHD) progression remains unknown.

Methods: To establish scleroderma-like cGVHD model, lethally irradiated BALB/C recipients were injected with 2.5 × 10^6 T cell-depleted bone marrow cells (TCD-BM) and 5 × 10^5 splenocytes from C57BL/6 mice. For systemic lupus-like cGVHD, C57BL/6 or BALB/c recipients were injected with 1 ×10^7 splenocytes from bm12 mice or 5 ×10^6 BMs together with 3 × 10^7 CD25- splenocytes from DBA2 mice, respectively. For xenograft GVHD, NSG-A2+ mice were transplanted with HLA-A2- human PBMCs. To explore the potential mechanisms, we profiled the immune cell responses in thymus and splenocytes by flow cytometry 6-8 weeks post transplantation. Murine and human naïve CD4 + T cells were sorted to detect the role of DMF on Tfh development. RNA-Seq and ATAC-seq was analyzed to investigate the mechanism of DMF involved in Tfh differentiation. Expression of indicated genes were determined by real-time PCR and immunofluorescence staining. Luciferase reporter assay was performed to assess the regulation of Nrf2 on IL-21 transcription. To validate the therapeutic potential of DMF in clinical cGVHD, peripheral blood mononuclear cells (PBMCs) from cGVHD patients were isolated and treated with DMF. Proportions of Tfh cells and cytokines production were assessed by flow cytometry.

Results: The survival of receipts administrated with DMF was significantly prolonged than controls, companied by a lower cGVHD clinical score. Percentages of CD4 + CD8+ cells in thymus were significantly elevated in recipients with DMF. Tfh cells and GC B cells were dramatically reduced in both scleroderma-like and lupus-like cGVHD model after DMF treatment. However, DMF treatment didn’t affect the populations of Th1, Th2, Th17 and Tregs. In vitro studies revealed that DMF directly inhibits both murine and human Tfh development by a dose depedent manner. RNA-seq showed that DMF significantly down-regulates Tfh cell-related genes, including Tcf7, Il6ra, Batf, klf2 and Il21, while up-regulates prdm-1 expression. Integrated RNA-seq and public Tfh ATAC-seq datasets, we found DMF reduced the accessibility of IL-21 gene, while enhanced the accessibility of prdm-1. Luciferase reporter assay showed Nrf2 significantly repressed the transcript activity of IL-21, but not prdm1. The inhibitory role of DMF on Tfh development were diminished after Nrf2 depletion. Interestingly, DMF treatment leads to a striking decrease in the frequency of IL-21 secretion and Tfh generation in human cGVHD PBMCs. Morepver, DMF significantly mitigates xenograft GVHD development.

Conclusions: DMF significantly restrains Tfh cell responses and mitigates cGVHD development, rather than interrupted extrafollicular CD4 T-cell differentiation. DMF treatment orchestrates Tfh transcriptional profile. DMF reduces IL-21 transcription by argument of Nrf2 expression, leading to a defective Tfh differentiation. Importantly, DMF treatment significantly suppresses IL-21 secretion and Tfh cell generation in PBMCs of active cGVHD patients. Collectively, our results revealed that DMF potently inhibits cGVHD development by repressing Tfh differentiation via Nrf2, paving way for the treatment of clinical cGVHD.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P290 SPECIFIC INHIBITION OF PATHOLOGICAL ANGIOGENESIS TO DECREASE ACUTE GVHD

Katarina Riesner 1, Lydia Verlaat1, Sarah Mertlitz1, Angela Jacobi2, Hadeer Mohamed Rasheed1,3, Pedro Vallecillo Garcia1, Beate Anahita Jung1, Ningyu Li1, Martina Kalupa1, Claudine Fricke1, Bertina Mandzimba-Maloko1, Laura Barrero1, Lars Bullinger1, Olaf Penack1

1Charité Universitätsmedizin, Berlin, Germany, 2Technische Universität, Dresden, Germany, 3Alexandria University, Alexandria, Egypt

Background: Immunosuppressive strategies to prevent or treat aGVHD are in standard clinical use despite their risk of facilitating infections and tumor relapse after allo-SCT. We have previously demonstrated pathological angiogenesis during acute GVHD (aGVHD), providing the rationale to use anti-angiogenic strategies. The major hurdle for a translational development is that current anti-angiogenic drugs are non-specific, meaning that they also inhibit physiological angiogenesis needed for regeneration and tissue repair after allo-SCT. Therefore, we searched targets that allow a specific inhibition of pathological angiogenesis during aGVHD while keeping physiological angiogenesis intact. In our previous work, we identified genes in the fatty acid metabolism (Cpt2, carnitine palmitoyltransferase II) and the pentose phosphate pathway (G6pd, glucose-6-phosphate dehydrogenase) to be differentially upregulated during aGVHD-related pathological angiogenesis (Blood. 2017 Apr 6;129(14):2021-2032).

Methods: We performed experiments in vitro and in preclinical aGVHD models using genetic as well as therapeutic inhibition of the target genes (Cpt2, G6pd) following the hypothesis that inhibiting pathological angiogenesis will lead to improved aGVHD.

In vitro: Cpt2 and G6pd were inhibited in endothelial cells (MCEC, HUVEC, TMNK1) genetically via CRISPR technology and therapeutically via perhexiline (1, 2.5 and 5 µM) and polydatin (300-1000 µM). Endothelial cell behavior was measured under “physiological” conditions (without stimulation) and under “pathological” conditions (stimulation with alloreactive T cells and serum) using MTT, Scratch, Fibrin 3D sprouting and tube formation assay. Metabolic phenotype of endothelial cells was assessed via Seahorse technology.

In vivo: We used carrageenan-induced paw edema and different established chemotherapy-based aGVHD mouse models (129→B6, B6→BDF) of genetically inhibited CPT2VE−/− mice (endothelial specific Cpt2 knockout) and therapeutically inhibited mice by administering i.p. 6 mg/kg/day perhexiline or 100 mg/kg/day polydatin. aGVHD was monitored by clinical scoring and by histopathological analysis of tissue sections. Tissue infiltration and vessel density was analyzed via fluorescence staining against CD4, CD8, CD31 and mechanical phenotype of endothelial cells was assessed via Microfluidic microcirculation mimetic.

Results: Under stimulated conditions mimicking pathological conditions of aGVHD, the therapeutic as well as genetic inhibition of Cpt2 and G6pd showed in in vitro assays profound reduction of endothelial cell dysfunction. Under pathological conditions, sprouting and tube formation as well as proliferation, apoptosis and activation were profoundly changed, combined with an altered endothelial metabolic phenotype. In preclinical models, the therapeutic inhibition of Cpt2 and G6pd showed anti-inflammatory effects in a model of paw edema as well as improved aGVHD mortality and morbidity in 2 separate aGVHD models by reducing target organ aGVHD and T cell infiltration. Reduced aGVHD morbidity was confirmed in endothelial-specific knockout mice of Cpt2. Reduced aGVHD was achieved by targeting early angiogenesis as Cpt2 inhibition led to decreased angiogenesis during early- and established aGVHD (d + 2, and d + 14). Finally, treatment normalized mechanical properties of endothelial cells from aGVHD mice.

Conclusions: Our data indicate that Cpt2 (fatty acid metabolism) and G6pd (pentose phosphate pathway) are involved in endothelial mechanisms leading to pathological angiogenesis and can be therapeutically targeted during aGVHD. Our results support the translational development of therapeutic strategies targeting pathological angiogenesis after allo-SCT.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P291 ENDOTHELIAL-TO-MESENCHYMAL TRANSITION IN CHRONIC GVHD

Katarina Riesner 1, Lydia Verlaat1, Sarah Mertlitz1, Hadeer Mohamed Rasheed1,2, Pedro Vallecillo Garcia1, Beate Anahita Jung1, Ningyu Li1, Martina Kalupa1, Claudine Fricke1, Bertina Mandzimba-Maloko1, Lars Bullinger1, Bruce Blazar3, Olaf Penack1

1Charité Universitätsmedizin, Berlin, Germany, 2Alexandria University, Alexandria, Egypt, 3University of Minnesota, Minneapolis, United States

Background: Current immunosuppressive chronic GVHD (cGVHD) treatments often cause side effects and rarely lead to complete resolution of severe disease forms. cGVHD is characterized by fibrosis and vascular rarefaction. Our understanding as to how this typical cGVHD phenotype develops, progresses or resolves is rudimentary hampering treatment strategies. Using established pre-clinical cGVHD models closely resembling the human phenotype (PMID: 36761159), we sought to investigate the responsible mechanisms and contribution of cGVHD-induced vascular damage to target organ fibrosis.

Methods: Human data: Serum samples obtained between 2014 and 2019 at Charité University Hospital Berlin from alloSCT patients diagnosed by NIH cGVHD criteria were analyzed by ELISA for indicators of fibrosis [transforming growth factor- β (TGF-β)] and endothelial injury [endoglin, follistatin, endostatin, connective tissue growth factor (CTGF) and endocan / endothelial-cell-specific molecule 1].

Murine models: Humanized (human PBMCs→NSG mice) and murine (B6→BDF) cGVHD models were employed. Distinct features included GCSF mobilized stem cells and splenocytes (murine model) and long experimental times (both models) allowing the development of a human-like phenotype in lung, skin, liver, and colon. cGVHD was monitored by clinical scoring and fibrotic cGVHD assessed by day+130 tissue histopathology. Endothelial dysfunction was characterized via immune fluorescence staining and flow cytometry against molecules displaying endothelial function [CD31, VE-Cadherin], endothelial damage [von-Willebrand factor (vWF)], fibrotic function [alpha-smooth muscle actin (aSMA), Fibroblast-specific protein-1 (FSP-1)], and vascular integrity [tight junction protein Zonula occludens protein-1 (ZO-1) and pericyte Neuron-glial antigen 2 (NG2)]. Endothelial-to-mesenchymal transition (EndoMT), leading to the formation of profibrotic myofibroblasts and smooth muscle cells, was determined via TGF-β stimulating assays of endothelial cells in vitro and ex vivo as well as flow cytometry and qPCR analysis of mesenchymal and endothelial cell markers. Lung and liver of cGVHD animals treated with the SMAD2/3 inhibitor, pirfenidone, that has anti-inflammatory, anti-fibrotic and antioxidant functions, were characterized for endothelial dysfunction as described above.

Results: Serum endothelial and fibrotic biomarkers such as TGF-β, endoglin, follistatin, CTGF and endocan were significantly increased in cGVHD patients indicating endothelial alterations during cGVHD. In line, endothelial dysfunction and damage was found in preclinical cGVHD, characterized by a loss of endothelial CD31 and VE-Cadherin as well as an increase of vWF. Endothelial integrity was disturbed by the loss ZO-1 and pericyte marker NG2 and circulating endothelial cells and endothelial progenitor cells occurred in cGVHD mice. Severe endothelial fibrosis was detected in cGVHD by co-staining of endothelial CD31 and mesenchymal aSMA and FSP-1. Mechanistically EndoMT was detected. Isolated lung and liver endothelial cells from cGVHD mice exhibited reduced levels of endothelial markers and increased levels of mesenchymal markers and were more prone to differentiate ex vivo after TGF-β stimulation. This transition was successfully prevented by administration of pirfenidone (a potent inhibitor of EndoMT) in preclinical cGVHD, which normalized the CD31+ vessel density and decreased endothelial fibrosis.

Conclusions: Our data indicates that EndoMT contributes to fibrosis and vascular rarefaction during cGVHD. This opens the opportunity to test treatment strategies for preventing or reversing vascular damage and associated EndoMT and fibrosis during cGVHD.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P292 RAPID ADAPTATION OF DONOR REGULATORY T CELLS (TREG) TO RECIPIENT TISSUES FOR THE CONTROL OF ACUTE GRAFT-VERSUS-HOST DISEASE (GVHD)

Franziska Pielmeier 1, David J. Dittmar1, Nicholas Strieder2, Alexander Fischer1, Michael Herbst1, Hanna Stanewsky1, Niklas Wenzl2, Eveline Röseler2, Rüdiger Eder1, Claudia Gebhard2, Lucia Schwarzfischer-Pfeilschifter1, Christin Albrecht1, Wolfgang Herr1, Michael Rehli1,2, Petra Hoffmann1,2, Matthias Edinger1,2

1University Hospital Regensburg, Regensburg, Germany, 2Leibniz Institute for Immunotherapy, Regensburg, Germany

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) carries the risk of graft-versus-host-disease (GvHD) caused by alloreactive conventional T cells (Tconv) in the graft. The adoptive transfer of donor CD4+CD25+Foxp3+ regulatory T cells (Treg) prevents the development of lethal acute GvHD and ameliorates ongoing disease, as shown by us and others in murine models as well as first clinical studies. Here, we explored the early selection and adaptation processes of in vitro expanded donor Treg during migration to lymphoid and non-lymphoid GvHD target organs to define pre-requisites for improved therapeutic interventions.

Methods: Splenic CD4+CD25+CD62L+ Treg cells were isolated from C57BL/6 WT or Foxp3gfp reporter mice by flow cytometric cell sorting and in vitro expanded for 11d through stimulation with anti-CD3/CD28-coated beads (polyTreg) or host-type (BALB/c) dendritic cells (alloTreg) in the presence of IL-2. BALB/c mice were lethally irradiated (8 Gy) and transplanted with 2.5 x 106 T cell-depleted bone marrow cells and 0.25 – 1 x 106 splenic CD4+CD25- Tconv cells from C57BL/6(CD45.1) mice to induce aGvHD. GvHD prophylaxis consisted of co-injection of allo or polyTreg (1:1 ratio with Tconv on d0). Seven days post BMT, donor Treg and Tconv were re-isolated from bone marrow, spleen, mLN, liver and colon of recipient mice and phenotypically and functionally analyzed by multiparametric flow cytometry. Furthermore, we performed bulk and single cell transcriptome and TCR repertoire analysis of donor Treg. To monitor long-term outcome and protection, mice were kept for 60 days, scored in regular intervals and analyzed at the end of the observation period.

Results: Both in vitro expansion protocols generated Treg products with similar phenotype but different TCR repertoire diversity, which was expectedly reduced in allo as compared to polyTreg. Long-term protection from lethal aGvHD was more pronounced with allo as compared to polyTreg and resulted in 94 % versus 60 % survival at d60 after HSCT with only 6.7 % of recipients surviving in the absence of Treg (n=15-16 mice/group). Analysis on d7 after alloHSCT revealed that both donor Treg populations strongly suppress donor Tconv expansion, with alloTreg being more effective than polyTreg. Applying comprehensive phenotypic, gene expression and TCR repertoire profiling, we show that upon migration to non-lymphoid host organs, donor Treg rapidly acquire organ-specific gene expression profiles resembling those of their tissue-resident counterparts. In addition, they upregulate a broad range of suppressive programs in the context of aGVHD, including several cytotoxic effector molecules such as granzymes and perforin. Dominant clonotypes were detected across different organs and even across different recipients, indicating that early migration and aGvHD protection in this complete MHC-mismatched mouse model is mainly driven by ubiquitously expressed allo-antigens. In contrast, transcriptional rewiring of the cells during organ infiltration happens largely independent of the TCR specificity but triggered by the respective microenvironment.

Conclusions: This study provides unique insights into donor Treg selection and adaptation in GvHD target organs and highlights protective features of Treg products that guide future approaches to further improve donor Treg-based interventions in GvHD.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P293 INHIBITION OF MYELOPEROXIDASE AMELIORATES MURINE ACUTE GRAFT-VERSUS-HOST DISEASE

Michelle Klesse 1, Sebastian Schlaweck1, Oliver Schanz1, Peter Brossart1, Annkristin Heine1

1University Hospital Bonn, Bonn, Germany

Background: Acute Graft-versus-Host disease (aGvHD) remains a common and potentially lethal complication after allogeneic hematopoietic cell transplantation (allo-HCT). Although classically described as a T cell-driven disease, neutrophils have recently emerged as possible early key players in the development of experimental aGvHD (Schwab et al., 2014; Hülsdünker et al., 2018). Specific inhibition of myeloperoxidase (MPO), an enzyme abundantly expressed by neutrophils, was previously shown to alleviate inflammation in cardiovascular diseases and rheumatoid arthritis (Tiyerili et al., 2016). Here, we investigated the inhibition of MPO as a possible treatment strategy in a mouse model for aGvHD.

Methods: Recipient BALB/c mice (H-2d) underwent total body irradiation (2x 2,5 Gy, d-1) and were intravenously injected with 5x106 T cell-depleted bone marrow and 6x105 CD3+ splenocytes from sex- and age-matched C57BL6 (H-2b) donor mice (d0). 4-aminobenzoic acid hydrazide (ABAH) was used as an irreversible MPO inhibitor and administered daily via intraperitoneal injection until end of experiment.

Results: We performed bioluminescence imaging of MPO and confirmed early neutrophil infiltration into the small intestine after allo-HCT, as was previously shown by Schwab et al. (2014). MPO activity peaked on d3 and was effectively reduced by ABAH in vivo.

When started prior to conditioning (d-1), ABAH treatment significantly improved the overall survival of recipient mice following allo-HCT compared to vehicle treatment. Administration of ABAH from d7, however, had no effects on overall survival and aGvHD development. ABAH treatment from d-1 also led to reduced weight loss and lower clinical scores in mice compared to vehicle treatment.

Histological examination of target organs revealed less pronounced tissue damage in mice treated with ABAH from d-1, which correlated with decreased plasma levels of liver enzymes AST and ALT. Blood cell counts and donor chimerism remained unaffected by ABAH (> 90 % after d65), suggesting appropriate hematopoietic reconstitution.

We then investigated systemic levels of pro-inflammatory cytokines and chemokines at different time points after allo-HCT. No significant changes could be detected in sera of ABAH-treated compared to vehicle-treated mice. Local effects within target organs will therefore be examined by multiplexed tissue imaging and spectral flow cytometry. These methods will also allow high-resolution analysis of immune cell composition, localization and interaction throughout aGvHD progression.

Conclusions: Overall, our data show that inhibition of MPO prior to conditioning and shortly after allo-HCT significantly improves survival and ameliorates aGvHD disease severity. These results provide further evidence that neutrophils are critically involved in the initiation of aGvHD and may present a promising target for the early intervention in aGvHD development. We further hypothesize that targeting innate immune cells could enable GvHD treatment approaches that preserve donor T cell functionality and, thus, anti-tumor activity.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P294 IFN-GAMMA INDUCES ACUTE GRAFT-VERSUS-HOST DISEASE BY PROMOTING HMGB1-MEDIATED NUCLEAR-TO-CYTOPLASM TRANSLOCATION AND AUTOPHAGIC DEGRADATION OF P53

Xu Yajing 1, Wang Shiyu1, Cheng Tingting1, Chen Xu1, Zeng Cong1, Qin Wei1

1Central South University, Changsha, China

Background: Acute graft-versus-host disease (aGVHD) is a common immune complication that can occur after allogeneic hematopoietic stem cell transplantation (allo-HSCT), which is associated with high morbidity and mortality. Our previous findings revealed that p53 was significantly downregulated in CD4 + T cells from patients with aGVHD compared with the non-aGVHD group. CD4 + T cells with insufficient p53 expression were over-activated and proliferated under stimulation by large amounts of IL-2, mediating inflammatory damage in various organs in aGVHD patients. Interferon (IFN) -γ, a potent proinflammatory cytokine produced by multiple types of cells, is a pivotal regulator of alloreactive T cell responses that mediate GVHD. IFN-γ can induce autophagy in various cell types, which is a vital catabolic process for proteins degradation. However, whether IFN-γ is a potential effector to induce the autophagic degradation of p53 in CD4 + T cells so that to promote the cells proliferation and participate in the development of aGVHD remains unclear.

Methods: Here, we first collected and isolated peripheral blood CD4 + T cells from aGVHD and non-aGVHD patients to detect the autophagic activity and the subcellular distribution of p53 and HMGB1. Then, we detected the effect of IFN-γ on the autophagic activity and proliferation of CD4 + T cells from healthy people. In addition, we detected the effect of IFN-γ and autophagy inhibitors on the expression and distribution of p53 and HMGB1 in normal CD4 + T cells. Finally, we detected the interaction between p53 and HMGB1 in normal CD4 + T cells after IFN-γ stimulation and measured the expression and distribution of p53 in normal CD4 + T cells transfected with HMGB1 interference/control plasmid following IFN-γ treatment.

Results: The plasma IFN-γ level and autophagic activity of CD4 + T cells in patients with aGVHD were higher than that in patients without aGVHD. Compared with non-aGVHD patients, the expression of p53 and HMGB1 in the nucleus of peripheral blood CD4 + T cell from aGVHD patients were significantly decreased, while in the cytoplasm, p53 was decreased and HMGB1 was increased. IFN-γ can induce the activation of autophagic flux in normal CD4 + T cells and promote the proliferation of normal CD4 + T cell by accelerating the nucleus-to-cytoplasm translocation and autophagic degradation of p53. IFN-γ can also induce the nuclear export and cytoplasmic accumulation of HMGB1 in normal CD4 + T cells. Moreover, IFN-γ enhanced the interaction between p53 and HMGB1 in normal CD4 + T cells. After knockdown of HMGB1, IFN-γ-induced nuclear-to-cytoplasm translocation and autophagic degradation of p53 were significantly inhibited.

Conclusions: Increased IFN-γ in the plasma induces the proliferation of CD4 + T cells and aGVHD development by promoting HMGB1-mediated nuclear-to-cytoplasm translocation and autophagic degradation of p53.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P295 DEFINING THE TRANSCRIPTOMIC LANDSCAPE OF ACUTE SKIN GVHD AT SINGLE CELL RESOLUTION

Callum Wright 1, Jason Lam1, Lucas Cortes1, Paul Milne1, Jamie Macdonald1, Laura Jardine1, Rafiqul Hussain1, Jonathan Coxhead1, Rachel Queen1, Megan Hasoon1, Adrienne Unsworth1, Michael McCorkindale1, Erin Hurst2, Amy Publicover2, Venetia Bigley1, Matthew Collin1

1Newcastle University, Newcastle upon Tyne, United Kingdom, 2Northern Centre for Bone Marrow Transplantation, Newcastle upon Tyne, United Kingdom

Background: Acute GVHD is caused by immune infiltrates involving complex interactions with tissue cells. Understanding the transcriptional events governing these processes is key to defining tissue-specific processes and revealing new therapeutic insights. We present a large single cell RNA-seq dataset describing the immune landscape in acute GVHD of human blood and skin – comparing changes in architecture to HSCT patients with no history of GVHD.

Methods: Skin and PBMC samples were obtained at aGVHD onset (n=10), with paired samples taken from 4 of these at GVHD resolution (n=1) or progression (n=3). Comparison was made with 6 skin and PBMC samples from HSCT patients with no history of GVHD (HSCT), and 5 untransplanted healthy controls. Samples were split into dermis and epidermis and single cell suspensions were prepared following enzymatic digestion. Cells were processed using the 10x 5’ scRNAseq pipeline for gene expression and TCR profiling, with 311,954 cells passing QC. Cell chimerism was assessed through identification of transcribed donor and recipient specific SNPs (cellsnp-lite/Vireo).

Results: In PBMC myelomonocytic cells were enriched in GVHD compared to HSCT by differential abundance analysis (Milopy). These were noted on flow cytometry as a low density neutrophil population indicative of haematopoietic stress but not previously described as a marker of GVHD. Monocytes were not numerically increased but comparing differentially expressed genes (DEGs) in GVHD relative to HSCT demonstrated massive upregulation of interferon-stimulated genes (ISG) indicating a primed state.

In dermis and epidermis, GVHD samples demonstrated a rich inflammatory infiltrate consisting of plasmacytoid dendritic cells (DC), myeloid DC2/3, migratory DC and recruited macrophages. Donor derived Langerhans cells were also observed in the epidermis. GVHD was associated with increases in NK cells, CD4 and CD8 T lymphocytes relative to HSCT. Cross-tissue myeloid cell integration demonstrated almost exclusive donor origin with the exception of a defined population of dermal resident macrophages.

Trajectory analysis (scVelo) demonstrated a pathway of replacement of recipient resident macrophages by recruited donor macrophages that was accelerated in GVHD. Recipient resident macrophages were reprogrammed by GVHD to upregulate genes associated with antigen presentation. Transcription factor profiling (pyScenic) identified regulons upregulated in GVHD macrophages including MXD1 and STAT4, with loss of SOX5 and MAFB. Lymphocytes in GVHD dermis and epidermis were mostly but not exclusively of donor origin, TCR analysis identified that GVHD was associated with an oligoclonal T cell expansion of clones that were present in blood, more abundant in dermis but had unique access to the epidermis in GVHD.

Conclusions: Cutaneous GVHD was associated with the appearance of myelomonocytic cells in blood and priming of monocytes with an ISG signature. In tissue massive reprogramming of the immune populations occurred with appearance of cells not seen in steady state (NK and pDC) and upregulation of multiple populations of epidermal Langerhans cells, inflammatory DC, migratory DC and recruited macrophages. T cell clonality revealed unique access of expanded oligoclonal populations to the epidermal compartment. In myeloid cells, gene expression changes were driven by multiple transcription factor circuits previously described in viral infection and rejuvenation that may present new therapeutic targets.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P296 IMPROVING THE THERAPEUTIC EFFECT OF HUMAN UMBILICAL CORD MESENCHYMAL STEM CELLS ON ACUTE GRAFT-VERSUS-HOST DISEASE BY CRISPLD2-OVEREXPRESSION

Qing Xu1,2, Ya Zhou1, Yuxi Xu1, Rui Wang1, Xiaoqi Wang1, Shijie Yang1, Qingxiao Song 1,2, Xi Zhang1,2

1Medical Center of Hematology. Xinqiao Hospital of Army Medical University (Third Military Medical University), Chongqing, China, 2Jinfeng Laboratory, Chongqing, China

Background: Acute graft-versus-host disease (aGVHD) is mediated by alloreactive donor T cells after allogeneic hematopoietic cell transplantation (allo-HCT). The liver and gastrointestinal tract (GI) are the prominent targets of aGVHD, and the severity of damage in the liver and GI determines the outcome of aGVHD. Mesenchymal stem cells (MSCs) have become potential cell therapies for aGVHD due to their low immunogenicity and immunosuppressive properties. Nonetheless, the therapeutic effect of MSCs on aGVHD is substantially impacted by their heterogeneity. Immediate action is required to reduce the heterogeneity of MSCs while enhancing their immunosuppressive activity. CRISPLD2+ HUC-MSCs are subsets of umbilical cord sources identified by single-cell RNA-seq. Cysteine-rich secreted protein LCCL domain 2 (CRISPLD2) is localized on the extracellular matrix and plays an essential role in immune regulation by inhibiting the secretion of inflammation-related factors.

Methods: PCR and western blot were used to validate the lentivirus transfection effect of CRISPLD2. To establish the MHC-mismatched GVHD model, the BALB/c recipients were conditioned with a total body irradiation of 7.5Gy. Then, the recipients were transplanted with 5 ×10 6 bone marrow cells and 1.25 ×10 6 spleenocytes of C57BL/6 mice. 1×106 HUC-MSCs or CRISPLD2-overexpressed HUC-MSCs were infused on days 0, 9, 15, and 23 post-HCT. Recipients were monitored for clinical signs of aGVHD, including body weight loss, diarrhea, and overall survival. T cells and HUC-MSCs were cocultured in vitro to test the immunosuppressed function of HUC-MSCs. HE staining was used to examine pathological changes in the target organs (liver, small intestine, and colon). Lymphocytes from the target organs of the recipients are collected for flow cytometry analysis.

Results: The lentivirus transfection successfully enhances the CRISPLD2 gene and protein expression level. In vitro, overexpression of CRISPLD2 on HUC-MSCs has no impact on the osteogenic and adipogenic differentiation potential. However, the immunosuppressive function of HUC-MSCs was enhanced through CRISPLD2 overexpression, as evidenced by the upregulation of IDO and IL-10 gene expression and the downregulation of GM-CSF expression. In addition, CRISPLD2 overexpressed HUC-MSCs inhibited T cell proliferation much more than the control group. In vivo, infusion of CRISPLD2 overexpressed HUC-MSCs effectively alleviated aGVHD suggested by extending overall survival and reducing damage to the aGVHD target organs (liver, small intestine, and colon).

Conclusions: CRISPLD2-Overexpression enhances the HUC-MSCs immunosuppression, and CRISPLD2-Overexpression HUC-MSCs infusion significantly alleviates aGVHD in the periclinal model. These data provide the scientific rationale for CRISPLD2-overexpressed modified HUC-MSCs as a clinical treatment for aGVHD.

Clinical Trial Registry: N/A.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P297 RUXOLITINIB USE DURING DONOR STEM CELL MOBILIZATION RESULTS IN RECIPIENTS IMMUNOMODULATION POST BONE MARROW TRANSPLANT IN MURINE MODEL AND MIGHT REDUCE GRAFT VERSUS HOST DISEASE

Vamsi Kota 1, Ahmet Alptekin1, Hasanul Chowdhury1, Mahrima Parvin1, Evila Sales2, Amanda Barrett3, Babak Baban2, Ali Arbab1, Ravindra Kolhe3

1Georgia Cancer Center at Augusta University, Augusta, United States, 2Dental College of Georgia, Augusta, United States, 3Augusta University, Augusta, United States

Background: Acute graft versus host disease (aGVHD) is major complication post allogeneic bone marrow transplant (BMT). Donor-derived T-cell response to recipient antigens initiate aGVHD. Conditioning regimen-induced tissue damage enhances this activation with local proinflammatory cytokines playing a role in stimulating the donor T cell responses. Newer approaches are essential to improve outcomes in post-transplant period by reducing incidence of aGVHD but essential to preserve the graft versus leukemia effect. Janus kinase (JAK) and signal transducers and activators of transcription (STAT) pathways play an important role in T cell activation and inflammation during aGVHD. Ruxolitinib, an oral selective inhibitor Jak1/2, was shown to reduce the incidence of aGVHD while preserving the graft versus leukemia (GVL) effect and approved for the management of aGVHD. There is limited research directed at altering the T cell composition harvested from the donor that plays a major role in aGVHD. We explored in a murine model, the effects of Ruxolitinib therapy during mobilization in donor mice and immune and cytokine alteration in the donor as well as recipient mice.

Methods: CD45.1 expressing C57BL/6 mice were used as donors, primed with standard-of-care G-CSF for 3 days and/or Ruxolitinib for 7 days before bone marrow collection. CD45.2 expressing BALB/c mice were used as recipients, bone marrow eradicated with 4 Gray irradiation, and bone marrow cells transplanted the following day from G-CSF-primed, Ruxolitinib-primed, and G-CSF and Ruxolitinib-primed donor mice. Additional irradiation only and control without any treatment groups were created in the recipient group. We evaluated cytokine profiles of 170 mice using cytokine membrane array and mRNA expression with a NanoString of 36 representative mice from different groups of donor and recipient, at different time points. We also conducted flow cytometry on bone marrow and peripheral blood cells to assess the T cell immunophenotype.

Results: Mobilizing donor cells with G-CSF caused a surge in pro- and anti-inflammatory cytokines, while Ruxolitinib treatment showed significantly lower cytokine expression in donor mice plasma and bone marrow in the cytokine membrane array as expected based on the known mechanism of action. This cytokine surge is not observed in mRNA levels in Nanostring experiments in donors. There were no observed concerns in engraftment by exposure to Ruxolitinib. Interestingly though, in recipient mice plasma, we observed increased cytokine expression with G-CSF-primed transplants, but a significant decrease with G-CSF and Ruxolitinib-primed transplants. The mRNA levels showed a similar pattern, an increase in cytokine in recipients of G-CSF-primed transplants and a decrease in G-CSF and Ruxolitinib-primed transplants in Nanostring.

Cytokine mRNA levels of recipient mice and control mice (SC Ctrl), detected with Nanostring 28 days after the transplant. Recipients received bone marrow cells from donors without priming (BM-only control) and primed with Ruxolitinib, G-CSF, Ruxolitinib, and G-CSF.

Conclusions: Our findings in the murine model showed priming donors with G-CSF and Ruxolitinib provides comparable engraftment and survival to G-CSF alone in recipients and presents a different and lower cytokine profile compared to G-CSF. This indicates that the use of G-CSF and Ruxolitinib in donors has the potential to reduce GVHD through immunomodulation.

Disclosure: Research funding provided by Incyte Pharmaceuticals.

11: Graft-versus-host Disease – Preclinical and Animal Models

P298 ROS DEFICIENCY OF RECEIPT DERIVED MDSCS EXACERBATES ALLO-REACTIVE T CELLS RESPONSES DURING HYPERACUTE GVHD

Yigeng Cao1, Jiali Wang1, Zihan Zhao1, Erlie Jiang 1, Yuanfu Xu1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China

Background: Hyperacute graft-versus-host disease (GVHD) was a severe and lethal disorder occured within 2 weeks followed by allogeneic haematopoietic stem-cell transplantation (allo-HSCT). Molecular mechanisms underlying hyperacute GVHD remain elusive. Here, we investigated the role of reactive oxygen species (ROS) in this fatal disease.

Methods: ROS in Patients received allo-HSCT were examined. The underling molecular mechanisms were investigated via a genetic NADPH-oxidase deficient - the chronic granulomatous disease (CGD) mice.

Results: In a cohort of 205 patients who received allo-HSCT, hyperacute GVHD was found to be associated with decreased probability of overall survival (P=0.02) and ROS levels in the peripheral blood were reversely correlated with hyperacute GVHD symptoms development. Using the CGD mouse model, we found that ROS deficiency of receipt exacerbates hyperacute GVHD. Mechanistically, the donor T cells triggered an immense allo-reactive T cell response in recipient animals, which gained killing capacity during hyperacute GVHD. MDSCs with impaired ROS production from CGD mice exhibited defective suppressive effects on CD8+ and CD4+ T cells. Pharmacological depletion of MDSCs by an anti-Gr1 antibody reproduced the hyperacute GVHD phenotype as seen in CGD mice. Conversely, a gain-of-function approach using a ROS agonist prevented development of hyperacute GVHD. Transcriptional analysis indicated that T-cell and MDSCs dysregulation during hyperacute GVHD.

Conclusions: Our data provide novel mechanistic insights into the NADPH oxidase-mediated ROS suppressive functions in development of allo-HSCT. On the basis of these findings, we propose a new therapeutic paradigm by delivery of ROS agonists for effective prevention and treatment of hyperacute GVHD.

Disclosure: Nothing to declare.

11: Graft-versus-host Disease – Preclinical and Animal Models

P299 DIAGNOSIS OF CUTANEOUS ACUTE GRAFT-VERSUS-HOST THROUGH CIRCULATING PLASMA MIR-638, MIR-6511B-5P, MIR-3613-5P, MIR-455-3P, MIR-5787, AND MIR-548A-3P AS PROSPECTIVE NONINVASIVE BIOLOGICAL INDICATORS IN ACUTE MYELOID LEUKEMIA

Marjan Yaghmaie 1, Marzieh Izadifard1, Mohammad Ahmadvand1, Hossein Pashaiefar1, Kamran Alimoghadam1, Amir Kasaeian1, Maryam Barkhordar1, Ghazal Seghatoleslami1, Mohammad Vaezi1, Ardeshir Ghavamzadeh2

1Tehran University of Medical Sciences, Tehran, Iran, Islamic Republic of, 2Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, Iran, Islamic Republic of

Background: There are currently no laboratory tests that can accurately predict the likelihood of developing acute graft-versus-host disease (aGVHD), a patient’s response to treatment, or their chance of survival. The complicated pathophysiology of GVHD contributes in part to the lack of confirmed aGVHD biomarkers. According to recent investigations, miRNAs may play a role as one of the non-invasive diagnostics, prognostic, or predictive biological markers of aGVHD.

The main goal of this research was to replicate the findings from two previous studies to establish circulating miRNAs as biological indicators for diagnosis and anticipation to monitor patients’ conditions or predict outcomes.

Methods: In a prospective cohort setting, we studied the incidence of cutaneous aGVHD in AML patients undergoing allo-HSCT from HLA-identical sibling donors at Shariati Hospital in Tehran, Iran during 2020-2023. Patients with cutaneous aGVHD (overall grade I) were labeled as the case group, while patients without cutaneous aGVHD were selected as the control group. Accordingly, the expression levels of six significant miRNAs (miR-638, miR-6511b-5p, miR-3613-5p, miR-455-3p, miR-5787, and miR-548a-3p) were evaluated by quantitative reverse transcription–polymerase chain reaction (RT-qPCR) in three different time points: before transplantation, on day 14 and 21 after transplantation.

Results: The levels of plasma miR-455-3p, miR-5787, miR-638, and miR-3613-5p were significantly downregulated, while miR-548a-3p, and miR-6511b-5p were significantly upregulated in individuals with cutaneous aGVHD in comparison to patients without GVHD. Additionally, the possibility for great diagnostic accuracy for cutaneous aGVHD was revealed by ROC curve analysis of differentially expressed miRNAs (DEMs).

Conclusions: The study findings encourage us to hypothesize that the aforementioned miRNAs may contribute to the predominance of aGVHD, particularly low-grade cutaneous aGVHD. The disclosure of these specific miRNAs in plasma as conceivable diagnostic biomarkers may offer insightful knowledge into the pathogenesis of cutaneous aGVHD, thus early intervention and personalized therapies.

Clinical Trial Registry: Not applicable.

Disclosure: No Conflict of Interest.

1: Haematopoietic Stem Cells

P300 THE IMPORTANCE OF CYTOGENETIC BONE MARROW TESTS IN POST-TRANSPLANT CARE OF PATIENTS AFTER ALLO-HSCT DUE TO ACUTE MYELOID LEUKEMIA OR MYELODYSPLASTIC SYNDROME- A SINGLE-CENTER ANALYSIS

Ewa Karakulska-Prystupiuk 1, Agnieszka Stefaniak1, Anna Kulikowska1, Krzysztof Madry1, Agnieszka Tomaszewska1, Piotr Boguradzki1, Tomasz Stokłosa1, Grzegorz Władysław Basak1

1Medical University of Warsaw, Warsaw, Poland

Background: Monitoring the AML and MDS patients response after allo-HSCT at the cytogenetic level is standard in our center and is performed in the following scheme: 3,6,12 months after transplantation. In this study, we analyzed the detected cytogenetic changes after allo-HSCT in relation to aberrations before allo-HSCT, regardless of whether there was a clinically obvious relapse or not.

Methods: Recipients with AML and MDS, under the care of a single-center were retrospectively analyzed. Cytogenetic analysis was performed on the bone marrow cells from a 24-hour unstimulated culture. Chromosomes were stained using the classic GTG and CBG banding methods. The FISH technique was used for molecular cytogenetic studies.

Patients were divided into three cytogenetic risk groups based on ELN 2022 for AML and IPSS-R 2012 for MDS, with the awareness that the lack of some molecular tests in AML prevents the proper classification of intermediate and standard risk patients. A high-risk group, included the unfavorable risk category for AML with the poor and very poor prognostic subgroups of MDS.

An intermediate-risk group included intermediate AML and MDS, while a favorable risk group contained favorable AML and good and very good prognostic subgroups of MDS.

Results: We analyzed 162 patients (with a median age of 62 years) after allo-HSCT in the years 2011-2023 due to AML- 126(78%) or MDS- 36(22%). For twelve patients, it was the second allo-HSCT. AML-MR (myelodysplasia-related) was diagnosed in over 25% of patients. Conditioning was myeloablative in 59% of patients. The donor was matched unrelated in 74% of patients, matched sibling- in 22%, or haploidentical relative in 4% of patients.

Before allo-HSCT, 31(19%), 84(52%) and 47(29%) patients had favorable, intermediate, and unfavorable cytogenetic disease risk, respectively.

In total, in the group of 162 patients undergoing transplantation, cytogenetic abnormalities were found in 47(29%) patients after a median time 8 months after transplantation.

In this group, 22/47(47%) had the same cytogenetic changes, in 9/47(19%) had a clonal evolution and in 16/47(34%) a new clone occurred.

The results are presented in Table, independently for patients with AML and MDS (due to different classification systems) and in total.

Table. Type of cytogenetic abnormalities detected after allo-HSCT in patients with AML, MDS and in total

AML

MDS

In total

Same changes

14

8

22(47%)

Clonal evolution

6

3

9(19%)

New clone

11

5

16(34%)

A new clone after allo-HSCT occurred in 40% of patients with a favorable karyotype at diagnosis, 31% with an intermediate, and 14% with an unfavorable karyotype. The clonal evolution was detected in 10% of patients with a favorable, 7%- intermediate, and 28%- unfavorable karyotype, while the same clone in 23%, 23% and 54%, respectively.

It is worth paying attention to the frequent occurrence of additional cytogenetic abnormalities involving chromosomes 7,8,13 and 20 in patients with a new clone.

Conclusions: Cytogenetic bone marrow tests after allo-HSCT allow the detection of cancer cell clones, which enables preemptive treatment.

It should be taken into account that clonal evolution occurs most often in the group of unfavorable karyotype at diagnosis.

The detection of new clones may indicate, among others, the evolution of CHIP clones, which requires further monitoring unless there are signs of clinically overt relapse.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P301 INFECTION BY HUMAN HERPES VIRUS 6 IN ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN THE POST-TRANSPLANT CYCLOPHOSPHAMIDE ERA

Paola Charry1, Virginia Rodríguez1, Ares Guardia1, Teresa Solano1, Jordi Arcarons1, Joan Cid1, Miquel Lozano1, Laura Rosiñol1, Carmen Martínez1, María Queralt Salas1, Francesc Fernandez-Avilés1, Mª Ángeles Marcos1, Montserrat Rovira1, María Suárez-Lledó 1

1Hospital Clínic of Barcelona, Barcelona, Spain

Background: HHV-6 is associated with a high frequency of reactivations after allo-HCT, but there is great difficulty in knowing its pathogenic capacity. The HHV-6B subtype is the main pathogen that causes sudden exanthema in childhood and remains latent in T lymphocytes and monocytes. In allo-HCT patients the most frequent manifestation is encephalitis (1%) and can also cause delay in hematopoietic engraftment. Involvement of other organs is very rare (hepatitis) or not recognized (gastrointestinal), due, in part, to the difficulty in making an accurate diagnosis, as well as in ruling out the chromosomal integration of HHV-6B.

Methods: A retrospective analysis was conducted on cases of HHV-6 infection in patients who underwent allo-HCT at the Hospital Clínic of Barcelona between January/2015 and November/2023 (n=496). The most significant change in transplant practice during this period was the adoption of PTCY-based prophylaxis for GVHD, initially for allo-HCT from haploidentical-donors and subsequently expanded to allo-HCT performed from all donor types.

During the early post-transplant period, HHV-6 PCR-test was conducted on plasma samples if the patient exhibited symptoms such as fever, rash, and/or cytopenias. In cases where neurological symptoms were present, the test was conducted on both plasma and cerebrospinal fluid. If symptoms in other organs such as hepatitis or gastrointestinal issues were present, a HHV-6 PCR-test was conducted on tissue samples after ruling out other potential causes (CMV/GVHD).

Results: HHV-6 infection was detected in 51 patients (global incidence of HHV-6: 10%). The highest incidence was observed during the period of 2020-2023 (24% vs. 1.3% in 2015-2019) and in haploidentical-HCT (22%). The median time for HHV-6 infection was 36 days after transplant (IQR 22-81days). At the time of detection, most patients had lymphopenia (median 200 [IQR 100-600]).

Fourteen patients (27.5%) had detectable viral load (VL) in blood (average initial-VL of 27.289 copies/mL; 1.709–105.913 range). Organic involvement was observed in 88.2% of the patients, with the gastrointestinal system being the most commonly affected (91.2%). Tissue VL exhibited high variability (average 1.893.223 copies/mL; 1.709–71.527.336 range). Three patients had multiorgan involvement (one patient had CNS and liver; two had liver and gastrointestinal involvement), VHH-6 integration was ruled out in these patients. VHH-6B was isolated in 96% of cases.

Twenty patients (39.2%) resolved infection spontaneously without treatment. The remaining 31 patients (60.8%) were treated with foscarnet (64.5%) and val/ganciclovir (35.5%). All patients responded well to antiviral treatment as evidenced by the resolution of symptoms and/or a decrease in VL with a first line of treatment (5 patients needed a second line of antiviral). The median duration of treatment until the VL became negative was 8 days for those blood reactivation (IQR 8-14days) and 26.5 days for those with organic involvement (IQR 12-31.25days).

Conclusions: There is an increased incidence of HHV-6 infection with organic involvement other than encephalitis in allo-HCT in recent years, that could be related to the use of PTCY-prophylaxis and the immunosuppression that it entails. More studies are necessary to improve the usefulness of diagnostic techniques in the management of this infection.

Clinical Trial Registry: Does not apply.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P302 SECOND HAPLOIDENTICAL HAEMATOPOIETIC STEM CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE AS RESCUE STRATEGY FOR HIGH RISK CHILDREN WITH LEUKEMIA

Giorgio Ottaviano1, Maria Barazzoni2, Federica Acone2, Francesca Vendemini1, Giulia Prunotto1, Alex Moretti2, Pietro Casartelli1, Sara Napolitano1, Marta Verna1, Sonia Bonanomi1, Adriana Balduzzi 1,2

1Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 2Università degli Studi Milano-Bicocca, Monza, Italy

Background: Survival rate of children with leukemia who relapse after HSCT is extremely low. Despite the success of chimeric antigen receptor T cells has changed this paradigm, some patients who achieve remission still might benefit from a second transplant. An haploidentical (haplo) donor is readily available for the majority of patients, allows flexible and timely schedules, provides a degree of HLA disparity which might eventually enhance GvL. Here we aimed to evaluate outcome and toxicities of a second HSCT from an haplo donor using post-transplant cyclophosphamide (2nd-haplo PT-Cy), to mitigate the risk of rejection and alloreactivity.

Methods: We retrospectively reviewed clinical charts of children who received a 2nd-haplo PT-Cy transplant in a single referral paediatric HSCT centre (Fondazione IRCCS San Gerardo, Monza). Data on patients and transplants features, engraftment, acute and chronic GvHD, CMV/EBV/ADV viral reactivations, immune reconstitution and outcome were retrieved.

Patients

16 (100%)

Background disease

ALL

11 (69%)

AML

4 (25%)

JMML

1 (6%)

Disease status

CR1

2 (12%)

CR2

4 (25%)

CR3/CR4

8 (50%)

NA

2 (12%)

Conditioning

TBI-based

4 (25%)

Bu-based

2 (12%)

Treo-based

10 (63%)

GvHD prophylaxis

CSA + MMF + ATG

1 (7%)

CSA + MMF

15 (93%)

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (37)

Results: From 2014 to 2023, 16 patients (median age 9.5 years, range 2-17) received a 2nd-haplo PT-Cy transplant, using BM as stem cell source, due to relapse (14) or primary rejection (2) after the first HSCT. Patients and transplant characteristics are shown in table 1. After 2nd-haplo PT-Cy, only one patient showed a primary engraftment failure and underwent a third haplo PT-Cy due to a subsequent relapse. Neutrophils engraftment occurred between day +13 and day +20 (mean 16,7 days), while platelet engraftment was slightly delayed (range 17-133, median 41 days). An elevated incidence of severe aGvHD grade III-IV (33%) and chronic GvHD (43%) (Fig. 1A and 1B) was observed, whereas 2/16 patients had developed grade III-IV aGvHD after the first transplant, and only one experienced cGvHD. Ten out of 16 patients (62%) showed at least one viral reactivation that required treatment, with CMV being the most frequently reported (50%; ADV 23%, EBV, 21%) (Figure 1C). CD4 + T cell immune reconstitution (median 150/mmc) occurred within the first 6 months after transplant (Figure 1D). Transplant related mortality (TRM) remained extremely low (6%), and the major cause of treatment failure was disease relapse, with EFS being 57% in this highest risk cohort (Figure 1E and 1F).

Conclusions: Children with leukemia who relapse after HSCT face a dismal outcome. A 2nd-haplo PT-Cy transplant represented an efficacious rescue strategy, with minimal risk of TRM. The use of an haplo donor with PT-Cy might have favoured a high rate of GvHD and enhanced the anti-leukemic effect. We speculate that alloreactivity might have contributed to increase EFS, by eliciting graft-versus-leukemia, since historical outcome of second HSCT in children with leukemia showed significantly lower rate of survival (30%). However, larger set of data and comparison with a control cohort of non-haplo second transplant are needed to establish the best transplant strategy in such high risk patients.

Disclosure: nothing to declare.

1: Haematopoietic Stem Cells

P303 DISTINCT CLINICO-BIOLOGICAL FEATURES IN NPM1-MUTATED ACUTE MYELOID LEUKEMIA PATIENTS FOLLOWING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION

Xiaolin Yuan 1,2,3,4, Yibo W1,2,3,4, Xiaoyu Lai1,2,3,4, Lizhen Liu1,2,3,4, Jimin Shi1,2,3,4, Weiyan Zheng1,2,3,4, Jian Yu1,2,3,4, Yanmin Zhao1,2,3,4, Jie Sun1,2,3,4, Zhen Cai1,2,3,4, He Haung1,2,3,4, Yi Luo1,2,3,4

1Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Hangzhou, China, 3Institute of Hematology, Zhejiang University, Hangzhou, China, 4Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China

Background: NPM1 mutated AML has been reclassified as a distinct leukemia entity with unique biological and clinic-pathological features. The prognosis of NPM1-mutated AML may vary according to accompanying mutations or other clinical features. This study aimed to evaluate the genomic features, prognosis, and transplantation outcomes in the AML patients with NPM1 mutation who underwent allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Methods: A total of 115 consecutive NPM1-mutated AML patients who underwent allo-HSCT were investigated in this study. The primary endpoints were cumulative incidence of relapse (CIR), leukemia-free survival (LFS) and overall survival (OS).

Results: A total of 115 NPM1-mutated AML patients were enrolled in this study and the median age was 46 (IQR: 36-50) years. The most frequent co-mutations were FLT3-ITD (58.3%) and DNMT3A (40.0%), followed by IDH2 (18.3%), NRAS (13.9%) and IDH1 (11.3%). The overall 3-year CIR and LFS was 17.3% and 77.2%. The 3-year CIR of patients with FLT3-ITD was significantly higher than that without FLT3-ITD (24.3% vs. 7.0%; P=0.023). The 3-year CIR of patients with DTA (DNMT3A, TET2, and ASXL1) mutations was significantly higher than that without DTA mutations (24.7% vs. 11.2%; P=0.030). Patients who received matched sibling donor (MSD) allo-HSCT had the highest CIR compared with unrelated donor (URD) allo-HSCT and haploidentical (HRD) allo-HSCT (46.2%, 10.0% and 11.2%, P=0.002). The 3-year CIR in patient with pre-HSCT measurable residual disease negative (MRD-), MRD+ and non-remission was 10.9%, 30.0% and 72.2%(P<0.001). After multivariable adjustment, DTA mutations, pre-MRD+ or non-remission, MSD allo-HSCT remained statistically significantly associated with higher CIR as well as lower LFS and OS. In univariate and multivariate analyses, the development stage of AML did not influence significantly the outcomes.

Conclusions: The DTA (DNMT3A, TET2, and ASXL1) mutations identified a subgroup of patients with adverse prognosis in NPM1-mutated AML patients who received allo-HSCT. Our results suggest that HRD/URD transplants can achieve a stronger graft-versus-leukemia effect than MSD for NPM1-mutated patients.

Disclosure: There are no conflicts of interest to report.

1: Haematopoietic Stem Cells

P304 DIFFERENT SOURCES OF HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH THALASSEMIA MAJOR: RELATIONSHIP BETWEEN IMMUNE RECONSTITUTION, GRAFT-VERSUS-HOST DISEASE, AND VIRAL INFECTIONS

Jianyun Liao 1, Chaoke Bu1, Lan He1, Huaying Liu1, Zhiyong Peng1, Jujian He1, Weiwei Zhang1, Yuqian Xia1, Yuelin He1, Chunfu Li1

1Nanfang-Chunfu Children’s Institute of Hematology & Oncology, TaiXin Hospital, Dongguan, China

Background: To compare the relationship between immune reconstitution and graft-versus-host disease (GVHD) in three kinds of hematopoietic stem cell transplantation(HSCT) using different sources for thalassemia major patients: matched unrelated donor (MUD) HSCT, haploidentical (Haplo) HSCT with post-transplant cyclophosphamide (PTCy)-based prophylactic therapy, and TCRαβ-T cell-depleted HSCT (TDH).

Methods: A total of 198 patients with thalassemia major who underwent allogeneic HSCT in our center between January 2018 and December 2022 were included in this study. Among them, 56 cases were in the MUD group, 45 cases were in the Haplo group, and 97 cases were in the TDH group. Four-color flow cytometry was used to detect the proportions and absolute counts of lymphocyte subsets at the 12th month after transplantation for the three different donor types. The immune reconstitution was analyzed and compared with graft-versus-host disease, human cytomegalovirus, and varicella-zoster virus infections.

Results: 1. At 12 months after transplantation, there were significant differences (P<0.05) in the proportions of helper T cells (CD3 + CD4 + ), cytotoxic T cells (CD3 + CD8 + ), B cells (CD3-CD19 + ), NK cells (CD3-CD56 + ), and regulatory T cells among the three groups. The MUD group showed faster immune reconstitution, while no significant differences were observed between the other two groups.

2. There were significant differences (P<0.05) in the incidence of chronic GVHD among the three groups. The MUD group had a lower incidence of chronic GVHD.

3. When comparing the TDH group with the Haplo group, there was a significant difference (P<0.05) in the incidence of chronic GVHD, with higher incidence in the Haplo group.

4. There were significant differences (P<0.05) in the occurrence of viral infections when comparing all three groups. The MUD group had a lower incidence of viral infections, while no significant differences were observed between the other two groups.

Conclusions: The MUD group showed faster immune reconstitution and lower rates of GVHD and viral infections compared to the other two groups. In comparison between Haplo group and TDH group, the former group had a higher incidence of chronic GVHD.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P305 COMBINATION OF POST-TRANSPLANT CYCLOPHOSPHAMIDE WITH ANTITHYMOCYTE GLOBULIN AFTER HAPLOIDENTICAL ALLOGENEIC HEMATOPOIETIC STEM-CELL TRANSPLANTATION: A SINGLE CENTER ANALYSIS

Chiara Bernardi 1,2, Amandine Pradier1,2, Anne-Claire Mamez1, Federica Giannotti1, Sarah Morin1, Sisi Wang1,2, Astrid Melotti1,2, Yves Chalandon1, Federico Simonetta1,2, Starvoula Masouridi-Levrat1

1Division of Hematology, Geneva University Hospitals and Faculty of Medicine, University of Geneva, Geneva, Switzerland, 2Translational Research Center for Oncohematology, Faculty of Medicine, University of Geneva, Geneva, Switzerland

Background: In haploidentical allogenic hematopoietic stem-cell transplantation (HSCT), post-transplant cyclophosphamide (PTCy) is widely used for in vivo T cell depletion with great efficacy in preventing GvHD. Some groups have tried to combine antithymocyte globulin (ATG) with PTCy with the aim of reducing the risk of GvHD without compromising outcomes. However, little is known about the impact of PTCy/ATG combination on immune reconstitution and risk of infection related mortality (IRM) after haploidentical-HSCT.

Methods: We conducted a retrospective analysis including 81 adult patients who underwent haploidentical-HSCT with PBSC with or without ATG at our institution between 2013 and 2023. We analyzed transplant outcomes (OS, PFS and GRFS) as well as the immune-reconstitution of major lymphocyte subsets at different timepoints post-HSCT.

Results: 50 patients received PTCy alone and 31 received PTCy/ATG (Neovii; 10mg/kg for 1,2 or 3 days in 18, 10 and 3 patients, respectively). Median follow-up among survivors was 26 (range 1-98) and 29 (16-64) months, respectively. No major differences were observed between the two groups with the only exception of date of transplantation, the majority of patients in the PTCy/ATG group being transplanted after May 2019 (date at which the letermovir prophylaxis was introduced at our institution). No significant differences were observed in OS and PFS at 2 years (63%, 95% CI 50 -80% vs 81%, 95% CI 68-96% and 57%, 95% CI 44-74% vs 71%, 95% CI 57-89%, respectively). Patients receiving PTCy/ATG displayed a significantly improved GRFS at 2 years (48%, 95% CI 33-69%) compared to patients receiving PTCy alone (28%, 95% CI 17-46%, p-value= 0.029). Such difference was confirmed in a multivariable analysis taking into account the year of transplant (HR: 0.54, p=0.04). The 2 years cumulative incidence of relapse was not significant between the two groups while patients receiving PTCy/ATG displayed a significantly reduced NRM (6%, 95% CI 1-19%) compared to patients receiving PTCy alone (27%, 95% CI 15-41%). We found no differences in the cumulative incidence of aGVHD II-IV, III-IV or moderate/severe cGvHD between the two groups. We observed a slower immune-reconstitution of CD4 T cells, CD8 T cells and B cells when using the combination of PTCy/ATG with a significant reduction of the three subsets at day 30 (CD4: median of 8 (0-53) vs 35 (6-152) cells/µl, p=0.004; CD8: 16 (1-564) vs 47.5 (5-189) cells/µl, p=0.03; CD19: 0 (0-3) vs 0 (0-1), p=0.04). At 6 months since HSCT, CD4 T cell numbers were still significantly lower in the PTCy/ATG group (155 (22-633) cells/µl) compared to the PTCy alone (186 (22-508) cells/ul, p=0.03). Despite such differences in lymphocyte counts, the IRM was not significantly different between the two groups.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (38)

Conclusions: In our series, the prophylaxis based on combining PTCy and ATG appeared safe and was associated with an improved GRFS after haploidentical- HSCT using PBSC. Our results suggest that, despite a delayed immune reconstitution of major adaptive immune cell subsets, the addition of ATG was not associated to an increased risk of relapse or IRM after haploidentical-HSCT with PTCy.

Disclosure: Yves Chalandon has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution.

Federico Simonetta has received institutional consulting fees from BMS/Celgene, Incyte, Kite/Gilead; speaker fees from Kite/Gilead, Incyte; travel support from Kite/Gilead, Novartis, AstraZeneca, Neovii, Janssen; research funding from Kite/Gilead, Novartis, BMS/Celgene.

S. Masouridi-Levrat has received travel support from Gilead, BeiGene, Jazz and Sanofi all via the institution.

1: Haematopoietic Stem Cells

P306 TRIPLE STEM CELL INFUSION ALLEVIATE GRAFT VERSUS HOST DISEASE AND IMPROVE OUTCOME IN UNMANIPULATED HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION

Fang Hua1,2, Shan Zhang1, Xiao-Mei Zhang1, Yan Deng1, Ying Han1, Si-Han Lai1, Ying He1, Lei Ma1, Xu-Pai Zhang1, Dan Chen1, Yi Su1, Ling Zhang1, Hui Yang1, Rong Huang1, Guang-Cui He 1, Hao Yao1, Hai Yi1

1The General Hospital of Western Theater Command, PLA, Chengdu, China, 2Zigong First People’s Hospital, Zigong, China

Background: Haploidentical hematopoietic stem cell transplantation (haplo-HSCT) provides cure opportunity for patients requiring prompt allogeneic HSCT but failing to identify well-matched donor, but its outcomes are potentially impaired by increased transplant-related mortality (TRM).

Methods: We performed haplo-HSCT by using granulocyte colony-stimulating factor (G-CSF)-primed peripheral blood stem cells (PBSCs), umbilical cord-cultured mesenchymal stem cells (UC-MSCs) and third-party unrelated umbilical cord blood (UCB) stem cells. Modified “Beijing protocol” were performed in this study. All of the patients were transplanted by Busufan or TBI-based regimen. Anti-thymocyte globulin were used to T cell depletion in vivo. Cyclosporine, mycophenolate mofetil, and short course methotrexate were used to prevent graft-versus-host disease (GVHD).

Results: One hundred and sixty-five consecutive patients with hematological disorders undergoing haplo-HSCT from May 2021 to Nov 2023 were included inthis study. The median time of neutrophil engraftment were 12 days (range: 9–25 days), and the median time of platelet engraftment were 13 days(range: 6–50 days). Full haploidentical donor chimerism were obtained within 30 days. No evidence of UCB chimerism was found. Twenty-five patients developed acute GVHD. The incidence of grade II-IV and grade III-IV acute GVHD was 12.73% and 6.67%, respectively. Twenty-eight patients developed chronic GVHD, 10 were limited (6.06%) and 18 were extensive (10.91%). The 2 years overall survival (OS) rate is 72.96%. The median overall survival rate was not reached.

Conclusions: Haplo-HSCT performed by PBSCs, UC-MSCs and UCB “triple-infusion” achieved excellent outcomes, and need to explored in a larger cohort.

Disclosure: Fang Hua and Shan Zhang are contributed equally. Hao Yao, Guang-Cui He and Hai Yi are co-correspondence author.

1: Haematopoietic Stem Cells

P307 HIGH-DOSE CHEMOTHERAPY WITH AUTOLOGOUS BONE MARROW TRANSPLANTATION IN MULTI-REFRACTORY TESTICULAR GERM CELL CANCER: A RETROSPECTIVE SINGLE INSTITUTIONAL ANALYSIS

Andrea Tamayo1, Adolfo Sáez1, Reyes Mas1, Lucia Medina1, Guillermo Ramos1, Esther Parra1, Alberto Blanco1, Clara Cuellar1, Paula Lázaro1, Ana Jimenez1, Rafael Alonso1, Tycho Baumann1, Jose Sanchez Pina 1, Pilar Martinez1, Enrique Gonzalez1, Joaquin Martinez1, Maria Calbacho1

1Hospital 12 de Octubre, Madrid, Spain

Background: Testicular germ cell cancer (TGCC) generally affects young males, with outstanding results with chemotherapy, however some face therapeutic challenges when refractory to standard systemic treatments. This study aims to retrospectively assess the efficacy and safety of autologous bone marrow transplantation (ABMT) in patients with TGCC refractory to standard systemic therapies.

Methods: We conducted a retrospective analysis on a single institutional, cohort of male patients diagnosed with TGCC, refractory to standard systemic therapies regimens between January 2017 and November 2023. We included patients who underwent ABMT depending on a multidisciplinary committee decision. Survival analysis was performed using Kaplan-Meier curves to assess overall progression-free survival and overall survival outcome.

Results: We included 23 patients with a median age of 37 (Interquartile range [IQR] 17-57). The median follow-up was 11 months (IQR 3-32). Three patients had one pending ABMT during data collection, having undergone two out of the three scheduled. All patients (100%) presented febrile neutropenia which responded (100%) to standard treatment of G-CSF at day 5 post ABMT and broad-spectrum antibiotics. Moreover, five patients (21,7%) developed a graft syndrome and were treated with intravenous corticosteroids, as well as only two patient (7%) required ICU care, one due to hypotension and epistaxis, and one due to gastrointestinal complications and ultimate surgical requirement. The median time to an absolute neutrophil count of 500x1000m/l was 12 days (range: 6-33), and platelet count was of 15 days (range: 4-9). One patient (4,3%) was unable to recover from the induced thrombocytopenia and was diagnosed with refractory platelet thrombocytopenia. The crude mortality and progression rates were 21.7% and 52.2%, respectively. The main cause of mortality was disease progression and its complications, while no deaths were associated with treatment toxicity. Also, the median progression-free survival (PFS) was of 11 months (2-20). The median overall survival was not reached.

Conclusions: Our retrospective analysis conducted at a single institution highlights the potential efficacy of ABMT as a salvage therapy for patients with refractory TGCC resistant to standard systemic treatments. The observed positive outcomes in overall progression-free survival and undefined overall survival bring optimism to individuals facing limited treatment options. These promising results underscore the necessity for larger cohorts to validate and refine the role of ABMT in managing multi-refractory TGCC. The findings of this study offer hope to patients undergoing traditionally limited therapeutic alternatives.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P308 KIR-MISMATCH IMPACTS THE OUTCOMES AFTER HLA-HAPLOIDENTICAL STEM-CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE

Jairo Eduardo Niño-Ramirez1, Francisco Javier Gil-Etayo1, Marta Fonseca Santos1, Isabel Jiménez-Hernaz1, Ariadna Vicente-Parra1, Pilar Terradillos-Sánchez1, Ana Balanzategui1, Francisco Boix1, Miguel Alcoceba1, Almudena Navarro-Bailón1, Estefanía Pérez-López1, Mónica Cabrero1, Fermín Sanchez-Guijo1, Ramón García-Sanz1, Lucía López-Corral 1, Amalia Tejeda-Velarde1

1Hospital Universitario de Salamanca-IBSAL, Centro de Investigación del Cáncer-IBMCC (USAL-CSIC) and Centro de Investigación Biomédica en Red Cáncer (CIBERONC), Salamanca, Spain

Background: Natural killer cells (NKs) reconstitution process and KIR mediated-alloreactivity after haploidentical allogeneic hematopoietic stem-cell transplantation (haplo-HSCT) become relevant due to its contribution to the graft-versus-leukemia effect, although this benefit has been demonstrated predominantly with T-cell depleted transplant, while under the T-cell repleted protocol that includes administration of cyclophosphamide pos-transplant (PTCy) the benefit is still being debated. Here, we conducted a retrospectively single-institution study to examine the clinical benefit of predicted KIR-alloreactivity on this transplant modality and determine whether donor selection based on KIR-HLA interactions could be useful to improve transplantation outcomes.

Methods: A total of 138 patients with hematological malignant disorders receiving an haplo-HSCT with PTCy between 2013 and 2020 were enrolled. The donors were chosen using the classic criteria: absence of anti-HLA antibodies, CMV-status, younger, male gender and ABO compatibility. All patients were full-typed by NGS method (NGSgo® MX-11-3, GenDx) on MiniSeq, while donors typing was developed by SSO-PCR using Lifecodes® HLA-SSO Kit and by high-resolution when was necessary to unravel an allelic discrepancy. Pairs were also genotyped for KIR genes using commercially available Lifecodes® KIR-SSO kit.

Results: 71 (51.4%) individuals were diagnosed acute myeloid leukemia or myelodysplastic syndrome. At the time of infusion, 108 patients (78.3%) achieved partial or first complete remission (CR). HCT-CI was ≥3 in 31.2% and the Disease-Risk Index (DRI) was high or very high in 18.1% of cases. 45 patients (32.6%) previously received a stem-cell transplant and at least half of the cases received >2 lines of treatment before of haplo-HSCT. Patients were conditioned predominantly by reduced intensity (71%). All participants received an infusion of unmanipulated peripheral blood stem cells as the graft source. The median of graft composition was 6.45x106 CD34+ cells infused/µL.

Our findings revealed that higher HCT-CI was associated with poor 3-year survival post haplo-HSCT (HR=1.26, 95%CI: 1.08-1.48, p=0.003), while KIR3DL1-Bw4 mismatch (mm) improved overall survival (HR=0.40, 95%CI: 0.16-0.96, p=0.041). Also, we observed that multiple inhibitor KIR-mm (iKIR-mm) exhibited a significant increase in the rate of grade III-IV acute-GvHD compared to those with single iKIR-mm (HR=4.08, 95%CI: 1.46-11.38, p=0.007), accompanied by a risk associated with the graft cells load (HR=1.62, 95%CI: 1.11-2.35, p=0.011). Regarding to chronic-GvHD, KIR3DL1-Bw4 mm was again consolidated as parameter that confer protection without discriminating the event by its severity (HR=0.32, 95%CI: 0.11-0.92, p=0.036). In addition, recipient age between 60 and 65 years displayed the same protective behavior (HR=0.34, 95%CI: 0.12-0.91, p=0.033).

Finally, we detected exclusive protective effect on disease relapse after haplo-HSCT in recipients carrying various KIR-mm compared with KIR matched pairs (HR=0.39, 95%CI: 0.16-0.97, p=0.044). On the other hand, the following parameters showed a significant correlation on this endpoint: CR (HR=0.37, 95%CI: 0.15-0.92, p=0.033), high/very-high DRI (HR=2.57, 95%CI: 1.11-5.98, p=0.027), and ABO-incompatibility (HR=2.55, 95%CI: 1.11-5.85, p=0.02).

Conclusions: We can conclude that disparities in KIR genes and HLA ligands were strongly associated with improved overall survival and had an impact on post haplo-HSCT complications. Likewise, incorporate KIR-ligand information in the donor selection criteria when multiple haploidentical candidates are available could provide benefit the outcomes at least in this transplant type.

Disclosure: The authors declare no conflicts of interest.

1: Haematopoietic Stem Cells

P309 NOVEL AUTOMATIC APPLICATION FOR ENRICHMENT OF CD34 + CELLS FROM APHERESIS PRODUCTS USING THE CLINIMACS PRODIGY

Julia Dzionek 1, Stephanie Soltenborn1, Valeriya Olevska1, Svenja Oberbörsch1, Christin Bosbach1, Felix Hebbeker1, Burgund Kauling1, Juliane Raasch1, Carina Wenzel1, Heike Lahnor2, Katharina Krämer2, René Meißner2, Eleni Papanikolaou1, Andreas Bosio1

1Miltenyi Biotec, Bergisch Gladbach, Germany, 2Miltenyi Biomedicine, Bergisch Gladbach, Germany

Background: Hematopoietic stem cell transplantation (HSCT) is a curative treatment for many hematologic malignant and non-malignant diseases both in allogeneic transplantation and in the context of gene therapy. To this end, the number of the transplantable CD34+ cells as well as the purity of the graft are critical parameters for these therapeutic approaches. Lately, the stem cell boost has also emerged as a potent treatment modality not only in the classic HSCT field but in the context of CAR-T cell therapy as well, as a measure to manage cytopenias1-4.

Methods: Based on MACS Technology the CliniMACS Prodigy® LP-34-(320) System enables automatic production of an injectable CD34 enriched cell population which is simultaneously passively depleted by allogeneic reactive T cells. Central components of the new application, which utilizes tubing set TS 320, include a newly developed platelet removal step, a WBC-adjusted separation and the possibility of product release at a requested timepoint because the process allows storage of the labeled cells for up to 16h. The process is sufficient for the enrichment of 1.2x109 CD34+ cells from up to 120x109 total WBC. A new fluorescent flow analysis protocol suitable to analyze very low percentage of remaining T cells in the final product and a low percentage of CD34+ cells in the starting material was additionally developed.

Results: Inhouse evaluation runs (n=22) using mobilized leukapheresis products resulted in a mean depletion of 4.5 log (range 3.8 – 4.9) for T cells, a mean platelet depletion of 2.8 (range 2.2 – 3.7), a mean B cell depletion of 3.2 (range 2.1 – 3.5), and a mean purity of 91.2% (range 81.1 – 96.1) in the CD34 enriched product. Viability of CD34+ cells in the target product was above 99%, mean yield was 65% and mean WBC recovery 82%. The process duration is approximately within 3.2 – 5.5 hours, depending on the cell numbers, excluding the optional in-process storage.

Conclusions: The novel automated CliniMACS Prodigy LP-34-(320) enrichment process is a regulatory compliant, fully automated process in a closed fluidic system. The system delivers ready to use stem cell grafts by giving a flexible process endtime and minimal hands on time to enable most convenient staff resource planning. The system is capable to enrich CD34+ and to deplete T cells efficiently from apheresis products. In the CD34+ enriched fraction the mean CD34 purity was 91%, the mean CD34 yield was 65% and the CD34 viability was above 99%. The comparability to the existing CliniMACS plus system was shown in comparison runs. The submission to an European notified body for CE certification is an important next step.

Disclosure: All authors are employees of Miltenyi Biotec and / or Miltenyi Biomedicine.

1: Haematopoietic Stem Cells

P310 OUTCOMES OF ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH WISKOTT-ALDRICH SYNDROME

Samin Sharafian1,2, Su Han Lum1,3, Christo Tsilifis1,3, Terry Flood1, Eleri Williams1, Stephen Owens1, Sophie Hambleton1,3, Andrew Gennery1,3, Mary Slatter 1,3, Zohreh Nademi1,3

1Great North Children’s Hospital, Newcastle, United Kingdom, 2Mofid Children Hospital, Shahid Beheshti University of Medical Science, Tehran, Iran, Islamic Republic of, 3Translational and Clinical Research Institute, Newcastle University, Newcastle Upon Tyne, United Kingdom

Background: Haematopoietic stem cell transplantation (HSCT) is a curative treatment for Wiskott-Aldrich syndrome (WAS).

Methods: We examined the outcome of 27 children with WAS who received first HSCT at the Great North Children’s Hospital between January 1994 and June 2023. One patient who was referred for second transplant was excluded. Outcomes of interest were overall survival (OS) and event-free survival (EFS; event was defined as death, graft failure or second procedures), graft-versus-host disease (GvHD) and long-term disease outcomes. Busulfan-based conditioning was predominantly used before switching to treosulfan-based conditioning in 2007.

Results: Median age at diagnosis was 11.5 months (birth to 9.32 years) and median age at HSCT was 3.5 years (0.4-14.9). The interval between diagnosis and transplant was 9.6 months (2.4 months to 13.9 years). Conditioning was busulfan-cyclophosphamide (n=11), fludarabine-treosulfan (n=9), fludarabine-treosulfan-thiotepa (n=7), fludarabine-melphalan (n=1), and busulfan-fludarabine-thiotepa (n=1). Serotherapy was alemtuzumab (n=14), ATG (n=9) or none (n=4). GvHD prophylaxis was ciclosporin alone (n=4) or in combination with methotrexate (n=9) or MMF (n=13); none in three. Donors were matched related (MRD, n=6), matched unrelated (MUD, n=14), mismatched unrelated (MMUD, n =4) and mismatched related donor (MMRD, n=5). Stem cell source was bone marrow (BM, n=14), unmanipulated peripheral blood (PBSC, n=9), TCRab/CD19 depleted PBSC (n=4), CD34-selected marrow (n=1) and double cord (n=1). Median CD34+ cell dose was 7.1 x 106/kg (1- 50.9 x 106/kg). Median day to neutrophil and platelet engraftment was 16 days (10-41) and 18 days (9-51) respectively. Two developed veno-occlusive after receiving busulfan and none had transplant associated microangiopathy. Two patients developed grade 2 cutaneous GvHD, none had grade III-IV acute GvHD or chronic GvHD. Three (11%) developed CMV viraemia, 3 (11%) adenoviraemia, 3 (11%) EBV viraemia and 7 (26%) HHV6 viraemia.

Median duration of follow-up was 11 years (0.5-24). The 5-years OS and EFS was 89% (95%CI 69-96%) and 76% (55-89%) (Figure1). Details of 3 deceased patients were summarized in table 1. Three patients (Busulfan-Cyclophosphamide (2); Fludarabine-Treosulfan (1)) received second procedures: 1 CD34+ stem cell boost, 1 conditioned second transplant, 1 had CD34+ stem cell boost and then a conditioned second transplant.

Three patients developed post-transplant autoimmunity: 2 had immune thrombocytopenia (100% donor chimerism) which resolved with steroids and high dose immunoglobulin, and one patient (myeloid chimerism 2%; 35% T-cell chimerism) developed hypothyroidism and IgA nephropathy requiring a renal transplant. Median myeloid chimerism was 100% (2-100%) and median T cell chimerism was 100% (35-100%). All surviving patients with follow-up > 2 years are free from immunoglobulin replacement except one patient with myeloid chimerism of 2%. All had normal platelet counts except two patients with myeloid chimerism <= 5%.

Table 1: Details of deceased patients

Year

Age @ HSCT (year)

Pre-transplant issues

Transplant details

Cause of death

2000

7.4

CMV, EBV, Salmonella enteritis, recurrent pneumonia, vasculitis

Flu-Melphalan-ATGMUD, marrow

CMV pneumonitis and hepatitis

2006

1.1

HHV6, colitis

BuCy-AlemtuzumabMUD, marrow

Pulmonary hemorrhage and VOD

2017

12.7

CMV, EBV, Hepatitis B and C, colitis, osteomyelitis, hypothyroidism, AIHA

Fludarabine-Treosulfan-Thiotepa-ATG-RituximabHaplo, TCRab/CD19-depleted PBSC

Encephalitis

Conclusions: Our Cohort’s survival for WAS was good with a low rate of mild GvHD. Fludarabine-treosulfan-thiotepa has been adopted as our institutional standard conditioning for WAS to achieve robust myeloid chimerism.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (39)

Disclosure: There is no conflict of interest.

1: Haematopoietic Stem Cells

P311 STEM CELL TRANSPLANT ACTIVITY IN KYIV NATIONAL CANCER INSTITUTE IN THE CONDITIONS OF FULL-SCALE RUSSIAN AGGRESSION IN UKRAINE

Nazar Shokun 1, Alevtyna Burtna1, Maryna Bushuieva1, Moiseienko Kateryna1, Irina Kryachok1, Yana Stepanishyna1

1National Cancer Institute, Kyiv, Ukraine

Background: The whole-scale Russian invasion strongly impaired the availability of any kind of therapy for Ukrainian hematological patients including High dose chemotherapy (HCT) and stem cell transplantation (SCT) during the first months. However, many Ukrainian hematology centers including National Cancer Institute (NCI) continue to provide specialized care to patients even increasing workload despite the existing risks.

Methods: This study aimed analyzing dynamic of transplant activity in Kyiv NCI with a specific focus on the years 2022-2023 and some results of auto SCT. This report assesses the dynamics of transplant numbers in adults patients, 100-day transplant mortality rate (TMR), as well as the 1-year overall survival (OS) and progression-free survival (PFS) outcomes in adult patients who underwent HDCT+autoSCT in 2022-2023.

Results: Since December 2016 to November 2023 191 patients underwent autologous SCT in Ukrainian NCI. The median age at the time of SCT was 50.1 years (ranging 20 to 69) with a nearly equal distribution between males (51,7%) and females (48,3%). Primary diagnoses included multiple myeloma (MM) in 85 (44,5%) cases, Hodgkin’s lymphoma (HL) in 63 (33%) cases, various types of non-Hodgkin’s lymphomas (NHL) in 34 (17,8%) cases, such as peripheral T-cell lymphoma - 6 (3,1%), mantle cell Lymphoma - 7 (3,7%), primary CNS lymphoma - 4 (2%) and 9 other cases (4,7%). The «other»category included desmoplastic small round cell tumors, germ cell tumors, and Ewing’s sarcoma. Conditioning regimensused in lymphoproliferative diseases: LEAM for HL, NHL; Thiotepa+Lomustine and TBC for PCNSL, MEL 200 for MM and AL-amyloidosis. Median stem cell dose was 6,25x106/kg (2,3-34,5). Median neutrophil engraftment was 12 days (range 8-37), median platelet engraftment 16 days (range 9-62), respectively. The average annual number of SCTs increased by 162% to 44,5 in 2022 and 2023 compared to the previous years. In 2022, 31 auto-SCT were performed, and in 2023 this number increased significantly by 58% and reached 58 in 10,5 months, despite of ongoing conflict. The average follow-up time in this cohort of patients (2022-2023) was 355 days (CI 297,5-539,5). Specifically, in the years 2022 and 2023, the TRM rate was 1,14% for 88 transplantations. The 100 days-TRM rate was 3,66% for the entire cohortfor all years, mainly due to lymphoma’s rate. Survival rates were assessed for all hematologic malignancies, 1-year PFS was 89,4%, 88,9% and 73,4% for MM, HL, and NHL, respectively. The 1-year OS reached 90,6% and 90,5% for MM and HL and 76,5% for NHL, respectively.

Conclusions: Amidst the Russian invasion of Ukraine, the NCI showcased exceptional resilience, evidenced by an 87% surge in HDCT and SCT in 2023 compared to the previous year. One year survival rates and acceptable short-term mortality rates highlight the medical community’s capacity to sustain high-quality healthcare services even in the midst of war.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P312 HOME-BASED HEMATOPOIETIC CELL TRANSPLANTATION IS FEASIBLE AND SAFER IN TERMS OF NUTRITIONAL PARAMETERS, TOXICITY AND LENGTH OF HOSPITALIZATION STAY

Javier Cornago Navascués1, Laura Pardo Gambarte 1, Juan Carlos Caballero Hernáez1, María Carolina Dassen1, Ana Rodríguez-Calvo Parra1, Alberto Sánchez Donaire1, Jesús Ignacio Merlo Luis1, José Luis López Lorenzo1, Amalia Domingo González1, Pilar Llamas Sillero1, Laura Solán Blanco1

1University Hospital Fundación Jiménez Díaz, Madrid, Spain

Background: Patients undergoing hematopoietic cell transplantation (HCT) require extensive hospitalizations. Home HCT programs, wherein providers make trips to patients residence to perform assessments and deliver any necessary interventions, may enhance patient quality of life and improve outcomes.

Methods: We performed a retrospective and observational study in a tertiary and university hospital. 96 adult patients underwent autologous HCT between January 2021 and April 2023 in two modes: hospitalized and home-based transplants. Some variables were collected: antropometric, analitical and dynamometric parameters, not only at the admission but also at the discharge of procedure. For quantitative variables, we choose the median and quartiles for description and the Mann-Witney U test for comparison. Qualitative variables are described with frequencies and percentages and are compared with the Chi-square test or Fisher’s exact test. Statistical analysis were made by software SPSS v. 29.0.1.

Results: Of the 96 patients, 44 (45.8%) were women. The median age of our serie was 56.6 years (26-72). 79 (82.3%) HCT were performed at hospital and 17 (17.7%) in the home program. At admission, the two cohorts were comparable regarding age distribution, sex, hematologic malignancies, HCT-comorbidity index (CI), dynamometric parameters and blood test results. The table(qualitative) summarizes patient characteristics.

Albumin levels at discharge were higher in home-based HCT than in those hospitalized: 4 (3.2 - 4.9) vs. 3.5 (2 - 4.4), p < 0.01. Also home-based HCT patients presented significantly less mucositis 23.5% vs. 63,3% (p=0.006) and therefore less need for parenteral nutrition 5,9% vs. 83,5% (p < 0.001).

Patients who presented mucositis after HCT had significantly lower albumin levels at discharge 3.5 (2 - 4.5) vs. 4 (3 - 4.9) p=0.013, as well as worse muscle function with dynamometry values also significantly worse achieved at discharge: 26 (4 - 46) vs. 32 (16 - 56), p=0.005.

In hospitalized patients, older age was related with an increased risk of mucositis (OR 1.05; 1.0 - 1.11) p=0.03. This risk apparently disappeared in home-based program (OR 1.02; 0.92 - 1.17) p=0.68, so home transplant must be taken into account in older patients.

Home-based HCT patients also presented lower rate of fever and diarrhea and higher cholesterol levels at discharge. Moreover, they reported shorter length of hospitalization, less need for red blood cell transfusion (OR 0,13; 0,01 - 0,68; p=0,012) and later neutrophil engraftment (due to non-systematic use of G-CSF) without an increase in the incidence of infections. In fact, home HCT could decrease the risk of bacteriemia (OR 0,17; 0,01 - 0,93; p=0,039). There were no differences in need for ICU admission, hospital readmission in the first 14 days after discharge, relapse or death.

Variable

Hospitalized

Home-based

P value

>65 years old

1.0

No

61 (77,2%)

13 (76,5%)

Yes

18 (22,8%)

4 (23,5%)

Sex

1.0

Female

36 (45,6%)

8 (47,1%)

Male

43 (54,4%)

9 (52,9%)

Disease

0.1

Multiple myeloma

53 (67,1%)

13 (76,5%)

Lymphoma

26 (32,9%)

3 (17,6%)

Other

0 (0%)

1 (5,9%)

ICU admission

0.534

No

73 (92,4%)

17 (100%)

Yes

6 (7,6%)

0 (0%)

Readmission (14 days)

0.548

No

76 (96,2%)

16 (94,1%)

Yes

3 (3,8%)

1 (5,9%)

Relapse

1.0

No

70 (88,6%)

15 (88,2%)

Yes

9 (11,4%)

2 (11,8%)

Death

0.447

No

72 (91,1%)

17 (100%)

Yes

7 (8,9%)

0 (0%)

Parenteral nutrition

< 0.001

No

13 (16,5%)

16 (94,1%)

Yes

66 (83,5%)

1 (5,9%)

Oral mucositis

0.006

No

29 (36,7%)

13 (76,5%)

Yes

50 (63,3%)

4 (23,5%)

Fever

0.001

No

13 (16,5%)

10 (58,8%)

Yes

66 (83,5%)

7 (41,2%)

Diarrhea

0.008

No

4 (5,1%)

5 (29,4%)

Yes

75 (94,9%)

12 (70,6%)

Clost. Difficcile

1.0

No

72 (91,1%)

16 (94,1%)

Yes

7 (8,9%)

1 (5,9%)

Table. Comparison between hospitalized and home-based HCT (qualitative variables).

Conclusions: Home-based HCT offers a significant advantage in terms of not only less development of mucositis and need for parenteral nutrition, but also in shorter hospitalization stay, less transfusional requirements and infectious risk. Patients who present mucositis have significantly lower albumin levels at discharge and worse muscle strength.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P313 A RISK-ADAPTED STRATEGY TO SELECT AUTOLOGOUS OR ALLOGENEIC TRANSPLANT CONSOLIDATION IS EFFECTIVE IN MATURE T-CELL LYMPHOMA: A SINGLE CENTER EXPERIENCE

Alessio Maria Edoardo Maraglino1, Simona Sammassimo1, Viviana Amato1, Anna Vanazzi1, Paulina Maria Nierychlewska1, Ginevra Lolli1, Simonetta Viviani1, Tommaso Radice1, Fabio Giglio 1, Federica Gigli1, Rocco Pastano1, Corrado Tarella1, Enrico Derenzini1,2

1Division of Haemato-Oncology, Haematology Programme, IEO European Institute of Oncology IRCCS, Milano, Italy, 2University of Milan, Milano, Italy

Background: Treatment consolidation with either autologous or allogeneic stem cell transplantation (ASCT or alloSCT) has been proposed in order to improve outcome in Mature T-cell lymphomas (MTCL), with no consensus on the optimal transplant strategy. In this study we retrospectively reviewed the outcome of MTCL patients treated at the European Institute of Oncology (IEO) since 1995, with the aim of determining the impact of different consolidation strategies adopted in clinical practice over time.

Methods: Clinical data of MTCL pts treated at IEO from 1995 to 2021 were retrospectively reviewed. While in the first 2 decades (1995-2004 and 2005-2014) ASCT was increasingly used as first-line consolidation, starting from 2015 we introduced a risk-adapted strategy for transplant eligible pts ≤65 years, reserving ASCT consolidation only for pts in complete remission (CR) with no evidence of lymphoma contamination in the apheresis products. All patients achieving less than CR at the end of induction treatment (EOT), or with evidence of apheresis contamination underwent allo-SCT consolidation.

Factor

N(%)

Sex

Male

51 (55%)

Female

42 (45%)

MTCL subtype

Anaplastic Lymphoma Kinase (ALK)-positive

10 (11%)

Anaplastic Lymphoma Kinase (ALK)-negative

19 (20%)

Peripheral T-cell Lymphoma not otherwise specified

49 (53%)

Other

15 (16%)

Age, median (range)

59 (18-82)

Transplant eligible

56 (60%)

Results: Ninety-three MTCL pts were diagnosed and treated at IEO between 1995 to 2021 (Table 1). Median follow-up was 6.7 years (1-25). Excluding ALK pos ALCL, 56 transplant-eligible pts ≤65 years were considered, of whom 31 received transplant consolidation (23 ASCT, 8 allo-SCT). Response to induction therapy was a powerful outcome predictor (5-y OS 80% in CR pts vs 46% in PR pts vs 21% in non-responders, p=0.006). Notably, the CR rate at the end of induction did not change significantly over time (30% vs 34% vs 43% in the 1st, 2nd and 3rd period respectively p=0.7); however in the 1st decade only 4 of 18 pts received transplant consolidation (ASCT in all cases) vs 13 of 23 pts in the 2nd decade (13 ASCT, with 1 tandem ASCT-alloSCT) vs 14 of 15 pts in the 3rd period (7 allo-SCT and 7 ASCT) (p<0.0001). The 5-year OS of pts treated in the 3rd period was significantly higher (63%) compared to the 1st (37%) and 2nd decade (46%) (p=0.002). Accordingly, while transplant consolidation considered per se did not improve OS compared to the sole observation, the risk-adapted consolidation strategy was associated with a superior outcome compared to either observation or ASCT consolidation given before 2015, with 5-y OS of 63% vs 36% vs 43% respectively (p=0.01).

Conclusions: These data suggest that a risk adapted first-line consolidation strategy including allo-SCT could improve outcome in MTCL.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P314 EFFECTS OF THE PERIPHERAL EXPANSION OF CYTOTOXIC T LYMPHOCYTES ON OUTCOMES FOLLOWING HAPLOIDENTICAL ALLOGENEIC STEM CELL TRANSPLANTATION

Lucía García Tomás1, Oriana Jimena López Godino 1, Cristina Aroca Valverde1, Inmaculada Heras Fernando1, María Luisa Lozano Almela1

1University Hospital Morales Meseguer, Murcia, Spain

Background: The peripheral expansion of donor-derived cytotoxic T lymphocytes (CTL) during the early stages of allogeneic hematopoietic stem cell transplantation (HSCT) is a widely recognized physiological phenomenon in the process of immunologic recovery. However, the significance of this phenomenon in situations where it persists beyond the initial period of HSCT remains unknown. In this context, we aimed to assess the nature of CTL after haploidentical stem cell transplantation (haploHSCT) with post-transplantation graft-versus-host disease (GVHD) prophylaxis based on post-transplant cyclophosphamide (CyPT), as well as its impact on the occurrence of clinically relevant events.

Methods: We performed a retrospective analysis of 71 patients who consecutively underwent haploHSCT at our center between 2012 and 2022, including patients with at least one year of follow-up. We defined peripheral expansion of CTL (eCTL) as when the number of CD8 + T lymphocytes exceeded 2 x 10^9/L. The presence of clinically significant events was assessed on days +56, +180, and +365. Statistical analysis was conducted using the SPSS (v29.0.1.0) software.

Results: Thirteen patients met the eCTL criteria, while 58 were non CTL. Factors associated with CTL included having received a previous stem cell transplant (autologous or allogeneic, p= 0.04) and CMV reactivation (p= 0.01, Table 1). Analysis of relevant infectious episodes during each post-transplant period revealed that, overall, there were more episodes in the first year post-transplant in the group of patients who developed CTL. Additionally, they experienced more viral infections (CMV reactivation and respiratory infections) between days 0 and +30 (p= 0.01), fungal infections between days +30 and +180 (p <0.01), and bacterial infections between days +181 and +365 (p= 0.03).

With a median follow-up of 27 months (range: 14-46), the overall survival (OS) median for the entire cohort has not been reached. No differences were found in terms of OS based on the presence or absence of eLTC [p= 0.6; hazard ratio (HR)= 0.77; 95% confidence interval (CI), 0.22-2.6]. Although the median relapse-free survival has not been reached either, and there were no statistically significant differences in the CTL group [p= 0.3; HR= 0.5; 95% CI, 0.1-2.7], there was a trend towards a lower cumulative incidence of relapse in this group. CTL was also not associated with other complications of stem cell transplantation, including both acute and chronic GVHD or transplant-related mortality.

Conclusions: In the context of haploidentical stem cell transplantation with post-transplant cyclophosphamide (haploTPH with CyPT), CTL was associated with infectious episodes, particularly early viral infections (before +30), fungal infections (between +31 and +180), and bacterial infections beyond +181. No statistically significant differences were observed in terms of overall survival between both groups. It is anticipated that the impact of CTL on relapse may be corroborated in future studies with a larger number of patients.

Disclosure: No disclosure.

1: Haematopoietic Stem Cells

P315 OUTCOME OF ALLO-HSCT IN THERAPY RELATED MYELOID NEOPLASMS WITH A HISTORY OF OVARIAN CANCER PREVIOUSLY TREATED WITH CHEMOTERAPY AND PARP INHIBITORS

Alessio Maria Edoardo Maraglino1, Simona Sammassimo1, Patrizia Chiusolo2, Cristina Papayannidis3, Federica Gigli1, Filippo Frioni2, Chiara Sartor4, Viviana Amato1, Rocco Pastano1, Elisabetta Todisco5, Corrado Tarella1, Enrico Derenzini1,6, Fabio Giglio 1

1Division of Haemato-Oncology, Haematology Programme, IEO European Institute of Oncology IRCCS, Milano, Italy, 2Fondazione Policlinico Universitario A. Gemelli IRCCS, Roma, Italy, 3IRCCS Azienda Ospedaliero-Universitaria di Bologna Istituto di Ematologia “Seràgnoli”, Bologna, Italy, 4Istituto di Ematologia “Seràgnoli”, Università degli Studi di Bologna, Bologna, Italy, 5U.O. Ematologia ASST Valle Olona Busto Arsizio, Busto Arsizio, Italy, 6University of Milan, Milano, Italy

Background: PARP inhibitors (PARPi) improved ovarian cancer (OC) management, but are associated with an increased risk of therapy-related myeloid neoplasms (t-MN) with poor outcome.

Methods: We investigated the impact of allo-HSCT in pts with t-MN associated with a prior treatment including PARPi for OC. This is a multicenter (3 Centers) retrospective study evaluating outcomes of 11 consecutive pts affected by t-MN who underwent allo-HSCT between 2017 and 2023.

Results: t-MN developed after a median PARPi exposure of 22 months (15-52). All patients had previoulsy received platinum-based chemotherapy for OC. Median age at t-MN onset was 47 years (50-68). 5 pts (45%) were BRCA mutated. At diagnosis of t-MN 10 pts (90%) showed OC complete remission (CR), one patient had stable disease. 3 pts (26%) developed a myelodysplastic syndrome/Acute Myeloid Leukemia (MDS/AML) (2 with mutated Tp53 and 1 with myelodysplasia-related gene mutations with mutated RUNX1), 4 pts (37%) showed MDS with mutated Tp53 and 4 pts (37%) AML [2 with mutated Tp53, 1 not otherwise specified and 1 with myelodysplasia-related cytogenetic abnormalities]. 4 pts (37%) presented complex karyotype. 8 pts (74%) received cytarabine and an anthracycline (4 pts received CPX-351), 2 pts (16%) combination of hypomethylating agent and venetoclax and 1 pt (10%) with MDS with mutated Tp53 received upfront allo-HSCT. Following induction therapy, 6 pts (54 %) reached CR with negative minimal residual disease (MRD), 3 pts (26%) showed CR with MRD persistence by immunophenotyping and 1 pt (10%) was refractory. 8 pts (74%) received myeloablative conditioning, 3 pts (26%) reduced intensity. All pts received GVHD prophylaxis with post-HSCT cyclophosphamide, Mycophenolate Mofetil, FK506. 5 pts (45%) had haploidentical donor, 5 pts (45%) a matched unrelated donor and 1 pt (10%) had matched sibling donor. Donor engraftment occurred in 8 (74%) pts with full donor chimerism by day 30, reaching neutrophil and platelet graft with a median time of 17 (14-23) and 18 (10-42) days respectively. 3 pts (26%) developed steroid-sensitive cutaneous acute GVHD grades II-III. 3 pts (26%) showed graft failure: 1 patient showed AML persistence, 1 pt died for infections and 1 pt died 149 days after a second allo-HSCT for GVHD grade IV. After a median time of 180 days (90-1350) from allo-HSCT, 7 pts (64%) died [5 (45%) for t-MN relapse and 2 pts (16%) for transplant related mortality]. 1 pt (10%) was alive with persistent AML. 3 pts (26%) were alive and in t-MN CR. Only 1 pt (10%) developed a progressive OC at 42 months after allo-HSCT, now receiving treatment.

Conclusions: These data indicate that t-MN after PARPi often display high-risk genetics and poor outcome. However a minority of pts can be salvaged with allo-HSCT. In this light, the clinical history of these pts should not be considered a prohibitive factor for intensive treatment and allo-HSCT. Allo-HSCT can also offer the chance to continue OC-directed therapy that wouldn’t otherwise be feasible. Considering the expanding role of PARPi in different diseases the clinical need is expected to increase and new therapeutical approaches are necessary.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P316 EFFECTS OF LOW-DOSE HYPOMETHYLATING AGENTS ON THE PREVENTION OF RELAPSE AFTER TRANSPLANTATION IN PEDIATRIC HIGH-RISK AML

Qingwei Wang1, Shengqin Cheng1, Wei Gao1, Qi Ji1, Peifang Xiao1, Jun Lu1, Jie Li1, Shaoyan Hu 1

1Children’s Hospital of Soochow University, Suzhou, China

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is the only curative option for patients suffering from high-risk (HR) acute myelocytic leukemia (AML). However, 20% to 50% of patients face relapse, which is the main cause of death after HSCT. For HR AML, maintenance therapy post HSCT may improve relapse-free survival (RFS). Low-dose hypomethylating agents (HMAs) post HSCT have presented promising results for adult AML but limited data for children.

Methods: We conducted a retrospective study at the Children’s Hospital of Soochow University. Between August 2019 and April 2023, 173 AML children (according to CALS III-AML 18, Registration ID ChiCTR1800015883) due to HR (n=103), refractory or relapsed (RR, n=40), HR + RR (n=30) received HSCT (the second transplantation was excluded). There were 100 boys and 73 girls with a median age of 6 years and 9 months (range, 3 months-16 years). All the patients were in complete remission before HMAs post HSCT. 53 of the 173 children received HMAs maintenance (Decitabine were given as 10-15 mg/m 2 per day subcutaneously for 3 to 5 days every 28 days for at least 4 cycles; Azacitidine were given 32 mg/m 2 per day subcutaneously for 3 to 5 days every 28 days for at least 4 cycles). The median number of intervention was the 123th day after HSCT. The control group (n=120) had no HMAs intervention. All the patients were not lost to follow-up as of October 2023.

Results: Until the date of follow-up, the median number of cycles was 4. A total of 31 children achieved at least 4 cycles and 17 patients withdrew from maintenance therapy (abnormal liver function (n=4), abnormal renal function (n=1), pulmonary infection (n=1), MOG antigen-associated optic neuritis (n=1), GVHD (n=2), relapse (n=5), no specific reasons (n=3) and no serious adverse events occurred during maintenance treatment. The overall survival (OS) rate between HMAs group and control group is 94.3% vs 77.5% and the RFS rate between the two is 90.6% vs 66.7%. HMAs as maintenance can improve OS and RFS ([95% CI, 38.84-43.78; P=.006] and [95% CI, 31.45-37.23; P=.001]). For patients with different HR fusion genes, HMAs can help patients harbouring CBF-AML with KIT (P 0.044), KMT2A-rearranged (P=.037) but not improve patients harbouring FUS::ERG (P=.655) and CBFA2T3::GLIS2 (P=.181). There was no significant difference for graft versus host disease (GVHD), whether in aGVHD between HMAs group and control group (I aGVHD P=.405,II-III aGVHD P=.476, IVaGVHD P=1) or cGVHD between HMAs group and control group (localized cGVHD P=.578, generalized cGVHD P=1).

Conclusions: HMAs can prevent relapse after HSCT in pediatric HR AML and are safe and well tolerated for children, and play the graft versus leukemia effect while not increase the risk of GVHD. However, different HR fusion genes have different clinical effects, which helps us to identify patients who are more suitable for HMAs maintenance therapy.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P317 OUTCOMES OF PEDIATRIC CHRONIC MYELOID LEUKEMIA IN ADVANCED PHASE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH REDUCED-INTENSITY CONDITIONING REGIMEN

Polina Sheveleva1, Elena Morozova1, Anna Osipova1, Olesya Paina1, Olga Slesarchuk1, Tatyana Gindina1, Ekaterina Izmailova1, Ildar Barkhatov1, Tatyana Bykova 1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1

1RM Gorbacheva Research Institute of Pediatric Oncology, Hematology and Transplantation, Pavlov University, Saint Petersburg, Russian Federation

Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is still the only curative method of children and adolescents with chronic myeloid leukemia (CML) in advanced phase. The aim of study was to the analysis the efficacy of reduced-intensity conditioning regimen (RIC) to reduce transplant-related complications and mortality in pediatric CML in accelerated phase (AP) and blast crisis (BC).

Methods: Twelve patients (pts) with CML in advanced phases (AP – 9, lymphoid BC – 3) after a failure of treatment with TKIs (imatinib in 2 pts, switched to 2G-TKIs with dasatinib or nilotinib 10 pts) received allo-HSCT between 2008 to 2023. The median age at diagnosis was 13 years (5 to 18), median age at allo-HSCT – 18 years (range 8– 32). At the moment of allo-HSCT 9 pts were in chronic phase 2, 3 pts in AP2. Median interval from diagnosis to allo-HSCT – 38 months (range 5 –188). Indications for allo-HSCT were as follows: the lack or loss of molecular response (MR) and/or cytogenetic response (CyR) (n=5), lymphoid BC (n=3), T315I mutation (n=3), ASXL1 mutation (n=1). Karnofsky (Lansky) performance status was 80% in 3 pts, 90 % in 6 pts, 100% in 3 pts. RIC included Bu 8-10 mg/kg and Flu 150-180 mg/m2 (n=9, 75%) or Mel 140 mg/m2 and Flu 150-180 mg/m2 (n=2, 17%). The myeloablative conditioning regime (MAC) was done in 1 pt – Bu 16 mg/kg and Cy 120 mg/kg (n=1, 8 %). Matched related donor transplantation was done in 2 pts, matched unrelated (MUD) – in 5 pts, mismatched unrelated (MMUD) – in 4 pts, haploidentical – in 1 pt. In 9 pts (75 %) T-cell depletion was based on PTCY, in 3 pts (25%) – on anti-thymocyte globulin. Bone marrow (n=5) or peripheral blood (n=7) were used as stem cell source; median number of CD34+ cells of 7,4 (1,8 to 10,3) or 6,5 (5,0 to 13,6) ×106/kg weight of the recipient, respectively.

Results: Ten patients (83 %) achieved engraftment with median of 20 days (11 to 27); 9 of them achieved сomplete hematologic response (CHR) and CCyR, 6 pts achieved CMR оn day +100. Two patients received second allo-HSCT due to primary graft failure, 1 of them received third allo-HSCT. Grade III–IV acute GVHD was registered in 16 % (n=2) of cases. Severe chronic GVHD was not registered. One pts with ASXL1 mutation achieved only a partial hematologic response after allo-HSCT and started 3G-TKIs (asciminib). Four pts (33%) relapsed after allo-HSCT, among them 1 pt with hematologic relapse received chemotherapy + 2G-TKIs, 3 pts with molecular relapse received donor lymphocyte infusion + 2G-TKIs that resulted in CMR. Three pts died: grade IV aGVHD in 1 pt with MAC, infection (n=1) after third allo-HSCT and disease progression (n=1). OS after allo-HSCT was 75 % with a median follow-up of 38 months.

Conclusions: Allo-HSCT with RIC is an effective and relatively safe therapeutic option in children and adolescents with CML in advanced phases.

Disclosure: The authors have no conflicts of interest to declare.

1: Haematopoietic Stem Cells

P318 UPDATE ON DONOR DERIVED NK CELLS INFUSION FOLLOWING HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANT IN PEDIATRIC HIGH-RISK AND RELAPSED/REFRACTORY LEUKEMIA

Emanuela Rossitti1, Fabiana Cacace2, Flavia Antonucci1, Valeria Caprioli2, Maria Rosaria D’amico2, Giuseppina De Simone2, Maria Simona Sabbatino2, Mario Toriello2, Giovanna Maisto2, Roberta Penta de Vera d’aragona2, Francesco Paolo Tambaro 2

1AOU Federico II, Naples, Italy, 2AORN Santobono Pausilipon, Naples, Italy

Background: Natural killer (NKs) lymphocytes are cells of innate immunity, involved in early defense against tumor and infections. Their function is regulated by a balance between activator and inhibitory signals through surface receptors as the Inhibitory Killer-cell Immunoglobulin-like Receptors (iKIRs) that allow NKs to discriminate between healthy self and non-self cells. In haploidentical hematopoietic stem cell transplantation (haplo-HSCT), NKs play a key role in preventing early relapse and virus reactivation and exert an efficient GvL effect, without increasing GvHD incidence.

After haplo-SCT, donor NKs deriving from CD34+ stem cells are immature cells with poor cytolytic activity (CD56bright). Their progression toward more efficient and terminally differentiated CD56dim NKs is characterized by acquisition of KIRs, CD16 and CD57 and it is completed after 4-6 weeks. In patients undergoing haplo-HSCT, the infusion of phenotypically mature donor NKs could be useful in ensuring immunological control of disease and viral reactivations, before reconstitution of graft in the recipient.

We evaluated the outcome of four pediatric patients with high-risk leukemia who underwent two different platforms of haplo-HSCT plus add-back of donor NKs (NK + ), comparing them with five haplo-HSCT pediatric patients without add-back of NKs (Nk-).

Methods: Population study characteristics are summarized in the table. Median follow-up was 15 months after transplantation. Three patients had infant leukemia, two patients had relapsed-refractory disease. All the patients received a median of 3 lines of chemotherapy and one of them received a prior allo-HSCT. Donor was selected from parents based on genetic, immunological and clinical criteria. Four patients received αβ T/B cells depletion haplo-HSCT; one patient received PTCY haplo-HSCT. Characteristics of transplant, conditioning regimens and graft composition are summarized in the table. Patients underwent αβ T/B cells depletion haplo-HSCT received rituximab and letermovir for EBV and CMV prophylaxis, respectively. All patients received donor NKs infusion at median day of +35 after transplantation, except patient 5 because of the onset of hepatic acute GvHD. NKs count were monitored on day +7, +14, +21, +28 after transplantation and on day +7, +14, +21, +28 after NK infusion. Median NKs infused were 2x107/kg.

Results: In the Nk+ population, the median time to neutrophil and platelet engraftment was on day +14 and +10, respectively. CMV reactivation was significantly reduced in the NK+ as compared to NK- (p-value=0.017). ADV reactivation was observed in three NK+ patients but only one required antiviral therapy. None in both groups developed EBV reactivation. No significant differences in acute GvHD (p-value=0.76) and chronic GvHD (p-value=0.81) incidence between the two groups were observed. All the NK+ patients have currently full donor chimerism and are in hematological remission (OS and EFS 100%) while two NK- patients died for disease relapse (OS and EFS 60%).

Patient

1

2

3

4

5

Age at HSCT (years)

1.2

0.7

5

18

1.25

Sex

M

M

M

M

F

Diagnosis

ALL infant

ALL infant

ALL B

AML

AML infant

Disease characteristics

t(4;11) KMT2A/AF4

t(11;19) KMT2A/MLLT1

CD10+

M1, FLT3-ITD

M5

Disease status ad HSCT

CR2

CR1

CR2

CR1

CR1

Lines of treatment pre-HSCT

3

1

> 4

3

1

Previous HSCT

1

Donor

Madre

Madre

Madre

Padre

Padre

Transplantation

Type of Haplo-HSCT

Tαβ/B depletion

Tαβ/B depletion

Tαβ/B depletion

PT-CY

Tαβ/B depletion

Source of HSC

PBSC

PBSC

PBSC

BM

PBSC

Conditioning Regimen

ATG-TH-FLU-TREO-R

ATG-TH-BU-FLU-R

TBI-TH-FLU-ATG-R

TH-BU-FLU-EX

TH-BU-FLU-ATG-R

Graft composition

TNC x 108/Kg

17.16

11.69

5.34

5.8

15.2

CD34+ X 106/Kg

7.16

18.3

9.82

4.59

19.8

CD3+ TCRαβ+ X 106/kg

0.386

0.44

0.089

na

0.239

CD3+ TCRγδ+ x 106/kg

19.81

18.7

6.3

na

13.68

CD19+ x 106/kg

0.0031

0.07

0.01

na

0.045

NKs infusion

Day of NKs infusion

62

36

35

31

Na

CD16+CD56+ x 107/Kg

2.472

2.6

1.638

0.67

na

Outcome

Engraftment

ANC > 500/uL

13

14

20

19

13

PLT > 20000/uL

8

13

10

14

7

GvHD

aGvHD

Yes (Skin)

Yes (Skin)

No

No

Yes (liver)

Grade 1-2

Yes (day +5)

Yes (day +45)

na

na

Yes (day +33)

Grade 3-4

No

No

na

na

No

cGvHD

No

Yes (day +152)

No

No

No

Virus Reactivation

CMV

Yes ( + 15 months)

No

No

No

Yes (day +125)

ADV

Yes (day +280)

No

Yes (day +270)

Yes (day +162)

No

EBV

No

No

No

No

No

Treatment

Valganciclovir

na

No

Cidofovir

Valganciclovir

Relapse post HSCT

No

No

No

No

No

Follow-up (months)

23

16

15

10

4

Conclusions: Donor NKs infusion after haplo-HSCT could represent an effective option to improve transplant outcome and prevent virus reactivation in pediatric patients with high-risk leukemia. However, these promising preliminary results need to be confirmed in larger cohorts.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P319 COMPARISONS OF SAFERTY AND EFFICACY OF ALLO-HSCT AFTER CAR T-CELL OR CHEMOTHERAPY-BASED COMPLETE REMISSION IN PEDIATRIC T-ALL

Ruijuan Sun 1, Jianping Zhang1, Yue Lu1, Yanli Zhao1, Deyan Liu1, Xingyu Cao1, Min Xiong1, Jiarui Zhou1, Zhijie Wei1

1Hebei Yanda Lu Daopei Hospital, Langfang, China

Background: Patients often undergo consolidation allogeneic hematopoietic stem cell transplantation (allo-HSCT) to maintain long-term remission following chimeric antigen receptor (CAR) T-cell therapy. Comparisons of safety and efficacy of allo-HSCT following complete remission (CR) achieved by CAR-T therapy versus by chemotherapy for T-cell acute lymphoblastic leukemia (T-ALL) has not been reported. We compared long-term survival, incidence of acute graft-versus-host disease(aGVHD), and incidence of cytomegalovirusemia and Epstein-Barr virusemia in the two groups.

Methods: Between October 2010 and June 2023, we performed a parallel comparison of transplant outcomes in 90 consecutive pediatric T-ALL who received allo-HSCT after achieving CR with CAR-T therapy (n=28) or with chemotherapy (n=62). The median age was 9 (1-14) years old. Male to female was 68:22. 59 cases in the first complete remission (CR1) were transplanted. There were 14 patients in the CarT group and 45 in the chemotherapy group. 31 cases were in ≥CR2,14 patients in the CarT group and 17 in the chemotherapy group. The donors were identical sibling in 7, unrelated in 10 and haploidentical in 73. 76 cases underwent transplantation with conditioning regimen including total body irradiation (TBI), cyclophosphamide (CTX) and antilymphocyte globulin (ATG), 14 cases with busulphan (Bu), CTX and ATG. Graft versus-host disease (GVHD) prophylaxis was mainly with cyclosporine or tacrolimus, mycophenolate mofeil and short-course methorexate.

Results: With the median follow up 16 (1-37) months, 90 patients were successfully engrafted and had full donor chimerism. Efficacy measures 1 years following transplant were all similar in the CAR-T vs. the chemotherapy groups: cumulative incidences of relapse (CIR; 3.8% vs.5.2% p=0.846) and overall survival (OS;81.5% vs. 86.7%; p=0.572). The incidence of Grade II-IV acute graft-versus-host disease was similar in both groups (aGVHD 17.9% vs.16.1%;p=0.896). And the incidence of Grade III-IV aGVHD was similar in both groups too (7.6% vs.13.3%, p=0.421). The incidence of Cytomegalovirusemia and Epstein-Barr virusemia was different between groups(CMV 26.4%vs.39%;p=0.285;EBV 7.4% vs.12.9%;p=0.461).

Conclusions: In conclusion, our data indicate that, in Pediatric T-ALL patients, similar clinical safety and Efficacy outcomes could be achieved with either CD7 CAR T-cell therapy followed by allo-HSCT or chemotherapy followed by allo-HSCT. And Car T-cell therapy use before transplantation does not increase the incidence of acute GVHD or viremia.

Disclosure: No.

1: Haematopoietic Stem Cells

P320 EFFECT OF CYTOMEGALOVIRUS REACTIVATION ON RELAPSE AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN ACUTE LYMPHOBLASTIC LEUKEMIA

Mekni Sabrine1, Ben Abdeljelil Nour 1, Rihab Ouerghi1, Turki Ines1, Khayati Malek1, Ben Yaiche Insaf1, Frigui Siwar1, Torjemane Lamia1, Belloumi Dorra1, Kanoun Rimmel Yosra1, Ladeb Saloua1, Ben Othman Tarek1

1Faculty of Medicine of Tunis, University of Tunis El Manar, Tunis, Tunisia

Background: Previous studies have demonstrated the protective effect of cytomegalovirus (CMV) reactivation against relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT) for acute myeloblastic leukemia. However, this impact remains unclear, in acute lymphoblastic leukemia (ALL). The present study aimed to assess the effect of CMV reactivation on relapse after allo-HSCT in patients with ALL.

Methods: We conducted a retrospective study on patients with high-risk ALL who received allo-HSCT after myeloablative conditioning regimen (TBI-based or Busulfan iv-based) from MSD between 2018 and 2022. Patients who had undergone previous allo-HSCT were excluded. CMV serology was performed in donors and recipients before allo-HSCT. All patients underwent weekly monitoring for CMV reactivation by quantitative polymerase chain reaction (qPCR) assay from engraftment until 100 days after allo-HSCT. All patients received antiviral prophylaxis with acyclovir from day +1 to 6 months after allo-HSCT. Preemptive treatment was initiated when a viremia was higher than 150 copies/µg DNA. It included ganciclovir, valganciclovir or foscarnet depending on blood counts, gastrointestinal symptoms and renal function.

Results: A total of 66 patients were included (children: n=29, 44 % and adults n= 37, 56 %). The median age was 20 years (range, 5 – 50 years). Before allo-HSCT, 85 % of patients were in CR1, 15% were beyond CR1 and 85% had positive pre-allo-HSCT CMV serostatus. Conditioning regimen was TBI-based in 62% of patients. GVHD prophylaxis consisted on cyclosporine and short course of methotrexate in 85% of patients and cyclosporine alone in 15% of patients. Bone marrow was the most common stem cell source (67%). After allo-HSCT, 37% of patients developed CMV reactivation after a median time of 39 days (range, 19 – 100 days). After a median follow-up of 24 months (range, 1- 66months), patients with CMV reactivation had significantly lower 2-year cumulative incidence of relapse (CIR) compared with patients without CMV reactivation (14% vs 37%, respectively, p=0.03). In multivariate analysis, CMV reactivation remains an independent factor associated with reduced CIR (OR: 0.1, 95% CI [0.02 - 0.66], p=0.016). A statistically significant increase in CIR was identified in patients with EBMT score before allo-HSCT ≥ 2 (OR: 4.4, 95% CI [1- 17], p=0.038). There was a trend for higher non-relapse mortality (NRM) in patients with CMV reactivation (p=0.06) without negative impact on overall survival (p=0.9). No deaths were directly related to CMV disease. However, the NRM was mostly caused by infection after grades II to IV acute GVHD.

Conclusions: CMV reactivation was associated with reduced risk of relapse in patients with ALL without survival benefit, which was related to the increased incidence of NRM in these patients. Further studies are needed to validate the protective effect of CMV infection on ALL relapse.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P321 LOWER INCIDENCE OF CHRONIC GVHD WITH ATLG AS GVHD PROPHYLAXIS IN ALL PATIENTS IN FIRST COMPLETE REMISSION UNDERGOING ALLOGENEIC SCT COMPARED TO POST-TRANSPLANT CYCLOPHOSPHAMIDE

Normann Steiner 1, Radwan Massoud1, Evgeny Klyuchnikov1, Nico Gagelmann1, Johanna Richter1, Christian Niederwieser1, Kristin Rathje1, Ameya Kunte1, Jannik Engelmann1, Christina Ihne1, Tatiana Urbanowicz1, Iryna Lastovytska1, Cecilia Lindhauer1, Franziska Marquard1, Mirjam Reichard1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schäfersküpper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Silke Heidenreich1, Gabriele Zeck1, Ina Rudolph1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger1

1University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: For high-risk adult acute lymphoblastic leukemia (ALL) patients in first complete remission an allogeneic stem cell transplantation (alloSCT) is currently the best available treatment option for cure. Graft versus host disease (GvHD) is still a common transplant complication with high mortality and morbidity and prophylaxis regimen includes anti-T-lymphocyte globulin and more recently in a haploidentical setting post-transplant cyclophosphamide (PTCy).

Methods: Here, we retrospectively analyze the outcome of 147 high-risk ALL patients in first complete remission with conditioning regimen TBI 8-12 GY in combination with cyclophosphamide ± etoposide or with fludarabine in the period 1993 – 2023. Of the total cohort, 74 patients received ATLG and 73 patients PTCy with calcineurin inhibitor + /- mycophenolate mofetil.

Results: The 3-year overall survival was 64% in both groups (PTCy [95% CI 52% - 78%] and ATLG [95% CI 54% - 76%]; p=0.9). The 3-year leukemia-free survival after PTCy was 64% [95% CI 57% - 80%] versus 61% [95% CI 49% - 72%] after ATLG (p=0.5). GvHD relapse-free survival after PTCy was 34% [95% CI 25%-48%] versus 43% [95% CI 33% - 56%] after ATLG (p=0.2). Cumulative incidence of relapse ± standard error (SE) at three years was 12% ± 4% after PTCy and 19% ± 4% after ATLG (p=0.4). Cumulative incidence of non-relapse mortality at three years was 21% ± 5% after PTCy and 20% ± 4% after ATLG (p=0.8). Cumulative incidence of acute GvHD grades II-IV at 100 days was 46% ± 6% after PTCy and 33% ± 6% after ATLG (p=0.08). Cumulative incidence of chronic GvHD at two years was 55% ± 6% after PTCy and 26% ± 5% after ATLG (p<0.001). Incidence of infection, CMV reactivation, and TMA was significantly higher in the PTCy group (68%, 53% and 5%) as compared to the ATLG group (56%, 31% and 2%, respectively; p<0.05).

Conclusions: Our retrospective single-center analysis with a considerable number of patients demonstrates that alloSCT with TBI-CY-ATLG in comparison to TBI-FLU-PTCy leads to similar outcomes with a lower incidence of chronic GvHD and toxicity. Prospective studies comparing these two different treatment regimens should be conducted to confirm these data.

Disclosure: No conflict of interest.

1: Haematopoietic Stem Cells

P322 LONGITUDINAL FOLLOW-UP ON HUMORAL AND T CELL IMMUNE RESPONSES AFTER COVID-19 VACCINATION IN ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTED PATIENTS

Alma Wegener 1, Line Dam Heftdal1, Sebastian Rask Hamm1, Cecilie Bo Hansen1, Laura Pérez-Alós1, Andreas Runge Poulsen1, Dina Leth Møller1, Kamille Fogh2, Mia Pries-Heje1, Rasmus Bo Hasselbalch2, Sisse Rye Ostrowski1, Ruth Frikke-Schmidt1, Erik Sørensen1, Linda Hilsted1, Marietta Nygaard1, Niels Smedegaard Andersen1, Lone Smidstrup Friis1, Søren Lykke Petersen1, Ida Schjødt1, Brian Kornblit1, Henrik Sengeløv1, Henning Bundgaard1, Peter Garred1, Kasper Iversen2, Kirsten Grønbæk1, Susanne Dam Nielsen1, Lars Klingen Gjærde1

1Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark, 2Copenhagen University Hospital, Herlev and Gentofte, Herlev, Denmark

Background: Patients undergoing allogeneic hematopoietic cell transplantation (HCT) are immunocompromised and therefore vulnerable to infections including Coronavirus Disease 2019 (COVID-19). Understanding the immune responses to COVID-19 vaccines in these patients is essential to optimize future vaccination programs. We aimed to characterize the humoral and cellular immune response after administration of up to five COVID-19 vaccines in patients who had received an allogeneic HCT at our institution.

Methods: We included adult patients who had undergone allogeneic HCT at Rigshospitalet, Denmark between 1985 and 2021 and received up to five doses of COVID-19 vaccines between 2021 and 2022. Serum antibody levels against the S protein receptor-binding domain (RBD) were measured at baseline and at 2-, 6-, 12-, 18- and 24- months after first vaccination. A positive anti-RBD Immunoglobulin G (IgG) response was defined as IgG serum levels ≥225 arbitrary units (AU)/ml and a presence of ≥25% neutralizing antibody capacity. Plasma levels of interferon-gamma (IFN-γ) after whole blood T cell S1-based peptide stimulation with SARS-CoV-2-spike protein 1 were measured at 6- and 12-months and the threshold for assay positivity was set to 200 international milliunits (mIU)/ml. Breakthrough infections were defined as a positive COVID-19 diagnostic test at least 14 days after the first vaccine dose.

Results: A total of 103 patients were included, of which 52% were men. Median age at first vaccination was 64 years (quartile 1(Q1) - quartile 3(Q3), 52 to 69 years) and median time from HCT to first vaccination was 4.1 years (Q1-Q3, 2.4 to 7.6 years). Pfizer (Comirnaty) accounted for 98–99% of the first, second, third and fourth vaccine. Pfizer (Comirnaty), Pfizer (Omikron BA.1) and Pfizer (Omikron BA.4-5) accounted for 5%, 36%, and 59% of the fifth vaccine, respectively. Median time from first to fifth vaccination was 1.6 years (Q1–Q3, 1.6 to 1.7 years) and 54% of patients had completed all five vaccine doses. A positive IgG response was seen in 5% of patients at baseline (median neutralizing index 16.54%, Q1-Q3 7.28 to 31.76%), in 87% of patients at 2 months (median neutralizing index 98.53%, Q1-Q3 97.14 to 99.01%), in 85% of patients at 6 months (median neutralizing index 98.26%, Q1-Q3 95.05 to 99.04%), in 96% of patients at 12 months (median neutralizing index 99.35%, Q1-Q3 98.97-99.54 %), in 98% of patients at 18 months (median neutralizing index 99.66%, Q1-Q3 99.47-99.73%) and in all patients at 24 months (median neutralizing index 99.21%, Q1-Q3 98.64-99.51%). We observed a positive IFN-γ response in 55% of patients at 6 months and in 73% of patients at 12 months. Breakthrough infections with SARS-CoV-2 occurred in 40 (39%) patients.

Conclusions: All patients who had undergone allogeneic HCT and received up to five COVID-19 vaccines had a positive antibody response up to 24-months after first vaccination and 73% had a positive cellular response up to 12-months after first vaccination. However, break through infections occurred in more than one-third of patients.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P323 HAPLOIDENTICAL DONOR PERIPHERAL BLOOD STEM CELL TRANSPLANTATION USING THIRD-PARTY CORD BLOOD COMPARED WITH MATCHED UNRELATED DONOR TRANSPLANTATION FOR PATIENTS WITH HEMATOLOGIC MALIGNANCIES

Xia Ma1, Yan Chen1, Yi Liu1, Tingting Cheng1, Cong Zeng1, Xu Chen1, Shiyu Wang1, Juan Hua1, Yajing Xu 1

1Xiangya Hospital, Central South University, Changsha, China

Background: Compared with haploidentical hematopoietic stem cell transplantation (haplo-HSCT), combination of haplo-HSCT with third-party umbilical cord blood has previously shown to contribute to the faster engraftment and the lower incidence rates of hematological malignancies (e.g., disease relapse and graft-versus-host disease(GVHD)). However, few relevant studies have been conducted, especially on patients undergoing haploidentical donor peripheral blood stem cell transplantation supported by third-party cord blood (Haplo-cord-PBSCT).

Methods: A total of 156 patients with hematologic malignancies who underwent Haplo-cord-PBSCT (n= 104) or human leukocyte antigen (HLA)-matched unrelated donor peripheral blood stem cell transplantation (MUD-PBSCT) (n= 52) between January 2016 and December 2021 were retrospectively analyzed.

Results: The median neutrophil and platelet engraftment time was similar between the Haplo-cord-PBSCT and MUD-PBSCT groups. The cumulative incidence of 30-day neutrophil engraftment and 100-day platelet engraftment was comparable in the two groups. In both the groups, no significant difference was observed in the cumulative incidence of grade II-IV acute GVHD (aGVHD) at 100-day (29.1% vs.28.8%), grade III-IV aGVHD at 100-day (7.8% vs. 9.6%), chronic GVHD (cGVHD) (45.3% vs. 35.1%), and severe cGVHD (13.6% vs.12.9%) at 2-year. The 2-year cumulative incidence of relapse was 12.8% and 10.0% (P= 0.341), and that of non-relapse mortality (NRM) was 14.7% and 16.2% (P= 0.681) in the Haplo-cord-PBSCT and MUD-PBSCT groups, respectively. The probabilities of overall survival(OS), disease-free survival (DFS), and GVHD-free/relapse-free survival(GRFS) at two years were 82.2% vs. 75.5%,69.9% vs. 73.8%, and 55.3% vs. 64.7% (P>0.05), respectively.

Table 2. Comparison of transplantation outcomes between the Haplo-cord-PBSCT and MUD-PBSCT groups.

Parameter

Haplo-cord-PBSCT group

MUD-PBSCT group

P

Median time of neutrophil engraftment (range)

13(9-22)

13(10-24)

0.834

Median time of platelet engraftment (range)

15(7-103)

14(8-38)

0.816

Grade II–IV acute GVHD

29.1%(95%CI,20.1-38.1%)

28.8% (95%CI,17.2-41.6%)

0.965

Grade III–IV acute GVHD

7.8% (95%CI,3.6-14.0%)

9.6% (95%CI,3.5-19.5%)

0.725

Total chronic GVHD

45.3%(95%CI,21.9-48.7%)

35.1%(95%CI,11.4-26.7%)

0.237

Severe chronic GVHD

13.6% (95%CI,6.7-21.3%)

12.9% (5.1-24.3%)

0.840

Two-year probability of OS

82.2%(95%CI,74.8-90.3%)

75.5% (95%CI,64.2-88.7%)

0.276

Two-year probability of DFS

69.9%(95%CI,61.2-79.8%)

73.8% (95%CI,62.4-87.3%)

0.551

Two-year probability of GRFS

55.3%(95%CI,42.6-63.3%)

64.7% (95%CI,52.8-79.3%)

0.284

Two-year probability of relapse

12.8% (95%CI,7.0-20.5%)

10.0% (95%CI,3.6-20.2%)

0.341

Two-year probability of NRM

14.7%(95%CI,8.4-22.6%)

16.2% (95%CI,7.4-28.0%)

0.681

Conclusions: In conclusion, the clinical outcomes of Haplo-cord-PBSCT were similar to MUD-PBSCT, and Haplo-cord-PBSCT could be alternatively used for the treatment of patients with hematologic malignancies.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P324 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH PERIPHERAL T-CELL LYMPHOMAS: A RETROSPECTIVE MULTICENTER STUDY IN CHINA

Hongye Gao1, Jiali Wang1, Zhuoxin Zhang1, Hao Zhang2, Xin Du3, Dehui Zou1, Xianmin Song4, Erlie Jiang 1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China, 2Affiliated Hospital of Jining Medical University, Jining, China, 3Shenzhen Bone Marrow Transplantation Public Service Platform, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China, 4Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China

Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) has emerged as a potentially curative option in peripheral T-cell lymphomas (PTCL). However, its clinical application is limited by treatment-related mortality. With the advent of novel therapies, the landscape of PTCL treatment is evolving, necessitating a thorough examination of the role of allo-SCT as a conventional intervention.

Methods: This retrospective study collected data from PTCL patients who underwent allo-HSCT at four different medical centers in China. Endpoints, including overall survival (OS) and progression-free survival (PFS), were measured from the allo-HSCT procedure. The cumulative incidence of relapse/progression (CIR) and non-relapse mortality (NRM) were estimated using competing risk survival analysis methods. The annual hazard of incidence was defined as the rate of relapse/progression/death during a certain year after allo-HSCT for surviving patients.

Results: A total of 61 patients with PTCL who underwent allo-HSCT were included in the study. The median age at transplant was 38 years (range: 29-48), with a male-to-female ratio of 1:1.3 (Table 1). Hepatosplenic T-cell lymphoma (HSTCL) constituted the most prevalent histological type (n= 13, 16.4%). For the entire cohort, with a median follow-up of 32.5 months (95% CI: 21.1-49.1), the 2-year OS and PFS were 61.1% (95% CI: 48.6%-76.9%) and 58.1% (95% CI: 45.8%-73.7%), respectively. The 2-year CIR and NRM were 21% (95% CI: 11%-34.0%) and 19% (95% CI: 9.4%-30.0%), respectively. This comprehensive study delves into the prognostic outcomes of distinct subtypes. Hepatosplenic T-cell Lymphoma (HSTCL) emerges with the least favorable prognosis, showcasing a 52.4% 2-year OS rate and a limited median PFS of 13.6 months. In contrast, PTCL-NOS, demonstrates a robust 80% 2-year OS. ENKTL presents a moderately positive outlook, boasting a.

2-year OS of 63.5%, while AITL exhibits a 53.3% 2-year OS. Notably, Anaplastic Large Cell Lymphoma (ALCL), irrespective of ALK status, stands out with a remarkable 100% 2-year OS.

Age at transplant (HR: 1.08 per standard deviation, P= 0.008), HCT-CI score > 1 (HR: 7.05, P= 0.003), and non-remission status (SD/PD) (HR: 8.77, P= 0.030) were independently associated with unfavorable OS. Patients in CR before allo-HSCT achieved favorable outcomes, with 2-year OS and PFS of 84.9% and 76.0%, respectively. Conversely, a poor prognosis was observed in patients with SD/PD states before allo-HSCT, where the 2-year OS was only 36.6% (95% CI: 13.3%-NA) and 14.4% (2.8%-74.4%). For HSTCL patients, the 2-year OS and PFS were 61.1% (95% CI: 48.6%-76.9%) and 58.1% (95% CI: 45.8%-73.7%).

For patients with CR/PR, the 2-year OS was 74.6%, and the 2-year progression-free survival (PFS) was 72.4%. Previous treatment lines before allo-HSCT did not influence OS (P= 0.42) and PFS (P= 0.84). The annual incidence rate (relapse/progression/death) demonstrated a decreasing trend (range: 1.5%-4.6%) within the 24 months after allo-HSCT in this group.

Clinical parameters

Overall (n=61)

Clinical parameters

Overall (n=61)

Male (%)

26 (42.6)

Median lines of treatment before allo-HSCT > 2 (range) (%)

2 (1, 4)

Age at transplant (yo, median [IQR])

38.00 [29.00, 48.00]

Disease status at allo-HCT (%)

Histological subtype(%)

CR

24 (39.3)

AITL

6 (9.8)

PR

17 (27.9)

ALK-ALCL

2 (3.3)

SD/PD

17 (27.9)

ALK + ALCL

4 (6.6)

Unknown

3 (4.9)

ANKL

3 (4.9)

Donor type, no. (%)

ENKTL

10 (16.4)

Haploidentical related

31 (50.8)

HSTCL

13 (21.3)

HLA-identical sibling

18 (29.5)

PTCL-NOS

5 (8.2)

Matched unrelated donor

1 (1.6)

Other

18 (29.5)

Unknown

11 (18.0)

Stage at diagnosis (%)

Acute GvHD grade (%)

1~2

28 (45.9)

3~4

1~2

11 (18.0)

PIT (%)

3~4

11 (18.0)

Group1-2

36 (59.0)

Not reported

11 (18.0)

Group3-4

25 (41.0)

Chronic GvHD (%)

Prior ASCT (%)

5 (8.2)

No

30 (50.8)

HCT-CI score >1 (%)

20 (32.8)

Yes

13 (21.3)

Table1. Patients’ baseline

Conclusions: The study emphasizes the diverse characteristics of PTCL and implies potential advantages associated with allo-HSCT, especially in patients attaining CR/PR before transplantation. Given the backdrop of various treatment regimens, contemplating novel therapies aimed at achieving profound remission could prove beneficial.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P325 SAFETY AND EFFICACY OF POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR MUD, MMUD AND HAPLOIDENTICAL STEM CELL TRANSPLANTS IN PAEDIATRIC POPULATION

Gloria Miguel 1, Julia Marsal1, Maria Trabazo1, Cristina Rivera1, Montserrat Rovira2, Laura Jimenez1, Paula Mazorra1, Julio Ropero3, Ana Maria Infante4

1Pediatric Cancer Center, Barcelona, Spain, 2Hospital Clinic and Pediatric Cancer Center, Barcelona, Spain, 3Clinica FOSCAL, Colombia, Colombia, 4Hospital Universitario San Ignacio, Colombia, Colombia

Background: Graft-versus-host disease (GVHD) is a major complication in hematopoietic stem cell transplantation and increases the morbidity and mortality of these patients. Prevention of this complication remains a challenge in this setting. In this purpose, we focused this study in order to assess the safety and efficacy of post-transplant cyclophosphamide (PTCy) in the paediatric population undergoing to HSCT from matched unrelated donors (MUD; 10/10 and 9/10 antigen matches), mismatch unrelated donors (MMUD; 8/10 antigen matches) and haploidentical donors.

Methods: This retrospective study analyze 30 paediatric patients who received a HSCT at Sant Joan de Déu Hospital (Barcelona) between 2016-2023. The study included patients with hematological malignancies and non-oncological hematology diseases who received PTCy as prophylaxis for GVHD. Demographics, transplant details, incidence and severity of GVHD as well as acute complications during short-term follow-up were evaluated. Statistical analysis was performed using descriptive statistics, Pearson’s correlation, and t-tests.

Results: The mean age of the patients at the time of transplantation was 9.4 years. GVHD manifested in 45.16%, classifying it according to de Magic criteria. The skin was the most predominant affected organ with 10 cases (33.33%), including 4 cases of stage 1, 4 cases of stage 2 and 2 cases of stage 3. Only the two latters required systemic corticosteroids. The upper GI tract was affected in 4 cases (13.33%), being all stage 1. Regarding the lower GI tract, there were 5 cases (16.67%). These included 2 stage 1; 1 patient with stage 2; 1 patient with stage 3 and 1 stage 4. The liver was the organ with the lowest incidence of GVHD, with only 1 case (3.33%) grading stage 2. Although PTCy was administered to all patients, the dose was adjusted depending on undergoing a haploidentical transplant, MUD 9/10 or MMUD 8/10 (50 mg/kg/day x 2 days) versus MUD 10/10 transplant (40 mg/kg/day x 2 days). In all of the cases both, peripheral blood and bone marrow were used as source of stem cells. No significant increase in mortality or serious adverse effects were seen when comparing data with populations where this strategy was not applied. No events of renal failure, acute cardiotoxicity or hemorrhagic cystitis were observed, nor was graft failure evidenced in any case. Nevertheless, follow-up was less than 2 years (averaging 1.28 ±1.04 years).

Conclusions: PTCy is a safe and effective strategy for GVHD prophylaxis in paediatric HSCT. It is applicable not only in haploidentical transplants but also in MUD and MMUD using either PBSC or BM. In addition, it seems that in 10/10 MUD transplants a reduction of PTCy dose is safe. Studies with long-term follow-up in the paediatric population are needed.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P326 BEYOND BORDERS: HSCT’S GLOBAL SITUATIONS AND INDONESIA’S PURSUIT OF HEALTH EQUITY

Daniel Rizky 1, Ridho M. Naibaho2, Mohammed Bader Obeidat3, Mabruratussania Maherdika1, Budi Setiawan1, Eko Adhi Pangarsa1, Damai Santosa1, Catharina Suharti4, Marco A. Salvino5,6, Abel Santos Carreira7, Annalisa Paviglianiti7, Marta Peña7, Alberto Mussetti7, Anna Sureda7

1Dr. Kariadi General Hospital, Semarang, Indonesia, 2Mulawarman School of Medicine, A. W. Sjahranie Hospital, Samarinda, Indonesia, 3Royal Medical Services, Amman, Jordan, 4Diponegoro University, Semarang, Indonesia, 5PPGMS-UFBa/IDOR-Ba, Salvador, Brazil, 6Institut Catala d’Oncologia, Barcelona, Spain, 7Institut Català d’Oncologia. Hospital Duran i Reynals, IDIBELL, Universitat de Barcelona, Barcelona, Spain

Background: The campaign slogan “Close the Care Gap,” from World Cancer Day, seems challenging to accomplish in Indonesia, particularly concerning Hematopoietic Stem Cell Transplantation (HSCT) and cell therapy. HSCT is often the sole curative option for various hematological malignancies and non-malignant conditions. In this abstract, we will compare the Transplantation Ratio (TR) across different countries in the Asia Pacific and Europe.

Methods: Data on the number of HSCT procedures in Europe were obtained from the EBMT survey in 2021, while data for Asia Pacific countries were sourced from the Annual Meeting Asia Pacific Blood and Marrow Transplant 2023 reports. Population data were obtained from https://databank.worldbank.org. Transplant Ratio (TR) was calculated as the total number of HSCT procedures per 10 million inhabitants for each country, without adjusting for patients receiving treatment abroad.

Results: The EBMT survey reported a total of 47,412 HSCT procedures in 2021, with Asia Pacific contributing 34,363 procedures. Among 69 countries surveyed, the top five with the highest TR were Germany (954.61), Netherlands (926.25), Italy (901.03), Switzerland (893.91), and Israel (875.62). Conversely, the bottom five included Syria (6.57), Philippines (3.95), Bangladesh (2.42), Nigeria (0.33), and Indonesia (0.14). Despite a 357.14% increase in Indonesia’s TR to 0.64 in 2023, it remains significantly lower than global counterparts.

Given a hypothetical ideal TR of around 900, Indonesia would be faced with the monumental task of conducting approximately 25,000 transplants annually. This staggering figure represents more than a thousandfold increase compared to the current numbers. With a population exceeding 270 million, Indonesia currently has only three transplant centers, highlighting a significant health equity gap. Infrastructure, medication availability, support services, personnel training, the absence of a registry system, and inadequate funding need urgent attention. Approximately 90% of Indonesians rely on public health services, which do not comprehensively cover HSCT operational costs. Establishing HSCT centers in financially stable countries is feasible due to cost recovery not being a primary concern, yet this model proves challenging in Indonesia. Closing the care gap requires comprehensive support, particularly from the government. Adequate financing is essential, and this can take the form of government-regulated funding mechanisms and collaboration with non-government organizations for fundraising initiatives.

Conclusions: Despite a notable rise in the Transplantation Rate (TR) in the last three years, there’s still a big challenge in Indonesia. The current number of transplants, especially the TR, indicates that there’s much work to be done. Making sure everyone, no matter where they are, has a fair chance at the best treatment is crucial.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P327 EFFECTIVENESS OF THE SIMPLIFIED COMORBIDITY INDEX COMPARED TO THE HCT-CI AS A PREDICTOR OF TRANSPLANT RELATED MORTALITY AND OVERALL SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION

Maria Alejandra Nunez Atahualpa 1, Ruben Solis Armenta1, Anghela Milenka Mendoza Sánchez1, Araceli Leal Alanis1, Bogar Pineda Terreros1, Liliana Rivera Fong1, Brenda Lizeth Acosta Maldonado1, Luis Manuel Velero Saldaña1

1Instituto Nacional de Cancerología, Mexico City, Mexico

Background: Allogeneic hematopoietic stem cell transplantation (allo-HCT) is a potentially curative therapy for hematological malignancies and some autoimmune conditions. Patient comorbidities have an impact on the prognosis of patients undergoing bone marrow transplantation. There are comorbidity indices that attempt to predict transplant-related mortality (TRM) and overall survival (OS), the most used is the Haematopoietic Cell Tranasplantation-specific Comorbidity Index (HCT-CI), however, there are increasingly new tools such as the simplified comorbidity index (SCI) that with a smaller number of variables has proven to be useful to predict mortality.

Methods: Observational, comparative and retrospective study in patients over 18 years of age, who received allogeneic transplant at the National Cancer Institute in Mexico City, from January 2018 to July 2023 who were evaluated with HCT-CI and we compared results evaluating them with SCI.

Results: 84 patients undergoing transplantation were included, 43 (51.2%) were men, with a median age of 29 years (range 17 to 59 years). As a medical diagnosis; 33 (39.3%) had LAM, 37 (44%) LAL, 5 (6%) HL, 2 (2.4%) NHL, 4 (4.8%) LGC and 3 (3.6%) had some other disease. Regarding transplantation, 36 (42.9%) received a haploidentical transplant and 48 (57.1%) an HLA-identical transplant; In all cases CD34 + cells were obtained from peripheral blood; finally 75 (89.3%) underwent a myeloablative conditioning scheme and 9 (10.7%) received a reduced intensity scheme. In all of them, the HCT-CI indices were obtained observing the following distribution (0 point 36(42.9%), 1 point 33(39.3%), 2 points 15(17.9%)); while in the simplified HCT-CI score it was observed (0 points 66(78.6%), 1 point 12(14.3%), 2 points 4(4.8%) and 3 points 2(2.4%)). In the study population, the DFS was identified as 75% at 1 year and 73% at 2 years; while the OS was 74% at 1 year and 63% at 2 years.

In order to observe overall survival, a Kaplan-Meier analysis was carried out, which showed that there are no significant differences in survival depending on either the HCT-CI score at 1 and 3 years (Chi2= 1.699, p= 0.428) or of the simplified HCT-CI (chi2= 0.974, p= 0.807).

Days in hospital

PFS 2 y

TRM

2 y (%)

TRM

<100 days

OS 1 y (%)

OS 2 y (%)

OS 3 y (%)

HCT-CI

40 (15)

62

14

11

59

60

49

1

41(21)

77

12

20

75

63

63

2

38(17)

92

20

7

79

73

72

HCT-CI simplificado

39 (18)

75

14

9

71

66

61

1

42 (20)

62

17

17

58

58

58

2

38 (10)

67

50

33

50

33

3

72 (6)

50

50

50

50

50

50

Conclusions: The SCI is a useful tool to evaluate OS and TRM, with no statistical difference compared to HCT-CI, being simpler and more accessible. However, validation studies are required.

Disclosure: None.

1: Haematopoietic Stem Cells

P328 8 VS 12 GRAY TBI IN ALLOGENEIC SCT FOR ALL PATIENTS RESULTS IN SIMILAR SURVIVAL, BUT IMPROVED SURVIVAL FOR MRD + PATIENTS AFTER 12 GRAY

Normann Steiner 1, Radwan Massoud1, Evgeny Klyuchnikov1, Nico Gagelmann1, Johanna Richter1, Christian Niederwieser1, Kristin Rathje1, Ameya Kunte1, Tatiana Urbanowicz1, Jannik Engelmann1, Christina Ihne1, Iryna Lastovytska1, Cecilia Lindhauer1, Franziska Marquard1, Mirjam Reichard1, Gabriele Zeck1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schäfersküpper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Silke Heidenreich1, Ina Rudolph1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger1

1University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: For high-risk adult acute lymphoblastic leukemia (ALL) patients in first complete remission an allogeneic stem cell transplantation (alloSCT) is currently the best available treatment option for cure. Total body irradiation (TBI) is considered the preferable conditioning regime for ALL patients, several studies indicate a protective effect of TBI against relapses compared to other conditioning regimes in ALL patients and the optimal TBI dose for ALL patients is still not clearly defined.

Methods: Here, we compared outcome of 72 high-risk ALL patients in first complete remission with conditioning regimen TBI 8 Gy (n=22) or 12 Gy (n=50) in combination with fludarabine and post-transplant cyclophosphamide (PTCy) in the period 2014 – 2023.

Results: Overall survival at 2-years after 8 Gy was 65% [95% CI 46%-90%] versus 73% [95% CI 60% - 87%] after 12 Gy (p=0.3). Leukemia-free survival at 2-years after 8 Gy was 56% [95% CI 37%- 83%) versus 74% [95% CI 63% -88%) after 12 Gy (p=0.2). GvHD relapse-free survival at 2-years after 8 Gy was 32% [95% CI 16%-62%] versus 36% [95% CI 25% - 53%] after 12 Gy (p=0.7). Cumulative incidence of relapse (RI) ± standard error (SE) at 2-years was 27% ± 10% after 8 Gy and 4% ± 3% after 12 Gy (p=0.004). Cumulative incidence of non-relapse mortality (NRM) at 2-years was 14% ± 8% after 8 Gy and 20% ± 6% after 12 Gy (p=0.4). Cumulative incidence of acute GvHD grades II-IV at 100 days was 36% ± 10% after 8 Gy and 50% ± 7% after 12 Gy (p=0.3). Cumulative incidence of acute GvHD grades III/-IV at 100 days was 19% ± 9% after 8 Gy and 24% ± 7% after 12 Gy (p=0.5). Cumulative incidence of chronic GvHD (moderate/severe) at 2-years was 35% ± 11% after 8 Gy and 37% ± 7% after 12 Gy (p=0.8). MRD positive (+) patients (n=26) receiving 12 Gy (n=19) had a significantly better OS (89% vs 71%, p=0.01), LFS (83% vs 43%, p=0.001) and a lower relapse incidence (6% vs 43%, p= 0.02) compared to MRD+ patients receiving 8 Gy (n=7), respectively. MRD negative (-) patients (n=38) receiving 12 Gy (n=27) had similar OS (73% vs 80%, p=0.4), LFS (69% vs 76%, p=0.4), CRI (4% vs 18%, p=0.2) and a higher NRM (27% vs 0%, p=0.048) compared to MRD- patients receiving 8 Gy (n=11).

Conclusions: Our retrospective single-center analysis with a considerable number of patients demonstrates that 8 Gy TBI in comparison to 12 Gy TBI results in low NRM but high relapse with similar OS and LFS. In MRD+ patients, 12 Gy TBI leads to improved OS, LFS and low relapse rates. Prospective studies comparing these two different treatment regimens should be conducted to confirm these data.

Disclosure: No conflict of interest.

1: Haematopoietic Stem Cells

P329 POST-TRANSPLANT MAINTENANCE THERAPY WITH AZACITIDINE FOR HIGH-RISK MYELOID MALIGNANCIES: WHO BENEFITS THE MOST?

Linli Lu1, Xin Li 1, Qian Cheng1

1The Third Xiangya Hospital, Central South University, Changsha, Hunan, China

Background: Disease relapse is a major cause of treatment failure after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). The purpose of this study was to investigate the effectiveness and safety of using azacitidine (AZA) as a maintenance therapy to prevent relapse following allo-HSCT in patients with high-risk AML and MDS. Additionally, our study tried to preliminarily identify the patient populations who would benefit the most from AZA maintenance therapy.

Methods: We performed a retrospective study and evaluated all patients with high-risk AML and MDS who underwent allo-HSCT between 2016 and 2022. Among them, 53 patients received maintenance therapy with AZA approximately on days +60 to +100 after transplantation, and 10 of these patients received preemptive therapy. The rest of 86 patients were in the control arm.

Results: A total of 139 patients were enrolled, including 125 AML and 14 MDS, median age was 35 years, and 53.2% of patients were male. In the AZA group, the median number of AZA cycles received was 8. The 1-year relapse-free survival (RFS) was 92.5% in the AZA group compared to 88.4% in the control group(P=0.437), while the 1-year cumulative incidence of relapse (CIR) was 5.7% and 9.3%, respectively (P=0.44). During the follow-up period, a total of 8 patients in the AZA group experienced relapse, all of whom were in minimal residual disease (MRD) positive disease status pre-HSCT. A total of 17(32%) patients experienced any type of grade 3-4 adverse events (AEs), with hematological toxicities being the most common. In the preemptive therapy group, 70% patients achieved complete remission (CR) after AZA treatment.

Conclusions: Our findings confirm that AZA maintenance therapy following HSCT is well-tolerated and effective in preventing relapse for high-risk myeloid malignancies. Pre-transplant MRD status was identified as an important factor in affecting the efficacy of AZA maintenance therapy. Additionally, our study revealed that early intervention with AZA preemptive therapy during the molecular relapse stage could improve the prognosis for high-risk AML/MDS patients to some extent.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P330 PHYSICIAN PERSPECTIVES ON THE BURDEN AND CLINICAL MANAGEMENT OF CYTOMEGALOVIRUS (CMV) INFECTION IN TRANSPLANT RECIPIENTS IN CENTRAL AND EASTERN EUROPE

Radovan Vrhovac 1, Alicja Dębska-Ślizień2, Tina Roblek3, Aleksandar Biljić Erski4, Milena Todorović Balint5

1University Hospital Centre Zagreb, Zagreb, Croatia, 2Medical University of Gdansk, Gdansk, Poland, 3Takeda Pharmaceuticals, Ljubljana, Slovenia, 4Takeda Pharmaceuticals, Belgrade, Serbia, 5University Clinical Center of Serbia, Belgrade, Serbia

Background: Post-transplant (PT) refractory CMV infection remains a challenging complication and a major cause of morbidity and mortality in haematopoietic stem cell transplantation and solid organ transplantation recipients.

Methods: This real-world cross-sectional study surveyed physicians treating transplant recipients with refractory PT-CMV infections in Bosnia and Herzegovina, Bulgaria, Croatia, Estonia, Hungary, Latvia, Lithuania, Poland, Romania, Serbia, Slovenia and Switzerland. The primary objective was to estimate physician-reported prevalence of CMV infection in patients receiving a transplant in the past 12 months. Secondary objectives included physician-reported treatment regimens and their effectiveness and to identify challenges and unmet needs. Analyses were descriptive.

Results: In total, 164 physicians completed the survey, most of whom were nephrologists (23.8%) and haematologists (22.6%). Overall, 51.8% of physicians considered teaching hospitals as the primary setting for treating transplant recipients with PT-CMV and estimated that a higher proportion of patients were aged 41–55 years (34.2%), male (56.8%) and received immunosuppressive therapy (85.5%). Physicians estimated the incidence of PT-CMV among patients who had received a transplant in the past 12 months as 26.4%, of whom 35.8% had recurrent infection, 21.0% treatment intolerance and 12.9% refractory CMV infection. Physicians estimated that 58.0% of patients with PT-CMV were asymptomatic (Table 1). Physicians reported that 66.5%, 16.5% and 4.0% of their current PT-CMV patients received first-, second- and third- or later-line of therapy, respectively. Overall, physicians estimated that 13.0% of patients did not receive any PT-CMV treatment, which they primarily attributed to patient tolerability issues. When managing patients with PT-CMV, physicians considered the following treatment attributes to be extremely important: maintenance of CMV viraemia clearance (40.9%), lack of renal toxicity (40.9%), good symptom control (40.9%), lack of hepatotoxicity (36.0%) and rapid viraemia clearance (34.2%). Physicians reported intravenous ganciclovir and oral valganciclovir to be completely associated with rapid clearance of viraemia (32.3%) and convenient administration (37.8%), respectively, in transplant recipients with refractory PT-CMV infection. Overall, 42.1% of physicians were moderately-to-completely satisfied with the treatment of transplant recipients with PT-CMV. Approximately half of physicians reported having limited experience with newer anti-CMV options, such as treatment with maribavir (54.9%) and prophylaxis with letermovir (50.6%). Physicians’ reasons for dissatisfaction with conventional therapies included limited treatment options (47.7%), treatment resistance (28.5%), safety concerns (27.8%) and the duration (27.8%) and speed (25.2%) of viraemia clearance. The majority of physicians considered preventing early death (48.8%) as the most important treatment goal for transplant recipients with refractory PT-CMV infection. Overall, physicians were moderately satisfied with the ability of available therapies to meet treatment goals. However, they considered limited treatment options, inaccessible healthcare services and inadequate healthcare reimbursement as major treatment barriers to improve patient outcomes.

Table 1 Estimated prevalence of CMV infections and its subtypes in transplant recipients based on transplant types in the past 12 months in Central and Eastern Europe

Transplant Type

Overall

HSCT

Lung

Liver/Pancreas/Intestine

Heart

Kidney

PT-CMV*

Physicians reporting, n

90

30

14

16

11

34

Transplant recipients who developed PT-CMV in the past 12 months, %

26.4

37.0

21.3

24.1

10.4

26.6

Symptomatic CMV status (all reported CMV patients), n (%)

Overall, n

715

241

48

118

118

190

Symptomatic

294 (41.1)

81 (33.6)

18 (37.5)

65 (55.1)

42 (35.6)

88 (46.3)

Asymptomatic

415 (58.0)

159 (66.0)

29 (60.4)

52 (44.1)

75 (63.6)

100 (52.6)

Unknown symptom status

6 (0.8)

1 (0.4)

1 (2.1)

1 (0.9)

1 (0.9)

2 (1.1)

CMV subtypes (all reported CMV patients), n (%)

Overall, n

715

241

48

118

118

190

With treatment intolerance

150 (21.0)

46 (19.1)

10 (20.8)

39 (33.1)

19 (16.1)

36 (19.0)

With recurrent infection

256 (35.8)

82 (34.0)

21 (43.8)

48 (40.7)

46 (39.0)

59 (31.1)

Refractory PT-CMV

92 (12.9)

32 (13.3)

6 (12.5)

10 (8.5)

10 (8.5)

34 (17.9)

Others

211 (29.5)

80 (33.2)

10 (20.8)

20 (17.0)

42 (35.6)

59 (31.1)

Unknown Status

6 (0.8)

1 (0.4)

1 (2.1)

1 (0.9)

1 (0.9)

2 (1.1)

  1. CMV, cytomegalovirus; HSCT, haematopoietic stem cell transplantation; PT, post-transplant.
  2. *For PT-CMV results, infectious disease specialists reported on up to two transplant types; therefore, the number of physicians reporting for each transplant type sums to greater than 90.

Conclusions: This study provides real-world evidence from Central and Eastern Europe, bridging an existing knowledge gap and providing crucial epidemiological data on the prevalence, treatment practices and physicians’ perceptions of unmet needs in managing patients with refractory PT-CMV infection. These findings may assist physicians and other stakeholders in making informed clinical decisions, thereby facilitating reimbursement processes to improve patient outcomes and treatment accessibility.

Clinical Trial Registry: Not applicable.

Disclosure: This study was sponsored by Takeda Pharma OÜ.

Radovan Vrhovac received speaker fees from Takeda, AbbVie, Astellas, Pfizer, MSD, Novartis, Zentiva, Medis Adria and participated in advisory committees of Takeda, AbbVie, Servier and Swixx.

Milena Todorović Balint received speaker fees from MSD, Roche, Takeda, Astellas, Novartis and Pfizer and participated in advisory committees of MSD, Roche, Takeda, Astellas, Novartis and Pfizer.

Alicja Dębska-Ślizień is a consultant, speaker, advisory board member and investigator in clinical trials for AstraZeneca, Bayer, Astellas, Takeda, GSK, Boehringer Ingelheim and Chiesi.

Radovan Vrhovac, Milena Todorović Balint and Alicja Dębska-Ślizień received funding for protocol development and review of this study from Takeda.

Tina Roblek and Aleksandar Biljić Erski are employees of Takeda.

1: Haematopoietic Stem Cells

P331 THE DOSES OF POST-TRANSPLANTATION CYCLOPHOSPHAMIDE IN HAPLOIDENTICAL STEM CELL ALLOGRAFTS CAN BE REDUCED

Juan Carlos Olivares-Gazca1, María de Lourdes Pastelín-Martínez2,1, Merittzel Abigail Montes-Robles2,1, Moisés Manuel Gallardo-Pérez1, Edgar Jared Hernández-Flores1,3, Max Robles-Nasta1,3, Daniela Sánchez-Bonilla1, César Homero Gutiérrez-Aguirre4, Andrés Gómez-De-León4, David Gómez-Almaguer4, Guillermo José Ruiz-Delgado 1,3, Guillermo José Ruiz-Arguelles1,3

1Centro de Hematología y Medicina Interna, Clínica Ruiz, Puebla, Mexico, 2Universidad Anáhuac Puebla, Puebla, Mexico, 3Universidad Popular Autónoma del Estado de Puebla, Puebla, Mexico, 4Servicio de Hematología del Hospital Universitario “Dr. José Eleuterio González”, Monterrey, Mexico

Background: Allogeneic hematopoietic cell transplantation (HSCT) remains the most important curative modality for several hematologic malignancies, but an HLA-matched sibling or unrelated donor is not always available, particularly for ethnic minorities and multiethnic families. Haploidentical HSCT (Haplo-HSCT) is a solution and, since it can be safely conducted on an outpatient basis, the costs can be substantially reduced; toxicity, however, remains a major problem.

Methods: All patients given a Haplo-HSCT in the Centro de Hematología y Medicina Interna in Puebla, México employing the “Mexican” method to conduct Haplo-HSCT on an outpatient basis were included. The employed protocol is as follows: Fludarabine dose of 50 mg/m2 IV daily from Day -6 to Day -4 (total dose received 150 mg/m2), Pre-HSCT Cyclophosphamide dose of 500mg/m2 IV daily from Day -7 to Day -4 (total dose received 2000 mg/m2). Post-HSCT Cyclophosphamide daily from Days +3 to +4, conventional dose: 50 mg/kg IV; reduced dose 25-30 mg/kg IV. The employed protocol is registered in www.clinicalTrials.gov, identifier NCT05780554. Between March 2013 and July 2016, the transplants were done using the conventional doses (CD) of PTCy (50 mg/Kg) on d 3 and 4, whereas after July 2016, the allografts were done using reduced doses (RD-PTCy) 25-30 mg/Kg, d 3 and 4. Twenty-one patients received the CD, whereas 14 were given RD. All grafts were started as outpatients and could be completed as outpatients in 71% and 60% of cases respectively, for the groups of CD and RD.

Results: According to the statistical analysis, the two comparative groups (CD-PTCy versus RD-PTCy) had no significant differences in terms of age, sex and hematological recovery. Cytokine release syndrome and hematological toxicity were less frequent in the group of persons given RD-PTCy, whereas the prevalence of both acute and chronic graft versus host disease was similar in the two groups (Table 1). The median OS for the group CD-PTCy is 5.7 months meanwhile for the RD-PTCy is 6.4 months. The figure 1 depicts the Kaplan-Meier OS plots for the two subsets of patients, which were not statistically different. The RFS were better in the patients grafted with RD-PTCy (2.13 mo vs 5.13 mo). The median follow up for entire group is 4.53 months (IQR: 1.1 – 18.9 mo).

Table 1. Salient features of the two groups of patients given a haploidentical stem cell transplantation. PTCy= post-transplant cyclophosphamide. CD= conventional dose (50 mg/Kg d3 & 4). RD= reduced dose (25-30 mg/Kg d3 & 4) CRS= cytokine release syndrome. GVHD= graft versus host disease. OS= overall survival. RFS= relapse free survival. a= chi square. b= Mann-Whitney´s U. c= log-rank chi square.

PTCy, mg/Kg days 3 and 4

CD

RD

p

Number

21

14

Female

11

8

Male

10

6

0.78a

Median age

30

19

0.50b

Acute leukemia

57%

70%

0.49a

Outpatient

71%

60%

0.97a

Cardiac toxicity

0%

0%

CRS

48%

20%

0.06a

Days to > 500 neutrophils

15.5

14

0.52b

Days to > 20, 000 platelets

4.3

0.43b

Months to full chimerism

4

6

0.62b

Acute GVHD, II-IV

9.50%

28.50%

0.14a

Chronic GVHD

9.50%

7.14%

0.81a

Median OS

5.73 mo

15.67 mo

0.42c

Median RFS

5.13 mo

2.13 mo

0.66c

Median follow up (months)

4.53

2.1

0.73b

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (40)

Conclusions: These results indicate that the doses of PTCy can be significantly reduced without compromising the effectiveness of the Haplo-HSCT conducted in an outpatient basis and employing the “Mexican” reduced intensity method. This preliminary observation may be considered as an idea to conduct prospective randomized studies to explore the possibility of significantly reducing the doses of PTCy in the setting of Haplo-HSCT.

Clinical Trial Registry: NCT05780554.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P332 OUTCOMES WITH ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN THERAPY RELATED ACUTE MYELOID LEUKEMIA: A SYSTEMATIC REVIEW AND META-ANALYSIS

Moazzam Shahzad 1, Muhammad Kashif Amin2, Muhammad Fareed Khalid3, Iqra Anwar2, Michael Jaglal1

1H Lee Moffitt Cancer Center, University of South Florida, Tampa, United States, 2The University of Kansas Medical Center, Kansas City, United States, 3Danbury Hospital Connecticut, Danbury, United States

Background: Therapy-related acute myeloid leukemia (t-AML), a secondary malignancy from the cytotoxic effect of anti-neoplastic drugs and ionizing radiations, has limited available treatment options. Allogeneic hematopoietic stem cell transplant (allo-HCT) is a potentially curative treatment option in t-AML patients. This systematic review and meta-analysis report the outcomes of allo-HCT in t-AML patients.

Methods: Following PRISMA guidelines, a comprehensive literature search was performed on PubMed, Cochrane, and Clinicaltrials.gov using MeSH terms and keywords for ‘hematopoietic stem cell transplantation’ AND ‘outcome assessment’ AND ‘therapy-related acute myeloid leukemia’ from the date of inception to July 31, 2023. Nine original studies reporting outcomes of allo-HCT in adult patients with t-AML were included after screening 1271 articles. The inter-study variance was calculated using the Der Simonian-Laird Estimator. Proportions along with a 95% confidence Interval (CI) were extracted to compute pooled analysis using the ‘meta’ package by Schwarzer et al. in the R programming language (version 4.16-2).

Results: A total of 2437 patients were included for analysis. (Table 1) The median age of patients was 52 (13-89) years and 32% (n=777) were male. The median follow-up time was 20 (1-193) months and the median time from prior treatment to development of t-AML was 39 (1-377) months. The median time to transplant was 5 (0.3-65) months. The donor type was matched unrelated 46% (n=1070), matched related 32% (n=746), cord blood 15% (n=335), haploidentical 3%(n=69), mismatched related 2% (n=55), and mismatched unrelated 1.5% (n=34). The stem cell source was peripheral blood in 79% (n=849) and bone marrow in 21% (n=220) of the patients. Myeloablative conditioning was used in 49% (n=1266) of the patients and 51% (n=1300) received reduced intensity conditioning (RIC). At a median follow-up of 3 (2-5) years, the pooled median overall survival was 39% (95% CI 0.32-0.46, I2=90%, p<0.01, n=2418) while the pooled median relapse rate and pooled median non-relapse mortality was 39% (95% CI 0.36-0.43, I2=29%, p=0.0.21, n=1597) and 31.4% (95% CI 0.28-0.35, I2=25%, p=0.25, n=1273), respectively. The pooled incidence of acute graft versus host disease (GvHD) was 47.1% (95% CI 0.32-0.62, I2=95%, p<0.01, n=1630) and the pooled incidence of chronic GvHD was 25.7% (95% CI 0.12-0.43, I2=96%, p<0.01, n=1630).

Full size table

.

Conclusions: Allogeneic hematopoietic stem cell transplant is an effective treatment option for therapy-related acute myeloid leukemia. Timely transplants in carefully selected patients could improve outcomes of allo-HCT in t-AML patients. However, the data is limited, and prospective studies are needed to consolidate these findings.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P333 OUTCOME OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR THERAPY RELATED ACUTE MYELOID LEUKEMIA: MULTICENTER REAL-LIFE EXPERIENCE

Alessandra Sperotto1, Matteo Leoncin2, Chiara Cigana3, Roberta De Marchi1, Carla Filì4, Albana Lico5, Corinna Greco5, Marco Petrella6, Endri Mauro7, Eleonora De Bellis8, Ilaria Tanasi9, Federico Mosna10, Giulia Ciotti1, Cristina Skert2, Mariagrazia Michieli3, Alberto Tosetto5, Carmela Gurrieri6, Francesco Zaja8, Mauro Krampera9, Michele Gottardi1, Costanza Fraenza 2

1Oncohematology, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy, 2Unit of Haematology/Bone Marrow Transplantation, Unit “Ospedale dell’Angelo”, Venezia, Italy, 3CRO Aviano, National Cancer Institute, IRCCS, Aviano, Italy, 4Division of Hematology and Bone Marrow Transplant, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy, 5San Bortolo Hospital, Vicenza, Italy, 6Hematology, Azienda Ospedaliera di Padova, Padova, Italy, 7Cà Foncello Hospital, Treviso, Italy, 8Struttura Complessa Ematologia, Azienda Sanitaria Universitaria Giuliano Isontina Trieste, Trieste, Italy, 9Hemtology, Azienda Ospedaliera Universitaria Integrata di Verona, Verona, Italy, 10Hematology and BMTU, “San Maurizio” General Hospital, Azienda Sanitaria dell’Alto Adige (ASDAA), Bolzano, Italy

Background: Therapy related acute myeloid leukemia (t-AML) is a therapeutic challenge as a late complication of chemotherapy (CHT) and/or radiotherapy (RT) for primary malignancy and autoimmune diseases. While therapeutic advances have improved survival for many cancer patients, t-AML cases are increasing annually and account for 6-8% of all acute myeloid leukemia (AML) patients. Compared to de novo AML, t-AML patients have a dismal prognosis because of older age, higher proportion of unfavorable cytogenetic abnormalities and cumulative toxicity for the primary malignancy. As a curative approach, allogeneic hematopoietic stem cell transplantation (allo-HSCT) has been performed in t-AML, resulting in improved outcomes compared to patients who did not undergo transplant. The aim of our study is to analyzed t-AML treated in Hematological Centers of North-Est Italy between January ’19 to August ’23, comparing outcomes of transplanted vs not-transplanted patients.

Methods: t-AML patients aged above 18 years treated between January ’19 to August ’23 were included in the analysis. All patients provided written informed consent on data analysis. Statistical analysis was performed using NCSS (version 2022). We assessed the impact of allo-HSCT on overall and disease-free survival (OS and DFS) as a time – dependent covariable in multivariable regression analysis.

Results: A total of 117 t-AML patients treated in 10 Centers were analyzed. Patients and transplant’s characteristics are reported in Table 1.

The median age was 67 years, with 66.0% of the population over 65 years old. A prevalence of male sex, HCT-CI above 3, poor cytogenetic risk and TP53 mutation were observed. HCT-CI included a history of solid tumors: thus, scores tended to be high among t-AML patients. The most common primary malignancy was breast cancer among solid tumors (33-45.0%) and lymphomas among hematologic malignancies (24 -58.5%). Chemotherapy plus hormonotherapy plus immunotherapy was the prevalent treatment for primary malignancy (41.0%), followed by CHT plus RT (29.0%). Treatment for t-AML included: only supportive therapy – 12 (10.5%) patients; standard CHT (3 + 7 regimens) – 20 (17.0%) patients; hypomethylating agent (HMA) plus venetoclax – 27 (23.0%) patients; only HMA – 26 (22.0%) patients and CPX-351 - 32 patients (27.5%). Allo-HSCT was performed in 30 (25.5%) patients. The 3-year OS of the whole population was 31.0% (95% CI: 25.6 – 38.7%) with a median duration of 12.0 months (95%CI: 9.5 – 19.7). The 3-year DFS of transplanted vs not transplanted patients was 56.0% (95%CI: 49.0 – 61.0%) vs 24.0% (95%CI 18.0 – 31.0) – p value=<.0001.

The survival analysis identified age at t-AML, unfavorable karyotype, TP53 mutation, CPX -351 for induction and performing transplant as significant prognostic factors for OS. In multivariate analysis only performed transplant was an independent favorable prognostic factor for survival (Table 1).

Table 1. Patient’s and Transplant’s characteristics

Total N. of patients

117

Male/Female

67 (57.0%)/50 (43.0%)

Median age (range)

70 yrs (24 – 85)

Age >65yrs

67 (66.0%)

Previous disease

• Onco-hematological disease

41 (36.0%)

• Oncological disease

73 (64.0%)

Time to t-AML

60 months (12 – 396)

Cytogenetic Risk

107 patients

• Favorable

15 (14.5%)

• Intermediate

27 (25.0%)

• Adverse

65 (60.5%)

TP53 mutated

28/87 (32.0%)

Treatment for t-AML

• Supportive therapy

12 (10.5%)

• Standard chemotherapy

20 (17.0%)

• Hypomethylating + venetoclax

27 (23.0%)

• Hypomethylating alone

26 (22.0%)

• CPX-351

32 (27.5%)

Allogeneic stem cell Transplant

30 (25.5%)

Median age (range)

64.5 (52 – 74)

Age 70 yrs

5 (16.5%)

Male/Female

16 (53.5%)/14 (46.5%)

Donor:

• Match Unrelated

15 (50.0%)

• Haploidentical

10 (33.5%)

• Sibling HLA-ID

5 (16.5%)

Conditioning regimen

• Myeloablative

6 (20.0%)

• Reduced Intensity

24 (80.0%)

Status at transplant

• Complete remission MRD pos

10 (33.5%)

• Complete remission MRD neg

8 (26.5%)

• Active disease

12 (40.0%)

Univariate analysis for Overall Survival Pr>ChiSq HR 95%CI

• Age at t-AML (continuous variable). 0001 1.02 1.013 – 1.042

• Previous disease.54 1.1 0.79 - 1.53

• Treatment of primary disease 0.95 1.1 0.9 - 1.1

• Cytogenetic 0.00001 0.446 0.3 - 0.6

• TP53 mutation 0.001 0.45 0.23 - 0.83

• CPX-351 for induction 0.002 0.34 0.11 - 0.93

• Performing transplant <.00001 0.55 0.3 - 0.95

Multivariate analysis for Overall Survival Pr>ChiSq HR 95%CI

• Performing transplant <.0001 1.65 1.13 - 2.39

  1. HR= Hazard ratio; CI= confidence limits

Conclusions: dismal prognosis of t-AML could significantly be mitigated by allo-HSCT in our analysis, with a DFS (56.0%) even higher than previously reported in other series. Survival was also best for patients receiving CPX-351 for induction treatment with more patients obtaining complete remission (26 – 81.0%) and be able to perform transplant (20 – 62.5%).

Clinical Trial Registry: Not Applicable.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P334 OUTCOMES OF SECOND ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR HEMATOLOGIC DISEASES: A SINGLE-CENTER REAL-WORLD EXPERIENCE

Marta Castelli 1, Anna Grassi2, Alessandra Algarotti2, Maria Caterina Micò2, Federico Lussana1, Maria Chiara Finazzi1, Chiara Pavoni2, Gianluca Cavallaro2, Federico Mazzon1, Giuliana Rizzuto2, Benedetta Rambaldi2, Alessandro Rambaldi1

1University of Milan and Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy, 2Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) stands as the primary curative option for various hematological disorders. However, complications like graft failure and post-transplant relapse often require further therapeutic interventions. Despite its uncertain role and the absence of comparative randomized trials, a second HSCT (HSCT2) remains a potential strategy for achieving long-term survival. In this study, we report our single Center experience in performing HSCT2.

Methods: We retrospectively investigated clinical outcomes of patients who underwent HSCT2 in our Centre between September 1998 and September 2023 due to disease relapse or graft failure, either primary or secondary. Endpoints included are engraftment achievement, Overall Survival (OS), Progression-Free Survival (PFS), and Non-Relapse Mortality (NRM). All endpoints were measured from the time of HSCT2.

Results: A total of 61 patients underwent HSCT2, with a median age of 50 years (range: 23 – 69), as shown in Table 1. Forty-five patients (73.8%) had HSCT2 for disease relapse, while 16 (26.2%) for graft failure. Conditioning regimens were myeloablative in 12 patients (19,7%), non-myeloablative in 6 (9,8%) and reduced intensity in 43 (70,5%). In 33 patients (54.1%), a different donor was utilized for HSCT2. Concerning disease status at HSCT2, 36% were in complete remission (CR) and 64% had active disease, including one patient who achieved a partial response. The median time between the first HSCT and HSCT2 was 18.6 months (range: 1.1 – 255.7). Engraftment was achieved in 48 of 61 patients (78.7%), with a median time to engraftment of 16 days (range: 8 - 123). At a median follow-up of 8.36 months (range: 0.2 – 276.3), 18 patients were alive, while 43 died. Twenty-seven patients died for disease recurrence or progression. Among them, 7 out of 27 (26%) were in CR at HSCT2, while 20 out of 27 (74%) had active disease at HSCT2. Sixteen patients died from non-relapse causes such as Graft versus Host Disease (GvHD), infection, and organ toxicity. Overall, the 5-year cumulative incidence of NRM was 22%. The 5-year OS and PFS rates were 29% and 28%, respectively, as shown in Figure 1. Patients undergoing HSCT2 due to graft failure exhibited a 56% survival rate, while those treated for relapse demonstrated a 20% survival rate. Notably, the median follow-up considering alive patients was 5 years (range 0.1-23), providing an extended observation period for those who survived beyond the initial post-transplant period. Univariate analysis showed no significant association between disease status at HSCT2, a different donor from the first HSCT, or relapse within 12 months and outcomes, potentially due to limited subgroup sizes.

Table 1.

Patients’ and transplant main characteristics

61 patients

Median Age, years (range)

50 (23-69)

Diagnosis

Myeloid Malignancies, n° (%)

48 (79%)

Lymphoid Malignancies, n° (%)

9 (15%)

Aplastic Anaemia, n° (%)

4 (6%)

HSCT2 indication

Graft Failure, n° (%)

16 (26,3%)

Relapse, n° (%)

45 (73,7%)

Disease status at HSCT2

CR, n° (%)

22 (36%)

Less than CR, n° (%)

39 (64%)

Donor

Same as HSCT1, n° (%)

28 (45.9%)

Different, n° (%)

33 (54.1%)

Source of HSC

BM, n° (%)

4 (6.5%)

CB, n° (%)

6 (9.8%)

PB, n° (%)

51 (83.6 %)

Figure 1.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (41)

Conclusions: In line with data from literature, our single Center experience confirms that HSCT2 could provide long-term survival in selected patients with high-risk settings, such as post HSCT relapse and graft failure. This is particularly true for patients experiencing graft failure, who have a superior outcome with regards to relapsed patients. However, the NRM rate underscores the critical importance of an accurate patients’ selection. Further studies are warranted to refine patient selection criteria for HSCT2.

Disclosure: Federico Lussana: Amgen: Speakers Bureau, Pfizer Membership on an entity’s Board of Directors or advisory committees and Speakers Bureau, Incyte Speakers Bureau, Bristol Myers Squibb Membership on an entity’s Board of Directors or advisory committees and Speakers Bureau, AbbVie Membership on an entity’s Board of Directors or advisory committees, Clinigen Membership on an entity’s Board of Directors or advisory committees.

Alessandro Rambaldi: Astellas: Honoraria; Pfizer: Honoraria; Agmen: Honoraria; Omeros: Honoraria; Novartis: Honoraria; Kite-Gilead: Honoraria; Jazz: Honoraria; Celgene-BMS: Honoraria; Janssen: Honoraria; Roche: Honoraria; Incyte: Honoraria; ABBVIE: Honoraria.

Marta Castelli, Anna Grassi, Alessandra Algarotti, Maria Caterina Micò, Chiara Pavoni, Maria Chiara Finazzi, Gianluca Cavallaro, Giuliana Rizzuto, Benedetta Rambaldi, Federico Mazzon: nothing to declare.

1: Haematopoietic Stem Cells

P335 PERSPECTIVES OF GENERAL HEMATOLOGISTS ON A PROPOSED SHARED CARE MODEL FOR HSCT IN SAUDI ARABIA FOR SICKLE CELL DISEASE

Abdulrahman Alsultan 1, Mohsen Alzahrani2, Mohammed Essa3, Abdullah Aljefri4, Hatoon Ezzat5, Wasil Jastaniah6

1King Saud University, Riyadh, Saudi Arabia, 2King Abdulaziz Medical City, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia, 3King Abdullah Specialist Children’s Hospital, Ministry of National Guard Health Affairs, Riyadh, Saudi Arabia, 4King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia, 5Ministry of Health, Riyadh, Saudi Arabia, 6King Faisal Specialist Hospital and Research Center, Jeddah, Saudi Arabia

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for sickle cell disease (SCD). In Saudi Arabia, about 90,000 have SCD with 20% eligible for transplant. HSCT centers face a significant burden to meet the demand. The recent approval of gene therapy for SCD will add to this demand. In addition, giving priority to malignant conditions, aplastic anemia, and primary immune deficiency disorders delays access to HSCT for SCD. A shared care model between general hematologists and transplant centers can be implemented to expand access to HSCT (Khera et al. Blood Adv. 2017, Abel et al. Blood 2022, Fein et al. JCO 2022).

Methods: We conducted a survey to understand the perspectives of primary hematologists in Saudi Arabia, who frequently refer SCD patients for HSCT, regarding proposed shared care models and identify opportunities and challenges. The survey covered current resources at local hospitals, pre-transplant and post-transplant care sharing opportunities, and factors that may increase the interest of hematologists to participate in the shared care of HSCT of SCD patients.

Results: A total of 54 adult and pediatric hematologists from different settings, such as academic centers and community hospitals, were contacted, and 33 (61%) responded to the survey. The most frequent indications to refer patients to transplant were stroke (n=27, 82%), recurrent acute chest syndrome (n=22, 67%), and frequent vaso-occlusive crisis despite optimal dosing of hydroxyurea (n=20, 60%). However, nearly half of hematologists will refer patients to HSCT based on patient and family preferences regardless of the disease phenotype (n=18, 55%). Current resources at local hospitals and peri-transplant support potentials are summarized in the Table. Of note, only 16 (48%) can perform HLA typing to identify potential donors prior to referring patients. Most hematologists are willing to support in the pre-transplant phase, such as optimizing hydroxyurea dose, iron chelation, and performing exchange transfusion. However, few (n=7, 21%) are interested in following patients soon after engraftment. In addition, the knowledge of managing transplant-related complications varied among hematologists, as well as access to essential transplant medications. The majority (n=23, 70%) are willing to participate in the long-term care of SCD, such as administering vaccination and survivorship program, and are willing to comply with sharing necessary data to comply with regulatory requirements. The following interventions were believed to increase the interest of hematologists in participating in shared care: well-documented care plan 82%, availability of treatment protocols on the peri-transplant care of SCD 82%, periodic virtual meetings with the transplant center 79%, educating patients and caregivers about the shared care model 70%, availability of 24/7 hotline at transplant center 67%, short-term in-person training at transplant centers 45%, and financial incentives 30%.

Table: Current resources at local hospitals and peri-transplant support potentials by general hematologists

1) Current resources at local hospitals:

HEPA-filtered rooms

Intensive care unit

Trained nurses for central line care

Can perform Cholecystectomy

HLA-Typing

19 (58%)

29 (88%)

24 (73%)

28 (85%)

16 (48%)

Brain MRI/MRA/MRV

Screening for RBC alloimmunization

Leukocyte-depleted irradiated blood products

TDM for cyclosporine, tacrolimus, sirolimus

PCR monitoring for CMV, EBV, adenovirus

29 (88%)

26 (79%)

24 (73%)

11 (33%)

7 (21%)

2) Pre-Transplant support potential:

Counsel patients/families about HSCT

Optimize hydroxyurea dose

Administer intensive iron chelation

Arrange for an exchange transfusion

28 (85%)

33 (100%)

29 (88%)

27 (82%)

3) Post-Transplant support potential:

a. When will you feel comfortable sharing the care of transplanted SCD patients with the transplant team?

After engraftment

After day 100

After six months

After one year

Do not want to follow

7 (21%)

9 (27%)

5 (15%)

1 (3%)

11 (33%)

b. Do you feel comfortable assessing and managing the following transplant-related complications?

Infectious complications

VOD

TA-TMA

Acute GVHD

Chronic GVHD

21 (64%)

16 (48%)

16 (48%)

14 (42%)

14 (42%)

c. Do you have access to the following medications at your hospital?

Cyclosporine, tacrolimus, MMF, sirolimus

Ruxolitinib

Ganciclovir/Foscarnet

Pentamidine

IVIG

17 (52%)

13 (39%)

9 (27%)

14 (42%)

32 (97%)

Conclusions: General hematologists are open to a shared care model for HSCT in SCD, with most referring centers considering pre-transplant support feasible. However, challenges such as limited local resources and varying expertise in managing post-transplant-related complications exist. Well-documented care plans, treatment guidelines, and virtual meetings offer solutions. Collaborative efforts between hematologists and transplant centers could significantly improve access to HSCT for SCD patients in Saudi Arabia.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P336 DOES AGE MATTER? IMPACT OF DONOR AGE IN ENGRAFTMENT AND GRAFT FUNCTION AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION

Oana Enache1, Lavinia Eugenia Lipan2,1, Oana Gabriela Craciun2, Andra Georgiana Stoica2, Adela Ionela Ranete2, Ileana Constantinescu2,1, Alina Daniela Tanase 2,1

1University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania, 2Fundeni Clinical Institute, Bucharest, Romania

Background: Donor age influence on the outcome of hematopoietic stem cell transplant (HSCT) from a related donor (RD) has been of major concern in the last years since the introduction of PtCy and more frequent use of a RD.

Methods: After the approval by the ethics committee, we conducted a retrospective study of 129 patients who underwent HSCT from a RD (mainly first-degree relatives) between 2017 and 2022 in Fundeni Clinical Institute, Bucharest. The patients received peripheral blood stem cell grafts. The primary study endpoints were prevalence of primary graft failure (PGF), secondary graft failure (SGF) and poor graft function, and secondary endpoints were engraftment time and grade of mucositis. Due to the small numbers and the heterogenic distribution on the diseases, we don’t evaluate de PFS/OS.

Results: According to age donors were grouped into three categories: <30 (n=36), between 30-45 (n=59) and >45 years (n=34). Patients were treated mainly for AML (n=56), ALL (n=30) and the rest for other hematological malignancies (n=43), with equal distribution between the three subgroups. Matched-RD were almost equally distributed between age categories with 25 donors <30, 28 donors between 30-45, and 26 donors >45. Haploidentical donors were better represented in the 30-45 years category (n=21), and less in the other two <30 (n=11) and >45 (n=8). Reduced intensity conditioning regimen was preferred among the study population, indifferent of donor-age category with 25, 45 and 27 patients in each category <30, 30-45 and >45 years. Myeloablative conditioning regimen had almost equal distribution between subgroups (n=7 patients, n=11 patients and n=7 patients in first, second and third subgroup).

Median time to neutrophil engraftment was similar for the three donor age subgroups: 18 days for donors <30, 17 days for donors between 30-45 years and 18.5 days for donors >45. Median time to platelet engraftment was 16 days both for patients with a donor <30 and the ones >45 years, and 14 days for patients in the second subgroup. Mucositis grade III/IV was registered in 55.55%, 54.23% and 76.47% of patients in each subgroup (p=0.39 for donor-age <30 vs. 30-45 years, p=0.077 for donor-age <30 vs. >45 years). PGF was experienced by 2 patients in the second subgroup, and SGF by 2 patients in the first subgroup. Poor graft function was seen in 2 patients with a donor-age <30, 6 patients with donor-age 30-45 years (p=0.087) and in 5 patients with a donor age >45 (p=0.012).

Conclusions: In this analysis there is no significant difference in median time to platelet or neutrophil engraftment or mucositis grade, according to donor-age subgroup. Regarding primary and secondary graft failure, there were no statistically significant differences, but there was a higher prevalence of poor graft function in patients who received a graft from donors aged >30 years. Further studies are needed to evaluate the influence and importance of donor-age on poor graft function in the context of a related-donor HSCT.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P337 COMPARABLE RESULTS OF HAPLOIDENTICAL AND MATCHED RELATED HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR PATIENTS WITH BLAST PHASES CHRONIC MYELOID LEUKEMIA

Darina Zammoeva1, Renat Badaev 1, Yuliya Alekseeva1, Dmitriy Motorin2, Elza Lomaia1

1Almazov National Medical Research Centre, Saint Petersburg, Russian Federation, 2Russian Research Institute of Hematology and Transfusiology, Saint Petersburg, Russian Federation

Background: Over the past 20 years, significant advances were made in treatment of chronic myeloid leukemia (CML), but some patients are treatment refractory and progress to blast phase (BP). Allogeneic stem cell transplantation (allo-SCT) still remains the only potentially curative option for patients with BP CML.

Methods: Retrospective analysis included all patients who underwent allo-SCT in our center between 2010 and 2021 years with CML and history of BP according to the European LeukemiaNet criteria. The study was divided into two parts. Firstly, patients were divided according to donor on matched related and haploidentical group. For matched related donor allo-SCT (MRD) myelooblative conditioning was chosen and graft versus host disease (GVHD) prophylaxis was methotrexate, cyclosporine. In haploidentical allo-SCT group (Haplo) reduced intensity conditioning was used, accompanied by GHVD prophylaxis with posttransplant-cyclophosphamide, mycophenolate mofetil, cyclosporine. In case of allo-SCT performed in advanced stages of the disease, reduced intensive conditioning (RIC) regimen with prior FLAG-like cytoreduction was chosen.

Further all patients were divided on groups according to number of chronical phase (CP) or BP at the time of transplantation.

Primary endpoints were overall survival (OS) and relapse incidence (RI). Secondary endpoints were: cumulative incidence of acute GVHD and chronic GVHD, achievement of PCR-negativity.

Results: 25 patients were included in analysis:12 matched related and 13 haploidentical. Two groups were comparable by sex, age, lines of previous therapies and number of BP prior to allo-SCT. Characteristics of patients are presented in Table 1.

Haploidentical SCT (N 13)

Matched related SCT (N 12)

Gender:

p>0,05

Male

7

8

Female

6

4

Age, years

27-58

23-59

p>0,05

Median

39

41,5

MutationaI status:

Mutations found

7 (T3151=3)

3 (T3151=2)

Mutations not found

1

2

Not evaluated

3

6

Patients with >1BC

5

5

p>0,05

Time from Ds to BC, month

39 (4-189)

21 (0-123)

p>0,05

Time from BC to SCT, month

4,1 (0,5-9,9)

3,9 (2,0-94,3)

p>0,05

Lines of TKI therapy before BC

1 (1-3)

1 (1-4)

p>0,05

Conditionig:

p>0,05

Myelooblative

8

Reduced intensity

10

With previous cytoreduction

3

4

Status of disease before SCT:

p>0,05

CP

10

8

BP

3

4

No difference in time to neutrophil engraftment (MRD - 17 (13-32) vs Haplo - 18 (13-66) days, p>0,05) and platelets engraftment (MRD - 17 (13-89) vs Haplo 27 (14-111) days, p>0,05) was observed in two groups. No statistical difference in cumulative incidence of 6-months acute GVHD st II-IV (MRD: 10% vs Haplo: 54%, p=0,15), 6-months acute GVHD st. III-IV (MRD: 0% vs Haplo: 40%, p=0,35) and 1-y chronic GVHD (MRD: 30% vs Haplo: 15%, p>0,05) was observed.

We found significantly higher rate of PCR-negativity achievement by 1-month post Haplo SCT compared to MRD SCT (MRD: 10% vs Haplo: 46%, p=0,03) and comparable results at 6 months (MRD: 50% vs Haplo: 70%, p=0,28). Although most patients in MRD group received myelooblative conditioning and RIC in Haplo group, there was no difference in 6-month relapse incidence (MRD: 37% vs Haplo: 33%, p=0,8) and 1-year OS (MRD: 40% vs Haplo: 41%, p>0,05) (Figure 1).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (42)

Assessment of the effect of disease stage on outcomes of allo-SCT was conducted among all patients. On time of allo-SCT 9 patients were in 2nd CP (2CP), 8 patients in ≥ 3rd CP (3CP) and 7 patients were in active BP. Transplantation in 2CP shows best OS comparing to ≥3CP and BP: 1-y OS in 2CP – 75%, ≥3CP – 15%, BP – 15%; p=0,012. 6-months relapse incidence shows tendency to be lower in 2CP group also: 2CP – 10%, ≥3CP – 45%, BP – 60%; p=0.08.

Conclusions: Haploidentical SCT is a suitable option when MRD is not available for patents with BP CML. 2nd CP is the optimal time for allo-SCT.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P338 ANALYSIS OF THE EFFICACY OF LUSPATERCEPT IN THE TREATMENT OF ANEMIA AFTER ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION

Lei Dong 1, Wei Ma1, Liu Chan-Chan1, Zhang Fang-Fang1, Tian Yue-Feng1, Liu Ji-Cong1, Zhao Xiao-Zhen1, He Yang2, Cao Xing-Yu1

1HeBei YanDa LuDaoPei Hospital, LangFang, China, 2HeBei LuDaoPei Hospital, LangFang, China

Background: Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is a well-established and potentially curative therapy for some malignant and benign hematologic disorders. There is insufficient data on the use of luspatercept after transplantation. Our study aims to evaluate the efficacy and safety of luspatercept in treating post-transplant anemia.

Methods: We conducted a retrospective analysis of the clinical characteristics and outcomes of patients with post-transplant anemia who were treated with luspatercept at our center between January and November 2023.

Results: Ten patients were included in the study, with a median age of 37 years (range:13-67). The group consisted of 3 females and 7 males, The diagnosis included 6 cases of acute myeloid leukemia(AML), 2 cases of acute lymphoblastic leukemia (ALL), 1 case of MDS and 1 case of acute mixed lineage leukemia. The conditioning regimens used were TBI (n=4) or BU (n=6)-based. All patients underwent haploidentical transplantation. Six patients had active GVHD when luspatercept was administered. Two patients has cytomegalovirus and 0 patients has EBV. Plasma human microvirus B19-DNA was negative in all cases. All patients remained in complete remission and the chimerism of CD3-positive cells in peripheral blood and bone marrow were complete donor type. The median peripheral blood EPO levels were 118 uIU/ml (range: 81.9-438), with a reference value of 4.3-29.0uIU/ml. The median hemoglobin level before the application of luspatercept was 59.5g/L(range: 45.4-66.0). The median percentage of red lineage in bone marrow was 37.1% (range: 5-77). The median time for patients start to receive luspatercept treatment was 44.5 days (range: 17-334) post-transplant. Patients received a median one dose (range:1-3) of luspatercept. The efficacy and adverse effects of luspatercept was assessed after 4 weeks. Six patients showed a haemoglobin elevation of ≥20g/L, one case showed a haemoglobin elevation of <20g/L but >10g/L in 1 case, and four cases showed a haemoglobin elevation <10g/L. No patient experienced adverse events of grade ≥2.

Conclusions: The effectiveness of luspatercept in treating anemia after a transplant is promising. However, it is important to note that the small sample size of this study warrants further research with a larger sample size.

Disclosure: There’s no statement. Lei Dong, Wei Ma, Chan-Chan Liu, Fang-fang Zhang, Yue-feng Tian, Ji-cong Liu, Xiao-Zhen Zhao, Yang He, Xing-yu Cao.

1: Haematopoietic Stem Cells

P339 THE IMPACT OF THE TRAVELED DISTANCE TO THE TRANSPLANT CENTER ON THE CLINICAL OUTCOME OF ALLOGENEIC BONE MARROW TRANSPLANT PATIENTS

Jean El-Cheikh 1, Mustafa Saleh1, Khodr Terro1, Radwan Massoud2, Ghassan Bidaoui1, Layal Sharrouf1, Sara Chehayeb1, Toufik Sebai3, David Karam1, Ammar Zahreddine1, Rita Nehme1, Iman Abou Dalle1, Nour Moukalled1, Ali Bazarbachi1

1American University of Beirut Medical Center, Beirut, Lebanon, 2University Medical Center Hamburg Eppendorf, Hamburg, Germany, 3American University of Beirut, Beirut, Lebanon

Background: Numerous factors influence the outcome of Allogeneic Hematopoietic Stem Cell Transplantation (Allo-HSCT) in patients with hematologic malignancies. It is preferable to seek specialized centers with expertise in the intricate procedure as well as its complications. Nevertheless, the clustering of these facilities in Urban areas results in a disparity in healthcare access, necessitating patients to travel greater distances to receive treatment. The influence of travel distance on outcomes HSCT is a topic of ongoing debate. This retrospective study aims to investigate the impact of travel distance on post Allo-HSCT outcomes for adult patients who received treatment at the American University of Beirut Medical Center (AUBMC).

Methods: All adult patients (≥18 years of age) who underwent Allo-HSCT at AUBMC were considered eligible for inclusion in our study. The identified patients were categorized into three groups. The study included 108 Urban patients residing within a 20-kilometer radius of AUBMC. The cohort of patients residing more than 20 kilometers away from AUBMC, denoted as Rural (n=124), also encompasses patients who commute by car from Syria. The third group consists of patients who arrived in Lebanon by airplane (n= 43). Using univariate and multivariate analysis, we investigated the influence of the distance from the transplant center along with other independent variables, on the outcomes of Allo-HSCT.

Urban (˂ 20KM)

Median (range); N (%)*

Rural (˃20 KM)

Median (range); N (%)*

Abroad

Median (range); N (%)*

P-value

Patient / Donor related

Distance in Km

9.0 (1 – 14)

75.0 (21 – 363)

-

0.000

Age at Transplant (years)

47 (18-75)

39 (18-77)

37 (18-71)

0.014

Male sex

70 (65)

79 (64)

34 (79)

0.163

Disease

0.320

Leukemia

86 (37)

106 (46)

38 (17)

Lymphoma

22 (49)

18 (40)

5 (11)

Place of Diagnosis

0.042

AUBMC

57 (45)

56 (44)

13 (10)

Outside AUBMC

51 (34)

68 (46)

30 (20)

Donor Type

0.002

MRD

62 (34)

82 (45)

39 (21)

Haplo

41 (48)

40 (47)

4 (5)

MUD

5 (71)

2 (39)

Transplant related

Median Follow up, months

24 (0-128)

26 (0-147)

75 (1-135)

0.004

Conditioning Regimen

0.300

NMA/ RIC

70 (42)

75 (45)

22 (13)

MA

38 (35)

49 (45)

21 (20)

Results: A cohort of 275 adult patients who underwent Allo-HSCT was identified. Male predominance is seen in all three groups. The distribution of types of diseases was comparable across all three groups. A significant difference in age at transplant was noted (p-value= 0.014), with the Urban group (UG) having the highest (median =47), and the Abroad group (AG) having the lowest (median= 39). The UG had the shortest follow-up duration, whereas the AG had the longest (24 months and 75 months, respectively; p=0.004). Patients who received their stem cells from a MRD were 34% Urban, 45% Rural, and 39% from Abroad. While, both UG and Rural Group (RG) (48% and 47%) respectively received stem cells from a haploidentical donor unlike the AG only 5% (p=0.002). A trend toward statistical significance was noted in overall survival (OS) (p=0.073), disease-free survival (DFS) (p=0.069), and graft-versus-host disease and relapse-free survival (GRFS) (p=0.016) among patients from Urban, Rural, or Abroad groups. The UG exhibited OS, DFS, and GRFS rates of 54.4%, 45.2%, and 32.2%, respectively. In Rural areas, the OS, DFS, and GRFS were comparable at 56.2%, 49.1%, and 41.9% respectively. In the multivariate analysis, the RG exhibited superior DFS (HR: 0.88, 95% CI: 0.55-1.43), OS (HR: 0.93, 95% CI: 0.56-1.53), and GRFS (HR: 0.76, 95% CI: 0.49-1.17) compared to the UG.

Conclusions: Our study provides further evidence to the current body of literature, challenging the notion that the distance between patients and the transplant center has an independent impact on the clinical outcome of their treatment. The lack of a clear correlation between travel distance and clinical outcomes, as evidenced in our study, necessitates a reevaluation of prevailing assumptions regarding the potential influence of geographical distance on the outcomes of Allo-HSCT.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P340 THE CLINICAL ANALYSIS OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN THERAPY-RELATED MYELOID NEOPLASMS

Shu Yan1, Deyan Liu1, Fang Xu 1

1Hebei Yanda Ludaopei Hospital, Langfang, China

Background: Therapy-related myeloid neoplasms (t-MN) are unique myeloid tumors following radiation or cytotoxic therapy, often resistant to chemotherapy and associated with poor prognosis. Earlier studies reported 3-year disease-free survival (DFS) and overall survival (OS) rates of 24-33% and 25-35%, respectively.

Methods: Retrospective review of t-MN patients treated with HSCT at our hospital from January 2019 to October 2023.

Results: There were 21 cases of t-MN, including 5 cases of myelodysplastic syndrome (MDS), 14 cases of acute myeloid leukemia (AML), and 2 cases of mixed leukemia. Among the AML cases, 7 were classified as M5, 2 evolved from MDS, and 1 from CMML. There were 7 males and 14 females with a median age of 36 years (range 4-55 years). The primary tumors included 7 cases of breast cancer, 2 thyroid cancers, 2 liver cancers, and one each of kidney cancer, lung cancer, osteosarcoma, yolk sac tumor of the vagina, intracranial germ cell tumor, primitive neuroectodermal tumor, and embryonal rhabdomyosarcoma. The median interval between the primary tumor and secondary myeloid tumor was 24 months (range 5-96 months). Pre-transplant disease status included 3 partial responses (PR), 9 non-responses (NR), and 9 complete responses (CR). Transplant types were sibling matched in 5 cases and haploidentical in 16 cases. All transplants achieved hematopoietic engraftment, with a median white blood cell engraftment time of +13 days (range 10-20 days) and a median platelet engraftment time of +14 days (range 7-40 days). Post-transplant, 6 cases developed acute graft-versus-host disease (GVHD), 4 of which were grade III-IV. There were 7 cases of leukemia relapse, and 1 case of MRD relapse achieved remission again after treatment. As of the follow-up date of October 22, 2023, 13 out of 21 patients were alive, and 8 had died. The overall survival rates at 1, 2, and 3 year post-transplant were 70.59%, 58.26%, and 58.26%, respectively. The 1, 2, and 3-year overall survival rates for the CR + PR group versus the NR group post-transplant were 82.50% vs 66.67%, 73.33% vs 55.56%, and 73.33% vs 41.67%, respectively, with no significant difference between the two groups (P > 0.05%). Causes of death in 8 patients included 5 from leukemia relapse, 1 from NK cell infusion combined with intestinal, liver and lung GVHD due to MRD positivity, 1 from viral encephalitis, and 1 from infection shock combined with pulmonary alveolar hemorrhage after a second transplant for MDS relapse. 4 patients received maintenance therapy post-transplant, including 3 with azacitidine and 1 with decitabine, all of whom survived without disease.

Conclusions: t-MN has a poor prognosis, making allo-HSCT a preferred treatment option. Despite no significant survival rate differences between remission status groups, the remission group had higher survival rates. The limited case number and follow-up duration necessitate further research with larger samples and extended follow-up.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P341 HAPLOIDENTICAL TRANSPLANTATION FOR PATIENTS WITH FANCONI ANEMIA HAPLOIDENTICAL TRANSPLANTATION FOR PATIENTS WITH FANCONI ANEMIA

Eman Khattab 1, Hasan Hashem1, Mayada Abu Shanap1, Iyad Sultan1, Rawad Rihani1

1King Hussein Cancer Center, Amman, Jordan

Background: Fanconi anemia(FA) is a heterogeneous bone marrow failure disorder. Allogeneic stem cell transplantation(alloHSCT) is the only therapy that can correct the hematological manifestations of FA patients. Haploidentical transplantation has increased the pool of donors.

Methods: Retrospective analysis of FA patients, who underwent haploidentical allogeneic HSCT at King Hussein Cancer Center from January, 2005 until June, 2022.

Results: We are reporting the outcome of haploidentical alloHSCT for 8 FA patients, with overall survival 75%(95% CI: 31.5-93.1).

Five patients underwent upfront T-cell repleted with post-transplant cyclophosphamide(PT-CY). The preparative regimen included Fludarabine and single fraction total body irradiation(TBI), one patient has received cyclophosphamide which was omitted later due to toxicity, rabbit antithymocyte globulin was added to the last 2 patients, All patients received calcineurin inhibitors and Mycophenolate mofetil starting from day 5 post-transplant. The timing of immunosuppressants was adjusted for the last patient from day zero and 1 post-transplant, respectively.

Three patients were rescued after primary graft failure, one after related donor transplant using PT-CY, two patients after unrelated cord blood transplantation, Alemtuzumab in the stem cells bag was used for T-cell depletion.

The median time for neutrophil engraftment was 13 days(9-18), and 14 days(11-93)for platelet engraftment. Seven patients developed acute GVHD, six developed CMV reactivation, six patients developed chronic GVHD, and three patients are still receiving immunosuppressants more than 4 years post-transplant.

We report 3 deaths, one due to chemotherapy toxicity before engraftment, the second patient died 8 months post-transplant due to extensive chronic GVHD, and viral reactivation, and the third death was in the rescue group due to squamous cell carcinoma 5 years post-transplant.

Conclusions: T-cell repleted haploidentical donor transplantation with PT-CY is feasible for FA patients without a matched related, with increased risk of post-transplant complications, and requires extensive supportive care. Modification of the timing of r-ATG and the GVHD prophylaxis may decrease the toxicity.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P342 NK-CELL INFUSIONS TO PREVENT RELAPSE FOLLOWING HEMATOPOIETIC STEM CELL TRANSPLANTATION

Maryia Navumovich1, Tatsiana Shman1, Yuliya Mareika1, Volha Mishkova1, Katsiaryna Vashkevich1, Natalia Kirsanava1, Iryna Pakhomova1, Viktoryia Belabokava1, Dmitriy Prudnikov1, Lubov Zherko1, Nina Minakovskaya 1, Anzhalika Solntsava1

1Belarusian Research Center for Pediatric Oncology, Hematology and Immunology, Borovliany, Belarus

Background: One of the main treatment options for patients with high-risk hematological malignancies is allogeneic hematopoietic stem cell transplantation (HSCT). Significant progress has been made in reducing treatment-related mortality (TRM). Still, the relapse rate after transplantation remains virtually unchanged and, according to the literature, is 10-60% for patients with high-risk acute leukemia. To some extent, this is because T-cell recovery in most patients occurs 6-12 months after HSCT. We hypothesize that introducing allogeneic natural killer (NK) cells in the early post-transplantation period can increase the antileukemic effect of the graft and reduce the incidence of relapses after HSCT.

Methods: In 2023, in our Center, three patients (boys, median age 10.2 years) with hematological malignancies (acute myeloblastic leukemia (AML), myelodysplastic syndrome (MDS), mixed phenotype acute leukemia (MPAL)) underwent a myeloablative conditioning regimen allogeneic related (one compatible and two haploidentical) HSCT. The source of hematopoietic stem cells (HSC) was peripheral blood (PBSC) in 2 cases and bone marrow (BM) in 1 case.

Prevention of graft versus host disease (GvHD) was based on cyclosporine A (CSA) and post-transplant cyclophosphamide (ptCY) with mycophenolate mofetil (MMF) after haploidentical HSCT. Because the children were at high risk of relapse after allogeneic HSCT (blasts in the peripheral blood or high levels of minimal residual disease (MRD) at the time of HSCT), they received NK cells on days +7-12 (Table 1).

NK cells were obtained by expansion using feeder lines K-562-4-1BBL-mIL21 from HSC donors. Cell culture purity is 99%, the average dose of NK cells is 8x10*6 cells/kg, and the average dose of T cells is 0.01x10*6/kg.

Patient 1

Patient 2

Patient 3

Age

15,24

10,24

1,52

Diagnosis

MDS

MPAL

AML

Genetic markers

Monosomy 7

CBL, PTPN11

CBFb\MYH11

Blast cells before conditioning (%)

12.25

3

1.5 (MRD 0.45% FCM)

Conditioning

FLU 160 THIO 10 MEL 140 ATG 15

TBI 12 Gy VP 60

ATG 15

FLU 160 THIO 10 MEL 140 ATG 15

Donor/source

MSD\PBSC

MMFD\PBSC

MMFD\BM

GvHD prevention

CSA

CSA, ptCY, MMF

CSA, ptCY, MMF

Days of infusion

+10

+12

+7, +10

NK cell dose, х10^6\kg

10

10

10, 2.6

MRD, day +30

Not determined

0,01

0,07

Donor chimerism (STR) in BM and PB on day +30 (%)

100

100

100

Granulocytes > 0.5х10^9/l

+14

+12

+16

Platelets > 50х10^9\l

+16

+17

+34

Acute GvHD

No

Grade II (skin 3, GI 2)

Grade II (skin 3, GI 2)

Treatment

No

CSA, MMF, medrol, ruxolitinib, budenofalk, MSC transplantation

CSA, MMF, medrol

Chronic GvHD

No

Skin

No

Treatment

No

Ruxolitinib, medrol, ECP

No

Outcome

Alive in remission

Alive in remission

Alive in remission

Duration of observation

312

314

63

  1. FCM-flow cytometry, CBL – cord blood leukemia, PTPN11 - protein tyrosine phosphatase non-receptor type 11, CBFB – core binding factor beta, MYH - myosin heavy chain, FLU – fludarabine, THIO – thiotepa, MEL – melphalan, ATG – thymoglobulin, TBI – total body irradiation, Gy – gray, VP – Etoposide, MSD – matched sibling donor, MMFD – mismatched family donor, PB – peripheral blood, STR - short tandem repeat, GI- gastrointestinal, MSC – mesenchymal stem cells, ECP – extracorporeal photopheresis,

Results: All patients are alive and in remission (median follow-up 312 days).

The 1st patient did not have acute or chronic GVHD or significant infectious and toxic complications.

Two patients developed grade II acute GVHD (skin – 3, gastrointestinal – 2), which was successfully treated. One of these patients developed chronic GVHD (skin), which was treated. The second patient was diagnosed with CMV viremia.

None of the observations showed any adverse reactions to administering NK cells.

Conclusions: NK cell immunotherapy may be a practical addition to standard HSCT to reduce the risk of disease relapse in the early post-transplant period without increasing the risk of severe life-threatening acute GVHD. Further studies are needed in a larger group of patients for further evaluation.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P343 SURVIVAL OF PATIENTS WITH MULTIPLE MYELOMA WITH ASCT IN ONCOSALUD - AUNA BETWEEN 2015 AND 2021

Alonso Diaz1, Claudio J. Flores1, Shirley Quintana 2, Cesar Samanez2

1AUNA-IDEAS, Lima, Peru, 2Oncosalud - AUNA, Lima, Peru

Background: ASCT was introduced as a consolidation approach in multiple myeloma more than two decades ago and has been shown to achieve significant improvement in survival. Here we present the impact of autologous ASCT on the overall survival of patients with MM who received treatment in a private clinic (Lima, Peru).

Methods: We analyzed the medical records of 143 patients with multiple myeloma treated at Oncosalud-AUNA between 2015 and 2021. The clinical characteristics of the patients were compiled from electronic medical records. The MM stage was determined according to the Salmon Durie Staging System and the International Staging System (ISS). Overall survival (OS) was evaluated using the Kaplan-Meier method, and comparisons of survival curves were performed using the Logrank or Breslow test.

Results: The median age at diagnosis was 67 years (range: 37–94), 71.3% were over 60 years of age, and 49.0% were male. MM was IgA type in 37 (28.2%) patients, IgG in 86 (65.7%), and light chains in 8 (6.1%). The stage of MM, according to Salmon-Durie, was generally stage III (89%), and ISS was stage I in 35%, II in 29%, and III in 36% of the patients.

First-line treatment was generally VRD (bortezomib, lenalidomide, and dexamethasone) in 34.1% of patients and VTD (bortezomib, thalidomide, and dexamethasone) in 22.2%. 71.3% of patients received bortezomib-based treatment. The pre-HSCT response was CR in 29.6%, very good PR in 15.2%, PR in 8.8%, and NR in 14.4%. 26.6% of patients had ASCT, and 73.4% did not have ASCT.

The median follow-up was 4.4 years, and 35% of patients had died, reaching a median OS of 5.9 years (95% CI: 4.2 to 7.7). The 4- and 5-year OS rates were 66.8% and 59.8%; 91.4% in patients with ASCT, 58.3%, and 50.3% in patients without ASCT, presenting a significant difference (p < 0.001).

In patients younger than 65 years, the 4- and 5-year OS rates in ASCT (n = 33) were 93.4% and in non-ASCT (n = 27) were 41.9% and 27.9%, respectively, presenting a significant difference (p < 0.001), in which age, sex, Salmon-Durie, and ISS did not present a difference (p > 0.05) between both groups.

Conclusions: In our series, patients with MM who received ASCT had a better survival rate than those who did not receive it; these findings are similar to those reported in other series.

Disclosure: None.

1: Haematopoietic Stem Cells

P344 RESOLUTION OF VOD/SOS WITH DEFIBROTIDE AFTER HSCT AND PERSISTENT HEPATIC DEMAGE: MONOCENTRIC PEDIATRIC EXPERIENCE

Fabiana Cacace1, Emanuela Rossitti2, Valeria Caprioli1, Flavia Antonucci2, Maria Rosaria D’amico1, Giuseppina De Simone1, Maria Simona Sabbatino1, Francesco Paolo Tambaro 1

1AORN Santobono Pausilipon, Naples, Italy, 2AOU Federico II, Naples, Italy

Background: Veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) is a potentially life-threatening complication of hematopoietic stem cell transplantation (HSCT). Defibrotide is the only medication approved for the management of severe VOD/SOS after HSCT.

Methods: We retrospectively reviewed data from 135 HSCT procedures performed in our department from June 2017 to November 2023. Each case of VOD/SOS was diagnosed using pediatric EBMT criteria, along with a severity assessment. All the VOD/SOS patients were monitored by abdomen ultrasound, liver function tests (LFT) and blood coagulation parameters.

Results: Among 135 HSCT analyzed, 9 patients (7 males, 2 females) were diagnosed with VOD/SOS. Cumulative incidence in our center was 6,6% (9/135). The median age of VOD/SOS onset was 9,8 years (range 0.6 to 19 years). VOD occurred in 1 patient diagnosed with neuroblastoma following an autologous HSCT. Less than half of VOD/SOS patients met the criteria of severe VOD (3/9, 33%), only one patient was very severe, according to the severity classification. Concomitant endothelial complications were acute GVHD, acute GVHD and TA-TMA and acute GVHD, TA-TAM and PRES in 2, 1 and 1 patient, respectively. The median time of VOD onset after HSCT was 21 days (range 7 to 41 days). Transfusion-refractory thrombocytopenia and the unexplained weight gain in three consecutive days (despite use of diuretics) were the consistent symptoms of VOD/SOS (observed in 89% of cases), followed by hepatomegaly and ascites (observed in 78% and 55% of cases, respectively). All patients were treated with 25mg/kg/day defibrotide at VOD/SOS diagnosis. The median duration of treatment was 28 days (range 10 to 60 days) and the discontinuation was based on clinical resolution, while coagulation parameters, hepatomegaly were still persistent. The defibrotide response rate was 78% (7/9 patients). Instead, two patients died less than a month after VOD/SOS diagnosis: cause of death was multiorgan failure. Among patients with VOD/SOS resolution the most common causes of death were infection (2 patients) and hematological relapse (1 patient). The overall survival (OS) and 100-day transplant-related mortality (TRM) were 55% and 22%, respectively. LFT, coagulation parameters and abdomen ultrasound were performed to evaluate liver damage. While AST and ALT did not present significant alterations, D-dimer and von Willebrand factor were above their threshold limit until six months from VOD/SOS diagnosis. All patients showed hepatomegaly at the abdomen ultrasound.

Conclusions: VOD/SOS is a severe complication of HSCT. Defibrotide is the only treatment available; longer survival was observed in patients who presented resolution of VOD/SOS. In our cohort, LFT did not undergo alterations except when other complications (e.g. GVHD or iatrogenic toxicity) occurred. An increase in D-dimer and von Willebrand factor as sign of “inflammatory status” have been observed six months after resolution of VOD/SOS while hepatomegaly persisted as sign of hepatic damage. These results should be confirmed by larger cohort of patients in multicenter prospective studies. Correlation between imaging such as ultrasound and MRI and duration of treatment with defibrotide need further investigations.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P345 ACCURATE DIETARY EVALUATION AND ADEQUATE NUTRITIONAL THERAPY ARE INDISPENSABLE FOR BETTER OUTCOMES OF ALLO-HSCT

Yuki Mori 1, Koichi Nakase1, Toshimi Noma1, Tomoya Katsuta1, Ryuichiro Hiyama1, Yuki Goto1, Ryo Ueda1, Risa Hashida1, Kyoko Itakusu1, Kyosuke Saeki1, Masakazu Mori1, Yuichiro Nawa1

1Ehime Prefectural Central Hospital, Matsuyama, Japan

Background: The importance of nutritional intake for allogeneic hematopoietic stem cell transplantation (allo-HSCT)recipients, especially the role to ensure total calories, protein, and fat, has not been well understood. In our hospital, a registered dietitian has evaluated and calculated accurate nutritional intake including oral, nasogastric, and intravenous administration of allo-HSCT patients on a daily basis. Based on these data, we retrospectively analyzed the effect of nutrition therapies for outcomes of allo-HSCT recipients.

Methods: Sixty-seven patients who underwent their first allo-HSCT between June 2019 and April 2023 were analyzed. Background diseases of 67 patients were as follows: acute myeloid leukemia (n=26), acute lymphocytic leukemia (n=8), myelodysplastic syndromes (n=20), adult T-cell leukemia/lymphoma (n=6), aplastic anemia (n=2), and lymphoma (n=5). Pre-transplant disease status was in CR for 36 patients (53.7%) and non-CR for 31 patients (46.3%). Adequate nutritional therapy was defined as total energy intake meets basal energy expenditure (BEE) x 1.3, protein 1.0-1.5 g/kg body weight, and fat 20-30% of BEE.

Age, height, body weight, pre-transplant KPS, performance status, HCT-CI, Gastrointestinal acute GVHD, pre-transplant disease status (CR or non-CR), intensity of conditioning regimen, nutrition (BEE x 1.3), protein (weight x 1.0g), and fat (BEE × 20%) were analyzed. We analyzed nutritional data from day 0 to day 42. Nutritional parameters were treated as a time-dependent covariate converted from the date of transplantation and analyzed. Statistical software R was used for statistical analyses.

Results: Nutritional intake of BEEx1.3 or more (HR 0.41, 95%CI 0.18-0.93 P=0.034), sufficient protein (HR 0.36, 95%CI 0.17-0.76 P=0.007), and sufficient fat (HR 0.37, 95% CI 0.15-0.94 P=0.036) were the significant factors for better overall survival.

In addition to nutrition, age and disease status before transplantation were other factors that were associated with overall survival in this analysis.

Conclusions: The impact of pre-transplant malnutrition on clinical outcomes after allo-HSCT has been reported. Our study demonstrates the importance of providing adequate nutrition to improve the outcome of allo-HSCT patients. In addition to total energy intake, sufficient protein and fat intake are also essential. Our study is important in that it indicates the importance of daily intake of protein and fat, as well as energy levels, during hospitalization for allo-HSCT.

Disclosure: Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P346 ALLOGENOUS BONE MARROW TRANSPLANT IN HIGH-RISK CHRONIC MYELOID LEUKEMIA IN THE ERA TKI INHIBITOR: A SYSTEMATIC REVIEW

Nia Novianti Siregar 1, Resti Mulya Sari1, Andree Kurniawan2, Felix Wijovi2, Devina Adella Halim2, Patricia Angel2, Rivaldo Steven Heriyanto2

1Dharmais National Cancer Centre Hospital, Jakarta Barat, Indonesia, 2Pelita Harapan University, Tangerang, Indonesia

Background: To this day, bone marrow transplant remains the chosen curative option for Chronic Myeloid Leukemia (CML) that are resistant to Tyrosine Kinase Inhibitors (TKI), especially those that are considered to be in the high-risk part of the disease.

Methods: Data will be gathered from PubMed, PMC, Science Direct, and Scopus. Inclusion criteria were prospective randomized trials and non-randomized studies, that assessed transplant outcome of high-risk CML patients in era of TKI treatment from various centers. The included study’s quality was assessed using the Jadad scale.

Results: A total of 5 studies that include 688 CML patients were included. Across all 5 studies the most common indication for transplant was TKI treatment failure or resistance. After changing the curative option to transplant, favorable outcomes were observed with high survival rates, compared to TKI treatment. This results gives light to CML patients especially high risk patients that did not respond to TKI treatment, and can even be considered to those with suboptimal response. Assessment using Jadad scale yields good results for all studies.

Conclusions: Allogenous bone marrow transplant is a promising alternative for CML patients that is resistant to TKI treatments, providing higher survival and lower relapse mortality.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P347 LONELINESS AND RUMINATIONS IN CANCER PATIENT-CAREGIVER DYADS – ANALYSIS OF DAILY FLUCTUATIONS

Małgorzata Sobczyk-Kruszelnicka1, Aleksandra Kroemeke2

1Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland, 2Institute of Psychology, SWPS University, Warsow, Poland

Background: Several studies have found positive links between loneliness and symptoms of depression, anxiety, or distress. One of the factors responsible for these associations is ruminative thought. However, little is known about the mechanisms linking loneliness to different types of ruminations. The study aimed to investigate whether fluctuations in daily loneliness, reported each evening after hospital discharge following hematopoietic cell transplantation (HCT), are associated with fluctuations in passive and active ruminations (i.e., intrusive, “negative” and more reflective, “positive”, respectively) and co-ruminations (i.e., disclosing problems and speculating about them during conversation) in patient-caregiver dyads.

Methods: The sample consisted of 200 patient-caregiver dyads following the first autologous or allogeneic HCT. Participants made independent, daily reports of loneliness, passive and active ruminations, and co-ruminations for 28 consecutive days.

Results: Participants reported higher levels of all types of ruminations on days when they felt more lonely. Co-rumination had a beneficial effect: it alleviated the negative effects of feeling lonely by activating “positive” ruminations.

Conclusions: The results indicated a potential beneficial role of disclosing and discussing one’s problems with a close one in the patient-caregiver dyads after HCT. The results may be useful for future intervention studies on ruminative thoughts in recovery.

Disclosure: none.

1: Haematopoietic Stem Cells

P348 PATIENTS WITH FOLLICULAR LYMPHOMA AND CANDIDATES FOR AUTOLOGOUS TRANSPLANTATION: IS THE COLLECTION OF HEMATOPOIETIC PROGENITORS EFFICIENT AFTER THE FIRST LINE OF TREATMENT?

María Casado Sánchez1, Sara Hormaza de Jauregui1, Juan José Mateos Mazón1, Javier Arzuaga Méndez1, Xabier Martín Martitegui1, Paula María Zoco Gallardo 1, Laura Aranguren del Castillo1, Ariadna García Ascacibar1, Gorka Pinedo Martín1, María Elena Amutio Díez1, Juan Carlos García Ruiz1

1Hospital Universitario Cruces, Baracaldo, Spain

Background: First-line treatment based on R-CHOP and rituximab maintenance gives patients with follicular lymphoma a median progression-free survival (PFS) of 10 years according to current data from the PRIMA trial. When these patients relapse, even with the discovery of new therapies, the gold standard remains the consolidation of 2nd or subsequent complete response with high-dose QT and autologous hematopoietic stem cell transplantation (ASCT).

In order to avoid compromising the collection of hematopoietic progenitors in a subsequent line of treatment, our center proposes the performance of hematopoietic progenitor apheresis (HPA) after the first line of treatment in patients who are candidates for ASCT.

Methods: Retrospective study performed at Hospital Universitario Cruces, Barakaldo, Spain.

Data in the period from 01/01/2010 to 12/31/2021 on all patients with follicular lymphoma and under 65 years old in whom HPA has been performed after first-line chemotherapy with R-CHOP are collected.

The rate of ASCT in 2nd line or later in our cohort of patients is evaluated.

Results: A total of 48 patients under 65 years old diagnosed with follicular lymphoma and eligible for transplantation underwent HPA.

With a median follow-up of 60 months, the cumulative incidence of ASCT is 15.54% (95% CI 3.0% to 26.2%).

Currently, 81.25% (39) of the afferized patients have not undergone ASCT: because they have not relapsed to date (34), because they were not considered candidates at the time of relapse (2), because of death prior to transplantation (2) or because they opted for another line of treatment at relapse (1).

Of the patients who have not currently relapsed (34), 4 of them are no longer, due to age, candidates for ASCT. However, 25 (73.5%) have not reached the median PFS yet, according to the PRIMA study, and could therefore be potential candidates.

Conclusions: The results obtained in our study suggest a priori limited efficiency of performing HPA on patients with follicular lymphoma after the first line of treatment.

If we take into account the prolonged PFS of those patients treated with R-CHOP and rituximab maintenance, a feasible approach could be based on a correct selection of the candidate patient for this strategy, lowering the age limit for HPA.

However, a key point to consider in our approach will be to determine whether the current setting of new therapies will be able to displace the indication for ASCT.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P349 HAPLOIDENTICAL BONE MARROW TRANSPLANTATION IN A COLOMBIAN BMT UNIT: CLINICAL DESCRIPTION AND OUTCOMES

Jorge Cuervo sierra 1, Germán A. Velasquez-Quintero1, Juan Felipe Jaramillo-Alvarez1, Wang Nataly Montoya-Arbelaez1, Sandra Gisella Martinez-Jimenez1, Luz Maridier Tobon-Tobon1, Jurany Andrea Marin-Montoya1, Deisy Johana Giraldo-Gomez1, Luisa Fernanda Castro-Quintero1, Maira Alejandra Mesa-Giraldo1, Maribel Londoño-Buitrago1

1Clinica Somer, Rionegro, Colombia

Background: Bone marrow transplants presents significant delays in developing countries. The need to find an ideal donor can complicate the clinical situation of refractory/relapsed patients who need urgent BMT to obtain complete disease remission.

Haploidentical BMT allows to solve these problem and helps to achieve earlier and safe transplants.

Methods: We retrospectively analyzed all haploidentical bone marrow transplants in adults in a Colombian BMT unit during a six year period. Fifty three (n:53) patients received haploidentical BMT between January 2017 and December 2022.

Results: The median age of patients was 35 (range 17 to 59). The 69% were female (n:37) and 31% male (n: 16). The main indications for BMT were: acute leukemia 39 (AML 13, ALL 24 and biphenotypic 2), Lymphomas 6 (HL 3 and NHL 3), Mycosis Fungoides 2, Chronic myeloid leukemia 2, Aplastic anemia 3, Blastic Plasmacytoid Dendrytic Cell Neoplasm 2 and Myelodisplasic syndrome 1. The status at transplantation were: complete remission (n:38), partial remission (n:5), and active disease (n: 9). The median number of CD34 infused was 7,293 x 106/kg (range 4,325 to 10). All transplants were made using stem cells from peripheral blood after mobilization with GCSF. The conditioning regimen was myeloablative in 16 patients and RIC/NMA in 37. The most commonly employed conditioning regimen was Busulfan- Cyclophosphamide-Fludarabine (BuCyFlu) with or without total body radiation (TBI). Graft-versus-host disease prophylaxis was cyclosporine, mycophenolate mofetil and posttransplant high dose cyclophosphamide (PTCy) based. Seventy nine percent (79%) of patients had at least one episode of neutropenic fever. Of these 43,3% developed bacterial and 5,66% viral infections. SARS CoV2 represented 3,77%.

Citomegalovirus reactivation was found in 28 patients(52,8%) and was managed with gancyclovir/valgancyclovir.

Acute graft-versus-host disease (aGVHD) was documented in 16 patients (30,2%). Of these 11 patients had skin involvement, 7 gastrointestinal tract and 8 hepatic.

Chronic graft-versus-host disease was observed in 17 of 53 patients (32%). The median time for myeloid engraftment was 16 days (range 13 to 24).

Median following time was 461 days (maximum following time 2155 days). Sixteen patients had relapse of disease after transplant (32,0%).

Twenty one patients died (39,6%). The Non Relapse Mortality (NRM) was 17% (9 patients). Median survival was not achieved (95% confidence interval, 333 days - not achieved).

There was a trend towards lower mortality in patients who developed aGVHD requiring systemic management (Log rank test p=0.181), and there was a lower tendency to relapse in patients with aGVHD requiring systemic management (Log rank test p= 0.231). Relapse-free survival was 2155 days (confidence interval between 1023 days and not reached).

Conclusions: The activity of haploidentical BMT in developing countries remains an important option for the treatment of patients without a compatible related donor and presents an acceptable morbidity and mortality compared to more developed countries. The clinical outcomes were similar to that described at international studies.

Disclosure: Nothing to disclosure.

1: Haematopoietic Stem Cells

P350 HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN FANCONI ANEMIA: A SINGLE CENTER EXPERIENCE

Guzmán López de Hontanar 1,2,3, June Iriondo2,3, Josune Zubicaray2,3, Elena Sebastián2,3, Blanca Molina2,3, Sara Vinagre2,3, Marta González-Vicent2,3, Julián Sevilla2,3

1Hospital Universitario Príncipe de Asturias, Alcalá de Henares, Spain, 2Hospital Infantil Universitario Niño Jesús, Madrid, Spain, 3Fundación de Investigación Biomédica del Hospital Infantil Universitario Niño Jesús, Madrid, Spain

Background: Allogeneic hematopoietic stem cell transplantation is the standard treatment for rescuing bone marrow failure in Fanconi Anemia (FA). Historically, the best results have been obtained with an HLA-matched related donor. However, advances in conditioning regimes and graft-versus-host disease (GVHD) prophylaxis have increased the number of alternative-donor transplants, including haploidentical stem cell transplant. In the context of FA, effective results have been achieved with both post-transplant cyclophosphamide prophylaxis and CD19 + /TCRαβ+ graft depletion. Since a significant percentage of patients do not have an HLA-matched related donor, this option could be an effective alternative. We report our experience with haploidentical CD19 + /TCRαβ+ depleted stem cell transplantation in FA.

Methods: Between February 2020 and September 2022 two children aged 7 and 9 years old without matched-related donor received an haploidentical CD19 + /TCRαβ+ depleted transplant at our center. FA was confirmed through molecular diagnosis and chromosome breakage assay in both patients. Donor CD34+ hematopoietic progenitors were mobilized by subcutaneous administration of granulocyte colony-stimulation factors. Apheresis was performed with a Spectra Optia Cell Separator (Terumo BCT, Leuven, Belgium). Graft manipulation procedures were performed with a CliniMACS device (Miltenyi Biotec, Bergisch Gladbach, Germany). Conditioning regimen included 300cGy single dose total body irradiation on day -6, intravenous fludarabine 35mg/m2/24h and cyclophosphamide 10mg/kg/24h days -5 to -2. GVHD prophylaxis included cyclosporine 3mg/kg/24h initiated on day -3; rabbit antithymocyte globulin 5mg/kg/24h days -5 to -1. Antimicrobial prophylaxis included acyclovir, trimethoprim/sulfamethoxazole and micafungin, continuing after discharge with oral fluconazole.

Results: Conditioning was well tolerated in both patients. The first patient, a 7-year-old girl, experienced a capillary-leak syndrome successfully treated with methylprednisolone and tocilizumab, and a sinusoidal obstruction syndrome managed with defibrotide. The second patient, a 9-year-old girl, suffered a typhlitis treated with cefepime for 13 days until clinical resolution. Sustained primary engraftment was achieved in both patients, with time to neutrophil recovery of 9 days for both patients and platelet recovery of 8 and 11 days. None had acute or chronic GVHD. Monitoring of donor-recipient chimerism confirmed stable engraftment of donor hematopoiesis in both patients. The 7-year-old girl experienced an adenovirus infection, solved after cidofovir treatment, intravenous immunoglobulins, and CD45RA-depleted donor lymphocyte infusion. At a median follow up of 2.4 years both patients maintain normal blood counts.

Conclusions: While an HLA-matched related donor stem cell transplant continues to be the optimal treatment for bone marrow failure in FA, our results, in correlation with previous reports, confirm that haploidentical CD19 + /TCRαβ+ depleted stem cell transplant is a feasible and effective treatment in FA. GVHD rates were low, but viral infections represented a significant challenge and required substantial support including donor lymphocyte infusion. Nevertheless, toxicities are manageable in an experienced center and this modality represents a treatment opportunity in patients lacking an HLA-matched related donor.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P351 EARLY AUTOLOGOUS AND/OR ALLOGENEIC STEM CELL TRANSPLANTATION FOR ADULT PATIENTS WITH ADVANCED STAGE T- LYMPHOBLASTIC LEUKEMIA/LYMPHOMA OR BURKITT LYMPHOMA. A RETROSPECTIVE SINGLE-CENTRE ANALYSIS

Normann Steiner 1, Katherina Baier1, Denise Ritter1, Gabriele Hetzenauer1, Jakob Rudzki1, Stefan Köck1, Eberhard Gunsilius1, Brigitte Kircher1, Dominik Wolf1, David Nachbaur1

1University Hospital of Internal Medicine V (Hematology and Oncology), Medical University of Innsbruck, Innsbruck, Austria

Background: T-cell acute lymphoblastic leukemia/lymphoma (T-ALL/LBL) and Burkitt lymphoma (BL) are uncommon, highly aggressive diseases originating either from immature precursor T cells or from mature B cells in BL.

Methods: We retrospectively analyzed the outcome of an early autologous and/or allogeneic stem cell transplantation (SCT) concept in 28 patients with advanced stage T-ALL/LBL and BL after three to four remission induction/consolidation chemotherapy cycles.

Results: Considering only patients in first complete remission (CR), the 5-year overall survival (OS) and event-free survival (EFS) was 91% in patients with BL and 73% in patients with T-ALL/LBL with a 5-year relapse incidence (RI) of 9% in patients with BL and 27% in patients with T-ALL/LBL. All relapsing patients finally succumbed to the disease (n=10) or complications/toxicity after having received a salvage allogeneic transplant (n=5).

Conclusions: Despite the low patient number our retrospective single-centre analysis by incorporating an early intensive high-dose chemo-/radiotherapy strategy with either autologous or allogeneic stem cell transplantation, although preliminary, show promising long-term outcome comparable to conventional chemotherapy treatment strategies. Further studies are highly warranted to better define those patients who might benefit most from such a treatment approach.

Disclosure: No conflict of interest.

1: Haematopoietic Stem Cells

P352 MONOCYTES IN ALLO-HSCT WITH AGED DONORS SECRET IL-1/IL-6/TNF TO INCREASE THE RISK OF GVHD AND DAMAGE THE AGED HSCS

Xia Li 1, Wanying Zhang2, Yanan Wang2, Chentao Li2, Yibo Wu1, Xiaoyu Lai1, Yi Luo1, Pengxu Qian1, He Huang1

1Bone Marrow Transplantation Center, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Zhejiang University-University of Edinburgh Institute, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, China

Background: Aging has been considered a critical factor causing poor prognosis of allogenic hemopoietic stem cell transplantation (allo-HSCT), such as poor overall survival (OS) and high incidence of GVHD. However, the mechanisms underlying this phenomenon are still unknown.

Methods: To elucidate the underlying mechanisms, we comprehensively analyzed the effect of donor aging on poor outcomes by reintegrating our clinical data from patients after allo-HSCT and public single-cell RNA-seq data obtained from patients’HSC niche post-allo-HSCT. Moreover, we further analyzed the single-cell RNA-seq data of healthy individuals, focusing on HSCs, the HSC niche and their interactions during aging.

Results: Our clinical results demonstrated that old donors were more prone to acute GVHD (aGVHD), a life-threatening complication, limiting the overall survival (OS) of patients with allo-HSCT. Also, a pronounced accumulation of inflammation was observed in the bone marrow of patients treated with allo-HSCT. Our analysis revealed the presence of inflammation-related CXCL2 + HSC subpopulation during aging, characterized by the enriched interleukin-17 (IL-17), TNF, NF-κB, and MAPK signaling pathways, emphasizing the role of inflammation during aging. Shifting attention to the HSC microenvironment, CD14 monocytes and CD16 monocytes play pivotal role in inflammation accumulation and GVHD risk during aging. Specifically, we deciphered that IL-1/IL-6 and TRAIL (i.e., TNFSF10) ligand‒receptor pair serves as the crucial bridge between CD14/CD16 monocytes and hematopoietic stem/progenitor cells (HSPCs). The profound upregulation of these signaling during aging finally causes HSC dysfunction and lineage-biased differentiation.

Conclusions: Our findings provide the theoretical basis for achieving precise management of GVHD and enhancing the efficacy of allo-HSCT regimens for ageddonors.

Disclosure: The authors declare that they have no competing interests. This work was supported by grants from the National Natural Science Foundation of China (81900176 and 82130003), the Zhejiang Provincial Key Research andDevelopment Program (2021C03010), Sanming Project of Medicine in Shenzhen(SZSM202111004).

1: Haematopoietic Stem Cells

P353 SETTING UP THE FIRST PEDIATRIC BONE MARROW TRANSPLANTATION UNIT IN THE UAE AND THE EXPERIENCE OF COMPLETING THE FIRST 30 PEDIATRIC BMTS WITHOUT ANY MORTALITY

Zainul Aabideen 1, Sagar Mohan Nivargi1, Abdullah Odat1, Mohammed Alfar1, Charbel Khalil1, Rakeshkumar Shah1, Kesava Ramakrishnan1, Rajesh Phatak2, Ahmed Elsheikh1, Mohamed Abdelsamad1, Krishnappa Venkatesh1, Ghulam Mujtaba1, Dr. Georgey Koshy1, Amro El Saddik3, Humaid Alshamsi4, Fulvio Porta5, Lawrence Faulkner6, Mohammed Hamid1

1Burjeel Medical City, Abu Dhabi, United Arab Emirates, 2Burjeel Hospital-AbuDhabi, Abu Dhabi, United Arab Emirates, 3Burjeel Holdings, Abu Dhabi, United Arab Emirates, 4Burjeel Hospital For Advanced Surgery, Abu Dhabi, United Arab Emirates, 5Burjeel Holding, Abu Dhabi, United Arab Emirates, 6Cure 2 children foundation, Florence Area, Italy

Background: Hematopoietic stem cell transplantation (HSCT) is a lifesaving and curative treatment option for various medical conditions in children. HSCT was not available in the UAE in children till March 2022 when the first ever paediatric HSCT was done in Abu Dhabi. Objective of this study is to describe experience and outcome of HSCT in children at our centre.

Methods: We prospectively collected data of all children who underwent HSCT at our centre between March 2022 to July 2023.

Results: We performed 24 allogenic HSCT and 6 autologous HSCT over a period of 18 months from March 2022 to September 2023 with no mortality either related to transplant or related to relapse/progression of disease. The indications for allogeneic HSCT included Thalassemia (6/24), Sickle Cell Anemia (2/24), Pure Red Cell Aplasia (1/24), Sideroblastic Anemia (1/24), Congenital amegakaryocytic thrombocytopenia (1/24), Severe Combined Immunodeficiency Disease (n=5/24), Hemophagocytic Lymphohistocytic Histiocytosis (3/24), Chronic Granulomatous Disease (1/24), Wiskott Aldrich Syndrome (1/24), Acute Lymphoblastic Leukemia (n=2/24) and Acute Myeloid Leukemia (1/24). The autologous stem cell transplantation was carried out for neuroblastoma (5/6), Hodgkin’s Lymphoma (1/6). Out of twenty-four allogeneic HSCT, 17 were matched related donors while 7 were haploidentical related donor.

The median age was 3.9 years (0.3 – 14.5 years) for the cohort including allogeneic & autologous HSCT recipients. Bone marrow was the source of Stem Cells for all allogenic BMT patients. The mean time to neutrophil engraftment and platelet engraftment in allogeneic HSCT was 17.96 days and 13.65 days respectively (range 9 – 30 days & 6 – 30 days respectively). The mean time to neutrophil engraftment and platelet engraftment in autologous HSCT was 12.33 days and 13.66 days respectively (range 9 – 15 days & 12 – 15 days respectively).

We report no major complication in the cohort during hospital stay except two. Acute GvHD (aGvHD) limited to grade II skin was noted in 16.6% (4/24) patients. Grade I & grade IV gut GvHD was seen in 4% (1/24) patients each. Chronic skin GvHD was noted in 16.6% (4/24),. Thus, GvHD free survival rate is 91.6%.

The median follow-up duration is 261 days (range 60 – 600 days). At median follow up, we have 4% each of primary & secondary rejection (1/24), 75% full donor chimerism (18/24) and 16.6% mixed chimerism (4/24). Thus, we report a 100% overall survival (OS) & 93.33% disease free survival at 1 year of HSCT. As mentioned above our center has GvHD free survival rate of 91.66%. We also report 100% relapse free survival in HSCT performed for malignant disorders.

Conclusions: Our centre has made remarkable progress in developing paediatric allogeneic HSCT unit in the region. Our results are comparable to international standards. There has been no transplant related mortality so far. Paediatric HSCT is feasible in this country, and this would benefit the paediatric population in this country who were otherwise travelling abroad for HSCT, which is associated with obvious economic and logistic burden and hampers continuity of care which is challenging in these patients who undergo HSCT abroad.

Clinical Trial Registry: not part of trial.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P354 POST-TRANSPLANT CYCLOPHOSPHAMIDE ± ANTI-THYMOCYTE GLOBULIN IN PATIENTS WITH AML/MDN UNDERGOING ALLOGENEIC HCT WITH PBSC FROM HAPLOIDENTICAL DONOR: A SINGLE-CENTER EXPERIENCE

Francesca Cavallaro 1, Marta Canzi1,2, Fabio Serpenti1, Maria Cecilia Goldaniga1, Kordelia Barbullushi1, Giulia Galassi3, Francesco Onida4,2, Francesco Passamonti1,2, Giorgia Natascia Saporiti1

1Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy, 2Università degli Studi di Milano, Milano, Italy, 3ASST Santi Paolo e Carlo, Milano, Italy, 4ASST Fatebenefratelli-Sacco, Milano, Italy

Background: Post-transplant cyclophosphamide (pTCy) and anti-thymocyte globulin (ATG) are substantial and effective agents utilized as prophylaxis against graft-versus-host disease (GvHD). The combination of these drugs appears to be synergic in preventing GvHD, but data on strategies combining pCTy and ATG in patients undergoing haploidentical hematopoietic cell transplantation (Haplo-HCT) are limited. We hereby report the outcomes of patients undergoing Haplo-HCT who received pTCy with or without ATG at our Institution.

Methods: We retrospectively analyzed clinical data and outcomes of patients with acute myeloid leukemia (AML) or myelodysplastic neoplasms (MDN) who underwent Haplo-HCT and received GvHD prophylaxis with pTCy +/- ATG at our Center between July 2016 and August 2023. Overall (OS) and progression-free survivals (PFS) were analysed using the KM estimator and the log-rank test was used to assess differences between groups.

Results: As of November 2023, 29 patients underwent Haplo-HCT at our Institution for AML (n= 26) and MDS (n= 3). Table 1 shows patients and transplant characteristics. Median age at transplant was 61 years (range 29-74). Disease status at transplant was complete remission in 21 cases (72%), and active disease in 8 (28%). Stem cell source was peripheral blood in all but two patients. According to the transplant conditioning intensity (TCI) score, a low-intermediate score was used in 9/17 in the ATG group (53%) and 4/12 in the non-ATG-group (33%). GvHD prophylaxis consisted of MMF, pTCy (50 mg/kg, days +3, +4) and CsA in all patients but one, who received sirolimus. Seventeen patients (58%) received ATG at the dose of 2.5 mg/kg (days -3, -2), mostly after 2020 (15/17, 88%). No G3-4 aGvHD was observed and G1-2 GvHD occurred d in 6/17 patients who received ATG (35%) and in 6/12 patients who didn’t (50%), p=0.42. Chronic GVHD developed in 6 patients of the ATG group (35%, moderate/severe in 3) and 4 in the non-ATG group (33%, moderate/severe in 1), p=0,91. Patients experiencing CMV and EBV reactivation were 4 (23,5%) and 1 (6%) in the ATG-group and in 3 (25%) and 0 in the non-ATG-group, respectively. With a median follow-up of 22.6 months (19.3 in patients who received ATG and 43.0 in patients who didn’t), 2-year OS and PFS scores were respectively of 61% and 57% in the ATG-group and 81% and 78% in the non-ATG-group. Ten patients relapsed (10/29, 35%), 7/17 in ATG-group (41%) and 3/12 in non-ATG-group (25%). Ten patients died, 9 of whom from disease progression and one from transplant-related causes (in the non-ATG-group).

Table 1. Patient and transplant characteristics

All patients (N=29)

ATG-group (N=17)

No-ATG-group (N=12)

Sex, N (%)

Male / female

17 (59) / 12 (41)

11 (65) / 6 (35)

6 (50) / 6 (50)

Age at transplant, years, median (range)

61 (29-74)

65 (49-74)

59 (29-68)

Number of previous lines, median (range)

2 (0-7)

1 (0-3)

2 (0-5)

Diagnosis, N (%)

Acute myeloid leukemia

26 (89)

15 (88)

11 (92)

Myelodysplastic neoplasm

3 (11)

2 (12)

1 (8)

Disease status at transplant, N (%)

Complete response

23 (79)

13 (76)

10 (83)

Active disease

6 (21)

4 (24)

2 (17)

Donor age, years, median (range)

31 (18-61)

29 (22-56)

33 (18-61)

CMV status donor / recipient, N (%)

Positive / positive

14 (48)

9 (53)

6 (50)

Positive /negative

/

/

/

Negative /negative

10 (35)

3 (17)

4 (33)

Negative / positive

5 (17)

5 (29)

2 (17)

Conditioning, N (%)

TCI low-intermediate

13 (45)

9 (53)

4 (33)

TCI high

16 (55)

8 (47)

8 (67)

CD34+ infused, x10^6/kg, median (range)

5.2 (4.36-6.57)

5,2 (4,36-6,08)

5,15 (4,6-6,57)

Neutrophil engraftment, days from transplant, median (range)

20 (14-33)

18 (15-22)

21 (14-33)

Conclusions: In our experience, the addition of ATG to the classical haploidentical pTCy/CSA/MMF platform didn’t lead to a significant reduction of both acute and chronic GVHD nor to an increase of viral infectious complications. Even though not reaching statistical significance due to the limited sample size, we couldn’t rule out a negative impact on the PFS, possibly due to an increased relapse incidence in the ATG-group. In the haplo-HCT setting, a multicenter randomized prospective trial comparing GvHD prophylaxis strategies including ATG vs no-ATG is therefore warranted.

Disclosure: Nothing to declare.

1: Haematopoietic Stem Cells

P355 DESCRIPTION OF PATIENTS WITH RELAPSED/REFRACTORY HODGKIN LYMPHOMA WHO UNDERWENT TO ALLOGENEIC STEM CELL TRANSPLANTATION IN A REFERENCE CENTER IN BOGOTA, COLOMBIA

Carlos Fernando Gómez Calcetero 1, Angie Paola Guarin Castañeda1, Oscar Javier Peña Ardila1, Gloria Elena Mora Figueroa1, Cesar Lamadrid Sastre1, Enrique Pedraza Mesa1, Gerson Menoyo Caballero1, Gustavo Adolfo Martinez Salazar1, Licet Villamizar Gómez1

1Clinica de Marly SA, Bogota, Colombia

Background: Allogeneic stem cell transplantation (allo-SCT) may be a curative option for a group of patients who suffer from refractory or relapsed Hodgkin lymphoma (R/R LH) or in relapse after Autologous stem-cell transplantation (auto-SCT). To our knowledge, there are no Latin American series that include this specific group of patients.

Methods: An observational and analytical case-control study is proposed, nested in a retrospective cohort of patients with R/R HL who underwent Allo-SCT. In addition to describing the cohort, it was analyzed whether there is an association between the type of donor and overall mortality, the cumulative incidence of relapse, and the cumulative incidence of acute graft-versus-host disease (aGVHD).

Results: 17 patients were included, in 8 a match related donor (MRD) was used, in 8 an haploidentical donor and in one patient a match unrelated donor (MUR) was used. All received reduced intensity conditioning (RIC), the most used was Bu-Flu-Cy. GVHD prophylaxis was performed with post-transplant cyclophosphamide associated with mycophenolate mofetil and Tacrolimus. At one year, non-relapse mortality (NRM), overall survival (OS), and progression-free survival (PFS) were 11.7%, 83%, and 93.7%, respectively. The 1-year overall survival rate in MRD and haploidentical donor was 1.18 (95% CI 0.59-2.38), and 1.26 (95% CI 0.57-2.81) respectively. The 1-year progression/relapse-free survival rate in MRD and haploidentical donor was 1.04 (95% CI 0.50-2.18) and 1.26 (95% CI 0.57-2.18). 2.81) respectively. The mortality rate not associated with relapse at day +100 in MRD and haploidentical donor was 0 and 4.19 (95% CI 0.59-29.74) respectively. The cumulative incidence rate of acute GVHD at day +100 in MRD and haploidentical donor was 8.96 (95% CI 2.24-35.81) and 9.71 (95% CI 2.43 – 38 0.82) respectively.

Outcome

Overall

n= 17

MRD

n=8

MUD

n=1

Haploidentical donor

n=8

Wilcoxon

test p*

1-year overall survival rate

Event by patient - year (CI 95%)

1,20 (0,72-1,99)

1,18 (0,59-2,38)

1,00 (0,14-7,10)

1,26 (0,57-2,81)

0,778

1-year progression/relapse-free survival rate

Event by patient - year (CI 95%)

1,12 (0,66-1,89)

1,04 (0,50-2,18)

1,00 (0,14-7,10)

1,26 (0,57-2,81)

0,885

Mortality rate not associated with relapse at day +100

Event by patient - year (CI 95%)

1,86 (0,26-13,18)

4,19 (0,59-29,74)

0,778

Cumulative incidence rate of acute GVHD at day +100

Event by patient - year (CI 95%)

11,33 (4,72-27,22)

8,96 (2,24-35,81)

83,33 (11,74-591,6)

9,71 (2,43 – 38,82)

1,000

  1. MRD=match related donor
  2. MUD= match unrelated donor
  3. CI 95%= 95% confidence interval
  4. *= the log-rank test with Wilcoxon adjustment was used given the sample size.

Conclusions: In patients with relapsed/refractory Hodgkin lymphoma who relapse after autologous stem-cell transplantation (auto-SCT), allogeneic stem cell transplantation (allo-SCT) is a curative, safe and accessible option in Colombia. In our series we did not find differences between the different types of donor used, this is probably due to the use of post-transplant cyclophosphamide as prophylaxis for GVHD.

Disclosure: Nothing to declare.

23: Haemoglobinopathy

P356 TREOSULFAN-BASED HAPLOIDENTICAL T CELL DEPLETED HSCT FOR ADOLESCENTS AND YOUNG ADULTS (AYA) WITH ADVANCED STAGE SICKLE CELL DISEASE AND THALASSEMIA

Anja Troeger 1, Juergen Foell1, Katharina Kleinschmidt1, Gina Penkivech1, Tarek Hanafee-Alali1, Andreas Brosig1, Robert Offner1, Daniel Wolff1, Selim Corbacioglu1

1University Hospital Regensburg, Regensburg, Germany

Background: Inherited hemoglobinopathies, such as sickle cell disease (SCD) and transfusion dependent thalassemia (TDT) are associated with considerable morbidity and mortality. Availability of a matched sibling donor (MSD) bone marrow transplant as standard of care curative treatment is limited. Therefore, we explored T cell depleted haploidentical stem cell transplantation (T-haplo-HSCT) as alternative treatment to further reduce the risk of acute and chronic graft-versus-host disease (GvHD) in SCD and TDT.

Methods: 29 patients (pts) with advanced stage SCD, SCD-ß-Thal or TDT (median age: 18 years; range: 11-32) received a T-haplo-HSCT (CD3+/CD19+; n=16 or aß/CD19+ depleted; n=13) in Regensburg/Germany between July 2012 and July 2023. Indications for HSCT in SCD pts (n=22) included severe or moderate SCD complications such as recurrent pain crisis (>5/year), acute chest syndrome, neurological events, osteonecrosis, nephropathy, and transfusion-refractory allo-immunization; and in TDT severe transfusion-dependent iron overload with complications (n=7). In SCD only, conditioning was preceded by an exchange transfusion. SCD and TDT pts were conditioned with a regimen consisting of ATG-Grafalon (45mg/kg), treosulfan (42g/m2), thiotepa (10mg/kg) and fludarabine (160mg/m2). Immunosuppression (IST) with a calcineurin inhibitor (mainly tacrolimus; with target levels between 5ng/ml and 8ng/ml) and MMF, was maintained for a minimum of 180 days, based on chimerism and split-chimerism analyses.

Results: Pts. received a median of 10.3 x 106 T cell depleted CD34+ cells/kg (range: 4.8–29). Neutrophil engraftment was achieved after a median of 18 days. With a median follow-up was 70 months (range 5-137 mo), the overall survival (OS) was 93%. Two pts (one SCD and one TDT) needed a second haplo-HSCT due to primary graft failure and achieved disease-free survival.

Median chimerism off-IST was 99% (range 73.4–100%). T cell recovery with T cell counts >200/µl was achieved in all but two pt at a median of 152 days (range 34-516 d), with one pt requiring regulatory T cell infusions and one developing fatal viral infection. No severe cases of ≥°III aGvHD or extended cGvHD were observed. Viral reactivation occurred in 63% of pts. Two SCD pts died of HHV6-associated complications accompanied by hyperinflammation, macrophage activation and acute respiratory distress despite antiviral and anti-inflammatory treatment. One pt developed kidney failure after prolonged BKV reactivation. All remaining patients achieved transfusion independence in TDT and cessation of SCD-related complications. Meanwhile, one female patient gave birth to a healthy child. Overall, the treosulfan-based conditioning regimen was well tolerated with two cases of moderate VOD/SOS and only one case of CNS infarction in a SCD pt being observed in this high-risk population. 1/19 pts developed PRES early after conditioning while on immunosuppression with CSA and MMF.

RESULTS

T-haplo HSCT

OS (%); EFS (%)

93; 86

Follow-up (months); median (range)

70 (25-137)

Neutrophil engraftment (days); median (range)

18 (11-41)

Chimerism (%); median (range)

99 (74.3-100)

Immune reconstitution >200/µl CD3 + /µl (days); median (range)

152 (34-516)

Sepsis/ severe bacterial infection (outcome)

34% (resolved with antibiotic treatment)

Viral infections/reactivation (outcome)

62% viral reactivation; (1 kidney failure (BKV); 2 fatal HHV6-associated complications)

Conclusions: The safety and efficacy data of this high-risk AYA patient population with advanced stage hemoglobinopathies reveal that outcome with a T-haplo-HSCT can even exceed that of a MSD-HSCT with regard to a disease-free, GvHD free overall survival (EFS). However, diligent infectious and viral monitoring and early, effective treatment of viral reactivation, in particular HHV6 and CMV is mandatory in SCD pts.

Disclosure: DW: Received research support from Novartis and honoraria from Novartis, Mallinckrodt, Incyte, Sanofi, Behring, Neovii and Takeda.

23: Haemoglobinopathy

P357 COMPARISON OF THE UNRELATED TRANSPLANTATION RESULTS OF AN HLA FULLY MATCHED DONOR AND A HOMOZYGOUS MM DONOR AT ONE LOCUS IN PATIENTS WITH BETA THALASSEMIA

Koray Yalçın 1,2, Safiye Suna Çelen1, Seda Öztürkmen3, Hayriye Daloğlu4, Suleimen Zhumatayev5, Vedat Uygun6, Gülsün Karasu5, Akif Yeşilipek3

1Bahcesehir University, Istanbul, Turkey, 2Acıbadem University, Istanbul, Turkey, 3MedicalPark Antalya Hospital, Antalya, Turkey, 4Antalya Bilim University, Antalya, Turkey, 5MedicalPark Göztepe Hospital, Istanbul, Turkey, 6Istinye University, Istanbul, Turkey

Background: For patients with beta thalassemia, hematopoetic stem cell transplantation (HSCT) is presently the only curative therapy option. Results from transplants using HLA fully matched donors are considered very encouraging, however the impact of HLA homozygosity at 1 mismatch (MM) on the outcome is not yet clear. In this study, we aimed to compare the transplantation results of fully matched unrelated donors with the results of unrelated HLA homozygous MM at one locus in patients for whom a HLA fully matched donor could not be found.

Methods: The study included 149 children with beta thalassemia major who received hematopoietic stem cell transplantation (HSCT) from a matched unrelated donor between December 2012 to December 2022 in Medicalpark Antalya and Istanbul Goztepe Hospitals Pediatric HSCT Units. Data was obtained retrospectively from registry. All unrelated donors were screened for HLA with high-resolution typing and homozygous donor is defined as 1-MM in the graft-versus-host (GVH) direction for a heterozygous recipient. Thalassemia free survival (TFS), cGvHD free survival (TGFS), and overall survival (OS) were evaluated to compare the outcomes.

Results: Median follow-up was 57 (range 12-133) months. Regarding HLA matching, 127 of the patients had HSCT from donors who were fully HLA matched and 22 patients from donors who were homozygous MM for one antigen locus. Mismatched locus was in HLA A in 20 patients and HLA C in 2 patients.

Conclusions: Through allogeneic hematopoietic stem cell transplantation, many children worldwide have been cured of beta thalassemia. For many patients, however, the requirement to undergo HSCT from a donor who is 10/10 HLA compatible poses a barrier. A homozygous MM donor is accepted as 9/10 matched for graft versus host (GVH) and 10/10 for host-versus-graft (HVG) direction. However, among the patients in the MM group in our study, there was no statistical difference in cGVHD. Using donors homozygous for MM at one locus, we observed that the HSCT results were comparable to the transplant outcomes using donors that were 10/10 matched.

In conclusion, results from our study indicate that donors who are homozygous for MM at the HLA A and C loci have comparable HSCT outcomes as 10/10 matched donors. Prospective studies with large patient groups are needed to determine preferred MM loci and their efficacy and safety.

Disclosure: Nothing to declare.

23: Haemoglobinopathy

P358 CLINICAL OUTCOMES AND HEALTHCARE RESOURCE USE IN PATIENTS WITH SICKLE CELL DISEASE WITH RECURRENT VASO-OCCLUSIVE CRISES WHO RECEIVED HEMATOPOIETIC STEM CELL TRANSPLANTS IN FRANCE

Jessica Baldwin 1, Chuka Udeze1, Naxin Li1, Lyes Boulmerka1, Lila Dahal1, Giancarlo Pesce2, Nadia Quignot2, Heng Jiang2, Frederic Galactéros3

1Vertex Pharmaceuticals, Boston, United States, 2Certara France, Paris, France, 3Sickle Cell Referral Center, Internal Medicine Unit, Henri Mondor Hospital, Paris, France

Background: Sickle cell disease (SCD) is characterized by abnormal sickle hemoglobin that causes vaso-occlusive crises (VOCs), hemolysis, and end-organ damage. Allogeneic hematopoietic stem cell transplant (HSCT) is currently available as a curative option for some patients. There is limited research on the outcomes in patients with SCD who undergo HSCT in Europe. This study describes the clinical outcomes and healthcare resource use (HCRU) for patients with SCD with recurrent VOCs who received HSCT in France.

Methods: This longitudinal, retrospective cohort study utilized the French National Health Data System database (SNDS, Système National Des Données De Santé). Patients with SCD were identified based on an inpatient claim or registration in the long-term condition database (ALD, Affection Longue Durée) with a diagnosis of SCD between January 1, 2012, and March 1, 2019. Patients with SCD with recurrent VOCs were required to have ≥2 VOCs per year in any 2 consecutive years after the first qualifying SCD claim. Eligible patients who had evidence of hereditary persistence of fetal hemoglobin (HPFH) in their medical record were excluded. Patients with SCD with recurrent VOCs who received HSCT were identified based on relevant procedure codes. The index date was the date of transplant. Patients were followed from the index date until death, or end of study period (March 1, 2020). Demographics were assessed at the index date. Clinical complications and HCRU were summarized in the post-index period. An analysis of the broader SCD patient population who received HSCT was also conducted.

Results: Overall, 29,771 patients with SCD were identified and 182 underwent HSCT and met additional inclusion criteria. Of the 29,771 patients with SCD, a total of 4,813 patients met the criteria for SCD with recurrent VOCs and 91 of these patients underwent HSCT and met additional inclusion criteria. For patients with SCD with recurrent VOCs (n=91), mean age at the transplant was 14.8 years, and 44.0% (n=40) were female. A substantial proportion of patients who underwent HSCT experienced transplant-related complications (33/91; 36.3%) following transplant, including graft versus host disease (GvHD) (28/91; 30.8%). Mean length of stay for the index HSCT admission was 58.4 days. In the 1 year following transplant 114.8 total days were spent in the hospital on average, with 89 of those days occurring in the first 100 days after transplant.

Outcomes in the overall SCD cohort who received HSCT (n=182) were consistent with those with recurrent VOCs who received HSCT, including proportion of patients developing GvHD (63/182; 34.6%) and the mean total days in the hospital during the first 100 days and 1 year after transplant (100 days: 84.0 days; 1-year: 110.4 days).

Conclusions: Only a small proportion of patients with SCD or SCD with recurrent VOCs undergo HSCT. Many patients who undergo transplants experience clinical complications related to HSCT including GvHD, which can be serious and potentially life threatening, highlighting the need for additional curative therapies for these patients.

Disclosure: Baldwin, Jessica: Vertex Pharmaceuticals employee.

Udeze, Chuka: Vertex Pharmaceuticals employee.

Li, Nanxin: Vertex Pharmaceuticals employee.

Boulmerka, Lyes: Grants or contracts and stock options at Vertex and Amgen.

Dahal, Lila: Vertex Pharmaceuticals employee.

Pesce, Giancarlo: Full-time employee of Certara with stock options and Certara received consulting fees from Vertex Pharmaceuticals.

Quignot, Nadia: Full-time employee of Certara with stock options and Certara received consulting fees from Vertex Pharmaceuticals.

Jiang, Heng: Full-time employee of Certara with stock options and Certara received consulting fees from Vertex Pharmaceuticals.

Galactéros, Frederic: Nothing to declare.

23: Haemoglobinopathy

P359 SINGLE-CENTER EXPERIENCE OF STEM CELL TRANSPLANTATION IN PATIENTS WITH SEVERE SICKLE CELL DISEASE

Miriam P. Klahr 1, Amer Assal1, Christian Gordillo1, Ran Reshef1, Monica Bhatia1, Diane George1, Markus Y. Mapara1

1Columbia University Irving Medical Center, New York, United States

Background: Allogeneic hematopoietic stem cell transplant (HSCT) is currently considered to be the only curative treatment option for patients with severe SCD. However, gene therapy approaches appear promising and have recently received FDA approval. Here, we present our institutional experience using alternative donors (AD) in comparison to patients with matched related donors (MRD).

Methods: Eligibility for HSCT included frequent vaso-occlusive crises and evidence of end-organ damage. Non-myeloablative conditioning with alemtuzumab and 3 Gy total body irradiation (TBI) was used for MRD HSCT (n=12). AD consisted of matched unrelated, mismatched unrelated, or haploidentical donors. Patients undergoing AD transplants (n=10) were treated with either an in-house IRB-approved research protocol using CD34 selection (Alemtuzumab [54 mg/m2], fludarabine [180 mg/m2], and melphalan [140 mg/m2]) (n=7) or a standardized protocol with Alemtuzumab, TBI and post-transplant Cyclophosphamide (PTCy) (n=3). All allo-patients received G-CSF mobilized peripheral blood. All patients underwent RBC exchange to achieve Hgb S <30% before the start of conditioning. Data is reported using n (%) or median (range).

Results: Median follow-up was 50.3 months (9.2-107.7) for MRD recipients and 57.1 months (4.7 - 85.1) for AD. Median age in years was 29 (21-42) for MRD recipients and 26 (18-44) for AD recipients. The cohorts included four patients on hemodialysis (HD) (MRD n=1, PTCy = 3), and the MRD group included one patient with a renal allograft from the same donor. 2/4 of patients on HD underwent a successful cadaveric donor kidney transplant following the allogeneic stem cell transplant. All patients were homozygous for hemoglobin S except one with hemoglobin Sβo-thalassemia in the MRD group and another heterozygous for hemoglobin S and C in the non-MRD group. All patients engrafted with no cases of graft failure. GVHD occurred in 4/12 MRD patients and 3/10 AD patients. All GVHD cases were steroid-responsive and resolved. Four patients in the non-MRD group developed PRES. Overall survival was 100% in MRD vs 90% (9/10) in AD patients. One patient on the CD34-selected protocol died at 554 days due to sepsis. The mean length of stay (LOS) for transplant admission was not different between MRD (35 days [27 – 71]) and AD recipients (42 days [36 -70]). The number of hospital readmissions per patient within the first year of transplant showed marked variation, with a median of 2 (0-5) readmissions per patient in the MRD group and 3 (1-9) for AD. We studied markers of hemolysis (LDH, haptoglobin, absolute Reticulocyte count, and indirect bilirubin) post-transplant as a surrogate marker for clinical efficacy. We observed a decrease in all hemolysis markers at six months and one-year post-transplant.

Conclusions: Alternative donor transplants are feasible even in patients with impaired organ function (e.g., ESRD) but are associated with more transplant-related complications (e.g., PRES, longer LOS) compared to MRD transplants.

Disclosure: Markus, Mapara - Consulting: Ossiumhealtth, Incyte, BluebirdBio, CRISPR/Vertex.

Amer, Assal - Consulting: Gamida cell, Guidepoint INC; Honoraria: Sanofi.

23: Haemoglobinopathy

P360 HAEMATOPOIETIC STEM CELL TRANSPLANTATION IN A LOW RESOURCE COUNTRY NIGERIA. A 10 YEARS REPORT FROM A SINGLE CENTER

Nosakahare Bazuaye 1, Nancy Ojiemhangbe2

1Igbinedion University Teaching Hospital, Benin, Nigeria, 2Celltek Healthcare Medical Center, Benin, Nigeria

Background: Nigeria has one of the highest incidence of sickle cell disease in the world (2-3% of a population of over 200 million). Also common are other non –malignant and malignant diseases that require HSCT. Activities of HSCT is low in Africa/East Mediterranean accounting for only 3.3% of global HSCT (over 1.5 million in 2017). Despites documented challenges, the first successful HSCT in Nigeria was performed by Bazuaye et al in 2011 at the Federal University of Benin Teaching Hospital (UBTH) and thereafter at a public private partnership at celltek healthcare medical center Nigeria. This is a report from 2011 to 2022 from a single transplant team in Nigeria.

Methods: Sixteen of the patients with Sickle cell disease had HLA matched sibling donors (12/12 match), six were Haplo transplant (HLA > 6/12) and stem cell source used was bone marrow in nineteen patients and peripheral stem cells in three patients for those with ABO mismatch. The transplant protocol for matched sibling donor consists of reduced intensity conditioning (RIC) with Anti-thymocyte globulin (ATG), Fludarabine and oral Busulphan. Immunosuppression was with cyclosporine and Mycophenolate Mofetil. Haplo protocol was with Fludarabin, dexamethasone, Busulphan and post-transplant day +3 and +4 cyclosphosphamide. Immunosuppression was with Tarcolimus and Mycophenolate Mofetil (MMF). All patients with multiple Myeloma had autologous peripheral stem cell transplantation.

Results: Total HSCT was twenty seven, twenty one (77.8%) with Sickle cell disease, five (18.5%) with Multiple myeloma and one (3.7%) with Aplastic Anaemia. Transplant activities in UBTH was 3 (11%) while in public private collaboration with celltek healthcare medical center it was 24 (89%) The average age for patients with sickle cells disease was 12.1 years (04 – 19 years), Multiple Myeloma 52 years (45-59 years) and Aplastic Anaemia 25yrs. Donors Hb phynotype for sickle cell disease patients was HbAS (38%) and HbAA (62%). Neutrophil and platelet engraftments occurred within 12 – 19 days (mean 15.2days) and 14 – 23 days (mean 19.1 days) respectively. Overall survival (OS) was 22 (81 %) and 2(7.4%) rejection with high persistent fetal haemoglobin and has been stable with clinical cure.

Conclusions: There is need for collaboration of Private public partnership in Nigeria to increase the number of HSCT centers. Haplo HSCT may become a possible source of donors and the use of single peripheral non cryopreserved stem cell dose in place of double tandem has made it possible for many developing countries to perform autologous HSCT for Multiple Myeloma patients. Overall survival is comparable to data from other centers.

Disclosure: No disclosure and no conflict of interest to declare.

23: Haemoglobinopathy

P361 IMPACT OF THE FIRST ONLINE WEBINAR OF THE WBMT NURSING GROUP IN COLLABORATION WITH THE LAGOS UNIVERSITY TEACHING HOSPITAL AND SICKLE CELL FOUNDATION OF NIGERIA

Adetola Kassim 1, Reggie Belkhedim2, Sebastian Galeano3, Dietger Niederwieser3, Damiano Rondelli4, Abijah Princy B5, Amala Lucas6, Ainy Azra7, Cristina Vogel8, Diana Villalobos9, Eugenia Arjona10, Felicia Michael11, Francisca Negrete12, Gloria Ceballo13, John Murray14, Jorge Morales15, Josephine Suganya6, Kishwar Sultana16, Komal Mundhe17, Latha Gracelin6, Lu Yin18, Megan Hogg11, Michelle Kenyon19, Niti Lotey20, Nouf Shwikan21, Paola Llamas22, Rita Nehme23, Sandeep Elizabeth24, Suman Kubal17, Trish Joyce25, Yvonne Panek-Hudson25, Adriana Seber26, Annette Akinsete27, Edamisan Temiye28, Oluseye Akinstete28, Mahmoud Aljurf21

1Vanderbilt University Medical Center, Nashville, United States, 2Eastern Mediterranean Blood and Marrow Transplantation, Saudi Arabia, Saudi Arabia, 3Worldwide Network for Blood and Marrow Transplantation WBMT, Liebefeld, Switzerland, 4College of Medicine, University of Illinois at Chicago -UI Health, Chicago, United States, 5Christian Medical College, Velore, India, 6Christian Medical College, Vellore, India, 7Akbar Niazi Teaching Hospital, Islamabad, Pakistan, 8Latin American Bone Marrow Transplant Nursing Group, Sao Paulo, Brazil, 9Hospital Doctor Luis Calvo Mackenna, Santiago, Chile, 10Enfermera Hospital U y P LA FE, La Fe, Spain, 11Westmead Hospital, New South Wales, Sydney, Australia, Sydney, Australia, 12Clínica Universidad de los Andes, Santiago, Chile, 13Hospital del Niño Doctor Jose Renan Esquivel, Panama, Esquivel, Panama, 14The Christie NHS Foundation Trust, London, United Kingdom, 15Doctor Luis Calvo Mackenna Hospital, Santiago, Chile, 16AF Bone Marrow Transplant Centre, Rawalpindi, Pakistan, Rawalpindi, Pakistan, 17Advanced Centre for Treatment, Research and Education in Cancer (ACTREC), Tata Memorial Centre, Mumbai, India, 18The first affiliated hospital of Soochow University, China, Suzhou, China, 19Kings College Hospital, London, United Kingdom, 20Alberta Health Services, Canada, Alberta, Canada, 21King Faisal Specialist Hospital & Research Centre, Jeddah, Saudi Arabia, 22Dr Luis Calvo Mackenna Hospital Santiago, Providencia, Región Metropolitana, Santiago, Chile, 23American University of Beirut, Beirut, Lebanon, 24Sankalp India Foundation, Bangalore, India, 25Peter MacCallum Cancer Centre, Victoria, Australia, 26Pediatric Hematopoietic Cell Transplantation, Hospital Samaritano Higienopolis - Americas, Sao Paulo, Brazil, 27Sickle Cell Foundation of Nigeria, Lagos, Nigeria, 28Paediatric Haematology/Oncology, Lagos University Teaching Hospital, Idi-Araba, Lagos, Nigeria

Background: The Worldwide Network for Blood and Marrow Transplantation is a non-governmental (NGO) umbrella organization in the field of hematopoietic stem cell transplant (HSCT) and, in collaboration with the World Health Organization (WHO), has taken up the challenge of collecting and disseminating global HSCT activity data regularly. Information on indications, the use of different technologies, donor types, and trends over time provide a sound basis for physicians and nurses to provide appropriate patient counseling and for healthcare agencies to develop the necessary infrastructure. Informed by global activity survey data, the WBMT performs worldwide workshops to support the development of new HSCT programs and to optimize existing programs. The ability to share accumulated experience covering a wide range of strategies, successes, and pitfalls continues to be vital in improving global HSCT access for needy patients. The Nigeria BMT nurses’ online workshop was a 2-day online comprehensive program designed to cover the basics and current approaches to managing BMT patients at the pre-transplant, transplant, and post-transplant stages. The goal was to provide a overview and education in HSCT geared toward low-middle-income countries without access to expensive educational programs organized by experts whose populations have poor access to HSCT.

Methods: The free online Webinar was approved and hosted by the leadership and ethics committee of the WBMT, Lagos University Teaching Hospital, and Sickle Cell Foundation of Nigeria. This 2-day workshop was hosted from 16-17 November 2023. This was a virtual workshop coordinated across 11 time zones. 26 expert nurse speakers delivered scientific presentations from different regions of the world.

Results: Attendance was across 7 continents, 37 countries, and 413 attendees. A total of 259 attendees from 6 countries from North and South Africa, 6 from the Asia-Pacific region, 36 from Central Asia, 20 from East and Western Europe, 15 from Latin America, 64 from the Middle East, and 13 from North America. The 2-day online workshop Program highlights included: I, introduction to HSCT, HLA, and pre-transplant preparations; ii, donor issues, cell sources, apheresis, and cryopreservation; iii, stem cell infusion, transfusion, complications, and management; iv, the role of nurses, pharma care, follow-ups; v, the role of careers quality management of patients and vi, the sickle cell patients. The workshop also features question-and-answer sessions that address unique challenges in each HSCT center, geographic area, and region. Other topics of interest included fertility preservation and survivorship after HSCT, especially in low-middle-income countries. 86.4% of attendees rated content as very relevant; 88.5% felt content impacted their knowledge, and >90% overall rated as excellent and would attend a similar event /webinar workshop.

Conclusions: The Nigeria BMT nurses’ online workshop was a huge success, with a positive impact on improving the fund of knowledge of participants in the area of HSCT.

The workshop curriculum focused on current advances across various disciplines and specialties, addressing disparities and impacting providers and nurses, especially from low-income countries. This workshop is a model for teaching HSCT care to healthcare professionals & trainees and is relevant to a broad medical audience across the globe.

Clinical Trial Registry: Not applicable.

Disclosure: None.

23: Haemoglobinopathy

P362 MATCHED-RELATED HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR INTERMEDIATE AND HIGH RISK THALASSEMIA PATIENTS USING REDUCED INTENSITY CONDITIONING REGIMEN WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE AS GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS. A SINGLE CENTER EXPERIENCE FROM JORDAN

Ayad Ahmed Hussein 1, Bayan Majed Alaaraj1, Tariq Ahmed Abdelghani1, Nour Awni Ghanem1, Hadeel Hassan Al-Zoubi1, Dina Mohammad Abu Assab1

1Bone Marrow and Stem Cell Transplantation Center, Istishari Hospital, Amman, Jordan

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is curative modality of treatment for patients with Thalassemia major (TM). Reduced-intensity conditioning (RIC) is associated with lower transplant-related toxicity; especially with intermediate and high risk patients. Post-transplant cyclophosphamide (PT/Cy) has improved engraftment and decreased the risk of graft-versus-host disease (GVHD).

Methods: This is a retrospective analysis of all patients with intermediate and high risk TM according to Pesaro classification, who received HSCT from matched related donor (MRD) at Dr. Ayad Bone Marrow and Stem Cell Transplantation Center, Istishari Hospital, Jordan from March 2019 till January 2023.

Results: Five patients with median age of 12 year (7-17) were included. Three were males. All patients received rabbit anti-thymocyte globulin (rATG) 0.5 mg/kg on day −10 and 1.5 mg/kg on days −9 to −7, thiotepa 10 mg/kg on day -7, fludarabine 30 mg/m2/day IV on days −6 to −2, cyclophosphamide 14.5 mg/kg/day IV on days −6 to −5, and 300 cGy TBI in a single fraction on day-1. The graft versus host disease (GVHD) prophylaxis was with post-transplant cyclophosphamide (PT/Cy) at 50 mg/kg/day IV on days +3 and +4 post-HSCT. Mycophenolate Mofetil and tacrolimus were started on day +5 and stopped on days +45 and +365 post-HSCT, respectively. The median CD34+ cell dose was 7.3×106/kg (2.35- 9.5×106/kg). Three patients received peripheral blood stem cells (PBSC). The median time for Neutrophil and platelet engraftment was 14 (13-17) and 26 (15-43) day respectively. Grade I–II mucositis occurred in all patients. None of the patients needed ICU admission. One patient experienced idiopathic pneumonia syndrome (IPS) that managed properly with systemic steroid and supportive care. Four patients had CMV reactivation, which was managed preemptively. Two patients developed acute grade I skin GVHD. No patients have developed chronic GVHD. At a median follow-up time of 30 month (11-56), all patients are alive, transfusion independent with donor-type Hb-electrophoresis.

Conclusions: The use of RIC with PT/Cy in MRD-HSCT for intermediate and high risk patients with TM is scarcely reported. This approach is safe, effective and associated with a favorable outcome with very low incidence of GVHD. More prospective studies with a larger sample size are required.

Disclosure: Nothing to declare.

23: Haemoglobinopathy

P363 ABO INCOMPATIBILITY DID NOT IMPACT TRANSPLANT OUTCOMES FOLLOWING REDUCED INTENSITY HAPLOIDENTICAL BONE MARROW TRANSPLANTATION FOR PATIENTS WITH SICKLE CELL DISEASE: MULTICENTER EXPERIENCE

Danielle Murphy 1, Pavina Akhom1, Karina Wilkerson1, Katie Gatwood1, Lindsay Orton1, Jim Connelly1, Reena Jayani1, Olalekan Oluwole1, Bhagirathbhai Dholaria1, Nancy Clayton-Long1, Brittney Baer1, Bipin Savani1, James Jerkins1, Andrew Jallouk1, Mohsen Alzahrani2, Ali D. Alhamari3, Josu de la Fuenta4, Erfan Nur5, Heidi Chen1, Adetola Kassim1

1Vanderbilt University Medical Center, Nashville, United States, 2King Abdulaziz Medical City, Riyadh, Saudi Arabia, 3King Faisal Specialist Hospital, Riyadh, Saudi Arabia, 4St Mary’s hospital, Imperial College, London, United Kingdom, 5Amsterdam University Medical Center, Amsterdam, Netherlands

Background: Common immune complications of ABO-incompatible allogeneic hematopoietic stem cell transplantation (HSCT) include pure red cell aplasia (PRCA) and early or delayed hemolysis. Data on the impact of ABO incompatibility on the incidence and severity of graft-versus-host disease (GVHD), engraftment, and survival is conflicting. HSCT is currently the only widely available curative therapy for SCD, which is offered as a standard of care for patients with a matched sibling donor (MSD). Still, donor availability is limited to a minority of patients. Haploidentical bone marrow transplant (haplo-BMT) has increased the donor pool, allowing the application to more eligible patients. ABO incompatibility adds to the immunologic barriers of an HLA mismatched transplant. We report the impact of donor–recipient ABO blood type matching on the outcomes of SCD patients receiving haplo-BMT.

Methods: The study was conducted as part of a multicenter learning collaborative (ClinicalTrials.gov identifier NCT01850108). All patients received common haplo-BMT conditioning. Primary graft failure (GF) was defined as <5% donor myeloid/lymphoid or whole blood (WB) chimerism at Day+30 post-BMT. Secondary GF was defined as <5% donor myeloid/lymphoid or WB chimerism with prior documentation of >5% donor cells at Day +30. ABO incompatibility was defined using donor->recipient blood types: major (A->O, B->O, AB->O, AB->A, AB->B), minor (O->A, O->B, O->AB, A->AB, B->AB), or bidirectional (A->B, B->A) mismatches.

Results: A total of 84 patients with SCD underwent related haplo-BMT. The mean age was 18.5 years (range 14 months to 47 years), with 44 patients </= 18 years and 40 patients >18 years; 39 patients were male (46.5%), and 45 were female (53.5%). ABO-incompatible transplant was minor in 14% (6/84), major in 10.5% (8/84), bidirectional in 16.8% (5/84), and ABO compatible was 58.7% of patients (65/84).

Average neutrophil engraftment was 19 days for ABO-compatible and 21 days for ABO-incompatible patients. Average platelet engraftment was 28 days for ABO compatible and 26 days for ABO incompatible. Grade III and IV Acute Graft Versus Host Disease (aGVHD) occurred in 4 ABO compatible patients (grades III, n=2, IV n=2) and 5 ABO incompatible patients (grades III n=5, IV n=1). Moderate to severe Chronic Graft Versus Host Disease (cGVHD) was observed in 4 total ABO compatible patients (moderate n=4, severe n=0) and 3 ABO incompatible patients (moderate n=1, severe n=2). Primary graft failure did not occur amongst any ABO incompatible patients, but did occur amongst 4 total ABO incompatible patients (n=4). Secondary graft failure did occur amongst one ABO incompatible patient and occurred amongst 4 total ABO incompatible patients (n=4). There were 2 deaths among the compatible group and 4 among the incompatible groups.

No statistical difference was seen between ABO incompatibility and engraftment, graft failure, acute or chronic GVHD or survival.

Conclusions: ABO-incompatibility status did not have a confounding effect on transplant outcomes following haploidentical BMT for SCD. This analysis validates a previous study conducted by these authors with a larger sample size including the pediatric population. More extensive prospective studies are necessary to validate this observation.

Clinical Trial Registry: https://clinicaltrials.gov/study/NCT01850108.

Disclosure: Nothing to declare.

23: Haemoglobinopathy

P364 DURABLE ENGRAFTMENT AFTER PHARMACOLOGICAL PRE-TRANSPLANT IMMUNE SUPPRESSION FOLLOWED BY REDUCED-TOXICITY MYELOABLATIVE HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN HIGHLY HLA-IMMUNIZED ADULTS WITH SICKLE CELL DISEASE

Sabine Fürst 1, Emmanuelle Bernit2, Faezeh Legrand1, Angela Granata1, Samia Harbi1, Raynier Devillier1, Benjamin Bouchacourt1, Thomas Pagliardini1, Valerio Maisano1, Djamel Mokart1, Claude Lemarié1, Boris Calmels1, Christophe Picard3, Agnès Basire3, Borje S. Andersson4, Didier Blaise1

1Institut Paoli Calmettes, Marseille, France, 2CHU Gouadeloupe, Pointe à Pitre, Guadeloupe, 3Etablissement Français du Sang, Marseille, France, 4University of Texas MD Anderson Cancer Center, Houston, United States

Background: Allogeneic stem cell transplantation (Allo-SCT) is the only rapidly available curative treatment modality in patients with severe sickle cell disease. The development of reduced-toxicity myeloablative conditioning (RT-MAC) regimen and the use of partially matched family donors with post-transplantation cyclophosphamide (PT-Cy) have widened the access to Allo-SCT. Antibodies against donor-specific HLA (DSA) increase the risk of engraftement failure in HLA mismatched Allo-SCT. We report the results of five patients with SCD, whereas three with DSA, who underwent an unmanipulated haploidentical stem cell transplantation (Haplo-SCT) after a busulfan-based RT-MAC regimen with PT-Cy. To reduce the risk of engraftment failure, a sequential two courses pharmacological pre-tranplant immune suppression (PTIS) phase was added prior to the conditioning regimen.

Methods: Only first-degree family members were evaluated after high resolution HLA-typing. The stem cell source was unmanipulated bone marrow with a target cell dose of 4x106 CD34+ cells/recipients body weight.

The DSA level was determined by using the LUMINEX technique (One Lambda, Inc., West Hills CA, USA). The values were expressed in mean fluorescence intensity (MFI). DSA were considered positive if the MFI value was ≥ 1000.

All five patients received two courses of a pharmacological PTIS phase prior to the start of the conditioning regimen. this initially was based on two sequential courses of Flu and Dxm, but was later modified to instead of Flu consider Cy and ATG to address the concern for Flu contributing to neurotoxicity in patients with cerebral sequelae. In patients with positive DSA the Flu-/Cy- + Dxm was fortiifed with rituximab and bortezomid followed by plasmapheresis to eliminate preformed DSA.

The transplant conditioning consisted of RT-MAC based on Flu and IV Bu with PT-Cy GVHD prophylaxis. We administered “early”ATG for all patients. The first patient received thiotepa (TT) in combination with Flu and Bu. The regimen was modified for the following patients by removing TT and including Bu by pharmacokinetic dose guidance targeting an average AUC after Bu 110 mg/m2 of 4,500 mMol-min daily over four days, or total course AUC of 18,000 mMol-min. All patients received GVHD prophylaxis with PT-Cy 50 mg/kg/day on days +3 and +4. Cyclosporine A (CSA) 1.5 mg/kg/twice a day and mycophenolate mofetil (MMF) 15 mg/kg/three times a day were administered from day +5.

Results: All patients engrafted successfully. the procedure was well tolerated. None of the patients developed acute GVHD, whereas one developed moderate chronic GVHD. After a median follow-up of 5 years, all patients were free of pain with excellent quality of life.

Table 1. Outcome

Patient 1

(HbSβ + )

Patient 2

(HbSβ + )

Patient 3

(HbSS)

Patient 4

(HbSS)

Patient 5

(HbSS)

ANC > 0.5 x 109 days

+26

+31

+16

+24

+21

PLT > 20 x 109 days

+38

+38

+26

+104

+26

Toxicity

muco gr 1

muco gr 2, ATG sickness

muco gr 2

muco gr 3, mild liver

muco gr 2, engraftment syndrome

Infections

HHV6

CMV, HHV-6, pneumonia

CMV, E. coli septicemia, C. difficile colitis, EBV

CMV, pyelonephritis

Pneumonia, otitis

Covid-19

a / c GVHD

(≥ mod)

-/-

-/-

-/moderate

-/-

- / -

Chimerism %

82

99

99

99

99

Stop IS, days

+372

+419

+1143

+514

+332

FUP, years

9

6

4

4

2

Conclusions: Adult patients with severe SCD should not be excluded from the possibility of a Haplo-SCT even it the presence of high titers of DSA. The pharmacological PTIS followed by a reduced toxicity ATG-Fludarabine-IV Busulfan-based conditioning regimen is feasible and safe and allows stable long-term engraftement and excellent disease control. Further studies are needed to confirm these promising resuts shown in our patients and to allow a widened access of this platform technology to patients with SCD.

Disclosure: Nothing to declare.

23: Haemoglobinopathy

P365 CLINICAL OUTCOMES AND HEALTHCARE RESOURCE USE IN PATIENTS WITH TRANSFUSION-DEPENDENT Β-THALASSEMIA WHO RECEIVED HEMATOPOIETIC STEM CELL TRANSPLANTS IN FRANCE

Jessica Baldwin1, Chuka Udeze 1, Nanxin Li1, Lyes Boulmerka1, Lila Dahal1, Giancarlo Pesce2, Nadia Quignot2, Heng Jiang2, Frederic Galactéros3

1Vertex Pharmaceuticals, Boston, United States, 2Certara France, Paris, France, 3Sickle Cell Referral Center, Internal Medicine Unit, Henri Mondor Hospital, Paris, France

Background: β-thalassemia is an inherited blood disorder caused by mutations in the β-globin gene, resulting in reduced, or absent expression of hemoglobin. Transfusion dependent β-thalassemia (TDT) is characterized by ineffective red blood cell production that leads to chronic anemia, need for regular red blood cell transfusions and iron chelation therapies to manage iron overload, and organ complications. Allogeneic hematopoietic stem cell transplant (HSCT) is currently available as a curative option for some patients. There is limited research on outcomes in patients with TDT who undergo HSCT in Europe. This study describes the clinical outcomes and healthcare resource use (HCRU) of patients with TDT who underwent HSCT in France.

Methods: This longitudinal, retrospective cohort study utilized the French National Health Data System database (SNDS, Système National Des Données De Santé). Patients with β-thalassemia were identified based on an inpatient claim or registration in the long-term condition database (ALD, Affection Longue Durée) with a diagnosis of β-thalassemia between January 1, 2012, and March 1, 2019. Patients with TDT were required to have ≥8 RBCTs per year in any 2 consecutive years after the first qualifying β-thalassemia claim. Eligible patients who had evidence of hereditary persistence of fetal hemoglobin, alpha thalassemia or sickle cell disease in their medical record were excluded. Patients who received HSCT were captured using relevant procedure codes. The index date was the date of transplant. Patients were followed from the index date until death, or end of study period (March 1, 2020). Demographics were assessed at the index date. Clinical complications and HCRU were summarized in the post-index period. A broader analysis of patients with β-thalassemia who underwent HSCT was also conducted.

Results: Overall, 9,913 patients with evidence of β-thalassemia in their medical records were identified and 83 underwent HSCT. Of the 9,913 patients with β-thalassemia, a total of 474 patients met additional criteria to be classified as TDT and 28 of these patients with TDT underwent HSCT. For patients with TDT (n=28), mean age of transplant was 9.2 years, and 57.1% (n=16) were female. A substantial proportion of patients with TDT who underwent HSCT experienced transplant related complications following HSCT, including graft versus host disease (GvHD) (10/28; 35.7%). Mean length of stay (LOS) for the index admission was 59.1 days. In the 1 year following transplant 91.5 total days were spent in the hospital on average, with 76.2 of those days occurring in the first 100 days after transplant.

Outcomes in the overall β-thalassemia cohort who received HSCT (n=83) were consistent with the cohort of TDT patients who received HSCT, including proportion of patients developing GvHD (33/83; 39.8%) and the mean total days spent in the hospital during the 100 days and 1 year after transplant (100 days: 79.6 days; 1 year: 100.1 days).

Conclusions: Only a small proportion of patients with β-thalassemia or TDT undergo HSCTs. Many patients who undergo transplants experience clinical complications related to HSCT, notably GvHD, which can be serious and potentially life threatening, highlighting the need for additional curative therapies for these patients.

Disclosure: Baldwin, Jessica: Vertex Pharmaceuticals employee.

Udeze, Chuka: Vertex Pharmaceuticals employee.

Li, Nanxin: Vertex Pharmaceuticals employee.

Boulmerka, Lyes: Grants or contracts and stock options from Vertex and Amgen.

Dahal, Lila: Vertex Pharmaceuticals employee.

Pesce, Giancarlo: Full-time employee of Certara with stock options and Certara received consulting fees from Vertex Pharmaceuticals.

Quignot, Nadia: Full-time employee of Certara with stock options and Certara received consulting fees from Vertex Pharmaceuticals.

Jiang, Heng: Full-time employee of Certara with stock options and Certara received consulting fees from Vertex Pharmaceuticals.

23: Haemoglobinopathy

P366 THE IMPACT OF PANEL REACTIVE ANTIBODIES IN PEDIATRIC BETA THALASSEMIA PATIENTS UNDERGOING STEM CELL TRANSPLANTATION

Gülay Sezgin1

1Çukurova University Medical School Division of Pediatric Oncology/Pediatric BMT Unit, Adana, Turkey

Background: With the developments in regular supportive treatment and iron chelation treatments for patients diagnosed with thalassemia major (transfusion dependent), life expectancy has improved significantly in recent years and gene therapies are promising. Although HSCT carries transplantation risks, it is currently the only curative standard treatment to avoid lifelong treatments and possible complications of iron overload. Anti-HLA antibodies have been found to be associated with rejection and low survival, especially in solid organ transplantations, and there are studies showing that they cause graft failure, low survival and engraftment delay in terms of HSCT. In this study, we aimed to evaluate the effect of anti-HLA status on graft failure, graft rejection, engraftment times, platelet and erythrocyte transfusion refractoriness and survival in patients diagnosed with TM (transfusion dependent) who underwent HSCT by us.

Methods: Eighty patients with beta thalassemia who received their first HSCT between January 1, 2015 and December 31, 2022 were enrolled in the study. All patients were transfusion dependent. Informed consent was obtained from their legal guardians. Ethical approval was obtained from the Institutional Review of Board of Çukurova University. Demographic and clinical characteristics of the patients (gender, age, stem cell source, donor type, HLA compatibility, ABO compatibility, number of CD34+ cells in the product, number of RBC transfusions before transplantation, splenectomy status, panel reactive antibodies) were recorded. PRA positive and negative patients were compared concerning the incidence of graft failure, graft rejection, engraftment times, platelet and erythrocyte transfusion refractoriness and survival (OS and EFS).

Results: 46 were anti-HLA positive (62.2%) Anti-HLA was found to be negative (37.8). When the final status of the patients was examined, it was seen that 53 patients were alive (66.3%) and 27 patients were dead (33.8%).

It was observed that the anti-HLA positive group received more ES transfusions before transplantation (p: 0.001), and there was no difference between them and the anti-HLA negative group in terms of engraftment time, post-transplant TS and ES support graft status and patient final status (p>0.05). It was observed that neutrophil engraftment occurred earlier in patients who underwent splenectomy than in those who did not undergo splenectomy (p:0.002), and the need for ES support after transplantation was less (p:0.030). There was no effect on graft status, patient final status and survival (p>0.05). A weak negative correlation was observed between the number of transplanted CD34+ stem cells and engraftment times (p<0.05). There was no significant difference between the groups in terms of ES and TS transfusion, graft status and patient final status after transplantation (p>0.05). Overall survival (OS) was 85% at day 100, 66.3% at 2, 5 and 8 years, and event-free survival (EFS) was 56.3% at 2 years, 5 and 8 years.

Conclusions: There was no relationship between anti-HLA status and neutrophil, platelet and erythrocyte engraftment. Although anti-HLA positive patients required more post-transplant TS and ES transfusions, this was not significant. In our study, no relationship was observed between anti-HLA positivity and graft status and mortality. There was no difference in survival between the anti-HLA positive and negative groups.

Disclosure: Nothing to declare.

23: Haemoglobinopathy

P367 SICKLE CELL DISEASE WITH SEVERE ACUTE CHEST SYNDROME AND HEPATOPATHY: CAN ALLOGENEIC SCT BE PERFORMED IN SUCH DESPERATE CLINICAL SITUATIONS? A CASE REPORT

Celia Kaffenberger 1, Eva Rettinger1, Shahrzad Bakhtiar1, André Willasch1, Roland Schrewe1, Michael Merker1, Birgit Knoppke2, Jean-Hugues Dalle3, Jan-Henning Klusmann1, Peter Bader1, Andrea Jarisch1

1Goethe University, University Hospital Frankfurt, Frankfurt am Main, Germany, 2University Hospital Regensburg, Regensburg, Germany, 3Assistance Publique-Hopitaux de Paris and University of Paris, Paris, France

Background: Sickle cell disease (SCD) often presents with severe organ manifestations, among others in the liver, affecting 40% of children with SCD due to recurrent crises and ischemia.1 Severe liver complications, such as acute sickle cell hepatic disease, sickle cell intrahepatic cholestasis, and acute hepatic sequestration, occur in 6% of cases, leading to potentially irreversible damage and challenging treatment involving monthly exchange transfusions.2 This case report elucidates the complicated nature of SCD which can be associated with severe hepatopathy. This leads to therapeutic challenges and decision-making complexities.

Methods: We present the case of a 4-year-old boy with SCD (HbSS) and associated severest hepatopathy which could only be controlled with regular erythrocyte apheresis and finally resolved by allogeneic stem cell transplantation.

Results: The patient developed a severe acute chest syndrome secondary to RSV infection, that lead to acute respiratory distress syndrome with the need for ECMO-therapy. The protracted intensive course ended in severe hepatopathy. Further diagnostics, including MRI and liver biopsy, revealed acute cholangitis and reversible liver damage. After exclusion of further differential diagnoses, SCD-associated hepatopathy was determined. A matched unrelated donor 9/10 was found, but due to extreme hepatopathy with bilirubin being as high as 54 mg/dl a hematopoietic stem cell transplantation (HSCT) in this state was deemed impossible. Despite effective erythrocyte apheresis with HbS levels <10% post apheresis every second week, the hepatopathy showed minimal improvement, making HSCT the only treatment option left. Still, transplantation in this case was associated with a much higher risk. One major risk being complete liver failure with the need for a liver transplantation. Based on this we considered it very difficult to use a matched unrelated donor, because a possible split liver transplantation would be impossible. As a consequence performance of haploidentical HSCT was considered. The patient’s father, a potential liver donor, was ineligible due to underlying medical conditions, prompting the mother’s selection as the donor. After extensive work-up of the mother, a conditioning regimen with Thiotepa, Treosulfan, and Fludarabine was chosen. Post-transplant cyclophosphamide was used for GVHD prevention. Already during conditioning the bilirubin started to continuously decline.

The patient achieved engraftment at day +22 for white blood cells and day +23 for platelets with complete donor chimerism and good bone marrow regeneration. The post-transplant course was complicated to veno-occlusive disease of the liver, which could finally be resolved after treatment with defibrotide. GVHD of the gut and skin up to overall Grad II was treated with methylprednisolone.

In addition, alloimmune-neutropenia and infections were successfully addressed, resulting in a final decrease in bilirubin levels to 3 mg/dl without further need of apheresis.

Conclusions: This case demonstrates the successful use of stem cell transplantation in a pediatric patient with a severe sickle cell-associated hepatopathy, shedding light on the SCD complications, the treatment decisions, and potential complications. The presented case proves that patients with severe and complicated courses of SCD can be cured by HSCT. Careful and interdisciplinary considerations are important to manage such devastating and desperate cases.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P368 OUTCOMES FOLLOWING HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR MONOGENIC INFLAMMATORY BOWEL DISEASE

Xiaowen Zhai 1, Ping Wang1, Wenjin Jiang1, Hongsheng Wang1, Ying Huang1, Xiaowen Qian1

1Children’s Hospital of Fudan University, Shanghai, China

Background: Monogenic inflammatory bowel disease (IBD) is caused by monogenic variants with Mendelian inheritance patterns, which is typically rare, severe, and refractory to conventional therapy. Allogeneic hematopoietic stem cell transplantation (HSCT) can serve as a curative treatment option. This study aimed to assess the outcome of patients with monogenic IBD after HSCT.

Methods: Pediatric patients with refractory monogenic IBD who underwent HSCT between January 2015 and October 2023 were retrospectively analyzed.

Results: One hundred and thirty-four patients with refractory monogenic IBD received HSCT, including 85 male and 49 female. Median age at HSCT was 15 months (IQR, 9–28 months). The cohort included IL-10R deficiency, chronic granulomatous disease (CGD), Wiskott–Aldrich syndrome (WAS), immune dysregulation, polyendocrinopathy, enteropathy X-linked (IPEX), lipopolysaccharide-responsive and beige-like anchor protein (LRBA) deficiency, CD40 ligand deficiency, Activated phosphoinositide 3-kinase δ syndrome (APDS), Leukocyte adhesion deficiency type 1 (LAD1), CARD11 deficiency, and one patient without specific genetic diagnosis. IL10RA, CYBB and WAS were top 3 mutant genes. Unrelated cord blood (UCB) was the main donor (76.1%), and the other donors included matched sibling donor (MSD) (6.0%), matched unrelated donor (MUD) (8.2%), mismatched related donor (MMRD) (6.7%) and mismatched unrelated donor (MMUD) (3.0%). Over a median follow-up of 29 months (IQR, 5–53 months), the 1-year and 3-year overall survival rate (OS) were 72.8% (95%CI 65.6%–80.8%) and 70.3% (95%CI 62.9%–78.6%), respectively. Patients with IL-10R deficiency had a lower OS rate compared with the others (67.1% versus 84.0%). Patients who received HSCT from MSD and MUD had higher OS rates compared with patients who received HSCT from MMUD, UCB and MMRD (100%, 90% versus 75%, 66.4%, 63.5%). All survivors achieved stable engraftment and gastrointestinal symptoms were resolved.

Conclusions: HSCT can cure some monogenic IBD. Patients with IL10R deficiency should proceed with caution and be under intensive monitoring after transplant. MSD and MUD are the preferred donor selection.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P369 CLINICAL CHARACTERISATION AND OUTCOME OF 125 CASES WITH GRISCELLI SYNDROME

Adriel Roa-Bautista1, Mahreen Sohail2, Reem Elfeky3, Maaike Kusters 3

1Manchester University Hospital NHS Foundation Trust, Manchester, United Kingdom, 2Barts Health NHS Trust, London, United Kingdom, 3Great Ormond Street Hospital for Children National Health Service (NHS) Foundation Trust, London, United Kingdom

Background: Griscelli type 2 syndrome (GS2) is caused by homozygous or compound heterozygous mutation in RAB27a gene. Classical phenotype presents with grey/silver hair, hypopigmented skin, and life-threatening haemophagocytic lymphohistiocytosis (HLH) with haematopoietic stem cell transplantation (HSCT) being the only curative option.

Methods: We systematically searched PubMed for relevant literature, covering publications from the earliest available date up to first of August 2023, with a particular focus on GS2 publications using keyword” Griscelli syndrome type 2”, “RAB27A”, “Hypopigmentation”, and “Silver-grey hair” and gathered data on patients’ characteristics, clinical presentation, molecular genetics, functional immunology tests and management; HSCT versus conservative approach.

Results: A total of 125 unique GS2 cases were identified (54.4% male) with a median age for diagnosis of 3 (range 0-42) years. Consanguinity was reported among 62% of cases. Positive family history of GS2 phenotype was seen among 45%, and 13% having sibling death due to HLH or infections. 80% of the cases had HLH as initial presentation at a median age of 1.83 years; including isolated systemic HLH, isolated CNS HLH and combined systemic and CNS HLH; 38%, 17% and 45%; respectively. 25 cases have no HLH symptoms and were mainly picked-up through positive family history. Silver-grey hair and hypopigmented skin were present among 62% and 31% of cases; respectively. More than 2/3 of the cases had no functional immunology testing done (NK killing, granule release assay), and HLH diagnosis was made on clinical criteria with GS2 diagnosis being based on genetics activated due to HLH or family history.

HLH was treated by HLH-94 in 18%, HLH-2004 in 12% or off protocol/not described in the remainder of cases. 41 patients (32.8%) underwent HSCT at median age of 2.25 (0.10-16) years. Donor choices were variable with the majority receiving a haploidentical donor transplant; 25/41. Mortality rates were 15% (6/41) among HSCT recipients versus 62.7% (37/59) among those who received conservative management (p <0.0001). Main cause of death among HSCT recipients was infection while HLH was the main cause of death among patients who were treated conservatively.

Conclusions: CNS HLH was recorded in more than 50% of GS2 cases described. Classical skin and hair abnormalities were absent in more than one third of GS2 patients at time of HLH presentation and thus should not rule out the diagnosis. Post-HSCT survival was superior to conservative management, suggesting adequate HLH control followed by early HSCT is needed. Asymptomatic cases picked-up through family history/genetic screening may benefit from pre-emptive HSCT, but access to functional immunological testing is required. High mortality related to HLH remains concerning and emphasises the need for better molecular characterisation and clinical prognostic factors to guide management decisions. Access to functional immunological testing for HLH pathways should be standard of care for primary HLH work-up.

Disclosure: Authors declare no conflict of interest.

25: Immunodeficiency Diseases and Macrophages

P370 OUTCOMES FOR ALLOGENEIC SCT FOR PATIENTS WITH INBORN ERRORS OF IMMUNITY UTILIZING REDUCED INTENSITY CONDITIONING REGIMEN WITH TARGETED BUSULFAN

Hannah Lust1, Olatundun Williams2, Morris Kletzel1, Reggie Duerst1, Jennifer Schneiderman1, William Tse3, Sonali Chaudhury 1

1Ann & Robert H. Lurie Children’s Hospital, Chicago, United States, 2Columbia University Irving Medical Center, New York, United States, 3University of Louisville, Norton Children’s Hospital, Louisville, United States

Background: Reduced intensity conditioning (RIC) for pediatric patients with inborn errors of immunity (IEI) receiving allogeneic stem cell transplant (alloSCT) has improved outcomes, though the optimal drug exposure remains unclear.

Methods: Conditioning consisted of busulfan (2 days), fludarabine 180mg/m2, and rATG 8mg/kg (Bu2/Flu/ATG). Prior to 2007, busulfan target cumulative AUC was 32mg/L*h (cohort 1); increased to 41mg/L*h from 2007 onward, and thiotepa (5 mg/kg) was added in cord blood (UCB) transplants (cohort 2) given high observed rates of graft failure with busulfan AUC 32mg/L*h. Preferred graft source was mobilized peripheral blood stem cell (PBSC) followed by MMF and cyclosporine as GVHD prophylaxis. Busulfan pharmacokinetic measurements were obtained with test dose and after 1st targeted dose.

Results: Between 2000 and 2023, 76 patients with IEI received Bu2/Flu/ATG conditioning (table 1). The group included 34 patients with SCID and 42 with other IEI, of whom 19 had primary immune regulatory disorders (PIRD). The median CD34 dose was 6.25 x 10E6 cells/kg. 5yr OS was 80% overall, 82% for SCID, 78% for other IEI, and 73% for PIRD (fig 1A, P=0.59). 5yr OS was lower for UCB/bone marrow (BM) graft recipients compared to PBSC - 66% vs 88% (P=0.01). Improved outcomes with 5yr OS 95% were observed in SCID patients receiving PBSC vs 58% in UCB recipients (fig 1B, P=0.02). Graft source did not impact survival in non-SCID IEI (79% PBSC vs 76% CB/BM). Patients receiving HLA-matched PBSC had 5-year OS of 86% vs 73% for those receiving a mismatched graft (P=0.11). Patients treated in cohort 1 experienced lower 5yr OS – 63% vs 85% in cohort 2 (P=0.07). Nine deaths occurred prior to D + 100, with 5 deaths in cohort 1 (3 attributed to primary graft failure) and 4 deaths in cohort 2 (due to infection or VOD). Seven deaths occurred after D + 100, largely related to infection. Only 2 patients (3%) experienced VOD/SOS. Acute GVHD of any grade was seen in 22% of patients, with only 3 patients (4%) experiencing > grade 2 aGVHD. At 1-year post alloSCT, 82% achieved stable full donor T-cell chimerism and 62% achieved full myeloid chimerism. Immune reconstitution was robust with CD3 count >1000 in 80%, CD4 count >500 in 86%, and CD19 count >200 in 90% with IVIG independence in 77% at 1-year post-alloSCT. Of survivors with available data, 97% exhibited adequate response to vaccines (94% of SCID, 100% of other IEI). With continuing long term follow up, 5 patients have autoimmune complications, and 10 patients were diagnosed with endocrinopathy.

Table 1: Patient & Transplant Characteristics

Overall, N=76

SCID, N=34

Other IEI, N=42

Median age at HCT, months (range)

7.5 (1-266)

3 (1-97)

28.5 (2-266)

Median follow up, years (range)

7.7 (0.2-21.4)

10.1 (0.2-17.3)

6.3 (0.5-21.4)

Female

20 (26)

14 (41)

6 (14)

Male

56 (74)

20 (59)

36 (86)

Donor type, N (%)*

MRD

19 (25)

3 (9)

16 (38)

MUD

21 (28)

12 (36)

10 (24)

Mismatched

35 (46)

18 (53)

16 (38)

HLA matching, N (%)^

Fully matched (PB, BM 8/8 or CB 6/6)

42 (56)

16 (49)

26 (62)

1 Ag mismatch (PB, BM 7/8 or CB 5/6)

23 (31)

12 (36)

11 (26)

>1 Ag mismatch (PB, BM <7/8 or CB <5/6)

10 (13)

5 (15)

5 (12)

Peripheral blood

51 (67)

22 (65)

29 (69)

Umbilical cord blood

22 (29)

12 (35)

10 (24)

Bone marrow

3 (4)

3 (7)

Median busulfan cumulative exposure^, mg/L*h (range)

39.5 (23.9-71.7)

38.1 (23.9-46.6)

40.3 (28.5-71.7)

Cohort by busulfan target, thiotepa

Cohort 1, target AUC 32mg/L*H, years 1992-2006

16 (21)

9 (26)

7 (17)

Cohort 2, target AUC 41mg/L*H, years 2007-2023

60 (79)

25 (74)

35 (83)

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (43)

Conclusions: We demonstrate excellent 5yr OS of 88% utilizing busulfan/fludarabine-based RIC and PBSC grafts in this IEI patient population with low rates of graft failure. Rates of severe aGVHD and cGVHD were very low despite the prevalence of PBSC grafts. We also demonstrate very good 5yr OS in patients with PIRD utilizing this RIC regimen, suggesting they may not require more intensive conditioning. While we noted some mixed chimerism in the long-term, immune reconstitution in all disease groups was encouraging.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P371 OUTCOME OF THE PATIENTS DIAGNOSED WITH SEVERE COMBINED IMMUNODEFICIENCY THROUGH NEWBORN SCREENING IN CATALONIA (2017-2023)

Laura Alonso1, Andrea Martin-Nalda1, Maria Isabel Benitez Carabante 1, Ana Argudo-Ramirez2, Luz Uria-Ofialdegui1, Judit Garcia-Villoria2, Melissa Panesso1, Jacques Gabriel Riviere1, Pere Soler-Palacin1, Cristina Diaz de Heredia1

1Hospital Universitari MaternoInfantil Vall d´Hebron, Barcelona, Spain, 2Hospital Clinic, Barcelona, Spain

Background: In Catalonia, newborn screening (NBS) for severe combined immunodeficiency (SCID) by T-cell receptor excision circles (TRECS) was initiated in January 2017. This is the first European region to universally include this testing.

Data of the patients diagnosed with SCID, their characteristics, treatment and outcomes, following the implementation of this NBS program are presented.

Methods: Retrospective analysis of the patients diagnosed with SCID in the NBS program in Catalonia. Data between January 2017 and 31/11/2023 were analysed.

Results: Demographics

Of the 413.207 newborns screened, 8 children were diagnosed with SCID; incidence 1:51651. All the patients were referred to the Children’s Hospital at Vall d´Hebron Barcelona Hospital Campus. One patient presented X-linked SCID, had poor donor options and underwent gene therapy; one patient presented FOXI3 athymia and received a thymus trasplant. These 2 patients are alive and well. The remaining six patients were considered candidates for HCT, five of them have already been transplanted and one is awaiting HCT.

Characteristics of the patients who received an HCT:

Patient characteristics and pre-HCT evolution (Table 1)

Full size table

The patients were 4 males and one female. The SCID phenotype was T- B- NK+ in all. One had no genetic confirmation but DNA repair syndrome was suspected, three had RAG2 mutations, and 1 patient PNP deficiency.

None of the patients developed overt Omenn syndrome requiring systemic therapy; two received topical steroids for skin rash. Three experienced gastrointestinal symptoms and were diagnosed with cow´s milk protein intolerance. There were no significant pre-HCT infections.

HCT process

Patients were 1.8 to 3 months of age at HCT and their weight ranged from 4 to 5.5Kg. None of them presented relevant active complications before HCT.

Stem cell donors were matched sibling donors (n=2) unrelated cord blood (n=2), and haploidentical (n=1).

Conditioning therapy consisted of ATG, fludarabine and busulfan in 4 cases. Busulfan AUC was adjusted in all of them to 55-65 mg*h/l. The patient with suspected DNA repair deficiency received ATG, fludarabine and cyclophosphamide.

All the patients engrafted. The patient with the haploidentical HCT experienced pancytopenia and declining chimerism and received CD34+boost and DLI with favourable outcome.

Two patients developed mild venooclusive disease of the liver despite prophylaxis with defibrotide that resolved with supportive therapy. On patient who received an UCB transplant developed engraftment syndrome.

There were no cases of Grade >2 acute GVHD or severe chronic GVHD.

Two patients required rituximab for EBV reactivation. None experienced CMV infection. Two patients developed pericardial effusion and one, autoimmune hemolytic anemia.

Outcomes.

Median follow up after HCT is 3.5 years (0.05-5.5).

Four patients are alive, three with stable chimerism, one of them 3 weeks post-HCT and already engrafted. Two patients have successfully stopped immunoglobulin replacement therapy and one is off immunosuppresion but still requires immunoglobulin replacement. One patient died of septic shock in the context of refractory hemolytic anemia 9 months-post-HCT.

Conclusions: These results provide preliminary evidence of the benefits of including SCID in newborn screening programs. The patients benefitted from early treatment and arrived to the curative therapy in good clinical condition.

Disclosure: None.

25: Immunodeficiency Diseases and Macrophages

P372 HSCT AS A CURATIVE TREATMENT OPTION IN PATIENTS WITH A PRIMARY IMMUNODEFICIENCY DUE TO IL2RB-DEFECT: DESCRIPTION OF A SMALL COHORT

Ommo E. Mauss 1, Christo Tsilifis2,3, Eva-Maria Jacobsen1, Manfred Hönig1, Ansgar Schulz1, Mary A. Slatter2,3, Sophie Hambleton2,3, Klaus-Michael Debatin1, Klaus Schwarz4,5, Ulrich Pannicke4,5, Andrew R. Gennery2,3, Mehtap Sirin1

1University Medical Centre Ulm, Ulm, Germany, 2Great North Children’s Hospital, Newcastle upon Tyne, United Kingdom, 3Newcastle University, Newcastle upon Tyne, United Kingdom, 4German Red Cross Blood Service Baden-Württemberg and Hessen, Ulm, Germany, 5University Ulm, Ulm, Germany

Background: A novel Inborn error of Immunity caused by genetic variants in IL2RB has been first described by Zhang et al in 2019. Since then, no further data regarding HSCT as a curative option in these patients has been published. Here, we report on three patients who presented at the age of four to 18 months with the characteristic clinical phenotype of eczema, lymphadenopathy, failure to thrive and EBV or CMV viraemia with elevated IgG-levels and low regulatory T-cells. For detailed patient data see table 1.

Full size table

.

Methods: Patients were transplanted between the age of seven to 25 months in Ulm and Newcastle. Patient A received PBSC from a 12/12 MUD (CMV + ) after conditioning with Treo/Flu, Alemtuzumab. Patient B was initially transplanted from a 6/12 MMFD (father, CMV + ) after TCRα/ß-depletion and conditioning with Treo/Flu/TT, ATG, Rituximab. She experienced a secondary graft failure 6 months after transplant and was successfully re-transplanted with PBSC from a 9/12 MFD (mother, CMV + ) after conditioning with Treo/Flu/TT, Alemtuzumab. Patient C received PBSC from a 10/12 MUD after conditioning with Bu(75)/Flu, Alemtuzumab, Rituximab. All patients received MMF and CSA as GvHD prophylaxis.

Results: Patient A reactivated CMV on d + 14 and died of CMV pneumonitis on d + 34 (Patient B1 in Zhang et al, 2019). Patient B had asymptomatic CMV viraemia prior to transplantation treated with ganciclovir. She developed mild GvHD of the skin (treated with steroids, IFX, ECP) and experienced secondary graft failure 6 months post-transplant. She received a second HSCT under ongoing steroid treatment for her GvHD and is now free of disease with full donor chimerism 36 months after her initial transplantation. She reactivated CMV after both transplantations, but never developed CMV disease under antiviral treatment. EBV viraemia in patient C was treated with Rituximab prior to transplant. She had mild engraftment syndrome and VOD and developed a mild GvHD of the skin which was treated with steroids, MTX, Ruxolitinib. 6 months after transplant, she is free of disease with 10% autologous T-cells, but full donor chimerism for CD3-negative cells and granulocytes. Her eczema and lymphadenopathy significantly improved during conditioning and skin fully recovered without scarring over the first month post-transplant.

Conclusions: In conclusion, we were able to demonstrate that HSCT is a curative treatment option in patients with IL2RB-defects. However, early diagnosis appears to be crucial as infectious complications (mainly CMV and EBV viremia) appear early in the course of disease and heavily impact treatment, complications and mortality. Despite severe eczema of the skin in our patients prior to transplant, no cases of severe GvHD were observed in our cohort. Symptoms of immune dysregulation (eczema, lymphadenopathy, enteritis) drastically decreased during transplantation and further stabilised with ongoing immune reconstitution. Interestingly, split donor chimerism for CD3+ cells seems to be sufficient to control associated immune dysregulation, presumably associated with reconstitution of the T-reg compartment. It will be vital to collect data of additional patients to optimize treatment plans and properly assess transplant outcomes. We suggest to retrospectively analyse similar historic patients to identify additional cases of IL2RB-defects.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P373 WHICH ALTERNATIVE TO CHOOSE? - A COMPARATIVE STUDY BETWEEN MATCHED UNRELATED AND HAPLOIDENTICAL DONOR TRANSPLANTS IN CHILDREN WITH INBORN ERRORS OF IMMUNITY IN LOW-MIDDLE-INCOME COUNTRIES

Anuraag Reddy Nalla 1, Revathi Raj1, Ramya Uppuluri1, Venkateswaran Swaminathan1, Kavitha Ganesan1, Suresh R D1, Anupama Nair1, Vijayshree M1, Indira Jayakumar1, Ravindra Prasad Thokala1, Usha Suriyanarayanan1

1Apollo Cancer Hospitals, Chennai, India

Background: Hematopoietic stem cell transplant (HSCT) has emerged as a curative option for inborn errors of immunity (IEI) in children. An HLA-matched unrelated donor or a haploidentical-related donor are the options in children without a matched sibling donor. The possibility of finding an HLA-matched unrelated donor (MUD) is 16 % for the Indian population and 100% for haploidentical donor. Patients with severe combined immune deficiency (SCID) are diagnosed during infancy and are a unique group of patients. Our study aimed to analyze the impact of the type of IEI and donor source on survival in low-middle-income countries.

Methods: We conducted a retrospective analysis of the children with IEI who underwent HSCT at our center between June 2008 and May 2023. We collected data about underlying diagnosis, donor source, conditioning, engraftment, graft versus host disease (GVHD), viral reactivation, and survival. Data analysis was performed using SPSS software.

Results: One hundred ninety-eight children underwent HSCT at our center, of which the donor was a matched family donor (MFD) in 70, MUD in 42 and haploidentical family donor in 86 children. We analyzed the data on 128 children who had an alternate donor HSCT with follow up ranging from 12 to 147 months.

In MUD HSCT, 36 patients engrafted (85.7%) and 9 (21.4%) had graft rejection - with 6 (14.2%) primary and 3 (7.1%) secondary graft rejection. We documented acute GVHD in 9/33 (27%) and chronic GVHD in 18/33 (54%) children. Cytomegalovirus and adenovirus reactivation was seen in 26 (61.9%) patients. The major cause of mortality was sepsis at 40% and GVHD at 45%. The survival for children with SCID was 30%, HLH 78% and non CGD phagocytic defects was 100%.

Among haploidentical donor HSCTs, 73 (84.8%) patients engrafted, and 21 (24.4%) had a graft failure – 13 had primary and 8 had secondary graft failure. We documented acute GVHD in 11 (15%) patients and chronic GVHD in 12 (16.4%). Viral reactivation was seen in 51 (59.3%) of patients. Among the 37 (43%) patients who died in this cohort, sepsis (54%) and viral reactivation (37.8%) were the most common causes of death. The survival for SCID 54.5%, CGD 66.6% and WAS at 50%.

There was no difference in overall survival in the MUD cohort (52.4%) versus haploidentical cohort (57%) with a p value of 0.38. The survival in SCID was inferior at 54% compared to non SCID cohort at 69% (p value 0.03). The rates of chronic GVHD were significantly higher in MUD at 54.5% versus haploidentical at 18.4% - p value (<0.001).

Table 1: Survival in alternate donor HSCTs in different IEI.

Disease (n)

MUD HSCT

Haploidentical donor HSCT

Severe combined immunodeficiency (SCID) (53)

10

22

Survival: 3 (30%)

12 (54.5%)

HLH and HLH like syndromes (37)

9

17

Survival: 7 (77.8%)

10 (58.8%)

Other immune dysregulation – (IPEX, LRBA, XIAP, IL 10 R deficiency, XLP)

2

6

Survival: 1 (50%)

4 (66.6%)

Chronic granulomatous disease (CGD)

4

12

Survival: 1 (25%)

8 (66.6%)

Other phagocytic defects: (LAD, congenital neutropenia)

3

5

Survival: 3 (100%)

2 (40%)

Hyper IgE syndromes

2

3

Survival: 2 (100%)

Antibody deficiency: (XLA, Hyper IgM)

2

4

Survival: 2 (100%)

2 (50%)

Wiskott Aldrich syndrome (WAS)

10

12

Survival: 3 (30%)

6 (50%)

Conclusions: There was no difference in overall survival between the MUD and haploidentical donor HSCTs in our IEI cohort. Children with SCID present with significant comorbidity and hence have inferior survival. We need to use the earliest available optimal alternate donor to prevent a delay in HSCT. We need strategies to reduce mortality due to sepsis and viral reactivation in the entire alternate donor cohort and morbidity due to chronic GVHD in our MUD cohort.

Clinical Trial Registry: NA.

Disclosure: NOTHING TO DECLARE.

25: Immunodeficiency Diseases and Macrophages

P374 MUD-HSCT IN A RARE COMBINED IMMUNODEFICIENCY WITH ARPC1B DEFICIENCY-CASE REPORT

Devyani Surange 1, Rajiv Kumar1, Rajan Kapoor1, Sanjeev Khera1

1Army Hospital Research and Referral, New Delhi, India

Background: Recently, a novel syndrome of combined immune deficiency, infections, allergy, and inflammation has been attributed to mutations in the gene encoding actin-related protein 2/3 complex subunit 1B (ARPC1B), which is a key molecule driving the dynamics of the cytoskeleton and was reported for the first time in 2016. To date, only a few individuals have been diagnosed with ARPC1B deficiency worldwide with 3 cases from Nepal so the exact incidence and prevalence is not known.

Methods: We herein report a case of ARPC1B deficiency who underwent MUD-HSCT successfully with no evidence of GVHD and was discharged on Day+16 and is now on follow-up.

Results: A 7-year-old male with Nepalese ancestry born of non-consanguineous marriage presented at 2 months of age with history of multiple hospital admissions with failure to thrive, recurrent bloody stools, haematuria, gum bleeding, and eczematous rashes. Over 3 years, the child was admitted multiple times with recurrent infections (Pneumonia, chronic suppurative otitis media). Physical examination revealed maloccluded teeth, dental caries, and stunted growth. CBC showed anemia and thrombocytopenia. Immunoglobulin profile revealed Low S. IgG with high S. IgA and S. IgE. Faecal calprotectin and S. IgA TTG were raised along with increased CRP and ESR. Sigmoidoscopic biopsy was suggestive of eosinophilic colitis. Lymphocyte subset analysis showed reduced circulating CD3 + , CD4 + , and CD8 + T cells and platelet function assay revealed mild dysfunction. Based on the above findings, NGS-WES was done and ARPC1B deficiency-Homozygous was detected. He was initiated on Prednisolone, MMF, and sirolimus and put on monthly IVIG therapy. Due to the lack of matched sibling donors and HSCT being the established curative treatment for this deficiency, a 12/12 MUD match was found. He was detected HCV positive and was treated. Pre-transplant, he had an active infection (Biofire positive for H.influenzae and Rhinovirus), Raised S. Galactomannan, and CMV-DNA -7036 IU/ml. He was initiated on IV antibiotics, antifungals, and antivirals and was taken up for transplant with myeloablative conditioning with Fludarabine (40mg/m2 x 4 days) and Fractionated Busulfan (3.2mg/kg/day x 4 days). GVHD prophylaxis used was PTCy+Tacrolimus+MMF. CD34 stem cell dose was 4.1 x 106/kg. The patient had an uneventful peri-transplant period with platelet engraftment on Day+14, neutrophil engraftment on Day+13, and was discharged on Day+16.

In a study by Stefano Giardino et al (2022), the outcomes of 7 patients who underwent allo-HSCT for ARPC1B deficiency was described. In the study, the donor type was MUD in 2 cases, and the conditioning regimen used was (Flu-Bu-ATG and Flu-Bu). GVHD prophylaxis was PTCY-MMF-FK506 in one patient and CsA-Mtx in other. Engraftment rates were similar with 100% donor chimerism and no evidence of GVHD.

Conclusions: Although careful monitoring, antimicrobial prophylaxis, and adequate treatment are mandatory to prevent and counter infections, the immune dysregulation contributing to systemic inflammation requires immunosuppression. Whereas both ARPC1A and ARPC1B are present in tissue cells, ARPC1B is the only ARPC1 isoform in hematopoietic cells. The unique and variable combination of clinical features makes ARPC1B deficiency a complex disease entity for which HSCT is considered a curative treatment option.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P375 ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION (HCT) IN TWO PEDIATRIC PATIENTS WITH CENTRAL NERVOUS SYSTEM-RESTRICTED FAMILIAL HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS

Alison Cusmano1, Ariane Soldatos1, Jessenia Campos1, Katherine Townsend1, Corina Gonzalez1, Allison Grimes2, Sara Makkeyah3, Maha Mohammed3, Hoda Tomoum3, Luigi Notarangelo1, Laila Selim4, Jennifer Kanakry1, Dimana Dimitrova 1

1National Institutes of Health, Bethesda, United States, 2University of Texas Health San Antonio, San Antonio, United States, 3Ain Shams University, Cairo, Egypt, 4Cairo University Children’s Hospital, Cairo, Egypt

Background: Familial hemophagocytic lymphohistiocytosis (HLH) is increasingly identified in patients presenting with HLH as extensive genetic testing becomes more rapidly and readily available. When disease is restricted to the central nervous system (CNS) without systemic manifestations, however, making the initial diagnosis of HLH is challenging, and referral for curative allogeneic HCT may be delayed, potentially resulting in irreversible neurological injury. Herein, we report two successful pediatric HCTs for CNS-restricted familial HLH in the setting of PRF1 deficiency.

Methods: P1, 5yo female, was previously healthy with normal development before presenting at 3yo with progressive gait abnormalities. P2 presented at 10yo with ataxia, speech difficulties, severe headaches, and urinary incontinence. Both P1 and P2 received therapies for alternative suspected diagnoses before genetic sequencing revealed biallelic mutations in PRF1, leading to the diagnosis and directed treatment of CNS-restricted HLH. (Table 1) P1’s healthy HLA-matched sibling was considered as a potential donor but found to be affected despite remaining asymptomatic. P1 and P2 were enrolled on NCT03663933 (Clinicaltrials.gov) and received T-replete peripheral blood stem cell transplants with reduced intensity conditioning using distally-timed equine antithymocyte globulin, pentostatin, hyperfractionated cyclophosphamide, and 2 days of busulfan (total treatment exposure target of 46 mg x h/L). P1’s donor was her haploidentical father, while P2 received a graft from her HLA-matched sister, both carriers of a single pathogenic PRF1 mutation. Graft-versus-host-disease (GVHD) prophylaxis included high dose posttransplantation cyclophosphamide, mycophenolate mofetil, and tacrolimus.

Table 1. Clinical characteristics of patients pre-and post-HCT.

P1

P2

Age

3y9m at presentation

10y1m at presentation

4y7m at diagnosis

10y3m at diagnosis

5y9m at HCT

10y9m at HCT

Genetic defect

Compound heterozygous PRF1 mutations:

Homozygous PRF1 mutations:

c1081A>T, p.Arg361Trp (missense)

c.673C>T, p.Arg225Trp (missense)

c.1122G>A, p.Trp374* (nonsense)

Perforin expression reduced

Perforin expression reduced

Clinical presentation and prior therapies

Abnormal gait progressing to inability to walk

Anarthria with intact receptive language

Movement disorder

Suspected multiple sclerosis:

• Systemic corticosteroids

• High dose IVIG x2

• Plasmapheresis x 6 sessions

• Rituximab x5

• High dose cyclophosphamide x 3 doses

HLH:

• HLH-2004 protocol x2 including IT MTX

• Ruxolitinib

• Emapalumab pre-HCT for elevated CXCL9

Cerebellar ataxia

Loss of expressive language (improved with therapy)

Urinary incontinence (resolved with therapy)

Anxiety and mood lability

Suspected ADEM vs. lupus cerebritis ( + ANA, anti-Sm; low C3/C4):

• Systemic corticosteroids

• Cyclophosphamide (1500mg/m2 lifetime dose)

• High dose IVIG

• Mycophenolate mofetil

HLH:

• HLH 2004 protocol x2 including IT MTX

• Anakinra x1.5mos

Treatment-related complications

Steroid-induced osteoporosis

Steroid-induced intraocular hypertension, low bone mineral density, and visceral adiposity

Adrenal insufficiency

Hypogammaglobulinemia

HCT-CI score

3

CSF evaluations (ref. range)

Pre-HCT a

Post-HCT (day + 60)

Pre-HCT a

Post-HCT (day + 60)

WBC count, cytopathology

1/uL

Pre-HLH diagnosis: 5/uL lymphocytes

2/uL

Lymphocytes, monocytes

1/uL

Lymphocytes, monocytesb

1/uL

Lymphocytes, monocytes, some with reactive changes (irregular nuclear contours)

Protein (15-40)

29 mg/dL

22 mg/dL

24 mg/dL

22 mg/dL

Cytokine panel

IL-6 ↑

normal

normal

normal

Neopterin (7-40)

111 nmol/L ↑

33 nmol/L

20 nmol/L

NA

Peripheral blood (ref. range)

Pre-HCT

Post-HCT

Pre-HCT

Post-HCT

CXCL9 (<647)

7696 pg/mL ↑

424 pg/mL (day +60)

977 pg/mL ↑ c

NA

Soluble IL2R (175-858)

1301.6 pg/mL ↑ cd

725.7 pg/mL (day +60)

640.4 pg/mL

756.2 pg/mL (day +60)

IL-18 (<468)

573 ↑

NA

537 ↑

NA

Ferritin (14-79)

140 mcg/L d

88 mcg/L (day +60)

32 mcg/L

NA

Bone marrow biopsy

No hemophagocytosis

No hemophagocytosis

No hemophagocytosis

Rare hemophagocytosis of no clinical significance

Chimerism at last follow up (% donor)

CD3: 100%

Myeloid: 100%

CD3: 87%

Myeloid: 100%

Outcome

Alive, off immunosuppression at 117 days post-HCT

No brain MRI evidence of increased inflammation at day +41, no signs of systemic HLH

Improvement in speech, tracking gaze

Alive at 73 days post-HCT

No brain MRI evidence of increased inflammation at day +28, no signs of systemic HLH

Improvement in cognition, memory and ambulation

  1. Abbreviations: ADEM, acute disseminated encephalomyelitis; HCT-CI, HCT comorbidity index; HLH, hemophagocytic lymphohistiocytosis; IT MTX, intrathecal methotrexate; IVIG, intravenous immunoglobulin; NA, not available.
  2. aevaluations within 1 month of beginning conditioning for HCT unless otherwise noted
  3. bprior cytopathology showed rare hemophagocytosis at time of relapse
  4. c patient with concurrent urinary tract infection
  5. dnormal at initial presentation

Results: Both patients engrafted on day +13. With 117 and 73 days follow up respectively, transplant-related complications have been few. P1’s course has been complicated by grade 1, skin only acute GVHD, resolved without systemic therapy, and non-traumatic tibial fracture in the setting of steroid-induced osteoporosis, while P2 developed mild BK virus-associated hemorrhagic cystitis and bacterial urinary tract infections in the setting of pre-existing vesicoureteral reflux. Post-HCT, neither patient has had any evidence of systemic HLH or required any HLH-directed therapy. Additionally, neither has suffered any neurologic decline since HCT, nor is there post-HCT evidence of further inflammation on MRI neuroimaging. With intense multimodal rehabilitation therapy, P1 shows improved ability to vocalize and track, while P2 shows improved cognition, memory, and ambulation.

Conclusions: As has been shown in HCT recipients with systemic primary HLH, serotherapy-containing RIC is the preferred HCT approach in order to minimize toxicity and is sufficient for engraftment and phenotype reversal. Use of alkylating agents which penetrate the blood brain barrier, such as busulfan or melphalan but not treosulfan, may theoretically offer disease-stabilizing benefit when the CNS alone is involved. Importantly, siblings or other relatives within consanguineous families should undergo genetic testing prior to serving as donors even if they are apparently asymptomatic. Prognosis for neurologic recovery in the presence of severe pre-HCT injury remains uncertain, and early diagnosis of primary CNS-restricted HLH with prompt referral for allogeneic HCT is critical for optimizing patient outcomes.

Clinical Trial Registry: NCT03663933 (Clinicaltrials.gov).

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P376 SIL2-RΑ AND ST2 LEVELS BEFORE HEMATOPOIETIC STEM CELL TRANSPLANTATION ARE ASSOCIATED WITH MORTALITY IN HLH PATIENTS

Anne B. Verbeek 1, Anja M. Jansen-Hoogendijk1, Erik G J. von Asmuth1, Marco W. Schilham1, Arjan C. Lankester1, Emilie P. Buddingh1

1Leiden University Medical Center, Leiden, Netherlands

Background: Overall survival after hematopoietic stem cell transplantation (HSCT) for hemophagocytic lymphohistiocytosis (HLH) has plateaued at ~70%. A better understanding of inflammatory dynamics may create opportunities for intervention. In this study we aimed to longitudinally evaluate cytokine dynamics in children transplanted for HLH and to evaluate whether levels of drug-targetable cytokines are associated with survival after hematopoietic stem cell transplantation (HSCT).

Methods: We measured 25 cytokines using a Luminex immunoassay in 35 HLH patients at 6 different timepoints: before start of conditioning (TP1), after conditioning but before HSCT (TP2), at day 7 after HSCT (TP3), at the time of neutrophil engraftment (TP4), at day 100 after HSCT (TP5) and, if applicable, in the week before the patient died (TP6). Protein levels over time were visualized using median and interquartile range values at each timepoint. We identified proteins which optimally discriminated between patients who survived and who died using a recursive partitioning survival decision tree.

Results: Patient and transplant characteristics are shown in Table 1. Overall survival at 1-year post-HSCT was 60%. Some cytokines such as sIL-2Rα, interferon-gamma inducible CXCL16 and ST2 were upregulated after conditioning. Other cytokines, such as IL-6 and CXCL8 were upregulated in the weeks after transplantation. Soluble interleukin-2 receptor alpha (sIL2Rα) at TP2 best discriminated between patients who did and did not survive all 11 patients with a sIL2Rα level below 1150 pg/mL were alive at 1 year after HSCT, whereas overall survival in the 24 patients with high sIL2Rα levels was only 40% (Log-rank p = 0.0044). The second best discriminating protein was Suppression of tumorigenicity 2 (ST2) at TP2 with a cutoff value of 45000 pg/ml (lower (n = 18) OS 85% vs higher (n = 17) OS 30%, Log-rank p = 0.0015). In all patients that died, there was a pronounced hyperinflammatory profile in the days before death.

Table 1: Patient and Transplant characteristics

N = 35

Age (median, IQR)

1.7 (0.9, 4.9)

Patient sex (n, %)

Male

26 (74%)

Female

9 (26%)

Donor type (n, %)

Mismatched unrelated donor

12 (34%)

Matched unrelated donor

7 (20%)

Matched related donor

6 (17%)

Mismatched related donor

5 (14%)

Unrelated donor (matching unknown)

5 (14%)

Graft type (n, %)

Bone marrow

18 (51%)

Cord blood

9 (26%)

Peripheral blood stem cells

8 (23%)

Overall survival at 1 year post-HSCT (n, %)

21 (60%)

AGvHD cumulative incidence at 1 year post-HSCT (n, %)

3 (8.6%)

Conclusions: HLH patients have poor survival after HSCT. High SIL2Rα and ST2 levels immediately after conditioning were associated with increased mortality after HSCT in children with HLH. Interventions to decrease inflammation during conditioning may result in a better outcome. Future trials should confirm these findings.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P377 HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN FOR PRIMARY IMMUNE DEFICIENCIES: THE EXPERIENCE OF SINGLE CENTER

Utku Aygunes1, Ali Antmen 1, Ilgen Sasmaz1,2, Barbaros Karagun1, Duygu Turksoy1

1Acibadem Adana Hospital, Adana, Turkey, 2Cukurova University, Adana, Turkey

Background: Primary immune deficiencies (PID) are disorders that for the most part result from mutations in genes involved in immune host defense and immunoregulation. These conditions are characterized by various combinations of recurrent infections, autoimmunity, lymphoproliferation, inflammatory manifestations, atopy, and malignancy. Hematopoietic stem cell transplantation (HSCT) is the curative option for many primary immune deficiency disorders at this time. We aimed to present the results of HSCT for children with PID at Acibadem Adana Hospital Pediatric Bone Marrow Transplantation Unit as a single center experience.

Methods: Between 2012 and 2023, 45 patients with PID received an allogeneic BMT at Acibadem Adana Hospital Pediatric Bone Marrow Transplantation Unit. Medical records, complications and treatment modalities were evaluated.

Results: A total of 545 allogeneic hematopoietic cell transplantations were performed at Acıbadem Adana Hospital Pediatric Bone Marrow Transplantation Unit in Turkey from 2013 to 2023. Forty-five patients underwent transplantation for primary immunodeficiency disorders. Two patients with CARD9 deficiency and XIAP deficiency underwent a second HSCT due to engraftment failure. The median age at transplantation was 5.7 years (range, 2 months to 17 years). There were 15 patients with severe combined immune deficiency, 6 patients with Kostmann syndrome, 5 patients with chronic granulomatous disease, 4 patients with Griscelli syndrome, 5 patients with Wiskott Aldrich syndrome, 5 patients with MHC Class II deficiency, 2 patients with DOCK 8 deficiency, one patient with XIAP deficiency, one patient with CARD9 deficiency, one patent with LRBA deficiency, one patient with ADA deficiency. Reduced-toxicity conditioning was applied to the patients. A two-month-old SCID patient was not given a conditioning regimen. Twenty seven patients received bone marrow stem cells; 20 patients received peripheral blood stem cells (PBSC). Sixteen patients had HSCT from an unrelated donor. Four patients had haploidentical transplantation from mother and father. Engraftment times were shorter with patients who underwent PBSC. Five patients died in the first 100 days due to severe GVHD and sepsis. The 5 year overall survival rate was found to be 80%.

Conclusions: HSCT has the potential of providing definitive correction for most PID. The recent advances in HSCT, including reduced toxicity conditioning agents and graft manipulation, have drastically improved the outcome of HSCT in PID. In the last 5 years, increased awareness, availability of diagnostics based on flow cytometry, genetic tests, improved supportive care, use of reduced toxicity conditioning, and success of haploidentical donor HSCT have improved access to HSCT for children with PID in Cukurova region, Turkey. The 5 year overall survival rate was found to be 80% in our center. We present results on children with PID who underwent HSCT in our center and the factors that influenced the outcome of these PID patients.

Disclosure: No.

25: Immunodeficiency Diseases and Macrophages

P378 THE OUTCOMES OF UMBILICAL CORD BLOOD STEM CELL TRANSPLANTATION FOR COMBINED IMMUNODEFICIENCY DISEASE IN CHILDREN WITH CD40LG MUTATION

Xiaowen Qian 1, Ping Wang1, Wenjin Jiang1, Xiaowen Zhai1

1Children’s Hospital of Fudan University, National Children’s Medical Center, Shanghai, China

Background: CD40 ligand (CD40L) deficiency is a combined immunodeficiency disease (CID) caused by CD40L gene (CD40LG) mutation, which is localized on the X chromosome, thus affecting mainly male individuals. CD40L was initially described to play an essential role during membrane–membrane interaction between activated CD4 + T cells and B cells, and CD4 + T and antigen-presenting cells (APCs). CD40L deficiency is a severe Inborn Errors of Immunity (IEI) characterized by susceptibility to life-threatening infections. We report the results of UCBT performed in 10 CID patients with CD40LG mutation between 2018 and 2022 at a single center.

Methods: Retrospective analysis was performed on consecutive children diagnosed as CID with CD40LG mutation in our center from January 2018 to December 2022. All patients received a myeloablative conditioning regimen consisting of busulfan, fludarabine and cyclophosphamide, and prophylaxis for graft-versus-host disease (GVHD) was tacrolimus only. The clinical and laboratory data before and after umbilical cord blood stem cell transplantation were evaluated.

Results: en children with CD40LG mutation receiving UCBT were included in the analysis. All the children are boys, median age at transplantation was 14 months (range, 6 to 100 months), median body weight was 10.5 kg (range, 7.7 to 23.2 kg). The median of total nucleated cells infusion amount was 9.68x107/kg (5.93~17.87x107/kg), CD34+ median amount is 3.28x105/kg (1.72~10.7 x105/kg). Among the 10 boys, 8 were achieved complete engraftment, the median time to neutrophil and platelet engraftment were 18.5 (rang, 17-52) and 33 (rang, 17-68) days. At a median follow-up of 33 (rang,1-66) months, 2 children died on early transplantation caused by infection, 8 children survived without disease and obtained immune reconstruction. 2 children occured graft failure, one of them successfully received a second unrelated donor SCT 4 months later and alive. 4 children developed acute GVHD, with 2 episodes of grade III-IV aGVHD. Chronic GVHD (skin) occurred in 1 patient.

Conclusions: Unrelated UCBT should be considered as potential curative methods in children with CD40LG mutation.

Clinical Trial Registry: None.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P379 BACILLE CALMETTE-GUÉRIN (BCG) VACCINE-ASSOCIATED COMPLICATIONS FOLLOWING STEM CELL TRANSPLANTATION IN PATIENTS WITH INBORN ERRORS OF IMMUNITY (IEI) – SINGLE TERTIARY CENTRE EXPERIENCE FROM SOUTH INDIA

Stalin Ramprakash 1, C P. Raghuram1, P. Anoop1, Neha Singh1, Jyothi Janardhanan1, Sagar Bhattad1

1Aster CMI hospital, Bangalore, India

Background: Disseminated BCG infection is a potential life threatening complication in countries where universal BCG vaccination at birth is the norm, but routine newborn screening programme for inborn errors of immunity does not exist. We studied the clinical profile of BCG vaccine related complications in children who received BCG vaccination and undergone stem cell transplantation for Inborn errors of Immunity in such a setting.

Methods: Case records of children less than 18 years of age who underwent stem cell transplant for Inborn errors of Immunity in our centre between November 2017 to October 2022 were retrospectively reviewed. All patients had received BCG vaccination at birth and had at least 1 year post-transplant follow up period at time of analysis. Indications for transplant and details BCG related complications and treatment were analysed.

Results: Out of 35 children transplanted for inborn errors in immunity 8 children (23%) developed BCG related complications. Among the 8 patients who developed BCG related complications 4 had disseminated BCG infection (BCGiosis) which was diagnosed prior to transplant and all of them were diagnosed to have T-B+ Severe combined immunodeficiency(SCID). Two of those patients were NK+ and the remaining 2 were NK-ve. Out of these 4 patients with disseminated BCG one died as direct consequence of disseminated BCG, as the infection progressed in spite of antituberculosis treatment in the immediate post-transplant period and another succumbed to severe GVHD. Among the two survivors one patient needed ATT until day 659 post-transplant as an earlier attempt to stop ATT resulted in reactivation of disseminated BGG and the other child is still on ongoing ATT and off all immunosuppression on day +230 post-transplant. Among the 4 patients with local BCG infection (BCGitis) 3 were diagnosed with Chronic Granulomatous disease (CGD) and remaining one was confirmed as RAG2 SCID. None of those localised BCG progressed to disseminated BCG post transplantation. Three patients (2 with disseminated BCG and 1 with local infection with RAG2 SCID developed) BCG- Immune reconstitution Inflammatory Syndrome (BCG-IRIS). One patient received low dose steroids to control the inflammation and other two were treated with IL-6 blockade (Tocilizumab) and all of them fully recovered with treatment.

Conclusions: BCG associated complication are often encountered during stem cell transplantation of children with IEI especially among patients with SCID and CGD. Disseminated BCG may need prolonged treatment to achieve complete control of the infection post-transplant. Prompt recognition of BCG-IRIS and distinguishing it from disseminated-BCG is crucial in achieving optimal outcomes.

Clinical Trial Registry: Retrospective review.

Disclosure: “Nothing to declare”.

25: Immunodeficiency Diseases and Macrophages

P380 UNSUCCESSFUL HSCT IN 2 ADA2 DEFICIENCY PATIENTS ASSOCIATED WITH HIGH LEVELS OF ANTI-HLA ANTIBODIES

Amit Dotan 1, Marganit Benish1, Hila Rosenfeld Keidar1, David Hagin1, Sabina Edelman1, Ronit Elhasid1

1Tel Aviv Sourasky Medical Center, Tel Aviv, Israel

Background: Deficiency of adenosine deaminase 2 (DADA2) is a recessively inherited disease characterized by systemic vasculitis, early-onset stroke, bone marrow failure, and/or immunodeficiency. Based on 30 published cases, hematopoietic stem cell transplantation (HSCT) represents a potential curative treatment for DADA2, with 2-year overall survival of 97%. However, the actual outcome might be different, especially in the presence of anti-HLA antibodies as their occurrence in DADA2 and their contribution to graft rejection have not been described.

Methods: A retrospective analysis of the outcomes of allogeneic HSCT in 2 children with DADA2 and anti-HLA antibodies.

Results: Case 1: A 3-year-old girl was referred for pure red cell aplasia (PRCA) and was diagnosed with DADA2. Pre-transplant evaluation showed high levels of anti-HLA antibodies, with mean fluorescence intensity (MFI) of up to 15,900 for anti-HLA class I, and up to 4500 for anti-HLA class II, including donor-specific anti-HLA antibodies (DSA) with MFI of 11,400. As there was no alternative donor available, she underwent desensitization using bortezomib and intravenous immunoglobulins (IVIG). This resulted in disappearance of DSA, but no change in levels of the non-DSA anti-HLA antibodies. Following conditioning with busulfan, fludarabine and ATG, she underwent peripheral blood HSCT from a 9/10 matched unrelated donor. Shortly after neutrophil engraftment (day 15) a decline in donor chimerism was observed (from 94% to 70% donor cells) in parallel to pancytopenia. Repeated anti-HLA antibody testing remained mostly unchanged with no DSA detected but similar high levels of non-DSA anti-HLA antibodies. Pre-collected autologous stem cells were then administered with autologous neutrophil engraftment 12 days post-infusion.

Case 2: A 1.5-year-old boy was referred for PRCA and myelodysplastic syndrome and was eventually diagnosed with DADA2. Multiple anti-HLA antibodies were identified, with an MFI of up to 17,300 for anti-HLA class-I and up to 18,700 for anti-HLA class-II, including anti-potential-donor DSA. Therefore, a matched related cord blood was chosen as an alternative stem-cell source. The conditioning regimen included busulfan, cyclophosphamide and melphalan. Here too, Despite the absence of anti-cord DSA a desensitization protocol with bortezomib, IVIG and plasmapheresis followed by rituximab and IVIG was given. However, this resulted in non-significant reduction in anti-HLA antibodies levels. Unfortunately, transplant resulted in primary graft failure, and he was eventually infused with pre-collected autologous stem cells, with neutrophil engraftment 10 days later.

Conclusions: Multiple high-level anti-HLA antibodies, as observed in these 2 DADA2 cases, are rare in children, and may reflect DADA2-related immune dysregulation, resulting in increased sensitivity to foreign HLA exposure and overproduction of anti-HLA antibodies. Thus, testing for anti-HLA antibodies should be considered for DADA2 patients who are candidates for HSCT.

In addition, graft failure in the presence of anti-HLA antibodies, despite no DSA and without overt alternative etiology, suggest that anti-HLA antibodies may still play a role in the rejection process, probably through targeting mismatched antigens not routinely tested.

Collecting autologous stem cells as backup for graft failure in the absence of spontaneous autologous marrow recovery may serve as a life saving measure at such high-risk situations.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P381 FATAL NEUROLOGICAL COMPLICATION POST HSCT IN A PEDIATRIC PATIENT WITH IMMUNODEFICIENCY DUE TO NFKB2 MUTATION

Paula Catalán 1,2, Cristián Sotomayor2, Cecilia Poli3,4, María Angélica Wietstruck1,2

1Pontificia Universidad Católica de Chile, Santiago, Chile, 2Hospital Clínico Universidad Católica Red Salud UC Christus, Santiago, Chile, 3Hospital Roberto del Río, Santiago, Chile, 4Universidad del Desarrollo - Clínica Alemana, Santiago, Chile

Background: In the era of genetic testing, inborn errors of immunity and their HSCT timing became a challenging decision considering the post HSCT evolution and complications.

Methods: Case report.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (44)

Results: Nine y/o boy with a history of retinal vasculitis and hypogammaglobulinemia diagnosed at 7y/o. A complete study was performed to rule out infections and autoimmune diseases, EBV CMV were negative. INVITAE testing report: “One Pathogenic variant identified in NFKB2. NFKB2 is associated with autosomal dominant DAVID syndrome and CVID. One Pathogenic variant identified in VPS13B. VPS13B is associated with autosomal recessive Cohen syndrome”.

He only presented isolated warts with good response to topical treatment. No history of major infections nor autoimmunity. He was under weekly methotrexate (15 mg/m2), IVGG (500 mg/kg) every 5 weeks and periodic ophthalmology visits. He has not required topical treatment in the last 8 months (RE atrophic macula, LE occlusive vasculitis, peripheral retinal scars).

The HSCT indication was assessed considering the evidence of functional NK deficiency in the setting of associated disease with a confirmed pathogenic NFKB2 mutation.

Pre-HSCT work-up: CMV-, O-, Lansky score 90%.

No MSD nor 10/10 donor in the WDMA unrelated search. The selected donor was the only 9/10 available (HLA-A MM), permissive DP, M, CMV-, ABO incompatible (B + ).

Conditioning regimen: Melphalan (140 mg/m2), fludarabine (160 mg/m2) and TT (10mg/kg).

GvHD prophylaxis: Alemtuzumab, CNI and short MTX schedule.

Engraftment: WBC d + 26, platelets d + 26, RBC d + 32, Chimerism > 95% d + 30.

He was discharged on day+43, he required periodic G-CSF and weekly transfusions.

He was readmitted because of febrile neutropenia, low inflammatory parameters, no microbiological isolation, persistent fever, he received methylprednisolone 2 mg/kg/day, since day+105.

On day +108, he presented 6th nerve palsy. MRI: Alteration of the left lenticular signal with striatocapsular vasogenic edema with thalamus-mesencephalic extension. Infectious disease ruled out in CSF, BMA and blood. He remained under wide spectrum antimicrobial treatment, Cyclosporin A withdrawal.

One week later, with neurological impairment and chorioretinitis.

New MRI: “progression of the left lenticular signal with major involvement of the brainstem and subcortical insular region, multiple lesions enhanced by contrast”. Spinal MRI OK.

He underwent a brain biopsy: microglia proliferation, perivascular lymphocytic infiltrate, inflammatory pattern. Negative IHC and ID tests (viral, fungal, parasitic). He started ruxolitinib and Anakinra.

He evolved with progressive neurological impairment, no evidence of recovery, with decreasing chimerism.

He died 5 months post-transplant (day+152).

Conclusions: NFBK2 expression is not limited to HSC. This PID is a rare disease, with limited experience in HSCT. It was proposed as a curative treatment, but with risk of graft failure.

We report this case for analysis of the outcome and complications.

Disclosure: Nothimg to declare.

25: Immunodeficiency Diseases and Macrophages

P382 ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) TO CHILDREN WITH PRIMARY IMMUNODEFICIENCY –THE HELLENIC EXPERIENCE

Eleni Ioannidou 1, Aikaterini Kaissari1, Anna Paissiou1, Christina Oikonomopoulou1, Anna Komitopoulou1, Michalis Kastamoulas1, Georgia Stavroulaki1, Ioannis Grafakos1, Vassiliki Kitra1, Ioulia Peristeri1, Stylianos Grafakos1, Georgios Vessalas1, Maria Theodosaki1, Eftychia Petrakou1, Irene Sfougataki1, Dimitra Kattemi1, Alexandra Papasarantopoulou1, Evangelia Farmaki2, Rediona Cane3, Marianna Tzanoudaki3, Evgenios Goussetis1

1Stem Cell Transplantation and Cellular Therapies Unit, “Marianna V. Vardinogianni – Elpida” Pediatric Oncology Center, “Agia Sofia” Children Hospital, Athens, Greece, 2“Hippocration” Hospital of Thessaloniki, Thessaloniki, Greece, 3“Agia Sofia” Children Hospital, Athens, Greece

Background: Primary immunodeficiencies (PIDs) represent a wide and heterogeneous group of diseases affecting the immune system and causing infections, autoimmunity, lymphohyperplasia, hyperinflammation or malignancies. HSCT can offer long term cure to patients suffering from PID.

Methods: Patients diagnosed with Primary Immunodeficiency transplanted from 1994 to 2022 were included in the study.

Results: 81 children with PID (median age 3,6 ys – range 0,1-16,9 ys) have been transplanted at our center. The diagnosis was Severe Combined Immunodeficiency (SCID) (n=38), Wiscott Aldrich (n=15), Chronic Granulomatous Disease (n=12), other (n=16). The donor was matched relative donor (n=30), voluntary unrelated donor (VUD) (n=39) and haploidentical donor (n=12). The conditioning regimen was Busulfan based in 44 cases combined with Cyclophosphamide (n=29) or Fludarabine (n=15) and Treosulfan (42g/m2 or 36g/m2 if <1y old) combined with Fludarabine in 31 cases. The remaining cases did not receive conditioning (n=3) or received other regimens (n=3). Graft source was bone marrow (n=46), peripheral graft (n=27), cord blood (n=8).

The Overall Survival (OS) rate was 80% and Event Free Survival (EFS) probability where event was defined as death or second procedure was 67% in the entire cohort. OS and EFS for SCID patients were 61% and 93% respectively. OS and EFS for non-SCID patients were 86% and 82% respectively.

Transplantation Related Mortality (day 100) was 8%. The majority of deaths were due to infection (46%) and GVHD (15%). OS probability was significantly higher for children transplanted after 2000 (78% vs 44% p=0,022) whereas conditioning, age at transplantation, donor type or graft source did not affect survival in multivariate model analysis. Eight patients (SCID n=1, non-SCID=7) rejected the graft and underwent second HSCT.

Twenty percent of SCID patients conditioned with Busulfan achieved stable full donor chimerism and 5% rejected the graft whereas 50% of SCID patients conditioned with Treosulfan achieved stable full whole blood donor chimerism and none rejected the graft.

Seventy five percent of non-SCID patients conditioned with Busulfan achieved stable full donor chimerism and 5% rejected the graft whereas 26% of non-SCID patients conditioned with Treosulfan achieved stable full whole blood chimerism and 31% rejected the graft.

Cumulative incidence (CI) of second transplant was higher for non-SCID patients conditioned with Treosulfan (HR 14,5, 95% CI 3,2-65,3). Eleven patients presented VOD, all of them were conditioned with Busulfan and 2 of them died from VOD. Acute Graft Versus Host Disease (GVHD) presented 34% of the patients but only 10% had grade III-IV aGVHD.

Conclusions: Transplantation offers cure to the majority of patients with PID. As Treosulfan was associated with higher possibility of rejection in non-SCID whereas, Busulfan had higher toxicity, a tailored approach should be adopted for the selection of the conditioning in each particular variant of PID.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P383 POSTTRANSPLANT COMPLICATIONS IN A CASE OF LATE-ONSET FAMILIAL HEMOPHAGOCYTIC LYMPHOHISTIOCYTOSIS TYPE 2

Delfien Bogaert 1, Siel Daelemans2, Tessa Wassenberg2, Filomeen Haerynck1, Victoria Bordon1, Catharina Dhooge1

1Ghent University Hospital, Ghent, Belgium, 2University Hospital Brussels, Brussels, Belgium

Background: Hemophagocytic lymphohistiocytosis (HLH) is a severe hyperinflammatory condition caused by excessive release of pro-inflammatory cytokines. Central nervous system (CNS) involvement is often seen. HLH can be life-threatening due to rapid progression to multi-organ failure. The term primary HLH is used when there is a genetic predisposition, such as familial HLH (FHL) types 1-5. The initial treatment of HLH is aimed at damping the cytokine storm. However, the only curative treatment for primary HLH is allogeneic hematopoietic stem cell transplantation (HSCT).

Methods: We report the posttransplant complications in a patient with FHL type 2.

Results: The patient is a girl of 12 years old born to non-consanguineous parents of Central-African origin. At the age of 11, she had two episodes of encephalitis presenting as headache, vertigo and altered consciousness. Acute disseminated encephalomyelitis (ADEM) was suspected. She was treated with high-dose prednisolone. At the age of 12, she presented again with encephalitis associated with fever, dyspnea and cytopenia. High-dose prednisolone was given and additional diagnostics were performed. Genetic testing revealed a compound heterozygous mutations in PRF1. This confirmed the diagnosis of perforin deficiency or FHL type 2 (FHL2). FHL2 is known to result in recurrent and refractory HLH, typically with CNS involvement. Therefore, the previous episodes of encephalitis were retrospectively diagnosed as central HLH. Diagnosis of FHL2 is an indication for HSCT, even in late-onset cases, because there is a high risk of progressive disease.

The patient was transferred to our center for further treatment. Immunosuppression was switched to dexamethasone and cyclosporine in accordance with the HLH-2004 protocol. A donor search was initiated. During that time, she suffered from a CMV infection for which she was treated with ganciclovir. Both her brothers were HLA-identical; since one of them carried the PRF1 mutation, the PRF1 wild-type brother was selected as stem cell donor. Myeloablative conditioning with Busulfan (AUC 85-95), Fludarabine (160 mg/m²) and ATG (10 mg/kg) was given. The immediate posttransplant period was uncomplicated. Neutrophil engraftment was seen at D + 26, donor chimerism was >95%. There was no CMV reactivation, and no signs of graft versus host disease (prophylaxis with cyclosporine and methotrexate). At 3 months posttransplant, she developed immune-mediated thrombocytopenia and anemia (positive anti-HPA antibodies, positive Coombs); cyclosporine was switch to sirolimus. At 5 months posttransplant, she showed progressive cytopenias with a hypocellular bone marrow, despite > 95% donor chimerism. Immune-mediated graft rejection was suspected and high-dose prednisolone was started. At 6 months posttransplant, she developed fever and tremor and had elevated levels of CRP and ferritine. HLH was suspected, immunosuppression was switched to dexamethasone and cyclosporine. Lumbar puncture was normal. Infectious work-up was negative. She quickly deteriorated with a severe intracerebral hemorrhage and epileptic seizures. ATG was associated. Unfortunately, the intracerebral hemorrhage expanded and she developed a gram-negative sepsis. She deceased 6,5 months posttransplant.

Conclusions: We reported a patient with late-onset FHL2 mainly suffering from central HLH, that underwent HSCT. Six months posttransplant she died from hemorrhagic cerebral HLH. Severe posttransplant inflammatory complications remain a clinical challenge in primary HLH.

Clinical Trial Registry: not applicable.

Disclosure: Nothing to declare.

25: Immunodeficiency Diseases and Macrophages

P384 HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PRIMARY IMMUNODEFICIENCY DISEASES: SINGLE CENTRE EXPERIENCE

Nayera El-Sherif 1, Heba G.A. Ali1, Sara Makkeyah1, Safa Sayed1, Nesrine Radwan2, Dalia ElGhoneimy2, Fatma S.E. Ebeid1, Sally Gouda2

1Ain Shams University, Pediatric Hematology/Oncology & BMT Unit, Cairo, Egypt, 2Ain Shams University, Pediatric Allergy, Immunology and Rheumatology, Cairo, Egypt

Background: Outcome of Hematopoietic stem cell transplantation (HSCT) for primary immune deficiency (PID) has significantly improved with the increased use of alternative donors, the new stem cell sources, and the use of less toxic, reduced-intensity conditioning (RIC) regimens as well as the graft manipulations techniques.

Methods: Our Goal is to describe our one-year experience in starting HSCT program of PID patients at Ain Shams University in Cairo, Egypt, balancing high quality concurrently with lowering the costs.

Methods: A case series of four patients (3 males and one female) over a period of one year from January 2023 to December 2023 with confirmed diagnosis of PID using international criteria, all patients were provided a written consent as per institutional practice for allogeneic-HSCT, their mean age at the time of HSCT was 19 months±9.6 (range 9-32 months); their diagnosis were as follow: Three patients had severe combined immunodeficiency (SCID) and one diagnosed with chronic granulomatous disease (CGD). They all had matched related donors; two SCID patients matched with their mothers, while one SCID and one CGD patients matched with their older brothers. All received Fludarabine-Busulfan (Flu/Bu)/rabbit ATG as conditioning with cyclosporine (CSA) and methotrexate as GVHD prophylaxis. One SCID patient had transplacental maternal engraftment and autoimmune haemolytic anaemia for which he received 2 doses of rituximab pretransplant. Another SCID patient was on prophylactic antituberculosis treatment for previous BCG vaccine.

Results: Conditioning was generally well tolerated. Neutrophil engraftment occurred around day 12-14 post-transplant. All patients had achieved initial full donor chimerism; two SCID patients converted to mixed chimerism by day 90. All SCID patients were maintained on IVIG replacement. Dihydrorhodamine assay of CGD patient remained normal at day +180 post-transplant. One patient developed grades I-II acute skin GVHD. Another SCID patient had several CMV reactivation; he died after 5 months post-transplant of sepsis and multiorgan failure. BCGitis occurred in the third SCID patient after 3 months of transplant and resolved with quadruple anti-tuberculous treatment.

Conclusions: Although, starting HSCT program of PID patients was challenging, yet the integration of knowledge between immunologists and transplant specialists and the individualized plans based on the characteristics of every patient made it successful.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P385 HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR INBORN ERRORS OF IMMUNITY (IEI): GAP BETWEEN NEED AND REALITY IN A LIMITED RESOURCE SETTING: A FIVE-YEAR SINGLE-CENTER EXPERIENCE

Stalin Ramprakash 1, Neha Singh1, Jyothi Janardhanan1, Harish Kumar1, Chetan Ginegeri1, C P. Raghuram1, P. Anoop1, Sagar Bhattad1

1Aster CMI Hospital, Bangalore, India

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) plays a pivotal role as a curative intervention for a spectrum of inborn errors of immunity (IEI). Despite its proven efficacy, the reality persists that not all eligible patients are able to avail this therapy. We present a comprehensive review of our institution’s five-year experience, focusing on patients with diverse IEI who, although deemed suitable candidates for allogeneic HSCT, failed to undergo HSCT. We decided to study the clinical profile and outcomes of patients with IEI where HSCT was indicated as a curative treatment but they could not undergo HSCT.

Methods: Four hundred and fifteen patients with various inborn errors of immunity (IEIs) were diagnosed in the Pediatric Immunology and Transplant Unit of our hospital during the study period of February 2017 to November 2023. Based on the clinical, immunological, and genetic results, 208 patients were found to be eligible for HSCT as per EBMT/ESID inborn errors working party guidelines. Out of 208 patients, 51 (46 patients at our center and five patients elsewhere) underwent or are undergoing HSCT (the majority received some financial assistance through various sources) and hence were excluded from the study. The profile of the remaining 157 patients was analyzed in detail.

Results: We had 157 patients, which included 52 females and 105 males in our cohort. Out of 157 patients, the largest number had severe combined immunodeficiency (n = 37), followed by chronic granulomatous disease (n = 31), primary immune regulatory defect (n = 18), and DOCK8 deficiency (n = 17). The most common reasons for failure to undergo HSCT were parental refusal (n = 82), clinical deterioration and death of patients awaiting donor search and transplant (n = 48), and delay in crowdfunding or financial constraints (n = 27). The reluctance of parents to proceed with HSCT was multifactorial, with financial burden and risk-benefit considerations playing a significant role. Overall mortality in the non-transplant group was 76/157 (48.4%), while 10 patients (6.3%) were lost to follow-up, which may mean that those families may have abandoned treatment. One patient with Wiskott-Aldrich syndrome developed B-cell lymphoma and was transferred to the hematology-oncology unit. Presently, 70 patients (44.6%) are under follow-up with ongoing medical problems such as recurrent infections and autoimmunity.

Diagnosis of transplant eligible patients who did not proceed to transplant

Severe Combined Immunodeficiency (SCID)

37

Chronic Granulomatous Disease. (CGD)

31

Primary Immune Regulatory disorders (PIRD)

18

DOCK8 deficiency

17

Mendelian Susceptibility to Mycobacterial Disease (MSMD)

14

Leukocyte Adhesion Defect (LAD)

11

Wiskott-Aldrich syndrome (WAS)

8

Activated PI3 kinase delta syndrome (APDS)

5

CARMIL2 Defect

4

Hemophagocytic lymphohistiocytosis (HLH)

3

Hyper IgM syndromes (HIGM)

3

C1q deficiency

3

Bone marrow failure (category IX inborn errors of immunity)

3

Conclusions: The high overall mortality rate of 48.4% in the non-transplant group highlights the importance of timely intervention and the challenges of managing patients with IEI when transplantation is not feasible. Furthermore, 6.3% of patients were lost to follow-up, emphasizing the need for robust support systems and continuity of care. The findings from this study underscore the importance of a comprehensive approach involving healthcare professionals, support services, and public awareness campaigns to facilitate timely HSCT and improve outcomes for patients with IEI.

Clinical Trial Registry: Retrospective study.

Disclosure: “Nothing to declare”.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P386 AN INNOVATIVE PLATFORM APPROACH FOR THE DEVELOPMENT OF EX-VIVO HEMATOPOIETIC STEM AND PROGENITOR CELL-GENE THERAPY FOR THE TREATMENT OF LYSOSOMAL STORAGE DISEASES WITH SKELETAL INVOLVEMENT

Stefania Crippa1, Pamela Quaranta1, Margherita Berti1, Luca Basso-Ricci1, Ludovica Santi1, Giada De Ponti1, Raisa Jofra Hernandez1, Claudia Forni1, Ilaria Visigalli1, Paola Albertini1, Rossella Parini1, Serena Scala1, Alessandro Aiuti2, Maria Ester Bernardo 2

1San Raffaele Telethon Institute for Gene Therapy, Milano, Italy, 2IRCCS San Raffaele Scientific Institute, Milano, Italy

Background: Lysosomal storage diseases (LSDs) are a group of genetic diseases caused by lysosomal dysfunction leading to the accumulation of undegraded substrates in different organs, including nervous and skeletal systems, which are not fully addressed by currently approved treatments. Previous data obtained by our Institute proved the superior safety and efficacy of Hematopoietic Stem and Progenitor Cell-Gene Therapy (HSPC-GT) for the treatment of LSD-skeletal and neurological symptoms. Building on these experiences and the similar features of LSDs, we are developing a platform approach of HSPC-GT for the treatment of LSDs characterized by severe skeletal involvement. In this framework, chemistry, manufacturing and controls, non-clinical and clinical development plans have been optimized to generate a platform dataset complemented by specific disease data aimed at launching a single combined clinical trial, where the selected products are simultaneously tested in all the platform diseases.

Methods: We generated 3rd-generation lentiviral vectors (LVs) encoding for each disease-specific enzyme (LV-GALNS, -GLB1, and -MAN2B1). We texted the LV-related toxicity and the transduction efficiency in human HSPCs from healthy donors transduced with a single hit of each LV using CyclosporinH as a transduction enhancer. Untransduced (UT) cells were used as controls. We evaluated in vitro toxicity as proliferation and clonogenic capacity. We determined the transduction efficiency as vector copy number (VCN) by digital droplet PCR and enzymatic activity. We further study the biodistribution of transduced cells into transplantation models to exclude LV toxicity in vivo.

Results: We performed a horizon scanning of 13 LSDs and selected Mucopolysaccharidosis IVA, Mucopolysaccharidosis IVB and Alpha-Mannosidosis as platform diseases. Human HSPCs transduced with the different LVs showed a proper clonogenic capacity and proliferated similarly to UT cells when expanded as myeloid liquid culture. We measured a similar mean VCN in myeloid cells and at the single colony level upon transduction with the different LVs. Transduced cells significantly overexpressed the therapeutic enzymes at the intracellular level, while we measured a low level of the enzymatic activity in the extracellular space of LV-GLB1 and -MAN2B1 transduced cells. We optimized the LV-GLB1 transgene sequence and the transduction protocol for LV-MAN2B1 to improve the extracellular release and favor the cross-correction of diseased cells. Importantly, all transduced cells differentiated into osteoclasts releasing supraphysiological level of therapeutic enzyme, possibly serving as circulating and tissue-resident source of enzymes. The medium conditioned by myeloid cells and osteoclasts derived transduced HSPCs restored the enzymatic activity in patients’ fibroblasts, underlying the capacity of transduced cells to cross-correct affected non-hematopoietic cells. Finally, we studied the toxicity of LV-GALNS and -GLB1 in humanized models of transplantation. Transduced HSPCs engrafted and reconstituted the hematopoietic organs similarly to UT cells, demonstrating that the LVs did not impact on HSPCs functionality in vivo. Importantly, the level of enzymatic activity was higher in the bone marrow of mice transplanted with transduced cells compared to control mice.

Conclusions: Our preliminary results strongly support the HSPC-GT platform for LSDs, moving from one single to simultaneous multi-disease development, saving time and costs and making GT more sustainable for ultra-rare diseases.

Disclosure: None.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P387 HEMATOPOETIC STEM CELL TRANSPLANTATION WITH CURRENT PERSPECTIVE IS SAFE FOR MUCOPOLYSACCHARIDOSIS TYPE VI PATIENTS

Vedat Uygun1, Gülsün Karasu2, Koray Yalçin 3,4, Seda Öztürkmen5, Safiye Suna Çelen3, Hayriye Daloğlu6, Suleimen Zhumatayev2, Akif Yesilipek5

1Istinye University, Istanbul, Turkey, 2MedicalPark Göztepe Hospital, Istanbul, Turkey, 3Bahcesehir University, Istanbul, Turkey, 4Acıbadem University, Istanbul, Turkey, 5MedicalPark Antalya Hospital, Antalya, Turkey, 6Antalya Bilim University, Antalya, Turkey

Background: It is uncertain whether hematopoetic stem cell transplantation (HSCT) would be a successful therapy for MPS VI instead of enzyme replacement therapy (ERT). The reason for avoiding HSCT is the toxicity of the conditioning regimen and subsequent complications. Most of the data about HSCT is based on small number of case studies, the largest of including patients transplanted mostly before 2000, and resulted in a relatively high rate of GVHD and mortality. This highlights the necessity of transplant procedures carried out in a recent cohort in light of new advances and less toxic agents.

Methods: This was a single-center retrospective review of 17 pediatric MPS VI patients undergoing allogeneic HSCT between November 2021 and July 2023. All conditioning regimens were myeloablative, comprising busulfan in weight-based doses or treosulfan based on body surface area. Fludarabine was used in all patients at a dose of 150 mg/m2. If available, thiotepa was used at a dose of 10 mg/kg. All patients were administered a calcineurin inhibitor in combination with a short course of methotrexate for GVHD prophylaxis. Antithymocyte globulin was administered in the pre-transplantation period for all patients. Engraftment days, complications, GVHD, and survival data were recorded. As the follow-up was short for showing physical improvement, we only recorded the change in the 6-minute walk distance test (6-MWT) before and after HSCT.

Results: Patient and transplant characteristics are summarized in Table 1. Patients were transplanted median 6 years after diagnosis, mostly with a matched unrelated donor. All patients were engrafted and had full or mixed chimerism at the last follow-up. Twelve patients had their enzyme levels assessed at six months following HSCT, and all were within normal ranges. Only one patient experienced high-grade acute GVHD, and no patient had moderate or severe chronic GVHD. The most frequent complication was CMV viremia and, to a lesser extent, engraftment syndrome, autoimmunity, and hemorrhagic cystitis. All of the complications, including GVHD, were resolved after treatment, excluding one patient’s autoimmune hemolytic anemia. Because of the lack of regular multi-disciplinary follow-up of patients before HSCT and refusal to do the test after HSCT, only 9 patients performed the 6-MWT for comparison, and it was observed that all patients’ test performance was improved in a median 9-month follow-up.

Conclusions: HSCT is not generally considered an alternative treatment in patients with MPS VI, especially because enzyme therapy is already known to be beneficial and toxicity may be high due to cardiac and respiratory problems. However, as all our patients were alive on the last follow-up, high complications such as GVHD and mortality in previous studies were not observed in our study, and adequate enzyme levels were achieved without enzyme replacement therapy after transplantation. The walking tests of all evaluable patients showed significant improvements over pre-HSCT results.

Our conditioning regimen, which did not consist of cyclophosphamide and TBI, contrary to previous reports, is a promising regimen that has a very low toxicity profile. The effect of HSCT on clinical improvement should be observed in further follow-up.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P388 HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM MATCHED AND MISMATCHED DONORS WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE AND ANTITHYMOCYTE GLOBULIN IN CHILDREN WITH PRIMARY IMMUNODEFICIENCIES

Alexandr Bazaev1, Alexandra Laberko 1,2, Yulia Skvortsova1, Elena Gutovskaya1, Svetlana Kozlovskaya1, Anna Vasilieva1, Larisa Shelikhova1, Galina Novichkova1, Alexey Maschan1, Michael Maschan1, Dmitry Balashov1

1Dmitriy Rogachev National Medical Research Center for Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation, 2Raisa Gorbacheva Memorial Research Institute of Children Oncology, Hematology and Transplantation, First Pavlov State Medical University, St. Petersburg, Russian Federation

Background: Hematopoietic stem cell transplantation (HSCT) is widely used in primary immunodeficiencies (PID), and risks of graft-versus-host disease (GVHD) remain high. Use of post-transplant cyclophosphamide (PTCY) for GVHD prophylaxis revolutionized the outcomes of HSCT from mismatched related donor (MMRD). Another well-known option for GVHD prevention is antithymocyte globulin. Here we analyzed the results of HSCT with PTCY/ thymoglobulin in PID who received HSCT from different types of donors.

Methods: PTCY was used in 30 children with PID (SCID-7, WAS-5, SCN-4, CGD-3, others PID-11) who received HSCT from 2022 to 2023 in our center. The median age at HSCT was 1.35 years (range 0.13-15.4). In 7 patients matched related donors (MRD), in 12 matched unrelated donors (MUD) and in 11 MMRD were used.

All patients received treosulfan 30-42g/m2 and fludarabine 150mg/m2, 25 patients additionally received melphalan 80-140mg/m2 (n=14), thiotepa 10mg/kg (n=3) or cyclophosphamide 30-100mg/kg (n=8). In 8 patients (WAS-5, SCN-2, CGD-1), conditioning included plerixafor 720mcg/kg. All patients received thymoglobulin (Genzyme) 5-7mg/kg (days -6, 5, 4). In all patients, bone marrow was used as a stem cell source (the median of NC was 6,25x108/kg, CD34 + 8,22x106/kg). PTCY was given at +3, 4 days at dose 25 mg/kg/d in MUD/ MRD and 50 mg/kg/d in MMRD.

For GVHD prophylaxis 6 patients received calcineurin inhibitor as a monotherapy (MUD 1, MRD 2, MMRD 3) and 1 in combination with methotrexate/mycophenolate mofetil (MUD), 25 ruxolitinib (24 7-10 mg/m2 and 1 20 mg/m2 (MUD 10, MRD 6, MMRD 9). Nine patients additionally received abatacept 10 mg/kg (days +7, 14, 28, 45, 60).

Results: The median follow–up was 0.64 years (range 0.11-1.3). Engraftment was reached in 28 (93%). The median time of neutrophil engraftment was 23.5 days (range 16-35), platelet 19 days (range 11-86). Cumulative incidence of graft failure (GF) (non-engraftment in 2 and graft rejection in 2) was 0.14 (95%CI 0.05-0.36). Among the patients with GF (WAS-1, SCID-1, SCN-1, SDS-1), 1 died, 3 received second allogeneic HSCT.

Acute GVHD was seen in 11 (36%) patients: 8 grade I-II (MUD 2, MRD 3, MMRD 3), 3 grade III-IV (MUD 1, MMRD 2). Incidence of acute GVHD was 0.42 (95%CI 0.27-0.65), with no difference between different donors: 0.5 (95%CI 0.26-0.95) in MMRD, 0.43 (95%CI 0.18-1) in MRD and 0.33 (95%CI 0.15-0.74) in MUD, p=0.6. Three patients developed chronic GVHD (MMRD 2, MRD1), the incidence was 0,18 (95%CI 0,06-0,5). Risk of CMV reactivation was 0.38 (95%CI 0.24-0.61), 6 patients developed CMV disease (4 pneumonia, 1 retinitis, 1 gastritis). No other serious viral infections, requiring therapy, were observed.

Liver venocclusive disease was observed in 4 (13%) patients. All patients received defibrotide with complete resolution in 3 (1 died of infections).

Overall survival was 0.86 (95%CI 0.74-0.99). Four patients died (3 SCID from infections, 1 Schwachman-Diamond syndrome and MDS had rejection of 2 grafts and died of infections post-third HSCT).

Conclusions: PTCY in combination with serotherapy is a safe and effective method of GVHD prophylaxis in patients with PID. Importantly, no difference in GVHD incidence and severity was seen between MUD, MSD and MMRD.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P389 CONDITIONING FOR SECOND HSCTS FROM THE SAME DONOR AFTER MYELOID AUTOLOGOUS RECONSTITUTION IN PATIENTS WITH IEI AND THALASSEMIA: IMMUNOSUPPRESSION IS NOT ALWAYS REQUIRED

Sarah Bauer1, Beatrice Zwiebler1, Michael H. Albert2, Jörn-Sven Kühl3, Mehtap Sirin1, Kerstin Felgentreff1, Ingrid Furlan1, Klaus-Michael Debatin1, Eva-Maria Jacobsen1, Ansgar Schulz1, Manfred Hoenig 1

1University Medical Center Ulm, Ulm, Germany, 2Dr. von Hauner Children’s Hospital, LMU Munich, Munich, Germany, 3University Hospital Leipzig, Leipzig, Germany

Background: Autologous reconstitution of the myeloid lineage after allogeneic HSCT in patients with Inborn Errors of Immunity (IEI) and thalassemia can be associated with incomplete immunological reconstitution or recurrence of the primary disease and the need for second HSCT. T cells -with their ability of peripheral expansion- frequently remain at least partially of donor origin for prolonged periods of time. Ideally, conditioning before a second HSCT from the same donor in this situation should preserve established donor T-cell function, cause limited toxicity, and allow for durable myeloid engraftment. We hypothesized that this aim could be achieved with a purely myeloablative, non-immunosuppressive regimen.

Methods: In a retrospective multicenter analysis we reviewed data of eleven (female n=6) patients with SCID (n=4), CGD, Griscelli syndrome, Reticular Dysgenesis, Thalassemia, WAS, Leukocyte Adhesion Deficiency I and unknown IEI (all n=1), who underwent HSCT in three centers between1995 and 2020. After initial myeloid engraftment, all patients experienced partial or complete autologous reconstitution of myeloid cells with remaining donor T-cells of 9-100%.

Results: Second stem cell grafts were administered 0.5 to 25 years (median 2.2 years) after the initial transplantation. Patients received a conditioning regimen containing predominantly myelosuppressive drugs (Treosulfan n=4, Treosulfan/Thiotepa n=2, Busulfan n=2, Busulfan/Melphalan n=1) or radioimmunotherapy (n=2) avoiding immunosuppression or serotherapy (except in one patient with autoimmune phenomena before his 2nd HSCT) before the administration of T-cell replete (n=4), T-cell reduced (n=2) or T-cell depleted (n=5) grafts from the same donor. Limited toxicity was observed and neutrophil engraftment occurred at median day +16 ( + 13 to +40). No severe acute (>II°) or chronic GvHD occurred. All patients survived with a follow up of 6 months to 13 years (median 3.5 years), with complete (n=6) or stable mixed (n=4) myeloid chimerism. One patient did not benefit from the procedure and remained without myeloid engraftment. All but two patients were durably cured from the primary disease.

Conclusions: In summary these data demonstrate that for this cohort a selective myelosuppressive conditioning regimen for a second transplant from the same donor after autologous myeloid reconstitution is a feasible approach to achieve stable myeloid engraftment with low toxicity, treatment related morbidity/ mortality, or infectious complications.

Disclosure: The authors have no conflicts of interest to disclose.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P390 SINGLE CENTER OBSERVATION ON CLINICAL PRESENTATION, TRANSPLANTATION AND OUTCOME OF SEVEN PATIENTS WITH ZAP-70 DEFICIENCY

Felix Immanuel Maier 1, Philipp Friederichsen1,2, Ansgar Schulz1, Eva-Maria Jacobsen1, Klaus-Michael Debatin1, Katharina Kleinschmidt3, Selim Corbacioglu3, Jürgen Föll3, Klaus Schwarz4,5, Ulrich Pannicke4, Manfred Hönig1

1University Medical Center Ulm, Ulm, Germany, 2Schulthess Klinik Zuerich, Zuerich, Switzerland, 3University of Regensburg, Regensburg, Germany, 4University of Ulm, Ulm, Germany, 5Institute for Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Service Baden-Württemberg-Hessen, Ulm, Germany

Background: We report on the presentation, transplantation, immunological reconstitution and follow-up in a cohort of seven patients with ZAP-70 deficiency, who received HSCT between 1996 and 2012 in our institution.

Methods: Data were collected retrospectively. Immunophenotyping was determined by FACS and T-cell in vitro function by radioactive thymidine incorporation. Chimerism was determined using STR, HLA or XX/XY-FISH analysis.

Results: The age at presentation was between 5 and 372 days. All but one patient had symptomatic mostly opportunistic infections (CMV n=2, PCP n=3) at diagnosis. T-cell counts were found within or even above normal age matched reference values in 5/7 patients. Cytotoxic T-cells though were considerably reduced and below the normal range or absent in 6/7 patients. Naïve T cells were reduced as was T-cell in vitro proliferation. B-cell counts were normal in all, NK-cell counts in 5/7 patients.

The majority of patients (5/7) had a positive family history for immunodeficiency but only one patient was tested for PID in the first days after birth. Parental consanguinity was reported (6/7) or suspected (1/7) in all families and homozygous mutations in ZAP70 were detected accordingly in all patients.

HLA-identical family donors were identified for two patients, matched unrelated donors with a 10/10 match were identified for 3 and mismatched family donors for 2 patients.

Both patients transplanted from MFDs did not receive any conditioning as did another patient who was transplanted with a special regimen with stem cells from a MUD and DLIs from his formerly transplanted HLA-identical brother. One patient received his graft from a MUD after a RIC regimen containing Melphalan and Fludarabin. The remaining 3 patients were treated with myeloablative regimens containing Busulfan or Treosulfan in combination with Fludarabin and Thiotepa.

Acute GvHD ≥II° was observed in 4 patients. One patient developed chronic GvHD and finally died from lower intestinal bleeding based on cGvHD. All other patients survived HSCT with a follow up between 7 and 18 years. All 6 long-term survivors have stable and prolonged donor cell engraftment. Two patients have no myeloid engraftment, who were both transplanted without conditioning.

One patient, with pretransplant hepatic disease of unknown origin who had received RIC, developed complete donor chimerism and normal immune function but developed chronic liver disease of unknown origin and finally needed a liver transplant and is currently alive and well. Another patient with mixed donor chimerism and recurrent autoimmune cytopenia received a second transplant 12 years after his first HSCT. Non-hematological sequelae included neurological disease due to pretransplant CMV-infection, pulmonary disease, thyroid dysfunction and cutaneous dyspigmentation post GvHD.

Conclusions: Despite a positive family history, patients in this cohort were mostly diagnosed with clinically relevant opportunistic infections. Successful HSCT from a MFD without conditioning can allow for stable immune reconstitution but does not necessarily result in myeloid engraftment. As patients with severe T-cell deficiency due to genetic variants in ZAP70 are reported not to be detected by neonatal screening, typical findings in immunophenotyping need to be identified for early diagnosis to avoid pretransplant morbidity. If toxicity is tolerable, pretransplant conditioning is recommended.

Disclosure: Conflicts of interest: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P391 POLYMORPHISM OR RISK ALLELE? PRF1 A91V IN TRANS WITH A “SEVERE” PRF1 MUTATION

Oliver Wegehaupt 1,2, Oleg Borisov3, Florian Oyen4, Jasmin Mann1, Despina Moshous5, Geneviève de Saint Basile5, Kimberly Gilmour6, Wenying Zhang7, Rebecca Marsh7, Eberhard Gunsilius8, Laine Hosking9, Sharon Choo9, Katharina Wustrau10, Sujal Gosh11, Kai Lehmberg4, Anna Köttgen3, Stephan Ehl1

1Center for Chronic Immunodeficiency, Medical Center, University of Freiburg, Freiburg, Germany, 2Center for Pediatrics, Division of Pediatric Hematology and Oncology, Medical Center, University of Freiburg, Freiburg, Germany, 3Institute of Genetic Epidemiology, Medical Center, University of Freiburg, Freiburg, Germany, 4Division of Pediatric Stem Cell Transplantation and Immunology, University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 5Imagine Institute, Université de Paris, INSERM U1163, Paris, France, 6Great Ormond Street Hospital, London, United Kingdom, 7Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children’s Hospital Medical Center, Cincinnati, United States, 8Medical University Innsbruck, Innsbruck, Austria, 9The Royal Children’s Hospital, Melbourne, Australia, 10University Medical Center Ulm, Ulm, Germany, 11Center of Child and Adolescent Health, Heinrich-Heine-University, Düsseldorf, Germany

Background: Familial hemophagocytic lymphohistiocytosis type 2 (FHL2) is caused by mutations in the perforin gene (PRF1). Null mutations lead to absent cytotoxicity and predispose to hemophagocytic lymphohistiocytosis (HLH). Hypomorphic variants allow residual cytotoxicity and can cause later onset of HLH, neuroinflammation or lymphoma. PRF1 A91V, carried by 4% of the population leads to reduced perforin expression and cytolytic activity, but does not affect health or longevity even in homocygocity. Uncertainty remains whether this is also true if A91V pairs with a loss-of-function PRF1 allele (“X”). Since FHL2 is an attractive target for newborn screening, we study the clinical significance of the A91V/X constellation.

Methods: (1) We recruit A91V/X individuals through our networks and literature; (2) We gather epidemiological data on A91V/X carriers using the UK Biobank; (3) We investigate the functional consequences of A91V/X for cytotoxicity.

Results: Perforin expression in A91V/X lymphocytes was reduced variably depending on the “X”, while CTL cytotoxicity was normal in all 4 investigated A91V/X constellations. Of 33 PRF1 A91V/X individuals reported by the HLH network, 24 were identified because of FHL-2 related symptoms (14 HLH, 6 neuroinflammation, 4 lymphoma) with variable disease onset (mean 24y; range: neonatal–72y). Surprisingly, all 9 individuals identified by family screening of early-onset FHL2 patients were asymptomatic (mean 36.3y; range: 4-81y). In the UK Biobank, A91V/X was carried by 173/470.000 individuals (0.04%), none of whom had a clinical history of systemic HLH by the age of 40 years. However, we identified a slight enrichment of individuals with splenomegaly/cytopenia, neuroinflammation or lymphoma in A91V/X carriers compared to healthy individuals (8/9439 versus 165/51917;OR 2.6; p=0.045).

Conclusions: Our preliminary data indicate that PRF1 A91V/X does not entail a predictable risk for developing HLH, but we can not exclude that under certain conditions (e.g. certain triggers, nature of the “X”-mutation) HLH can be induced more readily than in controls. Furthermore, we provide evidence for a mildy increased risk for potentially FHL-2 related disease manifestations on a population level. Overall, these findings argue against preemptive HSCT in these individuals. The question remains if a first episode of HLH is sufficient to justify HSCT.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P392 IMMUNE-MEDIATED CYTOPENIA AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN CHILDREN WITH INHERITATED METABOLIC DISORDERS

Hirotoshi Sakaguchi 1, Tetsumin So1, Yoshihiro Gochio1, Akihiro Iguchi1, Takao Deguchi1, Daisuke Tomizawa1, Motomichi Kosuga1, Kimikazu Matsumoto1

1National Center for Child Health and Development, Tokyo, Japan

Background: Hematopoietic cell transplantation (HCT) is now expanding to non-malignant diseases such as inherited metabolic disorders (IMDs). Post-transplant, immune-mediated cytopenia (IMC), manifested as hemolytic anemia, thrombocytopenia, and/or neutropenia, is a significant complication with limited insight into its prevalence, risk factors, and management. Herein, we aimed to elucidate the incidence, management and prognosis of IMC after HCT in children with IMDs.

Methods: Between February 2020 and October 2023, 11 consecutive patients with IMDs (Hurler syndrome, n=2; Hunter syndrome, n=8; mucolipidosis-III, n=1) underwent allogeneic HCT (bone marrow, n=9; cord blood, n=2) at the National Center for Child Health and Development, Tokyo, Japan. The median age at the time of diagnosis and HCT was 21 months (range, 1–44 months) and 36 months (range, 13–70 months), respectively. All but one patient received with the targeted busulfan (Bu)-based myeloablative conditioning regimen (Bu with 200 mg/kg of cyclophosphamide, n=5; Bu with 150 mg/m2 of fludarabine, n=5) with serotherapy (2.5–5 mg/kg of rabbit anti-thymocyte globulin, n=6; 0.8 mg/kg of alemtuzumab, n=4); the other patient received a conditioning regimen consisting of 150 mg/m2 of fludarabine, 180 mg/m2 of melphalan, and 3 Gy of total body irradiation. In addition, the last five patients received 375 mg/m2 of rituximab prior to the conditioning regimen. All but one patient received tacrolimus and short-term methotrexate (15 mg/m2 on day 1, 10 mg/m2 on days 3, 6, and 11) for GvHD prophylaxis; the other patient, who had anaphylactic reactions to both tacrolimus and cyclosporine A, received post-transplant cyclophosphamide (PT-Cy, 50 mg/kg/day on days 3 and 4) and mycophenolate mofetil (from day 5).

Results: All patients achieved neutrophil recovery; the median day of engraftment was 16 days (range, 15–21 days) and were alive at the last observation date (December 10, 2023), with a median follow-up of 528 days (41–1382 days). However, one patient developed secondary graft failure on day 31 and was rescued by a haploidentical bone marrow transplantation from his father with PT-Cy setting. Overall, 4 of 11 patients (36 %) developed IMC with a median time to onset of 85 days (56–123 days); two patients had immune-mediated thrombocytopenia, one had immune-mediated hemolytic anemia, and the other had Evans syndrome. Of 4 patients with IMC, two patients developed IMC despite receiving rituximab prior to the conditioning regimen. Patients with grade 2 or higher acute GvHD who received systemic steroids did not develop IMC, whereas IMC was more common, although not statistically significant, in patients without GvHD or who had completed systemic steroids for GvHD. Three of the four patients with IMC were rapidly resolved with systemic steroids, while one patient with severe thrombocytopenia was resistant to steroids, IVIg, rituximab, plasma exchange, fostamatinib, and eltrombopag and was finally resolved with daratumumab.

Conclusions: The risk of post-transplant IMC in IMDs patients is high, and may be preventable with more aggressive steroid use in these non-malignant disease, which needs to be validated in a prospective study.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P393 PROPHYLACTIC ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR CSF1R-RELATED LEUKOENCEPHALOPATHY- A CASE REPORT

Malu Lian Hestdalen 1, Morten Andreas Horn2, Camilla Dao Vo1, Ingerid Weum Abrahamsen1, Tor Henrik Anderson Tvedt1, Tobias Gedde-Dahl1, Anders Eivind Myhre1

1Rikshospitalet, Oslo University Hospital, Oslo, Norway, 2Ullevål, Oslo University Hospital, Oslo, Norway

Background: Leukoencephalopathy related to pathogenic variants in the colony stimulating factor 1R (CSF1R) gene typically manifests as progressive dementia accompanied by motor symptoms resulting in death within few years of symptom onset. The inheritance pattern is autosomal dominant with high penetrance. Allogeneic hematopoietic stem cell transplantation (HSCT) may influence progression of leukoencephalopathy in patients with pathogenic CSF1R variants. However, the treatment’s effectiveness appears to be delayed by six months or more. Consequently, symptomatic patients may experience substantial decline before they start to benefit from the treatment. We present the outcomes of prophylactic HSCT in an asymptomatic carrier.

Methods: The patient was a healthy 31-year-old woman, descendant of a family where all affected members had succumbed to the disease. She, daughter of NO2 in the report by Rademakers et al in Nature Genetics 2011, carried the same mutation as her deceased relatives: heterozygosity for the pathogenic CSF1R variant NM_005211.3(CSF1R): het c.1754-2A>G. The patient displayed no abnormalities on her MRI scans and was asymptomatic.

After thorough deliberation, with a special emphasis on ethical considerations, HSCT was performed with a reduced-intensity conditioning regimen, consisting of anti-thymocyte globulin, thiotepa, fludarabine, cyclophosphamide, and total body irradiation. A bone marrow graft from a 9/10 HLA-matched unrelated male donor was used, graft-versus-host disease prophylaxis consisted of post-transplantation cyclophosphamide, sirolimus, and mycophenolate mofetil.

Results: The initial clinical course was uneventful, with full donor chimerism and no indications of graft-versus-host disease observed within the first year. Neurological and psychological assessments, as well as cerebral MRIs, showed no abnormalities up to +12 months post-transplant. A lumbar puncture at +12 months revealed 99% donor chimerism in the CSF.

However, 13 months post-transplant, she encountered a self-limiting yet extended episode of brain and spinal cord inflammation, accompanied by seizures, mild myelopathy, and mild cognitive impairment. After thorough investigations, we concluded that the most plausible explanation was an allo-immune response, although lacking conclusive evidence. She recovered fully without any medical intervention.

As of 3.5 years post-transplant, she remains in good health without cognitive or neurological impairment, except for a suspected attention deficit disorder. There are no signs of leukoencephalopathy on MRI scans, and she has no symptoms of graft-versus-host disease.

Conclusions: To our knowledge, this is the first report of prophylactic HSCT in a patient harboring a pathogenic variant in CSF1R. At 3.5 years post-treatment, there is no evidence of leukoencephalopathy. However, it is important to note that this does not conclusively establish the effectiveness of the intervention, as all affected family members of our patient have succumbed to the disease after symptom onset in their late thirties. Whether asymptomatic individuals with pathogenic CSF1R variants should be considered for prophylactic HSCT remains unanswered and raises challenging ethical concerns due to potential treatment related morbidity and mortality.

This case, with a shorter follow-up duration, was previously presented at the European Academy of Neurology Congress in June 2022 and published as a comment in Movement Disorders in May 2022 (Movement Disorders 2022 May;37(5):1108-1109. https://doi.org/10.1002/mds.29011.).

Disclosure: Anders E. Myhre:

Consultant/advisor: Takeda, Bristol Myers Squibb, Astra Zeneca, Immedica.

Speaking fees: Novartis, Takeda, Unimedic.

Malu Lian Hestdalen:

Advisor: Astra Zeneca.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P394 CLINICAL PROFILE AND TRANSPLANT OUTCOMES OF 41 PATIENTS WITH INBORN ERRORS OF IMMUNITY: A SINGLE CENTER EXPERIENCE FROM SOUTH INDIA

Stalin Ramprakash 1, Neha Singh1, C P. Raghuram1, P. Anoop1, Jyothi Janardhanan1, Harish Kumar1, Chetan Ginigeri1, Fulvio Porta2, Sagar Bhattad1

1Aster CMI Hospital, Bangalore, India, 2Ospedale dei Bambini, Brescia, Italy

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for various inborn errors of immunity (IEI) but can be challenging due to late presentation and diagnosis and financial constraints. We report the experience of transplanting 41 patients affected by IEI with at least 6 months of post-transplant follow-up over a five-year period at Aster CMI Hospital, Bangalore, India. We studied the clinical profile and transplant outcomes of 41 consecutive patients with inborn errors of immunity.

Methods: Three hundred seventy-eight patients with various Inborn errors of immunity (IEIs) were diagnosed in the pediatric immunology unit of our hospital during the study period of February 2017 to June 2023. Forty-one patients (10.8%) underwent hematopoietic stem cell transplants during the same period. Their profiles were analysed in detail.

Results: During the study period, 41 patients with various IEIs were transplanted, including 29 males and 12 females. The most common IEI to undergo transplant was severe combined immunodeficiency (SCID) (n = 12), followed by leukocyte adhesion defect (n = 7) and chronic granulomatous disease (n = 7). Patients with rarer diseases, including B-cell expansion with NF-κB and T-cell anergy (BENTA), C1q deficiency, CARMIL2 deficiency, CTLA4 haploinsufficiency, IL10RA deficiency, and MAP3 K14 deficiency, were also transplanted. The median age at transplant was 48 months. Twenty-four patients (56%) received a haplo-identical transplant using post-transplant cyclophosphamide as a GVHD prophylaxis strategy. Of the remaining 17 patients, five (12%) received human leukocyte antigen (HLA) matched sibling donor transplant (MSD), another five (12%) received HLA matched related donor transplant, six patients received matched unrelated donor transplant (15%), and one (2%) patient received 9/10 matched unrelated donor transplant. Twenty-five patients (61%) had an infection at the onset of the transplant that was not able to be eradicated by conventional treatment, while 16 patients (39%) had some form of ongoing autoimmunity. Twenty-nine patients (70.7%) achieved full chimerism, seven patients (17.1%) had mixed chimerism, and five patients had engraftment failure and had to undergo a second transplant procedure. The mean engraftment of neutrophils was achieved at 15 days (range 12–20), while the mean engraftment of platelets was achieved by day 20 (range 11–36 days). Diarrhoea (n = 18, 43.9%) was the most common infectious manifestation in the cohort, followed by CMV reactivation (n = 13, 31.7%) and pneumonia (n = 11, 26.8%). Graft versus host disease was the most common non-infectious manifestation (n = 12, 29.3%), gut being most commonly involved (n = 5), followed by veno-occlusive disease (n = 9, 22%), and thrombotic microangiopathy (n = 6, 15%). The overall survival was 63.4% (n = 26) for the entire cohort, whereas it was 75% for the fully matched cohort and 58% for the haploidentical cohort.

Clinical Characteristics

Total Number 41

Stem cell source

Bone marrow - 26

Peripheral blood stem cells - 15

Type of transplant

Matched - 16

Haplo-idientical - 24

9/10 matched - 1

Conditioning regimen

Fludarabine Busulfan based - 29

Fludarabine Treosulfan based - 10

Busulfan -cyclophophamide based - 2

Autoimmune manifestations prior to HSCT

Inflammatory Bowel disease - 5

HLH- 5

Arthritis – 2

Others - 5

Infectious complications related to HSCT

(Febrile neutropaenia not considered)

Diarrhoea- 18

CMV - 13

Lung infection - 11

Others- 9

Non-Infectious complications related to HSCT

GVHD- 12

VOD/SOS - 9

TMA - 6

Others - 9

Conclusions: This study highlights the challenges faced in transplanting children with Inborn Errors in Immunity in resource-limited settings. The factors adversely affecting transplant outcomes seem to be late presentations with ongoing infections at the time of transplant, autoimmunity, and the lack of availability of fully HLA-matched donors.

Clinical Trial Registry: Restrospective study.

Disclosure: “Nothing to declare”.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P395 HEMATOPOIETIC CELL TRANSPLANTATION AND SPLIT LIVER TRANSPLANTATION IN A PATIENT WITH ERYTHROPOIETIC PROTOPORPHYRIA - A CASE REPORT

Lasse Jost 1, Ulrich Stölzel2, Daniel Seehofer3, Katharina Egger-Heidrich4, Kristina Hölig4, Thomas Stauch5, Desiree Kunadt4, Detlef Schuppan6, Johannes Schetelig4, Nils Wohmann2, Martin Bornhäuser4, Friedrich Stölzel1

1University Hospital Schleswig-Holstein Kiel, Kiel, Germany, 2Klinikum Chemnitz, Chemnitz, Germany, 3University Hospital Leipzig, Leipzig, Germany, 4University Hospital Dresden, Dresden, Germany, 5Labor Volkmann, Karlsruhe, Germany, 6University Hospital Mainz, Mainz, Germany

Background: Erythropoietic Protoporphyria (EPP) is a rare genetic disorder characterized by deficient ferrochelatase (FECH) activity, leading to the accumulation of protoporphyrin in erythrocytes and various tissues. In some patients, EPP progresses, despite taking all therapeutic measures, to liver cirrhosis already at young age.

Methods: This abstract outlines the case of a patient with EPP, who was diagnosed at the age of two years after developing skin symptom and laboratory testings revealing increased levels of protoporphyrin IX in red blood cells. Although light protection, vitamin D supplement, oral beta carotene and later cholestyramine and time-shifted ursodesoxycholic acid as treatment strategies were followed, the patient progressed to liver cirrhosis. As the only curative option, hematopoietic cell transplantation (HCT) was considered. Since no HLA matched unrelated donor could be found, the father was concidered as a haploidentical donor after excluding EPP in him. HCT was postponed due to the Covid-19 pandemic. While HCT was postponed, liver decompensation developed in 2021, making a liver transplantation necessary. The patient underwent a split liver transplantation from his father, followed by a haploidentical bone marrow transplantation. Primary graft failure occured, a hematopoietic cell transplantation with peripheral blood stem cells was therefore performed 9 months later.

Results: Following liver transplantation as well as bone marrow and afterwards hematopoiectic cell transplantation from peripheral blood stem cells, the patient exhibited significant improvement in liver function and a reduction in protoporphyrin levels, eventually no elevated protoporphyrin concentrations were detectable. The patient was able to go outside due to a disappearance of the light sensitivity and was able to take part in outdoor activities such as a local maypole festival for the first time in his life.

Conclusions: In pediatric patients as well as one adult case, the approach of a liver transplantation followed by HCT has been shown effective. This case of paternal split liver transplantation in an adult with liver failure followed by HCT is another reminder of the therapeutic options in protoporphyria-related liver failure.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P396 SUCCESSFUL THIRD ALLOGENEIC STEM CELL TRANSPLANTATION FOLLOWING TWO GRAFT REJECTIONS IN A PATIENT WITH CHRONIC GRANULOMATOUS DISEASE AND HEPATIC/PARAPHRENIC ABSCESSES

Daniela Sperl 1, Martin Benesch1, Ursula Posch1, Konrad Rosskopf1, Volker Strenger1, Sebastian Tschauner1, Markus G. Seidel1, Wolfgang Schwinger1

1Medical University Graz, Graz, Austria

Background: In patients with chronic granulomatous disease (CGD) an allogeneic stem cell transplantation is the curative approach with a known life-threatening risk for graft rejection.

Methods: A 5-year-old patient with CGD (CYBB/gp91phox) developed hepatic/paraphrenic abscesses (Actinomyces naeslundi) and abscess-forming cervical lymphadenitis (Candida crusei) and infection-associated nephrotic syndrome.

Adequate local control of the abscesses was considered prerequisite for the planned stem cell transplantation, the hepatic/perihepatic abscesses were partially resected, the abscess-forming cervical lymphadenitis was biopsied and he was put on antimicrobial and steroid treatment for 8 months.

Results: At the age of 6 years he received a CD3 + TCRαβ+ depleted and CD34+ selected PBSCT from a matched unrelated donor (9/10 match) following conditioning with fludarabine, treosulfan, thiotepa and anti-thymocyte globulin. Acute graft rejection occurred on day +15. Despite immediate cessation of immunosuppression, initiation of corticosteroids and one donor lymphocyte infusion on day +15 (1 x 105/Lymphocytes kg body weight) rejection could not be reversed.

Thus, the patient underwent a second haplo PBSCT (CD3 + TCRαβ+ depletion/CD34+ selection) from his haploidentical father 2 weeks after graft rejection (on day +34 after the first HCT). Conditioning consisted of fludarabine, thiotepa, cyclophosphamide, thymoglobulin and total lymphoid irradiation.

Second allograft rejection occurred at day +10 necessitated a third allogeneic PBSCT from his haploidentical mother. Intensified immunosuppression was achieved by adding alemtuzumab, low-dose total body irradiation, fludarabine/melphalan and post-transplant cyclophosphamide. Since this patient failed two previous CD3 + TCRαβ+ depleted allografts, unmanipulated PBSC were chosen as stem cell source. Stable neutrophil engraftment was observed since day +13, the last platelet transfusion was given on day +15.

Due to slightly decreasing donor chimerism 7 repetitive donorlymphocyte infusions were applied monthly (each containing 5 x 104 Lymphocytes/kg recipient’s bodyweight) leading to an ongoing stable full donor chimerism and no signs of graft versus host disease (GvHD) so far. The follow-up magnetic resonance images showed resolved abscesses and the patient is up to now one year after his third stem cell transplantation in an excellent health status without any medication. All three PBSCT were performed within 3 weeks to minimize WBC aplasia after graft rejection.

Conclusions: In CGD patients infectious complications may trigger graft rejections due to strong immune stimulation. In cases of graft rejections even second and third allogeneic stem cell transplantations may be performed successfully.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P397 CHRONIC GRANULOMATOUS DISEASE DO NOT NEED MOST OF THE TIMES NGS FOR DIAGNOSIS

Marianna Maffeis 1, Marta Comini2, Alessandra Beghin2, Federica Bolda2, Elena Soncini1, Giulia Baresi1, Stefano Rossi1, Fulvio Porta1, Arnalda Lanfranchi2

1Pediatric Oncohematology and Bone Marrow Transplant (BMT) Unit, Children’s Hospital, Brescia, Italy, 2Pediatric Stem Cell Laboratory, ASST Spedali Civili, Brescia, Italy

Background: Chronic Granulomatous Disease (CGD) is an inborn error of immunity due to genetic defects in subunits of the NADPH oxidase complex of PMN. The PMN antimicrobial activity depends on the production of the reactive oxygen species (ROS). ROS damage bacterial membrane of microorganisms and cause their death. Clinical symptoms are always very suggestive.

NADPH oxidase plays a key role in the oxidative burst and defect in the NADPH oxidase protein cause the lack of ROS production and the absence of neutrophil intracellular killing mechanism.

The evaluation of oxidative burst and granulocyte activity represents an essential part of the screening of patients suspected of CGD. This is a first line functional screening, rapidity and efficacy permit to direct patient to therapy or curative treatment such as HSCT. Timing for HSCT is one of the most difficult challenge for the success of transplant, patients less than 5 years have a best prognosis; prompt diagnosis becomes critical.

Methods: The functional diagnosis of CGD occurs with specific laboratory test: TETRAZOLIUM NITROBLUE (NBT) test and Dihydrorhodamine 123 (DHR 123) TEST which is based on the flow cytometric measurement of the fluorescence emitted by rhodamine 123 in stimulated cells. PMNs are activated through PMA (Phorbol Myristate Acetate), the activation induces the assembly of the protein at the cell membrane and the production of H202 that causes the oxidation of DHR 123 in rhodamine 123, a fluorescent compound which is measured by flow cytometry.

Results: From 1996, the Pediatric Stem Cell Laboratory has screened, using the NBT or DHR 123 test, more than 3750 patients with a potential diagnosis of CGD. 52 children didn’t show a response after stimulation with PMA and therefore diagnosed as CGD Molecular analysis confirmed the diagnosis due to the following gene mutations (NCF1, NCF2, NCF3, CYBB).

The first HSCT in a CGD was in 1990. Nowadays, 21 patients were transplanted.

We performed 26 HSCT (and 4 boost), 21 of 26 HSCT after using myeloablative conditioning regimen.

The source of HSCT was bone marrow in 13 cases, PBSC in 10 and cord blood in 3 patients. 14 HSCT from MUD, 9 from HLA-identical familiar donors and 3 form haploidentical donors.

6 patients died due overwhelming infections. The median age at transplant was higher than 5 years in this group of patients. The median age at transplant for alive patients was 3 years.

GVHD complications were negligible in MUD as well as in haploidentical HSCT recipients since we applied a method with CD34+ selection and a controlled T cell add-back.

Chimerism analysis post HSCT showed 17 patients with total donor chimerism, 3 patients with mixed chimerism and only 1 patient with autologous reconstitution.

Conclusions: Today, HSCT should be considered standard of care for CGD patients.

Our experience demonstrates that HSCT is successful if performed at younger age and in absence of a severe history of infectious disease. Therefore, HSCT has to be performed immediately after the DHR 123 test result.

Disclosure: Nothing to declare.

24: Inborn Errors, Granulocyte and Osteoclast Disorders

P398 BLINATUMOMAB IS AN EFFECTIVE AND LOW TOXICITY BRIDGE TREATMENT TO HEMATOPOIETIC STEM CELL TRANSPLANTATION IN ACUTE LYMPHOBLASTIC LEUKEMIA ASSOCIATED WITH SHWACHMAN-DIAMOND SYNDROME

Fabiana Cacace1, Giovanna Giagnuolo1, Pio Stellato1, Emanuela Rossitti2, Valeria Caprioli1, Flavia Antonucci2, Maria Rosaria D’amico1, Giuseppina De Simone1, Maria Simona Sabbatino1, Rosanna Parasole1, Giuseppe Menna1, Francesco Paolo Tambaro 1

1AORN Santobono Pausilipon, Naples, Italy, 2AOU Federico II, Naples, Italy

Background: Shwachman- Diamond Syndrome (SDS) is a rare multisystem disease characterized by skeletal abnormalities, pancreatic exocrine insufficiency and bone marrow dysfunction. Patients affected by SDS are at risk for severe cytopenia and predisposition for myeloid malignancies.

Methods: We report our experience of a young girl diagnosed with SDS who developed ALL at the age of 15-years.

Results: At the age of 14-months the patient was diagnosed with SDS. Therefore, hematological follow up was performed in order to exclude myelodysplastic/AML evolution and treatment for gastroenterological-nutritional complications was started.

In September 2020, the patient was diagnosed with B-Cell-ALL. She started treatment with the dose reduced AIEOP-BFM ALL 2017 protocol, consisting of vincristine and daunoblastin without asparaginase to avoid pancreatic toxicity. She received just day 8 dose of chemotherapy due to prolonged cytopenia after only one vincristine and daunoblastin administration. Therefore, chemotherapy was discontinued and Blinatumomab was administered for 2 cycles.

A complete remission (CR) with undetectable MRD was achieved and patient was consolidated with allogeneic SCT.

The conditioning regimen consisted of 12 GY-TBI plus etoposide and GVHD prophylaxis consisted of Cyclosporin A without short course MTX, omitted to reduce toxicities.

In September 2021, the patient had a molecular relapse demonstrated by two consecutive bone marrow evaluation. She received treatment with blinatumomab (1 cycle) and 4 DLI again achieving a CR. She is currently in CR 34 months after SCT.

Conclusions: SDS is associated with predisposition to develop MDS or AML. Children with SDS develop AML in 12-25% of cases, while ALL rarely occurs. Only two cases of lymphoid malignancies in patients with SDS have been described: 1 patient in 1978 with ALL and a postmortem SDS diagnosis based on pancreatic histology and 1 patient with primary mediastinal B cell lymphoma. The latter was treated with the R-CHOP regimen and he was in remission at last follow up 15 months after diagnosis. Our case is the third ever described lymphoid malignancy in patients with SDS, and the only case with ALL currently alive and in CR.

Second ALL can be very aggressive disease with poor prognosis. There is no experience in the treatment of ALL in patients with SDS. The BFM inspired protocols showed the importance of the dose density and dose intensity in the treatment of ALL. In order to achieve the CR, an ALL patient should receive on time the entire treatment. These aspects represented the problems for the treatment of ALL in our “unique/rare” case. She, in fact, could not receive asparaginase due to pancreatic toxicity and she could not strictly adhere to the treatment schedule.

Besides, blinatumomab was the best treatment option for lower risk for toxicity and is a well tolerable treatment with no gastrointestinal/pancreatic side effects and low-grade hematological toxicities (no prolonged neutropenia) that allowed the treatment administration and adherence to treatment schedule and a bridge to transplant.

Allogeneic-SCT was performed to consolidate the CR based on the consideration that our case was a secondary ALL and because of high efficacy of combination therapy of blinatumomab and SCT in pediatric ALL.

Disclosure: Nothing to declare.

13: Infectious Complications

P399 BREAKTHROUGH INVASIVE FUNGAL DISEASE IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHINA: A MULTICENTER EPIDEMIOLOGICAL STUDY (CAESAR 2.0)

Yu-Qian Sun 1, Chuan Li1, Dan-Ping Zhu1, Jia Chen2, Xiao-Yu Zhu3, Nai-Nong Li4, Wei-Jie Cao5, Zhong-Ming Zhang6, Hai-Long Yuan7, Xiao-Xia Hu8, Xiao-Sheng Fang9, Hong-Tao Wang10, Yue Yin11, Ye-Hui Tan12, Xiao-Jun Huang1

1Peking University People’s Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China, 2The First Affiliated Hospital of Soochow University, Suzhou, China, 3The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China, 4Hematopoietic Stem Cell Transplantation Center, Fujian Institute of Hematology, Fujian Provincial Key Laboratory on Hematology, Fujian Medical University Union Hospital, Fuzhou, China, 5The First Afliated Hospital of Zhengzhou University, Zhengzhou, China, 6The first affiliated hospital of guangxi medical university, Nanning, China, 7The First Affiliated Hospital of Xinjiang Medical University, Xinjiang Institute of Hematology, Urumqi, China, 8State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 9Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, China, 10Shengjing Hospital of China Medical University, Shenyang, China, 11Peking University First Hospital, Beijing, China, 12The First Hospital, Jilin University, Changchun, China

Background: The wide use of mold-active antifungal prophylaxis in recent years might have led to the shift of epidemiology of invasive fungal disease (IFD) after allogeneic hematopoietic stem cell transplantation (allo-HSCT). However, there is no large-scale study about the epidemiology of breakthrough IFD with the prophylaxis of mold-active drugs. In the last Chinese large-scale epidemiological study (China Assessment of Antifungal Therapy in Hematological Disease, CAESAR) performed 10 years ago, more than one half patients received fluconazole as prevention (Y.Q Sun, et al. Biol Blood Marrow Transplant. 2015(21): 1117-1126) while mold-active drugs were widely used in clinical practice nowadays. Therefore, it is necessary to launch an updated study to investigate the epidemiology of breakthrough IFD in China. This study (CAESAR 2.0) aims to provide updated epidemiology of IFD in patients undergoing allo-HSCT.

Methods: This study was a multicenter, retrospective observational study performed in 12 allo-HSCT centers in China. Consecutive adult patients undergoing allogeneic HSCT during Jan, 2021 to Dec, 2021 were retrospectively reviewed. Patients with previous IFD before allo-HSCT or patients not received antifungal prophylaxis were excluded to this analysis. IFD was diagnosed according to the European Organization for Research and Treatment of Cancer and Mycoses Study Group (EORTC/MSG) 2019 criteria. Follow-up was completed on Dec 31, 2022. All patients were followed-up for at least one year or death before last follow-up.

Results: A total of 1837 patients were reviewed, including 1194 (65.0%) haploidentical stem cell transplantation, 408 (22.2%) sibling donor transplantation, 149 (8.1%) unrelated donor transplantation and 86 (4.7%) cord blood transplantation. The antifungal prophylaxis was most common with voriconazole (43.7%), followed by posaconazole (34.5%), caspofungin (13.4%), micafungin (3.0%), fluconazole (3.5%), itraconazole (1.6%) and amphotericin (0.4%), respectively. Two hundred and two (11.0%) IFD were documented, including 22 (10.9%) proven, 98 (48.5%) probable and 82 (40.6%) possible IFD. The incidence of proven+ probable IFD at one year after allo-HSCT was 6.5%. The median time of IFD was 39.5 (0-358) days after transplantation. The most common site of infection was lung (73.3%), followed by blood (18.3%). Pathogens were identified in 46.1% IFD cases, and were mainly Candida (37.5%), Mucor (28.6%), Aspergillus (16.1%) and Pneumocystis jirovecii (14.3%). The IFD attribute mortality was 22.5%, and the overall survival of IFD was 52.5%. Factors associated with IFD in multivariate analysis were failure of platelet engraftment, development of acute GVHD and EBV reactivation.

Conclusions: The risk of breakthrough IFD after allo-HSCT was still high with the prevention of mold-active antifungal drugs. Candida, Mucor, Aspergillus, and Pneumocystis jirovecii were the most frequent pathogens. It suggests that the epidemiology of IFD seems to be different with 10 years ago.

Disclosure: Fundings: This work was supported (in part) by the National Natural Science Foundation of China (grant no. 8227010768), National Key Research and Development Program of China (2021YFC2500300) and funding from Pfizer (tracking number: 74291157).

13: Infectious Complications

P400 CHANGES IN THE EPIDEMIOLOGY OF INVASIVE FUNGAL DISEASES IN HSCT OVER ALMOST A QUARTER OF A CENTURY

Marina Popova 1, Yuliya Rogacheva1, Valentin Zagranichnov2, Olga Pinegina3, Vladislav Markelov1, Aleksander Siniaev1, Irina Baranova1, Yulia Rodneva1, Alisa Volkova1, Ilya Nikolaev1, Alena Zaytseva1, Alexander Shvetsov1, Anna Spiridonova2, Oleg Golochtapov1, Yulia Vlasova1, Anna Smirnova1, Natalia Mikhaylova1, Maria Vladovskaya1, Sergey Bondarenko1, Alexander Kulagin1

1RM Gorbacheva Research Institute, Pavlov University, Saint-Petersburg, Russian Federation, 2Pavlov University, Saint-Petersburg, Russian Federation, 3Morozov Children’s City Clinical Hospital, Moscow, Russian Federation

Background: HSCT recipients, especially allo-HSCT, are at high risk of invasive fungal diseases (IFD). The introduction of new target therapy, transplantation technics, improvement of supportive care, diagnosis, prevention and treatment of IFD influence on the portrait of the HSCT recipient and the epidemiology of IFD. Changes in the epidemiology of IFD in HSCT is a subject of controversy.

Methods: This retrospective, observational study included all adult patients with HSCT from all donor types and stem cell sources between 2000 and 2023. With the aim of explore changes in the epidemiology of IFD 2842 HSCT recipients of CIC725 were enrolled in to study in two periods: cohort 1 from 2000–2010 (365 HSCT: 237 allo-HSCT and 119 auto-HSCT) and cohort 2 from 2014–2023 (2486 HSCT: 1701 allo-HSCT and 785 auto-HSCT). We estimated the incidence, etiology structure and pathogens structure of IFD comparing two cohorts separately for auto- and allo-HSCT. All probable and proven cases of IFD according to EORTC/MSG criteria during the 1 year after HSCT were included into analysis.

Results: Incidence of IFD in cohort 1 after allo-HSCT was 23,2%, invasive aspergillosis (IA) – 19%, invasive candidiasis (IC) – 2,5%, mucormycosis (MM) – 1,3%, other rare IFD – 0,4%; after auto-HSCT incidence of IFD was 10,9%, IA – 9,2%, IC – 1,7%, there was no MM and other rare IFD registered. Incidence of IFD in cohort 2 after allo-HSCT was 13,3%, IA – 10,2%, IC – 1,8%, MM – 0,6%, other rare IFD – 0,7%; after auto-HSCT incidence of IFD was 2,2%, IA – 0,9%, IC – 0,2%, MM – 0,5%, other rare IFD – 0,5%. We registered a significant decrease the incidence of IFD (<0,001) and IA (<0,001) in allo-HSCT recipients, a significant decrease the incidence of IFD (<0,001), IA (<0,001) and IC (p=0,031) in auto-HSCT recipients in cohort 2. The etiology structure of IFD in cohort 1 after allo-HSCT presented by: IA – 82% > IC – 11% > MM – 5% > other rare IFD – 2%; after auto-HSCT: IA – 85% > IC – 15%. The etiology structure of IFD in cohort 2 after allo-HSCT presented by: IA – 77% > IC – 14% > other rare IFD – 5% > MM – 4%; after auto-HSCT: IA – 41% > other rare IFD – 24% > MM – 23% > IC – 12%. The significant changes in the etiological structure was registered in auto-HSCT by the increased the MM and other rare IFD (p= 0.022), and decreased the proportion of IA in allo-HSCT (p<0,001) was found in cohort 2. The changes in pathogens structure of IFD are presented in table 1.

Table 1. Changes over time of causes of IFD in HSCT recipients (top three pathogens of IA, IC, MM, rare fungal agents)

Causes of IFD

(cultures)

Allo-HSCT n (%)

Auto-HSCT n (%)

Cohort 1

Cohort 2

p

Cohort 1

Cohort 2

p

Invasive aspergillosis

45

92

11

5

Aspergillus fumigatus

27 (60)

34 (36,9)

0.124

6 (54,5)

3 (60)

0.915

Aspergillus non-fumigatus

18 (29)

58 (62)

0.031

3 (27,3)

2 (40)

0.898

Aspergillus niger

9 (20)

33 (35,8)

0.159

2 (18,2)

1 (20)

0.944

Aspergillus flavus

2 (4,4)

10 (10,8)

0.248

1 (9,1)

1 (20)

0.596

Invasive candidiasis/candidemia

6

31

2

2

Candida albicans

3 (50)

6 (19,4)

0.246

1 (50)

1 (50)

1.000

Candida non-albicans

3 (50)

25 (80,6)

0.525

1 (50)

1 (50)

1.000

Candida parapsilosis

14 (45,2)

0.109

1 (50)

0.362

Candida krusei

1 (16,7)

5 (16,1)

0.978

-

Mucormycosis

2 (66,7)

8 (34,8)

0.511

4 (50)

1.000

Rizopus arrhizus

4 (17,4)

0.474

2 (25)

1.000

Rhizopus microsporus

1 (33,3)

0.015

1 (12,5)

1.000

Rhizomucor pusillus

1 (33,3)

2 (8,7)

0.300

1 (12,5)

1.000

Other rare pathogens

1 (33,3)

15 (65,2)

0.571

4 (50)

1.000

Cryptococcus neoformans

1 (33,3)

1 (4,3)

0.135

-

Paecilomyces variotii

6 (26,1)

0.383

1 (12,5)

1.000

Trihosporon asahii

5 (21,7)

0.425

1

Conclusions: Over the past almost quarter of a century, we observed significant decrease the incidence of IFD due to decreasing the IA incidence and proportion in the etiology structure together with significant increase in share of Aspergillus non-fumigatus in allo-HSCT and decrease the incidence of IFD due to decreasing the IA and IC incidence and the increased the MM and other rare IFD in the proportion of the etiology of IFD in auto-HSCT.

Disclosure: Nothing to declare.

13: Infectious Complications

P401 OUTCOMES FROM BRONCHOSCOPY AND BRONCHOALVEOLAR LAVAGE IN HEMATOPOIETIC CELL TRANSPLANT RECIPIENTS

Colin Hayes 1, Benison Lau1, Kengo Inagaki1, Mike Triebwasser1, John Magenau1, Mary Riwes1, Mark Vander Lugt1, Ghada Abusin1, Sung Choi1, Sarah Anand1, Monalisa Ghosh1, John Maciejewski1, Attaphol Pawarode1, Gregory Yanik1

1University of Michigan Medical Center, Ann Arbor, United States

Background: Bronchoscopy with bronchoalveolar lavage [BAL] is commonly performed as a diagnostic tool in patients with pulmonary complications following hematopoietic cell transplantation [HCT]. We report a single center experience of patients undergoing BAL post-HCT, including the incidence and types of pathogens identified, and the clinical impact of the BAL.

Methods: Case records from HCT recipients that underwent BAL between 2001-2022 were identified using a validated free text search tool (EMERSE), followed by review of electronic medical records. Demographic and basic transplant-related data, including timing of the BAL (post-HCT), identification of pathogens, changes in clinical management as a result of the BAL, and bronchoscopy-related complications were identified. If more than one BAL was performed on a patient, only data from their initial BAL was entered.

Table 1

Pathogenic Yield of BAL in HCT Recipients

Total Cases

% Pathogenic

p-value

BAL

All

717

32.8

N/A

Age (Years)

0-20

98

28.6

0.447

21-40

147

31.3

0.746

41-60

285

29.5

0.230

> 60

187

41.1

0.014*

Conditioning Regimen

Full Intensity

565

32.4

0.750

Reduced Intensity

152

34.2

Primary Disease

Malignant

662

32.0

0.220

Non-Malignant

55

41.8

Transplant Type

ALLO

589

34.3

0.125

AUTO

128

25.8

Days HCT to BAL

0-30

153

26.8

0.151

31-100

148

31.8

0.748

101-365

249

28.5

0.132

> 365

167

45.5

0.001*

Results: Seven hundred and seventeen of 4980 HCT recipients (14.4%) underwent BAL during this time period, with the median age 52.0 years (range 0.0-75.9 years). BAL were more frequently performed in recipients of allogeneic versus autologous grafts (p <0.001), recipients of reduced versus full intensity conditioning regimen (p <0.001), and those 21-40 years in age (p <0.001). At least one pathogen was identified in 235 (32.8%) BAL, including fungal (n= 100), viral (n= 98), bacterial (n= 64), or mycobacteria (n= 29) pathogens. The most prevalent individual pathogens included Aspergillus sp (n= 53), Cytomegalovirus (n= 34), Pneumocystis jirovecii (n= 23), and Mycobacterium avium (n= 22). The pathogenic yield was higher if the BAL was performed > 365 days post-HCT (p= 0.001), or in patients > 60 years of age (p= 0.014) (Table 1). The BAL led to changes in antimicrobial therapy in 40.5% of cases, with changes in immunosuppressive therapy (including systemic corticosteroids) in 6.4% of BAL. Overall, BAL were well tolerated, with complications occurring in 5.9% of cases, including hemorrhage (2.9%) and hypoxia (0.7%). In patients not requiring mechanical ventilation prior to the BAL (n= 631), 2.1% required prolonged intubation (> 24 hours) after BAL procedure.

Conclusions: To our knowledge, this represents the largest case series of BAL performed in HCT recipients. Pathogens were identified in approximately 33% of cases, with the pathogenic yield highest in BAL performed > 365 days post-HCT. The performance of BAL had high utility in HCT recipients, impacting clinical management in nearly 50% of cases.

Clinical Trial Registry: Not applicable.

Disclosure: Colin, Hayes, Nothing to declare.

Benison, Lau, Nothing to declare.

Kengo, Inagaki, Nothing to declare.

Mike, Triebwasser, Nothing to declare.

John, Magenau, Nothing to declare.

Mary, Riwes, Nothing to declare.

Mark, Vander Lugt, Nothing to declare.

Ghada, Abusin, Nothing to declare.

Sung, Choi, Nothing to declare.

Sarah, Anand, Nothing to declare.

Monalisa, Ghosh, Nothing to declare.

John, Maciejewski, Nothing to declare.

Attaphol, Pawarode, Nothing to declare.

Gregory, Yanik, Nothing to declare.

13: Infectious Complications

P402 LETERMOVIR PROPHYLAXIS FOR CYTOMEGALOVIRUS INFECTION IS THE POTENTIAL RISK OF EBV-POSITIVE POSTTRANSPLANT LYMPHOPROLIFERATIVE DISORDERS AFTER HAPLOIDENTICAL STEM-CELL TRANSPLANTATION

Xuying Pei 1, Xiaojun Huang1

1Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China

Background: Letermovir, a novel antiviral targeting the viral terminase complex of cytomegalovirus, has been reported highly effective in reducing the risk of CMV-related complications after allogeneic stem cell transplantation (allo-SCT). However, published data suggested that cellular reconstitution might delayed after letermovir administration, partialy related to decreased CMV antigen exposure. Whether the delay in cellular immune reconstitution whould increase other viral associated complications is unknown.

Methods: We prospectively analyzed 363 consecutive haploidentical SCT recipients who received prophylactic letermovir. An additional cohort of 363 historical haplo-SCT recipients who received PCR-guided preemptive therapy were included as controls matched on the propensity score. We compared the kinetics of double-stranded DNA viremia and related disease after allo-SCT in the ear of letermovir administration, and also evaluated the multivirus-specific immune reconstitution.

Results: We observed that prophylactic letermovir significantly reduce the 100-day and also 180-day cumulative incidence of CMV viremia and refractory CMV infection. Though the rates of EBV, ADV, BKV, HSV and HHV6 viremia were comparable in letermovir and preemptive therapy cohorts, the cumulative incidence of posttransplant lymphoproliferative disorders (PTLD) at 180-day was significant higher in letermovir recipients (8.73% VS 2.77%, P=0.001).

Conclusions: Our findings demonstrate that prophylactic letermovir treatment is highly effective in reducing the risk of CMV-related complications, but is the potential risk of EBV-positive PLTD after haplo-SCT.

Disclosure: Nothing to declare.

13: Infectious Complications

P403 DIAGNOSIS OF MUCORMYCOSIS IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION USING METAGENOMIC NEXT-GENERATION SEQUENCING: A SINGLE-CENTER CLINICAL STUDY

Fang Xu 1, Jianping Zhang1, Min Xiong1, Yanli Zhao1, Deyan Liu1, Xingyu Cao1, Zhijie Wei1, Jiarui Zhou1, Ruijuan Sun1, Yue Lu1

1Hebei Yanda Lu Daopei Hospital, Langfang, China

Background: Mucormycosis present a formidable diagnostic and prognostic challenge in allogeneic hematopoietic stem cell transplantation (allo-HSCT).

Methods: We conducted a retrospective analysis of 4303 allo-HSCT cases performed at Hebei Yanda Ludaopei Hospital between January 1, 2019, and December 31, 2022. Proven or probable mucormycosis cases were identified from one month pre-transplantation to post-transplantation, accompanied by an analysis of their clinical characteristics and identification of prognostic factors. Metagenomic next-generation sequencing (mNGS) was employed concurrently with traditional culture for examining body fluids or tissues.

Results: A total of 53 cases (1.23%), comprising 46 probable and 7 proven cases, of mucormycosis were identified. Among them, 29 were male and 24 were female, with a median age of 31 (3-65) years. The distribution of underlying diseases included 23 cases of acute myeloid leukemia, 14 cases of acute lymphoblastic leukemia, 6 cases of myelodysplastic syndrome, 5 cases of aplastic anemia, and 5 cases of other hematologic disorders. Transplant donors were haploidentical(n=39), match-unrelated(n=10), and sibling(n=4). Infections within one month pre-transplantation were observed in 8 cases, while the remaining 45 cases experienced post-transplantation infections at a median time of 133 (0-910) days. Pathogens identified included Rhizopus spp. (n=25), Rhizomucor spp. (n=16), Absidia spp. (n=7), Cunninghamella spp. (n=4), and Mucor spp. (n=1). All pathogens were identified using mNGS, with specimens primarily sourced from blood (41 cases), sterile tissues (6 cases), and non-sterile sites (10 cases). The median reads number for blood NGS was 150 (3-46276). Only one patient had a positive culture. The predominant infection site was the lungs (39 cases), followed by disseminated (6 cases), rhino-orbital-cerebral (5 cases), gastrointestinal tract (2 cases), and skin-soft tissue (1 case), with 6 cases involving central nervous system invasion. Clinical manifestations were primarily fever, accounting for 85% of cases. All patients had antifungal prophylaxis before infection, with azole drugs accounting for 71.7%. Among the patients, 25 were neutropenia during fungal infection, 19 had acute or chronic graft-versus-host disease(GvHD), and the remaining patients experienced relapse, thrombotic microangiopathy, viral infections, among others. Treatment modalities included combination therapy in 43 cases, monotherapy in 10 cases, and surgical intervention in 6 cases. The overall treatment response rate was 52.8%. As of July 1, 2023, the median follow-up time was 65 (1-1422) days, with a 30-day overall survival (OS) rate of 60.4±6.7% and a 2-year OS rate of 31±6.5%. A total of 36 deaths occurred, with causes including fungal infection in 21 cases, relapse in 6 cases, GvHD in 6 cases, others in 3 cases. Univariate analysis indicated that disseminated infection (p=0.03%), central nervous system invasion (p<0.01%), second transplantation (p=0.03%), concurrent GvHD (p=0.01%) and neutropenia (p=0.03%) were adverse prognostic factors, while surgical intervention (p=0.012%) suggested a favorable prognosis. Multivariate analysis revealed central nervous system invasion (p=0.001%) and second transplantation (0.004%) as independent risk factors for adverse outcomes.

Conclusions: The overall prognosis of mucormycosis in allo-HSCT remains poor, emphasizing the critical role of mNGS in rapid pathogen identification. Central nervous system involvement and second transplantation were identified as independent risk factors for adverse outcomes.

Disclosure: Nothing to declare.

13: Infectious Complications

P404 MARIBAVIR USE FOR REFRACTORY CMV INFECTION/DISEASE: THE RESULTS FROM THE FRENCH COMPASSIONATE PROGRAMME

Catherine Cordonnier 1, Nassim Kamar2, Philippe Gatault3, Faouzi Saliba4, Fanny Vuotto5, Lionel Couzi6, Cinira Lefevre7, Michèle Maric7, Sophie Alain8

1Assistance Publique-Hopitaux de Paris (AP-HP), Henri Mondor Hospital and University Paris-Est-Créteil, Créteil, France, 2CHU Rangueil and INSERM U1043, IFR–BMT, University Paul Sabatier, Toulouse, France, 3Hôpital Bretonneau et Hôpital Clocheville, Tours, France, 4Hôpital Paul Brousse, University Paris Saclay, Villejuif, France, 5CHU Lille, Lille, France, 6Hôpital Pellegrin, Bordeaux, France, 7Takeda France, Paris, France, 8Limoges University Hospital and UMR Inserm 1092, University of Limoges, Limoges, France

Background: Maribavir, a benzimidazole riboside, inhibits CMV replication by inhibiting the UL97 protein kinase. Its indication is “treatment of CMV infection and/or disease that are refractory (with or without resistance) to one or more prior therapies in patients who had a hematopoietic stem cell transplant (HSCT) or solid organ transplant (SOT)”. The recommended dose is 400 mg twice daily for 8 weeks based on the summary of product characteristics (SmPC). In France, patients were treated with maribavir, through a compassionate use program (CUP), from November 2021 to April 2023, prior to the granting of European marketing authorization.

Methods: The main objectives of the CUP data collection were to describe patient characteristics at baseline, maribavir use, effectiveness, and safety data using descriptive statistics. Effectiveness was assessed by viral clearance (ie., CMV DNA concentration below the lower limit of quantification, ie, <137 IU/mL for plasma or <411 IU/ml for whole blood) at week 8. Effectiveness was estimated within all CUP patients, and separately within the SOLSTICE equivalent sub-group (i.e. patients with same eligibility criteria as the SOLSTICE pivotal trial [NCT02931539]). These analyses were conducted using an intention-to-treat (ITT) approach (including all treated patients), and a sensitivity analysis approach excluding lost to follow-up and deceased patients.

Results: In total, at baseline, on 84 approved requests 82 patients were treated with maribavir, with a median age of 58.7 years, and 69.0% of male patients. Most patients had a SOT (n=71, 84.5%; mainly kidney transplants n=55; 77.5%) and 15.5% of them had a HSCT (n=13). Twenty-four patients (29.0%) had cytopenia (n=17; neutrophil <1000/mm3 or hemoglobin <8 g/dl) and/or severe renal failure (n=13; creatinine clearance ≤30 mL/min/1.73 m²) and were included in the CUP (such patients were excluded from the SOLSTICE trial). CMV disease was observed in 31 patients (36.9%), with 67.9% of them having involvement of the gastrointestinal tract. More than 1/3 of patients (n=36; 42.8%) had a previous episode of CMV infection before treatment access request. Resistance testing was performed for 80 (95.2%) patients, of which 59 (78.7%) had identified mutations in UL97 (55.9%) and in UL54 (32.2%) genes. Median treatment duration was 8.4 weeks, which is aligned with the SmPC. Viremia clearance at week 8 was observed in 41.5% of patients (n=82) using the ITT approach, and 47.9% (n=71) using sensitivity analysis approach. Within the SOLSTICE subgroup (n=59), viremia clearance at week 8 was observed in 44.1% of patients using the ITT approach, and 51.0% in the sensitivity analysis approach (n=51). There were no new safety signals with respect to the maribavir SmPC.

Conclusions: This was the first analyses of maribavir use outside the SOLSTICE pivotal trial, in France. The population analyzed differed from the SOLSTICE population in that it had a higher proportion of SOT recipients, with CMV disease, more severe biological criteria and with identified antiviral resistance. Despite the more severe CUP patient profiles, the results showed coherent efficacy and safety findings with the pivotal study.

Disclosure: C. Cordonnier has received speakers fees and participated to advisory boards for Cidara, Equillium, Gilead, MSD, Mundipharma, Takeda.

N. Kamar has received speakers fees and participated to advisory boards for Astellas, AstraZeneca, Biotest, CSL Behring, Chiesi, ExeViR, Hansa, Merck Sharp and Dohme, Glasgow Smith Kline, Novartis Pharma, Sanofi, Sandoz, Takeda.

P. Gatault has received speakers fees or participated to advisory boards for AstraZeneca, Astellas, Bayer, Chiesi, Biotest, GSK, Sandoz, Takeda: Lectures.

F. Saliba has received speakers fees or participated to advisory boards for Novartis, Chiesi, Biotest, Takeda, Gilead, Mundipharma.

S. Alain received research funding as a scientific expert and site principal investigator: Altona, BioMérieux, Biotest, GlaxoSmithKline, Merck, Merck Sharp & Dohme, Qiagen, Shire/Takeda; honoraria for lectures paid to institution: Biotest, Merck Sharp & Dohme, IQone, Takeda; support for attending meetings: BioMérieux, Biotest, QCMD, Takeda; advisory board (unpaid): QCMD. Primary investigator for this study in France.

F. Vuotto has received participated to advisory boards for Takeda.

C. Lefevre and M. Maric are part of Takeda company.

- This work was supported by Takeda France.

13: Infectious Complications

P405 COMPREHENSIVE SCREENING AND SENSITIVITY-GUIDED TREATMENT SIGNIFICANTLY REDUCES THIRTY-DAY MORTALITY OF METALLO-Β-LACTAMASES PRODUCING ENTEROBACTERALES (MBL-E) INFECTION FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION: INSIGHTS FROM A PROSPECTIVE STUDY

Yi-Han Yang1, Meng Lv1, Qi Wang1, Jing Liu1, Xiao-Lu Zhu1, Xiao-Dong Mo1, Yu-Qian Sun 1, Yu Wang1, Lan-Ping Xu1, Xiao-Hui Zhang1, Xiao-Jun Huang1,2

1Peking University People’s Hospital, Beijing, China, 2Peking-Tsinghua Center for Life Sciences, Beijing, China

Background: Enterobacterales producing Metallo-β-lactamases (MBL-E), resistant to the latest beta-lactamase inhibitors (such as avibactam), have been associated with the highest observed excess thirty-day mortality (approximately 30%, Clin Infect Dis. 2023) in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT). The optimized strategy for addressing MBL-E infections has yet to be established.

Methods: Sequential allo-HSCT patients (n=5226) between July 2018 and June 2023 underwent screening for MBL-E colonization using rectal, nasopharyngeal, and axillary swabs. Antimicrobial susceptibility testing was conducted through the Vitek 2 automated system (mCIM+eCIM) before Dec 2021, and post-Jan 2022, the NG-Test CARBA 5 (NG Biotech, Guipry, France) was employed for carbapenemase detection (NDM, IMP, VIM, etc.). Antibiotic regimens were chosen based on clinical judgment and guided by in vitro active antibiotics (OAAs), including aztreonam (ATM) +/- ceftazidime-avibactam (CZA), tigecycline, colistin, high-dose meropenem, and aminoglycoside. The primary endpoint was thirty-day mortality post-diagnosis.

Results: Eighty-five consecutive patients (1.6% of the total cohort) infected by MBL-E, along with an additional 12 patients screened positive for MBL-E, were included in the study. Underlying disease AML (n=44; 45.4%); ALL(n=31; 32.0%); MDS(n=8; 8.23%); SAA(n=6; 6.19%);other disease(n=7). Donor type: haploidentical (n=78; 80.4%), matched sibling donor (n=14;14.4%);matched unrelated donor (n=5, 5.1%). Overall, there were 22 (21.6%) cases of bloodstream infections (BSIs) or central venous catheter-related infections, 7 (7.22%) hospital-acquired/ventilator-associated pneumonias (HAP/VAPs), 4 (4.12%) complicated urinary tract infections (cUTIs), and 36 (37.1%) intra-abdominal/gastrointestinal/perianal infections.

The primary endpoint was the 30-day mortality, which was only 2.4% (0.5-7.5%), and the 3-year non-relapse mortality was 16.0% (8.4-25.7%).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (45)

The same MBL-E predicted infection in 45 patients (52.9%) identified during screening, with a median time from screening to infection of 13 days (2 to 66 days). The median days of neutrophil engraftment were 14.5 days post-HSCT (9 to 35 days). Of the 68 patients (58.8%) diagnosed with MBL infection before neutrophil engraftment, the median days of infection were 4 days before infusion (-12 to -1). A total of 125 bacterial strains were identified, including 56% Escherichia coli (n=70), 16% Klebsiella pneumoniae (n=20), 17.6% Enterobacter cloacae (n=22), 4% Citrobacter freundii (n=5), and Morganella morganii subsp. Morganii (n=1), Enterobacter hormaechei (n=1), Proteus mirabilis (n=2), Citrobacter amalonaticus (n=1), Klebsiella oxytoca (n=2), Citrobacter farmeri (n=1). Of these, 38.4% (n=48) were sensitive to aztreonam (ATM), 90.3% (n=102) were sensitive to tigecycline, 89.9% (n=80) were sensitive to colistin, and 31.2% (n=39) had a meropenem MIC between 4 to 8. In strains with a known gene phenotype(n=39), 95% (n=37) were NDM, 5% (n=2) were IMP, and none was VIM. Among MBL-E infected patients, 90.6% received OAAs-supported treatments, 10.6% (n=9) received ATM, 15.3% (n=13) received CZA + ATM, 15.3% (n=13) received colistin-containing regimens, and 49.4% (n=42) received tigecycline.

Conclusions: In this extensive cohort study, screening for MBL-E infection and sensitivity-guided treatment substantially improved outcomes and significantly decreased 30-day mortality compared to previous findings.

Clinical Trial Registry: NCT03756675.

Disclosure: Nothing to declare.

13: Infectious Complications

P406 PENTAGLOBIN® AS EARLY ADJUVANT TREATMENT FOR FEBRILE NEUTROPENIA IN HEMATOLOGICAL PATIENTS COLONIZED BY CARBAPENEM-RESISTANT ENTEROBACTERIACEAE OR PSEUDOMONAS AERUGINOSA: INTERIM ANALYSIS OF GITMO STUDY “PENTALLO”

Daniela Clerici 1, Alessandra Picardi2, Patrizia Chiusolo3, Nicola Di Renzo4, Anna Paola Iori5, Mario Delia6, Ilaria Cutini7, Francesca Bonifazi8, Michele Malagola9, Raffaella Cerretti10, Maria Goldaniga11, Francesca Patriarca12, Daniele Vallisa13, Attilio Olivieri14, Roberto Sorasio15, Stefania Bregante16, Angelo Michele Carella17, Alessandro Busca18, Consuelo Corti1, Eliana Degrandi19, Fabio Ciceri1, Massimo Martino20, Corrado Girmenia5

1IRCCS Ospedale San Raffaele, Milano, Italy, 2AORN A. Cardarelli, Napoli, Italy, 3Fondazione Policlinico A. Gemelli IRCCS, Roma, Italy, 4Ospedale “Vito Fazzi”, Lecce, Italy, 5Policlinico Umberto I, Roma, Italy, 6A.O.U. Polyclinic Consortium of Bari, Bari, Italy, 7AOU Careggi, Firenze, Italy, 8IRCCS University Hospital of Bologna, Institute “L. and A. Seràgnoli”, Bologna, Italy, 9“ASST-Spedali Civili” Hospital of Brescia, Brescia, Italy, 10PTV Foundation - Tor Vergata Polyclinic, Roma, Italy, 11Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milano, Italy, 12S. Maria della Misericordia Hospital, Azienda Sanitaria Universitaria Friuli Centrale, Udine, Italy, 13Local Health Authority of Piacenza, Piacenza, Italy, 14Ospedali Riuniti di Ancona, Ancona, Italy, 15Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy, 16IRCCS Ospedale Policlinico San Martino, Genova, Italy, 17Fondazione IRCCS Casa Sollievo della Sofferenza, San Giovanni Rotondo, Italy, 18AOU Citta’ della Salute e della Scienza di Torino, Torino, Italy, 19Trial Office GITMO Gruppo Italiano per il Trapianto di Midollo Osseo, cellule staminali emopoietiche e terapia Cellulare, Bologna, Italy, 20Grande Ospedale Metropolitano “Bianchi-Melacrino-Morelli”, Reggio Calabria, Italy

Background: Despite improvements in the use of antibacterial therapy, infections sustained by multi-drug-resistant (MDR) gram-negative bacteria (GNB) still represent a lifethreatening complication in hematological neutropenic patients. In this setting colonization by MDR-GNB is a crucial predictive factor for bloodstream infection (BSI) sustained by the colonizing pathogen. Experiences in intensive care unit patients suggested that the addition of an immunological adjuvant therapy as Pentaglobin® (IgM-enriched immunoglobulin) to antibacterial targeted therapy may have a synergistic effect on further decreasing mortality.

The aim of the study is to investigate early addition of Pentaglobin® to the best available antimicrobial therapy to reduce mortality in neutropenic febrile hematological patients colonized by carbapenem-resistant Enterobacteriaceae (CRE) or Pseudomonas aeruginosa (PA).

In a previous GITMO study, overall 50% of colonized patients were alive at 120 days from a BSI with a 40% mortality rate at 30 days.

Methods: Prospective multicenter interventional study (Pentallo, EudraCT 2018-001344-57) conducted in centers of the Gruppo Italiano Trapianto di Midollo Osseo (GITMO). The study enrolls consecutive adult patients (target 120 patients) suffering from acute leukemia candidates to intensive chemotherapy and patients candidates to allogeneic HSCT (alloHSCT), with a documented colonization (rectal/pharyngeal swab) or previous BSI sustained by CRE or any PA during the three months preceding chemotherapy or alloHSCT. At the onset of the first neutropenic fever, patients receive an antimicrobial treatment active against the MDR colonizing strain in combination to Pentaglobin® (5 ml/kg body weight daily on three consecutive days).

Aim of the study is to demonstrate a 50% reduction in 30-days mortality for carriers developing a pre-engraftment BSI sustained by CRE or PA, and an increase by 20% in overall survival at 120-days from the treatment.

Results: Enrollment is ongoing since December 2019. To date 106 patients have been enrolled and 88 patients are evaluable (76 allo-HSCT). 4 of 88 patients did not receive Pentaglobin (3/4 patients did not develop fever). At 30-days and 120-days follow-up 84 and 80 patients, respectively, are evaluable.

Patients were colonized by CR-Klebsiella pneumoniae (CR-KP, 59/84), other CRE (3/84) and PA (22/84, 7/22 MDR).

All cases of microbiologically documented infections (n=43) were BSI: CR-KP (23/43), PA (5/43), other non-MDR GNB (6/43), Gram-positive bacteria (9/43).

The colonizing pathogen was isolated from blood in 23 of 59 CR-KP carries and in 4 of 22 PA carriers.

Overall 3 of 28 (10.7%) patients with a CRE or PA bacteremia died within 30 days from the first febrile neutropenia episode. The infection was considered the cause of death in 2 of the 3 cases. All six patients with septic shock sustained by CR-KP survived at 30 days.

Overall 64 (80.0%) patients were alive at 120 days. Out of 16 patients who died during the first 120 days only 3 died due to an infectious cause (2/3 sustained by a CR-KP).

Conclusions: Preliminary data on 84/120 patients show promising results in term of favorable outcome of neutropenic high risk febrile hematologic patients colonized by CRE or PA treated with Pentaglobin® as adjuvant of the best early antibiotic empiric therapy.

Clinical Trial Registry: EudraCT 2018-001344-57.

Disclosure: None directly related to the abstract.

13: Infectious Complications

P407 MUCOSITIS-ASSOCIATED BLOODSTREAM INFECTIONS IN HAEMATOLOGY PATIENTS WITH FEVER DURING NEUTROPENIA

Nick de Jonge 1,2,3, Jeroen Janssen1,2,3, Paula Ypma4, Alexandra Herbers5, Arne de Kreuk6, Wies Vasmel6, Jody van den Ouweland7, Aart Beeker8, Otto Visser9, Sonja Zweegman1,3, Nicole Blijlevens2, Michiel van Agtmael1, Jonne Sikkens1

1Amsterdam UMC, Amsterdam, Netherlands, 2Radboudumc, Nijmegen, Netherlands, 3Cancer Center Amsterdam, Amsterdam, Netherlands, 4HAGA hospital, The Hague, Netherlands, 5Jeroen Bosch Ziekenhuis, ’s-Hertogenbosch, Netherlands, 6OLVG, location West, Amsterdam, Netherlands, 7Canisius Wilhelmina Ziekenhuis, Nijmegen, Netherlands, 8Spaarne Gasthuis, Hoofddorp, Netherlands, 9Isala, Zwolle, Netherlands

Background: Haematology patients with high-risk neutropenia are prone to mucosal-barrier injury-associated laboratory confirmed bloodstream infections (MBI-LCBI). We assessed risk factors for MBI-LCBI including candidaemia in neutropenic haematology patients with fever.

Methods: This prospective observational study was performed in six dedicated haematology units in the Netherlands (NCT02149329). Eligible haematology patients had neutropenia <500/mL for ≥7 days and had fever. MBI-LCBIs were classified according to Centers for Disease Control (CDC) definitions and were followed until the end of neutropenia >500/mL or discharge.

Results: We included 416 patients from December 2014 until August 2019. We observed 63 MBI-LCBIs. MBI-LCBIs occurred predominantly in the second and third week after starting chemotherapy. Neither clinical mucositis scores nor the blood level of citrulline at fever onset was associated with MBI-LCBI. In the multivariable analysis, MASCC-score (odds ratio [OR] 1·16, 95%-confidence interval[CI] 1·05 to 1·29 per point decrease), intensive chemotherapy (OR 3·81, 95%-CI 2·10 to 6·90) and Pichia kudriavzevii (formerly Candida krusei) colonization (OR 5·40, 95% CI 1·75 to 16·7) were retained as risk factors for MBI-LCBI, while quinolone use seemed protective (OR 0·42, 95%-CI 0·20 to 0·92). Citrulline level (OR 1·57,95%-CI 1·07 to 2·31 per mmol/L decrease), active chronic obstructive pulmonary disease (OR 15·4, 95%-CI 1·61 to 147) and colonization with fluconazole-resistant Candida (OR 8·54, 95%-CI 1·51 to 48·4) were associated with candidaemia.

Conclusions: In haematology patients with fever during neutropenia, hypocitrullinaemia at fever onset was associated with candidaemia, but not with bacterial MBI-LCBI. Patients with intensive chemotherapy with a low MASCC-score and colonization with Pichia kudriavzevii had the highest risk of MBI-LCBI.

Clinical Trial Registry: NCT02149329.

Disclosure: None.

13: Infectious Complications

P408 DESCRIPTIVE ANALYSES OF PULMONARY FUNCTION CHANGES BEFORE AND AFTER COVID-19 IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS. AN INFECTIOUS DISEASE WORKING PARTY STUDY

Jose-Luis Piñana 1, Per Ljungman2, Stephan Mielke2, Gloria Tridello3, Helene Labussiere4, Carlos Solano1, Anna Torrent5, Regis Peffault De Latour6, Nicolaus Kroeger7, Maria Jesus Pascual Gascon8, Adriana Balduzzi9, Lutz Peter-Muller10, Jürgen Kuball11, Amjad Hayat12, Urpu Salmenniemi13, Fabio Benedetti14, Dina Averbuch15, Rafael De La Camara16

1Hospital Clinico Universitario of Valencia, Valencia, Spain, 2Karolinska Comprehensive Cancer Center, Stockholm, Sweden, 3Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy, Verona, Italy, 4Centre Hospitalier Lyon Sud, Lyon, France, 5ICO-Hospital Universitari Germans Trias i Pujol, Badalona, Spain, 6Saint-Louis Hospital, BMT Unit, Paris, France, 7University Hospital Eppendorf, Hamburg, Germany, Hamburg, Germany, 8Hospital Regional de Málaga, Spain, Malaga, Spain, 9Centro Trapianto di Midollo Osseo - Clinica Pediatrica, Monza, Italy, 10Martin-Luther-Universitaet Halle-Wittenberg, Halle, Germany, Halle, Germany, 11University Medical Centre, Utrecht, Netherlands, Utrecht, Netherlands, 12Galway University Hospital, Galway, Ireland, 13HUCH Comprehensive Cancer Center, Helsinki, Finland, 14Policlinico G.B. Rossi, Verona, Italy, 15Faculty of Medicine, Hebrew University of Jerusalem; Hadassah Medical Center, Jerusalem, Israel, 16Hospital de la Princesa, Madrid, Spain

Background: In the context of SARS-CoV-2, the emergence of post-COVID-19 pulmonary fibrosis in the general population has been documented. Previous reports indicate that over a third of COVID-19 patients develop fibrotic abnormalities, impaired Diffusing Capacity of the Lungs for Carbon Monoxide (DLCO), and reduced total lung capacity (TLC). However, data on pulmonary function impairment in allogeneic stem cell transplant (allo-SCT) recipients after COVID-19 is lacking. This study aims to describe changes in pulmonary function tests (PFTs) before and after COVID-19.

Methods: In this retrospective multicenter cohort study, 102 allo-SCT recipients diagnosed with COVID-19 between March 2020 and August 2021, reported to the EBMT-IDWP-COVID-19 registry, were included. These recipients had pre-transplant (pre-Tx) (n=97) and/or post-transplant but pre-COVID-19 (n=65) PFTs. Among them, 55 recipients also had PFT data 2 to 6 months (median 133 days, range 22-374 days) after recovering from SARS-CoV-2 infection. 35 recipients (34.3%) had pre-Tx or pre-COVID-19 and post-COVID PFT for the analyses. PFT parameters included forced expiratory volume per second (FEV1), forced vital capacity (FVC), FEV1/FVC ratio, DLCO, and total lung capacity (TLC). Pre-transplant PFTs were classified according to the pulmonary hematopoietic cell transplant comorbidity index (PF-HCT-CI).

Results: The cohort comprised allo-SCT recipients with median age of 56 years. The median time from SCT infusion to COVID-19 diagnosis was 538 days, and the severity of COVID-19 varied, with 47 recipients experiencing lower respiratory tract disease, and 61.4% requiring hospitalization. PF-HCT-CI was normal in 43%, moderately altered in 29%, and severe in 28% of recipients. Overall, FEV1, FVC, FEV1/FVC, TLC, and DLCO exhibited median declines post-COVID-19 compared to pre-transplant or pre-COVID-19 levels in evaluable recipients, indicating impaired PFTs. Specifically, there was a significant decrease in FEV1 in 54.9% of cases, translating to a magnitude of -20.5%. FVC exhibited a reduction in 55.3% of cases, with a magnitude of -13.7%. FEV1/FVC ratio experienced a reduction in 25% of cases, amounting to a magnitude of -5.5%. TLC decreased in 56.8% of cases, with a magnitude of -13%. Additionally, DLCO values decreased in 69.4% of cases, with a magnitude of -17%. Regarding functional pulmonary patterns, the percentage of individuals with obstructive patterns exhibits an increase from Pre-Tx at 9.5% to Pre-COVID at 20.3%, with a subsequent decrease post-COVID to 9.1%. On the other hand, the percentage of individuals with restrictive patterns shows an increase from Pre-Tx at 7.4% to Pre-COVID at 15.6%, followed by a subsequent rise post-COVID to 23.6% whereas the percentage of individuals with mixed patterns demonstrates a minimal rise from Pre-Tx at 1.1% to Pre-COVID at 4.7%, with a subsequent decrease post-COVID to 3.6%. Finally, abdnormal DLCOc rate shows an increase from Pre-Tx at 36% to Pre-COVID at 42%, followed by a subsequent rise post-COVID to 53%. Functional pulmonary patterns revealed a higher increase of restrictive patterns post-COVID-19. Pre-transplant PF-HCT-CI was not associated with COVID-19 survival (normal 86% vs moderate/severe 88%, p= 0.9).

Conclusions: Most PFT parameters decrease after COVID-19, predominantly marked by DLCO abnormalities, and the most common post-COVID-19 pulmonary pattern alteration was restrictive. Pre-transplant PFTs did not influence mortality after COVID-19.

Clinical Trial Registry: NA.

Disclosure: NA.

13: Infectious Complications

P409 EPSTEIN-BARR VIRUS-RELATED POSTTRANSPLANT LYMPHOPROLIFERATIVE DISORDERS IN THE LETERMOVIR ERA

Jingtao Huang1, Luxiang Wang1, Zengkai Pan1, Zilu Zhang1, Jiayu Huang1, Chuanhe Jiang1, Xiaoxia Hu 1

1Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China

Background: Letermovir is an antiviral drug that significantly decreases cytomegalovirus (CMV) reactivation after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Still, its effects on Epstein-Barr virus (EBV) reactivation and overall benefit for transplant recipients remain uncertain.

Methods: A total of 227 consecutive patients who received allo-HSCT between April 2021 and August 2023 were retrospectively screened. The eligibility criteria included: (1) with a life expectancy ≥3 months; (2) with complete medical records. According to whether the patients received letermovir prophylaxis, the eligible patients (n=222) were divided into two groups: letermovir (n=128) and control group (n=94). We investigated the cumulative incidences of EBV reactivation and posttransplant lymphoproliferative disorders (PTLD) in that wo groups, and identified the risk factors for PTLD. The immune response of the patients with letermovir prophylaxis was evaluated with flowspot assay which measured the interferon-γ (IFN-γ) released by peripheral blood mononuclear cells (PBMC) stimulating with phytohaemagglutinin (PHA).

Results: The cumulative incidences of EBV reactivation within the first 100 days post-transplantation between the letermovir group and the control group were comparable (34.38% vs. 41.49%, P = 0.0852). However, among all patients, the incidence of PTLD was significantly increased in the letermovir group compared with those in the control group (10.16% [13/128] vs. 2.13% [2/94], P = 0.0371). All PTLD patients experienced EBV reactivation at a median of 5 days (range: 0-213 days) before PTLD, and the incidence of EBV-related PTLD was significantly elevated due to the letermovir prophylaxis. (28.26% [13/46] vs. 4.44% [2/45], P = 0.0055). The multivariate logistics analysis identified letermovir as a risk factor for PTLD (odds ratio = 5.13 [95% confidence interval: 1.33-33.93], P = 0.0374). The median interval from transplantation to PTLD was 62 days [range:48-300 days] and we then focus on the early recovery of the immune response after HSCT with letermovir prophylaxis. Of the patients without EBV reactivation (EBV-), the anti-viral capacity of PBMC measured with flowspot has a steady elevation during the first 5 months post-HSCT, especially with a peak at 3 months. A similar rush of median IFN-γ secretion of PBMC was observed in the 2nd month in patients with EBV reactivation but without PTLD (EBV + PTLD-). In contrast, the recovery of immune response of PTLD patients (EBV + PTLD + ) was impaired, and the recovery peaking was not observed within the first 3 months. In the comparison of the IFN-γ secretion capacity of EBV- patients, EBV + PTLD+ patients and EBV + PTLD- patients, patients with EBV + PTLD+ had a lower median value of IFN-γ secretion at the 2nd month compared with the EBV + PTLD- patients (6.955 [range: 0.34-106.96] vs. 42.13[range: 0.61-435], P = 0.0451).

Conclusions: Letermovir prophylaxis might potentially increase the risk of EBV-associated PTLD. EBV reactivation promoted the recovery of the immune response. The immune response was significantly impaired in patients with EBV-related PTLD.

Disclosure: Nothing to declare.

13: Infectious Complications

P410 PATTERN OF SENSITIVITY OF STOOL SURVEILLANCE OVER 13 YEARS AND IMPACT OF STOOL SURVEILLANCE GUIDED SELECTION OF ANTIBIOTIC IN ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANT PATIENTS

Keshav Garg1, Akanksha Chichra1, Davinder Paul1, Anant Gokarn1, Sachin Punatar1, Sumeet Mirgh1, Nishant Jindal1, Lingaraj Nayak1, Sujata Lall1, Vivek Bhat1, Sadhana Kannan1, Bhausaheb Bagal1, Navin Khattry1, Laxma Reddy 2

1ACTREC, Tata Memorial Centre, Mumbai, India, 2Tata Memorial Hospital, Mumbai, India

Background: Allogeneic hematopoietic cell transplant (AHCT) patients are at risk for serious bacterial infections. Our centre has a policy of doing stool surveillance cultures (SSC) weekly from time of admission for AHCT till first discharge to guide antibiotic selection during febrile episodes. We aimed to compare the impact of empirical versus SSC guided antibiotic use during febrile episodes and change in stool sensitivity patterns over a 13-year period.

Methods: This single centre retrospective analysis included AHCT patients between January 2008 -December 2020. SSCs were done at admission and weekly till first discharge and during febrile episodes if readmitted till Day +100 post-transplant. Patients were divided into 4 groups based on baseline SSC- pan sensitive (PS), extended spectrum beta lactamases (ESBL), carbapenem resistant organisms (CRO)and vancomycin resistant enterococcus (VRE). Those with baseline PS SSC received empirical antibiotics (cefoperazone-sulbactam or cefipime-tazobactam with amikacin) in first febrile neutropenia (FN) episode. In the three baseline resistant groups, first line antibiotics were empirical or SSC guided. Change in SSC sensitivity pattern was analysed over two time periods-Cohort 1(2008 - 2014) & Cohort 2 (2015- 2020). The change in SSC sensitivity over first 100 days was captured as persistent PS, persistent resistant, PS to ESBL and/or CRO and PS to CRO (Defined in Legend of Table 1).

Results: Three hundred and fifty eight patients with hematological malignancies were included (ALL-110, AML-110, CML-55, MDS-34, others-49). The median age of the cohort was 27 years (range 3-57 years). There were 269 matched related, 67 haploidentical and 22 matched unrelated donor transplants. Majority of the patients received reduced intensity conditioning. Baseline SSC was PS in 232, ESBL in 78, CRO in 37 and VRE in 11.

Of 348 patients who developed FN in first admission, 136 patients (40%) had defervescence to first line antibiotics. Of the 123 patients who had baseline resistant SSC and developed FN, empirical antibiotics were used in 72 and SSC guided in 51. The use of SSC guided antibiotics led to defervescence with first line antibiotics in 78% vs 18% with empirical antibiotics (P=0.0001). Blood culture positivity during the first episode of FN was seen in 53 (15%) patients; this was higher in empirical vs SSC guided antibiotics (68% vs 31%, P=0.05).

In the two time periods, Cohort 1 & Cohort 2, baseline SSC sensitivity pattern remained similar (Table 1), however more patients changed from sensitive to resistant strains in sequential weekly cultures in Cohort 2 till day +100.

Table 1: Baseline stool surveillance culture sensitivity and change in resistance pattern over two time periods

Characteristic

Cohort 1(1st January 2008-31st December 2014) n=193

Cohort 2(1st January 2015-31st December 2020) n=165

P value

Baseline Stool Surveillance

PS

119(62%)

PS

113(68%)

NS

ESBL

45(23%)

ESBL

33(20%)

CRO

21(11%)

CRO

16 10%)

VRE

8(4%)

VRE

3(2%)

Change in SSC Pattern

n=126

n=112

Persistent PS

83(43%)

Persistent PS

45(27%)

0.0028

Persistent Resistant SSC

4(2%)

Persistent Resistant SSC

PS to ESBL and/or CRO

30(16%)

PS to ESBL and/CRO

41(25%)

0.09

PS to CRO

9(5%)

PS to CRO

26(16%)

0.0008

Conclusions: Stool surveillance culture guided use of first line antibiotics lead to higher rate of defervescence than use of empirical antibiotic approach. Also, more patients have blood culture positivity in the empirical group as possibly organisms remain resistant to empirical antibiotics and grow in blood culture. Incidence of resistant stool cultures has increased over a 13 year period which should raise an alarm on the growing bacterial resistance. More study needs to be done to explore ways to stem this pattern.

Clinical Trial Registry: not applicable.

Disclosure: nothing to declare.

13: Infectious Complications

P411 CYTOMEGALOVIRUS (CMV) LOAD PREDICTS CMV DISEASE AND MORTALITY INDEPENDENT OF POSTTRANSPLANT IMMUNOSUPPRESSION AFTER HEMATOPOIETIC CELL TRANSPLANTATION (HCT)

Alicja Sadowska-Klasa 1,2, Danniel Zamora1, Hu Xie1, Elizabeth R. Duke1, Margaret L. Green3, Masumi Ueda Oshima1,3, Alpana Waghmare1,3,4, Joshua A. Hill1,3, Brenda M. Sandmaier1,3, Keith R. Jerome1,3, Wendy M. Leisenring1,3, Michael Boeckh1,3

1Fred Hutchinson Cancer Center, Seattle, United States, 2Medical University of Gdansk, Gdansk, Poland, 3University of Washington, Seattle, United States, 4Seattle Children’s Hospital, Seattle, United States

Background: CMV viral load has been accepted as a surrogate marker for CMV disease after HCT, and recent interventional trials have used it as primary endpoint. However, the data supporting the surrogacy claim are derived from older cohorts and trials when transplantation techniques differed significantly, and overall mortality (OM) and non-relapse mortality (NRM) were higher. CMV viral load is now used as part of the clinically significant CMV infection endpoint in ongoing and planned clinical trials of novel agents in HCT recipients. The purpose of this study was to define the impact of CMV viral load on CMV disease in the most recent era characterized by different graft versus host disease (GvHD) prevention strategies and improved survival. We specifically analyzed how immunosuppression as measured by lymphopenia affects the correlation between CMV viral load and clinical outcomes.

Methods: CMV seropositive patients undergoing 1st allogeneic HCT in two time periods (1/1/2007-2/28/2013, 3/1/2013-12/31/2017) and weekly CMV DNA PCR surveillance were analyzed. Pre-emptive therapy was administered according to institutional guidelines. No patient received letermovir. Multivariable Cox proportional hazards models were used to estimate the association between CMV viral load by day 100 at different thresholds with CMV disease, OM and NRM up to 1 year post HCT. Landmark analyses in day 100 survivors were used to depict the risk of CMV disease and mortality associated with CMV viral load and lymphopenia using cubic splines.

Results: Of 1539 patients transplanted in the study period, 1394 survived >100 days after HCT and were included in the final analyses. Among these, 1068 (76%) experienced any level of CMV reactivation and 126 (9%) developed CMV disease by one year post HCT. There was a noticeable decline in both OM and NRM in the more recent cohort (1y OM 29.1% vs 25.9%, 1y NRM 26.1% vs 22.8%). Among day 100 survivors, higher viral load was associated with OM (data not shown) and NRM at one year post HCT. CMV viral load was associated with CMV disease and NRM, independent of lymphopenia. However, in the more recent period, the effect of viral load on OM and NRM was the most pronounced in patients with severe lymphopenia by 100, and an interaction between these variables was detected (p=0.03 and 0.06, respectively). The association between viral load and CMV disease was strong and independent of lymphopenia in both periods. In a multivariable model adjusted for age, GvHD presence, GvHD prophylaxis protocols and lymphopenia, CMV viral load was associated with a statistically significant increased risk of CMV disease at all levels, with the highest risk when viral load exceeded 1000 IU/mL (aHR 15.0 95% CI 7.2-30.6), however, the association with OS and NRM (while significant in univariate models) no longer reached statistical significance across all levels (Table 1).

Table 1. Multivariable analysis of factors affecting NRM by 1 year post HCT among day 100 survivors transplanted before 2/28/2013 and after 3/1/2013.

Before 2/28/2013

After 3/1/2013

Covariates

Categories

HR (95% CI)

P-values

HR (95% CI)

P-values

Max viral load (IU/mL) before day 100

Negative

1

1

Positive to 150

1.21 (0.70-2.10)

0.502

1.00 (0.52-1.92)

0.998

>150-1000

2.05 (1.23-3.44)

0.006

1.79 (1.02-3.12)

0.041

>1000

1.84 (1.02-3.33)

0.043

1.08 (0.55-2.09)

0.831

Acute GVHD before day 100

Grade 0-2

1

1

Grade 3-4

2.01 (1.34-3.02)

<.001

3.66 (2.30-5.84)

<.001

Cell source

Bone Marrow

1

1

Cord Blood

2.40 (1.16-4.96)

0.019

2.02 (0.84-4.85)

0.116

PBSC

1.03 (0.61-1.73)

0.909

1.79 (0.88-3.66)

0.111

GHVD prophylaxis

MTX/CNI

1

1

MMF/CNI + PTCy

0.42 (0.26-0.66)

<.001

1.78 (1.05-3.02)

0.033

Sirolimus-based + Other

0.35 (0.15-0.85)

0.021

1.11 (0.58-2.10)

0.758

Type of donor

Matched related

1

1

Haploidentical

1.63 (0.68-3.89)

0.273

2.01 (1.00-4.02)

0.05

Other

1.28 (0.84-1.95)

0.254

1.27 (0.76-2.12)

0.357

Absolute lymphocyte count at day 100 (cells/uL)

≥300

1

1

100-300

2.01 (1.24-3.25)

0.004

1.87 (1.13-3.11)

0.015

<100

6.52 (3.60-11.8)

<.001

8.43 (3.86-18.4)

<.001

Disease risk

Low

1

1

Intermediate

0.83 (0.39-1.75)

0.619

1.27 (0.70-2.32)

0.429

High

2.33 (1.58-3.43)

<.001

1.26 (0.78-2.02)

0.345

HCT CI Score

0-2

1

1

≥3 (including unknown)

1.10 (0.78-1.55)

0.599

1.37 (0.91-2.05)

0.13

Age at Transplant

As continuous

1.03 (1.01-1.04)

<.001

1.01 (1.00-1.02)

0.051

  1. Abbreviations: GvHD- graft versus host disease, PBSC – peripheral blood stem cells, MTX – methotrexate, CNI – calcineurin inhibitors, MMF – mycophenolate mofetil, PTCy – post transplant cyclophosphamide, HCT CI - hematopoietic cell transplantation-specific comorbidity index, NRM - non relapse mortality

Conclusions: CMV viral load continues to be a strong predictor of CMV disease with modern transplantation techniques, however, its impact on OS and NRM appears to be more restricted to lymphopenic patients in recent years.

Disclosure: J.A.H. has served as a consultant for Moderna, Allovir, Gilead, Karius and Takeda and received research funding from Allovir, Gilead, Karius, Oxford Immunotec and Merck.

M.B. Merck – Research support, Moderna – Consulting, research support, Allovir – Advisory Board, Symbio – Consulting, EvrysBio – Advisory Board.

13: Infectious Complications

P412 ADENOVIRUS INFECTION IN ADULT ALLOGENEIC TRANSPLANT RECIPIENTS WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE

Maria Chiara Quattrocchi 1, Giorgio Orofino1, Edoardo Campodonico1, Alessandro Bruno1, Lorenzo Lazzari1, Daniela Clerici1, Francesca Farina1, Simona Piemontese1, Elisa Diral1, Oltolini Chiara1, Raffaele Dell’Acqua1, Sara Racca1, Raffaella Milani1, Elisabetta Xue1, Sara Mastaglio1, Consuelo Corti1, Jacopo Peccatori1, Maria Teresa Lupo-Stanghellini1, Antonella Castagna1, Fabio Ciceri1, Raffaella Greco1

1IRCCS Ospedale San Raffaele, Milano, Italy

Background: In recent years, post-transplant cyclophosphamide (PTCy) has been increasingly adopted as GvHD prophylaxis in haploidentical (haplo) and HLA-matched HSCT. A major drawback of PTCy is the increased risk of viral reactivations, such as CMV and HHV6. Despite the potential impact on morbidity and mortality in HSCT recipients, adenovirus (AdV) infection represents a significant “gap” in the literature. In this context, data on adult population are scarce.

Methods: We carried out a single center analysis to better understand the impact of AdV infection and its outcome in adult patients undergoing allogeneic HSCT in the PTCy era. We analyzed a total of 757 adult patients who underwent HSCT from January 2015 to December 2022 at our center. All patients received PTCy in association with sirolimus (n=31), MMF (N=28) and ATG (n=1) as GvHD prophylaxis. All of them received antiviral prophylaxis with acyclovir and, since 2020, also with letermovir. PCR was used for AdV-DNA determination in biological samples. Until 2018, AdV-PCR was carried out only in case of clinical suspicion; afterwards, we chose to perform universal weekly monitoring on peripheral blood during the early-phase after HSCT, even in asymptomatic patients. We performed AdV-PCR together with immunohistochemistry on histological samples, in case of tissue biopsy done within clinical practice.

Results: Overall, we detected 31 cases of AdV-reactivation, with organ involvement in 21 of them. Patients (median age 47 years) were affected by myeloid (68%) and lymphoid malignancies (32%). Donors were: 55% haplo, 42% matched-unrelated donor (MUD) and only 3% matched-related. Most patients received a myeloablative conditioning regimen (68%). Median time from HSCT to AdV positivity was 53 days (range 3-271). The virus was isolated in plasma samples (n= 26), bone marrow aspirates (n=8), gut biopsy specimens (n=14), urine samples (n=7) and cerebrospinal fluid (n=1). We reported the following clinical manifestations: fever in 4 patients, colitis in 14 patients, cystitis in 7 patients, delayed engraftment in 7 patients and encephalitis in 1 patient. The median absolute CD3+ lymphocyte count at the time of infection was 143 cells/μL (range 2-1334). Ten patients had concomitant acute GvHD, mainly grade III-IV (70%). Overall, 21 patients received specific antiviral treatment according to center guidelines, mainly based on intravenous immunoglobulin and cidofovir. Brincidofovir was administered as compassionate use in case of contraindications (n=5) or unavailability (n=1) of cidofovir. More than 75% of patients achieved complete response, At a median follow-up of 468 days after HSCT (range 31-2235), 26 patients solved the infection while 12 patients died due to AdV infection (n=3) or other non-relapse causes (n=9).

Conclusions: AdV infection in adult HSCT patients is a relevant clinical issue which deserves attention as potential cause of morbidity and mortality. Our analysis reported cases of AdV infections with organ involvement in adult patients undergoing HSCT with PTCy. We reported AdV mainly in the context of haplo and MUD transplants, myeloablative conditioning regimen, low CD3+ lymphocyte counts, grade III-IV acute GvHD. AdV monitoring is warranted in the era of potentially curative AdV treatments including antiviral agents and expanded antiviral specific T-cells products.

Clinical Trial Registry: NA.

Disclosure: None.

13: Infectious Complications

P413 NON-TUBERCULOUS MYCOBACTERIAL INFECTIONS AMONG HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS: A MULTICENTER EUROPEAN CASE-CONTROL STUDY BY THE INFECTIOUS DISEASES WORKING PARTY OF EBMT

Maria Stamouli 1, John Snowden2, Alienor Xhaard3, Awatif AlAnazi4, Antonio Perez Martinez5, Moshe Yeshurun6, Nicolaus Kröger7, Jürgen Kuball8, Fanourios Kontos1, Jakob Passweg9, Stephan Mielke10, Mohsen Al Zahrani11, Luis Miguel Juarez-Salcedo12, Joanna Drozd-Sokolowska13, Fabio Benedetti14, Lucrecia Yañez Sansegundo15, Alessandra Biffi16, Maria Teresa Lupo Stanghellini17, Pavel Jindra18, Alexander Kulagin19, Nuno Miranda20, Urpu Salmenniemi21, Petr Sedlacek22, Gloria Tridello23, Inge Verheggen23, Dina Averbuch24, Rafael de la Camara12

1Attikon University Hospital, Athens, Greece, 2Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom, 3Saint-Louis Hospital, Hematology transplantation, Paris, France, 4King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia, 5Hospital Universitario La Paz, Madrid, Spain, 6Beilinson Hospital, Petach-Tikva, Israel, 7University Medical Center, Hamburg, Germany, 8University Medical Centre, Utrecht, Netherlands, 9University Hospital Basel, Basel, Switzerland, 10Karolinska University Hospital, Stockholm, Sweden, 11King Abdul - Aziz Medical City, Riyadh, Saudi Arabia, 12Hospital de la Princesa, Madrid, Spain, 13Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland, 14Policlinico G.B. Rossi, Verona, Italy, 15Hospital Universitario Marqués de Valdecilla, Santander, Spain, 16Pediatric Hematology, Oncology and Stem Cell Transplant Division, Padua University Hospital, Padua, Italy, 17Ospedale San Raffaele s.r.l, Milano, Italy, 18Charles University Hospital, Pilsen, Czech Republic, 19RM Gorbacheva Research Institute, Pavlov University, Petersburg, Russian Federation, 20Inst. Portugues Oncologia, Lisboa, Portugal, 21HUCH Comprehensive Cancer Center, Helsinki, Finland, 22University Hospital Motol, Prague, Czech Republic, 23EBMT Leiden Study Unit, Leiden, Netherlands, 24Pediatric Infectious Diseases, Faculty of Medicine, Hebrew University of Jerusalem, Hadassah Medical Center, Jerusalem, Israel

Background: Until recently, non-tuberculous mycobacterial (NTM) infections were considered a rare complication that received little attention. Currently though, there have been numerous reports of NTM as rapidly emerging pathogens in the hematopoietic stem cell transplant (HSCT) population. However, data regarding NTM infections in patients undergoing both autologous (auto) and allogeneic (allo) HSCT remain scarce.

Methods: This is a retrospective case-control (1:2) study on post-HSCT NTM infections from 01/01/2005 until 30/09/2022 (22 transplant centers;15 countries). Fifty-five cases and 110 controls were included (100 male/65 female,153 allo-SCT/12 auto-SCT with a median age of 41.1 years, range:0.3-75.8). The study aims to investigate the clinical and microbiological characteristics of NTM infections in HSCT patients, assess treatment options and identify risk factors for these infections.

Results: Fifty-five cases of NTM infections were identified: 51 cases in allo-HSCT and 4 cases in auto-HSCT recipients (34 male/21 female) with a median age of 40.3 years (min-max:0.3-75.8). Underlying diseases were acute leukemia (n=17), MDS/MPN (n=15), plasma cell disorders (n=7), lymphoma (n=5), other (n=11). NTM infections occurred a median of 314 days post-HCT (1 day –19.1 years). Infection was affecting a single site in 37 patients (67.3%). In 11 cases (20%) two sites and in 7 cases (12.7%) ≥3 sites were involved. The most frequently involved organs were lungs (40/55;72.7%) and soft tissue (10/55;18.2%). In 16/55 cases (27.3%) blood cultures were positive for NTM. In 12 cases bacteremia co-existed with other sites of infections, mostly lung (n=7) and soft tissue (n=4). Diagnosis was made by culture (37/55;67.3%), microscopy (10/55;18.2%) and PCR (14/55;25.5%). The NTM involved were: M. Avium (12;21.8%), M. Chelonae (11;20%), M. Intracellulare (8;14.5%), M. Abscessus (5;9.1%), M. Fortuitum (5;9,1%), other (14;25%). Acute graft versus host disease (GvHD) was observed in 25/55 patients and chronic GvHD in 23/55 patients. In 42/55 cases (76.4%) patients were receiving immunosuppressive therapy, most frequently cyclosporine (21;38.2%), corticosteroids (14;25.5%) and ruxolitinib (10;18.2%). Several antimicrobial agents were used in the management of NTM infection, the most frequent being: clarithromycin (22;40.0%), ethambutol (19;34.5%), rifampicin (15;27.3%), azithromycin (15;27.3%) and amikacin (11;20.0%). Hospitalization due to NTM infection was required in 25/55 patients, while 23/55 patients were hospitalized when NTM infection occurred. In 12 cases patients were admitted in ICU. Twenty-two out of 38 evaluable patients had a clinical and/or microbiological cure of NMT infection. Relapse of NTM infection occurred in 3 cases. Overall survival at 12 months, 24 months and 36 months was 51.7%, 43.8% and 39.6% respectively. Death due to NTM infection occurred in 10 patients (18.2%). Acute GvHD (OR 3.9, 1.1-13.7, p=0.03) and diagnosis different from Acute Leukemia (OR 4.9, 1.3-17.8, p=0.02) are significant risk factors for NTM infection in the multivariate model, but these findings are considered exploratory due to low sample size.

Conclusions: Non-tuberculous mycobacterial infections were associated with significant morbidity and mortality in HSCT patients. These infections usually appeared late post-HCT and manifest as pulmonary or soft tissue infections with frequent bacteremia and multiple-site infection. Acute GvHD and diagnosis different from Acute Leukemia were identified as possible risk factors for NTM infection.

Clinical Trial Registry: N/A.

Disclosure: Maria Stamouli: MSD, Pfizer, Astellas, Gilead.

John Snowden: Jazz, Vertex and Medac.

13: Infectious Complications

P414 BACTEREMIA IN THE FIRST + 100 DAYS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: MICROORGANISMS, RESISTANCE AND PROGNOSTIC IMPACT

Merce Aren1, Rosa Pacheco2, Mireia Morgades1, Nadia Güell1, Gerard García-Cirera1, Cristina Blanco-Montes1, Vitor Botafogo1, Maria Izquierdo1, Anna Torrent1, Christelle Ferrà3, Maria Josefa Jiménez1, Josep Maria Ribera1, Juan Manuel Sancho1, Maria Dolores Quesada2, Maria Huguet 1

1Institut Català d’Oncologia-Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain, 2Hospital Universitari Germans Trias i Pujol, Badalona, Barcelona, Spain, 3Hospital del Mar, Barcelona, Spain

Background: Bacteremias are very common in allogeneic transplant (allo-HSCT) recipients and are associated with high early morbimortality. The aim of this study was to analyze the prevalence of blood stream infection in the 100 days following allo-HSCT, the microbiological characteristics, the role of antimicrobial prophylaxis, empirical treatment and its impact on evolution.

Methods: We included 358 allo-HSCT procedures (342 were a first allo-HSCT) performed between March 2010 and December 2022 at our center. The characteristics of patients, the episodes of bacteremia and their evolution were analyzed, as well as the impact of quinolone prophylaxis and the overall survival associated with bacteremia.

Results: Of the 358 allo-HSCT, 144 (40%) had ≥1 episode of bacteremia in the first 100 days post-transplant. The clinical characteristics of donors were analyzed (Table 1) and a higher proportion of haploidentical donors and greater use of post-trasplant cyclophosphamide (PT-Cy) as prophylaxis for graft-versus-recipient disease (GvHD) were observed in the group with blood stream infections. We considered the use of PT-Cy separately and observed that it was associated with a higher number of bacteremia, 58.8% (47/80) vs 34.9% (33/80), p<0.001.

A total of 185 episodes of bacteremia were studied. The median time of onset was 11 days [0-98], most of them being nosocomial (93%) and of unknown origin although abdominal (26.5%) and catheter (17%) foci stood out. Gram-negative bacilli (GNB) were isolated in 48% of cases; 36% gram-positive cocci (GPC); 3% fungi and 13% polymicrobial infections. Resistance to quinolones was observed in 53% of the microorganisms, 32% to piperacillin-tazobactam and 13% were multiresistant.

In our center, antimicrobial prophylaxis was used until 2019 so in 62% of bacteremia episodes, patients were under levofloxacin. Its use was associated with a lower number of blood stream infections, 34.5% (88/255) vs 54.4% (56/103), p=0.001, without observing a decrease in mortality due to infection or improvement in overall survival (OS) but a large increase in quinolones resistance, 75.3% (55/73) vs 22.2% (12/54), p<0.001. Bacteremia or levofloxacin prophylaxis has not been significantly associated with GvHD.

In 36% of bacteremias, empirical antibiotherapy was inadequate due to intrinsic or acquired resistance. The need for admission to the critical care unit was 26% and mortality associated with blood stream infection was 29%. OS at 1 year (95% CI) was 40% (32%-48%) in the group that presented ≥1 bacteremia in the first 100 days post-HSCT vs 55% (48-61%) in the group that did not. At 5 years, 33% (25%-41%) vs 38% (31%-45%).

Table 1. Clinical characteristics of allo-HSCT recipients.

Procedures without bacteremia (n=214)

Procedures with bacteremia (n=144)

P

Age, years (median [extremes])

49,5 [15 - 69]

50 [16 - 70]

0,896

Genre

Male

111 (52%)

89 (62%)

0,063

Female

103 (48%)

55 (38%)

Hematologic disease

Acute leukemia

136 (64%)

82 (57%)

0,315

Lymphoproliferative syndrome

34 (16%)

24 (17%)

Othera

44 (20%)

38 (26%)

Disease status atallo-HSCT

Complete Response

148 (69%)

95 (66%)

0,611

Active disease

66 (31%)

49 (34%)

Type of donor

Related donor

94 (44%)

46 (32%)

<0,001

Unrelated donor

90 (42%)

50 (35%)

Haploidentical

21 (10%)

36 (25%)

Umbilical cord

8 (4%)

12 (8%)

HLA Matching

Identical

151/206 (73%)

80/133 (60%)

<0,001

Mismatch

34/206 (17%)

17/133 (13%)

Haploidentical

21/206 (10%)

36/133 (27%)

Type of conditioning

Myeloablative

92 (43%)

66 (46%)

0,595

Non-myeloablative

122 (57%)

78 (54%)

Cyclophosphamide post-HSCT

Yes

33 (15%)

47 (33%)

<0,001

No

181 (85%)

97 (67%)

  1. aMyelodysplasic syndrome (47), multiple myeloma (4), myeloproliferative neoplasm (19), aplasia (8), granulocytic sarcoma (2), autoimmune bicytopenia (2).

Conclusions: In post-transplant patients bacteremia has a great impact on survival. In our center, most bacteremia were caused by GNB, had a high rate of resistance and inadequate empirical antibiotherapy. Haploidentical transplantation and using PT-Cy is associated with a higher risk of bacteremia. Antibiotic prophylaxis seems to be associated with a lower rate of blood stream infection but a large increase in antibiotic resistance without translating to a benefit in terms of survival.

Disclosure: Nothing to declare.

13: Infectious Complications

P415 ISAVUCONAZOLE THERAPEUTIC DRUG MONITORING-DRIVEN TREATMENT IN A COHORT OF PAEDIATRIC PATIENTS UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANTATION: A SINGLE-CENTRE EXPERIENCE

Carmen Dolores De Luca 1,2, Chiara Rossi1, Chiara Rosignoli1, Federica Galaverna1, Francesca Del Bufalo1, Marco Becilli1, Michele Massa1,2, Raffaele Simeoli1, Emilia Boccieri1, Barbarella Lucarelli1, Antonio Torelli1, Daria Pagliara1, Bianca Maria Goffredo1, Mattia Algeri1, Franco Locatelli1,3, Pietro Merli1

1Bambino Gesù Children’s Hospital, Rome, Italy, 2Sapienza University of Rome, Rome, Italy, 3Catholic University of the Sacred Heart, Rome, Italy

Background: Azoles are a class of antifungal agents characterized by a wide variability in spectrum of action, pharmacokinetics and toxicities, metabolized by hepatic cytochrome P450 enzymes and therefore susceptible to several drug interactions. Isavuconazole has a broad range of activity (molds, yeasts and Mucor) with a half-life of 130 hours and a high volume of distribution. The use of isavuconazole in invasive fungal diseases (such as invasive aspergillosis, invasive candidiasis and mucormycosis) is currently off-label in the pediatric population.

Methods: The aim of this retrospective study is to evaluate the safety, pharmacokinetics, adverse events and efficacy of isavuconazole for treatment and prophylaxis of invasive fungal infections (IFIs) in 46 pediatric patients undergoing hematopoietic stem cell transplantation (HSCT) between 2021 and 2023.

Therapeutic drug monitoring (TDM) of isavuconazole was performed using liquid chromatography-mass spectrometry.

Indication for isavuconazole treatment was related to the likelihood of IFI according to EORTC/MSGERC consensus definitions (IFI proven, probable, possible).

Results: Of the forty-six patients, forty-two received isavuconazole as treatment for IFIs (systemic, pulmonary and sinus aspergillosis together with other IFIs) and four as prophylaxis. Three patients had proven IFIs and thirty-nine had probable IFIs. The characteristics of the population are shown in Table 1.

After loading doses, patients received a maintenance dose of isavuconazole between 5 and 10 mg/kg/day with dose adjustment in accordance with TDM (Figure 1). Twenty-nine out of forty-six patients had a plasma level of isavuconazole below the therapeutic range at least once during the course of antifungal treatment. The median time that isavuconazole plasma levels remained within the therapeutic range was thirty-four days at a median dose of 5 mg/kg/day for a median treatment duration of one hundred and ten days.

The overall response rate (ORR) to treatment with isavuconazole was 80.4% (37/46); of these thirty-seven patients, 54% (20/37) achieved a complete resolution of IFIs after one month from treatment start.

Grade 3-4 adverse events (according to CTCAE v5.0) were reported in only 13% of patients (6/46) and included: somnolence, hypertransaminasemia, polyneuropathy, possibly related to the drug.

No patients died for fungal invasive infections and isavuconazole-related complications.

Exploring drug interactions, the most notable reported was between isavuconazole and nirmatrelvir-ritonavir antiviral treatment, a potent inhibitor of CYP3 A4, which resulted in an increase in isavuconazole plasma levels and the consequent decision to temporarily discontinue antifungal therapy.

Noteworthy, chemotherapy had no effect on isavuconazole plasma levels, and calcineurin inhibitors induced an increase in isavuconazolemia in only three out of twenty-six patients.

Table 1

n° of patients

46

Sex

F

15 (32.6%)

M

31 (67.4%)

Disease

ALL

13 (28.3%)

AML

17 (36.9%)

BDs

9 (19.6%)

Other (RCC, NB, NHL)

7 (15.2%)

Phase of disease

CR

26 (56.5%)

AD

20 (43.5%)

Type of Transplant

Haploidentical

19 (41.3%)

MUD

21 (45.7%)

Sibling

6 (13.0%)

Indication to isavuconazole treatment

Systemic Aspergillosis

11 (23.9%)

Neutrophils/Lymphocytes at start of isavuconazole therapy (median) [n° cell/mmc]

960/510

Chemotherapy during Isavuconazole treatment

38 (82.6%)

IFIs persistent/reactivation

15 (32.6%)

Relapse/refractory disease

10 (21.7%)

aGvHD concomitant with isavuconazole treatment

6 (13.0%)

cGvHD treated during isavuconazole treatment

3 (6.5%)

Death

9 (19.6%)

  1. AD: active disease; aGVHD: acute graft versus host disease; BD: benign diseases; cGVHD: chronic graft versus host disease; CR: complete response; IFIs: invasive fungal infections; MUD: mismatched unrelated donor; NB: neuroblastoma; NHL: Non-Hodgkin lymphoma; RCC: refractory cytopenia of childhood.
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (46)

Conclusions: Isavuconazole is a safe and effective drug for the treatment of IFIs caused by Aspergillus, as well as by other molds and yeasts also in children. The manageability of the drug is supported by both a low frequency of side effects and low drug interactions, this making its use particularly attractive in settings such as HSCT.

Disclosure: Nothing to declare.

13: Infectious Complications

P416 THINK TO CHECK PREVIOUS SEROLOGIES WHEN CMV OR TOXOPLASMA SEROLOGIES ARE NEGATIVE AT TRANSPLANT

Christine Robin 1, Ludovic Cabanne1, Florence Beckerich1, Anne Le Bouter1, Francoise Foulet1, Catherine Cordonnier1

1Henri Mondor Hospital, Creteil, France

Background: Because of the risk of reactivation and its subsequent post-transplant morbidity and mortality for these two pathogens, identifying the CMV and toxoplasma serostatus before HCT is crucial to choose optimal prophylactic and preemptive strategies. However, some hematological diseases and therapies may decrease the antibody titers overtime, inducing a possible false seronegativity at transplant in previously seropositive patients. This may preclude to adopt the best individual strategy after transplant. The aim of this study was to evaluate the rate of negativation of CMV and toxoplasma serology between the diagnosis of the underlying disease and the pretransplant check-up.

Methods: Among the 405 patients who consecutively received an allogeneic HCT between 01/01/2012-30/06/2022 in our department, 295 patients in whom CMV and/or toxoplasma serologies were available both at diagnosis and at transplant were selected for the present study. Factors associated with seronegativation were analyzed among initially seropositive patients. The “limit” values were those defined by the manufacturer of the test and extended by the laboratory according to measurement uncertainty and were considered as positive. Qualitative variables were described as numbers (%) and compared using the chi-2 test or Fisher exact test as appropriate. Quantitative variables were described as median (interquartile range, Q1-Q3) and compared using the Kruskall-Wallis test. Univariate and multivariate analyses were performed using logistic regression.

Results: The clinical characteristics of the patients are shown in table 1. Thirty patients (10%) had received rituximab (n=7), blinatumomab (n=12) and/or other monoclonal antibodies (MoAbs) during the treatment of the underlying disease. Among the 287 patients with available CMV serology at diagnosis and at transplant, 174 (61%) were initially CMV + . Among them, 4 (2.3%) patients became seronegative before transplant. None factor was significantly associated with CMV seronegativation.

Among the 167 patients with available toxoplasma serology at diagnosis and at transplant, 111 (66%) were initially toxoplasma + . Among them, 6 (5.4%) patients became seronegative before transplant. In univariate analysis, female sex, acute lymphoblastic leukemia (ALL) or lymphoproliferative disorder (LPD) and LPD, and the use of monoclonal antibodies were associated with toxoplasma seronegativation. The rate of seronegativation for toxoplasma was 4/21 (19%) in the ALL/LPD patients, and 3/10 (30%) in those who received MoAbs. There was a strong association between ALL/LPD and the administration of monoclonal antibodies. In multivariate analyses, two models were performed: both monoclonal antibodies (OR 14.0 [2.37-82.56], p=0.004) and ALL/LPD (OR 10.35 [1.75-61.09] p=0.01) were significantly associated with toxoplasma seronegativation. The overal results were not significantly different when considering the limit values as negative (data not shown).

Characteristics of the 295 patients

N=295

Sex (n=295)

Female

131 (44%)

Male

164 (56%)

Age at transplant (n=295)

Mean

52.1

Median

56.1

IQ25-75

42.6 - 63.9

Ranges

16.4 - 73.8

Underlying disease (n=295)

Acute leukemia

247 (84%)

Lymphoproliferative disease

23 (8%)

Myeloproliferative neoplasm

19 (6%)

Non-malignant disease

6 (2%)

Acute lymphoblastic leukemia and lymphoproliferative diseases (n=295)

60 (20%)

Complete Remission at transplant (n=256)

197 (77%)

HLA match (n=295)

Identical sibling

87 (29%)

Mismatched relative

38 (13%)

Unrelated

170 (58%)

Rituximab or other Monoclonal antibody (MoAb) (n=295)

30 (10%)

Timing between last dose of MoAb and alloHCT (days) (n=27)

Mean

61.6

Median

43

IQ25-75

27 - 63

Ranges

10 - 265

Previous autologous HCT (n=295)

20 (7%)

CMV serology of the recipient at diagnosis (n=287)

Negative

113 (39.5%)

Limit

4 (1.5%)

Positive

170 (59%)

CMV serology at transplant (n=287)

Negative

98 (34%)

Limit

12 (4%)

Positive

177 (62%)

Toxoplasma serology of the recipient at diagnosis (n=167)

Negative

56 (34%)

Limit

2 (1%)

Positive

109 (65%)

Toxoplasma serology at transplant (n=167)

Negative

58 (35%)

Limit

2 (1%)

Positive

107 (64%)

Conclusions: Two per cent of previously CMV-seropositive patients and 5% of previously toxoplasma-seropositive patients became seronegative at the pretransplant checking and may not benefit of specific prophylactic or preemptive management. For CMV- or toxoplasma-seronegative patients at transplant, it is important to check serology at diagnosis each time possible, especially in patients with ALL or lymphoproliferative disorders or those who received monoclonal antibodies. The serology at the diagnosis of the underlying disease should be the one taken in consideration for adapting the management of the patient after transplant.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

13: Infectious Complications

P417 IMMUNE POPULATIONS PREDICT CONTROL OF CMV REACTIVATION AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Paula Díaz-Fernández 1,2,3, Valle Gómez García de Soria1,2,3, Javier Sevilla-Montero1,2, Ana Marcos-Jiménez1,2, José María Serra López-Matencio1,2, Laura Cardeñoso1, Ángela Figuera Álvarez1,2,3, Rafael de la Cámara1,2, Cecilia Muñoz-Calleja1,2,3

1Hospital Universitario de la Princesa, Madrid, Spain, 2Instituto de Investigación Sanitaria Princesa, Madrid, Spain, 3Universidad Autónoma de Madrid, MadridSpain, Spain

Background: CMV reactivation is a common complication of allogeneic hematopoietic stem cell transplantation (alloHSCT). Easily applicable biomarkers are needed to identify patients who will reactivate the virus.

Aim: We aimed to define a model that could predict CMV reactivation and the need for antiviral treatment.

Methods: This is a prospective study of 59 alloHSCT patients transplanted at our institution between 2017 and 2019 who received high-dose acyclovir as CMV prophylaxis and were weekly monitored up to 100 days after HSCT.

Donor/recipient (D/R) CMV serostatus was as follows: D + /R+ (n=39; 66.10%), D-/R+ (n=15; 25.42%) and D + /R- (n=5; 8.48%). D-/R- pairs were not included.

Patients were retrospectively classified into three outcome groups: non-reactivating patients (NR; n=12; 20.34%), CMV-reactivating patients with no need for antiviral treatment to clear the viremia (viremia controllers-VC; n=16; 27.12%) and patients requiring antiviral treatment (viremia non-controllers-VNC; n=31; 52.54%). CMV reactivation was defined as any detectable CMV DNA in plasma.

Those groups were assigned a postransplant reference week. For VC and VNC, this corresponded to the CMV DNA peak within the first reactivation episode and the start of viral treatment, respectively. NR were assigned the seventh week as their reference, based on the median reference week for the VC and VNC groups.

Percentages of peripheral blood lymphocyte subpopulations based on the expression of HLA-DR, CD25 and NKG2C molecules were identified by flow cytometry the week prior to the reference week.

Results: The percentage of CD8 T cells expressing HLA-DR (CD8 + DR+ cells) was higher in VNC compared to NR (p<0.0005) and VC (p=0.0120) on the week before the reference point. Conversely, the percentage of NKG2C + NK cells was increased in the VC when compared to both NR (p=0.0111) and VNC (p=0.0224). Recipient’s age and D/R CMV serostatus also showed significant differences amongst the three groups.

We therefore established a multinomial logistic regression model with the four aforementioned variables serving as predictors, aiming to anticipate patients’ NR, VC or VNC outcome group. NR patients served as the reference category for OR calculations. This multivariate model revealed that higher percentages of CD8 + DR + T cells were associated with VNC (OR=1.0985, p=0.0072) when compared to NR. A higher percentage of NKG2C + NK cells was associated with VC (OR = 1.4768, p=0.0187). D + /R- serostatus decreased the risks of being VC (OR=2.1984 · 10-5, p < 0.0001) or NVC (OR=0.0275, p=0.0418) compared to NR, but increased the risk of being NVC (OR=1.408·103, p<0.0001) compared to VC.

Our model showed a patient classification accuracy of 79.7% and a ROC AUC of 0.894. Finally, external 4-fold cross-validation showed an accuracy of 67.7% and a ROC AUC of 0.754.

Conclusions: We have designed and cross-validated a highly accurate model capable of stratifying patients within NR, VC and VNC in the first 100 days after alloHSCT. This may aid in anticipating CMV reactivation and its need for treatment with the consequent benefit for the patients’ transplant outcome and clinical care. Further data under letermovir prophylaxis are needed to confirm these findings.

Disclosure: Nothing to declare.

13: Infectious Complications

P418 NEW ANTIMICROBIAL STEWARDSHIP PROGRAM TOOL TO REDUCE THE EMPIRICAL ANTIBACTERIAL THERAPY IN HAPLO-HSCT

Yuliya Rogacheva1, Marina Popova 1, Alexandr Siniaev1, Yuliya Vlasova1, Ivan Moiseev1, Sergey Bondarenko1, Alexander Kulagin1

1RM Gorbacheva Research Institute, Pavlov University, Saint Petersburg, Russian Federation

Background: The increase in antibiotic resistant bacteria infections in allo-HSCT recipients forces the implementation of rational empirical antibacterial therapy (EABT) protocols. Fever early after HLA mismatched allo-HSCT, especially from haploidentical donor (haplo-HSCT), may be associated with cytokine release syndrome (CRS). According to standard antimicrobial stewardship program, most of the patients are prescribed excessive EABT. The differential diagnosis of fever due to infections or CRS after haplo-HSCT is a challenging now. We report data of the study using clinical and laboratory sign of CSR, procalcitonin and lactate as a markers of differential diagnosis and start of EABT for patients after haplo-HSCT with post-transplant cyclophosphamide (PTCY) based graft-versus-host disease prophylaxis.

Methods: Study group included 64 patients after haplo-HSCT with PTCY, where new protocol was introduced. The control group consisted of 61 haplo-HSCT recipients with PTCY, in whom EABT was started without differential diagnosis of fever. Patients and HSCT characteristics are outlined in the table 1. First endpoint of the efficacy was the proportion of haplo-HSCT recipients where EABT was not initiated, the safety endpoints were the incidence of bloodstream infection (BSI), severe infections/sepsis and the 30-day overall survival.

Table 1. Patients and HSCT characteristics

Characteristics

Study group

Control group

P

n=64

n=61

05.2020-12.2022

01.2018-04.2020

Age, years, median (range)

34 (18-64)

32 (18-66)

NS

Gender, male, n (%)

43 (67,2)

40 (65,6)

0,849

Diagnosis, n (%)

>0,05

AML

31 (48)

30 (49)

ALL

24 (37,5)

16 (26,2)

MPN

4 (6,3)

4 (6,5)

АA

3 (4,6)

3 (4,9)

Other

2 (3,6)

8 (13,4)

Relapse/active disease, n (%)

12 (19)

17 (28)

0,228

Myeloablative conditioning regimen, n (%)

40 (62,5)

40 (65,5)

0,721

Neutropenia grade 4 on Day 0 – Day +4, n (%)

8 (12,5)

7 (11,4)

0,861

CRE-colonization, n (%)

3 (4,6)

3 (4,9)

0,952

CRS, n (%)

grade 1-2

18 (28)

-

grade 3-4

8 (12,5)

-

  1. AML - acute myeloid leukemia, ALL - acute lymphoblastic leukemia, MPN - myeloproliferative neoplasms, AA - aplastic anemia, MAC - myeloablative conditioning, CRE - carbapenem-resistant Enterobacteriaceae, CRS - cytokine release syndrome

Results: Fever during first 4 days after haplo-HSCT developed in 27 (42%) patients of the study group and 18 (30%) in the control group, p=0,140. EABT was prescribed in 2 (7,4%) patients on D + 2 in study group (lactate – 5,6 mmol/l (n=1) and procalcitonin > 2 μg/l (n=1)) and in 18 (100%) patients on D0-D + 4 in the control group. Prescribing of the EABT was reduced in 92,6% of patients in study group (p<0,001). In the following pre-engraftment period febrile neutropenia (FN) required the EABT occurred in 22 (88%) patients of study group with the median 10 (5-14) days. Nine (33%) and 8 (44%) patients developed BSI in the study and control group, respectively (p=0,452). The median day of BSI diagnosis was 14 (5-18) and 15 (3-30) days, respectively. Severe infections/sepsis evolved in 5 (18,5%) and 6 (33%) patients in study and control group (p=0,258). Overall survival during 30 days after haplo-HSCT in patients who developed fever in D0-D + 4 in study and control groups was 100% vs 78.9%, p=0,013.

Conclusions: The proposed protocol for the differential diagnosis of fever leads to significant reduction EABT in early period after haplo-HSCT. There was no difference in the safety end-points. Improvement of overall survival in the study group can be attributed to the results of applying the сolonization-guided protocol for EABT (O099 Popova et al., the 47th Annual Meeting 2021).

Disclosure: Nothing to declare.

13: Infectious Complications

P419 LETERMOVIR IS SAFE AND EFFECTIVE FOR TREATMENT AND PREVENTION OF CMV REACTIVATION IN PAEDIATRIC ALLOGENEIC STEM CELL TRANSPLANT

Nathanael Lucas 1, Abbey Forster1, Malcolm Guiver1, Ramya Nataraj1, Madeleine Powys1, Noor Barotchi1, Omima Mustafa1, Srividhya Senthil1, Robert Wynn1

1Manchester University NHS Trust, Manchester, United Kingdom

Background: Cytomegalovirus is a major cause of morbidity, and mortality in paediatric haematopoietic stem cell transplant (HSCT). Letermovir is an antiviral that binds the CMV-terminase complex and is both licensed and commissioned for prophylaxis of CMV reactivation in post pubescent children undergoing HSCT. It is not commissioned for pre-pubertal children in the UK. We aim to describe our experience with letermovir as a prophylactic (PPX) and treatment (TRT) agent in both the pre-pubertal (<12 years) vs post-pubertal (≥12 years and ≤18 years) paediatric population HSCT.

Methods: We completed a single-centre, retrospective cohort study of all CMV-seropositive children aged <18 that received Letermovir during HSCT in our unit between November 2021 and November 2023. Letermovir was accessed by meeting commissioning criteria or individually funded request methods therefore use was not uniformly PPX or TRT. A complete chart review including demographics, PPX versus TRT, letermovir treatment course, liver function tests, CMV viral loads, and sentinel safety events were recorded.

Results: Twenty-three children met inclusion criteria. Median age was 11 (Range 3-17), with 13 Males (57%). All were CMV serologically positive for prior to HSCT, and 20 had negative viral loads prior to transplant. Fifteen received Letermovir as PPX, and 8 TRT. The PPX group was mainly post-pubescent with only one patient under 10 (Mean age 13.1, Range 4-18). In the PPX group 10/15 (66%) had negative viral loads throughout HSCT, three had transient reactivation following cessation of letermovir and two had reactivation during treatment. None of the prophylactic group had CMV related morbidity. The TRT group were younger (N=8, Mean age 6.25, Range 4-10), received longer courses on average (TRT: 156 days, PPX: 127 days). All TRT patients also had conventional therapy, with five having clear benefit from addition of Letermovir. The median highest CMV viral load log was 3.95 (Range 2.85-4.89). Four patients in this group died for non-CMV related reasons. One patient had successful treatment of CNS CMV infection. Only one patient stopped Letermovir –due to rising LFT’s and a diagnosis of liver GVHD. No side effects, including no myelosuppression were noted in chart review for any patients.

Conclusions: We present the biggest cohort in the paediatric population, demonstrating letermovir is a safe, effective, and useful medication in reducing Cytomegalovirus induced morbidity and mortality in paediatric HSCT. In our cohort using Letermovir as prophylaxis decreases duration of therapy, decreases therapy failure, and had better long term outcomes. As a treatment where other therapies have failed it can induce responses, including in CNS disease. The drug urgently needs to be made available as prophylaxis for all seropositive children undergoing HSCT.

Disclosure: Nothing to declare.

13: Infectious Complications

P420 DUAL METAGENOMICS NEXT-GENERATION SEQUENCING FOR FIRST-LINE DIAGNOSIS OF BLOODSTREAM INFECTION IN HEMATOLOGIC PATIENTS WITH FEBRILE NEUTROPENIA: A MULTICENTER PROSPECTIVE STUDY

Yuqian Sun 1, Jingrui Zhou1, Yue Yin2, Jianping Zhang3, Meixiang Zhang4, Yun He1, Wei Gai5, Xiaohui Zhang1, Yu Wang1, Lanping Xu1, Kaiyan Liu1, Xiaojun Huang1

1Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China, 2Peking University First Hospital, Beijing, China, 3Hebei Yanda Lu Daopei Hospital, Hebei, China, 4Peking University International Hospital, Beijing, China, 5WillingMed Technology (Beijing) Co., Ltd, Beijing, China

Background: Bloodstream infection (BSI) is a lethal complication in hematologic patients with febrile neutropenia (FN). Unfortunately, blood culture (BC) can only identify pathogens in 20-30% of patients with BSl. We aim to evaluate the diagnostic performance of dual metagenomics next-generation sequencing (mNGS) as a first-line method in BSI.

Methods: This study was prospective performed in four Chinese hematologic centers since October 2021. For consecutive patients aged ≥15 years with hematologic diseases, patients were screened at the onset of neutropenia. At the onset of FN, peripheral blood specimens were collected per subject for simultaneous BC and dual mNGS (separate sequencing for plasma and blood corpuscle). All patients will be followed for 14 days, or until death before 14 days. The intervention was per the attending physician. The results of dual mNGS bio-informatic analysis were determined according to the reference literature, the abundance in each sample and relative to other samples in the cohort. Clinical physician and mNGS analysis team were double-blind. The primary endpoint of this study was the diagnostic performance of dual mNGS. The positive agreement was defined as mNGS identified at least one same pathogen as initial BC. The negative agreement was defined as both mNGS and BC were negative. And the adjudication of the result of BC, mNGS, and clinical diagnosis was evaluated by 4 independent specialists in hematology. Clinical adjudication categorized mNGS results as: Definite, mNGS identified at least one pathogen also found by BC or standard microbiological testing within 7 days; Probable, mNGS result was concordant with a clinical diagnosis of BSI in FN; Possible, mNGS result was consistent with BSI but not considered a common cause of FN; Unlikely, mNGS result was not a plausible cause of BSI in FN.

Results: A total of 300 FN events were enrolled, including 62 definite BS, 61 probable BSI, 116 infectious FN other than BSI, 55 non-infectious FN events and 6 FN of indeterminate cause. The sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV) of dual mNGS were 95.2%, 94.6%,95.2%, and 94.6%, respectively. The diagnostic time of dual mNGS was significantly shorter than BC (39.7 + 15.0 vs. 119.8 + 31.9 h, p < 0.0001). Real-time availability of dual mNGS results could have allowed early optimization of agents in 52.3% events, by addition or escalation of antibacterial in 31.7%, and antimicrobial narrowing in 20.6%.

Conclusions: The findings suggest that a novel dual mNGS approach has excellent diagnostic performance for the early diagnosis of BSl in FN patients with hematological disorders. The study will promote early diagnosis and better management of the patients.

Clinical Trial Registry: ClinicalTrials.gov ID: NCT05149547.

Disclosure: Nothing to declare.

13: Infectious Complications

P421 CYTOMEGALOVIRUS-SPECIFIC T CELL IMMUNITY RECONSTITUTION AFTER LETERMOVIR WITHDRAWAL IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION RECIPIENTS

Luxiang Wang1, Jingtao Huang1, Jiayu Huang1, Zilu Zhang1, Zengkai Pan1, Chuanhe Jiang1, Sujiang Zhang1, Xiaoxia Hu 1

1National Research Center for Translational Medicine, State Key Laboratory of Medical Genomics, Shanghai Institute of Hematology, Ruijin Hospital, Shanghai JiaoTong University School of Medicine, Shanghai, China

Background: Reactivation of cytomegalovirus (CMV) leads to significant morbidity and mortality following allogeneic hematopoietic stem cell transplantation (allo-HSCT). Letermovir (LTV) has substantially reduced the risk of clinically significant CMV infection (csCMVi) in CMV seropositive recipients of allo-HSCT. LTV discontinuation after day 100 (d100) has been reported to increase the risk of late-onset csCMVi, causing by impaired reconstitution of CMV-specific T immunity.

Methods: A total of 127 transplant recipients was retrospectively analyzed between August 2022 and October 2023. CMV IFN-γ FlowSpot (CMV-F) assay was applied to measure CMV-specific T cell response at d60 and d90 after allo-HSCT, and one-month interval, thereafter. The patients were divided into two groups: 47 patients who stopped LTV within d100 without CMV-F reference (LTV-N group) and 80 patients who discontinued LTV when CMV-F ratio ≥1.5(LTV-F group). The median age was 52 (range 16-69) and 42 (range 18-69, p = 0.08), respectively, for the LTV-N and LTV-F group. Most of the patients (LTV-N vs. LTV-F, 91.4% vs. 95.0%, p = 0.43) were conditioned with myeloablative regimen. 89.3% of LTV-N group (n=42) and 75.0% of LTV-F group (n=60) received graft from haploidentical (HID) donor (p = 0.32).

Results: Cumulative incidence of csCMVi at d100, d180 and d365 was 3.12%, 11.00%, and 19.26%, respectively, for the entire cohort. Three patients (2.3%) developed CMV disease within day 180, and 7 patients (5.5%) developed CMV disease between day 180-365. The median duration of LTV exposure was 98 (range: 83-112) and 117 days (range: 56-216, p = 0.09), respectively, for LTV-N and LTV-F groups. Within 60 days after LTV withdrawal, the cumulative incidence of late-onset csCMVi was 16.27% and 3.22%, respectively, in LTV-N and LTV-F group. The median occurrence time for late-onset csCMVi was d130 (range: 110-150) and d204 (range: 127-278), respectively, for LTV-N and LTV-F group (p = 0.02). CMV-specific T cell reconstitution was markedly delayed in patients with late-onset csCMVi compared to those without (CMV-F ratio: 0.50 vs. 1.20, p = 0.03) by d120. The CMV-F value was more stable for patients who did not experience late-onset csCMVi in serial monitor timepoints. Logistic‐regression analysis showed that a lower elevated value of CMV-F at d120 was associated with an increased risk for late-onset csCMVi (HR 0.32; 95% CI 0.052-0.987, p = 0.15). In multivariate analyses, patient age (>55 year, HR 1.50; 95% CI 1.006-1.111, p = 0.04), donor CMV serostatus (>69.5AU/ml, HR 0.108; 95% CI 0.005-0.711, p = 0.049) and steroid exposure (>1mg/kg, HR 2.529; 95% CI 0.740-8.811, p = 0.137) were associated with late-onset csCMVi in HID allo-HSCT.

Table 1 Multivariate analysis.

Variables

HR

95% CI

p

patient age (>55 years)

1.052

1.006-1.11

0.040

CMV serostatus (>69.5AU/ml)

0.108

0.005-0.711

0.049

exposure to steroid (>1mg/kg)

2.529

0.740-8.811

0.137

Conclusions: In summary, late-onset csCMVi after letermovir withdrawal should be closely monitored in allo-HSCT patients, if not, in HID HSCT. For high-risk patients for late-onset csCMVi, prolonged letermovir prescription is warrant. CMV-specific T cell response test is a reliable indicator to direct prolonged usage of letermovir for late-onset csCMVi prophylaxis.

Disclosure: This work was supported by the National Key Research and Development Program of China (2022YFC2502600 to X.H.), the National Natural Science Foundation of China (82170206 to X.H., 82000156 to L.W.), Shanghai Municipal Health Commission Project of Disciplines of Excellence (20234Z0002 to X.H.) and Shanghai Sailing Program grant (20YF1439700 to L.W.).

13: Infectious Complications

P422 CMV REACTIVATION AND ITS IMPACT ON SURVIVAL AFTER ALLOGENEIC STEM CELL TRANSPLANTATION IN LETERMOVIR ERA – A SINGLE CENTRE STUDY

Lana Desnica 1, Nadira Durakovic1, Zinaida Peric2, Zrinka Bosnjak1, Ranka Serventi Seiwerth1, Dubravka Sertic1, Ante Vulic1, Mirta Mikulic1, Violeta Rezo Vranjes1, Pavle Roncevic1, Drazen Pulanic1, Nurka Rustan1, Zorana Grubic1, Radovan Vrhovac1

1University Hospital Center Zagreb, Zagreb, Croatia, 2University Hospital Center Rijeka, Rijeka, Croatia

Background: CMV infection remains a major cause of morbidity and mortality after allogeneic stem cell transplantation (allo-HSCT) despite decades of challenging treatment and diagnostic advances. Prophylaxis with letermovir significantly reduced the rate of clinically significant CMV infections and all-cause mortality.

Methods: CMV reactivation free and overall survival was analysed in 262 consecutive patients who underwent allo-HSCT from 8/2019 to 12/2022 and received letermovir prophylaxis.

Results: Median patients’ age was 50 years (range 17-71, SD 13,4), 51% were men, with 60% of the transplants from unrelated donors (75% 10/10, 25% 9/10), followed by related (24%) and haploidentical in 16% of patients. Most patients received a PBSC graft (81%) preceded by an RIC or NMA conditioning regimen (76%). Sixteen patients received 480 mg of letermovir daily, and all others 240 mg while on cyclosporine for GvHD prophylaxis. Patients started letermovir from a median of 8 days (range 0-28, SD 6,2) after allo-HSCT until they finished a total of 84 tablets or until CMV reactivation. With a median follow-up of 260 days (range 1-1630), cumulative incidence of CMV reactivation in the whole cohort was 38.2%, and overall survival on the study at 4.5 years was 55.6% (median follow-up 605, range 16-1630 days). Patients with CMV reactivation had worse overall survival (48.2% vs 58.9%, p=NS). Incidence of CMV reactivation was highest in patients receiving haploidentical transplants with PTCy, followed by 10/10 MUD, 9/10 MUD and MRD transplants (63.3%, 41.3%, 33.3%, 19.1%, p<0.001), respectively. Patients receiving transplants from CMV-seronegative donors had higher reactivation rates than patients receiving transplants from CMV-seropositive donors (50.7 vs 30.8%, p=0.002). Patients with low but detectable viral loads (<137 IU/ml) were also started on letermovir prophylaxis, and were found to have higher cumulative incidence of CMV reactivation compared to patients who were CMV DNA negative (47.5 vs 35.6%, p=0.029). Age and intensity of conditioning did not influence CMV reactivation. However, in our cohort patients’ gender had an impact on CMV reactivation, women reactivating more often than men (47.2 vs 29.6%, p=0.01). Furthermore, patients starting letermovir before day 8 had not only more CMV reactivations than patients who started prophylaxis later (46.4 vs 30.7%, p=0.02) but also worse overall survival (53.8 vs 61.5%, p=0.016).

Conclusions: This retrospective single-centre study revealed a >20% decrease in CMV reactivations compared to historic results before letermovir prophylaxis was introduced. This study also confirmed that haploidentical transplants with PT-Cy are accompanied with the highest rates of CMV reactivation. Interestingly, with women reactivated CMV more often than men, a finding that requires additional research. Finally, the finding that patients who started letermovir early (before day 8) reactivated CMV more often could probably be explained by earlier discontinuation of their prophylaxis (before day 100), probably contributing to their worse outcomes.

Disclosure: no disclosures.

13: Infectious Complications

P423 IMPACT OF QUANTIFERON-CMV (QNF-CMV) ON LATE ONSET CLINICALLY SIGNIFICANT CMV INFECTION (CSCMVI) AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT)

Chiara Maria Dellacasa 1, Roberto Passera1, Carolina Secreto1, Luisa Giaccone1, Irene Dogliotti1, Jessica Gill1, Davide Stella1, Aurora Martin1, Federica Ferrando1, Sofia Zompi1, Alessandro Busca1

1AOU Città della Salute e della Scienza di Torino, Torino, Italy

Background: Late onset (after day 100) csCMVi still represents a challenging issue in allogeneic HSCT patients receiving Letermovir (LTV) prophylaxis. Monitoring for T cell response through CMV specific T cell assays may predict patients’ susceptibility to csCMVi.

The aim of this retrospective single center study was to evaluate the impact of day 100 QFN-CMV assay on csCMVi development.

Methods: Overall, 89 consecutive patients received an allogeneic HSCT at our Institution between March 2021 and March 2023.

All CMV seropositive patients (n =78, median age 58 years, range 21-71) received LTVprophylaxis from day + 7 to day + 100 post HSCT, and were included in the study.

The donor was a matched sibling in 16 cases, unrelated in 44 and haploidentical in 18 patients. In total 51 pairs (65%) were CMV D + /R+ and 27 (35%) were D-/R + .

44 patients received myeloablative and 34 received reduced intensity conditioning regimen.

GVHD prophylaxis included anti-thymocyte globulin (ATG) in 37 patients and post transplant Cyclophosphamide in 36 patients.

QFN-CMV assay was performed in 56 out of 78 patients at day 100: in 29 cases (52%) QFN-CMV was reactive and in 27 (48%) it was non-reactive/indeterminate.

CsCMVi was defined as CMV disease or CMV viremia leading to pre-emptive treatment.

Results: A grade II-IV acute GVHD was diagnosed in 36 (46%) patients and a moderate or severe chronic GVHD was detected in 29 (37%) patients.

5 patients died from transplant complications before day 100.

With a median follow up of 433 days (range 14-972), cumulative incidence of cs-CMVi was 20.5% at 6 months and 24.7% at 12 months post HSCT. After LTV discontinuation, csCMVi was detected in 19 patients (24%), at a median time of 147 days (range 125-350) post transplant: 12 of them had a non reactive/indeterminate QFN-CMV at day 100. 5 patients developed organ disease (three gastrointestinal and two lung disease). Lymphocyte counts at the time of csCMVi ranged from 150 to 8400/mmc (median 610/mmc). All the 19 patients were receiving immunosuppressive treatment and 16 of them had active chronic GVHD at the time of cs-CMVi diagnosis. Nine patients experienced more than one csCMVi episode (range 1-4) and two of them developed Valganciclovir resistence.

Death attributable to csCMVi occurred in one patient with pneumonia due to CMV and SARS-CoV2.

A non reactive/indeterminate QFN-CMV value at day 100 was the only identified risk factor for csCMVi (p = 0.021). A trend towards a stronger susceptibility to csCMVi was observed in patients with acute (p = 0.09) and chronic GVHD (p = 0.059), and in D-/R+ patients (p =0.09). Numbers are too small to define an impact of QFN-CMV values on post transplant mortality.

The sensitivity and specificity of reactive QFN-CMV at day 100 were 63.2% and 72.2% for predicting freedom from csCMVi after day 100; the positive and negative predictive value were 82.8% and 48.1% respectively.

Conclusions: QNF-CMV may represent a useful tool to predict post transplant specific immune reconstitution against CMV, potentially leading to risk-adapted therapeutic decisions. Additional prospective studies are needed to confirm these data.

Disclosure: Nothing to declare.

13: Infectious Complications

P424 THERAPEUTIC DRUG MONITORING (TDM) OF PIPERACILLIN IN PATIENTS WITH NEUTROPENIC FEVER AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Sabrina Kraus1, Prisca Rauen1, Josip Zovko1, Hermann Einsele1, Güzin Surat 1, Torsten Steinbrunn1

1University Hospital of Würzburg, Würzburg, Germany

Background: Patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT) frequently suffer from neutropenic fever (NF) which is a serious complication that requires timely and sufficiently dosed anti-infective treatment. Altered and variable pharmacokinetics due to critical illness can lead to inadequate free serum concentrations (FSC) which may facilitate antibiotic treatment failure, drug resistance, and increased toxicities. To account for these limitations of standardized drug application, we implemented therapeutic drug monitoring (TDM) of continuous piperacillin (PIP) infusion for patients with NF after allo-HSCT. This retrospective observational study assessed feasibility of TDM, pharmacological target attainment, and clinical outcome of infection in patients after allo-HSCT.

Methods: Data from n=44 adult patients who developed NF after undergoing allo-HSCT at the University of Würzburg Medical Center between October 2020 and March 2022 were included in our study. Empirical PIP/tazobactam (TAZ) treatment was administered as a continuous infusion and dose levels were regularly adjusted according to in-house antimicrobial stewardship (AMS)-guidelines specifically developed for TDM. Pharmacological target attainment was defined for the FSC of PIP of 80 mg/l (= target concentration (TC), corresponding to 5x MIC P. aeruginosa). Clinical outcome of infection was determined by successful withdrawal of PIP/TAZ monotherapy vs. antibiotic treatment escalation.

Results: At first assessment, the median FSC of PIP was 52 mg/l (IQR 41-68 mg/l), and at mid-treatment, the median FSC of PIP was 65.5 mg/l (IQR 47-90 mg/l). 9 patients (20%) reached TC at first assessment, 10 patients (23%) reached TC within the first three days, and 27 patients (61%) reached TC at their individual peak concentration. For 34 of the patients (77%), the infusion starting rate (562.5 mg/h) had to be increased subsequently. Conversely, 6 patients (14%) reached FSCs of 100 mg/l or above at their first assessment and required a dose reduction. No clinically apparent adverse effects attributable to PIP/TAZ treatment were documented. Microbiological assessment identified a pathogen in the bloodstream of 24 patients (55%), with gram-negative bacteremia occurring in 13 (30%) of these patients, most frequently due to K. pneumoniae (6 patients/40%) and E. coli (3 patients/20%). P. aeruginosa was not detected in any case. PIP/TAZ monotherapy effectively resolved infections in 21 of the patients (48%).

Conclusions: This study, to our knowledge, is the first to document TDM in patients with NF after allo-HSCT. PIP target attainment for the defined TC was feasible and TDM was safely applicable in our patient cohort. However, close monitoring of FSCs with timely dose adjustments was required to reach the set TC for PIP. Our study also suggests that higher PIP/TAZ infusion starting rates and/or shorter monitoring intervals may be warranted for earlier target attainment. Moreover, the pre-defined TC as well as additional sub-criteria may need to be re-examined for the best clinical benefit for patients receiving TDM.

Disclosure: Nothing to declare.

13: Infectious Complications

P425 ESTABLISHING EBV LOAD THRESHOLDS FOR PREDICTING PERSISTING EBV-DNAEMIA AND ASSOCIATED DISEASE IN HAEMATOPOIETIC CELL TRANSPLANT RECIPIENTS

Svenia Schmid 1, Andrea Erba1, Rainer Gosert1, Hans H. Hirsch2, Jakob Passweg1, Joerg Halter1, Nina Khanna1, Karoline Leuzinger1

1University Hospital Basel, Basel, Switzerland, 2University of Basel, Basel, Switzerland

Background: Quantification of EBV-DNA load in blood by nucleic acid testing (NAT) is routinely done to monitor EBV replication dynamics after hematopoietic cell transplantation (HCT). However, there is a lack of well-defined EBV-DNA load thresholds reliably predicting persistent EBV-DNAemia and subsequent development of EBV-associated disease, thus guiding the timely initiation of pre-emptive therapy.

Methods: We conducted a retrospective analysis of EBV-DNA load results in whole blood (WB) and time-matched plasma (PL), analysed from January 2010 to July 2023, and normalized EBV-DNA loads using two cellular markers (aspartoacylase (ACY) gene copy number and lymphocyte absolute counts (LYM)). Different absolute and normalized EBV-DNA load thresholds were evaluated to identify patients at risk for developing persistent EBV-DNAemia and EBV-associated disease.

Results: In this retrospective study, we identified 218 HCT-recipients with longitudinal EBV-DNA load results available (n≥50). Out of these, 19 (9%) did not exhibit any detectable EBV-DNAemia. The remaining 199 patients demonstrated EBV-DNAemia exceeding the lowest EBV-DNA load threshold of 1’000 copies/mL (corresponding to 10% percentile). Utilising this threshold allowed us to identify 60% of HCT-recipients experiencing persistent EBV-DNAemia, defined as exceeding a given EBV-DNA load threshold in two consecutive measurements. In contrast, an EBV-DNA load of >70’000 copies/mL (corresponding to 90% percentile) was only exceeded in 58 patients and determined persistent EBV-DNAemia in 52% of them. Normalization of EBV-DNA load thresholds by cellular markers did not improve predictive accuracy. Given that both absolute and normalized EBV-DNA load thresholds demonstrated limited predictive accuracy in assessing the risk of patients to exhibit persistent EBV-DNAemia, we analysed the dynamics of EBV-DNAemia in 133 time-matched whole blood and plasma samples from 58 HCT-recipients. The overall concordance of EBV-DNA detection by NAT was 16%, with a median quantification difference of 3.8 log10 copies/mL. One patient (1.7%) developed a relapse of an EBV-associated lymphoma, which was detected three weeks earlier in WB than in PL.

Finally, we evaluated the specificity of various EBV-DNA load thresholds in WB and PL for ruling out EBV-related disease in HCT-recipients. In comparison to the current EBV-DNA load threshold of 50’000 copies/mL used at our tertiary care centre, which has a specificity of 97%, the specificity of EBV-DNAemia in WB varied considerably, ranging from 11% to 99%, depending on the selected threshold. Conversely, EBV-DNAemia at any threshold in PL exhibited a high specificity of 87%, which further increased to 99% for an EBV-DNA load threshold of 7’000 copies/mL.

Conclusions: Our retrospective study highlights the challenges in establishing effective EBV-DNA load thresholds for predicting persistent EBV-DNAemia and EBV-related disease in HCT recipients.

Normalization of EBV-DNA loads with cellular markers did not markedly improve predictive accuracy. The specificity of EBV-DNAemia in WB for EBV-related disease varied considerably based on the EBV-DNA load threshold, while a higher specificity was noted for EBV-DNAemia in PL. This variability highlights the necessity for more precisely defined EBV-DNA load thresholds to accurately interpret EBV NAT results, guiding early detection and management of EBV-related complications in HCT recipients.

Disclosure: Nothing to declare.

13: Infectious Complications

P426 CLINICAL OUTCOMES OF STRONGYLOIDIASIS IN HEMATOPOIETIC CELL TRANSPLANT (HCT) RECIPIENTS

Avneet Kaur 1, Deepa Nanayakkara1, Ryotaro Nakamura1, Randy Taplitz1, Jana Dickter1, Stephen Forman1, Monzr Malki1, Sanjeet Dadwal1

1City of Hope National Medical Center, Duarte, United States

Background: Strongyloides is a helminth that can cause localized or disseminated infection with increased morbidity and mortality in HCT recipients. The diagnosis of strongyloidiasis can be challenging and delayed.

Methods: Institutional review board (IRB) approved retrospective study of HCT recipients tested for strongyloides between January 2010 till September 9th, 2023. Data was collected on demographics, underlying malignancy, conditioning regimen, graft versus host disease (GVHD), immunosuppression (IS), chemotherapy, type of HCT, time from transplant, diagnosis modality, eosinophilia, symptoms, imaging, association with HTLV, clinical outcomes including mechanical ventilation (MV) and mortality.

Results: One hundred thirty-one HCT patients were tested for strongyloides post-HCT based on clinical suspicion and 13 tested positive with one or more of the following methods: IgG by enzyme-linked immunosorbent assay (ELISA), stool for ova and parasite (O + P), bronchoalveolar lavage (BAL) cytology, and tissue biopsy. Majority had allogenic HCT (10/13[77%]) and three had autologous HCT. Ten (77%) males, six (46%) Hispanics, three (23%) Asians, two Caucasians, and two patients declined to answer. Median age at diagnosis was 58 years. Median time from HCT to diagnosis of infection was 73 days (range: 8-233).

Nine (69%) patients tested positive by ELISA and 4 (31%) tested positive with more than one method. Six (46%) patients had GVHD and 4 (31%) were receiving corticosteroids at diagnosis of strongyloidiasis. Three patients (23%) had disseminated infection 6 (46%) had co-infections with other pathogens; bacteremia in 3/13 (23%) patients. Peripheral eosinophilia was noted in 8 (62%), 3 of them were on corticosteroids simultaneously. Nine (71%) had abnormal radiographic findings with 3 having findings due to disseminated infection documented by larvae on BAL cytology and 3 (23%) required MV. None were associated with HTLV. The constellation of symptoms observed were cough, abdominal distention with cramps, ileus, pruritis, rash, and diarrhea. Patients with localized infection were treated with oral (PO) ivermectin. One patient who tested positive by ELISA had negative work-up (multiple stool O + P) was not treated and has done well. Patients with disseminated infection were initially treated with PO ivermectin and upon approval of compassionate use by the US Food and Drug Agency & IRB approval, received veterinary formulation of subcutaneous (SQ) ivermectin. One patient also received albendazole along with PO and SQ ivermectin for disseminated disease. SQ ivermectin was well tolerated and led to rapid improvement in 2 of 3 patients with disseminated infection. Treatment duration was 14 days for disseminated infection. Overall mortality was 15% (2/13) but was higher with disseminated infection (1/2) at 50%.

Conclusions: Strongyloidiasis occurs early post-HCT posing diagnostic challenges due to multiple factors such as false negative serology, absence of eosinophilia, and presence of GVHD of the gut which can confound the clinical scenario. SQ ivermectin should be considered early on in patients with disseminated infection.

Disclosure: None.

13: Infectious Complications

P427 MONITORING OF CMV-SPECIFIC CELL-MEDIATED IMMUNITY - PROGNOSTIC FACTORS AND CLINICAL VALUE

Jonas Wißkirchen 1, Diana Wolff1, Isabelle Ries1, Oliver Kriege1, Pascal Wölfinger1, Beate Hauptrock1, Matthias Theobald1, Eva Wagner-Drouet1

1University Medical Center Mainz, Mainz, Germany

Background: Despite Letermovir prophylaxis, reactivation of cytomegalovirus (CMV) remains a relevant complication after allogeneic hematopoietic stem cell transplantation (HSCT). Monitoring CMV-specific cellular immunity may identify patients at risk and could guide screening and therapy.

Within a clinical trial, CMV T-Track® (a standardized IFN-γ ELISpot assay) conducted after a first CMV reactivation identified patients at risk for recurrent reactivations with a positive predictive value of 80%.

Here we analyzed the feasibility of CMV-T-Track® within clinical routine as well as influencing factors and the predictive value.

Methods: We analyzed 173 patients during 365 days after HSCT within the clinical routine at our center. Screening for CMV was performed by CMV-qPCR, CMV specific immunity was assessed by CMV T-Track®. In all patients CMV T-Track® was conducted at d + 100. In case of a negative result, an additional test was performed at d + 200 and +300, respectively.

Patients: Conditioning regimen contained T cell depleting (TCD) agents: ATG in 125, Alemtuzumab i.v. in 28, ptCy in 12 patients. 8 patients received no TCD.

CMV-serostatus: Donor(D)+/recipient(R)+ in 125, D + /R- in 11, D-/R+ in 34 and D-/R- in 3 patients.

All CMVpos recipients received Letermovir prophylaxis until d + 100.

Results: At d + 100 168/173 tests produced a robust result, 5 tests were invalid.

109 patients had a positive T-Track® at d + 100, with a higher percentage in patients with a CMVpos donor (99/131 (76%) vs. 10/37 (27%)).

The rate of positive T-Track® was best in pTCy 11/12(92%), similar after ATG 83/124 (67%) as without TCD 5/8 (63%), and the lowest after Alemtuzumab 10/24 (42%).

In case of a CMVneg donor, 9/10 (90%) patients with a positive T-Track® d + 100 had CMV-viremia before d + 100, whereas only 3/27 (11%) without CMV immunity.

In patients with a CMVpos donor, 42/99 (42%) had a CMV-viremia leading to a positive T Track®, in contrast to 9/32 (28%) with a negative T-Track®.

18/20 (90%) patients with a negative test result d + 100 developed a positive T-Track® at d + 200 after CMV-viremia between d + 100 and d + 200. 24 patients were persistently negative at d + 300.

Despite a positive T-Track® at d + 100, 28/109 (26%) patients needed preemptive treatment for CMV reactivation. In most of them immunosuppression was initiated after d + 100: 20/28 (71%) high dose corticosteroids, 1/28 (4%) Ruxolitinib.

Excluding patients, that received additional immunosuppression after d + 100 only 7/89 (8%) developed a CMV reactivation despite a positive d + 100 T-Track® (negative predictive value: 0,921), whereas 25/59 (42%) patients with a negative T-Track® at d + 100 had a subsequent CMV reactivation.

Conclusions: 109/173 (64%) patients showed immunity against CMV at day +100. Excluding patients that received additional immunosuppression thereafter, only 7/89 (8%) developed a subsequent CMV reactivation.

With this T-Track® d + 100 has a negative predictive value of 0,921.

CMV negative donor was the strongest predictor for a negative result. Alemtuzumab was the only TCD with a negative impact.

CMV-viremia enhances CMV-immunity and is mandatory in patients with a CMVneg donor to establish CMV-immunity.

CMV T-Track® is feasible in clinical routine. This may help to identify patients with sufficient CMV-immunity at d + 100 to guide monitoring intervals or therapeutic decisions (e.g. prolonged prophylaxis/preemptive therapy).

Disclosure: Nothing to disclose.

13: Infectious Complications

P428 USE OF LETERMOVIR IN PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS UNDER AGE OF 12: A RETROSPECTIVE MULTI-CENTRE STUDY ON BEHALF OF THE IEWP AND PDWP

Katharina Kleinschmidt 1, Giovanna Lucchini2, Jacques-Emmanuel Galimard3, Adriana Balduzzi4, Krzysztof Czyzewski5, Jowita Frączkiewicz6, Francesco Paolo Tambaro7, Jaroslava Adamcakova8, Andrea Urtasun9, Matthias Woelfl10, Maria Ester Bernardo11, Alessandra Biffi12, Gergely Kriván13, Iván Lopez Torija14, Petr Sedlacek15, Wolfgang Holter16, Tamara Diesch17, Michael H. Albert18, Agnieszka Sobkowiak-Sobierajska19, Marta Gonzalez Vicent20, Elif Ince21, Sarah May Johnson2, Roland Meisel22, Arnaud Dalissier3, Selim Corbacioglu1, Bénédicte Neven23, Krzysztof Kalwak6

1University of Regensburg, Regensburg, Germany, 2Great Ormond Street Hospital, London, United Kingdom, 3EBMT Paris Study Unit, Paris, France, 4University of Milan, Milan, Italy, 5Collegium Medicum UMK, Bydgoszc, Poland, 6Wroclaw Medical University, Wroclaw, Poland, 7Azienda Ospedaliera di Rilievo Nazionale, Naples, Italy, 8Comenius University and National Institute of Children’s Diseases, Bratislava, Slovakia, 9Clínica Universidad de Navarra, Pamplona, Spain, 10University Children’s Hospital of Würzburg, Würzburg, Germany, 11Ospedale San Raffaele, Milan, Italy, 12Padua University Hospital, Padua, Italy, 13United St. Istvan and St. Laszlo Hospital, Budapest, Hungary, 14Hospital Santa Creu i Sant Pau, Barcelona, Spain, 15University Hospital Motol, Prague, Czech Republic, 16St. Anna Kidnerspital, Vienna, Austria, 17University Children´s Hospital of Basel, Basel, Switzerland, 18Dr. von Hauner Children’s Hospital, Munich, Germany, 19University of Medical Sciences, Poznan, Poland, 20Niño Jesus Children`s Hospital, Madrid, Spain, 21Ankara University Faculty of Medicine, Ankara, Turkey, 22Medical Faculty Heinrich Heine University, Duesseldorf, Germany, 23INSERM UMR 1163-Institut Imagine, Paris, France

Background: Letermovir is licensed for prophylaxis of CMV infection in adults in allogeneic hematopoietic stem cell transplantation (HSCT) and there is very limited data on its use, both for prophylaxis and treatment in HSCT recipients below the age of 12 years. This study aimed to summarize in large retrospective series the experience of letermovir in children <12 years in the setting of HSCT with focus on feasibility of administration, toxicity and efficacy.

Methods: The present study is an EBMT data-collection based retrospective multicenter analysis (2019-2023). 89 pediatric patients with a median age of 7.7 years (range: 0.2-11.9) at initiation have received letermovir as primary (N=72) or secondary (N=17) prophylaxis. Letermovir has been administered as oral (83.1%), IV (10.1%) or combined (6.7%) formulation. For patient characteristics refer to Table 1. Conditioning regimens were busulfan-based in 15.8% of patients, treosulfan-based in 43.8%, TBI-based in 30.3% and other in 10.1%. GvHD-prophylaxis consisted of Cyclosporine A alone or in combination in 69.7% of patients, combination of MMF and Tacrolimus in 18%, and others (12.4%). In-vivo T-cell depletion (TCD) has been performed in 35% of patients with either ATG (31.5%) or Campath (4.5%), while 10.1% of patients have received ex-vivo TCD.

Results: With a median follow-up of 1.1 year (95% CI= 0.9-1.3), the 1-year cumulative incidence of CMV reactivation was 19.9% (95% CI=12.2-29.1) for all patients, and 21.8% (95% CI=12.9-32.3) during primary and 12.2% (95% CI=1.9-32.9) during secondary prophylaxis, respectively.

From CMV-seropositive patients, among the 34 with a CMV-seronegative donor, 12 showed a CMV-reactivation and 4 died without CMV-reactivation. Among the 40 with a CMV-seropositive donor, only 4 had a CMV reactivation and 4 died (no CMV reactivation).

Regarding the 14 CMV-seronegative patients, only one received HSCT from a CMV-negative donor with letermovir being administered as secondary prophylaxis. The patient is alive without reactivation. The remaining 13 patients received HSCT from a CMV-positive donor, 1 had a reactivation and one died (no CMV-reactivation).

Overall survival at 1 year was 87.8% (95% CI=77.5-93.5) with 90% (95% CI=78.8-95.5) during primary and 79.1% (95% CI=47-92.9) during secondary prophylaxis, respectively. Among the patients with CMV-reactivation, one died after relapse. Other causes of death were viral/bacterial infections not CMV-related (3), relapse (2), GvHD (2), and others (2). No adverse events were reported except for one patient experiencing gastrointestinal toxicity. Treatment of CMV reactivation/disease was performed with ganciclovir (4), valganciclovir (8), foscarnet (3) or virus-specific T-cells (1), partly in combination. Two patients developed CMV-disease, both fully resolved. All but one patient with reactivation experienced complete viral clearance until end of follow-up.

Variables

Modalities

N=89

Primary prophylaxis (N=72)

Secondary prophylaxis (N=17)

Patient sex

Female

32 (36)

22 (30.6)

10 (58.8)

Male

57 (64)

50 (69.4)

7 (41.2)

Diagnosis for HSCT

ALL, AML, MDS and Lymphoma

47 (52.8)

36 (50)

11 (64.7)

PID, HLH, IEM

16 (18)

14 (19.4)

2 (11.8)

Bone marrow failure

14 (15.7)

11 (15.3)

3 (17.6)

Haemoglobinopathies

12 (13.5)

11 (15.3)

1 (5.9)

Number of HSCT

First

86 (96.6)

69 (95.8)

17 (100)

Second or third

3 (3.4)

3 (4.2)

0 (0)

Donor Type

Unrelated donor

50 (56.2)

39 (54.2)

11 (64.7)

Mismatched related donor

29 (32.6)

26 (36.1)

3 (17.6)

Matched related donor

10 (11.2)

7 (9.7)

3 (17.6)

Stem Cell source

Bone marrow

34 (38.2)

28 (38.9)

6 (35.3)

Peripheral blood

54 (60.7)

43 (59.7)

11 (64.7)

Cord blood

1 (1.1)

1 (1.4)

0 (0)

CMV donor to patient

D: positive; P: positive

40 (45.5.)

29 (40.8)

11 (64.7)

D: negative; P: positive

34 (38.6)

30 (42.3)

4 (23.5)

D: positive; P: negative

13 (14.8)

12 (16.9)

1 (5.9)

D: negative; P: negative

1 (1.1)

0 (0)

1 (5.9)

D: positive; P: missing

1 (1.1)

1

Table 1. Patient characteristics. PID primary immunodeficiencies; HLH hemophagocytic lymphohistiocytosis; IEM inborn errors of metabolism.

Conclusions: Letermovir was tolerated well and demonstrated a very high efficacy both in primary and secondary prophylaxis in children <12 years. Breakthrough reactivations/infections were infrequent and very well treatable. Consequently, the success of CMV prophylaxis with letermovir for children and infants below 12 years of age warrants licensing of this drug in due time.

Disclosure: No conflict of interest to declare.

13: Infectious Complications

P429 CONVENTIONAL VERSUS NGS-BASED PATHOGEN DIAGNOSTICS IN FEBRILE ALLOGENEIC STEM CELL TRANSPLANT PATIENTS: A PROSPECTIVE SINGLE-CENTRE STUDY

Sophie Weil 1, Madlen Amersbach1, Jochen J. Frietsch1, Sabrina Kraus1, Christoph Schoen2, Johannes Forster2, Philipp Reuter-Weissenberger2, Philip Stevens3, Oliver Kurzai2, Hermann Einsele1, Daniel Teschner1,4

1University Hospital Wuerzburg, Würzburg, Germany, 2Institute for Hygiene and Microbiology, Julius Maximilians University of Würzburg, Würzburg, Germany, 3Noscendo GmbH, Duisburg, Germany, 4University Medical Centre of the Johannes Gutenberg University, Mainz, Germany

Background: The aim of our study was to analyse whether next-generation-sequencing (NGS)-based pathogen diagnostic added to the conventional diagnostic standard of care (SOC) can contribute to identify infectious causes for febrile episodes in patients undergoing allogeneic haematopoietic stem cell transplantation (HSCT).

Methods: After informed consent, adult patients undergoing or after allogeneic HSCT were included in the study. During their first febrile episode, blood samples were drawn for blood cultures (BC) and NGS-based DISQVER®-diagnostic. Depending on individual symptoms, additional diagnostic procedures were performed according to the SOC. NGS-based results were provided in real-time and reviewed for clinical relevance. Additionally, diagnostic findings were correlated with the patients’ characteristics and clinical course.

Results: NGS-based diagnostic was performed in 71 febrile HSCT patients between February and September 2023. Of these, 50 patients were included in the final analysis. These patients showed a median age of 54 years, and the majority were male (28/50; 56%). The underlying disease was primarily acute myeloid leukaemia (AML; 29/50; 58%). Patients predominantly received a reduced intensity conditioning (RIC; 39/50; 78%) combined with anti-thymocyte globulin (ATG; 19/50; 38%). 29/50 (58%) patients experienced a febrile episode at a median of 32 days after HSCT of whom 15/29 (52%) presented with neutropenia. 21/50 (42%) patients developed fever during conditioning therapy prior to the HSCT of whom 19/21 (90%) received ATG at this time point. In total, clinically relevant pathogens were detected in 28/50 (56%) cases. Of these, 20/28 (71%) were identified NGS-based, 6/28 (24%) by BC, and 23/28 (82%) by all available diagnostic SOC procedures (including BC). 5/28 (18%) pathogens were found by NGS only, whereas 8/28 (29%) were detected solely by SOC. However, 6 out of these 8 pathogens were found in non-blood diagnostic samples. The identified pathogen spectrum did not substantially differ between NGS-based and SOC detection methods. The diagnostic yield was higher by trend in febrile patients treated with RIC versus myeloablative conditioning (MAC; 59% vs. 45%). In febrile patients under ATG treatment (19/50; 38%) NGS-based diagnostic was negative in 18/19 (95%) cases (95% [ATG] vs. 5% [non-ATG]; p<0.0001). To evaluate a potential impact of NGS-based results on antibiotic use in febrile ATG patients, a matched-pair analysis with 17 febrile non-NGS ATG patients in 2022 was performed. Here, febrile ATG patients in our study showed shorter duration of antibiotic therapy (median 7,5 days), compared to febrile non-NGS ATG patients in 2022 (median 11 days). 30-day survival was 90% (45/50) without any trend between both diagnostic groups.

Conclusions: Combination of NGS-based and conventional diagnostics approaches might increase the diagnostic yield in febrile allogeneic HSCT patients. Furthermore, NGS-based approaches have the potential to identify pathogens in blood, which otherwise can be detected by more invasive SOC methods only. Whether the addition of NGS-based approaches might enable to distinguish between infectious and non-infectious febrile episodes potentially resulting in reduced empiric anti-infective treatment needs to be further studied.

Disclosure: Daniel Teschner: received honoraria and institutional funding from Noscendo GmbH.

Philip Stevens: is an employee of Noscendo GmbH.

13: Infectious Complications

P430 MULTINATIONAL STUDY ASSESSING TREATMENT PATTERNS, OUTCOMES, AND HEALTHCARE RESOURCE UTILIZATION IN HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS WITH CYTOMEGALOVIRUS INFECTION AND INTOLERANCE TO ANTI-CYTOMEGALOVIRUS THERAPIES

Genovefa Papanicolaou1,2, Robin Avery3, María Laura Fox 4,5, Karl S. Peggs6, Luís Veloso7, Tien Bo8, Ishan Hirji8, Kimberly Davis8

1Memorial Sloan Kettering Cancer Center, New York, United States, 2Weill Cornell Medicine, New York, United States, 3Johns Hopkins University, Baltimore, United States, 4Vall d’Hebron Universitari Hospital Campus, Barcelona, Spain, 5Universitat Autònoma de Barcelona, Barcelona, Spain, 6University College London Hospitals NHS Foundation Trust, London, United Kingdom, 7CTI Clinical Trial & Consulting Services, Lisbon, Portugal, 8Takeda Development Center Americas, Inc., Lexington, United States

Background: Cytomegalovirus (CMV) infection is a major cause of mortality and morbidity after hematopoietic stem cell transplant (HSCT). CMV infection management can be limited by intolerance to anti-CMV agents including ganciclovir, valganciclovir or foscarnet due to related toxicities.

Methods: In this multicenter, retrospective study, de-identified patient data from HSCT recipients with CMV infection refractory or resistant to treatment, or with intolerance to anti-CMV agents were collected from 14 transplant centers in France, Germany, Italy, Spain, UK, and the US between January 2014 and December 2021. This analysis described treatment patterns, clinical outcomes, and healthcare resource utilization (HCRU) in the subgroup of HSCT recipients with CMV infection who had intolerance to anti-CMV agents. The first CMV episode in which the patient was considered intolerant to anti-CMV treatment was the index episode (first intolerant CMV episode). Data were collected and analyzed descriptively.

Results: Overall, 86/250 patients (median age, 57 years; male, 56%) were intolerant to anti-CMV agents. These patients experienced a total of 160 CMV episodes during the study and were most commonly treated with valganciclovir. Valganciclovir was most frequently used for all CMV episodes and at the time of identification of intolerance, followed by foscarnet, and ganciclovir (Table). For all CMV episodes, 83% (n=71) of patients were treated with ≥2 therapies. The majority of patients (98%) had ≥1 dose change of anti-CMV therapy, reasons included resolved viremia, lower maintenance dose, inadequate response and/or adverse events. Dose changes were most commonly with valganciclovir (75%), foscarnet (58%), and ganciclovir (49%). Neutropenia and renal toxicity were the most common clinical factors leading to intolerance (Table). Intolerance was due to myelosuppression in 51% of patients and nephrotoxicity in 33% of patients. Of 67 myelosuppression events reported following intolerance diagnosis, neutropenia 73% (n=49), thrombocytopenia 36% (n=24), and leukopenia 18% (n=12) were the most common. All-cause mortality, and mortality 1-year post-intolerance identification were 51% and 47%, respectively, with a median follow-up from first intolerant CMV episode of 438 days. During their first intolerant CMV episode, 76% (n=65) of patients achieved viremia clearance (median [Q1–Q3] time to viremia clearance 41 days [24–65]; n=63). CMV recurrence was experienced by 40% (n=34) of patients, with a median (Q1–Q3) time to CMV recurrence of 33 days (16–68). Overall, 2% (n=2) of patients had graft failure and 64% (n=55) of patients experienced ≥1 graft versus host disease event (classed as acute, chronic, or severe in 49, 15, and 7 patients respectively). Thirty-eight percent (n=33) of patients had ≥1 CMV-related hospitalization (median [Q1–Q3] length of stay, 17 days [9–29]), and there were 50 CMV-related hospitalizations in total.

Table. Characterization of intolerance to anti-CMV agents during the first intolerant CMV episodes in HSCT recipients

n=86

CMV treatment used at intolerancea identification, n (%)

Valganciclovir

38 (44.2)

Foscarnet

27 (31.4)

Ganciclovir

11 (12.8)

Days on anti-CMV treatment prior to first intolerancea identification, median (Q1–Q3)

12 (6–21)

Clinical factors leading to intolerancea during the first intolerant CMV episode,b,c n (%)

Neutropenia

39 (45.3)

Renal toxicity

25 (29.1)

Thrombocytopenia

16 (18.6)

Leukopenia

11 (12.8)

Nausea/vomiting

11 (12.8)

Days from HSCT until first intolerant episode, median (Q1–Q3)

57 (35–85)

  1. CMV, cytomegalovirus; HSCT, hematopoietic stem cell transplant.
  2. aIntolerance was defined as the inability to tolerate antiviral therapy due to adverse events or being considered as at risk of experiencing a serious adverse event to currently available antiviral agents.
  3. bClinical factors and events with frequency >10%.
  4. cMore than one clinical factor could be reported in the same patient.

Conclusions: Findings from this real-world study highlight the particularly high burden of CMV infection for HSCT recipients who develop intolerance (primarily neutropenia) to anti-CMV agents. As well as being at risk of adverse outcomes, including mortality and morbidity, a notable proportion of transplant patients failed to achieve viral clearance and/or experienced CMV recurrence. There is a need for CMV therapies that achieve and maintain viral clearance with improved safety profiles in this population.

Disclosure: Genovefa Papanicolaou: consultant/other fees from Allovir, Amplyx, Armata, Cidara, CSL Behring, MSD, Octapharma, Symbio, Takeda, and Vera; research funding from MSD.

Robin Avery: consultant (no personal financial remuneration) for Takeda; grant/research support from AiCuris, Astellas, AstraZeneca, Chimerix, Merck, Oxford Immunotec, QIAGEN, Regeneron, and Takeda.

María Laura Fox: consultant/other fees from AbbVie, Bristol Myers Squibb, Novartis, Sanofi Biotest, and Sierra Oncology - GlaxoSmithKline.

Karl S. Peggs: consultant/other fees from Bristol Myers Squibb and Takeda.

Luís Veloso: employee of CTI Clinical Trial & Consulting Services which received fees from Takeda.

Tien Bo: employee of Takeda Development Center Americas, Inc. and Takeda stockholder.

Ishan Hirji: employee of Takeda Development Center Americas, Inc. and Takeda stockholder.

Kimberly Davis: employee of Takeda Development Center Americas, Inc. and Takeda stockholder.

13: Infectious Complications

P431 EFFICACY OF LETERMOVIR IN PREVENTING CYTOMEGALOVIRUS REACTIVATION AFTER CORD BLOOD TRANSPLANTATION

Naoki Okada 1, Hiroyuki Muranushi1, Kazuya Okada1, Takayuki Sato1, Takeshi Maeda1, Tatsuhito Onishi1, Yasunori Ueda1

1Kurashiki Central Hospital, Kurashiki, Japan

Background: Cord blood transplantation (CBT) is associated with a high risk of cytomegalovirus (CMV) infection. Letermovir (LTV) is a prophylactic agent for CMV reactivation (CMV-R). However, there are limited data on the efficacy of LTV in preventing CMV-R after CBT and the incidence of delayed-onset CMV-R after LTV discontinuation. The purpose of this study was to evaluate the efficacy of LTV in preventing CMV-R after CBT.

Methods: A single-center retrospective observational study of 84 adult patients who received CBT as their first transplantation for acute myeloid leukemia, acute lymphoblastic leukemia, or myelodysplastic syndrome between February 2016 and September 2022 was conducted. Four patients who were seronegative for CMV and one patient who received CMV preemptive therapy on day 0 were excluded. Outcomes were compared between patients who received LTV (LTV group), which became the standard of care in January 2019, and those who did not receive LTV (non-LTV group) because they received CBT by the end of December 2018. The primary endpoint was the 100-day cumulative incidence of CMV-R. The 1-year cumulative incidence of CMV-R, cumulative incidence of CMV disease, and other transplant-related outcomes were also evaluated. Event rates were estimated using the Kaplan-Meier method for overall survival and Gray’s method for other endpoints. CMV was monitored weekly using a pp65 CMV antigenemia assay (C10/11 method), and CMV-R was defined as the start of CMV preemptive therapy. LTV was administered for three months after CBT and the dose was 240 mg per day because of the use of cyclosporine as a calcineurin inhibitor.

Results: There were 45 patients in the LTV group and 34 patients in the non-LTV group (Table 1). The cumulative incidence of CMV-R was significantly lower in the LTV group at day 100 (11.1% vs. 82.4%, p < 0.001) and at 1 year (45.3% vs. 82.4%, p < 0.001), but the incidence of delayed-onset CMV-R after LTV discontinuation was as high as 34.2%. The cumulative incidence of CMV disease was comparable between the LTV and non-LTV groups (0% vs. 5.9% at day 100, 6.8% vs. 5.9% at 1 year; p = 0.927). Multivariate analysis showed that the use of LTV reduced the cumulative incidence of CMV-R (hazard ratio: 0.17, 95% confidence interval: 0.08-0.37, p < 0.001). Overall survival, relapse, non-relapse mortality, and neutrophil engraftment were not significantly different between the two groups. The LTV group tended to have a higher 100-day cumulative incidence of platelet engraftment (91.1% vs. 79.4%, p = 0.0504) and a lower 100-day cumulative incidence of grade 3-4 acute graft-versus-host disease (GVHD) (11.1% vs. 26.5%, p = 0.0866). Among patients without death or relapse by day 100, the LTV group tended to have lower 2-year cumulative incidences of chronic GVHD (27.2% vs. 44.4%, p = 0.0504) and moderate and severe chronic GVHD (7.8% vs. 18.5%, p = 0.105).

Table 1. Patient characteristics

Total

(N = 79)

LTV

(n = 45)

Non-LTV

(n = 34)

p value

Age, years

55.00 [43.00, 64.00]

57.00 [47.00, 64.00]

51.50 [40.25, 61.75]

0.191

Men

46 (58.2)

22 (48.9)

24 (70.6)

0.067

Disease

AML

35 (44.3)

25 (55.6)

10 (29.4)

0.028

ALL

17 (21.5)

10 (22.2)

7 (20.6)

MDS

27 (34.2)

10 (22.2)

17 (50.0)

rDRI

Low to intermediate

49 (62.0)

29 (64.4)

20 (58.8)

0.646

High to very high

30 (38.0)

16 (35.6)

14 (41.2)

HCT-CI

Score 0

51 (64.6)

26 (57.8)

25 (73.5)

0.391

Score 1 to 2

18 (22.8)

12 (26.7)

6 (17.6)

Score ≥3

10 (12.7)

7 (15.6)

3 (8.8)

Conditioning regimen

MAC

50 (63.3)

28 (62.2)

22 (64.7)

1

RIC

29 (36.7)

17 (37.8)

12 (35.3)

  1. Data are median [interquartile range] or number (%).
  2. ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; HCT-CI, hematopoietic cell transplant-comorbidity index; LTV, letermovir; MAC, myeloablative conditioning; MDS, myelodysplastic syndrome; rDRI, refined Disease Risk Index; RIC, reduced intensity conditioning.
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (47)

Conclusions: LTV is effective in preventing CMV-R after CBT. Because of the high incidence of delayed-onset CMV-R, close monitoring is necessary after LTV discontinuation and long-term prophylaxis beyond day 100 should be considered.

Disclosure: Yasunori Ueda declares honoraria from Sanofi. The other authors have no conflict of interest to declare.

13: Infectious Complications

P432 SARS-COV-2 CYCLE THRESHOLD LEVELS AND COVID-19 OUTCOME IN ALLOGENEIC STEM CELL TRANSPLANTATION. AN INFECTIOUS DISEASE WORKING PARTY STUDY

Jose Luis Piñana Sanchez 1, Per Ljungman2, Alexander Kulagin3, Maria Teresa Lupo Stanghellini4, Carlos Solano5, Gloria Tridello6, Jiri Mayer7, Maria Suarez-Lledó8, Hélène Labussière-Wallet9, Robert Zeiser10, Jennifer Clay11, Anna Bergendahl Sandstedt12, Inmaculada Heras13, Krzysztof Kalwak14, Jose Luis Lopez15, Judith Schaffrath16, Adriana Balduzzi17, Fabio Benedetti18, Amjad Hayat19, David Gallardo20, Harry Koene21, Jose Rifón22, Paul Gerard Schlegel23, Nina Knelange24, Dina Averbuch25, Rafael De la Camara26

1Hospital clinico universitario, Valencia, Spain, 2Karolinska Comprehensive Cancer Center, Karolinska University Hospital Huddinge and Karolinska Institutet, Stockholm, Sweden, 3RM Gorbacheva Research Institute, Pavlov University, Petersburg, Russian Federation, 4Ospedale San Raffaele s.r.l., Milano, Italy, 5HOSPITAL CLINICO UNIVERESITARIO DE VALENCIA, Valencia, Spain, 6Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy, 7University Hospital Brno, Brno, Czech Republic, 8Hospital Clinic, Barcelona, Spain, 9Centre Hospitalier Lyon Sud, Lyon, France, 10University of Freiburg, Freiburg, Germany, 11St James University Hospital Leeds, Leeds, United Kingdom, 12University Hospital Linkoeping, Linkoeping, Sweden, 13Hospital Morales Meseguer, Murcia, Spain, 14Wroclaw Medical University, Wroclaw, Poland, 15Fundación Jiménez Díaz, Madrid, Spain, 16Martin-Luther-Universitaet Halle-Wittenberg, Halle, Germany, 17Centro Trapianto di Midollo Osseo - Clinica Pediatrica, Monza, Italy, 18Policlinico G.B. Rossi, Verona, Italy, 19Galway University Hospital, Galway, Ireland, 20Institut Català d`Oncologia, Girona, Spain, 21St. Antonius Hospital, Nieuwegein, Netherlands, 22Clínica Universitaria de Navarra, Pamplona, Spain, 23University Children`s Hospital, Wuerzburg, Germany, 24EBMT office, Leiden, Netherlands, 25Faculty of Medicine, Hebrew University of Jerusalem; Hadassah Medical Center, Jerusalem, Israel, 26Hospital de la Princesa, Madrid, Spain

Background: Prior studies in general hospitalized and not hospitalized population showed that cycle threshold (Ct) values correlate with COVID-19 progression to severe illness, with abnormal biochemical and/or haematological parameters, with higher probability of a positive viral culture and with mortality. Ct values provide a relative measure of viral quantity in the specimen, but do not provide the actual quantity. In hematological patients lower Ct correlates with COVID-19 severity and COVID-19 duration. However, data on the effect of Ct values in stem cell transplant recipients (allo-SCT/auto-SCT) is scarce.

Methods: This retrospective study aimed to investigate the effect of SARS-CoV-2 Ct values at the diagnosis on the COVID-19 outcome in a cohort of 113 allo-SCT recipients and 26 auto-SCT with available Ct values at diagnosis registered in the EBMT-COVID-19 database between 2020 and 2022. Clinical differences between recipients with Ct values below and above the median were assessed. The study divided patients into two groups based on their Ct values: those below the median (n=69) and those above the median (n=70). The median Ct value for the entire cohort was 22.30. The study also evaluated COVID-19-related variables, including pneumonia, hospitalization, oxygen support, ventilation, high-flow oxygen therapy, intensive care unit (ICU) admission, and COVID-19-related death.

Results: Recipients with Ct values below the median were generally older (median age 50 years) compared to those with Ct values above the median (median age 33 years) (p= 0.004). Recipients receiving peripheral blood showed higher rate of low Ct values compar4ed to bone marrow (56% vs 38%, p= 0.06) There was no significant differences in Ct values between allo-SCT and auto-SCT. Regarding allo-SCT recipients, the proportion of recipients with low Ct values was higher in male recipients allografted from female donors (28% vs 12%) (p= 0.04), in those who received an unrelated donor graft (58% vs 36%) and in frail recipients (performance status >1) (34% vs 10%, p= 0,006). Vaccination status at the time of COVID-19 did not significantly differ between the two groups. We did not observe statistical differences in the hospital admission, pneumonia rate and in intensive care unit admission between those below and above the median Ct value (29% vs 27%, p= 0.2; 49% vs 37%, p= 0,1; 17% vs 12%, p= 0.4, respectively). However, higher rate of oxygen supports was observed in those with low Ct values (31% vs 11%, p= 0,008) but this fact did not translate into significant differences in 6- and 12-weeks overall survival (94% vs 86% and 91% vs 81%, respectively p= 0.1).

Conclusions: While low Ct values were associated with certain demographic and clinical factors, including older age and specific stem cell sources, the impact on severe clinical outcomes such as hospitalization and mortality was not consistently observed. The findings emphasize the complexity of COVID-19 outcomes in this specific patient population, suggesting that additional factors beyond Ct values may contribute to the disease course. Further research is warranted to validate these findings and explore additional factors influencing COVID-19 outcomes in stem cell transplant recipients.

Disclosure: no coi.

13: Infectious Complications

P433 SINGLE-DAY TRIMETHOPRIM-SULFAMETHOXAZOLE PROPHYLAXIS FOR PNEUMOCYSTIS PNEUMONIAE AND TOXOPLASMOSIS INFECTION IN HEMATOPOIETIC STEM CELL TRANSPLANTATION PEDIATRIC PATIENTS

Maria Speranza Massei 1, Ilaria Capolsini1, Elena Mastrodicasa1, Grazia Gurdo1, Carla Cerri1, Francesco Arcioni1, Maurizio Caniglia1, Katia Perruccio1

1Pediatric Oncology-Hematology, Santa Maria della Misericordia Hospital, Perugia, Italy

Background: Trimethoprim/sulfamethoxazole (TMP-SMX) given 2-3 times weekly is the drug of choice for the primary prophylaxis of Pneumocystis jirovecii pneumoniae (PCP) and Toxoplasmosis infection in adults and children allogeneic hematopoietic stem cells transplantation recipients. Some data indicate that immunosuppressed children with an underlying haematological condition or cancer, may benefit equally from a once-weekly regimen. In this report we present a pediatric cohort of sixty patients who received a hematopoietic stem cells transplantation (HSCT) and received the one-day/week of TMP-SMX after transplant in the Perugia Pediatric Oncology-Hematology Unit.

Methods: Sixty pediatric patients, median age of nine years (range, 2-26), underwent HSCT between February 2014 and November 2023. Everyone received the 1-day/week prophylaxis regimen with TMP-SMX from day +50 after transplant for 6 months or until the patient is no longer receiving immunosuppression, whichever is longer, until immune reconstitution has occurred. Twentynine had acute lymphoblastic leukemia, eleven acute myeloid leukemia, one acute undifferentiated leukemia, five Fanconi anemia, seven severe aplastic anemia, three Non-Hodgkin Lymphoma, three Hemoglobinopathies, one Haemophagocytic familial syndrome; 41 were males, 19 females. 30/60 (50%) patients underwent HLA-haploidentical transplantation (everyone with regulatory and conventional T-cell adoptive immunotherapy), 17/60 (28%) mismatched unrelated donor, 13/60 (22%) HLA-identical sibling donor. 18/60 patients (30%) developed acute graft versus host disease (aGvHD). 42/60 (70%) are alive and 41/60 disease free (68%), 18/60 died, 9/18 for non-relapse mortality (NRM) and 9/18 for relapse. The median follow up is 5.1 years (range 9.8 years-1 month).

Results: None patient developed PCP neither Toxoplasmosis infection, not even patients (18/60) with immunosuppressive therapy for GvHD. 9/60 patients dead for NRM, but none for PCP or Toxoplasmosis infection.

Conclusions: These data in 60 pediatric patients, of which 50% HLA-haploidentical HSCT and 30% immunosuppressed for aGvHD, showed that a single-day course of prophylaxis with TMP-SMX may be sufficient to prevent PCP and Toxoplasmosis infection in patients underwent allogeneic hematopoietic stem cell transplantation. This strategy might have applied also for PCP prophylaxis in other patients with other conditions that induce a strong immunosuppression.

Disclosure: Nothing to declare.

13: Infectious Complications

P434 AN INTERIM ANALYSIS OF FOVOCIP: A MULTICENTER RANDOMIZED TRIAL OF FOSFOMYCIN VERSUS CIPROFLOXACIN FOR FEBRILE NEUTROPENIA PREVENTION IN HEMATOLOGIC PATIENTS

Ahinoa Fernández Moreno1, Ana Julia GonzalezHuerta2, Paula López de Ugarriza1, Cristina Muñoz3, Rebeca Rodríguez Veiga4, Laura Solán Blanco5, Maria Luisa Calabuig6, Karem Humala7, Juan Manuel Bergua Burgués3, Marta Polo Zarzuela8, Maria Luz Amigo9, María Izquierdo de Miguel3, Eva González Barberá4, Marina Medel Plaza5, Guillermo Ruiz Carrascoso7, Laura López Gonzalez8, Irene Weber9, Pau Montesinos Fernández4, Ana Maria Fernández Verdugo2, Javier Fernández Dominguez2, Teresa Bernal 1

1Instituto de Investigación Sanitaria del Principado de Asturias, Oviedo, Spain, 2Hospital Universitario Central de Asturias, Oviedo, Spain, 3Hospital San Pedro Alcántara, Caceres, Spain, 4Hospital La Fe, Valencia, Spain, 5Hospital Fundación Jiménez Díaz, Madrid, Spain, 6Hospital Clinico de Valencia, Valencia, Spain, 7Hospital La Paz, Madrid, Spain, 8Hospital Clinico San Carlos, Madrid, Spain, 9Hospital Universitario Morales Messeguer, Murcia, Spain

Background: Multidrug resistant Gram negative bacterial (MRGNB) infections represent a major public health threat. In hematological patients, gut colonization by MRGNB is common, occurs during hospitalization and chemotherapy exposure and increases the risk of difficult to treat blood stream infections. Fluoroquinolone (FQ) prophylaxis efficacy in these patients has been questioned in the last years due to the potential risk of increasing gut colonization and its lack of efficacy in settings with high prevalence of MRGNB. However, no randomized study has formally demonstrated that FQ prophylaxis is no longer valid and clinical trials exploring other alternatives are scarce.

Methods: We designed a Phase III randomized, controlled, clinical trial, open label parallel group with a 1:1 ratio, aimed to demonstrate the non-inferiority of oral fosfomycin (F) versus oral ciprofloxacin (C) for febrile neutropenia (FN) prevention in patients with acute leukemia (AL) or hematopoietic cell transplant (HSC) receptors. Primary end-point was febrile neutropenia. Secondary end-point was rate of gut colonization by MRGNB. In both arms, prophylaxis was stablished to stop in case of FN, absolute neutrophil count recovery (ANC) (>0.5 x109/L) or limiting adverse event (AE). This trial is expected to recruit 156 patients. We present the interim results of the first 50 patients included in the trial.

Results: Among the 50 patients, 25 were randomized to F. Underlying diagnosis was AL in 23/50 (46%) and SCT and 27/50 (54%) patients. Median age was 59 years (IQR 51-66) years. Regarding sex, 28/50 (56%) were males. Underlying disease in patients randomized to F was AL in 12/25 (48%) and SCT in 13/25 (52%).

FN requiring intravenous antibiotics developed in 47/50 (94%) patients, of whom 20/25 (80%) received C and 19/25 (76%) F, P=0.7. Time to FN was 9 and 8 days in C and F arms, respectively, P=0.45. Cumulative Incidence (CI) of FN is shown in Figure 1. Only 2 patients, both in ciprofloxacin arm, developed gut colonization by MRGNB (ESBL- and/or carbapenemase-producing Enterobacterales). Rate of AE requiring prophylaxis discontinuation was 2% in both arms.

This study has been funded by Instituto de Salud Carlos III through the project FIS21/1590 and cofunded by the European Union.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (48)

Conclusions: Interim analysis showed the non-inferiority of F vs C in the prevention of FN, as well as the safety and low selection of MRGNB of this alternative, however the completion of recruitment has to be performed to draw final conclusions.

Clinical Trial Registry: Clinicaltrials.gov: NCT05311254.

EudraCT Number: 2021-000354-25.

Disclosure: The authors disclose no conflict of interest.

13: Infectious Complications

P435 IMPACT OF LETERMOVIR PRIMARY AND SECONDARY PROPHYLAXIS IN A PEDIATRIC COHORT

Francesca Vendemini1, Francesca Romani1,2, Sonia Bonanomi1, Giorgio Ottaviano1, Paola de Lorenzo3, Maria Grazia Valsecchi2, Sergio Maria Ivano Malandrin4, Marta Verna 1, Giulia Prunotto1, Pietro Casartelli1, Adriana Cristina Balduzzi1,2

1Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy, 2Università degli Studi Milano-Bicocca, Monza, Italy, 3Tettamanti Center, Università degli Studi Milano-Bicocca, Monza, Italy, 4Microbiology Laboratory, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy

Background: CMV infection is associated with increased morbidity and mortality after HSCT. Letermovir primary prophylaxis against CMV infections has become a standard of care in adult HSCT recipients due to its efficacy and high tolerability. However, letermovir is not licensed in pediatric patients and data on its use for prophylaxis in HSCT recipients are limited.

Methods: Retrospective analysis of allogeneic HSCT performed between January 2014 and November 2022 at the pediatric Hematopoietic Stem Cell Transplantation Unit of the Fondazione IRCCS San Gerardo dei Tintori in Monza (Italy) was performed.

Since April 2021 (post-letermovir cohort), off-label primary prophylaxis with letermovir has been used from day +1 after HSCT in CMV+ recipient (R + ) who have received a transplant from CMV negative donor (D-). During the same period, R+ transplant from D+ who developed CMV reactivation treated with pre-emptive therapy (PET) and who were at high risk of subsequent reactivation (haploidentical or mismatched HSCT, steroid dose ≥ 1 mg/kg/day), received secondary prophylaxis with letermovir.

Letermovir doses were: 4 mg/kg/day in children weighing <30 kg, 120 mg/day for weight 30-50 kg, 240 mg/day for weight > 50 kg (all patients received concomitant cyclosporin).

Fisher’s exact test was used to compare the characteristics of the two cohorts. Cox regression analysis was performed to investigate the influence of primary prophylaxis with letermovir.

Results: Two hundred and eighty-two allogeneic HSCT were included: 239 and 43 patients belonged to the pre-letermovir and post-letermovir cohorts, respectively. The CMV D/R serological pair and the characteristics at baseline were balanced between the two cohorts, with the exception of a substantial increase in haploidentical HSCT and peripheral blood as stem cell source in the post-letermovir cohort (Table 1).

In the pre-letermovir cohort, CMV reactivation developed in 23/57 (40%) transplants D-/R + . In the post-letermovir cohort, 9 D-/R+ patients received primary prophylaxis with letermovir (median duration 114 days, 35-235) and none developed CMV reactivation.

The analysis of the association between the type of GvHD prophylaxis and CMV reactivation in both cohorts, excluding D-/R+ transplants in which letermovir was used, showed, as expected, that haploidentical HSCT was associated with the highest risk of CMV reactivation, followed by allogeneic HSCT with ATG as GVHD prophylaxis and sibling HSCT with cyclosporin and methotrexate. In multivariate analysis, primary prophylaxis with letermovir significantly reduced the risk of CMV reactivation (p=0.004), even after adjustment for the type of HSCT (higher number of haploidentical transplants in the post-letermovir cohort), maintaining significance (p=0.003).

In the pre-letermovir cohort, a second reactivation after the end of PET was observed in 11/39 (28%) D + /R+ transplants. In the post-letermovir cohort, 11 children D + /R+ were treated with letermovir secondary prophylaxis (median duration 112 days, 61-237) after the end of PET and none experienced CMV reactivation.

None of the patients treated with letermovir discontinued therapy due to the occurrence of side effects.

Table 1: Clinical characteristics of the patients. Higher proportion of haploidentical HSCT and peripheral blood as stem cell source was observed in the post-letermovir cohort.

Pre-letermovir

Post-letermovir

P Value

Hazard ratio

Total

No.

239

43

282

Male sex

No. (%)

153 (64%)

26 (60%)

ns

179 (63%)

Age at HSCT (years)

Median

8,87

8,52

ns

8,76

Range

0,9-18,2

0,8-18,7

0,8- 18,7

Diagnosis

Acute lymphoid leukemia

103 (43%)

16 (37%)

119(42%)

Acute myeloid Leukemia

48 (20%)

9 (21%)

57 (20%)

Chronic myeloid leukemia

5 (2%)

1 (2%)

6 (2%)

Non-Hodgkin lymphoma

8 (3%)

1 (2%)

9 (3%)

Hodgkin lymphoma

8 (3%)

-

8 (2%)

Severe aplastic anemia

15 (6%)

2 (5%)

17 (6%)

Hemoglobinopathies

19 (8%)

7 (16%)

26 (9%)

Others

33 (14%)

7 (16%)

40 (14%)

Transplant Type

Matched related donor

78 (33%)

11 (26%)

ns

89 (32%)

Matched unrelated donor

134 (56%)

22 (51%)

ns

156 (55%)

Haploidentical

27 (11%)

10 (23%)

0.047

0.003*

37 (13%)

Stem cell source

Bone marrow

223 (93%)

30 (70%)

ns

253(90%)

Peripheral blood

11 (5%)

12 (28%)

0.0001

23 (8%)

Cord blood

5 (2%)

1 (2%)

ns

6 (2%)

Donor/Recipient CMV Serology

D + /R+

117 (49%)

22(51%)

ns

139(49%)

D + /R-

25(10%)

7(16%)

ns

32 (11%)

D-/R+

57 (24%)

9 (21%)

ns

66 (23%)

D-/R-

40 (17%)

5 (12%)

ns

45 (16%)

GvHD requiring systemic therapy

No

106 (44%)

15 (35%)

ns

121 (43%)

Yes

133 (56%)

28 (65%)

ns

161 (57%)

Death

No

187 (78%)

36 (84%)

ns

223 (79%)

Yes

52(22%)

7(16%)

ns

59 (21%

  1. *Cox regression analysis investigated the impact of transplant type and letermovir on the occurrence of CMV reactivation: primary prophylaxis with letermovir significantly reduced the risk of CMV reactivation even when the model was adjusted for the type of HSCT.

Conclusions: Letermovir proved to be effective as both primary and secondary prophylaxis in preventing CMV infection and was well tolerated in pediatric patients. Our results suggest that letermovir effectively reduces CMV reactivation events in high-risk patients.

Clinical Trial Registry: -.

Disclosure: Nothing to declare.

13: Infectious Complications

P436 EPSTEIN-BARR VIRUS REACTIVATION AND POST-TRANSPLANT LYMPHOPROLIFERATIVE DISORDER (PTLD) AFTER IMPLEMENTATION OF ANTITHYMOCYTE GLOBULIN AS GVHD PROPHYLAXIS. A SINGLE CENTER EXPERIENCE

Tor Henrik Anderson Tvedt1, Mats Remberger1,2,3, Ingerid Weum Abrahamsen1, Camilla Dao Vo 1, Anders Eivind Leren Myhre1, Tobias Gedde-Dahl1

1Oslo University Hospital, Oslo, Norway, 2Uppsala universitet/ Institutionen för medicinska vetenskaper, Uppsala, Sweden, 3Akademiska sjukhuset/ Kliniska forsknings- och utvecklingsenheten, Uppsala, Sweden

Background: Antithymocyte globulin (ATG) increases the risk of Epstein-Barr virus (EBV) related diseases including transient lymphoproliferation and potentially fatal post-transplant lymphoproliferative disorder (PTLD). At Oslo University hospital (OUS), ATG was added to methotrexate/cyclosporine GVHD prophylaxis in 2016 for both unrelated (4-6 mg/kg) and matched related donors (2-4 mg/kg). The current study analyses incidence of EBV reactivation and PTLD along with associated mortality.

Methods: All patients treated with allogeneic stem cell transplantation at OUS from 2016 to 2023 were included in the analysis. The primary objectives were the cumulative incidence (CUI) of EBV reactivation and PTLD, and survival, non-relapse mortality (NRM), and relapse in patients with EBV-reactivation and PTLD. Only patients alive at day 30 post-transplant were included in the analysis.

Results: The study included 769 patients, with 51.2% having acute leukemia, 29.4% with MDS or MPN, 12.2% with CLL or lymphoma, 4.7% with non-malignant disorders, and 2.5% other disorders. There were 13.0% matched related donors, 5.7% haploidentical donors, 12.5% mismatched unrelated donors (MMUD), and 68.8% matched unrelated donors (MUD). Graft source was peripheral blood stem cells in the majority of cases (87.6%). ATG was used in 75.2% of transplantations, while post-transplant cyclophosphamide was used in 11.4%.

The CUI for EBV reactivation was 53.3% (95% CI: 49.7-56.8) at 100 days post-transplant, 63.9% (95% CI: 60.3-67.3) at 1 year, and 66.0% (95% CI: 62.4-69.3) at 2 years, while the CUI for PTLD was 6.8% (95% CI: 5.2-8.8) at 100 days post-transplant, 7.7% (95% CI: 5.9-9.7) at 1 year, and 8.0% (95% CI: 6.2-10.1) at 2 years. The 1-year CUI for EBV reactivation and PTLD after ATG exposure was 74.0% (95% CI:70.0-77.5) and 10.0% (95% CI:7.7-12.7) respectively compared with 34.1% (27.3-41.1) and 0.5% (95% CI: 0.1-2.9) for patients not exposed to ATG.

Similar 2-year survival was observed in patients with PTLD (66.8%, 95% CI: 52.9-77.5) and no EBV reactivation (62.4%, 95% CI: 55.7-68.4), while survival was significantly higher in patients with EBV reactivation, 79.3% (95% CI: 74.9-83.0, p-value <0.001). Correspondingly, significantly higher NRM was observed in the PTLD (25.6%, 95% CI: 15.1-37.3) and non-EBV population (23.5%, 95% CI: 18.5-28.8) compared to patients with EBV-reactivation (6.2%, 95% CI: 4.1-8.7). No difference in relapse rate was observed among the three groups.

Factors significantly associated with PTLD in univariate analysis were MMUD (p-value 0.01 HR 2.13 95% CI: 1.18-3.84), donor age (p-value 0.03, HR 0.98 95% CI: 0.96-0.99), ATG (p-value 0.001, HR 9.91 95% CI:2.42-40.6) bone marrow graft (p-value 0,035, HR 0.12 95% CI:0.02-0.86) and CD34 dose (p-value 0.01 HR 1.12 95% CI:1.03-1.22). Multivariate analysis including exposure to ATG and donor type found that ATG (p-value <0.006 HR 7.56 95%CI: 1.80-31.72) and MMUD (p-value <0.001 HR 2.75 vs. MUD reference, 95%CI: 1.52-4.96) were significantly associated with PTLD.

Conclusions: While EBV occurs in a majority of patients receiving ATG, the overall incidence of PTLD was low. The observation that patients with EBV reactivation had a better outcome than patients without EBV reactivation might be due to confounding factors but warrants further investigation.

Clinical Trial Registry: N/A.

Disclosure: Tor Henrik Anderson Tvedt: Consultant/advisor Jansen, Sanofi, Novartis, Takeda, BMS.

Mats Remberger: None.

Anders Eivind Myhre: Consultant/advisor: Takeda, Bristol Myers Squibb, Astra Zeneca, Immedica. Speaking fees: Novartis, Takeda, Unimedic.

Ingerid Weum Abrahamsen: None.

Camilla Dao Vo: None.

Tobias Gedde-Dahl: Takeda, Novartis, Incyte.

13: Infectious Complications

P437 IMPACT OF INVASIVE FUNGAL DISEASE UPDATED DEFINITIONS ON ALLOGENEIC HEMATOPOIETIC TRANSPLANTATION OUTCOME IN ADULT PATIENTS - POST-HOC ANALYSIS OF NATION-WIDE CROSS-SECTIONAL 2012-2014

Patrycja Mensah-Glanowska 1, Agnieszka Piekarska2, Monika Adamska3, Joanna Drozd-Sokolowska4, Anna Waszczuk-Gajda4, Agnieszka Tomaszewska5,4, Agnieszka Wierzbowska6, Marek Hus7, Joanna Manko5, Sylwia Bartzel-Palinska8, Lidia Gil3

1Jagiellonian University Medical College, Krakow / University Hospital in Krakow, Krakow, Poland, 2University Clinical Center/ Medical University of Gdansk, Gdanska, Poland, 3Poznan University of Medical Sciences, Poznan, Poland, 4Medical University in Warsaw, Warsaw, Poland, 5Institute of Hematology and Transfusion Medicine, Warsaw, Poland, 6Medical University of Lodz, Lodz, Poland, 7Medical University, Lublin, Poland, 8University Hospital in Krakow, Krakow, Poland

Background: This study is a retrospective analysis of a Polish cross-sectional study, conducted over three years (2012-2014), focusing on patients at transplant centers who underwent Hematopoietic Stem Cell Transplantation (HSCT) for hematological malignancies. The aim was to comprehensively assess Invasive Fungal Disease (IFD) occurrences in adult allogeneic HSCT patients, examine death-related risk factors linked to IFD, and evaluate the impact of the updated 2022 IFD diagnostic criteria on patient reclassification and treatment outcomes.

Methods: The primary patient cohort, derived from a nationwide Polish retrospective study, included 3205 adults from 11 of 13 transplant centers, comprising 2135 autologous HSCT and 1070 allogeneic HSCT patients. In the allogeneic cohort, 135 IFD episodes were diagnosed. The analysis was restricted to patients available for follow-up, resulting in a final dataset of 102 patients across 7 centers, with data collected until February 2016. Initially, IFD classification followed the 2008 EORTC/MSG consensus. However, considering the 2019 revisions, diagnoses were reevaluated using existing data. Patients receiving mold-active prophylaxis with oral posaconazole, showing multiple IFD risk factors and chest HRCT imaging results, but negative serum GM antigen, were categorized as probable IFD cases. Primary clinical outcomes measured were overall mortality, 3-month mortality and IFD-related mortality.

Results: In the analysed population of 102 patients with IFD, the primary analysis identified 43 possible cases, 38 probable IFD cases, and 18 confirmed cases. In light of the changes introduced in 2019, the data available allowed for a re-evaluation of the level of IFD diagnosis. The level of IFD diagnosis, following the criteria established in 2008, indicated similar 3-months all-cause mortality rates in patients with probable and proven IFD – 47.4% and 50 %, respectively, while the rates were lower in the cohort with possible IFD (27.9%). However, using the revised 2019 IFD diagnosis criteria, which segregates patients into possible, probable, and confirmed categories, the 3-monts mortality rates were 16%, 36%, and 52.4%, respectively. In the multivariate analysis, we examined risk factors associated with mortality among alloHCT patients who developed IFD during the post-transplant period. Neutropenia, already recognized as a main risk factor for IFD development, was also identified as a risk factor for both short-term and long-term all-cause mortality in this patient group. The risk of death related to IFD was influenced by the disease as defined by the revised EORTC/MSG criteria. Moreover, gastrointestinal IFD localization and myeloablative conditioning were linked to a decreased risk of all-cause mortality, whereas the use of voriconazole in IFD treatment was associated with a higher risk of all-cause mortality within the first three months from IFD diagnosis.

Conclusions: The reclassification of IFD diagnosis according to the 2019 EORTC/MSG criteria provides a more precise estimation of mortality risk in alloHCT patients diagnosed with IFD.

Disclosure: Nothing to declare.

13: Infectious Complications

P438 SURVEILLANCE BLOOD CULTURES FROM TUNNELLED CENTRAL VENOUS CATHETERS ON ADMISSION ARE OFTEN POSITIVE, GUIDE MANAGEMENT AND HELP INFORM REPORTING OF HOSPITAL ACQUIRED INFECTIONS

Debbie Barrow1, Krzysztof Ciesielski1, Seamus McDermott1, Grant McQuaker1, David Irvine1, Annie Latif1, Ailsa Holroyd1, Christine Peters1, Dimitris Galopoulos1, Andrew Clark 1

1Queen Elizabeth University Hospital, Glasgow, United Kingdom

Background: Tunnelled central venous catheters (tCVC) provide secure central access for patients undergoing prolonged periods of neutropenia during transplant procedures, which are often complicated by sepsis and mucositis. Patients like tCVC as they avoid the need for frequent venepuncture and they are often essential peri to allow delivery of high dose chemotherapy, blood product support or TPN. However, they are potential sites of serious infection, often acting as sanctuary sites, protected from systemic antibiotics. Hospital acquired infections (HAI) in tCVC are common and difficult to manage. The Scottish Allograft centre has performed blood cultures on admission for several years driven and co-ordinated by the Advanced Nurse Practitioner (ANP) team. Interpretation of results and management is a MDT process led by dedicated microbiologists and transplant physicians. This study represents a review of this practice.

Methods: On admission, aerobic and anaerobic blood cultures were taken from each lumen of patients’ indwelling tCVC. Cumulative infection rates and type of infections in tCVC were documented in all allograft and selected autograft recipients over calendar year 2023. A high level overview was then carried out of the impact of positive blood cultures on patient management, specifically antibiotic use and line removal, as well as their impact on infection control practices.

Results: We present data from 100 patients (87 allograft and 13 autograft recipients) screened in 2023. These patients were drawn from all Scottish Boards. No Health Board or line insertion centre had a preponderance of infections or high numbers of serious infections. Infection rates were 29%. Multiple different bacteria were isolated from tCVC in 9 patients. Specific Bacteria isolated are listed in Table 1. Most isolates are gram positive skin commensal organisms, with Staphlococcus Epidermidis by far the commonest. Eleven Gram negative isolates were identified in 7 patients. No positive fungal cultures were seen.

Bacterial Isolates from Admission Blood Cultures

Gram Positive Bacteria

Number of Patients

Gram Negative Bacteria

Number of Patients

Staph. Epidermidis

13

Klebsiella Pneumoniae*

3

Staph. Hominis

3

Klebsiella Oxytoca

1

Staph. Haemolyticus

1

Serratia Liquefacens*

1

Staph. Warnerii

1

Moraxella Osloensis

1

Staph. Lugdenensis

1

Stentrophomonas Maltophilia*

1

Enterococcus Faecalis

1

Acidovorax sp

1

Cutibacterium Acnes

1

Campylobacter Jejuni

1

Strep. Mitis

1

E. Coli

1

Corynebacterium Jeikeium

1

Acinetobacter Ursingii*

1

Corynebacterium Amycolatum

1

Corynebacterium Tuberculostearicum

1

Dermacoccus Nishinomiyaensis

1

Micrococcus Luteus

1

Clostridium Perfringens

1

Microbacterium Oxydans

1

Bacillus Lichenoformis

1

The type of infection often impacted on management. Targeted Antibiotics, taking account of ESBL (n=2) and CRE (n=1) isolates were started immediately and six tCVC were removed- 5 for gram negative infections, including all with Klebsiella, Serratia, Acinebacter and stenotrophomanas species- several of which had multiple different isolates in the same cultures and one for C. Perfringens. Only two of these patients were symptomatic. In other patients with documented infection the majority were gram positive. All had vancomycin included in their antibiotic plan (in the absence of VRE, when Daptomycin was included) to be started early if febrile. However, antibiotics were with-held and cultures repeated in the majority who remained afebrile. Most of the organisms identified can be HAI and it was extremely useful to exclude these infections on admission.

Conclusions: Asymptomatic tCVC infection is commonly found in patients on admission for Allogeneic or autologous transplant. A substantial number have their immediate peri-transplant management altered by these cultures. It is vital to exclude pre-existing infections at time of admission when determining the true extent of HAI in a unit. Infection control investigations of outbreaks of infection might otherwise over-report or draw erroneous conclusions. Conversely, any pattern seen in infection from referring teams may prove useful.

Clinical Trial Registry: None.

Disclosure: none.

13: Infectious Complications

P439 ROLE OF HUMAN HERPESVIRUS 6 REACTIVATION IN T-DEPLETE TREG/TCON ALLOGENEIC STEM CELL TRANSPLANTATION

Valerio Viglione 1, Loredana Ruggeri2, Antonella Mancusi1, Sara Tricarico2, Alessandra Cipiciani1, Rebecca Sembenico1, Matteo Caridi1, Zorutti Francesco1, Anna Castaldo1, Tiziana Zei2, Roberta Iacucci Ostini2, Simonetta Saldi1, Roberto Castronari2, Silvia Bozza1, Cynthia Aristei1, Antonella Mencacci1, Maria Paola Martelli1, Alessandra Carotti2, Antonio Pierini1

1University of Perugia, Perugia, Italy, 2Santa Maria della Misericordia Hospital, Perugia, Italy

Background: The role of Human Herpesvirus 6 (HHV-6) in conventional allogeneic human stem cell transplantation (allo-HSCT) is not fully elucidated, but HHV-6 reactivations have been associated to increased risk of graft failure, hepatitis, encephalitis, delayed post-transplant immune reconstitution and higher incidence of GVHD. HHV-6 incidence and role in immunosuppression-free T-deplete HSCT with adoptive immunotherapy with regulatory and conventional T cells (Treg/Tcon allo-HSCT) is not known.

Methods: We retrospectively analyzed 163 consecutive allo-HSCT performed in Perugia, Italy, from January 2019 to October 2023. HHV-6 DNAemia was assessed by quantitative PCR on whole blood twice a week from day +4 to day +28.

Results: Sixty-six (40%) patients underwent unmanipulated HSCT with post-transplant cyclophosphamide (PTCy allo-HSCT) and 97 (60%) underwent Treg/Tcon allo-HSCT. The 2 groups were similar regarding patients’ age, sex, and type of donor.

HHV-6 reactivation occurred in 96% of Treg/Tcon allo-HSCT patients and in 66% of PTCy allo-HSCT patients (p<0.0001). Median peak viraemia was higher (p<0.0001), earlier (p<0.0001) and more persistent (p<0.0001) in Treg/Tcon allo-HSCT (7909 copies/uL, range 20-1146359; median onset on day +11, range +3- + 21; median duration 12, range 3-21) than in PTCy allo-HSCT (183 copies/uL, range 4-58208; median onset +18, range 7-28; median duration 6, range 3-18). Despite such results, there was no higher incidence of graft failure in Treg/Tcon allo-HSCT (4% vs 6%, p=NS). No patient experienced encephalitis.

HLA-haploidentical transplant showed higher (p<0.0001) rates of HHV-6 reactivation compared to HLA-matched, with similar onset and duration in Treg/Tcon Allo-HSCT. HLA-haploidentical and HLA-matched reactivation were similar for peak viraemia, onset and duration in PTCy Allo-HSCT.

There were no differences in HHV-6 reactivation incidence, peak viraemia and transplant outcomes according to age, sex, type of conditioning regimen in Treg/Tcon allo-HSCT.

We found peak viraemia did not relate with transplant outcomes such us engraftment, non-relapse mortality (NRM), GvHD, and relapse. We analyzed viraemia/white blood cell (V/W) ratio as it might be considered a better parameter to evaluate HHV-6 active infection. Patients with high HHV-6 V/W ratio (above median value that was 15) experienced a delay of neutrophil engraftment in both haploidentical (median 12.8 days vs 13.7; p=0.04) and HLA-matched (median 13.9 vs 16.2; p=0.03) Treg/Tcon allo-HSCT unmodified by antiviral therapy with Foscarnet (p=NS).

No differences were found in platelet engraftment, incidence (p=NS) and grade (p=NS) of acute GVHD.

Patients with high V/W had a higher incidence of early, generally mild, hepatic toxicity (GOT: p=0.008; GPT: p=0.02). All patients had full resolution and no higher incidence of hepatic GVHD and VOD was observed.

Treg/Tcon allo-HSCT patients had a fast immune reconstitution, unaffected by both high viraemia or ratio, with a high rate of T-cell clones specific against HHV-6 that were detectable starting at day +30.

Conclusions: Almost all Treg/Tcon allo-HSCT patients reactivated the virus but the only significant clinical manifestation was a short delay in neutrophil engraftment. Fast immune recovery and the absence of pharmacological immunosuppression might have helped to clear the infection in all patients. Further studies will need to evaluate the real need of treatment for patients that reactivate HHV-6 after Treg/Tcon allo-HSCT.

Disclosure: No conflict of interest to declare.

13: Infectious Complications

P440 INTERLEUKIN-6 IS SIGNIFICANTLY INCREASED IN SEVERE PNEUMONIA AFTER ALLO-HSCT AND MIGHT INDUCE LUNG INJURY VIA IL-6/SIL-6R/JAK1/STAT3 PATHWAY

Jingrui Zhou1, Leqing Cao1, Rui Ma1, Yun He1, Danping Zhu1, Na Li1, Xiaosu Zhao1, Xiaojun Huang1, Yuqian Sun 1

1Peking University People’s Hospital, Peking University Institute of Hematology, Beijing, China

Background: Severe pneumonia after allogeneic hematopoietic stem cell transplantation (allo-HSCT) was associated with rapid progression and high mortality. It was known that cytokines including Interleukin-6 (IL-6) play a critical role in immune-mediated organ injury in patients with severe COVID-19, we speculated that cytokines might also play an important role in the pathogenesis of severe pneumonia after allo-HSCT. In addition, the detailed mechanism by which IL-6 contributes to the development of acute lung injury remains to be fully elucidated. Therefore, our study aims to examine the role of IL-6 and its potential mechanism in the development of severe pneumonia after allo-HSCT.

Methods: Blood sample from patients with severe pneumonia after allo-HSCT was prospectively collected to detect serum cytokines. A mouse model of acute lung injury induced by lipopolysaccharide (LPS) was constructed to investigate the impact of IL-6 blockade, and the possible mechanism of IL-6 in lung injury. Also, an in vitro experiment using primary murine PMVECs was performed to investigate the mechanism of immune-induced lung injury.

Results: Forty consecutive patients with pneumonia after allo-HSCT were prospectively enrolled, with 20 severe pneumonia and 20 non-severe pneumonia. The serum IL-6 levels in the severe pneumonia group were significantly increased compared to the non-severe group (median 83.92 vs. 37.27 pg/ml, P<0.001), and were positively correlated with disease progression. The mouse model demonstrated that early blockade of IL-6 in vivo could significantly reduce acute lung injury and improve survival. In vitro experiments demonstrated that the IL-6 trans-signaling complex (IL-6/soluble IL-6R) caused more severe damage to mouse pulmonary microvascular endothelial cells (PMVECs) than its classical signaling pathway, and soluble glycoprotein 130 (IL-6 trans-signaling inhibitor) and JAK inhibitor ruxolitinib could effectively block the JAK1/STAT3 pathway activated by IL-6 trans-signaling in mouse PMVECs and downstream inflammatory responses, respectively. Tocilizumab (IL-6 receptor antagonist) was used in one patient with severe pneumonia who failed to conventional methods, and the clinical condition was successfully improved.

Conclusions: IL-6 was significantly increased in severe pneumonia after allo-HSCT and is associated with disease progression. The injury might be induced by IL-6/sIL-6R/JAK1/STAT3. This is a preliminary study, however provides a new perspective that targeting the IL-6 trans-signaling pathway might be potentially useful in treating severe pneumonia/acute lung injury after allo-HSCT.

Disclosure: Nothing to declare.

13: Infectious Complications

P441 ECHINOCANDIN PROPHYLAXIS IS ASSOCIATED WITH BREAKTHROUGH TRICHOSPORON AND CANDIDA INFECTIONS AND A LIMITED ROLE OF GALACTOMANNAN MONITORING AMONG ALLOGENEIC STEM CELL TRANSPLANT RECIPIENTS

André Dias Américo 1, João Antônio Gonçalves Garreta Prats1, Hegta Tainá Rodrigues Figueiroa1, Juliana Matos Pessoa1, Eurides Leite da Rosa1, Fauze Lutfe Ayoub1, Moyses Antônio Porto Soares1, Isabella Silva Pimentel Pittol1, Fabio Rodrigues Kerbauy1,2, Phillip Scheinberg1, José Ulysses Amigo Filho1

1BP, Beneficência Portuguesa de São Paulo, São Paulo SP, Brazil, 2Universidade Federal de São Paulo, São Paulo, Brazil

Background: The choice of prophylactic antifungal agents in allogeneic hematopoietic stem cell transplantation (HSCT) is debated due to variable invasive fungal disease risks. While azoles are recommended, echinocandins present advantages in tolerability and fewer drug-drug interactions. Micafungin has been more widely studied in the HSCT setting. Here we report a single center experience in allogeneic HSCT patients on echinocandin prophylaxis, mostly anidulafungin, aiming to contribute real-world data on efficacy and safety.

Methods: A retrospective study of allogeneic HSCTs that occurred from 2018 to 2021. Prophylaxis failure was defined as breakthrough infection, death, or antifungal switch. Invasive fungal infections were assessed per EORTC standards. Data included patient characteristics, transplant details, and their outcomes. All patients were admitted to a unit equipped with HEPA filters and were monitored with serial galactomannan and episodes of febrile neutropenia and under suspicion of invasive fungal diseases were investigated according to current standard guidelines and practice. All patients received Cumulative incidence of invasive fungal infection was analyzed until day 35 post-HSCT.

Results: There were 145 transplants carried out at our center, of which 120 patients received echinocandins (anidulafungin, n = 106, micafungin, n = 14) as prophylaxis, most patients received a myeloablative conditioning regimen (54.2%), haploidentical donor was most frequently utilized (44.2%). Neutropenia lasted for 15 days (median, IQ 25-75, 9-20 days) and 7 patients failed to engraft. Prophylaxis failed for 25 (17.5%) patients (proven IFI n = 9, possible IFI n = 7, deaths unrelated to IFI n = 4, antifungal switch due to primary physician’s preference n = 4, esophageal candidiasis n = 1). The cumulative incidence of IFI was at 7.6% (see table 1). Breakthrough fungal infections were all proven, predominantly caused by yeasts (Candida, n = 5, and Trichosporon, n = 3) rather than molds (Aspergillus, n = 1, Fusarium, n = 1, Mucorales, n = 2). Survival after invasive fungal disease diagnosis was 55.6% (95% CI, 30.9-99.7) at 42 days of follow up, and liposomal amphotericin B was commonly used as salvage therapy. In this cohort, 113 (94.1%) patients collected galactomannan (GLM) serially, only one patient presented with a positive serum galactomannan throughout the whole observation period, a case of fungal sinusitis caused by Fusarium spp. which occurred several days after initiation of salvage antifungal therapy.

Table 1. Clinical Outcomes

Outcome

n (%)

Proven Ivasive Fungal Disease

9 (7.6)

Yeast

5

Mold

2

Yeast and Mold

2

Probable Invasive Fungal Disease

Overall Prophyalxis Failure

25 (17.5)

Proven IFD

9

Possible IFD

7

Esophageal candidiasis

1

Death unrelated to IFD

4

Physician’s choice

4

Positive serum Galactomannan

1

Preemtive Galactomannan guided therapy

1

Conclusions: Echinocandin prophylaxis during the neutropenic phase of allo-HSCT was associated with a particular pattern of breakthrough yeast infections including Trichosporon and echinocandin-sensitive Candida species. This distinct epidemiology should be contemplated in protocols of empirical and/or salvage antifungal therapy. Most of the patients in our study received anidulafungin as primary prophylaxis, a less well studied echinocandin in HSCT. A pre-emptive strategy with GLM monitoring during echinocandin prophylaxis might be of limited use for the diagnosis of IFD. No patient had a positive GLM test prior to the clinical suspicion or diagnosis of IFD and throughout the study observation period. This may be partly because most breakthrough infections in our study were caused by yeasts.

Disclosure: André Américo received speaker fees from Janssen, Knight Pharmaceutical and MSD.

João Prats received speaker fees from Knight Pharmaceutical and MSD.

Eurides Rosa received speaker fees from Aztra Zeneca and MSD.

Fauze Ayoub received speaker fees from Janssen.

Phillip Scheinberg has done scientific presentations for Novartis, Roche, Alexion, Janssen, AstraZeneca; Grants/Research Support: Alnylam, Pfizer; has received grants or research support from Alnylam, Pfizer; has received consultancy fees from Roche, Alexion, Pfizer, BioCryst, Novartis, Astellas; and has been a speaker for Novartis, Pfizer, Alexion.

Hegta Figueiroa, Fábio Kerbauy, Isabella Pittol, Moyses Soares, José Ulysses Filho, Juliana Pessoa have no conflicts of interest to declare.

13: Infectious Complications

P442 REAL WORLD EXPERIENCE OF EPSTEIN-BARR VIRUS(EBV) SCREENING FOR PERSISTENT VIRAEMIA POST HAEMATOPOIETIC STEM CELL TRANSPLANT (HSCT)

Abigail Downing 1, David Davies1, Keith Wilson1,2, Emma Kempshall1, Sophie Bertorelli1, Serena Linley-Adams1, Damith Dhanasekara1, Rey Consolacion1, Susannah Froude3, Samantha Ray3, Wendy Ingram1

1University Hospital of Wales, Cardiff, United Kingdom, 2Cardiff University, Cardiff, United Kingdom, 3Public Health Wales, Cardiff, United Kingdom

Background: EBV viraemia is common following HSCT and screening is recommended post-transplant for high-risk patients. Risk factors for EBV related PTLD published in the 2015 ECIL-6 guidelines are allogeneic HSCT with any of the following risk factors: T cell depletion (in vivo or ex vivo), EBV serology mismatch, cord blood transplant, HLA mismatch, acute/chronic GVHD requiring intensive immunosuppressive therapy, high EBV viral load and splenectomy. Prospective monitoring should be commenced within 4 weeks and continued until 4months, with longer screening in those with poor T cell reconstitution. It is often difficult to know when to stop, especially in the context of persistent prolonged low level viraemia. Our aim was to evaluate screening practices to enable rationalisation of screening and reduce the financial burden of unnecessary screening.

Methods: We retrospectively identified 86 patients who received 89 allogeneic HSCT between 2020-2021. All 86 patients had prior serological evidence of EBV infection. Serial EBV PCR results were collated and results grouped as per the risk factors identified above.

Results: Fifty-nine patients (67%) developed EBV viraemia during screening. 64.4% (n=38) reactivated within 100 days, 18.6% (n=11) between 101-180 days, and 16.9% (n=10) after day 180. Between methods of T cell depletion, reactivation occurred most commonly in the ATG group with 77.8% (n=28), compared to 58.1% (n=25) and 50% (n=3) in the CAMPATH and PTCy groups respectively. 67.7% of the serologically matched group developed viraemia, compared to 57.1%(n=4) in the serologically mismatched group. 17 of the transplants were HLA mismatched, with 53% reactivating. Of those with viraemia, 61% had GVHD; 53% (n=31) acute and 25% (n=15) chronic. In those without viraemia GVHD was present in 50%. Median CD4 at day 90 in those with reactivation was 90 x 106/L (range 19-527) compared with 45 x 106/L (range 23-1494) for those without.

In patients with viraemia pre day 100 (n=38), 22 (58%) continued to have viraemia post day 100 and 18 (47%) had ongoing viraemia at day 180. Nine with viraemia pre day 100 required treatment, starting at a median of 176 (range 55-393) days: 4 started treatment pre day 100, 1 between 100-179 days and 4 after day 180. Only 1/21 (5%) patient with new viraemia post day 100 required treatment with a rapidly rising level, treatment initiated on day 119.

Screening past day 180 identified 42 patients with viraemia. Four patients treated after day 180, with a maximum viral load range from 9699-66966 x 106/L. Twenty-eight had persisting viraemia from pre day 180 with a median maximum viral load of 4914 x 106/L (range 453-116313). None of the 10 developing new viraemia after day 180 required treatment, median maximum viral load was 1014 x 106/L (range 291-18453).

EBV viraemia

Total

Yes

No

Overall numbers

89

59 (66.3%)

30 (33.7%)

Stem cell source

MUD

75

47 (62.7%)

28 (37.3)

Sibling

13

12 (92.3%)

1 (7.7%)

Haplo

1

1

T cell depletion

Campath

43

25 (58.1%)

18 (41.9%)

ATG

36

28 (77.8%)

8 (22.2%)

PTCy

6

3 (50%)

3 (50%)

T Replete

4

3 (75%)

1 (25%)

HLA disparity

10/10

72

50 (69.4%)

22 (30.6%)

Mismatch

17

9 (52.9%)

8 (47.1%)

Serological status (R/D)[KW2]

+/+

65

44 (67.7%)

21(32.3%)

+/-

7

4 (57.1%)

3 (42.3%)

-/+

-

-

-/-

-

-

+/unknown

17

11 (64.7%)

6 (35.3%)

GVHD

Overall

51

36

15

Acute

44

31

13

Chronic

21

15

6

Conclusions: Screening past day 180 identified 42 patients with ongoing viraemia, many of whom had stable low level viraemia. The additional burden of screening in patients >180 days post HSCT resulted in 1170 PCRs with an estimated cost £67000. We propose that screening can stop in these patients, even in the presence of stable viraemia, providing absence of clinical PTLD concerns.

Disclosure: Emma Kempshall- Medac- Funding(Honoraria).

Wendy Ingram- Medac- Funding(Honoraria), Novartis- Funding(Honoraria), Sobi- Funding(Honoraria), Gilead- Funding(Honoraria), Synairgen- Funding(Honoraria), Incyte- Funding(Honoraria).

Keith Wilson- Novartis- Funding(Honoraria), Jazz Pharmaceuticals- Funding(Honoraria), Gilead- Funding(Honoraria), Celgene- Funding(Honoraria).

13: Infectious Complications

P443 EPIDEMIOLOGY OF VIRAEMIA IN CHILDREN UNDERGOING CORD BLOOD STEM CELL TRANSPLANT (CB-SCT) WITH NON-MALIGNANT DISEASES: A RETROSPECTIVE COHORT STUDY

Patel Zeeshan Jameel 1, Denise Bonney1, Omima Mustafa1, Malcolm Guiver1, Madeleine Powys1, Ramya Nataraj1, Srividhya Senthil1, Robert Wynn1

1Royal Manchester Children’s Hospital, Manchester, United Kingdom

Background: Children undergoing CB-SCT are at a significant risk of developing viraemia during and post their transplant. We describe the experience of paediatric CB-SCT from a single tertiary centre.

Methods: All children undergoing CB-SCT for non-malignant diseases between 2014-2023 at Royal Manchester Children’s Hospital were included. Data from subjects with positive blood polymerase chain reaction (PCR) results for cytomegalovirus (CMV), adenovirus (ADV) and Epstein-Barr virus (EBV) during transplant and 12 months post CB-SCT was extracted using paper/electronic case sheets.

Results: In the 9-year period, a total of 53 children underwent 55 allogenic CB-SCT for curative treatment of their underlying non-malignant disease. Various indications for CB-SCT included mucopolysaccharidosis type 1 – Hurler (MPS IH), Fanconi anaemia, Chediak-Higashi syndrome, Wolman disease, Krabbe disease and LRBA deficiency. Of the 55 CB-SCT episodes, 15 (27.27%) were complicated by post-transplant viraemia. CMV viraemia was detected in 9, ADV in 5 and EBV in 1. One CB-SCT episode was complicated by dual viremia with CMV and ADV.

Cumulative incidence is as shown in Figure 1. Data collected has been summarised in Table 1. Virus reactivation is early after CB transplant.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (49)

CMV

ADV

EBV

Incidence

16.36% (9/55)

11% (5/55)

1.8 % (1/55)

Mean age (months ± SD)

58.67 ± 15.55

11.2 ± 2.322

12

Sex distribution (M: F)

8:1

2:3

0:1

Diagnosis of included patients

MPS IH (4/9), Fanconi anaemia (1/9), Chediak-Higashi syndrome (1/9), Wolman disease (1/9), LRBA deficiency (1/9)

MPS IH (4/5), Krabbe disease (1/5)

MPS IH (1/1)

Treatment received

Foscarnet, Ganciclovir, Valganciclovir

Cidofovir, Brincidofovir

Rituximab

Complications secondary to viraemia

Disseminated CMV (1/9), CMV pneumonitis (1/9), CMV encephalitis (1/9)

Disseminated ADV disease (1/5)

None

Virus related mortality

Disseminated CMV (1/9)

None

None

Overall mortality

33.33 % (3/9)

None

None

GVHD

Skin GVHD (3/9)

Skin GVHD (2/5)

Skin GVHD (1/1)

Mean (±SD) CD3 count at 4 weeks (cells/μL)

213.44 ± 218.41

299.6 ± 410.53

157

Mean (±SD) CD4 count at 4 weeks (cells/μL)

73 ± 57.43

153 ± 270.30

71

Mean (±SD) CD8 count at 4 weeks (cells/μL)

132.11 ± 168.58

151±158.89

84

CMV viraemia was complicated by pneumonitis (1/9), encephalitis (1/9) and disseminated disease (1/9). Single anti-viral (ganciclovir/valganciclovir) was effective in 4/9 cases while Foscarnet was required as a second ant-viral agent in 5/9 cases. Letermovir was used for post-treatment CMV recurrence in 2/9 children. Response was defined as two consecutive blood PCR results less than Log 3. Mean duration for anti-viral response in CMV viremia was 38.6667 ± 20.55 days. Overall mortality in children was 3/9, out of which only 1 was directly accounted for by disseminated CMV disease. Mean CD 3, CD 4 and CD 8 cell count at 4 weeks post CB-SCT were 213.44 ± 218.41, 73 ± 57.43 and 132.11 ± 168.58 days respectively.

ADV viremia was seen in 5/55 CB-SCT episodes. ADV viremia was complicated by disseminated disease in 1/5. Single agent was effective in all 5 children with viremia. Four of these viremia episodes were treated with cidofovir, administered as 1 mg/kg three times a week therapy and oral brincidofovir was given successfully to one patient. Mean duration for anti-viral response in ADV viremia was 21.2 ±8.027 days. Mean CD 3, CD 4 and CD 8 cell count at 4 weeks post CB-SCT were 299.6 ± 410.53, 153 ± 270.30 and 151±158.89 days respectively. EBV viremia was seen in 1/55 CB-SCT episodes. No complications were seen resulting from EBV viremia. Excellent response was seen as the child completely cleared EBV after a single dose of rituximab.

Conclusions: This retrospective cohort from a single centre reports that despite T-cell depletion and with CB infused T-cells being naïve, the incidence of infection is modest, and the response to therapy is generally good. The maximum incidence of viraemia was seen early and within the first 50 days. The results are expected to be still better once letermovir is available for prophylaxis of CMV infection.

Disclosure: Nothing to declare.

13: Infectious Complications

P444 PREVALENCE OF VZV REACTIVATION AND EFFECTIVENESS OF VACCINATION WITH RECOMBINANT ADJUVANTED ZOSTER VACCINE IN ALLOGENEIC HEMATOPOIETIC STEM CELL RECIPIENTS: A SINGLE-CENTER ANALYSIS

Ewa Karakulska-Prystupiuk 1, Magdalena Feliksbrot-Bratosiewicz1, Maria Król1, Agnieszka Tomaszewska1, Grzegorz Władysław Basak1

1Medical University of Warsaw, Warsaw, Poland

Background: Secondary immunodeficiencies in allo-HSCT recipients increase the risk of viral reactivation, making vaccinations a vital issue. There are no reports on the effectiveness of recombinant vaccine against herpes zoster after allo-HSCT.

Methods: The analysis included 149 recipients of allo-HSCT transplanted in years 2012-2022, mainly due to hematological malignancies (>95%).

Recombinant adjuvanted herpes zoster vaccine(RZV) was used in patients in accordance with the current recommendations of the Advisory Committee on Immunization Practices(ACIP).

Vaccination included two doses of the vaccine given 1-2 months apart. The ELISA method was used to serologically assess the titer of VZV IgG antibodies (Ratio: negative ≤0.8, equivocal 0.8-1.1, positive >1.1. Peripheral blood lymphocyte subpopulations were analyzed using flow cytometry (reference value for the absolute count of total CD3 + CD4+ was 309-1139 cells/µl). Post-vaccination complications were graded according to CTCAE5.0 criteria. Patients gave written consent to the intervention.

Results: The prevalence of VZV reactivation in the entire group before vaccination:.

VZV reactivation was diagnosed in 49 out of 149(33%) patients including 5(3%) patients with reactivation within the first year after transplantation and the remaining 44(30%) within the subsequent three years.

At that time, most of patients did not receive acyclovir prophylaxis anymore, including five patients who stopped recommended prophylaxis despite receiving immunosuppressive treatment (3 - due to chronic GvHD, 2 - for its prevention).

The most common clinical manifestation of VZV reactivation involving intercostal nerves was diagnosed in 40(81%) patients. The remaining patients had unusual locations, including 3 patients with cranial nerves involvement, 2 - with ophtalmicus, 2 – with ulnar nerves, 1- with genital involvement, and 1 with a disseminated form of herpes zoster. Four other patients required hospitalization: one due to disseminated herpes zoster, second- ophtalmicus and two other- for infectious complications (1 - pneumonia, 1 - bronchitis). Postherpetic neuralgia was an essential complication in many of them.

The assessment of the vaccinated group:.

16 patients recipients (median age: 43) received two doses of RZV (median time 3 years after transplantation (range, 1-6 years). There were 8-seronegative, 2-seropositive, and 6-unassessed patients before vaccinations. Thirteen of them have been vaccinated at an interval of 1 month, and the remaining three- at an interval of 2 months. Several patients complained about mild pain, erythema, swelling, or fatigue- CTCAE grade 1 after injection. During vaccination, four patients received chronic immunosuppressive treatment due to severe (3 patients) and moderate (1 patient) chronic cGvHD. The serological post-vaccination response was confirmed in 10 recipients with Ratio 2- 8. Half of vaccinated patients had the absolute count of total CD3 + CD4+ cells below LLN (median of counts- 260/µl).

Despite vaccination (and an increase in VZV IgG titer), three patients developed herpes zoster. These patients were not receiving immunosuppressive treatment at the time of illness, but one of them had received rituximab a year earlier.

Conclusions: Herpes zoster occurs mainly in the late period after allogeneic hematopoietic stem cell transplantation, in over 30% of recipients who have completed acyclovir prophylaxis. The obtained preliminary results indicate that RZV vaccination after allo-HSCT is safe but not always effective.

Disclosure: Nothing to declare.

13: Infectious Complications

P445 BK VIRUS-ASSOCIATED HEMORRHAGIC CYSTITIS IN POSTTRANSPLANT CYCLOPHOSPHAMIDE-BASED ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION (HCT) FOR IMMUNE DEFICIENCY OR DYSREGULATION

Dimana Dimitrova 1, Christi McKeown1, Scott Napier1, Anita Stokes1, Alison Cusmano1, Ruby Sabina1, Jennifer Sponaugle1, Elisabetta Xue1, Jeffrey Cohen1, Alexandra Freeman1, Luigi Notarangelo1, Gulbu Uzel1, Vladimir Valera Romero1, Christopher Kanakry1, Jennifer Kanakry1

1National Institutes of Health, Bethesda, United States

Background: Allogeneic HCT recipients may develop BK virus (BKV)-associated hemorrhagic cystitis (BKV-HC), early post-HCT. Yet, BKV levels in urine and blood are not routinely monitored pre- or post-HCT.

Methods: Quantitative urine and blood BKV PCR were prospectively monitored in patients with immune deficiency or dysregulation treated according to either of two HCT trials at our center (NCT02579967, NCT03663933). Engrafted children and adults (median age 25y, range 4-66) with at least 28 days follow up (n=74) were included in this analysis.

Results: Of 60 patients considered at-risk for BKV-HC based on BKV detection in urine/blood at any timepoint, 41 (68%) developed BKV-HC. Of recipients treated on the busulfan-containing trial arms, 41 of 55 (75%) at-risk patients developed BKV-HC (Table 1). By contrast, when busulfan was not used, 0 of 5 at-risk patients developed BKV-HC (p=0.0006). To assess aggregate contribution of busulfan and cyclophosphamide exposure, which have both been implicated as risk factors for BKV-HC, cyclophosphamide equivalent dose (CED) was calculated for each patient. Patients who developed BKV-HC received a greater CED than patients who did not (p=0.0016), but there was no correlation between CED and overall BKV-HC symptom duration nor BKV-associated gross hematuria duration.

At 2 weeks post-HCT, urine BKV levels of recipients who developed HC were significantly higher than in those who did not, Mann-Whitney p=0.007 (Figure 1). However, early immune reconstitution as defined by absolute lymphocyte count at days +14 and +21 did not correlate with development, post-HCT onset day, or duration of BKV-HC. At BKV-HC onset, median level of BKV in urine was 9.51 compared to 7.09 log10 IU/mL upon HC resolution, with only 16% patients achieving urine BK clearance by day 100 even if asymptomatic. Median day of symptom resolution coincided with viremia resolution.

With neutrophil engraftment at median day +17 (range 13-42), median onset of BKV-HC symptoms was day +23 (range +6-74), and median duration was 24 days, with 6 (15%) patients reporting ongoing symptoms beyond day +100. Patient discomfort and supportive care needs were significant, especially during gross hematuria (n=35; median duration 14 days, range 1-174). Four required continuous bladder irrigation, while one patient with a particularly severe course required multiple cystoscopies for clot evacuation but no intravesicular therapy. This patient developed eosinophilic cystitis 4 years later, but the potential contribution of prior BKV-HC is unknown. No patients received antiviral or cellular therapies for BKV-HC, although 4 received cidofovir/brincidofovir for adenovirus infection. Tissue-proven disease outside the bladder occurred in only 1 patient, who developed BKV nephropathy, along with radiographic and cytopathologic (SV40+ cells on bronchoalveolar lavage) evidence of BKV pneumonitis. In addition, maximum BK viremia >4 log10 IU/mL was not associated with increased incidence of thrombotic microangiopathy as has been shown in other studies.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (50)

Table 1. Patient and HCT characteristics in association with BKV-HC

Total patients

At-risk patients

BKV-HCd

Diagnosis, n total patients (%)

74 (100%)

60 (81%)

41 (68%)

RAG 1/2 deficiency

10 (14%)

8 (80%)

6 (75%)

APDSa

8 (11%)

6 (75%)

2 (33%)

Other nonmalignant disorder (with LPD/lymphoma)b

28 (38%)

21 (75%)

15 (71%)

Other nonmalignant disorder (no LPD/lymphoma)c

28 (38%)

23 (82%)

16 (70%)

Busulfan-containing (RIC) HCT platforms, n (%; marrow/PBSC):

68 (93%, 36/32)

55 (81%, 32/23)

41 (75%, 27/14)

PCBu2; GVHD ppx: PTCy100, sirolimus, +/- MMF

45 (66%, 36/8)

36 (65%, 32/4)

31 (86%, 27/4)

PCBu2; GVHD ppx: PTCy100, MMF35, sirolimus

23 (51%, 21/2)

18 (78%, 18/0)

15 (83%, 15/0)

PCBu2; GVHD ppx: PTCy100, MMF18, sirolimus

12 (27%, 8/4)

9 (75%, 7/2)

9 (100%, 7/2)

PCBu2; GVHD ppx: PTCy100, sirolimus (no MMF)

10 (22%, 7/3)

9, (90%, 7/2)

7 (78%, 5/2)

Equine ATG, PCBu2; GVHD ppx: PTCy100, MMF25, tacrolimus

23 (34%, 0/23)

19 (83%, 0/19)

10 (53%, 0/10)

Busulfan-free (NMA) HCT platforms, n (%; marrow/PBSC)

6 (8%; 3/3)

5 (%; 2/3)

PCBu2; GVHD ppx: PTCy100, MMF35, sirolimus

3 (50%; 2/1)

2 (67%; 1/1)

PCBu2; GVHD ppx: PTCy50, MMF35, sirolimus

1 (17%; 1/0)

1 (100%; 1/0)

Equine ATG; PCBu2; GVHD ppx: PTCy50, MMF25, tacrolimus

2 (25%; 1/1)

2 (100%; 1/1)

Outcomes by graft type (RIC platforms only), n (%)

T-replete marrow

36 (53%)

32 (89%)

27 (84%)

T-replete PBSC

32 (47%)

23 (72%)

14 (61%)

  1. Abbreviations: Bu2, 2 days of pharmacokinetically dosed busulfan, target daily AUC 3600-5600 micromol*min; GVHD, graft-versus-host disease; LPD, lymphoproliferative disorder; MMF, mycophenolate mofetil through day +18, +25, or +35; NMA, nonmyeloablative; PBSC, peripheral blood stem cells; PC, pentostatin and hyperfractionated cyclophosphamide; PTCy100, posttransplantation cyclophosphamide, 50mg/kg/day on days +3 and +4; PTCy50, posttransplantation cyclophosphamide, 25mg/kg/day on days +3 and +4; RIC, reduced intensity conditioning.
  2. a4 APDS patients had lymphoma/LPD as a HCT indication
  3. bGenetic defects: MAGT1 deficiency (4), CTLA4 haploinsufficiency (2), IKZF1 gain of function (2), other known defect (8), unknown defect (12)
  4. cGenetic defects: ADA2 deficiency (4), PRF1 deficiency (3), STAT3 deficiency (3), IFNGR1 deficiency (2), other known defect (8), unknown defect (8)
  5. dPercentages expressed as percent of at-risk patients

Conclusions: In this cohort, the use of busulfan and possibly cyclophosphamide seems to be an important co-factor in BKV-HC risk, as also noted by others. Day +14 urine BKV levels were significantly higher in patients who ever developed symptomatic BKV-HC, but early post-HCT lymphocyte counts did not correlate with symptom evolution.

Clinical Trial Registry: Clinicaltrials.gov: NCT02579967, NCT03663933.

Disclosure: Nothing to declare.

13: Infectious Complications

P446 INFECTION PREVENTION MEASURES TAKEN FOR ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT PATIENTS DURING THE COVID-19 PANDEMIC AND THEIR IMPACT ON CLINICAL OUTCOMES

Sarah Noetzlin1, Michael Bader2, Andriyana Bankova3, Dominik Schneidawind3, Jakob Passweg2, Anne-Claire Mamez1, Federica Giannotti1, Sarah Morin 1, Federico Simonetta1, Stavroula Masouridi-Levrat1, Dionysios Neofytos1, Yves Chalandon1

1Hôpitaux Universitaires de Genève, Geneva, Switzerland, 2Universitätsspital Basel, Basel, Switzerland, 3Universitätsspital Zürich, Zürich, Switzerland

Background: Respiratory infections are the second most common infection site during the allogeneic hematopoietic stem cell transplant (HSCT) procedure, with a significant morbidity and mortality risk. Strict isolation protocols might diminish this risk. During the COVID pandemic, supplementary infection prevention measures have been implemented, with complete visitors ban to the HSCT unit. We hypothesize that this measure might have not only prevented SARS-CoV-2 infections, but also all common respiratory tract infections (RTI), with a potential effect on non-relapse mortality.

Methods: We performed a national, multi-center, retrospective cohort study including all consecutive adult allogeneic HSCT recipients in the three allogeneic HSCT-centers in Switzerland between 2019 and 2021. The primary objective was the incidence of RTI of all causes (viral, bacterial, fungal) from conditioning until hospital discharge after engraftment. Secondary outcomes included gastro-intestinal infections, need for transfer to the intensive care unit (ICU), all-cause non-relapse mortality, and grade≥2 acute/chronic graft-versus-host disease (GvHD). Outcomes were compared between group-1 (01.01.2019-31.12.2019, pre-COVID-era) and group-2 (01.04.2020-30.30.2021, COVID-era).

Results: Overall, 203 and 221 HSCT recipients were included in group-1 and group-2, respectively. The groups were well balanced, with a median age of 58 years, and two third of male patients. RTI were significantly more frequent in group-1 (43/203, 21.2%) compared to group-2 (23/221, 10.4%, p=0.003). Non-SARS-CoV-2 viral RTI (vRTI) were significantly decreased in group-2 (2/221, 0.9%) versus group-1 (16/203, 7.9%, p<0.001), including: rhinovirus (8/18, 44.4%), picornavirus (3/18, 16.7%), parainfluenza virus (2/18, 11.1%), and non-SARS-CoV2 coronavirus (1/18, 5.6%). No difference was observed in the incidence of bacterial (group-1: 11/203, 5.4%; group-2: 10/221, 4.5%; p=0.82%) or fungal RTI (group-1: 16/203, 7.9%, group-2: 11/221, 5.0%; p=0.24) between the two groups. In multivariable analysis, the only two factors significantly reducing the RTI incidence included transplantation in the latter study-period (OR 0.44, 95%CI 0.26-0.77, p=0.004) and peripheral blood stem cell source (OR 0.44, 95%CI 0.20-0.99, p=0.048). No effect of the study period was observed on gastro-intestinal infections (group-1: 6/203, 2.96%, group-2 10/221, 4.52%, p=0.453). ICU transfer were less frequent in the COVID period, albeit not statistically significantly (group-1: 18/203, 8.87%, group-2: 10/221, 4.52%, p=0.080). Although acute and chronic GvHD rates were similar in both groups, non-relapse mortality was lower at 1-year post-transplant in group 2 (4.5% vs 11.3%, p=0.0056).

Group 1

Group 2

P

N

%

N

%

203

221

Respiratory infection

Absence

160

78.82

198

89.59

0.003

Presence

43

21.18

23

10.41

Respiratory infection pathogen

Viral

16

7.88

2

0.90

<0.000

Bacterial

11

5.42

10

4.52

0.823

Fungal

16

7.88

11

4.98

0.238

Respiratory infection location

Upper airways

9

4.43

5

2.26

0.009

Lower airways

34

16.75

18

8.14

Viral Respiratory infection location

Upper airways

8

3.94

1

0.45

0.016

Lower airways

8

3.94

1

0.45

Gastro-intestinal infection

Presence

6

2.96

10

4.52

0.453

Gastro-intestinal infection pathogen

Viral

2

0.90

0.500

Bacterial

6

2.96

8

3.62

Conclusions: Visitor ban in the HSCT unit significantly decreases the risk of RTI, mainly vRTI, and 1-year all-cause non-relapse mortality. More data on restrictive visitor policies and clinical outcomes are urgently needed.

Disclosure: Y.C. has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution.

D. N. has received research support from MSD and Pfizer and consulting fees from Roche Diagnostics, MSD, Pfizer, Basilea, and Gilead.

S.M has received travel support from Travel Gilead, BeiGene, Jazz and Sanofi all via the institution.

F.S. has received consulting fees from BMS/Celgene, Incyte, Kite/Gilead; speaker fees from Kite/Gilead, Incyte; travel support from Kite/Gilead, Novartis, AstraZeneca, Neovii, Janssen; research funding from Kite/Gilead, Novartis, BMS/Celgene.

13: Infectious Complications

P447 EFFECT OF EARLY ADMINISTRATION OF ANTI-MRSA DRUGS ON THE COURSE OF FEBRILE NEUTROPENIA IN ALLO-SCT

Anna Akaogi 1, Junya Kanda1, Fumiya Wada1, Yasuyuki Arai1, Chisaki Mizumoto1, Toshio Kitawaki1, Kouhei Yamashita1, Akifumi Takaori-Kondo1

1Kyoto University, Kyoto, Japan

Background: In febrile neutropenia (FN), the empiric use of anti-methicillin-resistant Staphylococcus aureus (MRSA) drugs is generally not recommended; however, in cases where mucosal damage is present during quinolone antibiotic administration, their empirical use is recommended, particularly in many allogeneic hematopoietic stem cell transplantation (allo-SCT) patients.

Methods: We conducted a retrospective analysis of 184 allo-SCT patients to evaluate the impact of early administration of anti-MRSA drugs on FN and transplantation outcomes. Patients receiving anti-MRSA drugs within 3 days of fever onset were defined as the early decision group. Those treated on or after day 4 or untreated constituted the late decision group. The primary endpoint was the success rate of fever resolution at day7 after FN onset. Secondary endpoints included engraftment rate, hospital stay duration, grade II-IV acute graft-versus-host disease (aGVHD) incidence. The number of days from the onset of FN to fever resolution was compared as continuous variables using the Mann-Whitney U test. Multivariate logistic regression analysis was employed to assess the association between background factors and the fever resolution rate on day 7 after FN onset. Engraftment, discharge and aGVHD rates were analyzed using cumulative incidence methods and these were compared with Gray test. Fine and Gray proportional hazards models were used for multivariate analysis of aGVHD rates.

Results: The median age at the time of transplantation was 50 years, with 108 patients undergoing myeloablative conditioning and 76 receiving reduced-intensity conditioning. Transplant sources included 100 bone marrow cases, 18 peripheral blood cases, and 66 cord blood cases. The duration from FN onset to fever resolution was significantly shorter in the early decision group than in the late decision group (P = 0.041). Following multivariate analysis, a negative correlation persisted between the late decision group and successful fever resolution (OR: 0.44, P = 0.023). There is no significant correlation between the timing of MRSA drug administration and the cumulative incidence rates of the development of aGVHD, engraftment and discharge. Multivariate analysis was performed, stratifying by degree of mucosal damage, age, disease status, HLA compatibility, and source of transplant, to examine the fever resolution rate and the incidence of aGVHD between the early and late decision groups. In the analysis limited to cases with severe mucosal disorders, OR for fever resolution in the late decision group compared to the early group was 0.36, which was lower than the OR in the analysis of the entire cohort. The subgroup analysis also highlighted an increased incidence of aGVHD in older patients in the late decision group (HR: 2.15, P = 0.042).

Conclusions: This report highlights the positive impact of early anti-MRSA drug administration on the resolution of fever in FN in allo-SCT. It also suggests that early use of them may reduce the risk of aGVHD in elderly patients.

Disclosure: Junya Kanda received honorarium from Amgen Inc., Janssen Pharmaceuticals Companies, Novartis International AG, Sanofi S.A., AbbVie Inc. and Megakaryon Corporation.

Junya Kanda has received a scholarship grant from Eisai Co., Ltd.

Toshio Kitawaki received honorarium from Novartis International AG.

13: Infectious Complications

P448 VISCERAL TOXOCARIAS IN THE EARLY POST-TRANSPLANT COURSE

Iacopo Bellani 1, Francesca Vendemini2, Sonia Bonanomi2, Sergio Foresti3, Guglielmo Marco Migliorino3, Gaia Kullmann3, Alex Moretti2, Sergio Maria Ivano Malandrin4, Adriana Balduzzi2,1

1School of Pediatrics, University of Milano Bicocca, Monza, Italy, 2Fondazione IRCCS San Gerardo of Tintori, Monza, Italy, 3IRCCS San Gerardo of Tintori, Monza, Italy, 4Microbiology Laboratory, IRCCS San Gerardo of Tintori, Monza, Italy

Background: Parasitic diseases are rarely documented after HSCT, but may be life-threatening. As for understudied infections, clinical management is often based on expert recommendations.

Visceral Toxocariasis, caused by migrating Toxocara larvae, is a worldwide zoonosis that occurs mainly in children asymptomatically. However, diagnostic difficulties and potentially fatal complications(1) require major awareness in transplanted patients for prompt recognition and appropriate therapy.

Methods: We report the case of visceral Toxocariasis in the second month after allogeneic HSCT in an Egyptian girl transplanted at 15 months of age, after treosulfan-fludarabine-thiotepa, from her HLA-identical sister, with no serotherapy, for an high-risk infant acute myeloid leukemia (AML).

Results: The girl achieved both stable engraftment and disease remission, but on day +27 she developed unresponsive fever, diarrhea and abdominal pain. Laboratory tests, blood and stool coltures, and ultrasound scans were negative, except for slightly increased inflammatory indices and hepatomegaly. After piperacilline/tazobactam was added to her anti-infectious prophylaxes (acyclovir, liposomal amphotericin BIW and trimethoprim/sulfamethoxazole), her diarrhea rapidly resolved. On day +35 a diffuse non-itching maculo-papular rash was reported and, in the suspect of acute GvHD, methylprednisolone was started, with poor clinical response and only 48-hour-apyrexia. Total body CT scan showed an excavation in the left lower pulmonary lobe, therefore linezolid was initiated. Continuous diclofenac infusion was required to control persistent fever.

On day +44 focal seizures occurred, with facial deviation, hypotonia and drowsiness, controlled by midazolam and levetiracetam. Electroencephalography recorded a left occipital focus, suggestive for infection, and brain MRI revealed cerebellar white matter hyperintensity. Predominant eosinophilia was detected in the CSF, with corresponding eosinophilia in the peripheral blood, that had increased slowly but progressively in the previous days. Direct microscopy on blood and CSF detected no pathogens, including parasites. Ophthalmic evaluation, parasitological examinations on the stools and toxoplasma serology resulted also negative.

The patient was treated empirically with albendazole and dexametasone. Finally, the diagnosis was confirmed by Immunoblotting detection of anti-Toxocara spp. antibodies on blood and CSF.

On day +52 a follow-up CT scan detected a cardiac granulomatous lesion of parasitic nature that was growing rapidly in almost the entire right atrium, obstructing the outflow through the tricuspid valve. Due to the risk of sudden death, heparin was started and the patient underwent cardiac surgery with sternotomy, but the mass could only partially be removed and heparin was continued.

After 6 week of treatment with albendazole, no signs of Toxocariasis recurrence were observed after discontinuation. Neurologically, the worsening of cerebellar lesions, with cortical and subcortical damage, resulted in severe psychomotor impairment and generalized hypotonia, with loss of head and trunk control and feeding difficulties, with only little improvement after rehabilitation.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (51)

Conclusions: Toxocariasis is a widespread disease and an emerging complication after HSCT, which should be considered in case of eosinophilia, unresponsive fever and progressive systemic involvement. Improving diagnostic and knowing the serological Toxocara status of HSCT recipients may be useful to detect a neglected infection.

(1) Haque E, et al. Parasitic infections in hematopoietic stem cell transplant recipients. Transpl Infect Dis. 2023 Nov; https://doi.org/10.1111/tid.14160, PMID: 37793057.

Disclosure: Nothing to declare.

13: Infectious Complications

P449 DE-ESCALATION ANTIBIOTIC STRATEGY IN MULTIDRUG-RESISTANCE BACTERIAL COLONIZED PATIENTS AFTER ALLOGENEIC STEM CELL TRANSPLANTATION. A 3-YEAR RETROSPECTIVE STUDY

Roberto Bono1, Giuseppe Sapienza 1, Stefania Tringali1, Cristina Rotolo1, Alessandra Santoro2, Laura Di Noto3, Orazia Di Quattro4, Caterina Patti5, Luca Castagna1

1U.O.S.D. UTMO AOOR Villa Sofia-Cervello, Palermo, Italy, 2Laboratorio di diagnostica integrata oncoematologica e manipolazione cellulare, Palermo, Italy, 3U.O.C di Medicina Trasfusionale e dei Trapianti AOOR Villa Sofia -Cervello, Palermo, Italy, 4U.O.C Microbiologia e Virologia AOOR Villa Sofia-Cervello, Palermo, Italy, 5U.O.C Oncoematologia AOOR Villa Sofia-Cervello, Palermo, Italy

Background: Colonization by multidrug resistance (MDR) bacterial and related bloodstream infections (BSI) are associated with high rate of mortality, reported to be up to 70%, in aplastic patients.

The 2020 ECIL guidelines suggested for these patients an empirical adjusted treatment. It has been suggested that a de-escalation approach could reduce the selective pressure limiting the risk for ultra-multiresistant strain appearance. The aim of this study was to describe the de-escalation approach in a cohort of patients colonized by MDR bacteria, receiving empiric antibiotic therapy (EAT) active against resistant strain, at first neutropenic febrile episode (NFE) after allo-SCT, in a high prevalence environment.

Methods: Starting from 2021, we adopted the de-escalation approach for management of patients colonized with MDR bacteria, mostly carbapemenase-producing Klebsiella pneumoniae (KPC), before allo-SCT. The strategy was to treat those patients at first febrile episode with active against KPC (ceftazidime-avibactam or meropenem-vaborbactam, based on ATB susceptibility), and to de-escalate to conventional antibiotics (piperacilline-tazobactam, cefepime) after 48h if blood cultures were negative for MDR bacteria. The first line EAT was piperacillin-tazobactam or cefepime. Microbiological inclusion criteria were: detection of stool MDR colonization and/or systemic infections sustained by MDR bacteria any time during the conventional treatment; detection of MDR bacteria in the stool during the work-up before allo-SCT. Other inclusion criteria were age >18y, allo-SCT from any donor, any conditioning regimen, any stem cell source. The primary end-point is the attributable mortality rate (AMR) to MDR sepsis in patients de-escalated. Secondary objectives are feasibility of de-escalation, MDR-directed ATB duration, survival.

Results: 79 patients were included and 22 (28%) satisfied microbiological inclusion criteria (Table). Two patients did not develop fever after allo-SCT. At first NFE, KPC-active EAT was started in 75% of patients (15/20), mostly with ceftazidime/avibactam+aminoglycoside (79%), and meropenem/vaborbactam in 21% (5/20). The median time of KPC-active EAT duration was 3.5 days (range 2-19). 5 out of 20 (25%) received piperacilline-tazobactam, but 4 of them were not colonized at time of BMT unit admission. In these patients, KPC translocation was not observed.

The KPC translocation rate in the pre-engraftment period was detected in 10% of patients (2/20). The 30-day mortality rate was 5% and 3-months not relapse mortality rate was 14%. KPC 3-months AMR was 5%. 1 patient relapse at day +100.

Conclusions: The results from this retrospective analysis suggests that KPC-AMR is low using targeted EAT and that de-escalation strategy is feasible and safe, allowing to reduce the treatment duration and probably preserving the emergence of ultra-resistant strains.

All patients

MDR colonized patients

N= 79

N= 22 (28%)

Median age

55 (19-73)

59 (41-73)

Sex M/F

44/35

14/8

Disease

AML

52 (66%)

18 (82%)

ALL

15 (19%)

3 (14%)

MDS

7 (9%)

1 (4%)

MPN

5 (6%)

/

Donor

MUD

22 (28%)

6 (23%)

Haplo

28 (35%)

8 (36%)

HLAid sib

14 (18%)

6 (32%)

mMUD

15 (19%)

2 (9%)

GVHD prophylaxis

PTCY-based

44 (56%)

10 (45%)

ATG-based

34 (43%)

12 (55%)

CSA+short MTX

1 (1%)

/

Conditioning regimen

MAC

67 (85%)

19 (86%)

RIC

12 (15%)

3 (14%)

Stem cell source

PBSC

75 (95%)

21 (95%)

BM

4 (5%)

1 (5%)

Disclosure: i have no disclosures.

13: Infectious Complications

P450 MARIBAVIR FOR TREATMENT OF REFRACTORY/RECURRENT CYTOMEGALOVIRUS INFECTION IN ALLOGENEIC HSCT RECIPIENTS

Aleksandr Siniaev1, Ivan Moiseev1, Marina Popova 1, Yulia Rogacheva1, Yulia Vlasova1, Sergey Bondarenko1, Alexander Kulagin1

1RM Gorbacheva Research Institute, Saint Petersburg, Russian Federation

Background: There is a lack of real-world data on maribavir therapy in recipients of allogeneic HSCT.

Methods: Our single-center study included 15 adult recipients of allogeneic HSCT with refractory or recurrent cytomegalovirus (CMV) infection. Detection of CMV drug resistance mutations was not performed. Detailed group characteristics are presented in Table 1. The median time to CMV infection onset was 36.5 days (range 20 – 141), with ganciclovir (n=14, 93.3%) being the first-line therapy of choice. Maribavir was administered as a second-line therapy in 10 cases (66.7%), and as a subsequent line of therapy in 5 cases (33.3%).

Study group (n=15)

Age, median (range)

43 (19 – 66)

Sex

Male

7 (46.7%)

Female

8 (53.3%)

Diagnosis

AML

8 (53.2%)

ALL

2 (13.5%)

MDS

2 (13.5%)

MM

1 (6.6%)

CLL

1 (6.6%)

AA

1 (6.6%)

Therapy group

Standard

11 (73.3%)

Salvage

4 (26.7%)

Donor

MMUD

9 (60%)

Haploidentical

4 (26.7%)

MUD

1 (6.7%)

MRD

1 (6.6%)

Stem cell source

PBSC

14 (93.3%)

BM

1 (6.7%)

Conditioning regimen

MAC

9 (60%)

RIC

6 (40%)

Post-transplant cyclophosphamide GVHD prophylaxis

15 (100%)

Time of CMV infection onset after 1st allo-HSCT, median (range), days

36.5 (20 – 141)

2nd HSCT (graft failure)

4 (26.6%)

Boost (poor graft function)

1 (6.6%)

Localization of CMV infection

Blood (plasma)

14 (93.3%)

CMV colitis

1 (6.7%)

CMV DNA levels at the start of 1st line treatment, median (range)

IU/ml

4390 (100 – 4656000)

Copies/ml

8530 (167 – 7760000)

First-line therapy

Ganciclovir

14 (93.3%)

Valganciclovir

1 (6.7%)

Refractory CMV infection

12 (80%)

Recurrent CMV infection

3 (20%)

Duration of 1st line therapy, median (range), days

12.5 (9 – 20)

2nd line therapy

Maribavir

10 (66.7%)

Leflunomide

2 (13.3%)

Foscarnet

1 (6.7%)

Ganciclovir

1 (6.7%)

Valganciclovir

1 (6.6%)

Duration of 2nd line therapy, median (range), days

25.5 (2 – 74)

  1. Abbreviations: AML, acute myeloid leukemia; ALL, acute lymphoblastic leukemia; MDS, myelodysplastic syndrome; MM, multiple myeloma; CLL, chronic lymphocytic leukemia; AA, aplastic anemia; MMUD, mismatched unrelated donor; MUD, matched unrelated donor; MRD, matched related donor; PBSC, peripheral blood stem cell; BM, bone marrow; MAC, myeloablative conditioning; RIC, reduced intensity conditioning; GVHD, graft-versus-host disease; CMV, cytomegalovirus.

Results: The median time from CMV infection onset to the administration of maribavir was 14 days (range 10 – 486). The median duration of maribavir therapy was 28 days (range 2 – 59). The median level of IU/mL at the start of maribavir treatment was 15,200 (range 10,453 – 335,169), and the median number of copies/mL was 12,250 (range 6,930 – 559,000). Clearance was achieved in 10 cases (66.6%) at 8 weeks from the start of therapy. Five patients (33.4%) deceased before reaching the 8-week mark, although 1 patient did achieve CMV clearance eventually. The median time to achieve clearance after starting maribavir treatment was 20 days (range 7 – 84). Five patients (33.4%) maintained CMV clearance 16 weeks after the start of maribavir treatment. After the end of treatment, 4 patients (26.6%) developed a recurrent episode of CMV infection. Grade 3-4 adverse events during treatment were observed in 14 cases (93.3%), but these were not considered related to maribavir therapy. There were 8 lethal outcomes (53.3%) in the group. The median follow-up time after the first HSCT was 179 days (range 47 – 1001). During maribavir therapy, we also observed the reactivation of other herpesviruses: human herpesvirus-6 in 8 cases (53.3%), Epstein-Barr virus in 2 cases (13.3%), herpes simplex virus types 1 and 2 in 1 case (6.6%), and 1 case (6.6%) of infection caused by varicella-zoster virus. The one-year overall survival rate in the group was 46.7% (95% CI 21.2 – 68.7).

Conclusions: Maribavir therapy demonstrated efficacy in achieving CMV clearance, comparable to the results of the SOLSTICE study. However, maintaining this clearance and managing CMV recurrence remains a challenge. Careful monitoring and management of other herpesvirus infections are important during maribavir therapy.

Disclosure: Maribavir was obtained through extended access program. COI: M.I.: honoraria from Takeda.

13: Infectious Complications

P451 REAL LIFE MANAGEMENT OF ANTIBIOTIC THERAPY IN HSCT RECIPIENTS – FOCUS ON DE-ESCALATION IN PRE-ENGRAFTMENT NEUTROPENIA, THE STUDY FROM THE EBMT INFECTIOUS DISEASES WORKING PARTY (IDWP)

Malgorzata Mikulska 1, Lotus Wendel2, Gloria Tridello2, Alexander Kulagin3, Anna Czyz4, Nabil Yafour5, Fabio Ciceri6, Burak Deveci7, Simona Sica8, Kristina Carlson9, Matthias Eder10, Agnieszka Tomaszewska11, Tunc Fisgin12, Michel Schaap13, Michal Karas14, Anastasia Pouli15, Elisabetta Calore16, Marta Gonzalez Vicent17, Mariagrazia Michieli18, Maria Jesús Pascual Cascon19, Alessandra Picardi20, Gergely Krivan21, Anna Torrent Catarineu22, Tessa Kerre23, Pedro Chorão24, Fabio Benedetti25, Alessandra Carotti26, Melissa Gabriel27, Rik Schots28, Amjad Hayat29, Antonio Perez Martinez30, Domenico Pastore31, Mauro Turrini32, Diana Averbuch33, Jan Styczynski34, Rafael de la Camara35

1University of Genova and IRCCS Ospedale Policlinico San Martino, Genova, Italy, 2EBMT, Leiden Study Unit, Leiden, Netherlands, 3RM Gorbacheva Research Institute, Pavlov University, Petersburg, Russian Federation, 4Wroclaw Medical University, Wroclaw, Poland, 5University Hospital of Oran, Oran, Algeria, 6IRCCS Ospedale San Raffaele s.r.l., Milano, Italy, 7Medstar Antalya Hospital, Antalya, Turkey, 8Fondazione Policlinico Universitario A. Gemelli IRCCS, Cattolica del Sacro Cuore, Rome, Italy, 9University Hospital Uppsala, Uppsala, Sweden, 10Hannover Medical School, Hannover, Germany, 11Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland, 12Altinbas University, Faculty of Medicine, Bahçelievler Medicalpark Hospital, Pediatric Bone Marrow Transplantation Unit, Istanbul, Turkey, 13Nijmegen Medical Centre, Nijmegen, Netherlands, 14Charles University Hospital, Pilsen, Czech Republic, 15St. Savvas Oncology Hospital, Athens, Greece, 16Padua University Hospital, Padova, Italy, 17Niño Jesus Children`s Hospital, Madrid, Spain, 18Centro di Riferimento Oncologico, Aviano, Italy, 19Hospital Regional de Málaga, Malaga, Spain, 20Cardarelli Hospital, Napoli, Italy, 21Central Hospital of Southern Pest, Budapest, Hungary, 22ICO-Hospital Universitari Germans Trias i Pujol, Badalona, Spain, 23Ghent University Hospital, Gent, Belgium, 24University Hospital La Fe, Valencia, Spain, 25Policlinico G.B. Rossi, Verona, Italy, 26Sezione di Ematologia, Perugia, Italy, 27The Children`s Hospital at Westmead, Sydney, Australia, 28Universitair Ziekenhuis Brussel, Brussels, Belgium, 29Galway University Hospital, Galway, Ireland, 30Hospital Universitario La Paz, Madrid, Spain, 31Perrino Hospital, Brindisi, Italy, 32Valduce Hospital, Como, Italy, 33Pediatric Infectious Diseases, Faculty of Medicine, Hebrew University of Jerusalem, Hadassah Medical Center, Jerusalem, Israel, 34Collegium Medicum UMK, Bydgoszcz, Poland, 35Hospital de La Princesa, Madrid, Spain

Background: Antimicrobial resistance contributes significantly to higher mortality in patients with infections during neutropenia. De-escalation strategy has been proposed to balance the need for prompt and active antibiotic therapy and to reduce the negative impact of prolonged administration of broad-spectrum agents.

The aim of this study was to report the real-life management of antibiotic therapy used in different HSCT centres, and to identify predictors of de-escalation.

Methods: This cross-sectional point prevalence study was performed in EBMT centres over single days of four non-consecutive weeks a month apart and focused on the use of antibiotics in inpatients being admitted to HSCT ward on these days who were with pre-engraftment neutropenia (< 500 PMN) at that moment or were neutropenic for at least 3 days during the previous 14 days. Data on antibiotic therapy in infectious episodes classified as microbiologically documented infection (MDI), clinically documented infection (CDI) or fever of unknown origin (FUO) were collected. The rates of de-scalation and discontinuation and its predictors were analysed.

Results: Overall, 251 patients from 32 centres in 16 countries were reported: 58 did not have fever or other signs/symptoms of infection; 193 patients with at least one episode of neutropenic fever were included in the study (single episode in 147 patients, more than one episode in 46 patients). Among 193 patients, 53% we male, median age was 54y, 20% were <18yo, 23% were transplanted for AML, 64% received alloSCT, 36% autoSCT. Among 123 alloSCT recipients: 18% donors were MR; PT-CY was used in 44%, myeloablative conditioning in 58%. Median time from HSCT to the first fever episode was 4 days (95% CI 3.2-4.6). Data for all 193 first episodes are reported.

113 patients (58.5%) had FUO, 40 (20.7%) had MDI (with BSI in 25, with severe clinical presentation in 11), and 40 had CDI; overall 30 (15.5%) had severe clinical presentation.

At the time of the first fever episode, 110 patients (57%) were already receiving antibiotic therapy, 75.5% of them fluoroquinolone prophylaxis, other either other prophylaxis or treatment of infection occurring pre-conditioning.

The treatment of the first neutropenic episode included piperacillin/tazobactam in 88 (46%) patients, cephalosporin in 52 (27%) and a carbapenem in 42 (21%); it consisted of a combination therapy in 87 (45%). The empirical therapy was changed in 105 (54.4%) of patients, and continued unchanged until engraftment in 45.6%.

De-escalation occurred in 66 (34.2%) patients, and it consisted of reducing the spectrum of antibiotics in 33 (17.1%) and/or discontinuation of antibiotic therapy in median 4 days before engraftment in 40 (20.7%).

The reason for not performing de-escalation was based on patient’s clinical situation 74% of cases, and on centre’s policy in 26%.

Among the analysed demographic, HSCT-related and fever-related variables, few identified predictors of de-escalation/discontinuation were the type of infection, the earlier timing of the first fever episode, center location and type of empirical therapy (Table 1).

Full size table

.

Conclusions: Real life rate of de-escalation/discontinuation remains low during pre-engraftment neutropenia.

Disclosure: None.

13: Infectious Complications

P452 INFECTIOUS COMPLICATIONS IN 549 BMTS DONE WITHOUT CENTRAL HEPA FILTRATION OR POSITIVE PRESSURE?

Rajpreet Soni1,2, Lawrence Faulkner 3,4, Priya Marwah1,2, Harshita Agarwal1, Deepa Trivedi5, Vaibhav Shah5, Mohan Reddy6, Neema Bhatt6, Stalin Ramprakash4, Rajat Kumar Agarwal4,7, Rachna Narain1,2, Ganesh Narain Saxena1,2

1SEAIT, Jaipur, India, 2Mahatma Gandhi University of Medical Sciences and Technology, Jaipur, India, 3Cure2Children Foundation, Florence, Italy, 4Sankalp India Foundation, Banglore, India, 5Sankalp-CIMS, Ahemdabad, India, 6Sankalp-BMJH Centre for Pediatric Hematology Oncology and BMT, Banglore, India, 7Jagriti InnoHealth Platforms Pvt. Ltd., Banglore, India

Background: To review infectious complication rates in allogeneic hematopoietic cell transplantations (HCT) carried out in non-HEPA-filtered single units.

Methods: A total of 549 consecutive HCTs, 435 matched-related (MRD group), and 114 partially matched-related (PMRD group) carried out from February 2012 to December 2022 in 3 BMT centers in India were reviewed. The median age for the MRD group was 8.1 (range 1-20.5) years and for the PMRD group was 7.1 (range 1.4 to 17.4) years. All patients had severe thalassemia, G-CSF-primed bone marrow was used as graft source. Conditioning consisted primarily of early ATG (day -12 to -10), busulfan and cyclophosphamide, with the addition of fludarabine and post-transplant cyclophosphamide in the PMRD group. Fluconazole and acyclovir was used as anti-infectious prophylaxis.

Results:

MRD group

PMRD group

Days with neutrophil counts < 200 µL

13(5-43)

14(11-48)

Days with neutrophil counts < 500 µL

17(9-55)

14(9-37)

Fever neutropenia with or without abnormal CRP or PCT

54%

46%

Central line-associated blood stream infection (CLABSI)

12%

14%

Multidrug-resistant germ CLABSI

7%

17%

CMV reactivation

13%

39%

BK virus infection

4%

12%

Invasive fungal disease

5%

13%

Possible

3%

1%

Probable

2%

12%

Conclusions: Our retrospective data review suggests that the rate of potentially airborne infections, primarily invasive fungal disease, is quite low and not much different from that reported from HEPA-filtered and positive pressure HCT units internationally, particularly for matched-related transplants in patients with thalassemia. We believe that MRD BMTs in patients with thalassemia can be safely performed in non-HEPA-filtered units. This observation in a large number of transplants might have a significant impact on the design, complexity and cost of transplant units as well as on BMT access, particularly in settings with limited resources and lacking universal health care coverage.

Disclosure: Nothing to declare.

13: Infectious Complications

P453 COVID-19 IN PATIENTS WITH HAEMATOLOGICAL DISEASES: THE ASSOCIATION OF RNAEMIA WITH CLINICAL OUTCOMES IN VACCINATED PATIENTS

Francisco Manuel Martin Dominguez 1, Sonsoles Salto-Alejandre2, Carmen Infante-Domínguez2, Mónica Carretero-Ledesma2, Natalia Maldonado-Lizarazo3, Pedro Camacho-Martínez2, Inmaculada Tallón-Ruiz4, Zaira Palacios-Baena3, Patricia Pérez-Palacios3, Rocío Álvarez-Marín2, José Antonio Lepe-Jiménez2, José Miguel Cisneros2, María Elisa Cordero2, Jerónimo Pachón2, Javier Sánchez-Céspedes2, José Antonio Pérez-Simón1, Manuela Aguilar-Guisado2

1University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS / CISC), Seville, Spain, 2Division of Infectious Diseases, Microbiology and Parasitology, Virgen del Rocío University Hospital, Seville, Spain // Institute of Biomedicine of Seville (IBiS), Seville, Spain, 3Unit of Infectious Diseases and Microbiology, Virgen Macarena University Hospital, Seville, Spain // Institute of Biomedicine of Seville (IBiS), Seville, Spain, 4Unit of Hematology, University Hospital Virgen Macarena, Instituto de Biomedicina de Sevilla (IBIS / CISC), Seville, Spain

Background: COVID-19 patients with hematologic diseases are more likely to have severe disease, with increased risk of intensive care admission and mortality. The aim of this study was to analyse how SARS-CoV-2 infection and RNAemia and the specific humoral immune response impact on the clinical evolution of patients with haematological diseases receiving chemotherapy or CAR-T therapy and HSCT recipients, and to determine the impact on the clinical outcome.

Methods: Multicentre cohort of consecutive adult COVID-19 patients with haematologic malignancies. IFN-α/IFN-γ serum levels (ELISA), neutralizing antibodies, and RNAemia by RT-PCR were evaluated at hospital admission. The primary outcome was all-cause mortality after COVID-19 diagnosis at days +30 and +90, and the secondary outcome the need of high-flow nasal oxygen. We also analysed the impact of RNAemia, immune and inflammatory variables on the outcomes. Multivariable Cox regression analyses were performed to assess the impact of every exposure variable on the primary and secondary end-points.

Results: 107 patients with COVID-19 were included, with a median age of 66 years, 59.8% male. The most common disease was non-Hodgkin lymphoma (53.3%). 58.9% patients had RNAemia at COVID-19 diagnosis and of them 41.3% had persistent RNAemia. 66.4% patients received antiviral therapy. 13.1% patients needed HFNO during follow-up. All-cause mortality at day +30 occurred in 9 (8.4%) and at day +90 in 21 (19.6%) cases.

The presence of RNAemia at diagnosis was associated with pneumonia (p=0.045), higher WHO clinical progression scale at diagnosis (p=0.042) and need for HFNO during admission (p=0.001), whereas persistent RNAemia was associated with pneumonia (p=0.051), RNAemia at diagnosis (p<0.001), need for HFNO during admission (p=0.002) and all-cause mortality at the end of the follow-up (day +90, p=0.005).

Taking into account Cox regression analysis, age > 70 years (p =0.03) was associated with all-cause mortality at day +90. A CURB-65 score > 2 at diagnosis in patients with pneumonia and persistent RNAemia were associated with mortality at day +30 (p=0.002 and p=0.044, respectively) and +90 (p=0.001 and p=0.009). In the Kaplan-Meier analysis, at day +90, the overall survival showed a trend to be lower (p=0.071) in patients with RNAemia at diagnosis and was lower (p=0.006) in patients with persistent RNAemia.

To find if RNAemia was independently associated to mortality at +30 and +90, a model of multiple Cox regression was developed. Persistent RNAemia (p=0.045 and p=0.052) and the age > 70 years (p=0.023 and p=0.004) were independently associated with all-cause mortality at day +30 and +90 (table 1).

Variable

HR (95% CI)

P

All-cause mortality at day + 30

Age older than 70 years

1.63 (1.07-2.49)

0.023

CURB-65 > 2 at admission

0.84 (0.47-1.50)

0.557

Persistent RNAemia

1.62 (1.01-2.61)

0.045

All-cause mortality at day + 90

Age older than 70 years

1.98 (1.24-3.17)

0.004

CURB -65 > 2 at admission

0.88 (0.48-1.60)

0.671

Persistent RNAemia

2.08 (1.20-3.60)

0.052

Table 1. Model of Cox regression analyses.

Conclusions: The results of this study suggest that RNAemia at the time of COVID-19 diagnosis and during the follow-up could be a useful independent biomarker allowing early patients’ stratification according to the expected disease evolution, and driving clinical decisions tailored on the specific needs of the individual patient.

Disclosure: Nothing to declare.

13: Infectious Complications

P454 COST-EFFECTIVENESS OF MARIBAVIR FOR POST-TRANSPLANT CYTOMEGALOVIRUS INFECTION THAT IS REFRACTORY TO ALTERNATIVE TREATMENTS FROM THREE HEALTHCARE SYSTEM PERSPECTIVES: ITALY, UK AND US

Stacey L. Amorosi1, Emtiyaz Chowdhury2, Riccardo Ressa2, Peter Cain3, Simone Corinti4, Bob G. Schultz5, Tien Bo 1

1Takeda Development Center Americas, Inc., Lexington, United States, 2Parexel, London, United Kingdom, 3Takeda UK, London, United Kingdom, 4Takeda Italia S.p.A, Rome, Italy, 5Takeda Pharmaceuticals U.S.A., Inc., Lexington, United States

Background: Maribavir has been demonstrated to be an effective and well tolerated treatment for patients with post-transplant refractory cytomegalovirus (CMV) infection; therefore, it is important to evaluate its economic value relative to alternative treatments to allow patient access to this novel therapy.

Methods: A two-stage economic model was developed to investigate the cost-effectiveness of maribavir versus alternative treatments (valganciclovir/ganciclovir, foscarnet, or cidofovir) for treatment of post-transplant refractory CMV infection, and adapted for the Italian, UK, and US healthcare systems. The first stage of the economic model involved capturing transitions between three health states over a period of 78 weeks. The three health states included clinically significant CMV (CMV DNA >lower limit of quantification [LLOQ] requiring treatment), non-clinically significant CMV (CMV DNA <LLOQ or CMV DNA >LLOQ not requiring treatment), and dead. From 78 weeks to a lifetime horizon (second stage of the economic model), patients moved to a two-state, alive/dead model structure with annual cycles. Transitions between health states were based primarily on data from the Phase 3 SOLSTICE trial (NCT02931539) augmented with data from observational studies, including real-world, multicenter, retrospective CMV outcomes studies involving European and US patients, and other published literature. Patients enrolled in SOLSTICE had a mean age of 53 years; 60% of patients had received solid organ transplant and 40% hematopoietic cell transplant prior to the trial. Health-related quality of life data from SOLSTICE were used to estimate health-state utilities and supplemented with data from a Takeda-led vignette study to inform transplant complications and other utilities. The adapted model in each country was validated with country-specific healthcare physicians and payors. Cost inputs were derived from relevant cost sources per country and payor system. Costs and outcomes were discounted by 3.5% in the UK model (as per the National Institute for Health and Care Excellence [NICE] reference case) and 3% in the Italian and US models.

Results: Maribavir incurred an incremental cost of €11,455 for an incremental quality-adjusted life year (QALY) gain of 0.313 in Italy, and £4,461 for an incremental QALY gain of 0.221 in the UK. It was cost saving (incremental saving of $8,197) for an incremental QALY gain of 0.21 in the US, resulting in a dominant incremental cost-effectiveness ratio (ICER) for maribavir compared with alternative treatments in the US (more efficacious and less costly, with a negative ICER). In Italy, the ICER of €36,626/QALY was below the willingness-to-pay threshold for an orphan drug, and in the UK the ICER of £20,163/QALY was within the NICE willingness-to-pay threshold range. Probabilistic sensitivity analyses indicated that maribavir was cost effective at the countries’ respective willingness-to-pay thresholds in 45% of iterations in Italy, 62% in the UK, and 100% in the US.

Conclusions: Maribavir has demonstrated superior CMV clearance among patients with post-transplant refractory CMV infection, a complication that increases morbidity and mortality risk. These economic analyses indicate that maribavir is also cost-effective, and in some cases cost-saving, across three different payor systems, Italy, UK, and US, compared with alternative treatments.

Disclosure: Stacey L. Amorosi: employee of Takeda Development Center Americas, Inc., and Takeda stockholder.

Emtiyaz Chowdhury: employee of Parexel, which received research funding from Takeda Development Center Americas, Inc., for this study.

Ricardo Ressa: employee of Parexel, which received research funding from Takeda Development Center Americas, Inc., for this study.

Peter Cain: employee of Takeda UK.

Simone Corinti: employee of Takeda Italia S.p.A.

Bob G. Schultz: employee of Takeda Pharmaceuticals USA, Inc. and Takeda stockholder.

Tien Bo: employee of Takeda Development Center Americas, Inc., and Takeda stockholders.

13: Infectious Complications

P455 INVASIVE FUNGAL INFECTIONS IN PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A 4-YEAR SINGLE INSTITUTION ANALYSIS WITH FOCUS ON PROPHYLAXIS

Federica Gironi1, Benedetta Elena Di Majo2,1, Ilaria Castelli1, Sonia Bonanomi1, Francesca Vendemini1, Giorgio Ottaviano1, Annalisa Cavallero1, Sergio Maria Malandrin1, Irene Maria Sciabica1, Guglielmo Marco Migliorino1, Sergio Foresti1, Marta Verna 1, Adriana Balduzzi2,1

1Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 2Università degli Studi di Milano-Bicocca, Milano, Italy

Background: Invasive fungal infections (IFIs) represent a serious complication after hematopoietic stem cell transplantation (HSCT) with a prevalence in literature from 1.2 up to 25% and a mortality rate of up to 80% in the most severe cases. In the immunocompromised patient IFIs often represent both a diagnostic and a therapeutic challenge, as clinical, radiological and microbiological characteristics are often non-specific. Duration of neutropenia after HSCT, the presence of severe acute graft versus host disease (aGVHD), a short time between a previous infection and HSCT and the use of a narrow-spectrum secondary antifungal prophylaxis agent are known risk factors for the development of IFIs.

Methods: 123 consecutive allogeneic hematopoietic stem cell transplantations performed between 2019 and 2022 in 113 patients at the Pediatric Hematopoietic Stem Cell Transplantation Unit at Fondazione IRCCS San Gerardo dei Tintori - Monza were retrospectively assessed. All patients had at least one year of follow-up by the end of October 2023. Clinical, radiological and microbiological data were collected to assess the prevalence and characteristics of IFIs occurring during the first year post-HSCT.

The primary aim of the study was to evaluate the effectiveness and possible toxicity of different types of antifungal prophylaxis to guide our local strategies. In particular, shifts from one prophylaxis to another were analyzed to assess possible antifungal-induced toxicity.

Results: The prevalence of pre-HSCT IFIs was 11/123 (9%), of which 1/13 (9%) presented a post-HSCT reactivation, while the prevalence of post-HSCT IFIs was 13/123 (10.6%). The mortality rate of IFIs was 23%.

In the first year after HSCT, an age >14 years (p=0.08), the use of rabbit antithymocyte globulins (ATG) during conditioning (p=0.2) and aGVHD after HSCT (p=0.2) were identified as possible risk factors for developing IFI.

IFIs diagnosis was challenging due to non-specific clinical and radiological presentation, scarce predictiveness of non-invasive microbiological tests during ongoing prophylaxis, and difficulties in performing invasive procedures in post-HSCT patients.

In our cohort, the initial prophylaxis defined according to patients’ risk was liposomal amphotericin B in 41%, fluconazole in 57% and isavuconazole in 2% of the patients, with a similar IFIs incidence with different types of drugs.

The shift rate from one prophylaxis to another was 71% for liposomal amphotericin B, 48% for fluconazole and 50% for isavuconazole (p=0.037). Liposomal amphotericin B was discontinued in 77% of the cases for nephrotoxicity (<0.001), with a mean diagnosis after 60±38 (2;167) days of therapy given at a dosage of 3 mg/kg for 2-3 times/week.

Conclusions: Our data were consistent with the literature in terms of IFIs incidence, mortality and associated risk factors. Different types of antifungal prophylaxes did not influence IFIs occurrence, but the administration of liposomal amphotericin B as prophylaxis was associated with a high incidence of nephrotoxicity during the second month after HSCT. This finding suggests that a potential benefit could be achieved by switching to azoles after the first month post-HSCT to prevent nephrotoxicity. This strategy has been yet implemented at our Centre since the beginning of 2023 and further analyses will establish its effectiveness in reducing toxicity.

Disclosure: Nothing to declare.

13: Infectious Complications

P456 ENHANCING SARS-COV-2 IMMUNITY EARLY AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION: A THREE-DOSE RBD–TT-CONJUGATED VACCINE APPROACH

Maryam Barkhordar 1, Mohammad Vaezi1, Leyla Sharifi Aliabadi1, Mohammad Ahmadvand1, Ardeshir Ghavamzadeh2

1Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran, Islamic Republic of, 2Cancer & Cell Therapy Research Center, Tehran University of Medical Sciences, Tehran, Iran, Islamic Republic of

Background: The pressing demand for timely SARS-CoV-2 immunization of hematopoietic stem cell transplant (HSCT) recipients in an endemic area presents numerous obstacles regarding selecting a vaccine platform appropriate for HSCT recipients being economical for widespread use in developing countries. Optimizing vaccine efficacy is of particular concern in patients undergoing HSCT, which mainly have an inadequate immune response to primary SARS-CoV-2 vaccination. This investigation aimed to explore the potential three-dose COVID-19 vaccination strategy following autologous (auto-) and allogeneic (allo-) HSCT with a recombinant receptor-binding domain (RBD)–tetanus toxoid (TT)-conjugated platform shortly after HSCT in the developing country of Iran.

Methods: This prospective study aimed to investigate the safety, immunogenicity, and predictive factors following a homologous prime-boost COVID-19 vaccination strategy including two primary doses of RBD–TT-conjugated SARS-CoV-2 vaccine followed by a homologous booster (third) dose administered at 4-week (± 1-week) intervals within 3–12 months post-HSCT. An immune status ratio (ISR) was measured at baseline and 4 weeks (± 1 week) after each vaccine dose using a semiquantitative immunoassay. Using the median ISR as a cut-off point for immune response intensity, we performed a logistic regression analysis to determine the predictive impact of several baseline factors on the intensity of the serologic response following the third vaccination dose. To evaluate the serologic response of this vaccine platform in healthy individuals, we used the results of 50 healthy volunteers (22 females, 28 males) with a mean (±SD) age of 37.92 years (±12.62) who had received two primary doses of the RBD–TT-conjugated SARS-CoV-2 vaccine with a similar protocol.

Results: A total of 66 patients including 36 allo-HSCT and 30 auto-HSCT recipients who completed the three-dose RBD-TT conjugated SARS-CoV-2 vaccine between 3–12 months after HSCT and had an available anti-spike serologic test at four predefined time points of baseline and after the first, second, and booster (third) doses were included in the analysis. The patients’ and transplant details are presented in Table 1. The ISR significantly increased during the three-dose regimen, reaching 3.87 (95% CI 3.25 to 4.48) with 91.66% seropositivity after the booster (third) dose in allo-HSCT and 3.42 (95% CI: 2.67- 4.17) with a 93.0% seropositivity after the booster dose in the auto-HSCT recipients. However, the ISR values at baseline and following the two primary doses of RBD–TT-conjugated SARS-CoV-2 vaccine were comparable for auto-HSCT and healthy participants and were considerably higher than allo-HSCT recipients. No serious adverse events (i.e., grades 3 and 4) were observed following the vaccination regimen in both allo-HSCT and auto-HSCT recipients.

Table 1

HSCT Type

Total

Auto-HSCT

Allo-HSCT

Number of Participants

66

30

36

Age in years, mean (±SD)

46.2 (±12.2)

50.9 (±11.1)

42.4 (11.8%)

Sex

Female

27 (40.9%)

12 (40%)

15 (42%)

Male

39 (59%)

18 (60%)

21 (58%)

Primary Disease

AML

-

27 (75%)

ALL

-

9 (25%)

Lymphoma

14 (47%)

-

MM

16 (53%)

-

Pre-HSCT COVID-19 Vaccination

Yes

38 (57%)

25 (83%)

13 (36%)

Pre-HSCT PCR-Positive COVID-19

Yes

32 (48%)

17 (57%)

15 (42%)

The time between HSCT and the start of vaccination in Days, median (Q1- Q3)

133 (106- 205)

136 (105- 177)

133 (107- 228)

  1. Data are presented as frequency (%), Auto-HSCT; Autologous Hematopoietic Stem Cell Transplantation, Allo-HSCT; Allogeneic Hematopoietic Stem Cell Transplantation, MM; Multiple Myeloma, SD; Standard Deviation.

Conclusions: We concluded that early vaccination of allo-HSCT recipients with a three-dose RBD–TT-conjugated SARS-CoV-2 vaccine is safe and could improve the early post-allo-HSCT immune response. In allogeneic patients, due to the immunosuppression drugs, the immune response to two initial doses is lower than in autologous patients and also in the normal population. But following the booster dose, this deficiency is compensated, and the immune response increases in a similar to autologous.

Clinical Trial Registry: The study was registered on ClinicalTrial.gov (NCT05185817) and Iranian Registry of Clinical Trials (IRCT20140818018842N23) and (IRCT20140818018842N22).

Disclosure: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

13: Infectious Complications

P457 USE OF LETERMOVIR IN PEDIATRIC POPULATION POST BONE MARROW TRANSPLANT

Cristina Rivera-Pérez1, Nuria Mas Malagarriga1, Silvia Simó Nebot1, Maria Trabazo del Castillo1, Gloria Miguel Llordes1, Laura Jimenez Prat1, Montserrat Rovira Tarrats1,2, Julia Marsal Ricoma 1, Cristina Rivera

1Hospital Sant Joan de Deu, Barcelona, Spain, 2Hospital Clínic Provincial, Barcelona, Spain

Background: Cytomegalovirus is an opportunistic virus that can cause life-threatening complications during hematopoietic stem cell transplantation (HSCT). Letermovir is an antiviral drug approved for the prophylaxis of CMV reactivation and disease in adult patients, but not yet approved in pediatrics. In our center, we have been using letermovir in at-risk patients as compassionate use since October 2021.

Methods: This is a retrospective observational study in which we analyzed the use of letermovir in 23 seropositive patients undergoing HSCT in Sant Joan de Deu hospital, Barcelona.

Results: We analyzed 23 patients with a mean age of 10.5 years (10 months - 17 years) included from October 2021 to December 2023. Twelve patients were male (52%). The reason for transplantation was: 9 B acute lymphoblastic leukemia, 2 T acute lymphoblastic leukemia, 1 undifferentiated leukemia, 1 T lymphoma, 1 myelodysplastic syndrome, 3 Fanconi anemias, 3 aplastic anemia and 2 immunodeficiencies. Nineteen patients received myeloablative conditioning. Bone marrow source was peripheral blood in 10 patients and bone marrow in 13 patients.

A total of 21 patients (91%) received T-lymphocyte depletion (6 patients ATG, 2 patients alemtuzumab, 12 patients post cyclophosphamide and one patient post cyclophosphamide and ATG). Nineteen patients received letermovir as primary prophylaxis (82%) and the median number of days of letermovir use was 140 days. Thirteen patients (56%) had CMV blips during the treatment without reactivation (defined as >1000 IU/mL). Only 4 patients had CMV reactivation (17%) and any patient developed CMV disease. The 240mg and 480mg commercial tablets were used, as well as 60mg and 120mg magistral formula tablets for patients with lower weights. No patient presented adverse effects attributable to the drug.

Conclusions: In our experience, this study demonstrates the safety of letermovir in the pediatric population of the drug in this population adjusted for weight for the prophylaxis of CMV. Letermovir was well tolerated in all cases. Further studies are needed.

Disclosure: No disclosure.

13: Infectious Complications

P458 REAL-WORLD OUTCOMES AND TREATMENT PATTERNS OF CMV INFECTIONS IN HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS, REFRACTORY OR INTOLERANT TO TREATMENTS IN EUROPE, CANADA, ISRAEL: INTERIM ANALYSIS

Johan Maertens 1, Matthew Pellan Cheng2, Avichai Shimoni3, Andreas Braun4, Nawal Bent-Ennakhil4, Irmgard Andresen4

1UZ Leuven, Leuven, Belgium, 2McGill University Health Centre, Montreal, Canada, 3Chaim Sheba Medical Center, Ramat Gan, Israel, 4Takeda Pharmaceuticals International AG, Zürich, Switzerland

Background: Contemporary real-world data on the management of cytomegalovirus (CMV) infection following allogeneic hematopoietic stem cell transplant (HSCT) are limited; therefore, the aim of this real-world study was to generate evidence on the burden of CMV infection and disease in HSCT recipients who had refractory CMV with or without resistance, or who were intolerant to current treatments (RRI).

Methods: This interim analysis of a multinational, non-interventional medical chart review pooled de-identified data from HSCT recipients (aged ≥18 years) with RRI CMV across 10 transplant centers in Austria, Belgium, Greece, Poland, Serbia, Canada and Israel; Patient data between 2016 and mid-2023 were analyzed descriptively. The index CMV episode was the first CMV episode in which the patient is considered RRI to anti-CMV therapy.

Results: Data from 31 patients (median age: 54.0 years; male: 61.3%; CMV seropositive recipients: 80.6%; acute leukemia as primary indication for HSCT: 61.3%) were analyzed. Initial CMV management approach after HSCT was pre-emptive monitoring (22 patients; 71.0%), prophylaxis (3 patients; 9.7%), or none (6 patients; 19.4%). All 3 patients with initial primary prophylaxis received letermovir. Overall, 7 (22.6%) patients received a prophylactic (either primary or secondary) anti-CMV agent. At the time of index CMV date (RRI identification), 26 (83.9%) patients were identified as refractory with or without resistance, and 5 (16.1%) patients as intolerant only. A total of 24 (77.4%) patients had pre-emptively treated index CMV episodes. Overall median (Q1–Q3) time from HSCT until index CMV episode was 43.0 (29.0–104.0) days. Overall median (Q1–Q3) duration of index CMV episode was 42.5 (32.5–60.0) days. For treatment of index CMV episodes, 3 (9.7%), 28 (90.3%), 13 (41.9%), and 3 (9.7%) patients received 1, ≥2, ≥3, or 4 therapies, respectively. Overall, 26 (83.9%) patients used ganciclovir, 24 (77.4%) used foscarnet, 18 (58.1%) used valganciclovir, 4 (12.9%) used foscarnet/ganciclovir combination, and 2 (6.5%) used letermovir or other dual therapy. A total of 11 (35.5%) patients did not achieve viremia clearance during the index CMV episode: 5/6 (83.3%) and 6/25 (24.0%) patients with symptomatic or asymptomatic episodes, respectively. Overall, 11 (35.5%) patients experienced a myelosuppression event during index CMV episodes; thrombocytopenia and neutropenia were the most common types, each occurring in a total of 6 (19.4%) patients. Of patients with myelosuppression, 6 (54.5%), 3 (27.3%), and 2 (18.2%) were receiving ganciclovir, foscarnet, or valganciclovir, respectively. Overall, 8 (25.8%) patients experienced a nephrotoxicity event during index CMV episodes; these patients were receiving foscarnet (5 [62.5%] patients), ganciclovir (1 [12.5%] patient), valganciclovir (1 [12.5%] patient), or dual therapy (1 [12.5%] patient).

Conclusions: This interim analysis highlights the real-world burden of CMV infection/disease for HSCT recipients and illustrates limitations of current standard of care for managing RRI CMV and the need for improved treatments. A notable proportion of patients experienced adverse events of myelosuppression and nephrotoxicity; these results are consistent with those from prior studies in this patient population.

Clinical Trial Registry: N/A.

Disclosure: Funding: This study was sponsored by Takeda Pharmaceuticals International AG.

Johan Maertens: Consulting fees from Shire/Takeda, as well as consulting fees and non-financial support from Amgen, Astellas Pharma, Basilea, Cidara, F2G, Schering-Plough, and Scynexis; and grants, consulting fees, and non-financial support from Bio-Rad, Gilead Sciences, Merck, and Pfizer outside the submitted work; and honoraria from Astellas, F2G, Gilead, Pfizer, Merck Sharp & Dohme, and Mundipharma.

Matthew P. Cheng: Grants from the Canadian Institutes of Health Research during the conduct of the study and is supported by the Fonds de Recherche du Québec – Santé. Personal fees from GEn1E Lifesciences and Nomic Bio as a member of the scientific advisory board, as well as honoraria from AstraZeneca, Takeda, Merck, and Pfizer. Research support from Cidara Therapeutics, Scynexis, and Amplyx Pharmaceuticals during the conduct of the study, but outside the submitted work Dr. Cheng is the co-founder of Kanvas Biosciences, Inc. and owns equity in the company. Pending patents: i) Methods for detecting tissue damage, graft versus host disease, and infections using cell-free DNA profiling; ii) Methods for assessing the severity and progression of SARS-CoV-2 infections using cell-free DNA.

Avichai Shimoni: No relevant disclosures (Nothing to declare).

Andreas Braun: Employee of Takeda and owns Takeda stocks.

Nawal Bent-Ennakhil: Employee of Takeda and owns Takeda stocks.

Irmgard Andresen: Employee of Takeda and owns Takeda stocks.

13: Infectious Complications

P459 THE RESPECT STUDY DESIGN: COMPARISON OF ONCE-WEEKLY REZAFUNGIN AGAINST STANDARD ANTIMICROBIAL PROPHYLAXIS IN ADULTS UNDERGOING ALLOGENEIC BLOOD AND MARROW TRANSPLANTATION

Johan Maertens 1,2, Federica Sora3,4, Drew J. Winston5, Shariq Haider6, Lourdes Vazquez Lopez7, Christine Robin8, Alexander Schauwvlieghe9, Dominik Selleslag9, Taylor Sandison10, Kieren A. Marr11,12,13

1KU Leuven, Leuven, Belgium, 2University Hospitals Leuven, Leuven, Belgium, 3Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy, 4Università Cattolica del Sacro Cuore, Rome, Italy, 5University of California Los Angeles Medical Center, Los Angeles, CA,, United States, 6Juravinski Hospital and Cancer Centre, McMaster University, Hamilton, ON, Canada, 7Hospital Clínico Universitario Salamanca, Salamanca, Spain, 8Henri Mondor Hospital, Creteil, France, 9AZ Sint-Jan Brugge-Oostende AV, Bruges, Belgium, 10Cidara Therapeutics, Inc., San Diego, CA, United States, 11Johns Hopkins University School of Medicine, Baltimore, Maryland, United States, 12Pearl Diagnostics, Baltimore, Maryland, United States, 13Sfunga Therapeutics, New York, United States

Background: Current guidelines for first-line antifungal prophylaxis in haematopoietic stem cell transplantation (HSCT) recipients recommend either fluconazole or voriconazole/posaconazole, with posaconazole recommended for patients who develop graft versus host disease (GVHD) and co-trimoxazole (trimethoprim/sulfamethoxazole) for Pneumocystis jirovecii pneumonia (PCP) (Maertens JA, et al. J Antimicrob Chemother. 2018;73(12):3221-3230; Maertens J, et al. J Antimicrob Chemother. 2016;71(9):2397-404; Dadwal SS, et al; Transplant Cell Ther. 2021;27(3):201-211). Resistance, safety and tolerability issues and drug interactions are key concerns for antifungal prophylaxis in this patient group. The ongoing ReSPECT study aims to evaluate the efficacy and safety of once-weekly intravenous (IV) rezafungin, a next-generation echinocandin in Phase 3 development, in the prevention of invasive fungal infection caused by Aspergillus, Candida or Pneumocystis in adults undergoing allogeneic HSCT, compared with standard antifungal prophylaxis.

Methods: ReSPECT is a Phase 3, international, prospective, multicentre, randomised, double-blind trial examining antimicrobial therapy in adults (aged ≥18 years) receiving human leukocyte antigen (HLA)-matched allogeneic HSCT from a family or unrelated donor, HLA-mismatched related/unrelated donor, or haploidentical donor. Subjects included to date were randomised 2:1 at HSCT to receive either rezafungin once-weekly IV infusion (Day 0: 400 mg; Day 7 onward: 200 mg) or standard prophylaxis comprising oral fluconazole (400 mg daily from Day 0) and trimethoprim/sulfamethoxazole (80 mg/400mg; daily from Day 30) through Day 90. Fluconazole may be switched to posaconazole (300 mg twice daily on the first day and 300 mg daily thereafter) in the event of acute GVHD. The primary efficacy endpoint is fungal-free survival (FFS) at Day 90. Key secondary endpoints will include discontinuation due to toxicity/intolerance, FFS in subjects with/without GVHD at Day 90 and time to invasive fungal disease or death. Overall mortality and mortality attributable to invasive fungal disease will be assessed, and safety endpoints will examine adverse events.

Results: To date (December 2023), 363 patients have been screened and 307 enrolled in the ReSPECT study from treatment centres in Belgium, Canada, France, Germany, Italy, Spain, Switzerland, the United Kingdom and the United States (Table 1). The current target for randomisation is 462 patients.

Table 1: ReSPECT trial recruitment to date (December 2023)

Full size table

.

Conclusions: The ReSPECT trial has recruited more than 50% of the planned enrolment. Site recruitment and enrolment is intended to continue. The primary endpoint of FFS at the Day 90 time point aims to assess efficacy during the acute phase of allogeneic HSCT and to minimise the potential for bias due to patient drop-out in this setting. The trial will be the first to evaluate the efficacy of an echinocandin in the prevention of Candida, Aspergillus and Pneumocystis infection in patients undergoing allogeneic HSCT.

Clinical Trial Registry: NCT04368559.

Disclosure: Johan Maertens: consulting fees from Amplyx, Cidara, Gilead, Pfizer, Scynexis, F2G, Mundipharma, Takeda, and Basilea; honoraria for lectures from Astellas, Gilead, Basilea, Mundipharma, Takeda, Medscape, Pfizer, and Shionogi; participation on Advisory Board from Cidara, Pulmocide, Shionogi, Basilea, Sfunga and Mundipharma.

Federica Sora: No relevant disclosures.

Drew J Winston: Funding from Cidara.

Shariq Haider: Research Funding from Avir Pharmaceuticals.

Lourdes Vazquez Lopez: No relevant disclosures.

Christine Robin: Mundipharma, Gilead, MSD.

Alexander Schauwvlieghe: No relevant disclosures.

Dominik Selleslag: No relevant disclosures.

Taylor Sandison: Employee at Cidara Therapeutics Inc.

Kieren Marr: Consultant, Cidara; Employee and Equity Holder, Sfunga, Equity Pearl Diagnostics.

13: Infectious Complications

P460 NO INFERIOR OUTCOME OF OMITTING FLUOROQUINOLONE PROPHYLAXIS IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Vanessa Nehrbaß 1, Krischan Braitsch1, Katharina Nickel1, Maike Hefter1, Katrin Koch1, Kathrin Rothe1, Florian Bassermann1, Jochen Schneider1, Katharina S. Götze1, Peter Herhaus1, Mareike Verbeek1

1Technical University of Munich, Munich, Germany

Background: Infectious complications remain one of the main causes of morbidity after allogeneic hematopoietic stem cell transplantation (alloHSCT). Prophylactic strategies in patients with severe neutropenia include various drugs including fluoroquinolones (FC), which are thought to prevent bacteremia by decreasing the prevalence of oral and intestinal pathogens, as these are common sites for bacterial translocations during neutropenia after mucosal barrier injury. In April 2019 a direct healthcare professional communication warned about FC side effects and concerns about promoting antibiotic resistance and clostridium difficile infections (CDI) arose. Further, early microbiota disruption during alloHSCT is linked to increased mortality and GvHD-incidence. In May 2019, these data ultimately led to a discontinuation of antibiotic prophylaxis in patients undergoing alloHSCT at our center. The aim of this retrospective study is to analyze the effect of omitting FC prophylaxis. We hypothesize non-inferiority with regard to incidence of bloodstream infections (BSI), GvHD-rates and overall survival (OS).

Methods: A total of 143 patients who underwent first alloHSCT between 2017 and 2022 at our center were included. Of these, 73 patients received FC prophylaxis, and 69 did not (non-FC). Patients were considered to be treated with antibiotic prophylaxis if ciprofloxacin was administered with 500mg BID from the day of admission to the first day of fever. Univariate comparisons between both groups were performed using Chi-square test and Fisher’s test.

Results: The median age at transplantation was 58 years (18-73) in the FC group and 53 years (19-72) in the non-FC group. Gender distribution was balanced in both groups. The majority of patients received a peripheral stem cell graft from a matched unrelated donor (MUD, 62% vs 52%), followed by MRD (16% vs. 17%) and haploidentical donors (15% vs. 15%). AML was the most common disease with 25 AML patients in the FC group and 32 in the no-prophylaxis group. Average duration of ciprofloxacin prophylaxis was 8 days (3-38) in the FC group (see Table 1). BSI were common but did not differ in between the groups (38 vs 35 cases, FC group vs non-FC group; p=0.87). CDI cases were rare in both cohorts but tend to be more frequent in the FC group (5 vs. 1; p=0.11). There was a trend towards higher rates of grade I/II aGvHD in the non-FC group (38 vs. 47 cases, FC group vs. non-FC group; p=0.06). Grade III/IV aGvHD rates were comparable among the groups (12 vs. 12 cases in each group; p=0.88). No differences in rates of cGvHD between the two groups were seen (15 vs 14 cases of mild-moderate cGvHD (p=0.97) and 3 vs. 2 cases of severe cGvHD (p=0.70), FC vs. non-FC group). In the univariate analysis, OS after one year was significantly higher in the non-FC group (p=0.014). Early transplant mortality was low in both groups with a 2-month OS of 93% and 96% respectively.

Table 1. Patient characteristics

FC group

non-FC group

number of patients

73

69

age

median (years, range)

58 (18-73)

53 (19-72)

sex

male (n, %)

45 (63%)

42 (61%)

female (n, %)

28 (38%)

27 (39%)

disease type

AML (n, %)

25 (34%)

32 (46%)

ALL (n, %)

7 (10%)

7 (10%)

MPN (n,%)

6 (7%)

8 (12%)

MDS (n, %)

9 (12%)

7 (10%)

Multiple Myeloma (n, %)

8 (11%)

5 (7%)

BNHL (n, %)

7 (10%)

1 (2%)

TNHL (n, %)

2 (3%)

4 (6%)

AA (n, %)

2 (3%)

0 (0%)

Other (n, %)

7 (10%)

5 (7%)

donor type

MRD (n, %)

12 (16%)

12 (17%)

MUD (n, %)

45 (62%)

36 (52%)

MMUD (n, %)

5 (7%)

11 (16%)

Haplo (n, %)

11 (15%)

10 (15%)

sex of donor

male (n, %)

56 (77%)

50 (72%)

female (n, %)

17 (23%)

19 (28%)

stem cell source

PB (n, %)

67 (92%)

65 (94%)

BM (n, %)

5 (7%)

1 (2%)

PB + BM (n, %)

1 (1%)

-

NA (n, %)

-

3 (4%)

duration of ciprofloxacin prophylaxis

median (days, range)

8 (3-38)

-

Conclusions: Omitting FC prophylaxis did not have a negative impact on OS, BSI or CDI. Superior OS is most likely explained by confounding factors such as the new approval of drugs – for example letermovir.

Disclosure: Nothing to declare.

13: Infectious Complications

P461 OUTCOMES, CHOICE OF ANTIBIOTICS AND THEIR EFFECTIVENESS IN ALLOGENEIC-HSCT RECIPIENTS COLONIZED WITH MULTIRESISTANT GRAM-NEGATIVE BACTERIA: A SINGLE-CENTER RETROSPECTIVE ANALYSIS

Igor Age Kos 1, Jana Speer2, Onur Cetin1, Vadim Lesan1, Angelika Bick1, Konstantinos Christofyllakis1, Manfred Ahlgrimm1, Sigrun Smola3, Andreas Link1, Torben Rixecker1, Anna K. H. Hirsch4,5, Joerg Thomas Bittenbring1, Fabian Berger3, Sören Becker6, Lorenz Thurner1, Moritz Bewarder1

1University of Saarland, Homburg, Germany, 2José Carreras Centrum, University of Saarland, Homburg, Germany, 3Institute for Virology, University of Saarland, Homburg, Germany, 4Helmholtz Institute for Pharmaceutical Research Saarland, (HIPS)–Helmholtz Centre for Infection Research (HZI); Germany, Saarbrücken, Germany, 5Saarland University, Saarbrücken, Germany, 6Institute of Medical Microbiology and Hygiene, University of Saarland, Homburg, Germany

Background: Multiresistant gram-negative bacteria (MRGN) and vancomycin resistant enterococci (VRE) commonly colonize patients with prolonged hospitalization. This is also often observed in patients undergoing allogeneic hematopoietic stem cell transplantation (allo-HSCT), in which its clinical significance may increase due to prolonged immunosuppression.

Methods: Retrospective, single-center study. The primary endpoint was 1-year non-relapse mortality (NRM). Patients were stratified according to the resistance profile of colonizing bacteria as follows: 3-MRGN if resistant to three out of four antibiotic classes (acylureidopenicillines, cephalosporins of the 3rd and/or 4th generation, carbapenems, and fluoroquinolones); 4-MRGN if resistant to all 4 classes. VRE colonization was also included in the survival analysis. Patients undergoing allo-HSCT without any colonization served as controls. Patients with MRGN colonization diagnosed after one year of transplantation were excluded from the analysis. Survival analysis was performed with the Kaplan–Meier method. Continuous variables were compared using Student’s t-test or Mann–Whitney’s test. The odds ratio was calculated using Fisher’s exact test. Multivariable analysis for survival was calculated using Cox proportional hazards test.

Table 1. Use of antibiotics according to colonization group

4 MRGN

3 MRGN

Control group

Number of patients

21

24

54*

Receiving antibiotics°, n

21 (100%)

23 (95,8%)

37(68,5%)

Duration of Antibiotic use°, days

28,6

21,3

16,5

Number of Antibiotics°

Median (range)

3 (4)

2 (3)

1 (4)

Firstline Piperacillin/Tazobactam, n

18 (85,7%)

19 (79,2%)

33 (89,1%)

Effectiveness of first-line therapy*, n

3 (14,2%)

4 (17,3%)

14 (37,8%)

If Piperacillin/Tazobactam first line, n

2 (11%)

3 (15,7%)

10 (30%)

If first therapy other, n

1 (33%)

1 (25%)

4 (100%)

Time to escalation, median days

5,2d

3,6d

9,1d

Second-line Meropenem, n

15 (83,3%)

16 (84,2%)

23 (74,2%)

Effectiveness, n

1 (6,7%)

13 (81,3%)

19 (82,6%)

Third-Line or more

17 (81%)

3 (12,5%)

8 (14%)

Ceftazidim/Avibactam

11 (64,7%)

1 (33,3%)

1 (12,5%)

Tigecyclin

11 (64,7%)

3 (100%)

7 (87,5%)

Aminoglycoside

5 (29,4%)

1(33,3%)

Colistin

5 (29,4%)

Penicillin Allergy

2 (9,5%)

1 (4,2%)

4 (7%)

Bacteremia

13 (62%)

13 (54,2%)

15(26,3%)

MRG responsible for the bacteremia

6 (28,6%)

4 (16,7%)

-

  1. °Antibiotics including: Penicillins, Cephalosporine, Carbapenem, Tigecyclin, Ceftazidime/Avibactam, Colistin and Cefiderocol. (Vancomycin, Linezolid and Daptomycine not included in the analysis)
  2. *Percentages referring to 54 patients instead of 57 in total as data on antibiotics was not available for 3 patients in the control group

Results: In total 166 patients were included in the analysis (table 1). 21 patients with 4MRGN, 24 with 3MRGN, 60 with VRE and 57 controls. Patients colonized with 4-MRGN had significantly higher 1-year-NRM compared to VRE-colonized and controls. 3-MRGN-colonized patients presented higher 1-year-NRM compared to VRE and controls. A Cox regression showed that colonization with 3 or 4-MRGN was the only independent risk factor for 1-year NRM in this cohort (p = 0.005). These results also translated into significant 1-year overall survival differences. Of note, acute myeloid leukemia was more frequent amongst 4-MRGN-colonized patients (60% vs 15% for controls). Colonized patients (3-MRGN + 4-MRGN) had significantly longer hospitalization during allo-HSCT (median of 41 vs 33 days, p = 0.008), longer duration of antibiotic treatment (median of 20 vs 9 days, p = 0.01), higher number of antibiotic lines of therapy (median of 2 vs 1 p < 0.001 - of note, only broad-spectrum antibiotics were included in the analysis), as well as higher frequency of bacteremia (57.7% vs 26.3% p = 0.02). The colonizing bacteria were responsible for the bacteremia in 28% of cases for 4-MRGN and 16.7% for 3-MRGN colonized patients. Piperacillin/tazobactam was the first-line therapy in 86% of patients colonized with 4 MRGN, 79.2% of 3-MRGN, and 89.1% of controls. The effectiveness of first-line therapy, defined as not followed by second-line therapy and surviving patient, was significantly lower in colonized patients (OR = 0,31; p = 0,04). We found no significant differences regarding engraftment times and aGVHD rates between colonized and non-colonized patients.

Conclusions: Patients undergoing allo-HSCT colonized with MRGN presented higher mortality and morbidity. The lower probability of resolution of infection after first-line treatment amongst this group (in our cohort most frequently piperacillin/tazobactam) suggests the need for broader coverage of the initial antibiotic treatment. Besides MRGN-adapted antibiotic strategies in case of infection, prevention of MRGN colonization during stem cell transplantation and induction therapy lines may also be important.

Disclosure: Authors declare no significant conflicts of interest regarding this abstract.

13: Infectious Complications

P462 LETERMOVIR AS TREATMENT AND SECONDARY PROPHYLAXIS OF CMV REACTIVATION IN LOW-WEIGHT PEDIATRIC PATIENTS

Chiara Mainardi 1, Cecilia Liberati1, Marica De Pieri1, Antonio Marzollo1, Manuela Tumino1, Maria Gabelli1, Marta Pillon1, Elisabetta Calore1, Alessandra Biffi1,2

1Padova University Hospital, Padova, Italy, 2Padova University, Padova, Italy

Background: Letermovir is approved for primary prophylaxis of cytomegalovirus (CMV) infection in adult CMV-seropositive recipients of matched unrelated donor (MUD) hematopoietic stem cell transplantation (HSCT). Emerging data from real-life experience and observational studies support the potential efficacy of Letermovir for the secondary prophylaxis or treatment of CMV disease in adult HSCT recipients, while data on pediatric population are lacking. Letermovir use in CMV refractory infection is questioned since it might induce viral mutations causing resistance. We report here two cases of successful Letermovir use in low-weight pediatric patients for asymptomatic CMV reactivation and CMV secondary prophylaxis after HSCT.

Methods: Clinical data were reviewed from patient data records. Parents consented to data collection.

Results: Patients’ and HSCT characteristics are reported in table 1.#1A 3-month-old female underwent umbilical cord blood (UCB) HSCT for Krabbe disease. On day +10 she developed CMV- asymptomatic reactivation (CMV-DNA >1000 copies/mL). Antiviral therapy with Foscarnet was started, CMV viral load (VL) peaked at 289.650 copies/ml on day +14, followed by a slow reduction without negativization albeit foscarnet escalation to maximum dose. On day +25, with a VL of 64070 copies/mL i.v. Letermovir (4 mg/kg/day) was associated to treatment, due to initial signs of Foscarnet renal toxicity and contraindication to Ganciclovir, because of ongoing engraftment. Such antiviral combination led to stable CMV-DNA negativity from day +39. Foscarnet was stopped on day +52, and secondary prophylaxis with Letermovir was continued (swithed to oral at discharge) until day +200 without further episodes of CMV reactivation.

#2.

A 13-month-old female received MUD HSCT for primary immunodeficiency (PID). Immediate post-HSCT course was uneventful and neutrophil[CL1] engraftment occurred on day +19. On day +35 she developed CMV-reactivation (low grade fever, CMV viraemia of 1881 copies/mL, no organ disease), and was started on Foscarnet. CMV-DNA viral load peaked to 2540 copies/mL on day +44 and persisted positive despite maximum Foscarnet dose-escalation, reaching negativity on day +50. Since the patient showed signs of renal toxicity, i.v. Letermovir 4mg/kg/die was added to antiviral treatment on day +53, Foscarnet was suspended on day +56. No further episodes of CMV reactivation were seen during secondary prophylaxis with Letermovir, which was well tolerated without any sign of toxicity.

#1

#2

Diagnosis

IEM (Krabbe disease)

PID

Age at HSCT (months)

3

13

Weight (kg)

6

9

Donor (stem cell source)

UCB

MUD (PBSC)

cell dose

TNC 9.2 x 10^7/kg

TNC 7.9 x 10^8/kg

CD34 + 2.32 x 10^6/kg

CD34 + 9.96 x 10^6/kg

Engraftment

ANC day +31

ANC day +19

PTL day +52

PTL day +19

Conclusions: The two reported patients experienced early CMV-reactivation with need for prolonged antiviral treatment with high dose Foscarnet to achieve viraemia control. Due to the well-known toxicity of such drug, a switch to a better tolerated treatment was needed. The use of Letermovir once virological drop was achieved, showed to be safe and effective in inducing and maintaining CMV negativity, preventing further reactivation at the last available follow-up.

Disclosure: Nothing to declare.

13: Infectious Complications

P463 COMPARISON OF BLOOD STREAMING INFECTIONS BETWEEN TUNNELED CENTRAL VENOUS CATHETERS AND NON-TUNNELED CENTRAL VENOUS CATHETERS IN ACUTE LEUKEMIA REMISSION INDUCTION – A SINGLE CENTER EXPERIENCE

Cheongin Yang 1, Seong Hyun Jeong1, Joon Seong Park1

1Ajou University Hospital, Suwon, Korea, Republic of

Background: In treating acute leukemia, reliable central vascular access is very important not only for administration of chemotherapy but also for conservative treatments. However, there is no clear evidence that tunneled central venous catheter (CVC) is superior among central venous catheters. We analyzed whether there was a significant difference in blood streaming infection depending on whether tunneled or not during acute leukemia induction.

Methods: A retrospective analysis was conducted on 114 patients diagnosed with acute leukemia at Ajou University Hospital from January 2020 to May 2023. Only patients undergoing intensive induction chemotherapy were included, of which 26 patients underwent tunneled CVC and 88 patients underwent non-tunneled CVC.

Results: In each group, blood streaming infections were reported in blood culture in 34.6% (n=9) of tunneled CVC and 30.7% (n=27) of non-tunneled CVC. In the tunneled CVC group, gram-positive bacteria were reported in 30.8% (n = 8) and gram-negative bacteria in 15.4% (n = 4), while in the non-tunneled CVC group, gram-positive bacteria were reported in 23.9% (n = 21) and gram-negative bacteria were reported in 9.1% (n = 4). The most common gram-positive bacteria in both groups was enterococcus faecium (tunneled group - 15.4%, n=4, non-tunneled group - 12.5%, n=11), and the gram-negative bacteria was escheria coli (tunneled group - 7.7%, n=2, non-tunneled group - 3.4%, n= 3). Blood streaming infections tended to occur more frequently in the tunneled CVC group, regardless of whether they were Gram-positive or Gram-negative. However, the results of the chi-square test did not show significance in the frequency of bacteremia by group (p=0.875) or in the frequency of gram-positive bacteremia (p=0.557). There was only one case in the tunneled CVC group where infection occurred at the catheter insertion site and had to be removed, and other than this, there were no side effects related to catheter insertion in both groups.

Conclusions: Rather, there was no statistically significant difference in the tunneled CVC group, but blood streaming infection tended to be more prevalent. There was one case in the tunneled CVC group that had problems after insertion of the catheter and had it removed regardless of bacteremia. If each catheter insertion can be done safely under image guidance, non-tunneled CVC can be a good alternative to tunneled CVC.

Disclosure: Nothing to declare.

13: Infectious Complications

P464 INCIDENCE OF INFECTIONS AND QUALITY OF IMMUNE RECONSTITUTION FOLLOWING CRYOPRESERVATION OF PERIPHERAL BLOOD STEM CELLS FROM HLA-IDENTICAL DONOR

Luca Barabino1, Stefania Bregante2, Massimiliano Gambella 2, Anna Ghiso2, Livia Giannoni2, Silvia Lucchetti2, Alberto Serio2, Riccardo Varaldo2, Antonella Laudisi2, Monica Passannante2, Roberta Murru3, Andrea Galitzia1, Alessandra Bo2, Emanuele Angelucci2, Giovanni Caocci1, Anna Maria Raiola2

1University of Cagliari, Cagliari, Italy, 2IRCCS Ospedale Policlinico San Martino, Genoa, Italy, 3Hematology and Stem Cell Transplantation Unit, Ospedale Oncologico A. Businco, ARNAS “G. Brotzu, Cagliari, Italy

Background: The COVID-19 pandemic raised the utilization of cryopreserved peripheral blood stem cells in allogeneic Hematopoietic Stem Cell Transplantation (HSCT), but limited data on infections and immune reconstitution using cryopreserved grafts are available.

Methods: We performed a comparative analysis of 46 patients who underwent an allo-HSCT with cryopreserved (CRYO) and 39 patients with fresh (FRESH) apheresis. The analysis was retrospective for 61 patients transplanted from January 1, 2016 to May 31, 2022 and prospective for 24 patients transplanted from June 1, 2022 to May 30, 2023. The Local Ethics Committee approved the protocol (346/2022). All patients had an HLA-matched sibling or unrelated donor. GVHD prophylaxis consisted of thymoglobulin, cyclosporine, and methotrexate. Patient characteristics are summarized in Table 1.

Among the objectives of this analysis, there were Infection Density (ID) and time and quality of immune reconstitution. ID represents the number of infections over time for patients experiencing recurrent infections. The assessment of bacterial, viral, and fungal ID was carried out across various time intervals (Days 0-100, Days 100-180, Days 181-365, Day 0-1.5 years, Day 0-2 years, Engraftment-1.5 years, Engraftment-2 years).

We further compare serum IgG, IgA, and IgM levels on days +30, + 60, and +100 and lymphocyte immunophenotyping of Peripheral Blood (PB) and Bone Marrow (BM) on days +30, + 60, + 100, and +180 in the CRYO and FRESH groups.

Table 1

CRYO

(n = 46)

FRESH

(n = 39)

p – value

AGE at transplant, mean (DS)

54.45 (±14.8)

55.39 (± 14.19)

0. 802

Sex, n (%)

• Male

25 (54.3)

28 (71.7)

0.15

• Female

21 (45.6)

11 (28.2)

Sorror Score

• 0

15 (32.6)

12 (30.7)

0.25

• 1-2

16 (34.7)

19 (48.7)

• >3

15 (32.6)

7 (17.9)

• Not assessable

-

1 (2.56

Disease, n (%)*

• ALL

3 (6.5)

3 (7.7)

0.301

• AML

21 (45.6)

20 (51.3)

• HL

4 (8.7)

-

• LLC

1 (2.1)

-

• MDS

5 (10.8)

9 (23)

• IMF

5 (10.8)

1 (2.6)

• MM

3 (6.5)

3 (7.7)

• NHL

4 (8.7

3 (7.7)

Disease Status, n (%) **

• CR

17 (37)

20 (51.3)

0.08

• R/R

20 (43.5)

8 (20.5)

• Active disease

9 (19.5

11 (28.2)

Incomplete Hematologic Recovery, n(%)

• NO iCR

31 (67.4)

27 (69.2)

> 0.99

• Yes iCR

15 (32.6)

12 (30.7)

Donor Age, mean (DS)

33.24 (± 11.84)

36,97 (±14,76)

0.359

Donor

• HLA identical Donor

9 (19.5)

20 (51.3)

0.028

• Matched Unrelated Donor

37 (80.5)

19 (48.7)

ABO, n (%)

• Major incompatibility

28 (60.8)

16 (41)

0.54

Donor/Recipient Sex Mismatch, n (%)

4 (8,6)

9 (23)

0.078

Condition regimen, n (%)

• Myeloablative

13 (28.2)

17 (43.6)

0.256

• Reduce Intensity

27 (58.7)

20 (51.2)

• Non Myeloablative

6 (13)

2 (5.12

Patient/Donor CMV status, n (%)

• -/-

5 (10.8)

4 (10.2)

0.722

• -/+

17 (36.9)

11 (28.2)

• +/+

19 (41.3)

21 (53.8)

• +/-

5 (10.8)

3 (7.7)

Splenomegaly at transplant (> = 13 cm), n (%)

• NO splenomegaly

33 (71.8)

35 (89.7)

0.06

• YES splenomegaly

13 (28.2)

4 (10.25)

• Mean diameter ± DS

16.45 ± 4.03

16.25 ± 2.62

WBC /Kg infused mean ± DS

8.12 ± 2.94

7.11 ± 2.88

0.67

CD34 /Kg infused mean ± DS

6.33 ± 2.32

5.47 ± 2.25

0.018

CD3 /Kg infused mean ± DS

256.82 ± 94.11

230.88 ± 98.36

0.136

Days to neutrophil recovery (500/mmc), n (%) mean ± DS

17.89 (± 4.3)

15.82 (± 2.25)

0.004

• NO take

1

1

Days to platelet recovery (20000/mmc), n (%) mean ± DS

30.27 (± 36.4)

15.0 (± 3.58)

<0.001

• NO take

1

1

  1. ALL = acute lymphoid leukemia; AML = acute myeloid leukemia; HL = Hodgkin Lymphoma; LLC = Chronic lymphocytic leukemia; MDS = Myelodysplastic syndrome; IMF = Idiopathic myelofibrosis; MM = Multiple Myeloma; NHL = non-Hodgkin Lymphoma; ** CR = complete remission; R/R = relapsed/refractory

Results: Clinical characteristics of patients, donors, and transplantations were outlined in Table 1. Neutrophil recovery time was longer in CRYO group (17.9 days vs FRESH 15.8 days, p<0.001).

Bacterial ID was similar in the CRYO and FRESH groups (0.32 vs 0,25, p=0.15) from Day 0 to 2 years post-transplant, but higher from engraftment to 2 years in the CRYO group (0.2 vs FRESH 0.09, p=0.03). Viral ID was significantly higher in the CRYO group within the first 1.5 years after transplantation (0.29 vs 0.17, p=0.02). Higher fungal ID was observed within the CRYO group, both in the initial 100 days (0.17 vs 0.026, p=0.037) and from Day 0 throughout the first 1.5 years (0.068 vs 0.012, p=0.01).

There was no difference in bacterial, viral, and fungal infection severity between the two groups (p=0.3). Infections were the primary cause of death for 5 of 13 deceased patients in the CRYO group and 4 of 13 in the FRESH group (p=0.532).

CRYO group demonstrated statistically lower immunoglobulin levels at days +60 and +100 (e.g. mean IgG+100 4.75 g/L vs FRESH 6.63 g/L, p=0.003), while no differences were noted in the PB and BM lymphocyte immunophenotyping.

Conclusions: Our research shows a delayed neutrophil engraftment, coupled with a postponed recovery of immunoglobulin levels in cryopreserved HSCT. The PB and BM lymphocyte immunophenotyping exhibited no differences between CRYO and FRESH groups. We observed increased bacterial ID in the CRYO group, which was not linked to delayed engraftment. Furthermore, viral and fungal ID was higher in the CRYO group according to the delayed engraftment and immunoglobulins recovery. Ongoing assessments of diverse immune reconstitution kinetics and infection susceptibility remain essential when utilizing cryopreserved grafts.

Disclosure: Nothing to declare.

13: Infectious Complications

P465 DETECTION OF RESPIRATORY VIRUSES AMONG PAEDIATRIC HSCT RECIPIENTS – A TEN YEARS SINGLE-CENTRE EXPERIENCE

Petr Hubacek 1,2, Petr Riha2, Ales Briksi2, Petra Keslova2, Renata Formankova1,2, Daniela Janeckova2, Miroslav Zajac2, Zdenek Kepka2, Petr Sedlacek1,2

12nd Medical Faculty of Charles University, Prague, Czech Republic, 2Motol University Hospital, Prague, Czech Republic

Background: Respiratory viruses remain an important cause of morbidity and mortality in the immunocompromissed host. However before the pandemics of SARS-CoV-2, only influenza virus attracted ussually more attention. Recently, especially due to better and easily assessible detection same as due to the possibility of virostatic treatment, respiratory viruses are more frequently identified as cause of the clinical problems. Our aim was to analyse the results of wide detection of respiratory viruses in symptomatic paediatric HSCT recipients.

Methods: Between I/2012 and XII/2022, there were 427 HSCT perfomed (337 (79%) allogeneic and 90 (21%) autologous) in 388 children at our center. In case of koryza, cough, sneezing and sore throat presence of respiratory viruses was tested. Between 1st September 2012 and 31st January 2023, we have obtained 334 respiratory tract samples from 104 boys and 55 girls (305 nasopharyngeal swabs, 14 BALs, 2 sputum and 13 low respiratory tract aspirates) sampled from D-10 (about start of conditioning therapy). Nucleic acids were extracted using Seeprep 12 kits (Seegene Inc., South Korea) and Zybio kits and EXM3000 machine (Zybio, Chongqing, China). Subsequent PCR detection of respiratory viruses (influenza A and B virus (FluA, FluB), parainfluenza viruses 1-4 (PIV), respiratory syncytial virus (RSV), human metapneumovirus (hMPV), human rhinovirus (HRV), human enteroviruses (EV), human bocavirus (HBoV), adenovirus, seasonal coronaviruses (SeasCoV) and later SARS-CoV-2) was performed using Anyplex™ II RV16 kits (Seegene Inc., South Korea) till 2015 and later by Respiratory Viruses 16-well (AusDiagnostics Pty Ltd, Mascot, Australia).

Results: Any positivity of tested viruses was detected in samples from 71 boys and 38 girls; 68.6% of tested children. Children were positive for FluA virus in 6 pts.(5.5% of positive), FluB virus in 2 pts.(1.8% of positive), RSV in 21 pts.(19.3% of positive), hMPV in 10 pts.(9.2% of positive), PIV in 18 pts.(16.5% of positive), HRV in 64 pts.(58.7% of positive), EV in 3 pts.(2.8% of positive), HBoV in 6 pts.(5.5% of positive), adenovirus in 14 pts.(12.8% of positive), SeasCoV in 14 pts.(12.8% of positive) and SARS-CoV-2 in 11 pts.(10.1% of positive). First detection was observed with median of 123 days after HSCT (range -9 to 2316) during the whole year with higher frequency of detection from September to March. Detection of more viruses during the period was detected in 42 patients. In patients with influenza and paramyxoviruses (RSV, hMPV and PIV), treatment with oseltamivir or ribavirine was started. In last years, new virostatics were used for treatment too. The only patient deceased clearly due to respiratory viruses was patient with severe combined immunodeficiecy. In this patient the primary diagnosis was already complicated with FluA infection treated with oseltamivir. After 4 weeks of therapy, treatment was stopped and patient was negative in the samples. During the conditioning therapy, patient presented with respiratory symptoms and hepatitis and his clinical status deteriorated despite the virostatic therapy; later resistance of FluA to oseltamivir (H275Y) was detected.

Conclusions: Our data documents the necessity of testing in wide respiratory panel due to high frequency of potencially harmful but treatable viral infection in HSCT setting.

Disclosure: Supported by the project for conceptual development of research organization 00064203 and by the project National Institute of Virology and Bacteriology (Programme EXCELES, ID Project No. LX22NPO5103).

13: Infectious Complications

P466 MUTUAL IMPACT AND CHARACTERISTICS OF VIRAL REACTIVATIONS AND IMMUNE RECONSTITUTION AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN. A SINGLE CENTER EXPERIENCE

Cristian Jinca 1,2, Andrada Oprisoni1,2, Anca Isac2, Andreea Pascalau2, Loredana Balint-Gib2, Margit Serban2, Mihaela Baica2, Smaranda Arghirescu1,2

1”Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania, 2Emergency Hospital for Children LOUIS TURCANU, Timisoara, Romania

Background: Viral reactivation (VR) occurs frequently after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in the context of immunodeficiency and immunosuppression, causing morbidity and complications. Delayed immune reconstitution (IR) has been suggested to impact on VR.

Methods: Fourty-nine patients under the age of 18 years receiving allogeneic HSCT in our center, have been enrolled in this study. 21 patients were transplanted for non-malignant diseases. Viral load screening was performed on a weekly basis by PCR for adenovirus (AdV), Epstein-Barr virus (EBV), human herpesvirus 6 (HHV6), cytomegalovirus (CMV), and BK polyoma virus (BKPyV) starting with the first week post-transplant until hospital discharge and afterwards in case of suspicion of reactivation. The IR was assessed for B, T and NK cells by flowcytometry at 1, 3, 6, 9, 12 months and at 2 years post-transplant. Statistical analysis was performed using the SPSS® Statistics software, version 25.

Results: Overall, 50% of patients presented viremia. The incidence of viral reactivation did not differ significantly between patients with malignant and those with non-malignant diseases. The most frequent reactivations in our cohort were for BKPyV and for CMV. CMV VR occurred in 26.5 % of patients (all reactivations occurred in patients with D + / R+ serology whereas patients with D-/R+ or D + /R- serology status was not associated with VR, possibly due to the prophylactic approach). One patient developed CMV pneumonia. Preemptive treatment with gancyclovir and reduction of immunosuppression were the mainstay of therapy in case of reactivation. 18% of children reactivated BKPyV, 25% of the them presenting symptomatic hematuria. The median time to first detection of BKPyV in urine was 31.5 days and median time to clearance of viremia was 82.5 days. Treatment consisted of supportive measures and reduction of immunosuppression. EBV reactivation occurred in 8.2 %. HHV6 reactivation occurred in 14 % of the patients. Persistence of HHV6 viremia was associated with progressive mixed chimerism in one patient with very severe aplastic anemia but chimerism improved after treatment with foscarnet. Viral co-infection occurred in 14 % of patients. The most delayed IR characterized CD19+ lymphocytes with a median time to reach the cut-off absolute value at 7.2 months. At 3 months after HSCT CD19+ and CD4+ cells did not reach the cut-off absolute counts, whereas a general trend to reach the cut-off level for absolute counts of CD8+ lymphocytes early after engraftment was observed in all patients except for patients receiving grafts from CMV negative donors. EBV VR was especially associated with high CD8+ counts. NK cells reached the cut-off level at a median time of 3.5 months post-HSCT. Patients with delayed reconstitution of the CD4+ cells presented a higher risk of VR.

Conclusions: Viral reactivations are common following allogeneic HSCT impacting on IR but severe complications are rather rare. On the other hand, timely reconstitution of CD3 + CD4+ lymphocytes seems to reduce the incidence of post-transplant viral reactivations. Patients with delayed immune reconstitution may benefit from more intensified viral load screening in order to personalize preventive strategies for viral infections.

Disclosure: Nothing to declare.

13: Infectious Complications

P467 CMV MONITORING AND LETERMOVIR PROPHYLAXIS IN ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A SINGLE CENTER EXPERIENCE

Yuta Katayama 1, Yu Kochi1, Takuya Nunomura1, Riichiro Ikeda1, Kenjiro Hino1, Ryota Imanaka1, Kohei Kyo1, Takeshi Okatani1, Mitsuhiro Itagaki1, Shinya Katsutani1, Tsuyoshi Muta1

1Hiroshima Red Cross Hospital and Atomic-bomb Survivors Hospital, Hiroshima, Japan

Background: Cytomegalovirus (CMV) infection is a complication with poor prognosis in allogeneic hematopoietic stem cell transplantation (allo-HSCT). Pre-emptive therapy guided by CMV DNA PCR monitoring has become standard practice, and recently, letermovir (LET) has been focused on prevention. This study retrospectively evaluated the efficacy of CMV monitoring in using LET administration at our institution.

Methods: We enrolled 55 patients who underwent allo-HSCT at our hospital between April 1, 2021, and July 31, 2022. CMV DNA was monitored using the cobas CMV6800/8800 System before and after transplantation. Monitoring was performed weekly for up to 14 weeks post-transplant and then weekly or every two weeks for up to 48 weeks. There were 7 patients with sibling allo-HSCT, 20 with unrelated allogeneic bone marrow transplantation, and 28 with cord blood transplantation (CBT). Recipients were CMV antibody positive in 48 patients. Thirty-seven donors were CMV antibody-negative, including 28 on CBT. ATG was used in 25 of the 27 patients who underwent allo-HSCT, excluding CBT. All patients who were CMV antibody-positive before transplantation received LET until day 100, whereas patients who were CMV antibody-negative before transplantation did not receive LET.

Results: Forty-eight patients were CMV antibody-positive before transplantation, and 6 of these patients died within 14 weeks after transplantation. In the 6 patients, no deaths were attributed to CMV infection. CMV DNA was detected in 18 patients within 14 weeks after transplantation, and CMV DNA was >150 IU/mL in 6 patients. CMV DNA was detected in 4 of these 6 patients in the pre-transplant examination. One of them had CMV DNA >150 IU/mL before transplantation. Five of the six patients had undergone CBT and all six were transplanted from CMV antibody-negative donors. Within 14 weeks after transplantation, LET prevented CMV DNA levels from reaching ≥150 IU/mL in 36 patients. After completing LET, 19 of the 36 patients had CMV DNA PCR ≥350 IU/mL. Of these 19 cases, CMV DNA was detected in 8 cases within 14 weeks. At the onset of CMV reactivation, CMV DNA levels were >500 IU/mL in 10 patients and >1,000 IU/mL in 5 patients. CMV DNA levels of >1,000 IU/mL occurred in 16 patients even during the pre-emptive therapy. CMV infections were observed in three patients: retinitis on day 336, enteritis on day 142, and gastritis on day 526. Seven patients who were CMV antibody-negative before transplantation were undetectable by CMV DNA before and after transplantation.

Conclusions: CMV DNA ≥ 150 mL/IU within 14 weeks after LET administration was more frequent in patients with CMV DNA detected before transplantation. Therefore, monitoring CMV DNA PCR before transplantation is necessary to predict CMV reactivation during LET administration. In this study, many cases of CMV reactivation were observed after discontinuation of LET, and CMV DNA levels exceeding 1,000 IU/mL were observed. It was considered that many patients underwent CBT or allo-HSCT used ATG, and many CMV antibody-negative donors were included in our study. It is recommended that LET be administered to such patients after 100 days post-transplantation.

Disclosure: Nothing to declare.

13: Infectious Complications

P468 LETERMOVIR COMBINED WITH GANCICLOVIR AS A PREEMPTIVE OR TREATMENT FOR CYTOMEGALOVIRUS INFECTION FOLLOWING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION, A SINGLE-CENTER RETROSPECTIVE REVIEW

Han Yao 1,2, Yimei Feng1,2, Ting Chen1,2, Lu Zhao1,2, Yuqing Liu1,2, Lidan Zhu1,2, Jia Liu1,2, Lu Wang1,2, Shichun Gao1,2, Huanfeng Liu1,2, Lei Gao1,2, Peiyan Kong1,2, Xi Zhang1,2,3

1Army Medical University affiliated Xinqiao Hospital, Chongqing, China, 2State Key Laboratory of Trauma, Burns and Combined Injury, Chongqing, China, 3Jinfeng Laboratory, Chongqing, China

Background: Cytomegalovirus (CMV) remains a cause of morbidity and mortality after hematopoietic stem cell transplantation (HSCT). CMV reactivation or disease with preventative letermovir application were observed in HSCT recipients. Associated specific mutations were identified with in the terminase complex region of the CMV genome. Few options for treatment of CMV infections and in many instances, therapeutic options are limited for toxicity of the agents used or transplant-related complications. This is often the case with conventional agents such as ganciclovir, foscarnet, and cidofovir. Given the differed mechanism of letermovir and ganciclovir, considering the organ toxicity of ganciclovir, we chose a tapered dose of ganciclovir combined with letermovir in preemptive or treatment of CMV infection. This study aims to improve the outcome of reactivation of CMV or CVM disease.

Methods: Data were retrospective analyzed on patients receiving allogeneic HSCT at our center from October 2022 to October 2023. Patients were divided into the preventive group, preemptive group, and treatment group according to occurrence of CMV reactivation or CMV disease. The baseline characteristic, CMV copies, disease outcome, overall survival (OS) within 1 year in each group, as well as comparison of adverse events among the groups were analyzed by R statistical software.

Results: Data were retrospective analyzed at our center from October 2022 to October 2023. Patients were divided into the preventive group (87 cases), preemptive group (20 cases), and treatment group (15 cases) according to occurrence of CMV reactivation or CMV disease. The baseline characteristic, CMV copies, disease outcome, overall survival (OS) within 1 year in each group, as well as comparison of adverse events among the groups were analyzed by R statistical software. During the 1-year follow-up of the 122 cases(preventive group, preemptive group and treatment group), the 1-year OS for each group was 92.5%, 90.3%, and 88.5%, respectively (P=0.67), with no statistically significant difference at baseline. At the end of the first week after treatment, 95% of patients in the preemptive group reached negativity of CMV copies, compared with only 76% of patients in the treatment group. At the end of the second week after treatment, all patients in the preemptive group reached negativity of CMV copies, and no one died; accordingly,97.5% of patients in the treatment group reached negativity of CMV copies, 1 out of 15 patients died of VOD and 1 died of severe lung infection, considering associated with CMV disease. In the course of treatment, all three groups of patients developed a certain degree of adverse events. While no significant differences in the three groups in adverse events such as gastrointestinal events, cardiac events, hematologic toxicity, liver and hepatic and renal insufficiency.

Conclusions: Letemovir and ganciclovir may be applied in preemptive measure against CMV reactivation or as a treatment for CMV disease in patients receiving allogeneic HSCT. Ganciclovir at an tapered dose in combination with letemovir is efficient, particularly in reducing the duration of infection and preventing the poor prognosis that followed. In terms of safety, this regimen helps lessen the harm that conventional treatment causes to the liver, kidneys, and severe hematologic toxicity.

Disclosure: Nothing to declare.

13: Infectious Complications

P469 THE CLINICAL MANIFESTATION, PROGNOSTIC FACTORS, AND OUTCOMES OF ADENOVIRUS PNEUMONIA AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Yuewen Wang 1, Xiaohui Zhang1, Lanping Xu1, Yu Wang1, Chenhua Yan1, Huan Chen1, Yuhong Chen1, Fangfang Wei1, Wei Han1, Fengrong Wang1, Jingzhi Wang1, Xiaojun Huang1, Xiaodong Mo1

1Peking University People’s Hospital, Beijing, China

Background: Adenovirus (ADV) infection is one of the important opportunistic viral infections after allogeneic hematopoietic stem cell transplantation (allo-HSCT). ADV pneumonia is one of the most severe types of ADV infection in HSCT recipients with high mortality. We aimed to identify the clinical manifestation, prognostic factors, and outcomes of ADV pneumonia after allo-HSCT.

Methods: Twenty-nine patients who underwent allo-HSCT at the Peking University Institute of Hematology and met the criterion of ADV pneumonia after allo-HSCT were retrospectively enrolled. ADV was detected with real-time quantitative polymerase chain reaction (RT-PCR). This work was supported by the National Key Research and Development Program of China (2022YFC2502606), Peking University People’s Hospital Research and Development Funds (RZ2022-02), Natural Science Foundation of Beijing (Z230016) and Tongzhou District Distinguished Young Scholars (JCQN2023009).

Results: The median time from allo-HSCT to the occurrence of ADV pneumonia was 99 days (range 17-609 days). The most common clinical manifestations were fever (86.2%), cough (34.5%) and dyspnea (31.0%). The 100-day cumulative incidence of ADV-related mortality was 40.4% (95%CI 21.1%-59.7%) and the probability of overall survival (OS) at 100 days after ADV pneumonia was 40.5% (95% CI 25.2%-64.9%). The 30-day cumulative incidence of ADV-related mortality for the patients with low-level (<106 copies/ml in plasma) and high-level (≥ 106 copies/ml in plasma) ADV DNAemia was 15.0% (95% CI 0-31.1%) versus 80.0% (95% CI 37.5%-100%) (P=0.005), respectively. The probability of OS at 30 days after ADV pneumonia was 75.0% (95% CI 58.2%-96.6%) versus 0% (P=0.003) for patients with low-level and high-level ADV DNAemia, respectively. In multivariate analysis, high-level ADV DNAemia (HR 6.39, 95% CI 1.43-28.67, P=0.015) was the only risk factor associated with ADV-related mortality and OS at 100 days after ADV pneumonia (HR 5.45, 95% CI 1.55-19.16, P=0.008).

Conclusions: We firstly reported the clinical manifestations, prognostic factors, and outcomes of ADV pneumonia after allo-HSCT and we further confirmed that this is a life-threatening post-transplant complication. New drugs or cytotherapy may help to further improve the clinical outcomes of these patients.

Disclosure: Nothing to declare.

13: Infectious Complications

P470 OPTIMIZING CMV PREVENTION IN STEM CELL TRANSPLANTS: HYBRID STRATEGY DELIVERS PROMISING REDUCTION IN REACTIVATION RATES

Amirabbas Rashidi1,2, Tanaz Sayar Bahri1,2, Maryam Barkhordar 1,2, Seied Asadollah Mousavi1,2, Ahmad Khajeh-Mehrizi3, Mohammad Vaezi1,2

1Research Institute for Oncology, Hematology and Cell Therapy, Tehrani University of Medical Sciences, Tehran, Iran, Islamic Republic of, 2Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Tehran University of Medical Sciences, Tehran, Iran, Islamic Republic of, 3Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran, Iran, Islamic Republic of

Background: Cytomegalovirus (CMV) reactivation poses a significant threat to allogeneic hematopoietic stem cell transplant (HCT) recipients, especially in the context of unrelated donors and various high-risk factors. This cohort study aimed to assess the effectiveness of an aggressive hybrid prophylaxis strategy in reducing CMV reactivation and associated complications in high-risk haploidentical and unrelated allo-HSCT recipients.

Methods: The study, conducted at Shariati Hospital from 2016 to 2021, enlisted 86 CMV-seropositive patients who underwent haploidentical and unrelated allo-HSCT. A hybrid prophylaxis approach was employed, entailing ganciclovir (5 mg/kg IV daily) from Day -8 to Day -2 before transplantation. Primary prophylaxis involved high-dose valacyclovir from Day -1 until +100 (2000 mg, PO, every 8 hours), with a substitution to Acyclovir (500/m2, IV, every 8 hours) if the patient could not tolerate oral medications. A historical pre-emptive control group was utilized for comparison.

Hybrid

n=15

Pre-emptive

n=20

Unadjusted HR (95% CI)

P value

Adjusted HR (95% CI) *

P value

CMV reactivation

8 (53.3)

18 (90)

0.37 (0.16-0.88)

0.025

0.12 (0.03-0.46)

0.002

Early

≤ +28 days post-BMT

6 (75)

15 (83.3)

0.41 (0.15-1.06)

0.066

0.14 (0.03-0.57)

0.006

Late

>28 -100 days post-BMT

2 (25)

3 (16.4)

0.28 (0.04-1.71)

0.16

0.26 (0.003-0.27)

0.57

High-level viremia > 1000 copy/ml CMV viral load

5 (30)

12 (60)

0.48 (0.15-1.54)

0.22

0.18 (0.03-0.96)

0.04

Results: Among the 86 patients initially considered, our final analysis centred on 35 eligible individuals. This included 15 patients (43%) from the hybrid cohort and 20 patients (57%) from the pre-emptive control group. Patient characteristics were comparable between the two groups, with the major difference being the number of CD34+ cells in the graft. Incidence and timing of CMV reactivation showed a lower cumulative incidence in the hybrid cohort compared to the pre-emptive group (53% vs. 90%, p = .019). The hybrid strategy was associated with a significant reduction in CMV reactivation risk (HR 0.12; 95% CI 0.03-0.46; p = .002). Patients in the hybrid cohort had fewer days on CMV-specific antiviral therapy (median 23% vs. 44%, p = .001). No significant differences were observed in time-to-engraftment, platelet recovery, or nonrelapse mortality.

Conclusions: The hybrid prophylaxis strategy demonstrated significant efficacy in reducing CMV reactivation and associated complications in high-risk haploidentical and unrelated allo-HSCT recipients. The strategy was well-tolerated, with no significant renal toxicity observed. These findings suggest that the hybrid approach may serve as a valuable option for CMV prevention in alternative donor HSCT recipients, pending validation through prospective randomized trials.

Disclosure: Nothing to declare.

13: Infectious Complications

P471 STRATIFICATION AND IMMUNOLOGIC ANALYSIS OF PATIENTS WITH SARS-COV-2 INFECTION AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION

Man Chen 1, Wei Zhao1, Hui Wang1

1Beijing Lu Daopei Hospital, Beijing, China

Background: In December 2022, Beijing was hit by the Omicron epidemic. Because their immunity is in a state of reconstruction and their immunity is low, covid-19 patients after hematopoietic stem cell transplantation need better prevention and treatment in the epidemic. we classified the post-transplant infection of COVID-19 clinically and analyzed the immune status of the infection course.

Methods: A total of 47 patients were collected from December 2022 to January 2023 in transplantion department of Beijing Lu Daopei Hospital, including 23 males and 24 females. Aged 33 (12, 44) years old, all cases of covid-19 infection after hematopoietic stem cell transplantation, including 30 mild cases, 11 medium cases, 6 severe cases, and no critical cases. Flow cytometry was used to analyze the changes of immune cell subsets and cytokines in the patients before and after the covid-19 infection and the difference in the mild, moderate and severe disease groups. All data are shown in Table 1.

Results: 1. The proportions of COVID-19 vaccination among the mild, moderate, and severe cases were 8/30, 5/11, and 3/6, respectively, with no significant difference among the groups (P=0.345). The proportions of GVHD at the time of infection among the mild, moderate, and severe cases were 12/30, 7/11, and 3/6(P=0.432). The percentages of blood oxygen saturation decrease among the mild, moderate, and severe cases were 0/30, 1/11, and 6/6, respectively (P<0.001). The proportion of pneumonia occurred among the mild, moderate, and severe cases were 0/30, 8/11, and 6/6, respectively(P<0.001). The proportions of decreased hemogram among the mild, moderate, and severe cases were 17/30, 9/11, and 4/6, respectively (P=0.346). The interval between transplantation and infection of covid-19 was 234 (90.5-459) days in mild patients, which was greater than 174.00 (94-190) days of moderate group and 174.50 (56.5-285.5) days of severe group patients, but the difference did not reach significance. The number of days for severe cases to turn negative in the covid-19 nucleic acid test was longer than that of the moderate and mild groups (P=0.024).

2. The infection-related indicators of patients in the severe group were significantly higher than those of the other two groups after infection of covid-19, and there were significant differences among the three groups: the level of CRP (P=0.001), PCT (P=0.005), and IL-6 (P=0.041) in the severe group was significantly higher.

3. we found that the ratio of central memory T cells (TCM) and naive T cells (NaiveT) in severe patients was was higher than in the mild and moderate disease group (P=0.013, P=0.010), among which CD8+NaiveT was significantly higher In the other two groups (P=0.017).

Conclusions: The severe group may be at an earlier post-transplantation stage than the mild group, thus the number of immune cells is less than that of the other two groups. But in the other hand, we also observed that NaiveT and TCM was higher than that of the other two groups, May be associated with a more severe clinical presentation.

Disclosure: Nothing to declare.

13: Infectious Complications

P472KLEBSIELLA PNEUMONIAE INFECTION BEFORE AND AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION IN THE ERA OF NOVEL ANTIBIOTICS: A COMPARATIVE STUDY

Eleni Gavriilaki 1, Damianos Sotiropoulos2, Ioannis Batsis2, Despina Mallouri2, Alkistis Panteliadou2, Nikolaos Spyridis2, Giorgos Karavalakis2, Paschalis Evangelidis1, Vasiliki Kanava2, Giannis Kyriakou2, Eleni Papchianou2, Christos Demosthenous2, Zoi Bousiou2, Anna Vardi2, Ioanna Sakellari2

1Aristotle University of Thessaloniki, Thessaloniki, Greece, 2G. Papanicolaou Hospital, Thessaloniki, Greece

Background: Novel antibiotics [ceftazidime/avibactam (C/A) and ceftolozane/tazobactam (C/T)] have shown efficacy in Carbapenemase-producing (KPC)-Klebsiella pneumoniae (Kp) infections. However, their impact on KP infections before and after allogeneic hematopoietic cell transplantation (alloHCT) has not been extensively evaluated.

Methods: We retrospectively studied consecutive patients with Kp infection before and/or after alloHCT (January 2008- December 2022). Data cut-off for follow-up was October 2023. AlloHCT was performed according to standard operating procedures of our JACIE-accredited center. Patients with colonization or infection pre-transplant received secondary prophylaxis pre and during transplant. Nurses, visitors and staff were carefully trained on infection control measures. Since 2019, C/A was administered as empirical therapy for KPC-Kp colonized patients. C/A and C/T were also administered according to antibiograms. Statistical analysis included the following factors: age, gender, disease phase at transplant (early, intermediate or advanced), donor, pre-transplant and post-transplant Kp infections and colonizations, KPC-Kp, severe acute and extensive chronic GVHD, relapse, treatment-related mortality (TRM) and overall survival (OS).

Results: In the early study period (2008-2018), Kp infections were found in 52/424 (12%) of alloHCT patients, compared to 24/233 (10%) in the current period (2019-2022).

Among patients with Kp infections, no significant difference was observed in pre-transplant characteristics between time-period. Pre-transplant Kp infections significantly increased the risk of post-transplant Kp infections (p=0.015), despite secondary prophylaxis. With a median follow-up of 18.7 months (range 0.4-212), cumulative incidence (CI) of severe acute GVHD was 40.5% and moderate/severe chronic 41.7%; both similar for two time-period. Acute GVHD was associated only with pre-transplant Kp infections (p=0.015), among pre-transplant and transplant factors.

One-year CI of TRM was independently associated with acute GVHD (p=0.007) and pre-transplant Kp infections (p=0.025). Overall survival (OS) was significantly associated only with disease phase at transplant (p=0.001).

Conclusions: Our study highlights the significant impact of Kp infections on TRM, despite the use of novel antibiotics. Nevertheless, Kp infections did not impact OS in our population. Novel antibiotics and secondary prophylaxis allowed the performance of alloHCT with an acceptable outcome.

Disclosure: Nothing to declare.

13: Infectious Complications

P473 OUTCOMES OF HYPERBARIC OXYGEN THERAPY IN LATE-ONSET HEMORRRHAGIC CYSTITS AFTER ALLO-HSCT: A RETROSPECTIVE ANALYSIS

Mariana Trigo Miranda 1, Joana Arana Ribeiro2, Diogo Alpuim Costa3,4,3, Carla D’Espiney Amaro4,3, Andreia Teixeira5,6, Inês Portugal Rodrigues7, Clara Gaio-Lima7, Carlos Pinho Vaz8, Óscar Camacho7

1Centro Hospitalar Tondela-Viseu, EPE, Viseu, Portugal, 2Unidade Local de Saúde da Guarda, EPE, Guarda, Portugal, 3Centro de Medicina Subaquática e Hiperbárica, Lisboa, Portugal, 4CUF Oncologia, Lisboa, Portugal, 5University of Porto, Porto, Portugal, 6Instituto Politécnico de Viana do Castelo, Viana do Castelo, Portugal, 7Unidade Local de Saúde de Matosinhos, Porto, Portugal, 8Instituto Português de Oncologia do Porto Francisco Gentil, EPE, Porto, Portugal

Background: Hemorrhagic cystitis (HC) is a common complication after allogeneic hematopoietic stem cell transplantation (allo-HSCT). It can present early, mainly due to regimen-related toxicity, or have late-onset, often caused by viruses such as BK polyomavirus (BKV), adenovirus (ADV), cytomegalovirus (CMV) or JC polyomavirus. Treatment is usually based on supportive measures like forced diuresis, bladder irrigation, platelet transfusion and sometimes the use of antivirals such as cidofovir. Hyperbaric oxygen (HBO) therapy works by promoting angiogenesis and fibroblast proliferation and has shown good results mainly in the setting of BKV associated HC after allo-HSCT. Our aim in this retrospective analysis was to review the outcomes of this therapy in allo-HCST patients from two Portuguese centers, highlighting response rates and significant associations.

Methods: Retrospective data was collected encompassing patients who underwent HBO therapy for late-onset HC after allo-HSCT spanning from 1998 to 2022 from two Portuguese medical centers. We looked for differences between groups of patients and their outcomes with a focus on the diagnosis, type and source of transplant and other patient related characteristics. The data analysis was conducted using SPSS v.27®. Significance was determined at p≤0.05.

Results: A sample of 78 patients were evaluated. 52.6% were male and the median age was 29 years-old (1-66), 20 children and 58 adults. The most prevalent diagnosis was acute myeloid leukemia (AML) in 26%, acute lymphoblastic leukemia (ALL) in 16.9% and myelodysplastic syndrome (MDS) in 11.7% of cases. 62.5% of transplants were from an unrelated donor and 85.9% were from a peripheral blood stem cells source (PBSC), 9.4% bone marrow (BM) and 4.7% umbilical cord. The median time to onset of hematuria was 33 days and the median time to initiation of HBO therapy was 17.5 days. 98.5% of patients had a viral agent detected in urine samples, being BKV the most prevalent (74.2%). The average number of sessions was 15 (2-85). 2 patients didn’t tolerate the treatment. Complete response was obtained in 71.8% of cases and no response occurred in 14.1%. When it comes to conditioning regimens, 95.5% contained either busulfan, cyclophosphamide or both. We found no relation between the diagnosis and the onset of symptoms. There was a significant difference between the BM and PBSC types (p-value = 0.018). Symptoms began earlier in transplants with a BM source rather than with PBSC. We also found better outcomes (CR + PR) in younger patients (p-value = 0.048).

Conclusions: In this retrospective study on HC after allo-HSCT, there was an overall response rate of 85.9% with HBO therapy. While consistent with findings from other retrospective studies, the lack of prospective randomized controlled trials limits recommendation for the use of this therapy. Of note, we found differences between graft type and between groups of patients who might have a better response to therapy. HBO therapy has emerged as a promising intervention in a dismal complication of allo-HSCT. Future research should focus on finding risk factors and the patients who might benefit the most from this therapy.

Disclosure: Nothing to declare.

13: Infectious Complications

P474 INVASIVE FUNGAL DISEASES IN PATIENTS WITH CLL TREATED WITH BRUTON TYROSINE KINASE INHIBITORS, A MONO-CENTRIC RETROSPECTIVE STUDY

Yuri Vanbiervliet 1, Robina Aerts1, Finn Segers1, Johan Maertens1, Ann Janssens1

1KU Leuven, Leuven, Belgium

Background: Patients with chronic lymphocytic leukaemia (CLL) are prone to infections due to impaired humoral and cellular immunity. Bruton tyrosine kinase inhibitors (BTKIs) have taken in a prominent place in the treatment of CLL. Recently, an increased incidence (ranging from 0.5% to 12.1%) of invasive fungal diseases (IFDs) in patients treated with BTKIs, has been reported, especially central nervous system infections. In this monocentric study we assessed the incidence of IFDs.

Methods: We retrospectively analyzed the medical records of patients with CLL who received BTKIs at the University Hospitals of Leuven between January 2013 and July 2023.

Results: One hundred and seventy-five patients were identified (median age at diagnosis 64 years). 16/175 (9.1%, NA=8) and 11/175 (6.3%, NA =8) presented with a high risk Rai or Binet score, respectively. Forty-five (44.6%, NA=56) of the cohort had a unmutated IgVH status and forty-three (24.6%, NA=21) had a loss/mutation of 17p/TP53. Mean follow-up time of the total cohort was 117 months, 32% of the patients died. Six (3.4%) patients underwent hematopoietic stem cell transplantation (HSCT). One in three patients (61/175, 34.8%) had hypogammaglobulinemia requiring IVIG-treatment. One in six (26/175, 14.8%) and one in ten (20/175, 11.4%) had diabetes mellitus and other pulmonary disease, respectively.

BTKIs were initiated 82 months (mean, range 0-252 months), after CLL diagnosis and after a median of one previous treatment line (range 0-5). For half of the patients (49.7%), BTKIs were started as first-line treatment. In almost half of them (49.4%) the BTKI was continued until last follow-up.

6/175 patients (3.4%) had recovered from a IFD before initiation of BTKIs. 4/175 patients (2.3%) received prophylaxis with trimethoprim/sulfamethoxazole (TPM/SMX) during treatment with BTKIs. In total, 8/175 (4.6%) patients on current or previous treatment with BTKIs were diagnosed with an IFD of whom one patient was first diagnosed with pneumocystosis and 14 months later with invasive aspergillosis (IA). None of these patients were receipt of HSCT. None of these patients were on antifungal prophylaxis. The lungs were the only infection site. Aspergillus fumigatus was the main isolated pathogen (77.8%), the remaining pathogens were Candida krusei and Pneumocystis jirovecii. 7/9 (77.8%) fungal infections occurred during treatment with BTKIs with a mean of 751 days (range 46-2053 days) after initiation. Two episodes occurred after discontinuation of BTKIs after a mean of 426 days (both patients were on active treatment with venetoclax). Concomitant risk factors were common: neutropenia (22.2%), corticosteroids use (66.7%), chemo-immunotherapy within 12 months (33.3%), critical-illness (66.7%) and severe viral pneumonia (33.3%). Strikingly, one patient was diagnosed with probable invasive aspergillosis without other risk factors, apart from the underlying CLL and active treatment with ibrutinib.

Full size table

.

Conclusions: Our monocentric retrospective study shows an incidence for IFDs of 4.6% in patients with CLL treated with BTKIs. Interestingly only pulmonary infections were seen without any case of central nervous system infection. The IFDs were mainly seen in the presence of concomitant risk factors. Our data do not support the routine use of antifungal prophylaxis in BTKI-treated CLL patients.

Clinical Trial Registry: NA.

Disclosure: YV: NA.

RA: NA.

FS: NA.

JM: NA.

AJ: Janssen, Abbvie, Astra Zeneca, Beigene.

13: Infectious Complications

P475 PREDICTORS OF INVASIVE FUNGAL INFECTION AND IMPACT OF OUTCOME AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION: SINGLE CENTER EXPERIENCE FROM THE KINGDOM OF SAUDI ARABIA

Lama AlHmaly 1, Ahmad Alharbi1, Moussab Damlaj1,2,3, Ahmed Alaskar1,2,3,4, Ayman Hejazi1,2,3, Hind Salama1,2,3, Abdulrahman Al raizah1,2,3, Abdullah S. Al Saleh1,2,3, Ayman Ibrahim1,2,3, Ayel Yahya1,2,3, Mohammed Bakkar1,2,3, Inaam Shehabeddine1, Suha Alkhraisat1, Amani Alharbi1, Isam Mahasneh1, Maybelle Ballili1, Mazen Ahmed1, Husam Alsadi1,2,3, Mohsen Alzahrani1,2,3,4, Bader Alahmari1,2,3

1King Abdulaziz Medical City, Ministry of National Guards - Health Affairs, Riyadh, Saudi Arabia, 2King Abdullah International Medical Research Center, Riyadh, Saudi Arabia, 3College of Medicine, King Saud bin Abdulaziz University for Health Sciences, Riyadh, Saudi Arabia, 4Saudi Scientific Society of Blood and Marrow Transplantation (SSBMT), Riyadh, Saudi Arabia

Background: Invasive Fungal Infections (IFIs) pose a significant threat to patients undergoing allogeneic hematopoietic cell transplantation (HCT). This study aimed to investigate the incidence and risk factors of IFI in this patient population and assess its impact on overall survival.

Methods: A total of 189 patients who underwent allogeneic HCT between 2015 until 2021 were included. Demographic characteristics, underlying diseases, transplant-related factors, and prophylactic measures were recorded. IFI was classified as proven, probable, and possible according to the recent EBMT infectious disease working party guidelines. Regression model was utilized to predict odds of IFI and mortality. Log-Rank KM curve used to estimate overall survival. Statistical significance was set at p < 0.05.

Results: The cohort had a median age of 27 years, with 60% being male. The most common underlying diseases were acute lymphoblastic leukemia (35.5%) and acute myelogenous leukemia (34.9%). Other characteristics are available in table 1. The overall Incidence of IFI (proven, probable, and possible) was 27.5%, with a median time of onset at 6.5 months after HCT. However, the incidence rate of proven and probable IFI was 10.5% with a median time of onset at 8.4 months after HCT. In multivariate analysis antifungal prophylaxis, significant steroid use, and transplant-associated microangiopathy (TMA) were significant predictors of proven and probable IFI, with odds ratios of 11.0 (p = 0.0011), 3.8 (p = 0.0394), and 23.6 (p = 0.0009), respectively. Relapse and proven and probable IFI emerged as significant predictors of mortality in multivariate analysis, with odds ratios of 6.9 (p < 0.0001) and 3.1 (p = 0.0357), respectively. After a median follow-up of 28.4 months, the overall survival (OS) for all patients was not reached. The median OS for the no IFI group was also not reached, however, the median OS for the proven and probable IFI group was 17 months, p-value of 0.042.

Table 1. Descriptive Statistics of the Participants in the Study (n=189)

Parameter

n

%

Age, Median (IQR)

27 (14-63)

Sex

Male

114

60

Female

75

40

Underlying disease

Acute myelogenous leukemia

66

34.9

Acute lymphoblastic leukemia

67

35.5

Myelodysplastic syndrome

5

2.6

Aplastic anemia

18

9.5

Non-Hodgkin Lymphoma

7

3.7

Hodgkin Lymphoma

10

5.3

Plasma cell dyscrasia

7

3.7

Myeloproliferative Neoplasm

2

1.1

Chronic myeloid leukemia

4

2.1

Other

3

1.5

Disease status at Allogenic HCT

In complete remission

140

74

Not in complete remission

49

26

Donor type

MRD

136

72

MUD

19

10

HAPLO

29

15.3

MMRD

2

1.1

MMUD

3

1.6

Graft type

PBSCs

174

92.1

BMH

15

7.9

Conditioning regimen

MA

108

57

RIC

64

NMA

17

9

ATG containing regimen

YES

64

33.8

NO

125

66.2

GVHD Prophylaxis

CNI/MTX

98

51.8

CNI/MMF

63

33.3

PTCY

33

17.5

Graft Failure

YES

3

1.6

NO

186

98.4

TMA

YES

9

4.8

NO

180

95.2

CMV reactivation needed treatment

YES

77

40.7

NO

112

59.3

Acute Grade 2-4 acute GVHD before IFI onset

YES

17

10

NO

172

90

History of chronic GVHD (needed systemic therapy) before IFI onset

YES

8

4.4

NO

181

95.6

  1. n: number, %: frequency

Conclusions: Proven and probable IFI significantly reduces overall survival after allogeneic HCT patients. Antifungal prophylaxis, steroid use, and TMA increase the likelihood of development of IFI. Understanding these risk factors can guide strategies for prevention and management of IFI for patients undergoing allogeneic HCT.

Disclosure: Nothing to declare.

13: Infectious Complications

P476 THE ROLE OF COLONIZATION WITH RESISTANT G-BACTERIA IN THE TREATMENT OF FEBRILE NEUTROPENIA AFTER STEM CELL TRANSPLANTATION

Tereza Sokolova1, Pavla Paterova1, Alzbeta Zavrelova1, Benjamin Visek1, Pavel Zak1, Jakub Radocha 1

1University Hospital Hradec Králové, Charles University, Faculty of Medicine in Hradec Králové, Hradec Kralove, Czech Republic

Background: Febrile neutropenia (FN) is one of the most common and most serious complications of chemotherapy treatment in haemato-oncology patients, with a risk of death if sepsis is the cause. The aim of this analysis was to evaluate the frequency of sepsis in patients with FN colonized with resistant G-bacteria (Extended spectrum β-lactamase (ESBL + ) positive AmpC β-lactamase positive, multidrug resistant (MDR) P. aeruginosa) and the choice of primary antibiotic in colonized patients.

Methods: This was a retrospective study analyzed data from patients who underwent hematopoietic stem cell transplantation from 01/2018 to 09/2022. Data were extracted from the hospital information system.

Results: Autologous transplants showed a higher incidence of mucositis (p<0.0001, OR 0.36 [95% CI 0.23-0.57]), a higher incidence of AmpC colonization (p=0.0081), which, however, according to the following analyses, proved no differences to the non-colonized patients, therefore, for further analyses, patients without colonization and with AmpC colonization were included in one group. Allogeneic transplantation showed a significantly higher incidence of FN (p<0.0001, OR 2.79 [95% CI 1.77-4.39]) and sepsis (p<0.0001, OR 2.78 [95% CI 1.42-5.46]). Carbapenem as the primary antibiotic of choice was chosen in 10.9% of non-colonized +/-AmpC patients, 31.5% of ESBL+ patients, and 0% of MDR P. aeruginosa patients. Patients with FN and MDR P. aeruginosa colonization had a high prevalence of sepsis (namely 100%, p = 0.0197). The spectrum of sepsis appeared to be different, with G-bacilli predominating in the ESBL+ group (p = 0.0123, OR 5.39 [95% CI 1.55-18.76]). Colonizer sepsis was present in 100% of sepsis with MDR P. aeruginosa colonization (p=0.002), all in allogeneic transplantation (p=0.0003), with a mortality rate of 33.3% (p=0.0384). The incidence of sepsis in patients with ESBL+ colonization was 25.9% (p=0.0197), with colonizer sepsis in 50% of sepsis cases (p=0.0002), most in allogeneic transplantation (p=0.0003).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (52)

Conclusions: The results show a significant risk of septic state in FN with MDR P. aeruginosa colonization, this state is almost exclusively caused by the colonizer. At the same time, a higher risk of G-sepsis has been demonstrated in patients colonized with ESBL+ bacteria.

Disclosure: Supported by MH CZ - DRO (UHHK, 00179906) and supported by the Cooperatio Program, research area ONCO.

13: Infectious Complications

P477 OUTCOME OF THE USE OF LETERMOVIR AS PROPHYACTIC TREATMENT FOR HSCT RECIPIENTS: A SINGLE CENTER EXPERIENCE

Giulia Baresi1, Marianna Maffeis 2, Elena Soncini1, Stefano Rossi1, Giulia Albrici1, Marta Comini3, Federica Bolda3, Alessandra Beghin3, Arnalda Lanfranchi3, Fulvio Porta1

1ASST Spedali Civili / Pediatric Oncohematology Unit and Pediatric Bone Marrow Transplant Center, Brescia, Italy, 2Pediatric Oncohematology and Bone Marrow Transplant (BMT) Unit, Children’s Hospital, Brescia, Italy, 3ASST Spedali Civili / Stem Cell Laboratory, Section of Hematology and Blood Coagulation, Clinical Chemistry, Brescia, Italy

Background: CMV reactivation is one of the most threatening complications after hemopoietic stem cell transplantation (HSCT). Treatment relies upon anti-CMV specific Immunoglobulin and antiviral drugs such as Ganciclovir, Foscavir or Cidofovir. Recently, antiviral prophylaxis with Letermovir has been proven effective preventing CMV reactivation.

Methods: Here we present 3 years’ experience with the prophylactic use of Letermovir on 22 patients undergoing HSCT for malignant and nonmalignant diseases at Pediatric Bone Marrow Transplant Center in Brescia. Criteria for starting therapy were positive IgG serology versus CMV in donor, recipient, or both. Letermovir was started at a dose of 6 mg/Kg (max dose 240 mg) at time of transplant and continued until day +100.

Results: Among treated patients, a total of 7 out of 16 patients (equal to 31,8 %) presented CMV reactivation, of whom 2 suffered from CMV-related pneumonia responsive to antiviral treatment. Patients who presented CMV reactivation were successfully treated with anti-CMV Immunoglobulin and Ganciclovir. Median time of reactivation was at day +23. Median time of viremia suppression was 16 days after first positive result. Patients are still alive, expect for one who expired for an invasive fungal infection. Reactivations were more associated with CMV-negative donors versus CMV-positive recipients. No drug-related adverse events were experienced.

Concomitant reactivations of other viruses were observed after starting Letermovir: 5 cases of Adenovirus,5 cases of Human herpesvirus 6(HHSV6), 5 cases of Polyomavirus (3 BK, 2 JC), 1 case of Epstein Barr Virus (EBV) and 1 case of Herpes simplex-1 virus (HSV1). None of these concomitant reactivations presented clinical complications and resulted responsive to specific antiviral treatment.

Conclusions: The experience of our center shows an efficacy of the prophylactic use of letermovir in patients undergoing HSCT, even if it showed reactivation events which, overall, allowed effective therapeutic management with rapid resolution without complications.

Disclosure: Nothing to declare.

13: Infectious Complications

P478 COMPARATIVE EVALUATION OF THE EFFECTIVENESS OF LETERMOVIR SINCE ITS USE IN PREVENTION OF CYTOMEGALOVIRUS (CMV) INFECTIONS IN ALLOGENEIC TRANSPLANT PATIENTS HEMATOLOGY DEPARTMENT OF CAEN UNIVERSITY HOSPITAL

Tchuanga Djialeu Yannick Noel 1, Gandhi Damaj Laurent2, Johnson Ansah Hyacinthe Atchroue2, Dina J.2, Parienti Jean Jacques2, Chantepie Sylvain2

1Institut d’Hématologie de Basse Normandie, Caen, France, 2CHU de Caen, Caen, France

Background: Cytomegalovirus (CMV) continues to be one of the most significant complications to arise after allogeneic hematopoietic stem cell transplant (HSCT). Primary infection or reactivation of latent CMV after HSCT contributes to significant morbidity and mortality. Letermovir is a recent antiviral agent indicated in the prevention of CMV infection in patients undergoing HSCT, in terms of both efficacy and tolerance. The aim of this study was to evaluate the rate of CMV infection in real life before and after letermovir approval.

Methods: This was a retrospective real-life cohort study at the University Hospital of Caen from 2015 to 2022. The first cohort was CMV+ recipient who received HSCT between 2015 to 2019 before letermovir era (no letermovir group).

The second cohort was CMV+ recipient receiving HSCT from 2019 to 2022 (letermovir group).

Results: 104 Allo-HCT CMV+ recipients with a median age of 60 years received either a matched graft (genoidentical or matched 10/10 unrelated donor, 50% of cases) or an alternative graft (haploidentical or unrelated 9/10 donor, 50%). Four patients died before 24 weeks. First cohort was not exposed to letermovir (n=67) and in the second cohort (n=37) Letermovir was used with a median duration of 78 days (28-239). The letermovir cohort was older than the non exposed cohort (median age 64 versus 54 years, p=0.002). The clinically significant CMV infection rate was 19.4% (n=13) in the non-letermovir-exposed group and 2.7% (n=1) in the letermovir-exposed group (p = 0.017). Regarding risk factors, age and absence of prophylaxis with letermovir increased the risk of post-transplant infection, as did subjects who underwent incompatible grafts without antiviral prophylaxis.

Conclusions: This retrospective study confimed the benefit of letermovir prophylaxis in CMV prevention in real life at our institution.

Clinical Trial Registry: no.

Disclosure: no.

13: Infectious Complications

P479 EXPERIENCE WITH THE USE OF MARIBAVIR IN HEMATOPOIETIC STEM CELL TRANSPLANT RECIPIENTS WITH REFRACTORY OR RESISTANT CYTOMEGALOVIRUS (CMV)

Mónica Fernández Pérez1, Candela Ceballos Bolaños 1, Itziar Oiartzabal Ormategui1, Nerea Arratibel Zalacain1, Pamela Millacoy Austenrritt1, Marina Aranguren Ostolaza1, Maria Cruz Viguria Alegria1, Carlos Manuel Panizo Santos1

1Hospital Universitario Donostia, Donostia, Spain

Background: Maribavir is an antiviral with activity against cytomegalovirus (CMV), approved for the treatment of adult and pediatric patients with CMV infection/disease post-transplant refractory to treatment (with or without genotypic resistance) with ganciclovir, valganciclovir, cidofovir, or foscarnet. It offers a more favorable safety profile compared to the dose-limiting side effects of previously available therapies.

Methods: This is a retrospective, single-center study conducted at the Donostia University Hospital (HUD). Seven patients undergoing hematopoietic stem cell transplantation (HSCT) between 1/05/2022 and 30/11/2023 with positive CMV load were analized. Recorded variables included gender, age, pathology, conditioning, graft-versus-host disease (GVHD) prophylaxis, CMV serology of donor and recipient, complications during HSCT, CMV reactivation date, drugs used, CMV copy number, and treatment-associated complications.

Results: Of the total analized patients, 28.60% were females, and 71.40% were males. The mean age was 56.57 years (30-67). Regarding hematological pathology, 28.60% had myelodysplastic syndrome (MDS), another 28.60% acute lymphoblastic leukemia (ALL), another 28.60% non-Hodgkin lymphoma (NHL), and 14.20% multiple myeloma (MM). Of the total, 85.80% underwent HLA-mismatched HSCT, and 28.60% underwent Haploidentical HSCT. Conditioning regimens included FluBu2 (28.60%), Flumel (28.60%), TBF (28.60%), and TBI-Cy (14.20%). Regarding GVHD prophylaxis, 57.14% received Tacrolimus + Mycophenolate Mofetil + Cyclophosphamide post-HSCT, 28.60% Tacrolimus + ATG + Methotrexate, and 14.20% Tacrolimus + Mycophenolate Mofetil + Methotrexate for the second HSCT.

Complications during HSCT included acute GVHD in 71.40% of patients and chronic GVHD in only 28.60%. All patients had pre-HSCT positive CMV serology, and in 85.80% of patients, the donor had positive CMV serology, with one of them receiving Letermovir as prophylaxis. The median CMV copies at reactivation were 3251 (1420-5710). The first-line treatment was Foscarnet in 71.40% and Valganciclovir in 14.28%. All patients failed first-line treatment, with 42.85% due to cytopenias, another 42.85% due to acute renal failure, and 14.30% due to lack of response. The median pre-Maribavir copies were 4075 (1060-7120). The median time to reach <35 CMV copies with Maribavir was 18 days (6-45). Adverse effects included dysgeusia in 57.14%, abdominal discomfort in 28.60%, and no adverse effects in 42.85%.

Conclusions: In our center, we observed that Maribavir is an effective drug for clearing CMV viremia in HSCT recipients with refractory or resistant CMV, with a good safety profile. The drug’s rapid action, as observed in our center compared to the literature, has led to its good acceptance and adherence by the patients themselves.

Disclosure: Nothing to declare.

13: Infectious Complications

P480 IS THERE A PLACE FOR ANTI-HCMV HUMAN IMMUNOGLOBULIN AS PRE-EMPTIVE THERAPY IN T-DEPLETE TREG/TCON ALLOGENEIC STEM CELL TRANSPLANTATION?

Valerio Viglione 1, Loredana Ruggeri2, Antonella Mancusi1, Sara Tricarico2, Alessandra Cipiciani1, Rebecca Sembenico1, Matteo Caridi1, Francesco Zorutti1, Anna Castaldo1, Simonetta Saldi1, Roberto Castronari2, Silvia Bozza1, Cynthia Aristei1, Antonella Mencacci1, Maria Paola Martelli1, Antonio Pierini1, Alessandra Carotti2

1University of Perugia, Perugia, Italy, 2Santa Maria della Misericordia Hospital, Perugia, Italy

Background: Human cytomegalovirus (HCMV) is a beta-herpesvirinae, which increases morbidity and mortality in immunocompromised individuals. The use of letermovir prophylaxis reduced HCMV clinically relevant infections in the early phase (<100 days) after allogeneic hematopoietic stem cell transplantation (allo-HSCT), but in some patients and in later phases HCMV might still cause disease. To date, the therapy of HCMV relies on (Val)Ganciclovir and Foscarnet. These therapies are effective, but also burdened by important side effects, such as myelotoxicity, lymphotoxicity or nephrotoxicity. Allo-HSCT with regulatory and conventional T-cell immunotherapy (Treg/Tcon allo-HSCT) is followed by fast immune reconstitution. We evaluated the role of specific anti-HCMV human immunoglobulin in this setting.

Methods: We retrospectively analyzed 86 consecutive Treg/Tcon allo-HSCT from October 2019 to October 2023.

All recipients with positive serology received prophylaxis with Letermovir from day +5 to +100.

HCMV DNAemia was assessed by quantitative PCR on whole blood twice a week from day -1 to day +28, once a week for subsequent 3 months and less frequently thereafter.

Results: HCMV-DNA has been detected in 11 patients (12%). All had positive serology for HCMV and received prophylaxis with Letermovir. Median time of reactivation was day +134 (39-163). Nine/11 patients received therapy with Ganciclovir.

We report the case of 2 HLA-haploidentical Treg/Tcon allo-HSCT, who received monotherapy with anti-HCMV human immunoglobulin as pre-emptive treatment. In both cases, donor had negative serology for HCMV.

A 62-years-old female with AML developed acute intestinal GVHD on day +34 and was treated with a long-course of steroids and vedolizumab. On day +180, we found 11604 HCMV-DNA copies/uL, rapidly increasing. At the time of reactivation, the patient had full donor chimerism, concomitant mild renal impairment (eGFR 60 mL/min/1.73m), grade 2 anemia, grade 3 thrombocytopenia, peripheral T-CD4+ 108/uL and T-CD8+ 904/uL.

A 58-years-old male with AML was found with 200913 HCMV-DNA copies/uL at day +157, exponentially increasing. The patient had full donor chimerism but concomitant poor graft function (grade 3 thrombocytopenia, grade 3 neutropenia, grade 2 anemia) and impaired immune reconstitution (peripheral T CD4+ 40/mmc, T CD8+ 81/mmc).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (53)

Considering the absence of symptoms but concomitant comorbidities, both patients were judged unfit to other antiviral therapy and received anti-HCMV human immunoglobulin 100 UI/kg for 6 doses every 2-3 weeks. Both patients had a reduction of 56% to 74% of circulating HCMV copies within 3-4 days after the first administration. They both reached and mantained a viraemia under 200 copies/uL after the third administration. Circulating HCMV disappeared on day +405 in the first case, on day +236 in the second.

In the figure, is reported the trend of HCMV-DNAemia (blue lines) with the administration of anti-HCMV human immunoglobulin (arrows).

No patient developed symptoms of HCMV infection or adverse reaction to the medication.

Conclusions: To date, the evidences on the clinical use of anti-HCMV immunoglobulin are still limited. There are evidence of a specific stimulatory effect on T-CD4+ and T-CD8+, but further studies are needed. The presented 2 cases had frailties that made unsafe the use of Ganciclovir or Foscarnet. In both of them, pre-emptive monotherapy anti-HCMV immunoglobulin was effective.

Disclosure: No conflict of interest to declare.

13: Infectious Complications

P481 FREQUENCY AND OUTCOME OF CYTOMEGALOVIRUS INFECTION IN RECIPIENTS OF HAEMATOPOIETIC STEM CELL TRANSPLANTATION – AN EXPERIENCE FROM PAKISTAN

Natasha Ali 1, Zurrya Khan1, Mohammad Usman Shaikh1, Zehra Fadoo1, Salman Adil1

1Aga Khan University, Karachi, Pakistan

Background: While therapeutic interventions, such as immunosuppressants, have made substantial contributions to improved transplant outcomes, infectious complications continue to impact the prognosis of HSCT recipients. Cytomegalovirus (CMV) stands as a significant pathogen responsible for substantial mortality in post-transplant patients. CMV infection can have unintended consequences affecting graft function, elevating the risk of graft versus host disease and acquiring other opportunistic infections. The objective of our study was to assess the frequency and outcomes of cytomegalovirus infection in patients undergoing hematopoietic stem cell transplant at our center.

Methods: This retrospective study was conducted at the Department of Oncology, Aga Khan University Karachi, Pakistan, from January 2015 to December 2021. We included patients who underwent HSCT and developed CMV infection post-transplant. All patients underwent pre-transplant assessments. During transplant admission, quantitative CMV PCR was done to detect CMV infection and at results above 1000 copies/ml, treatment with ganciclovir or valganciclovir was initiated. Descriptive statistics were employed to summarize patient demographics and transplant characteristics. Survival analysis was performed using the Kaplan-Meier method, and differences in survival were assessed using the log survival function charts. Cox regression analysis was employed to assess for factors affecting survival, with age group, gender, CMV status, ATG use, presence of GVHD, and mode of transplant being taken as moderator variables. A p-value of 0.05 was taken as the cutoff for statistical significance. Analysis was done on SPSS Statistics Version 26.

Results: We included 112 patients with 59 adults and 53 pediatric patients. Median age was 20 years. Acute leukemia was the most common indication (48%). GVHD occurred in 36% of patients. CMV PCR testing revealed 24/58 patients had levels exceeding 1000 copies.

Pre-transplant, on serology, 93% of tested donors had positive CMV IgG antibodies, while none had positive CMV IgM antibodies. Among patients, 98% had positive CMV IgG antibodies, and 4.1% had positive CMV IgM antibodies. In the cohort of 112 individuals, there were 58 new cases of CMV infection detected during the transplant period, resulting in a frequency of 52% with 97% of cases occurring within the first 100 days. CMV infection within 30 days occurred in 57%, between 30 and 100 days in 38% and after 100 days in 5% of patients.

CMV PCR positive patients had a mean survival of 43 months (95% CI: 31.3 to 55.2 months), while CMV PCR negative patients showed an estimated mean survival of 70 months (figure 1). Cox regression showed CMV positive adults with GVHD had a lower mean survival (table 1). CMV status (HR: 2.2, p-value: 0.04) and ATG use (HR: 2.3, p-value: 0.04) were associated with an increased hazard of death. All other variables were not significant.

Figure 1

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (54)

Table 1:

Mean survival in months, (95% CI)

Variable

CMV Detected

CMV Undetected

Significance

Age Group

Adults

18 (12 - 24)

58 (43 - 76)

Yes

Pediatrics

54 (39 - 69)

66(54 - 76)

No

Gender

Male

45.5 (31 – 60)

71 (57.5 – 85)

No

Female

31 (20– 42)

45 (27 – 63)

No

Type of transplant

Allogenic

45.3(32 – 59)

68 (55.4 – 81)

No

Haploidentical

Could not be estimated

Could not be estimated

-

Use of ATG

Yes

43.2 (23 – 63.6)

56 (41– 71)

No

No

32.3 (24.4 – 40.2)

67 (51.2 – 82)

Yes

GVHD

Yes

21.4 (15 – 28)

44.4 (33 – 56)

Yes

No

52 (37.2 – 67)

68 (53.5 – 82)

No

Conclusions: Overall frequency of CMV infection in our study was 52% with majority occurring in first 100 days of transplant. Adult population with GVHD and patients receiving ATG had a decreased mean survival time. CMV infection represents a significant cause of transplant complication. Understanding of factors worsening the prognosis of CMV infection is important to improve clinical practice.

Clinical Trial Registry: Not applicable.

Disclosure: The authors disclose no conflict of interest.

13: Infectious Complications

P482 CENTRAL VENOUS ACCESS DEVICE IN AUTOLOGOUS STEM CELL TRANSPLANT PATIENTS

Danah Chakfeh 1, Antonette Amao1, Baaba Bentil Tumi1, Josephine Crowe1

1Royal United Hospitals Bath NHS Foundation Trust, Bath, United Kingdom

Background: In order to achieve JACIE accreditation, the SCT centre must conduct clinical mandatory key audit measures. Compliance to such measures helps ensure safe patient care in line with best practice. Audits are documented, systematic evaluation to determine whether approved policies or Standard Operating Procedures have been properly implemented and are being followed. Audits are one of the essential tools to measure the effectiveness of the quality management system and its processes. Audits are conducted by an individual with sufficient knowledge in the process and competence in auditing to identify problems, but who is not solely responsible for the process being audited. Clinical audits are a part of the quality improvement process where current practice is compared with already set standards. The audit process is essential to improving the clinical service. In the Haematology Department, Royal United Hospital (RUH) Bath, audit is undertaken by the multidisciplinary team, including junior doctors in training. One of the mandatory clinical audits is the Central Vascular Access Device (CVAD) in Autologous Stem Cell Transplant Patients.

Methods: We compared our practice to standards from guidelines from internal guidelines, guidelines from other trusts, and NICE guidelines. Data collection and analysis was done using a data collection tool which identified the type of access used, duration of line in situ, complication rates such as thrombosis, infection and how these are managed. We reviewed a total of 18 patients undergoing autograft Stem Cell Transplant from January 2022 to December 2022. A combination of patient written records and computerised notes were used, in particular documentation at the time of the insertion and removal of CVAD, positive cultures and subsequent management.

Results: • 18 patients underwent ASCT.

• 72% of patients with CVAD had a infections where 33% of them were a confirmed line source.

• 5% of patients with CVAD had a thrombotic event.

• Documentation in the medical notes was suboptimal.

Conclusions: • Documentation needs to be improved by using a computer based system to improve accuracy.

• Collaboration to improve current pathway with haematology, radiology and microbiology.

• Prospective monthly audit in real time.

• Results will be used to inform action plan to reduce complications.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P483 FACTORS AFFECTING OUTCOME OF SALVAGE CHEMOTHERAPY AND AUTOLOGOUS STEM CELL TRANSPLANT IN PATIENTS WITH CLASSICAL HODGKIN LYMPHOMA RELAPSING OR PROGRESSING AFTER A FRONT-LINE PET-ADAPTED THERAPY

Simonetta Viviani1, Anna Vanazzi1, Samuele Frassoni2, Chiara Rusconi3, Andrea Rossi4, Lessandra Romano5, Caterina Patti6, Corrado Schiavotto7, Roberto Sorasio8, Vincenzo Marasco3, Laura Lissandrini7, Davide Rapezzi8, Daniela Gottardi9, Federica Cocito10, Antonio Mulé11, Salvatore Leotta5, Guido Gini12, Marco Sorio13, Enrico Derenzini1, Alessandro Rambaldi 4,14, Vincenzo Bagnardi2, Corrado Tarella1,14

1Istituto Europeo di Oncologia (IEO), IRCCS, Milano, Italy, 2Università Milano-Bicocca, Milano, Italy, 3Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy, 4ASST Papa Giovanni XXIII, Bergamo, Italy, 5Azienda Ospedaliera Policlinico Vittorio Emanuele, Università di Catania, Catania, Italy, 6Azienda Ospedali RiunitiVilla Sofia-Cervello, Palermo, Italy, 7Presidio Ospedaliero S Bortolo, Vicenza, Italy, 8Ospedale S Croce e Carle, Cuneo, Italy, 9A.O. Ordine Mauriziano, Università di Torino, Torino, Italy, 10Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 11Azienda Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy, 12AUO Ospedali Riuniti, Università Politecnica delle Marche, Ancona, Italy, 13Azienda Ospedaliera Universitaria Integrata, Verona, Italy, 14Università degli Studi di Milano, Milano, Italy

Background: PET performed after 2 cycles of chemotherapy (CT) (PET-2), is widely adopted to de-escalate or to intensify first-line treatment in patients with classical Hodgkin lymphoma (cHL). However, its predictive value for subsequent salvage therapy in patients who experienced failure of a front-line PET adapted strategy is unknown.

Methods: A retrospective observational multicenter study was performed on individual data of 184 patients with relapsed or refractory (R/R) cHL after first-line CT given according to a PET-adapted strategy. R/R patients were treated at 11 Italian centers with one or more lines of salvage CT followed by high-dose (HD)-CT and autologous stem cell transplant (ASCT) from 2009 to 2021.

We compared characteristics and outcomes of 125 R/R patients with PET-2 negative (PET-2 -ve) with those of 59 with PET-2 positive (PET-2 + ). Study endpoints included Progression-free survival (PFS) (defined as the time from the date of ASCT to progression or death after ASCT, whichever occurred first), Overall Survival (OS) (defined as the time from the date of ASCT to death from any cause or the last date for which information was available), and factors associated with ASCT outcome evaluated by univariate and multivariate analysis.

Results: Main patient characteristics at first relapse or progression according to PET-2 status are shown in Table 1.

Complete metabolic remission (mCR) was achieved before ASCT by 95 (76%) patients of the PET-2 -ve and 32 (54%) patients of the PET-2+ group. After a median follow-up of 36 months, the 3-year PFS and OS for the whole population were 73.4% (95% CI, 65.6-79.7%) and 89.1% (95% CI, 82.7-93.2%), respectively.

There was no significant difference in PFS or OS by type of salvage therapy received in the two groups. In patients who underwent ASCT after ≤ 2 vs > 2 lines of salvage therapy, 3-year PFS was 83.6% (95%CI, 74.5-89.7%) vs 47.2% (95%CI,15.0-74.3%) (P=0.006) in PET-2-ve compared to 63.3% (95%CI, 46.4-76.1%) vs 42.2% (95%CI, 11.9-70.4%) (P=0.024) in PET-2 + . The same figures for 3-year OS were as follows: 92.1% (95%CI, 84.1-96.2%) vs 79.6% (95%CI, 37.1-94.9%) (P=0.089) in PET-2-ve compared to 92.2% (95%CI, 77.6-97.4%) vs 51.4% (95%CI, 16.0-78.6%) (P=0.007) in PET-2 + .

In Cox regression multivariable analysis the need of more than 2 lines of salvage therapy before ASCT (HR 3.09, P=0.008), RT before ASCT (HR 3.10, P=0.036) and year of ASCT before 2013 (vs. 2017-2021: HR 2.48, P=0.043) were independent adverse prognostic factors for PFS. PET-2+ at front-line treatment (HR 1.83, P=0.083) and anemia at relapse or progression (HR 1.97, P=0.11) seemed associated with increased risk of failing ASCT, although they did not reach statistical significance.

Table 1

Patient characteristics

PET-2 negative (%)

PET-2 positive (%)

Total

125

59

Gender: Male/Female

64/61 (51/49)

28/31 (47/53)

Median age at relapse or progression, years (range)

34 (19-66)

34 (18-68)

Biopsy performed

84 (69)

24 (42)

Refractory disease

47 (38)

45 (76)

Early relapse (<12 months from end of first-line chemotherapy)

34 (27)

11 (19)

Late relapse (≥12 months from end of first-line chemotherapy)

44 (35)

3 (5)

Median time from end of first-line chemotherapy and relapse or progression, months (range)

7 (0-116)

2 (0-18)

Stage I-II/III-IV

71/54 (57/43)

33/26 (56/44)

B Symptoms

30 (24)

16 (27)

Extranodal involvement

33 (26)

14 (24)

Large nodal mass

15 (12)

7 (12)

ECOG PS ≤1 / > 1

111/14 (89/11)

54/5 (92/8)

Anemia

11 (9)

13 (22)

First salvage regimen: BeGEV

43 (34)

8 (14)

IGEV

42 (34)

20 (34)

Brentuximab Vedotin + /-Bendamustine

7 (6)

14 (24)

DHAP/DHAOX

21 (17)

14 (24)

Others

12 (10)

3 (5)

Conclusions: Good outcomes were documented in patients with relapse or progression after a PET-guided first-line therapy. Favorable prognostic factors were ≤ 2 salvage therapy lines, no need for salvage RT before ASCT and having received ASCT after 2013.

PET-2+ at front-line treatment was associated with significantly increased risk of failing ASCT in univariate analysis, although its value did not reach an actual statistical significance in multivariate analysis.

Disclosure: Authors do not report any conflict of interest in the present study.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P484 REDUCED RELAPSE INCIDENCE AFTER CHECK-POINT INHIBITORS RELATIVE TO BRENTUXIMAB VEDOTIN AS SALVAGE THERAPY BEFORE ALLOGENEIC STEM CELL TRANSPLANTATION FOR REFRACTORY/RELAPSED HODGKIN LYMPHOMA: A RETROSPECTIVE ANALYSIS

Jacopo Mariotti 1, Chiara Pinton1, Chiara de Philippis1, Daniele Mannina1, Barbara Sarina1, Daniela Taurino1, Armando Santoro1, Stefania Bramanti1

1IRCCS Humanitas Research Hospital, Rozzano (milan), Italy

Background: The outcome of patients with Hodgkin lymphoma either refractory to several lines of therapy or relapsing after autologous stem cell transplantation (R/R HL) is dismal and the only potential curative strategy is represented by allogeneic stem cell transplantation (Allo-SCT). Check point inhibitors (CPI) and anti-CD30 drug conjugate (brentuximab vedotin) represent the most common used salvage therapies for this subset of patients. In particular, CPI rescue before Allo-SCT was associated with a notable low incidence of relapse (18%) (Merryman RW, 2021), but also with a not negligeable incidence of acute and chronic graft-versus-host-disease (GVHD) that might hamper its beneficial graft versus tumor effect. Chen et al (2014) have shown a favorable outcome for patients undergoing Allo-SCT after bretuximab vedotin (BV) rescue, but this was not confirmed in a recent retrospective analysis by the EBMT (Barzarbachi, 2018). Given this background, we asked what is the most effective salvage regimen for R/R HL candidate to Allo-SCT.

Methods: From 30/4/2014 to 30/8/2022, 74 consecutive patients with R/R HL were treated with Allo-SCT either after CPI (N=46) or BV (n=28) at our center. The main objective of this retrospective study was to identify which rescue strategy was more beneficial in terms of efficacy, evaluated as post-transplant cumulative incidence of relapse (CIR), and safety, evaluated in terms of non-relapse mortality (NRM) and GVHD. Of note, patients receiving CPI may have received a previous treatment with BV: these subjects (=29) were enumrated in the cohort receiving CPI.

Results: The two cohorts were similar in terms of patients (sex, age, HCT-CI), disease (pre-transplant status, previous autologous transplant) and transplant characteristics (donor/recipient CMV serostatus, sex combination, graft source, donor type, conditioning regimen). Patients receiving CPI had received more lines of therapy compared with the BV cohort (median 6 vs 4, p<0.001) before Allo-SCT. With a median follow-up of 37,4 months, 2 patients had relapsed in the CPI cohort vs 9 in th BV group at a median of 7,8 months after transplant. CIR was significantly redcued in the CPI cohort vs the BV group: at 4 years, 5% vs 46% (p<0.001), respectively. NRM was higher after CPI salvage relative to BV, but did not reach statistical significance: at 4 years, 21% vs 7% (p=0.197). 6-months cumulative incidence of grade 2-4 and 3-4 acute GVHD were not different: 46% vs 32% (p=0.325) and 9% vs 4% (p=0.398), respectively. Moderate severe chronic GVHD was slightly higher in the CPI group: at 4 years, 15% vs 4% (p=0.220), respectively. Of note, GVHD/relapse free survival (GRFS) was again similar among the two groups: at 4 years, 55% vs 56% (p=0.965).

Conclusions: HL response achieved after CPI was more durable after Allo-SCT relative to BV. Nevertheless, pre-transplant treatment with CPI was associated with a slight, but not significant, increase of toxicity. CPI is a desirable pre-transplant salvage therapy relative to BV, but management of post-transplant side effects needs further improvement.

Disclosure: The authors have no conflict of interest to disclose.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P485 LOW NON-RELAPSE MORTALITY ASSOCIATED WITH ALLOGENEIC HAEMATOPOIETIC CELL TRANSPLANTATION AFTER CAR-T FAILURE IN PATIENTS AFFECTED BY LARGE B CELL LYMPHOMA

Chiara de Philippis1, Massimiliano Gambella2, Jacopo Mariotti 1, Anna Dodero3, Patrizia Chiusolo4, Alessia Castellino5, Laura Giordano6, Barbara Sarina1, Daniela Taurino1, Daniele Mannina1, Anna Maria Raiola2, Simona Sica4, Carmelo Carlo-Stella1, Emanuele Angelucci2, Armando Santoro1, Paolo Corradini3, Stefania Bramanti1

1IRCCS Humanitas Research Hospital, Rozzano, Italy, 2IRCCS Ospedale Policlinico San Martino, Genova, Italy, 3Hematology and Stem Cell Transplantation, Fondazione IRCCS Istituto Nazionale dei Tumori, Milano, Italy, 4Fondazione Policlinico A. Gemelli IRCCS, Roma, Italy, 5Santa Croce e Carle Hospital, Cuneo, Italy, 6IRCCS Humanitas Research Hospital- Humanitas Cancer Center, Rozzano, Italy

Background: Anti-CD19 chimeric antigen receptor (CAR) T-cells represent a major advance in the treatment of relapsed/refractory aggressive B-cell lymphomas. However, a significant number of patients experience failure and their prognosis is very poor. Previous reports highlight a possibly high toxicity rate, in terms of non-relapse mortality (NRM) and hepatic toxicities after allogeneic hematopoietic cell transplantation (allo-HCT) post CAR-T failure (1,2). The objective of the present study was to evaluate the role of allo-HCT after CAR-T therapy in Large B Cell Lymphomas patients.

Methods: We retrospectively analysed the outcome of 17 consecutive patients affected by Diffuse Large B Cell Lymphomas and Primary Mediastinal B Cell Lymphomas undergoing allo-HCT after CAR-T cell failure at 5 Italian centers between May 2020 and August 2023 in order to evaluate outcomes in terms of overall survival (OS), progression free survival (PFS), NRM and transplant specific complications.

Results: Patients’ characteristics are listed in Table 1. Median time from relapse post CAR-T to transplant was 6.2 months (range, 1.8-22.2). The median number of lines of therapy between CAR-T infusion and allo-HCT was one (range, 1-3). Bridge therapies to allo-HCT were glofitamab in 7 patients, lenalidomide in 4 patients, chemotherapy in 3 patients, radiotherapy in 1 patient, brentuximab + nivolumab in 1 patient and R-polatuzumab bendamustine in 1 patient. Disease status at allo-HCT was complete response in 60%, partial response in 23%, and progressive/stable disease in 17%. After a median follow-up of 23.4 months (range 4-35.9), the 6-mounths cumulative incidence (CI) of acute GVHD grade 2-4 was 32.5%, whereas the 1-year CI of moderate to severe chronic GVHD was 29.9%. No patients developed neither sinusoidal obstruction syndrome nor thrombothic microangiopathies. Median time to neutrophil recovery was 17 days (range, 14-28) and median time to platelet recovery was 19 days (range, 13-34). No patients experienced primary graft failure.

The 1- year CI of non-relapse mortality was 13.4% and the 1-year CI of relapse was 32.8%. The 1-year OS and PFS were 73.3% and 53.8%.

Patients characteristics

N 17 (%)

Median age (range)

54 years (24-69)

Disease

Diffuse Large B Cell Lymphoma

14 (82)

Primary Mediastinal B Cell Lymphoma

3 (18)

Previous CAR-T

Tisa-cel

8 (47)

Axi-cel

9 (53)

Previous auto stem cell transplantation

4 (23)

Previous lines of therapy (range)

5 (4-8)

Donor type

Identical sibling

1 (5)

Matched unrelated

5 (30)

Haploidentical

11 (65)

Stem cells source

Bone Marrow

6 (35)

Peripheral Blood Stem Cells

11 (65)

Conditioning regimen

Non myeloablative

10 (59)

Reduced toxicity

7 (41)

GVHD prophylaxis

PTCY, CSA and MMF

14 (82)

CSA, MTX, ATG

2 (12)

CSA, MTX, MMF

1 (6)

Conclusions: Our data suggest that allo-HCT after anti-CD19 CAR-T treatment failure is feasible with no signal of increased GVHD or other unexpected complications, in particular no hepatic toxicities were reported and the NRM at 1 year was lower than previously reported data. One-year allo-HCT outcomes following CAR T failure produced similar outcomes to what has been previously observed in LBCL with allo-HCT. More long-term follow-up is needed to confirm the durable remission rate and curative potential of allo-HCT after CAR T failure.

Disclosure: none.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P486 TREOSULFAN-BASED CONDITIONING AND SIROLIMUS-PTCY GVHD PROPHYLAXIS IN ALLOGENEIC STEM CELL TRANSPLANTATION FOR AGGRESSIVE B-CELL NHL

Lorenzo Lazzari 1, Alessandro Bruno1, Simona Piemontese1, Federico Erbella1, Piera Angelillo1, Maria Teresa Lupo Stanghellini1, Andrés Ferreri1,2, Fabio Ciceri1,2, Raffaella Greco1, Jacopo Peccatori1

1IRCCS San Raffaele Scientific Institute, Milan, Italy, 2Vita-Salute San Raffaele University, Milan, Italy

Background: Since the advent of novel therapies, such as CAR-T cells, allogeneic stem cell transplantation (alloSCT) is progressively less considered as a therapeutic option in patients (pts) with aggressive relapsed/refractory B-cell NHL due to excessive morbidity and mortality associated with the procedure. Recently, improvements in conditioning regimens led to a reduction in TRM, while PTCy has been widely and successfully used across different alloSCT settings.

Methods: Between June 2005 and December 2022, 40 heavily pretreated adult pts affected by B-cell NHL, namely DLBCL, PMBCL, and MCL, underwent first alloSCT at our Center using a treosulfan-based conditioning. Of these, 26 pts received sirolimus and PTCy (Sir-PTCy)-based GvHD prophylaxis and were considered for the primary analysis. Conditioning regimen consisted of treosulfan (TD 42 g/m2) and fludarabine (TD 150 mg/m2) in 14 pts (transplant conditioning intensity [TCI] intermediate); intensification with melphalan (TD 140 mg/m2) was applied on the remaining twelve (TCI high). We considered as a control group the 14 pts who mainly received standard MTX-based GvHD prophylaxis, with addition of ATG in 11 cases. Primary endpoint of the study was OS in the Sir-PTCy group.

Results: Pts and transplant characteristics are displayed in Table 1. Three pts received MRD, 14 MUD (with 2 MMUD), and 9 a MMRD unmanipulated peripheral blood stem cell alloSCT. At transplant, 17 pts were in CR, 6 were in PR, and 2 had PD. Two pts (1 PMCBL, 1 MCL) received a previous CAR-T cell therapy. Median follow-up was 41 months (range, 7-113). Median time to neutrophil and platelet engraftment was 19 (range, 12-42) and 22 days (range, 10-55), respectively; no graft failure was observed. OS and DFS at 3 years were 78.6% and 66.9%, respectively. One and 3-year relapse incidence was 3.8% and 17.7%, respectively. TRM was 7.7% at 100 days, 15.4% at 1 year and for the entire follow-up; overall, 4 pts died due to infections (1 CMV pneumonia, 1 COVID-19, and 2 bacteremia) and one from disease relapse. The 100-day cumulative incidence (CI) of acute GvHD grade≥2 and grade≥3 was 15.4% and 7.7%, respectively; 3-year CI of moderate and severe chronic GvHD was 41.8% and 4.6%, respectively. Undergoing alloSCT in CR projected better 3-year outcomes in terms of OS (94.1% vs 50%, p=0.01), DFS (72.8% vs 55.6%, p=0.09), and TRM (5.9% vs 33.3%, p=0.06). No statistical differences were found when pts were stratified according to disease subtype, donor type, HCT-CI, and intensity of conditioning. Seventeen pts are alive and disease-free at last follow-up, with only one pts on ongoing immunosuppression for GvHD. Compared to our control group, pts receiving Sir-PTCy displayed a significantly better OS (78.6% vs 35.7%, p<0.01; Figure 1) and DFS (66.9% vs 28.6%, p<0.01) at 3 years.

Sir-PTCy group (N=26)

MTX-based group (N=14)

P

Age at diagnosis, median year (range)

53 (28-65)

49 (28-63)

0.2

Age at alloSCT, median year (range)

55 (29-69)

51 (30-65)

0.3

Female sex, number (%)

7 (27%)

8 (57%)

0.08

Previous lines of therapy, median (range)

3 (2-4)

3 (2-4)

0.9

Previous autoSCT, number (%)

20 (77%)

13 (93%)

0.07

Disease subtype, number (%)

0.03

DLBCL

8 (30%)

10 (71%)

PMBCL

4 (15%)

-

MCL

14 (54%)

4 (29%)

Disease status at alloSCT, number (%)

0.3

CR

17 (65%)

6 (43%)

PR

6 (23%)

4 (29%)

SD

-

2 (14%)

PD

3 (12%)

2 (14%)

HCT-CI score, median (range)

2 (0-5)

2 (0-4)*

0.3

Time diagnosis to alloSCT, median months (range)

76 (25-416)

56 (21-222)

0.3

Donor type, number (%)

0.3

MRD

3 (12%)

3 (21%)

MUD

12 (46%)

6 (43%)

MMUD

2 (8%)

2 (14%)

MMRD

9 (34%)

3 (21%)

Intensity of conditioning, number (%)

0.08

TCI intermediate

14 (54%)

12 (86%)

TCI high

12 (46%)

2 (14%)

GvHD prophylaxis, number (%)

<0.001

PTCy+Sirolimus

3 (11%)

-

PTCy+Sirolimus+MMF

21 (81%)

-

PTCy+Sirolimus+MMF + ATG

2 (8%)

-

MTX + CSA

-

3 (21%)

MTX + CSA + ATG

-

7 (50%)

MTX+Sirolimus+ATG

-

4 (29%)

Infused CD34 + /Kg x106, median (range)

5,82 (2,98-7,08)

7,5 (5,27-12,24)

0.001

Infused CD3 + /Kg x108, median (range)

1,98 (0,72-3,65)

3,1 (1,31-6,89)

0.01

* 6 missing

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (55)

Conclusions: AlloSCT using a treosulfan-based conditioning combined with Sir-PTCy GvHD prophylaxis is safe and effective in the treatment of advanced B-cell NHL. Chronic GvHD was not negligible but treatable in almost all cases. Selected pts with DLBCL, PMBCL, and MCL may still benefit from the inclusion of alloSCT in their treatment algorithm.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P487 OUTCOME OF HIGH-DOSE CHEMOTHERAPY FOLLOWED BY AUTOLOGOUS HEMATOPOIETIC CELL TRANSPLANTATION IN PATIENTS WITH PERIPHERAL T-CELL LYMPHOMA AT A SINGLE INSTITUTION OVER 20 YEARS

Joanna Romejko-Jarosińska 1, Ewa Paszkiewicz-Kozik1, Marcin Szymanski1, Lidia Poplawska1, Anna Borawska1, Anna Dabrowska-Iwanicka1, Katarzyna Domanska-Czyz1, Agnieszka Druzd-Sitek1, Robert Konecki1, Martyna Kotarska1, Ewa Mroz-Zycińska1, Wlodzimierz Osiadacz1, Beata Ostrowska1, Monika Swierkowska1, Joanna Tajer1, Lukasz Targonski1, Elzbieta Wojciechowska-Lampka1, Jan Walewski1

1National Research Institute of Oncology, Warszawa, Poland

Background: Peripheral T cell lymphomas (PTCL) are rare, heterogenous group of diseases derived from mature T cells. The regimens such as CHOP or CHOP-like are usually employed in induction therapy. High dose chemotherapy followed by autologous hematopoietic cell transplantation (auto-HCT) is used to consolidate induction therapy or as standard treatment in relapsed refractory patients. Here we present the data of 79 consecutive PTCL patients, who underwent auto-HCT after the first or next lines of therapy at our centre between 2000-2022.

Methods: Our group includes patients with anaplastic large cell lymphoma (ALCL) ALK+ (n=11), ALK– (n=11), PTCL, not otherwise specified (NOS) (n=29), angioimmunoblastic T-cell lymphoma (n=11), subcutaneous panniculitis-like T-cell lymphoma (n=6), enteropathy-associated T-cell lymphoma (n=4), NK/T-cell subtype (n=6) and adult T-cell leukemia/lymphoma (n=1). Median (range) age at transplantation was 46 (19-66) years. At the time of diagnosis, 61% (n=48) of patients had performance status (PS) 0-1, 85% of patients (n=67) clinical stage III/IV disease, 63%- extranodal disease (n=50) and 34%-bone marrow involvement (n=27). IPI0-1, IPI2, IPI 3-5 scores were found in 29% (n=23), 29% (n=23), 42% (n=33) of patients, respectively. Complete remission (CR) at auto-HCT was confirmed in 57% of patients (n=45). Auto-HCT was performed after induction therapy in 47% of patients (n=37). Median time from diagnosis to auto-HCT was 9 months (range:4-71).

Results: At median follow up of 48 months (range: 4-275) from auto-HCT, the 5 year progression free survival (PFS) and overall survival (OS) were 50% (95%C.I.:39%, 61%) and 60% (95%C.I.:48%, 72%) respectively. Relapse after auto-HCT was confirmed in 50% of patients (n=40) with median time to relapse of 7 months (range:1-130). There were no significant differences in OS and PFS between ALCL and other PTCL subtypes; median OS: 75 vs 44 months, p=0.29 and median PFS: 24 vs 21 months, p=0.47, respectively. Number of lines prior auto-HCT did not have an impact on PFS and OS (p=0.8). Lack of CR at transplantation, IPI>1, CS III/IV were adverse risk factors for PFS and OS in univariate analysis. PS > 1 was unfavourable for OS. No CR at auto-HCT (Hazard ratio (HR):1.35, p=0.006), IPI>1 (HR:2.77, p=0.01), CS III/IV (HR 3.28, p=0.05) were adverse risk factors for PFS in multivariate analysis. IPI>1 (HR:2.56, p=0.01) and CSIII/IV (HR:3.05, p=0.04) was adverse factors for OS in multivariate analysis. The 5 year PFS and OS for CR versus no CR at transplantation were 62% vs 31%, (p=0.006) and 70% vs 47% (p=0.08), respectively. Only 65% of patients who relapsed after auto-HCT underwent other treatment (median treatment lines:2). Transplantation-related mortality did not occurred. Forty six percent (n=36) of patients died: due to progression (n=32) and due to other reasons (n=4), 54% of patients is still alive; in CR (n=35) and continuing treatment (n=8).

Conclusions: In conclusion: in our group more than 50% of PTCL patients who underwent auto-HCT achieved long-term remission, regardless of the number of previous treatment lines. We did not observe the impact of the subtype of PTCL on survival. Low clinical stage, IPI 0-1 and CR prior to transplantation were favourable factors for prolonged survival.

Disclosure: no conflict of interest.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P488 IMPROVING RESULTS OF ALLO-HCT FOR PATIENTS WITH RELAPSED/REFRACTORY HODGKIN LYMPHOMA. A SINGLE CENTER EXPERIENCE

Andrzej Frankiewicz 1, Małgorzata Ociepa-Wasilkowska1, Tomasz Czerw1, Monika Dzierżak-Mietła1, Magdalena Głowala-Kosińska1, Jerzy Hołowiecki1, Małgorzata Krawczy-Kuliś1, Włodzimierz Mendrek1, Iwona Mitrus1, Jacek Najda1, Maria Saduś-Wojciechowska1, Małgorzata Sobczyk-Kruszelnicka1, Sebastian Giebel1

1Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Gliwice, Poland

Background: Despite increasing efficacy of first line treatment of Hodgkin lymphoma (HL) and availability of various salvage regimens, there are still patients for whom allogeneic hematopoietic cell transplantation (allo-HCT) offers the only chance of cure. Historically, allo-HCT for HL was associated with high incidence of disease progression (PI) and high non-relapse mortality (NRM). The goal of this study was to analyze if the outcomes changed over time.

Methods: This was a single center retrospective study including all subsequent patients with relapsed/refractory (R/R) HL treated with allo-HCT between years 2012-2023. Results in the period 2012-2018 (group 1, n=47) were compared to more recent one i.e. 2019-2023 (group 2, n=37). Median recipient age was 35 (19-65) years. Allo-HCT was performed after a median 5 (2-8) lines of systemic therapy, including brentuximab vedotin (BV, 64%), nivolumab (32%), radiotherapy (52%) and autologous-HCT (77%). Disease status at allo-HCT was as follows: CR (43%), PR (20%), stable or progressive disease (37%). Proportion of patients in CR did not differ for study group 1 and 2 (40% and 43%, respectively). Transplantations were performed using either matched sibling (51%), unrelated (36%) or haploidentical (13%) donor. Post-transplant cyclophosphamide (PTCy) was used as a backbone of immunosuppression in 25% of cases.

Results: With the median follow-up of 30 months the probability of overall survival (OS) and progression-free survival (PFS) at 3 years for the whole study population was 63% ( + /-6) and 58% ( + /-6), respectively. In univariate analysis the probability of PFS at 2 years was significantly higher for group 2 compared to group 1 (77% vs. 49%, p=0.03; Figure 1). There was also a tendency towards increased OS (80% vs. 60%, respectively; p=0,13). PI at 2 years was reduced for group 2 compared to group 1 (7% vs. 31%, p=0.01) while no significant difference was observed for NRM (16% vs. 20%, respectively, p=0.65). In the whole study population CR status at allo-HCT compared to ≥PR was associated with improved PFS (74% vs. 50%, p=0.01) and OS (81% vs. 58%, p=0.025) as well as reduced PI (12% vs. 30%, p=0.01). In multivariate analysis allo-HCT performed between 2019-2023 was associated with improved chance of PFS (HR=0.37, p=0.02), reduced risk of progression (HR=0.14, p=0.008) and a tendency to improved OS (HR=0.45, p=0.07). CR status at allo-HCT was associated with improved PFS (HR=0.36, p=0.008) and OS (HR=0.36, p=0.02) as well as reduced risk of progression (HR=0.18, p=0.007). None of other variables included in the analysis (age, preceding BV, nivolumab, lines of treatment, donor type, use of PTCy, type of conditioning) influenced the study end-points.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (56)

Conclusions: Results of allo-HCT improved significantly over time with approximately 80% chance of survival for procedures performed in recent years. CR status at transplantation is the strongest predictor of outcome. Effective salvage regimens appear critical to increase a chance of cure with the use of allo-HCT.

Disclosure: No disclosures of interest.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P489 THE SINGLE-CELL LANDSCAPE EXPLORING ABNORMAL T CELL STATES AND DEVELOPMENTAL TRAJECTORIES IN HETEROGENEOUS NON-HODGKIN LYMPHOMA

Yuqing Wang 1, Cong Wang1, Chanmin Xiao1, Zheng Wang1, Xi Zhang1

1Army Medical University, Xinqiao Hospital, Chongqing, China

Background: According to the data published by WHO in 2023, the mobility of lymphoma is six point six-one hundred thousands. Lymphoma has the highest morbidity among hematological malignancies in Eastern Morocco, Korea and China. Non-Hodgkin lymphoma is not a single disease, but a group of diverse subtypes that have different molecular drivers, clinical outcomes, and therapeutic options. The mortality of T cell lymphoma is high and the molecular classification is unclear. Cutaneous T cell lymphoma (CTCL) is one of common types of T cell lymphoma. Currently, the advancement of single cell RNA sequencing (scRNA-seq) enables us to gain deeper insights into global patterns of transcriptomic changes at a single-cell resolution, which helps us to catch possible driver pathogenic molecules and find novel therapeutic target. However, most of scRNA-seq data focuses on B cells in non-Hodgkin lymphoma, rather than T cells.

Methods: We integrated all of public scRNA-seq databases on non-Hodgkin lymphoma. We made quality control and integrated the datasets from 10 publications via Seurat. We did pseudotime analysis through Monocle 2 and Monocle 3. pySCENIC was used to do transcriptional factor enrichment analysis and Cell Chat was used for cell-cell communication analysis.

Results: We integrated 10 public datasets of non-Hodgkin lymphoma. Eleven types of cells were identified. Among these datasets, cells from CTCL are most. In order to clear out T cell states in the development of CTCL. Four development trajectories were simulated. For CD4 single positive (SP) T cells, Trajectory 1 is central memory T (Tcm)-1 to regular T cell (Treg), and Trajectory 2 is Tcm-2 – resident memory T cell (Trm) – CD7 loss T cell. For CD8 SP T cells, one trajectory is Tcm-1 – Tcm-2 – effector T cell (Teff)-2, and another trajectory is Tcm-1 – Tcm-2 – effector T cell (Teff)-2. Through four developmental trajectories, active signals and exhaustive signals simultaneously upregulated, leading to dysfunctional immune responses. Finally, IRF8, SPI1 and EZH2 were identified to play a significant role during T cell state transition and may be novel target for CTCL.

Conclusions: Both CD4 and CD8 T cells from malignant tissues exhibited the characteristics of high proliferation and high exhaustion, which may cause T cell dysfunction and lead to tumorigenesis. IRF8, SPI1 and EZH2 may be driver molecules in CTCL, which are potentially novel targets for therapy.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P490 BEYOND TRANSPLANTATION: BRENTUXIMAB’S IMPACT ON LONG-TERM OUTCOMES AFTER AUTOLOGOUS STEM CELL TRANSPLANT (ASCT) IN ROMANIAN TRANSPLANT CENTERS

Lavinia Eugenia Lipan1,2, Andrei Colita3,2, Angela Dascalescu4, Anca Colita1,2, Zsofia Varady1, Laura Diana Stefan1, Miruna Tirnovan1, Alina Daniela Tanase 1,2

1Fundeni Clinical Institute, Bucharest, Romania, 2University of Medicine and Pharmacy “Carol Davila”, Bucharest, Romania, 3Coltea Hospital, Bucharest, Romania, 4Oncologic Institute, Iasi, Romania

Background: Brentuximab Vedotin (BV), a monoclonal antibody designed to target CD30 expressing cells, has demonstrated efficacy as maintenance therapy, contributing to an enhanced progression-free survival (PFS) in patients with high-risk Hodgkin Lymphoma (HL).

Methods: A retrospective multicenter-study was conducted on behalf of Romanian Society of Bone Marrow Transplantation, between January 2014 and December 2022 and include 210 patients who underwent ASCT for HL in Romania (Oncologic Institute Iasi, Coltea Hospital and Fundeni Clinical Institute from Bucharest). Main Objective of this descriptive analysis was to evaluate the impact of BV maintenance therapy on PFS and OS in HL patients treated with ASCT. 151/ 210 patients were classified as high-risk HL, and 99 out of 151 received BV maintenance therapy after ASCT, and 4 patients were excluded from the study after transplant-related death. Specialized software was used for data analysis after obtaining the approval of the Ethical Committee regarding data access.

Results: 55% of the studied population was male, and regarding histopathological subtypes, 69% of patients were identified with nodular sclerosis and 23% with mixed cellularity subtype. Complete remission (CR) at time of ASCT was registered in 44% (n=44) of patients in the BV maintenance group and in 40% (n=19) of patients without BV maintenance therapy. Progressive disease (PD) was present in 11% (n=11) of patients in the BV maintenance group, and in 6% (n=3) of patients without BV maintenance therapy.

Regarding the conditioning regimen LEAM was preferred in the BV maintenance group (87% of patients), while BEAM was preferred in the no-BV maintenance group (63% of patients), but not in the BV-maintenance group (8% of patients). CLV was used in 21% of patients who did not receive BV maintenance therapy. Significantly better outcomes were seen with BV maintenance therapy use with a 95% OS at 3 years, compared with no BV maintenance group where OS at 3 years was 85% (p<0.0012). PFS at 3 years was 70% in BV maintenance group vs. 58% in no BV maintenance group (p=0.52). In patients with progressive disease before ASCT in the BV maintenance group, 6 out of 11 patients attained CR following ASCT, whereas none in the non-BV maintenance group achieved CR.

Conclusions: The results observed in our cohort abide to the ones reported in literature, with better outcomes for patients in the BV maintenance group after ASCT both on OS and PFS. BV as maintenance may improve the outcome after ASCT in pre-transplant progressive disease patients. Further studies are needed to evaluate if the pre-transplant BV have an influence on outcome in patients receiving maintenance after ASCT in high-risk patients / chemo-refractory patients.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P491 ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION FOR T CELL LYMPHOMAS: A RETROSPECTIVE ANALYSIS FROM THE POLISH LYMPHOMA RESEARCH GROUP (PLRG)

Malgorzata Sobczyk-Kruszelnicka 1, Tomasz Czerw1, Joanna Drozd-Sokołowska2, Patrycja Mensah-Glanowska3, Agnieszka Piekarska4, Jarosław Dybko5, Anna Łojko-Dankowska6, Anna Czyż7, Jan Maciej Zaucha4, Sebastian Giebel1

1Maria Skłodowska-Curie National Research Institute of Oncology, Gliwice, Poland, 2Medical University of Warsaw, Warsow, Poland, 3Jagiellonian University Medical College, Kraków, Poland, 4Medical University of Gdańsk, Gdańsk, Poland, 5Lower Silesian Oncology Center, Wrocław, Poland, 6Poznan Univerity of Medical Sciences, Poznań, Poland, 7Wroclaw Medical University, Wrocław, Poland

Background: T-cell lymphomas are often associated with a poor prognosis despite the use of conventional chemotherapy and autologous hematopoietic cell transplantation (auto-HCT). Therapeutic progress in this group of patients is not as dynamic as in B-cell lymphomas. In case of refractory or relapsed disease patients can benefit from allogenic HCT (allo-HCT).

Methods: We evaluated retrospectively data of 39 patients (pts) who underwent allo-HCT due to refractory disease (26 pts) or relapsed after auto-HCT (13 pts) between year 2012 and 2023. They were treated in 6 transplantation departments of PLRG. The analysis included 25 men and 24 women. The histopathological type was: PTLC NOS – 25 pts (64%), other – 14 pts (36%). The median time from diagnosis to transplantation was 19, range 5-183 months. At the time of diagnosis 25 pts had the stage IV of the disease and general symptoms. The median age at allo-HCT was 46 years (19-65) and the disease stage was remission – 17 pts, active disease – 22 pts.

The donor type was HLA-identical sibling in 14, unrelated in 22 and haploidentical in 3 cases. The conditioning regimens were diverse: 79% regimens were myeloablative, 69% of them included TBI (dose 8 – 10 Gy). The immunosuppressive therapy consisted of cyclosporine + methotrexate in the vast majority of pts (77%). The median follow-up was 10 (1-145) months.

Results: All patients engrafted with median time of neutrophil and platelet recovery of 17 and 13 days, respectively. Twenty patients (51%) experienced acute GvHD (6 pts with grade 3-4). The incidence of chronic GvHD was 25% (10pts; mild – 3 pts, moderate – 4pts, severe – 3 pts).

The incidence of grade 4 infectious and non-infectious complications (CTCAE v. 5) was 5% and 8.

%, respectively. Fifteen patients died, all in the first year after transplantation. The cause of death was: relapse of the disease – 4 pts, transplant complications – 10 pts, other – 1 pts. The incidence of progression and TRM was 10% and 28%, respectively. The probability of PFS and OS at 36 months was 61% ( + /-8%). The probability of OS in patients transplanted in active disease after 36 months was 56% ( + /-11%).

Conclusions: Allo-HCT may offer a potential way of cure for patients with relapsed or refractory t-cell lymphomas. This analysis showed that the outcomes of patients transplanted without remission are only slightly worse than those treated in remission. Further research is needed to improve outcomes in this population of patients.

Clinical Trial Registry: no.

Disclosure: none.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P492 OPTIMIZING OUTCOMES: THE VITAL SIGNIFICANCE OF TRANSPLANTATION IN RESCUING PATIENTS DISCONTENTED WITH INITIAL PERIPHERAL T-CELL LYMPHOMA TREATMENT

Hongye Gao1, Zhuoxin Zhang1, Jiali Wang1, Yannan Jia2, Hao Zhang3, Xin Du4, Xianmin Song2, Yao Liu5, Dehui Zou1, Erlie Jiang 1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China, 2Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China, 3Affiliated Hospital of Jining Medical University, Jining, China, 4Shenzhen Bone Marrow Transplantation Public Service Platform, Shenzhen Second People’s Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China, 5Chongqing University Cancer Hospital, Chongqing, China

Background: Approximately 30%-50% of patients with Peripheral T-cell Lymphoma (PTCL) experience treatment failure during the initial stages, characterized by disease stability (SD), progression (PD), and dissatisfaction with partial response (PR). Salvage therapy with alternative regimens becomes imperative for these individuals. Subsequent treatment modalities, particularly in cases where transplantation is omitted (‘missing transplant’), remains unclear in terms of survival outcomes.

Methods: We collected data from five medical centers in China on patients with PTCL, confirmed through pathology reviews. Unsatisfactory initial treatment, defined by disease stability (SD), disease progression (PD), or partial response (PR) necessitating second-line treatment based on CT or PET/CT evaluations. Propensity score matching methods were employed across three groups using the ‘pm3’ package in R to mitigate biases and ensure a robust data analysis.

Results: The study enrolled 221 patients dissatisfied with their initial treatment, representing diverse subtypes, including AITL, ALCL, ENKTL, HSTCL, and PTCL-NOS. Over a median follow-up of 42.2 months (95% CI: 36.2-61.7), the entire cohort exhibited a 3-year overall survival (OS) of 30.5% (95% CI: 24.5%-37.9%). Treatment approaches included allo-HSCT for 33 patients, auto-HSCT for 26 patients, and chemotherapy or supportive treatment without transplant for 162 patients. The auto-HSCT group demonstrated the highest 3-year OS at 95.8% (95% CI: 88.2%-100%), followed by allo-HSCT at 45.7% (95% CI: 28.5%-73.4%). Patients without transplant experienced a notably lower 3-year OS of 15.9% (95% CI: 10.8%-23.3%). Addressing heterogeneity, a propensity score matching (PSM) model balanced baseline characteristics (pathology subtype, age, gender, first-line treatment response, and PIT risk stratification) across allo-HSCT, auto-HSCT, and chemo without transplant groups. After matching 13 patients in each group, the analysis revealed a significant HSCT benefit (p < 0.0001). In the high-risk group (IPI score ≥ 3), patients without transplant faced all-cause mortality within 1 year, while those with allo- or auto-HSCT achieved survival beyond 2 years. Median OS was unreached for auto-HSCT, 33.9 months for allo-HSCT, and only 2.95 months without HSCT. Even in the low-risk group (IPI score ≤ 2), patients demonstrated a survival benefit from HSCT. Among these patients, the 3-year OS was 100% for auto-HSCT, 66.7% for allo-HSCT, and only 33.3% for the chemotherapy group.

Conclusions: For PTCL patients dissatisfied with their initial treatment, HSCT is considered a crucial and indispensable salvage therapeutic intervention.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P493 LOW DOSE NIVOLUMAB IN COMBINATION WITH SALVAGE CHEMOTHERAPY BEFORE AUTO-HSCT IN PRIMARY REFRACTORY HODGKIN LYMPHOMA – A PILOT STUDY

Rakesh Reddy Boya 1, Pradeep Ventrapati1, Veni Prasanna Gedala1, Chandrasekhar Bendi2

1Apollo Cancer Centres, Visakhapatnam, India, 2Mahatma Gandhi Cancer Institute, Visakhapatnam, India

Background: Auto-HSCT is considered a standard of care for patients with relapsed/refractory classical HL. It is desirable to achieve a complete remission prior to HSCT with an appropriate salvage regimen. A significant proportion of primary refractory HL (up to 50%) will not be eligible for auto HSCT due to lack of chemo sensitivity to Gemcitabine and/or platinum based salvage regimens. Brentuximab based combinations and checkpoint inhibitors (Nivolumab, Pembrolizumab) showed potential as an effective salvage but are offset by the high financial burden they impose on a resource constrained setting.

Methods: This is a pilot study in transplant eligible patients of primary refractory HL in 2 south Indian cancer centres. A salvage regimen of low dose Nivolumab (fixed dose of 40mg – termed Nivo40) was used in combination with Gemcitabine and/or platinum based chemo (GDP/ICE/BEGEV) was assessed for efficacy before transplant. The measure of efficacy of salvage regimen was CR rate with three cycles and proportion of patients who can be taken up for HSCT.

Results: A total of ten patients were enrolled between June 2021 and May 2023. All patients had ABVD as initial treatment regimen with two patients escalated to BEACOPP after interim-PET suboptimal response. Three of the patients had progressive disease and rest of seven had residual active disease as disease status before salvage regimen. The salvage regimen used was Nivo40-ICE (n=4), nivo40-GDP (n=4), Nivo40-BEGEV (n=2). Five each of the patients had got the salvage as 2nd and 3rd lines of treatment, respectively. All ten patients had a response with this salvage strategy with complete remission in six patients (CR rate of 60%) and PR in rest of the four. None of the patients developed immune related AE.

HSCT with BEAM conditioning was undertaken for all the ten patients. One patient was lost due to transplant related mortality (neutropenic sepsis). Median follow up post HSCT was 20 months (range 6-30 months). Three of the nine patients who survived transplant period had relapse (at 6, 12 and 13 months) post HSCT. Two of the patients with relapse had PR as pre-HSCT disease status.

The average cost of salvage regimen was approximately two lakh rupees (2200 Euros).

Table1: Key Characteristics of the study population (excluding patient who had TRM)

s. no.

age

sex

line of treatment in which salvage was given

salvage regimen

disease status prior to HSCT

duration of follow up post HSCT

Relapse, Y/N

(If yes, duration of months post HSCT)

Treatment given at relapse

1

22

M

2

Nivo40-ICE

CR

30

N

2

25

M

2

Nivo40-ICE

CR

25

Y (13 mo)

single cervical node - RT given. continue to be in CR

3

10

M

3

Nivo40-BEGEV

PR

24

N

4

29

F

3

Nivo40-BEGEV

CR

22

N

5

36

M

3

Nivo40-GDP

PR

18

Y (12 mo)

Bendamustine base chemo

6

22

F

3

Nivo40-GDP

PR

13

Y (6 mo)

Brentuximab based - planning for Allo HSCT

7

11

M

3

Nivo40-GDP

CR

9

N

8

40

M

2

Nivo40-ICE

PR

6

N

9

10

F

2

Nivo40-ICE

CR

6

N

Conclusions: Low dose Nivolumab (Nivo40) with platinum and/or Gemcitabine based salvage chemo is an effective regimen in terms of efficacy and as a bridge to HSCT in primary refractory HL. It can be a low cost alternative to Brentuximab/ standard dose Nivolumab in patients in resource constrained settings.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P494 PREDICTORS OF DELAYED PLATELET ENGRAFTMENT AFTER AUTOLOGOUS STEM CELL TRANSPLANTATION IN NON-HODGKIN LYMPHOMA

Rita Costa e Sousa 1, Cátia Almeida1, João Gaião Santos1, Ricardo Ferreira1, Mafalda Urbano1, Maria Carolina Afonso1, Marília Gomes1, Adriana Roque1, Catarina Geraldes1

1Centro Hospitalar e Universitário de Coimbra, Coimbra, Portugal

Background: Delayed platelet engraftment (PE) is occasionally observed after autologous stem cell transplantation (ASCT) in non-Hodgkin lymphoma (NHL) patients (pts) and has been associated with worst outcomes mainly due to increased risk of life-threatening bleeding events. Data about clinical predictors of delayed PE following ASCT are scarce, and their identification is crucial for improvement of outcomes in these patients. The objective of this study was to identify risk factors for delayed PE post-ASCT, in pts with NHL.

Methods: Retrospective analysis of NHL pts, who underwent ASCT between 2007 and 2022, in a single center. Platelet engraftment (PE) was defined as a recovery of platelet count greater than 50×109/L without transfusion support for 3 consecutive days, and the first day of 3 consecutive days was regarded as the day of platelet engraftment. The influence of possible predictive factors for platelet engraftment was analyzed using a Gray test and Fine-Gray methods for univariate and multivariate analysis, respectively. All p-values were two-sided and p<0.05 was considered to be significant. Factors with a two-sided p<0.1 in univariate analysis were subjected to multivariate analysis. The cumulative incidence of platelet engraftment was calculated considering death as a competing risk.

Results: We included 146 pts, 68% male, median age at ASCT 54 years old (18-66), 93% B-lymphoma and 8% T or T/NK-lymphoma. At diagnosis, 57,4% (74/129) had stage III/IV, 64% (69/108) had extranodal involvement and 39% (38/98) had bone marrow involvement. The median pre-ASCT therapeutic lines were 2 (1-5), 11% had previous radiotherapy and 81% were in complete response. All pts received BEAM conditioning.

The median number of apheresis sessions was 1 (1-4), median platelet count in D-7 of ASCT was 168×109/L (46-450) and median infused CD34 + x106/kg was 2.76 (1.4-38.31). Median time for PE was 17 days (4-375), with median number of platelet transfusion units of 5 (0-35), 35 pts (18%) had not achieved PE at D + 60 and 7 (5%) died without platelet engraftment.

Univariate analysis revealed that number of therapeutic lines prior to ASCT (p=0.02), number of apheresis sessions (p=0.005), pre-transplant platelet count (p=0.005) and previous radiotherapy (p=0.03) were associated with time to platelet engraftment after ASCT. In multivariate analysis, only the number of therapeutic lines prior to ASCT ≥2 (HR 0.45 [95%CI 0.26-0.81]; p=0,007) and pre-ASCT platelet count >150×109/L (HR 1.75 [95%CI 1.17-2.61]; p=0.006) were independently associated with a longer time for PE after ASCT.

For a median follow-up of 70.6 months, median progression free survival (PFS) was 109.5 months and median overall survival (OS) was not reached. Time to PE was not associated with PFS (p=0.506) or OS (p=0.965).

In our cohort, a previous published score for predicting PE (Yamagushi et al. Leuk & Lymphoma 2019) was not associated with PE (p=0.2014).

Conclusions: In our population, the number of therapeutic lines before ASCT and low pre-transplant platelet count were the strongest predictors of delayed PE in NHL pts undergoing ASCT.

Disclosure: No conflicts of interest to report.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P495 NON-MYELOABLATIVE ALLOGENEIC TRANSPLANTATION AFTER TOTAL SKIN ELECTRON BEAM THERAPY (TSEBT) IN CUTANEOUS T-CELL LYMPHOMA. A SINGLE CENTER EXPERIENCE

Carlos De Miguel 1, Belén Navarro1, Irma Zapata1, Mercedes Hospital1, Guiomar Bautista1, Ana Bocanegra1, José Antonio García-Vela1, María Esther Martínez-Muñoz1, Silvia Monsalvo1, José Luis Bueno1, Isabel Salcedo1, Luis Gastón Roustan1, Jesús Romero1, Rafael Duarte1

1Hospital Universitario Puerta de Hierro, Majadahonda, Madrid, Spain

Background: Patients with refractory and advanced stage mycosis fungoides (MF) and Sézary syndrome (SS) have a poor prognosis with a life expectancy of less than 5 years. Allogeneic transplantation remains the only curative approach for these patients. Achieving control of the disease prior to transplant has been shown to increase the probability of long-term survival. Here, we report our experience with non-myeloablative allogeneic transplantation following total skin electron beam therapy (TSEBT) in our center during the last 5 years.

Methods: We retrospectively analyzed non-myeloablative allogeneic transplants after TSEBT in patients with cutaneous T-cell lymphoma (MF/SS) performed between July 2018 and July 2023 in our center.

Results: Seven non-myeloablative allogeneic transplants were included (1 MF, 6 SS); 71% men; median age 43 (24 - 56); 4 haploidentical, 2 matched related, 1 matched unrelated. The majority (86%) of the patients had stage IV disease and had received multiple prior systemic therapies (median 5; 3 - 6). All patients had in common the addition of TSEBT as part of the pre-transplant treatment using modified Standford technique with a total dose of 30 Gy in 24 sessions (1.25 Gy per session, 4 times per week). The “shadowed areas” (top of scalp, palms, soles, axillas, perineum and inframammary folds in women) received an additional dose with direct electron fields (boosts) of 10-15 Gy, depending on the degree of involvement (10 Gy at 500cGy per day (2 sessions) on disease-free areas and 15 Gy (3 sessions) in involved areas. Most of the patients (71%) received TSEBT immediately before transplant (1-2 months before conditioning). All the patients were in stable partial response (PR) before transplant. Conditioning was non-myeloablative in all cases (fludarabine in all, 3 with melphalan, 2 with busulfan and 2 with busulfan-cyclophosphamide). With a median post-transplant follow-up of 18 months (3-60), the 1 and 3 years overall survival (OS) were 100% and 86%, respectively. All patients achieved complete remission, most of them (86%) with undetectable minimal residual disease measured by flow cytometry. Transplant-related mortality (TRM) was 0% at 1 and 3 years after transplant. Two patients relapsed (28%), both at three months after transplant. One of them responded to donor lymphocyte infusion and one died from disease progression. The cumulative incidence of grade II-IV acute GVHD was 14% and the 2-year incidence of moderate-severe chronic GVHD was 0%. Only 1 patient (14%) had a cytomegalovirus infection and only 1 patient required ICU admission due to respiratory failure secondary to disease progression.

Conclusions: Non-myeloablative allogeneic transplant including skin-directed TSEBT is effective, well tolerated and potentially curative for patients with advanced stage MF/SS. Despite the low incidence of these diseases, more experience is necessary to confirm these data.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P496 ROLE OF AUTOLOGOUS STEM CELL TRANSPLANT IN PRIMARY AND SECONDARY CENTRAL NERVOUS SYSTEM LYMPHOMA – A SINGLE CENTER EXPERIENCE

Meredith Tan 1,2, Tertius Tuy1,2, Melinda Tan1,2, Chieh Hwee Ang1,2, Jeffrey Quek1,2, Than Hein1,2, Yunxin Chen1,2, Francesca Lim1,2, Chandramouli Nagarajan1,2, Jordan Hwang1, Jing Jing Lee1, Lalitha Krishnan1, Lyn Lee Wong1, Zi Jing Seng1, Yeow Tee Goh1,2, Yeh Ching Linn1,2, Colin Diong1,3, Yuh Shan Lee1,3, William Hwang1,2, Aloysius Ho1,2, Lawrence Ng1,2

1Singapore General Hospital, Singapore, Singapore, 2National Cancer Centre, Singapore, Singapore, 3Parkway Cancer Centre, Singapore, Singapore

Background: Introduction - Central Nervous System Lymphoma (CNSL), either primary or secondary, is an aggressive malignancy associated with inferior outcomes due to poor penetrance through the blood-brain barrier, impact on cognition, and heterogeneity of presentations1. For each group, consolidation with high dose chemotherapy followed by autologous stem cell transplant (ASCT) appear to have increased survival outcomes1,2,3.

Aim - To evaluate clinical outcomes of patients with primary and secondary CNSL, who underwent ASCT as either first-line consolidation or treatment for relapse or refractory disease.

Methods: 24 consecutive Primary and Secondary CNSL patients who underwent ASCT in Singapore General Hospital from January 2013 to August 2023 were retrospectively reviewed and analyzed.

Results: Twenty-four patients were included in this study comprising of 12 males and 12 females, with a median age at transplant of 57.5 years (28-70), of which 19 (79%) had an Eastern Cooperative Oncology Group (ECOG) Performance Status of 1 and below. Of the 24 patients, 8 (33.3%) were diagnosed with Primary CNSL, whilst 16 (66.7%) had Secondary CNSL. 19 (79.2%) patients had parenchymal involvement of the CNS, while the remaining 1 (4.2%), 2 (8.3%) and 2 (8.3%) patients had meningeal, ocular, and spinal involvement respectively. Of these 24 patients, 7 (29.2%) underwent ASCT as first-line consolidation, whilst 17 (70.8%) underwent ASCT as treatment to relapse or refractory (RR) disease, with 22 (91.5%) receiving TT-BCNU-Etoposide conditioning chemotherapy and 2 (8.3%) receiving TT-Busulfan-Cyclophosphamide conditioning chemotherapy. 13 (54.2%) patients had a disease status of Complete Response (CR) at the time of transplant, while 7 (29.2%), 3 (12.5%) and 1 (4.2%) patient were of Partial Response (PR), Stable Disease (SD) and Progressive disease (PD) status respectively.

At the median follow-up post transplant of 40 months (5-106), 11 (45.8%) patients relapsed, 6 (25%) demised from disease relapse, while 1 (4.2%) demised from treatment-related causes. The median progression-free survival (PFS) was 18 months (0.8-99) and median overall survival (OS) was not reached. The estimated 18-month overall survival was 73.3%. For primary CNSL, the median PFS and OS were both 9 months. For secondary CNSL, the median PFS and OS were 18 months and not reached, respectively. Patients (both primary and secondary CNSL) who underwent ASCT as first-line consolidation had an improved median PFS and OS compared to those who underwent ASCT at RR disease; not reached for both PFS and OS versus PFS and OS of 16 months (0.9-99) and 30 months (9-106), respectively.

Conclusions: Our analysis suggests that ASCT is a feasible option to consider for both primary and secondary CNSL, with relatively low toxicity whilst avoiding the toxicities associated with whole-brain irradiation. Secondary CNSL patients who underwent ASCT as first-line consolidation appear to have derived greatest benefit. Our study limitations are small sample size, heterogeneity of CNSL subtype and short duration of follow-up for certain subtypes. In the era of immune-cell therapy, sequencing, and selection in between ASCT and CAR-T cell therapy as consolidation for CNSL will be warranted for further evaluation.

Table 1: Study population baseline characteristics

Median (range)

n

Median age at transplant, years

57.5 (28-70)

24

ECOG

0-1 : 2

19 (79.2%) : 5 (20.8%)

Sex

Male : Female

12 (50%) : 12 (50%)

Diagnosis

Primary CNS Lymphoma

8 (33.3%)

Secondary CNS Lymphoma

16 (66.7%)

Site of CNS involvement

Parenchyma

19 (79.2%)

Meningeal / Ocular / Spinal

1 (4.2%) / 2 (8.3%) / 2 (8.3%)

BMT treatment stage

1st line consolidation

7 (29.2%)

RR

17 (70.8%)

Disease status at transplant

CR

13 (54.2%)

PR / SD / PD

7 (29.2%) / 3 (12.5%) / 1 (4.2%)

Treatment lines prior to transplant

MATRix

1 (1st line)

10 (RR)

RMVP/RMVP-AraC +/- RT

7 (1st line)

1 (RR)

Anthracycline-based

16 (1st line)

2 (RR)

RDHAP

0 (1st line)

7 (RR)

Others (CYVE/HiDAC/MTX/RT)

0 (1st line)

5 (RR)

Median lines of treatment prior to transplant

2 (1-5)

Conditioning chemo

TT/BCNU/Etoposide

22 (91.7%)

TT/Busulfan/Cyclophosphamide

2 (8.3%)

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P497 HIGH DOSE THERAPY FOLLOWED BY AUTOLOGOUS STEM CELL TRANSPLANTATION IN 236 PATIENTS WITH HODGKIN LYMPHOMA ON 25 YEARS PERIOD (1998-2022)

Sabrina Akhrouf 1, Hanane Bouarab1, Rihab Benouattas1, Farih Mehdid1, Nadia Rahmoun1, Mounira Baazizi1, Dina Ait ouali1, Nacera Ait Amer1, Farida Tensaout1, Rose Marie Hamladji1, Redhouane Ahmed Nacer1, Malek Benakli1

1Pierre and Marie Curie Center, Algiers, Algeria

Background: Classical Hodgkin’s lymphoma (cHL) has high rates of cure, but in 15% to 20% of general patients (pts) and between 35% and 40% of those in advanced stages, the disease will progress or will relapse after initial treatment. For pts refractory to first-line chemotherapy or who relapse after initial treatment, outcomes are considerably worse. Salvage high-dose chemotherapy (HDT) followed by autologous stem cell transplant (ASCT) is the standard of care.

Methods: A retrospective study was performed with data collected from patient charts. The analysis involved 236 cHL pts who were consecutively submitted to HDT followed by ASCT in a single institution from April 1998 to December 2022 (24 years). The median age is 27 years (13-64) and a sex-ratio of 1 (118M /118F). It is cHL Ann Arbor stage IV (98 pts), refractory and early relapse (86 pts), and late relapse (52 pts). Disease status at transplantation is: 124 pts in partial response (PR), 96 pts in complete response (CR) including 30 pts in unconfirmed CR (CRμ) and 16 pts in failure. Peripheral HSC was collected by cytapheresis after mobilization with G-CSF alone. The median rate CD34 + cells is 3.62 x 106/kg (0,94-16,8). Several HDT conditioning regimens were used: BEAM: 119 pts, BeEAM with Bendamustine: 100 pts, TUTSHKA (Busulphan, Cyclophosphamide): 10 pts and TEAM with Tiothepa : 07 pts). The median follow-up at 30/11/2023 is 91 months (10-290).

Results: The median time to graft (PNC > 0.5x109/l) was 12 days (08-25). Eight early deaths were observed including 7 infections, 1 pulmonary embolism and 1 heart failure (TRM: 3,3%). Of the 228 pts that can be evaluated in post-transplantation, 153 pts (67%) achieved a CR, 34 pts (15%) a CRμ, 35 pts (15,3%) achieved a PR and 6 pts achieved stable disease (2,7%). A relapse was observed in 55 pts (24%) after a median duration of 15 months (2-72). At 30 of November 2023, 160 pts (67,8%) are alive (153 in CR, 07 in PR), 63 pts (26,7%) died (45 pts from progression disease and other reason for 18 pts). Thirteen pts (5,5%) were lost to follow-up. The overall survival (OS) at 25 years is 63% and the event-free survival (EFS) is 58%.

Conclusions: Our retrospective analysis shows good outcomes in patients who had P/R disease. Patients can be cured with second-line salvage chemotherapy and with HDT followed by ASCT. The introduction of the novel agents, including antibody-drug conjugates and PD-1 blocking antibodies has transformed the management of cHL in multiple settings.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P498 NON-CRYOPRESERVED AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN HODGKIN LYMPHOMA. ELEVEN-YEAR SINGLE CENTRE EXPERIENCE FROM ORAN. ALGERIA

Nabil Yafour 1, Kamila Amani1, Nawel Bounoua1, Fatima Aoudia1, Amel Bendimerad1, Mohamed Amine Benaissa1, Nour El Houda Hassam1, Mounir Serradj1, Manel Maarouf1, Leila Charef1, Badra Enta Soltane1, Soufi Osmani1, Rachid Bouhass1, Abdessamad Arabi1, Mohamed Brahimi1, Mohamed Amine Bekadja1

1Établissement Hospitalier et Universitaire 1(er) Novembre 1954, Service d’Hématologie et de Thérapie Cellulaire, BP 4166 Ibn Rochd, Université d’Oran 1, Ahmed Ben Bella, Faculté de Médecine, Oran, Algeria

Background: Survival rates for patients (pts) with relapsed or refractory (R/R) Hodgkin lymphoma (HL) was significantly improved by autologous stem cell transplantation (ASCT). However, in the absence of post transplant maintenance treatment, approximately half of these pts relapse. The objective of this study was to evaluate the safety and effectiveness of non-cryopreserved ASCT in 1st line (consolidation), and in R/R HL pts.

Methods: This is a retrospective, single-center study. ASCT was performed as consolidation in advanced stage (III/IV) pts or in R/R HL pts. Mobilization was performed with G-CSF alone (filigrastim or lenogrsatim) at a dose of 15 μg/kg/day for 4 to 5 days. The minimum targeted dose of CD34+ at the end of the leukapheresis was 3x106 /kg. The graft was stored in a conventional blood bank refrigerator at 4°C until reinfusion on day 0. The viability of the hematopoietic stem cell was carried out at the end of apheresis, and before graft infusion. Intensification regimen were CBV (n=15 pts, 2009-2010), BEAM (n=45 pts, 2010-2016), Benda EAM, (n=5, 2015-2016), and EAM (n=41, 2016-2020) including a total dose of etoposide 800 mg/m2, aracytin 8g/m2, melphalan 140 mg/m2. All pts received G-CSF from day +5 until absolute neutrophil count >0.5 G/L. Antibacterial, antiviral and antifungal prophylaxis consisted of cotrimoxazole/sulfamethoxazol, aciclovir and fluconazole respectively. The primary endpoint was engraftment and transplant related mortality (TRM). Secondary endpoints were overall survival (OS) and progression-free survival (PFS). The probabilities of OS, PFS were estimated using the Kaplan Meier method and SPSS software.

Results: From 2009 to 2020, 106 HL pts underwent ASCT, including 53 pts (50%) in first line therapy as a consolidation, and 53 pts (50%) in R/R. The median age at transplant was 27 years (16-55 years). The number of therapeutic lines before ASCT was 2(1-5 lines). Pre-transplant status was complete remission (CR) in 102 pts (96%) and partial remission (PR) in 4 pts (4%).

The average number of apheresis was 2 (1-4), and that of CD34+ was 4.13 106/kg (1.41-21.05). Twelve pts (11%) required the use of G-CSF + Plerixafor (or copy) including 10 pts with R/R HL, and 02 pts with extended stages. The median time to neutrophil engraftment was 13 days (range 8–25 days), and the median time to platelet > 20 G/L was 13 days (range 3–42 days). Mucositis > grade 1 occurred in 71 pts (67 %). At day 100, the evaluation showed; CR in 99 pts (98%), PR in 2 pts (2%) and TRM in 5 pts (5%). At 13.6 years, median PFS and OS were 71% and 74% respectively. PFS and OS were 85% vs 55% (p=0.005), and 87% vs 57% (p=0.004) in pts who received ASCT as a consolidation and in R/R pts respectively.

Conclusions: Non cryopreserved ASCT is safety and effective in HL pts. First-line intensification may be a valid option in multi-visceral III-IV stage, particulary in the absence of post-transplant maintenance treatment, given the excellent results.

Disclosure: Authors declare no conflict of interest.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P499 AUTOLOGOUS AND ALLOGENEIC STEM CELL TRANSPLANTATION IN MATURE NK/T-CELL LYMPHOMA: A TWO-CENTER RETROSPECTIVE REAL-LIFE ANALYSIS

Michele Wieczorek 1, Giorgia Bonetto1, Beatrice Bugnotto2,1, Alessia Moioli2, Albana Lico1, Francesca Elice1, Marcello Riva1, Cristina Tecchio2, Mauro Krampera2, Carlo Borghero1, Alberto Tosetto1

1Hematology and Cell Therapy Unit, Ospedale “San Bortolo”, Vicenza, Italy, 2Hematology and Cell Therapy Unit, Verona University, Verona, Italy

Background: NK/T-cell lymphomas pose a unique therapeutic challenge as they are burdened with severe prognosis. Stem cells transplantation (SCT) is usually incorporated in treatment, often in first-line. Although it is considered to offer better outcomes, there is not yet a consensus on whether autologous or allogeneic SCT is more advantageous, or on the best timing, especially in relapsed disease or in aggressive subtypes. In this analysis we observed and compared the real-life benefit of transplant procedures in NK/T-cell lymphoma patients of two transplant centers.

Methods: We retrospectively collected data of adult patients affected by mature NK/T-cell lymphomas undergoing autologous or allogeneic SCT in 2 Bone Marrow Units of Northern Italy between Januanty 1st 2008 and December 1st 2023. The main endpoints were overall survival (OS), progression-free survival (PFS) and treatment-related complications.

Results: We identified 21 patients treated with autoSCT and 24 with alloSCT, with a global median follow up of 39 months and a PFS of 29 months. AutoSCT was performed in first-line treatment in 12 cases, in patients in complete (CR) or partial response (PR). The OS for the autoSCT group was 44 months, while the PFS was 10 months. At two years OS and PFS were 71% and 38% respectively. Thirteen (57%) patients relapsed after autoSCT: 5 underwent alloSCT, of which 4 obtained sustained CR and 1 died of acute graft-versus-host disease (GvHD); 4 obtained CR with standard treatment or in clinical trials; 4 died of disease progression. No patient was treated with sequential autoSCT and alloSCT without relapsing in-between. Patients undergoing alloSCT had a median OS and a PFS of 39 months, with a two-year OS and PFS of 67%. AlloSCT was administered from second-line treatment (9 cases - 38%) onward. At time of transplant 15 (63%) patients were in CR, while 4 (17%) had a stable of progressive disease. The donor was related in 5 (21%) cases, matched-unrelated for 12 (50%) and alternative donors (3 haploidentical and 5 with 1 mismatch) for the rest. Seventeen (71%) patients were affected by acute GvHD, severe in 4 cases, and 8 (33%) by chronic GvHD. Only 1 (4%) patient relapsed of systemic lymphoma after alloSCT, 2 patients relapsed of mycosis fungoides while sustaining remission of aggressive lymphoma and one of them restored RC after donor lymphocyte infusions. Six (25%) patients died: 2 of GvhD, 3 of infections, 1 of disease progression, with a cumulative transplant-related mortality of 21%. One patient developed treatment-related myelodysplastic syndrome.

No significant statistical difference was observed between OS or PFS in autoSCT and alloSCT.

Conclusions: In this retrospective series we could not demonstrate a clear advantage of one therapeutic choice. However, this analysis suggests that, although alloSCT is encumbered by higher rates of complications and mortality, it may offer longer remission and it should be considered as treatment for NK/T-cell lymphoma in eligible patients even without previous autoSCT. Prospective multicenter studies are needed, considering also the possibility of frontline alloSCT and focusing on life quality after transplant to propose the best care plan for such patients.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P500 PRETREATMENT WITH RITUXIMAB IS SUSPECTED TO INCREASE RISK FOR OCCURRENCE OF VENO-OCCLUSIVE DISEASE IN PATIENTS WITH AGGRESSIVE LYMPHOMA AFTER ALLOGENEIC TRANSPLANT

Ahmet Elmaagacli 1, Mathis Samuel Bittermann1, Farouk Dahmash1, Anju Singh1, Yana Shikova1, Vitaly Varyushkin1, Mathias Vierbuchen1, Hans Salwender1, Christian Jehn1

1AsklepiosKlinik St. Georg, Hamburg, Germany

Background: Veno-occlusive disease, also known as sinusoidal obstruction syndrome (VOD/SOS), is a potentially life-threatening complication of allogeneic transplantation (SCT) most commonly associated with high-intensity conditioning regimens and chemotherapies. Recently, gemtuzumab ozogamicin and Inotuzumab ozogamicin have been identified as high risk factors for VOD/SOS. Here we aimed to evaluate in this retrospective, mono-center study, if a pretreatment with rituximab is a potential risk factor for the occurrence of VOD/SOS in patients transplanted for aggressive lymphoma.

Methods: 60 patients transplanted for DLBCL who were pretreated as first line therapy with RCHOP and received DHAP as salvage therapy were compared to 23 patients with T-NHL receiving CHOP/CHOEP pretreatment as first line therapy and DHAP as salvage therapy. Patients characteristic are given in table 1. VOD/SOS was diagnosed only if the Baltimore criteria were fulfilled for adults with onset within the first 21 day after SCT, Bilirubin > 2mg/dL plus 2 or more of the following criteria: painful hepatomegaly, weigth gain > 5%, Ascites. All patients with VOD had received a defribrotide therapy.

Results: 11 of 60 patients with DLBCL who received a pretreatment with rituximab developed a VOD/SOS within 21 days after transplant while in none of the 23 patients with T-NHL a VOD/SOS was detected (p=0.03). Ten of 11 patients with VOD/SOS had a very severe grading according to the EBMT scale and died, while one patient with severe VOD/SOS survived. Multivariate analysis including LDH upper norm, gender, HLA-matched, high IPI >2, conditioning regimen with busulfan/cyclophosphamide/fludarabine (bu/cyc/flu), confirmed that rituximab was the only independent factor for the occurrence of VOD/SOS (ORR 34,7 (CI 95% 0,14 -867, p= 0,034). Conditioning with treosulfan/fludarabine (treo/flu) did not reduced the occurrence of VOD/SOS significantly compared to conditioning with bu/cyc/flu, (9,7% vs 15,4%, respectively). Estimates for OS was for treo/flu 57% for 4 years versus 41% for bu/cyc/flu. (n.s), while relapses/progress occurred in 55 % vs 39%, respectively (n.s.). TRM occurred after treo/flu conditioning in 17,5% vs 33,3% at 4 years (n.s.).

Conclusions: Here we report for the first time that patients with DLBCL who received CHOP-pretreatment with rituximab had a highly increased risk for VOD/SOS compared to patients with T-NHL treated with CHOP/CHOEP without rituximab. We suspect rituximab as a risk factor for the induction of VOD/SOS in patients after HSCT. More studies are required to confirm our findings.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P501 GENETIC PREDICTORS OF PROGNOSIS IN CHRONIC LYMPHOCYTIC LEUKEMIA: INSIGHTS FROM NEXT-GENERATION SEQUENCING

Emilia Jaskuła1,2, Anna Sobczyńska-Konefał2,1, Iga Jendrysik2, Marzena Wojtaszewska3, Monika Mordak-Domagała2, Krzysztof Suchnicki2, Mariola Sędzimirska2, Monika Jasek1, Lidia Karabon1, Jarosław Dybko 2

1L. Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wroclaw, Poland, 2Lower Silesian Oncology Center, Wroclaw, Poland, 3University Hospital in Rzeszow, Rzeszow, Poland

Background: Chronic Lymphocytic Leukemia (CLL) is distinguished by the clonal proliferation of B lymphocytes, marked by CD5 + , CD23 + , and CD19+ phenotypes. This heterogeneity is linked to various genetic abnormalities. Key diagnostic factors include cytogenetic alterations like del(13q), trisomy 12, del(11q), and del(17p), with the latter two associated with poorer outcomes. Recent advancements include Bruton’s tyrosine kinase (BTK) and B-cell lymphoma 2 (BCL-2) inhibitors. This has shifted focus to IGHV and TP53 mutations as prognostic markers. Consequently, in February 2023, the Lower Silesian Oncology Center initiated Next-Generation Sequencing (NGS) for CLL diagnostics.

Methods: This study involved 97 CLL patients (52 females, 45 males, median age 66.5 years) using NGS to assess IGHV and TP53 mutations. Concurrently, FISH and NGS identified the four prevalent cytogenetic aberrations. The cohort, spanning diagnoses from 2003 to 2023, included both prospective and retrospective patient samples. Techniques employed were LymphoTrackDx IGHV Leader Somatic Hypermutation Assay Panel – MiSeq, SureSeq CLL + CNV Panel for sequencing 13 CLL-related genes, and Laboratory Developed Tests (LDTs) conforming to ERIC standards.

Results:

  1. 1.

    Del(11q), chromosome 12 trisomy, del(13q) and del(17p) was found in 13%, 8%, 62% and 8% of patients, respectively.

  2. 2.

    NGS detected TP53 mutations in 19% (16 out of 83) of samples with VAF > 5%.

  3. 3.

    Common IGHV genes were IGHV1-69*01 and IGHV3-21*01 in 15% and 8% of patients, respectively. Dual IGHV clonality was observed in 9 patients.

  4. 4.

    55% had unmutated IGHV status (U-CLL), with 13 patients in aggressive CLL subsets (CLL#1, CLL#2, and CLL#6).

  5. 5.

    U-CLL patients had a median treatment-free survival (TFT) of 272 days versus 1002 days in patient with mutated IGHV status (M-CLL) (HR = 2.985; 95% CI: 1.745 to 5.107), independent of other genetic or cytogenetic alterations.

  6. 6.

    U-CLL with TP53 mutations showed reduced time to second therapy (TST) (median 201 days vs. 531 days in M-CLL, HR = 2.557; 95% CI: 1.260 to 5.188). TP53 mutations correlated with a median TST of 154 days versus 296 days (HR = 2.842; 95% CI: 1.093 to 7.391).

  7. 7.

    Patients with TP53 mutations/del(17p) often needed multiple therapy lines, with 85% with del(17p) and 69% with TP53 mutations versus 32% and 33% without these alterations, respectively.

Conclusions: Our analysis revealed that specific genetic mutations and chromosomal alterations are strongly associated with the disease’s course and patient prognosis. Particularly, the presence of unmutated IGHV and TP53 mutations emerged as key indicators of a more aggressive disease course and shorter treatment-free intervals. These findings underscore the critical role of advanced genetic testing in identifying patients who may require more intensive treatment and monitoring. Furthermore, the consistency and reliability of NGS in detecting these genetic markers establish it as an invaluable tool in the diagnostic and prognostic assessment of CLL, paving the way for more personalized treatment approaches.

Disclosure: This study was supported by DWD/6/0422/2022 grant from Polish Ministry of Education and Science.

The Authors have no conflicts of interest to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P502 A REAL WORLD EXPERIENCE WITH PD1 INHIBITORS IN RELAPSED/REFRACTORY HODGKINS LYMPHOMA

Disha Kakkar 1, Narendra Agarwal1, Tribikram Panda1, Aakanksha Singh1, Rohan Halder1, Roy J. Palatty1, Dinesh Bhurani1

1Rajiv Gandhi Cancer Institute and Research Centre, New Delhi, India

Background: There is no standard second line treatment for relapsed/refractory (R/R) Hodgkins lymphoma. There are multiple options such as PD-1 inhibitors, antibody-drug conjugate in combination with various salvage chemotherapies. We present a real world tertiary care centre experience with PD1 inhibitors in relapsed/refractory Hodgkins lymphoma.

Methods: It was an ambispective study and patients with R/R hodgkins lymphoma following failure of 1 line of therapy were eligible. Treatment consisted of PD1 inhibitors (pembrolizumab or nivolumab) in combination with a salvage chemotherapy or Brentuximab vedotin, which was at the physician”s discretion. The primary endpoint was ORR(overall response rate) and CR (complete remission) rate after 2-3 cycles of the designated treatment. The secondary endpoints included progression free survival and rate of bridging to ASCT (autologous stem cell transplantation).

Results: Among 26 patients enrolled, 24 were available for disease evaluation. The median age is 32.5 years, 57.7% are male,73% have advanced-stage disease and 50% have primary refractory disease. 38.5%,42.3% and19.2% of patients received pembrolizumab, nivolumab, and dual immunotherapy. The overall response rate was 100%, 100%, and 80% after pembrolizumab, nivolumab, and dual immunotherapy. Among 24 evaluable patients, 90%, 55.56% & 80% achieved CR after pembrolizumab, nivolumab, and dual immunotherapy. Among patients, 80%,88.89% & 60% underwent autologous stem cell transplantation after pembrolizumab, nivolumab and dual immunotherapy. With a median follow-up of 20 months, 90% of patients are maintaining remission status after pembrolizumab/nivolumab-based salvage. However, there was an increased risk of mortality in the peri-transplant period in patients treated with pembrolizumab due to sepsis and sepsis-related causes. Two patients succumbed to D + 5 and D + 6 due to human meta-pneumo virus-related ARDS (acute respiratory distress syndrome) and septic shock, respectively. While one patient had prolonged fever in the post-transplant period, after ruling out all infectious causes, a diagnosis of delayed immune checkpoint inhibitors related CRS (cytokine-related syndrome) was made and successfully treated with steroids. Progression-free survival is depicted graphically (0-Nivolumab-based salvage, 1-Dual immunotherapy, 2-Pembrolizumab-based salvage).

Conclusions: Second line therapy with PD-1 inhibitors is highly effective in combination with salvage chemotherapy or brentuximab vedotin, with a majority of patients could be bridged to autologous stem cell transplant. However, there was an increased risk of immune checkpoint inhibitor related adverse events in peri-transplant period, more so with pembrolizumab than nivolumab. Also, the higher response rates observed than most of the studies are due to the younger age of the patients, hence overcoming the lower response rates in older population due to T cell senescence. Also, most of our patients were not exposed to brentuximab and PD1 inhibitors was used as a second line treatment that might account for higher response rates observed in our cohort.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P503 PEGFILGRASTIM AFTER CONDITIONING WITH BEAM AND AUTOLOGOUS HEMATOPOIETIC PERIPHERAL BLOOD STEM CELL TRANSPLANTATION IN LYMPHOMA PATIENTS

Barbara Loteta 1, Giovanni Tripepi2, Pitino Annalisa3, Mercedes Gori3, Gaetana Porto1, Giovanna Utano1, G. Policastro1, Ludovica Santoro1, Maria Caterina Micò1, Massimo Martino1

1CTMO Grande Ospedale Metropolitano Reggio Calabria, Reggio Calabria, Italy, 2HCNR-IBIM, Clinical Epidemiology and Physiopathology of Renal Diseases and Hypertension of Reggio Calabria, Reggio Calabria, Italy, 3Institute of Clinical Physiology (IFC), Roma, Italy

Background: Neutropenia is a significant complication post-PBSCT, leading to increased infection risk. Granulocyte Colony-Stimulating Factor (G-CSF) is used to speed up neutrophil recovery. Pegfilgrastim, a modified form of G-CSF with prolonged action, is compared with daily filgrastim in supporting recovery after PBSCT.

Studies generally show:

  • Comparable efficacy: Both pegfilgrastim and filgrastim effectively reduce neutropenia duration, febrile episodes, and antibiotic use post-PBSCT.

  • Convenience: Pegfilgrastim requires a single dose per cycle, while filgrastim needs daily injections.

  • Safety: Both are generally well-tolerated, but individual responses may vary.

.

Choosing between pegfilgrastim and filgrastim depends on factors like patient-specific needs, cost, availability, and physician preference. Consultation with a healthcare provider experienced in post-transplant care is crucial for the best treatment decision, considering the most recent guidelines and literature.

Methods: Eighty-Six patients with Hodgkin’s Lymphoma and Non Hodgkin’s Lymphoma were treated with high-dose chemotherapy and PBSC between january 2020 and august 2023. These patients received a single 6-mg subcutaneous injection of pegfilgrastim on day +1 after transplantation.

These patients received the same treatment regimens as BEAM.

Results: FN occurred in 76.6% patients: in the 65% FUO. Grade 2-3 mucositis occurred in about 20% of patients and grade 2-3 diarrhea in about 16% of cases. The median time to neutrophil engraftment was 9 days (range 9-10). There were significant between groups differences as for gender, age at transplant, number of previous lines of therapies, platelets transfusions and units transfused, incidence of febrile neutropenia and its origin. No statistical difference was found between HD and NHL as for the time to reach the neutrophil engraftment. The RMST analysis confirmed that over 17 days of follow-up, neutrophil engraftment occurred, on average, 9.9 days after the transplant in HD and 9.7 days in NHL and the difference was statistically significant. Age, sex and previous lines of therapies-adjusted analyses also confirmed these results. FN occurred in 76.7% of patients and its incidence differs on the basis of lymphoma, and was higher in NHL than in HL. Univariate logistic analyses, show significant association with FN for lymphoma and for the number of previous lines of therapies (one vs two or more, OR 4.8 95%CI 1,03-22,59 p value =0,046). Multivariable model adjusted by variables associated at univariate analysis with FN, shows that only lymphoma is associated with FN but with large confidence interval due to relative low sample size.

Conclusions: In our patient cohort after PBSCT, pegfilgrastim showed comparable efficacy to filgrastim but offered better compliance due to its single dose. Notable differences emerged between high-dose (HD) and non-Hodgkin lymphoma (NHL) groups: younger age at transplantation (median 32 vs. 55 years), more Rituximab use in NHL, and varied treatment-related factors. NHL patients exhibited higher hospitalization rates, potentially elevating multidrug-resistant germ colonization and febrile neutropenia incidence. Continual monitoring aims to uncover further insights into febrile neutropenia rates among lymphoma subtypes post-PBSCT. This ongoing analysis seeks to elucidate underlying causes for tailored treatment approaches and improved patient outcomes in differing lymphoma subgroups.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P504 BEGEV AS SECOND-LINE SALVAGE THERAPY IN RELAPSED/REFRACTORY HODGKIN LYMPHOMA: A REAL‐LIFE EXPERIENCE

Ant Uzay 1, Elif Şenocak Taşçı1, Arda Ulaş Mutlu1, Barış Koşan2, Aybüke Görkem Koç1, Bülent Küçük2, S. Sami Kartı2

1Acıbadem Mehmet Ali Aydınlar University, Istanbul, Turkey, 2Acıbadem Atakent Hospital, Istanbul, Turkey

Background: The optimal goal in the management of refractory/relapsed Hodgkin lymphoma (HL) is to obtain minimal disease status before stem cell transplantation. The efficacy of BEGEV (bendamustin, gemcitabine, vinorelbine) regimen is well-established as first-line salvage therapy in HL in earlier studies. The search for novel treatment continues for patients with limited options in later lines. Herein, we aimed to report our clinical experience with BEGEV regimen in third- and fourth-line treatment in patients with HL who have relapsed after a standardized salvage regimen.

Methods: In this single-center retrospective trial, patients diagnosed with HL between 2018 and 2022 were analyzed. Patients who received BEGEV in third- or fourth-line treatment for recurrent/refractory disease were eligible. Patient’s demographic characteristics, pathology results, treatment and survival outcomes were recorded. Overall survival (OS), objective response rate (ORR), progression free survival (PFS) and toxicity were analyzed. p<0.05 was accepted as the statistical significance.

Results: A total of 19 Hodgkin lymphoma patients were included in this study. The median age of diagnosis was 33 (20-64) years. 15 (78.9%) patients were treated with BEGEV in the 3rd line and 4 (21.1%) in the 4th line. The risk score at the time of diagnosis was 1, 2, 3, and 4 for 6 (31.6%), 6 (31.6%), 6 (31.6%), and 1 (5.3%) patients, respectively. All patients had BEGEV-related complications. 9 (47.4%) patients had grade 3/4 complications. The most common complications were thrombocytopenia and anemia, reported in all patients. One patient had cardiotoxicity as grade 4 toxicity and had heart failure due to atrial fibrillation but was treated conservatively. One patient had grade 3 renal toxicity and required hemodialysis. Following salvage BEGEV treatment, 12 (63.2%) patients had a complete response, 5 (26.3%) patients had a partial response, and 1 (5.3%) patient had progression. A total of 17 patients underwent stem cell transplantation (SCT); 15 (78.9%) were autologous while 2 (11.1%) were allogeneic SCT. The mean PFS after the BEGEV regimen was 20.51 (14.76-26.27) months, and the OS was 28.48 (23.22-33.75) months.

Table 1. Follow-up Data

Mean (95% CI)

Follow-up

22.67 (10.25-35.11) months

Progression After BEGEV

8.35 (4.18-12.51) months

PFS After BEGEV

20.51 (14.76-26.27) months

OS After BEGEV

28.48 (23.22-33.75) months

Conclusions: To our knowledge, our study is the first to evaluate the efficacy and toxicity of BEGEV in later lines of patients with refractory/recurrent HL. Despite the accepted toxicity of this regimen in patients who had already received multiple lines of high-dose treatment, the high efficacy results make BEGEV a promising choice in treatment of HL patients whose treatment options are exhausted.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P505 EVALUATION IN SARDINIAN GENETIC UNIQUENESS POPOLATION: STEREOTYPED SUBSETS# AND MOLECULAR PROGNOSTIC MARKERS, CATEGORIZATION IN U-MUTATED POOR PROGNOSTIC GROUP OF CHRONIC LYMPHOCYTIC LEUKEMIA (CLL)

Fabio Culurgioni 1, Roberta Murru2, Alba Piras2, Wedad Salem Hamdi1, Manal Yaghmour1, Abdallah Alkhoujah1, Sara Alromaihi1, Aisha Alkaseri1, Nasimeh Azadi1, Giorgio La Nasa2, Einas Al Kuwari1

1Cellular Therapy Laboratory, National Center for Cancer Care & Research, Doha, Qatar, 2S. C. Ematologia e CTMO, Ospedale Oncologico “A. Businco”, ARNAS “G, Brotzu, Cagliari, Italy

Background: Prognostic factors are classified based on different types and varying prognostic significance. The categorization of molecular prognostic markers in Chronic Lymphocytic Leukemia (CLL) and stereotyped subsets is done based on their subtype and prognostic significance. Generally, subsets #1 and #2 categorize very aggressive diseases, while, in contrast, subset #4 is correlated with indolent disease courses.

The u mutated IGHV subset #1 (CLL#1) was identified in 2% of poor clinical outcome Caucasian CLL patients. Moreover, patients with subset #8, accounting for 0.8% of patients, have a poor outcome and are at high risk of Richter transformation among all CLL cases. On the other hand, mutations in SF3B1, TP53, BIRC3, and NOTCH1 were reported to be associated with a poor prognosis in CLL.

Methods: The aim of this study is to compare the normal classification of risk indices with other risk factors when evaluating a population with genetic uniqueness, in which previous studies appear to belong to haplotypes present exclusively in Sardinia.

The study included a total of 87 unmutated IGHV (U-IGHV) Chronic Lymphocytic Leukemia (CLL) patients from South Sardinia diagnosed at the Hematology Department of the Cancer Hospital in Cagliari between 2016 and 2022. DNAs extracted from peripheral blood were analyzed through Polymerase Chain Reaction (PCR) amplifications and subjected to Sanger sequencing analysis to evaluate the IGHV somatic mutational status. PCR reactions were employed to confirm the NOTCH1 mutational status, which was initially checked by Next-Generation Sequencing (NGS). The IGHV gene mutational analysis followed the recommendations of the European Research Initiative on CLL (ERIC), while Antigen Receptor Research Tools were used for subsets analysis.

Results: For the NGS evaluations, the hematologic panel of 32 customized genes were checked for sequencing patients with U-IGHV status and Subset#1.

In Unmutated IGHV results, in Sardinian patients, only 10.2% of them display subset CLL# expression. Among them, 52.9% showed subset #1, 11.8% were with subset #6, 5.8% were CLL#7, and 5.8% were CLL#64b. Only 5.8% of them were CLL#8. In contrast at published data, the frequency of Sardinian subsets is higher if compared to the Caucasian population. These results may suggest a change in the categorization of molecular prognostic markers in CLL, especially considering that subsets #8, #2, #1, and various genes strongly involved in the assessment of high-risk patients.

Another consideration in the evaluation of mutations of TP53 and BIRC3 reinforces the categorization of the high-risk group. In the patients included in our study, only 5.2% of CLL#1 patient expressed TP53, compared to the common 10.3%. Additionally, there was no expression of BIRC3 in the high-risk group. NOTCH1 (62.3%) showed a higher frequency compared to Sardinian populations.

Conclusions: In conclusion, the current categorization of very high-risk groups needs to be re-evaluated. Most patients in our study exhibit a high subset index CLL #1 at 47.36%, a significant difference compared to the commonly observed 2%, while subset #2 is entirely absent. The high expression of NOTCH1 (62.5%) in Unmutated IGHV (U-IGHV) patients shifts them to the high and very high-risk categories.

Disclosure: i don t have interest conflict.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P506 A NATIONWIDE STUDY OF CHRONIC LYMPHOCYTIC LEUKEMIA INCIDENCE AND MORTALITY IN THE REPUBLIC OF KOREA

Min Ji Jeon1, Hoonji Oh1, Kunye Kwak1, Eun Sang Yu1, Dae Sik Kim1, Chul Won Choi1, Byung-Hyun Lee1, Se Ryeon Lee1, Hwa Jung Sung1, Yong Park1, Byung Soo Kim1, Ka-Won Kang 1

1Korea University College of Medicine, Seoul, Korea, Republic of

Background: In East Asian countries, the incidence of chronic lymphocytic leukemia (CLL) is lower than in Western countries, and the use of novel agents is difficult due to various practical issues. Ibrutinib monotherapy for relapsed or refractory CLL has been reimbursed since 2018. As of September 2023, it just started to be covered by insurance for use as frontline therapy. In this current reality, we conducted a nationwide study to evaluate the incidence and mortality of CLL in the Republic of Korea for the establishment of a treatment scheme in the future.

Methods: Information on patients diagnosed with CLL between January 1, 2006 and December 31, 2021 was collected from the National Health Insurance Database, which contains data on more than 97% of insured patients.

Results: There has been a gradual increase in the incidence rate of CLL per 100,000 persons since 2006. Given the high incidence of CLL in individuals over the age of 60, it is believed that the aging of the population has contributed significantly to the increase in CLL incidence. The mortality rate began to rise, exceeding 10% from the age of 70 and showing a rising trend up to 30% with increasing age, and was higher in men than in women. The median overall survival of the 3,743 CLL patients identified in this study was 7.915 years (95% confidence interval: 7.389-8.309 years). Of a total of 3,743 patients, 542 (14.48%) received no treatment during follow-up, while 3,145 patients (84.02%) received any form of treatment, including those who received cytotoxic chemotherapy or novel agents. Allogeneic hematopoietic stem cell transplantation was performed in 56 patients (1.5%). Overall survival did not show significant differences between the no treatment, any treatment without transplantation, and transplantation groups.

Conclusions: Considering that any treatment, including transplantation, was administered only to the high-risk patient group with active and progressive disease, these interventions may equalize the overall survival of these patients with those in the low-risk group who do not require treatment. Further research is needed to determine whether early treatment initiation can lead to an increase in overall survival beyond the results of this study and to explore the potential benefits of allogeneic stem cell transplantation in patient populations in the era of novel agents.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P507 AUTOLOGOUS HEMATOPOETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH RELAPSED AND REFRACTORY PRIMARY MEDIASTINAL LARGE B-CELL LYMPHOMA

Alexander Galimov1, Andrey Kozlov1, Ilya Kazantsev1, Tatiana Yuhta1, Polina Tolkunova1, Natalia Mihailova1, Yuri Punanov1, Vadim Baykov1, Ivan Moiseev1, Alexander Kulagin1, Lyudmila Zubarovskaya 1

1RM Gorbacheva Research Institute, Pavlov University, Saint Petersburg, Russian Federation

Background: Primary mediastinal large B-cell lymphoma (PMBL) is a rare type of NHL in pediatric patients. Relapsed and refractory (R/R) PMBL is an even rarer phenomenon (10-25% of cases). Overall, children with R/R NHL have unfavorable prognosis but data in pediatric R/R PMBL are very limited. High-dose chemotherapy (HDT) with autologous hematopoietic stem cell transplantation (aHSCT) is used for remission consolidation in patients with chemosensitive R/R PMBL. The aim of the study was to evaluate the efficacy of HDT/ aHSCT in pediatric R/R PMBL.

Methods: Nine HDT/ aHSCT were performed in pediatric patients with R/R PMBL at the Pavlov First St. Petersburg State Medical University. The median age was 14 years (12 years to 37 years). There were 8 (89%) males and 1 (11%) female. All patients had stage 3 disease at onset. First-line treatment included protocols based on B-NHL-BFM (n=6), R-DA-EPOCH (n=2), CHOP (n=1). Seven patients (78%) had a refractory disease, two (22%) had a relapsed disease. Biopsy was performed in 2 cases (progression according to CT data in 1 case and Deauville 4 status in another case). Biopsy was not performed in cases when there was an unequivocal PET/CT-positive status (Deauville 5). Second-line treatment included R-ICE (n=5), R-DHAР (n=1), IGEV (n=1), immune checkpoint inhibitors (n=1), HDT/aHSCT (n=1). Before HDT/aHSCT, PET/CT-negative status (Deauville≤3) was in 4 (44.5%) cases, PET/CT-positive status (Deauville>3) - in 4 (44.5%) cases, not evaluated - 1 (11%) case. Two to four (median 2) lines of treatment preceded HDT/ aHSCT. HDT included conditioning regimen BeEAM - 7 (78%), R-BeEAM - 2 (22%). All patients with PET/CT-positive status before HDT/aHSCT (n=4) received posttransplant consolidative radiation therapy.

Patients’characteristics

n

Therapy

Disease status prior to HDT/aHSCT

1

R-DA-EPOCH, R-ICE №2, aHSCT

PR (CT)

2

B-NHL BFM, R-ICE №3, aHSCT+RT

PR (PET/CT)

3

B-NHL BFM, R-EPOCH №1, IGEV №2, Nivo №9 + BV №3, aHSCT+RT

PR (PET/CT)

4

B-NHL BFM, R-ICE №3, aHSCT

CR (PET/CT)

5

R-DA-EPOCH, R-ICE №2, aHSCT+RT

PR (PET/CT)

6

B-NHL BFM, Nivo №3, Pembro №3, aHSCT

CR (PET/CT)

7

B-NHL BFM, aHSCT+RT

PR (PET/CT)

8

B-NHL BFM + RT, R-ICE + BV №2, ABSCT, DHAP + BV №1, Pembro №2 + cytarabine (intrathecal), Nivo №2 + temozolomide №3 + cytarabine (intrathecal), venetoclax (3 month) + cytarabine (intrathecal), alloHSCT

CR (PET/CT)

9

CHOP №7, R-DHAР №3, aHSCT, NivoR №2 (allergy), PoloBR №6

CR (PET/CT)

  1. ICE - ifosfamide, etoposide, carboplatin; DHAP – cytarabine, cisplatin, dexamethasone, R – rituximab; Nivo – nivolumab; Pembro –pembrolizumab; PoloBR - bendamustine, polatuzumab vedotin, rituximab; IGEV - ifosfamide, gemcitabine, vinorelbine; BV - brentuximab vedotin; RT – radiation therapy, PR – partial response, CR – complete response

Results: Five-year overall survival and progression-free survival after HDT/ aHSCT were 100% and 85%, respectively. Nine of nine patients were alive at median follow-up after HDT/ aHSCT of 44 months (1-103). Two of nine patients experienced relapse after HDT/ aHSCT. One patient experienced early relapse (<12 months after remission), one patient - late relapse after 8 years. The patient with early relapse achieved remission after immune сheckpoint inhibitors followed by allogeneic hematopoietic stem cell transplantation. The patient with late relapse achieved another remission after regimen that consisted of bendamustine, polatuzumab vedotin, rituximab.

Conclusions: According to our study, R/R PMBL in pediatric patients has a relatively favorable prognosis. HDT/aHSCT is an effective treatment option for pediatric R/R PMBL. Relapse after aHSCT in children with R/R PMBL can be successfully treated with further chemoimmunotherapy.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P508 HAPLOIDENTICAL TRANSPLANTATION OF HEMATOPOIETIC STEM CELLS FOR PATIENT WITH POST-TRANSPLANT LYMPHOPROLIFERATIVE DISEASE

Vera Vasilyeva 1, Larisa Kuzmina1, Olga Aleshina1, Mariya Dovydenko1, Mikhail Drokov1, Irina Lukyanova1, Vera Troitskaya1, Elena Parovichnikova1

1National Medical Research Center for Hematology, Moscow, Russian Federation

Background: Post-transplant lymphoproliferative disease (PTLD) is one of the most common complications after organ transplantation. The main goal of PTLD treatment is to achieve stable remission of the disease and preserve the transplanted organ.

Methods: Patient B., in 1996, at the age of 9 diagnosed acute glomerulonephritis, and prednisolone therapy was started. Progression to the terminal stage of chronic renal failure (stage 5) was detected in 2003, hemodialysis was started. Transplantation of an allogeneic cadaveric kidney was performed in April 2011. Tacrolimus, mycophenolate mofetil, methylprednisolone were used as immunosuppressive therapy (IST). In the end of July 2017 patient had febrile temperature, a decreasing body weight, a dry cough, increasing abdominal volume, and irregularity in the menstrual cycle.

In August 2017 based on the obtained data during the examination the diagnosis of post-transplant lymphoproliferative disease (PTLD) was established: diffuse large B-cell lymphoma (PTLD-DLBCL) associated with IST with involvement of liver, spleen, stomach and bone marrow.

Despite the presence of a transplanted organ, taking into the variant of the disease and the generally accepted practice of managing such patients, IST were completely canceled from 24.08.2017.

Pre-phase with dexamethasone and cyclophosphamide was carried out and a positive trend was noted in decreasing size of the liver and spleen. From 31.08.2017 patient received 5 courses of CHOD in combination with rituximab. A complete remission of PTLD-DLBCL was achieved. Due to the risk of renal transplant rejection the patient was resumed immunosuppressive therapy with tacrolimus (2 mg / day) and prednisolone (5 mg / day) since April 2018. After 11 months of achievement of PET-CT-negative remission and 9 months after the resumption of the IST, a relapse of PTLD-DLBCL was noted.

IST was canceled, but the antitumor effect was not achieved. Three courses cytarabine + etoposide + lenalidomide were performed from 16.01.2019. The second PET-CT-negative remission of PTLD-DLBCL was achieved.

Program therapy for patients with the second remission of the DLBCL implies to consolidate remission by of autologous hematopoietic stem cell transplantation (HSCT). And for our patient it meant return IST to prevent kidney transplant rejection. Therefore, we decided to perform allogenic HSCT to induce tolerance of the donor immune system to the transplanted kidney. HSCT from a related haploidentical donor (mother) was planned with TCRαβ + /CD19+ depletion of the graft. The choice of such a transplant approach was determined by the fact that it does not use long-term IST.

Results: Pre-transplant conditioning regimen included: treosulfan/melphalan/ fludarabine. Haploidentical HSCT with TCRαβ + /CD19+ depletion was performed 14.05.2019 (CD34 + 10,8×106/kg). GVHD prophylaxis consisted of rituximab/ bortezomib / tocilizumab / abatacept and was completed on day + 28 after HSCT. The recovery of leukocytes was on day +13 after HSCT (3.0 × 109/l). The patient was discharged from the hospital on day + 21 after HSCT. Patient didn’t have any serious complications after haploidentical HSCT. At present, 4,5 years have passed after haplo-HSCT, and 12 years after transplantation of allogeneic cadaveric kidney.

Conclusions: Patient is in good status, without desaese relapse, renal rejection and any signs of GVHD.

Disclosure: no.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P509 ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANT IN LYMPHOMA: A SINGLE-CENTRE EXPERIENCE; A CASE SERIES

Jan Miko Aaron Baybay 1, Francisco Vicente Lopez1

1St Luke’s Medical Center, Bonifacio Global City, Philippines

Background: Allogeneic haematopoietic stem cell transplantation has a defined role in the management algorithms of non-Hodgkin lymphomas. Allogeneic haematopoietic stem cell transplant yielded durable remissions in patients with relapsed/refractory lymphomas. Graft-versus-lymphoma effect contributes to the treatment of the disease. However, allogeneic haematopoietic stem cell transplant can be complicated by acute graft-versus-host-disease, which has a significant effect on morbidity and mortality.

Methods: We performed a retrospective, single-centre study of adults with relapsed/refractory T-cell lymphoma and primary mediastinal B-cell lymphoma who underwent allogeneic haematopoietic cell transplant at St Luke’s Medical Center, Bonifacio Global City between July 2019 and 2023. Disease response evaluated using surveillance PET/CT scan. Patients underwent myeloablative regimen prior to allogeneic haematopoietic stem cell transplant according to institutional practice.

Results: Two patients with relapsed/refractory T-cell lymphoma and two patients with relapsed/refractory primary mediastinal B-cell lymphoma underwent allogeneic haematopoietic stem cell transplant. Median day of development of graft-versus-host disease was after 30 days, with skin as most affected organ. PET/CT scan showed regression of hypermetabolic activity of involved lymph nodes after allogeneic haematopoietic stem cell transplantation.

Conclusions: Adoptive immunotherapy using allogeneic haematopoietic stem cell transplant can provide high rates of durable disease control in high-risk lymphomas. Despite the expanding treatment options available for patients with lymphoma, allogeneic haematopoietic stem cell transplant should be considered in the management of patients who are sensitive to salvage therapy.

Clinical Trial Registry: None.

Disclosure: Nothing to declare.

26: Lymphoma and Chronic Lymphocytic Leukaemia

P510 THE FIRST CASE OF ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION AT DHARMAIS HOSPITAL – INDONESIAN NATIONAL CANCER CENTER

Dwi Wahyunianto Hadisantoso 1, Resti Mulya Sari1, Edel Herbitya1, Lyana Setiawan1, Hubertus Hosti Hayuanta1, Muhammad Raya Kurniawan1, Yanto Ciputra1, Annisa Annisa1, I Gusti Ngurah Agastya1, Della Manik Worowerdi Cintakaweni1

1Dharmais National Cancer Center Indonesia, West Jakarta, Indonesia

Background: Adult T-cell lymphoblastic lymphoma (T-LBL) and lymphoblastic leukemia (T-ALL) represent malignancies that arise from the transformation of immature precursor T cells. Studies suggest that T-LBL and T-ALL reflect different presentations of the same disease. The role of allogeneic hematopoietic stem cell transplantation (allo-HSCT) has been investigated in T-LBL, given the poor outcomes of this disease. Indonesia is a developing country that lacks many resources and facilities, thus hindering allo-HSCT service. We report our first case of allo-HSCT in a 29-year-old man Indonesian patient with T-LBL.

Methods: All patient’s siblings were screened for the availability of matched sibling donor (MSD). High resolution HLA typing was done by utilising the sequence-specific oligonucleotide (SSO) technologies with PCR. After a donor was identified, the thorough viral screening was done for both donor and patient. Filgrastim was used as mobilization agent and HSC were collected when blood CD34 level was above 20/mcl with the target of 4 – 6 million cells/kg patient body weight (BW). Busulfan-Cyclophosphamide (BuCy) regimen was chosen as the myeloablative conditioning (MAC). Graft versus host disease (GvHD) prophylaxis was given with the combination of MTX and ciclosporin A. After engraftment, the chimerism study was done by utilising Short Tandem Repeat (STR) analysis.

Results: A 40-year-old male sibling was identified having full-matched HLA. Both donor and patient had identical B+ blood group and positive results for anti-CMV IgG, anti-toxoplasma IgG, anti-rubella IgG, and anti-EBV VCA IgG. We collected donor’s HSC 15.47 millions/kg patient BW. There were no serious complications during the conditioning and HSC infusion process. The patient achieved engraftment status on day +19 with STR-PCR study at day +28 showed 100% chimerism.

Conclusions: We have performed our first allo-HSCT in a 29-year-old T-LBL case in Indonesia with limited facilities and resources in a developing country.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P511 PRE-EMPTIVE DONOR LYMPHOCYTE INFUSION AS OPTIMAL TREATMENT FOR RELAPSED ACUTE MYELOID LEUKEMIAS AND MYELODYSPLASTIC SYNDROMES FOLLOWING ALLOGENEIC STEM CELL TRANSPLANTATION:A FRENCH-ITALIAN EXPERIENCE WITH 103 PATIENTS

Eugenia Accorsi Buttini1, Cristina Doran2, Michele Malagola 1, Vera Radici1, Mirko Farina1, Marco Galli1, Gabriele Magliano1, Alessandro Leoni3, Federica Re3, Simona Bernardi3, Mohamad Mohty2, Domenico Russo1, Eolia Brissot2

1Cell Therapies and Hematology Research Program, University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy, 2Sorbonne Université Service d’Hématologie Clinique et Thérapie Cellulaire, Hospital Saint-Antoine, Centre de Recherche Saint-Antoine (CRSA), Paris, France, 3Research Center Ail (CREA), University of Brescia, ASST Spedali Civili di Brescia, Brescia, Italy

Background: Disease relapse represents the major reason of transplant failure in acute myeloid leukemias (AMLs) and myelodysplastic syndromes (MDSs). Ongoing developments to improve allogeneic stem cell transplantation (allo-SCT) outcome include post-transplant monitoring of minimal residual disease (MRD) associated with chimerism analysis and setting up a prompt intervention for emergent disease (pre-emptive therapy). This study aimed to assess the overall survival (OS) of AMLs and MDSs following allo-SCT relapse according to different therapeutic strategies.

Methods: We retrospectively analyzed a cohort of 553 AML and MDS patients allotransplanted between January 1, 2015, and December 31, 2021, at Saint-Antoine University Hospital, Paris (France) (n=420) and Spedali Civili di Brescia, Brescia (Italy) (n=133). 134 (24%) patients relapsed. Among these, 103 (77%) received subsequent treatment and were included in the study. Forty/103 (39%) underwent a donor lymphocyte infusion (DLI)-based regimen, with 9 receiving DLI alone and the rest combining DLI with a hypomethylating agent (HMA) (4 cases), HMA+ venetoclax (12 cases), FLT3 inhibitors (3 cases), intensive chemotherapy (5 cases), a second allo-SCT (5 cases), or other therapies (2 cases). The remaining 63/103 (61%) patients were treated with regimens excluding DLI: 10 received HMA, 27 HMA+venetoclax, 4 FLT3-inhibitors, 6 intensive chemotherapy, 9 a second allo-SCT, and 7 other therapies.

Results: With a median follow-up of 1.6 years, the 1-, 2-, and 5-year OS for the patients treated post allo-SCT relapse was 40%, 20%, and 15% compared to 6%, 3%, and 0% for patients who did not receive therapy, respectively (p<0.01). Furthermore, OS was significantly superior in patients treated in a pre-emptive setting compared to those treated for hematological relapse (OS at 1-, 2-, and 5-years: 60%, 36%, and 30% for pre-emptive treatment versus 26%, 12%, and 6% for hematological relapse (p<0.01). Regarding post-relapse treatment, patients receiving DLI-based regimens demonstrated a 1-, 2-, and 5-year OS of 55%, 32%, and 32%, respectively, compared to 27%, 16%, and 7% for patients treated with other therapies (p<0.01). Among DLI recipients, 50% were treated in a pre-emptive setting. Conversely, in the non-DLI group, only 22% were treated pre-emptively, indicating the more advanced relapse phase in this cohort. Finally, analyzing patients based on treatment strategy (pre-emptive therapy versus therapy in hematological relapse) and DLI administration (yes versus no), the optimal outcome occurred when relapse was promptly detected, and pre-emptive therapy was initiated, especially with DLI administration. Indeed, the 1-year OS for patients treated pre-emptively was 67% for those receiving DLI and 54% for those treated without DLI, while for patients with hematologic relapse, the 1-year OS was 43% for those treated with the DLI-based regimen and 17% for those treated without DLI (p<0.01). On multivariate analysis, DLI treatment and pre-emptive setting were independent factors associated with better OS (p=0.03 and p<0.01).

Conclusions: Our data demonstrate that relapse treatment with pre-emptive therapies improved outcomes, particularly when combined with DLIs, and underscore the importance of early detection through MRD monitoring.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P512 HLA-DR+REGULATORY T CELLS ARE ASSOCIATED WITH THE ONSET AND SEVERITY OF ACUTE GRAFT-VERSUS-HOST DISEASE AFTER HEMATOPOIETIC CELL TRANSPLANTATION

Kinga Hosszu 1, Devin McAvoy1, Moises Garcia-Rosa2, Charlie White1, Matthew Thomsen1, Evangelos Ntrivalas1, Elizabeth Klein1, Abdulrahman Alsultan1, Audrey Mauguen1, Kevin J. Curran1, Maria Cancio1, Andromachi Scaradavou1, Andrew Kung1, Joseph H. Oved1, Miguel-Angel Perales1, Andrew C. Harris1, Jaap Jan Boelens1

1Memorial Sloan Kettering Cancer Center, New York, United States, 2University of Puerto Rico, San Juan, Puerto Rico

Background: Recently, CD4(+) regulatory T cell (Treg) activity has been associated with the prophylaxis and therapeutic management of acute graft-versus-host disease (aGVHD) post-hematopoietic cell transplantation (HCT). Nevertheless, the intricate molecular pathways governing Treg efficacy remain poorly understood. Activated Tregs are marked by specific protein expression, notably HLA-DR, and represent a distinct terminally differentiated subgroup demonstrating heightened suppressive capabilities and swift cytokine secretion in vivo. We hypothesized that the pre-GVHD cytokine milieu influences Treg activation, and the resulting varied frequencies of activated Tregs modulate inflammatory processes during GVHD onset, thereby influencing disease severity.

Methods: All patients transplanted between June 2020 and February 2023 underwent a two-year standard flow cytometric immunophenotyping regimen (from day +14), utilizing CLIA-approved panels, including a dedicated Treg panel. Tregs, identified as CD3+CD4+CD25+CD127low, were analyzed for activation (HLA-DR+). Analyses were restricted to patients with Treg data predating the earliest aGVHD onset or with Treg values before the mean onset of GVHD (for patients without GVHD). Plasma samples at day+15 post-HCT underwent analysis using targeted proteomics via proximity extension assays (PEAs; Olink) for 184 proteins linked to inflammation and immune response. Spearman’s rank correlation tests explored associations between analyte values and aGVHD grades 2-4 at each time point. The primary outcome assessed the interplay between activated Tregs, cytokine levels, and aGVHD development within the initial 90 days.

Results: Among 101 transplanted patients, 51 with early Treg data preceding the onset of GVHD were included (median age: 12, range: 1–24 years); 14 (27%) received cord blood, 10 (20%) T-cell deplete peripheral blood stem cells, and 27 (53%) unmodified bone marrow, and 35% (16) patients developed aGVHD (grade 2-4). At day 15 post-HCT, preceding GVHD onset, various cytokines associated with decreased Treg activation and proliferation (SIT1, CD6, IL18, TNFB, TNFRSF9) exhibited significant correlations with aGVHD (Table 1). Patients before aGVHD onset experienced a rapid decline in Treg frequencies around day 25-30 post-HCT, and this decline was absent in patients without GVHD or those undergoing treatment for aGVHD. The Treg differentiation trajectory indicated a reduction in memory and activated, but not naïve, Treg frequencies before GVHD onset. Remarkably, patients with grade 2-4 GVHD displayed a decrease in activated Treg frequency, declining in a GVHD grade-dependent manner. These differences in Treg activation persisted at 1 year after HCT, indicative of a functionally suppressed Treg profile associated with aGVHD severity.

Conclusions: These findings underscore the pivotal role of HLA-DR+ activated Tregs in governing the initiation and severity of aGVHD, and emphasize the tight regulatory control exerted on Treg activation by distinct immunomodulatory cytokines. Our results contribute invaluable insights into the mechanistic underpinnings of aGVHD suppression by Tregs.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P513 MAINTENANCE THERAPY WITH OLAPARIB AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH TP53 MUTATED HEMATOLOGIC MALIGNANCIES

Zhihui Li 1, Teng Xu1, Yipei Guo1, Xianxuan Wang1, Xiaopei Wen1, Lei Wang1, Jingjing Wang1, Yanzhi Song1, Yongqiang Zhao1, Tong Wu1

1Beijing Gaobo Boren Hospital, Beijing, China

Background: The mutant TP53 protein interacts with caretaker proteins in DNA repair, affecting the repair of DNA double-strand breaks (DSB), and reducing the cell’s ability to repair DNA double-strand breaks. In cells, there is also repair of DNA single-strand breaks (SSB), which requires the participation of PARP. Using PARP inhibitors to block the tumor cells’ DNA single-strand repair (SSB) creates a combined lethality in tumor cells with TP53 gene mutations. PARP inhibitor may be a potential agent to treat TP53 mutated hematologic malignancies. In present study, the efficacy and safety of olaparib maintenance therapy after allogeneic hematopoietic stem cell transplantation (allo-HSCT) in patients with TP53 mutated hematologic malignancies are analyzed retrospectively.

Methods: Between September 2018 and November 2022, Seventy-one patients with TP53 mutated hematologic malignancies who underwent allo-HSCT in our hospital were included. The diagnosis included AML (17, 23.94%), B-ALL (36, 50.70%), T-LBL/ALL (8, 11.27%) and NHL (10, 14.08%). Twenty-nine relapsed patients (40.85%) received second-line treatment. Twenty-one patients(29.58%) underwent secondary transplant. The median age was 20.35 (1.6-64.7) years old. Before transplant, 33 patients (46.48%) had extramedullary diseases and 27 patients (38.03%) had chromosomal abnormalities. Disease status before transplantation was in CR (45 cases, 63.38%) and in NR (26 cases, 36.62%). Donor types included identical sibling (3, 4.23%), unrelated (17, 23.94%) and haploidential (51, 71.83%). Myeloablative conditioning regimen with TBI/fludarabine (40, 56.34%) based or busulfan/fludarabine (31, 43.66%) based was applied. Thirty-two patients (45.07%) received maintenance therapy with olaparib post-HSCT (olaparib group), and 39 patients (52.11%) did not receive any maintenance therapy after transplant (control group). No significant differences in clinical features were found between two groups. Olaparib treatment was initiated at a median of 67 (25-496) days at a median dose of 75 (50-150) mg twice a week. The median duration of olaparib maintenance therapy was 10(0.24-45) months.

Results: With a median follow-up 12.30 (13.72-21.28) months, overall survival (OS) and progress-free survival (PFS) were 56.36(43.78-67.16)% and 51.33 (39.04-62.34)%. For Olaparib group, OS and PFS were 90.63 (73.69-96.88)% and 84.38 (66.46-93.18)%, respectively. For control group, OS and PFS were 25.51 (12.44-40.85)% and 23.52 (11.33-38.21)%. Maintenance therapy with Olaparib significantly improved OS and PFS (P <0.001; P <0.001). Relapse rate was significantly lower in Olaparib group than that in control group (25% vs. 53.85%, p <0.014). Olaparib was temporarily discontinued in 8 patients (11.3%) and dose modified in 11 patients (15.5%) due to side effects. Grade 2 side effects occurred during Olaparib treatment, and no ≥ grade 3 side effects were observed. The most common adverse events were neutropenia, anemia, thrombocytopenia, epistaxis, neutropenic fever and fatigue, and all of them were tolerable and reversible. No organ toxicities or drug-related deaths were observed.

Conclusions: Studies have shown that post-transplant maintenance with olaparib in patients with TP53 mutation hematologic malignancies is safe and effective and can significantly reduce the recurrence rate and improve OS and PFS. Further prospective, randomized, controlled clinical trials with larger sample size should be needed in the future.

Disclosure: The authors declare no conflict of interest.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P514 PERSISTENCE OF MINOR HISTOCOMPATIBILITY ANTIGEN- AND VIRUS-SPECIFIC T CELLS IN LEUKEMIC REMISSION AFTER HEMATOPOIETIC ALLOGENEIC STEM CELL TRANSPLANTATION

Lisa Marie Schulz 1, Debora Basilio-Queiros1, Susanne Luther-Wolf1, Elke Dammann1, Michael Stadler1, Eva Mischak-Weissinger1

1Hannover Medical School, Hannover, Germany

Background: Leukemic relapse is still the major cause of mortality after allogeneic hematopoietic stem cell transplantation (HSCT). Immune responses against minor histocompatibility antigens (mHag) play an important role in relapse surveillance, evoking both graft-versus-host (GvHD) and the graft-versus leukemia (GvL) reaction. In a previous pilot study, we were able to show that the loss of cytotoxic T cells (CTL) specific for the hematopoietic restricted mHag-HA-1 correlated closely with an increase in recipient hematopoiesis and relapse of the underlying disease. In addition, our investigations showed that the presence of cytomegalovirus (CMV)-specific CTLs was associated with improved immune reconstitution, lower recipient hematopoiesis and persistent leukemia remission. Based on these data, we investigated the temporal and possibly functional relationship between CMV- and mHag-CTL reconstitution, chimerism and relapse incidence in a cohort of 52 patients after allogeneic HLA-matched HSCT.

Methods: Fifty-two patient/donor pairs were included to date since 2020. HLA-A*0201 positive patients were typed for the immunogenic and non-immunogenic mHag alleles of hematopoietic restricted mHag-HA-1 and -HA-2, as well as the ubiquitously expressed mHag-HA-8 and -H-Y using PCR analysis. Patients with immunologically relevant differences were suitable for further examinations. Samples were collected longitudinally on days +25, +50, +100, +150 and +300 after allogeneic HLA-matched HSCT. All 52 patients were analyzed for the presence of CMV- and mHag-specific CTLs via tetramer staining and subsequent flow cytometry, chimerism was measured by highly sensitive chimerism PCR. The degree of differentiation of the mHag-CTLs was examined by FACS analysis for markers CD45RA / CCR7 / CD27 / CD28 and CD57.

Results: The median age of the patients was 59 years (range: 20-72), 44 (84.6%) had acute leukemia, 27 (51.9%) received reduced intensity conditioning (RIC). Six (11.5%) patients experienced CMV reactivation (median: day +161.5; range: 28-262 days). A total of 37 (71.1%) patients developed acute GvHD (aGvHD), 12 of them developed aGvHD grade III-IV (mean: day +51; 21-127 days). Thirty-eight patients (73%) are currently alive (mean follow-up: 370 days; range 13-924 days). Seven (13.4%) patients have relapsed prior to day +300 (mean: 171 days; range 54-257), five of those have died. Decrease of mHag-CTLs consistent with the increase in host chimerism prior to relapse was observed in 85% (6/7) of the relapsed patients. The majority of all 52 patients (67%) displayed a correlation between presence of mHag-CTLs and host chimerism. Compared to patients in remission, relapsed patients tend to display a lower frequency of effector-memory T cells after day 100. Patients with a mHag-HA-2-mismatch showed significantly (p≤0.0001) higher mHag-CTL frequencies until day 50 compared to all other mHag-mismatches. CMV-CTLs counts were significantly (p≤0.05) lower after three months in relapsed patients compared to patients in remission.

Conclusions: We have established a monitoring protocol for the immune reconstitution of mHag-CTLs as well as CMV-CTLs to assess whether the presence, and most importantly, the persistence of these cell populations can be predictive of leukemia relapse. Our results indicate that presence of both CMV-CTLs and mHag-specific CTLs in patient/donor pairs correlates well with the prediction of relapse-free survival.

Clinical Trial Registry: None.

Disclosure: Authors declare no conflict of interest.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P515 SERIAL LINEAGE CHIMERISM ANALYSIS IMPROVES EARLY DIAGNOSIS OF GRAFT FAILURE AFTER ALLOGENEIC HAPLOIDENTICAL HSCT

Pilar Lancho Lavilla1, Ignacio Gómez Centurión1,2, Rebeca Bailén Almorox 1,2, Paula Fernández-Caldas1,2, Asunción Escudero1, Lucía Castilla1,2, Javier Anguita1,2,3, Ismael Buño1,2,4,3, Mi Kwon1,2,3, Carolina Martínez-Laperche1,2,4

1Hospital General Universitario Gregorio Marañón, Madrid, Spain, 2Instituto de Investigación Sanitaria Gregorio Marañón, Madrid, Spain, 3Complutense University of Madrid, Madrid, Spain, 4Genomic Unit, Hospital General Universitario Gregorio Marañón, Madrid, Spain

Background: Graft failure (GF) is an unusual threatening complication after allo-HSCT, associated with several factors, including use of mismatched donors. Monitoring of chimerism in peripheral blood (PB) and T lymphocytes (TL) has shown to contribute to its early detection.

The objective of this study was to analyse chimerism dynamics in patients who developed GF after haplo-HSCT and determine the relevance of the assessment of multilineage chimerism for early prediction of GF.

Methods: Data from 306 consecutive patients who underwent an haplo-HSCT in a single centre (2007-2023) were reviewed, and from those, patients diagnosed with GF were included.

Primary GF was defined as absence of neutrophil engraftment by day+28 with mixed chimerism (MC); and secondary GF was defined as initial engraftment and later development of severe cytopenia and MC.

T lymphocytes(CD3+) were purified by immunomagnetic means (AutoMACS, Miltenyi Biotec, Bergisch Gladbach, Germany) using antibodies against each lineage marker. The minimum purity of isolated leukocyte subsets was 95%, determined by flow cytometry.

Chimerism analysis was performed by STR-PCR (Mentype®Chimera®Biotype, Dresden, Germany) (sensitivity 1%). PB samples were obtained on day +14, + 21, +28, and every 2 weeks in MC or monthly in complete chimerism (CC).

Thereafter intervals varied according to physician decision. Relapse was ruled out in all cases with GF suspicion.

Results: Nine patients(2.9%) were diagnosed with GF after haplo-HSCT: 4(1.3%) primary GF and 5(1.6%) secondary GF (Table1). Donor specific antibodies were negative in all cases before transplant and at GF diagnosis.

Chimerism analysis in patients with primary GF diagnosis on day +14, +21 and +28 revealed persistent and increasingly high percentages of recipient (R) before GF was diagnosed, in PB (median percentage of R 75%(37.7-100), 90%(6-100), 97%(91-100) respectively) and mainly and earlier in TL (median percentage of R 97%; 93%(91-96); 85%(77-100)). All cases were salvaged with a second haplo-HSCT (3 same donor, 1 different). Median time to salvage therapy was 43(41-77) days from first transplant. Three patients achieved engraftment and CC. Overall survival (OS) at day+100 after salvage HSCT was 50% (two patients died from infection).

Chimerism analysis in patients diagnosed with secondary GF showed persistent high percentages of R, with a trend towards increasing dynamics in PB and mainly in TL. Median percentage of R 25, 10 days before and at GF diagnosis were 6%(4.4-33), 20%(14-93.9), 96%(51-97) in PB and 91%(43-95), 95%(25-97), 88.8%(64.6-96.3) in TL. As salvage therapy, one patient received donor lymphocyte infusion (DLI); two, second Haplo-HSCT; and two, DLI followed by HLA-identical-HSCT (one matched unrelated and one HLA-identical sibling who was unavailable before). Median time to salvage therapy was 28(8-130) days from diagnosis of GF. All patients achieved engraftment and CC. OS at day+100 after salvage was 80% (one patient died from infection).

Table 1. Characteristics of patients and transplants

PRIMARY GF

SECONDARY GF

Number of patients

4

5

Median age, y (range)

45 (28-63)

26 (22-62)

Male, n (%)

4 (100%)

4 (80%)

Diagnosis, n (%)

AML

1 (25%)

2 (40%)

ALL

1 (25%)

3 (60%)

MDN/MPN

1 (25%)

NHL

1 (25%)

Stem Cell Source, n (%) PB

4 (100%)

5 (100%)

Pre-transplant status, n (%)

CR MRD-

2 (50%)

3 (60%)

CR MRD+

1 (25%)

1 (20%)

Persistence of disease

1 (25%)

1 (20%)

Median infused CD34·106/kg (range)

6.8 (4.4-8)

6.6 (4.5-8.1)

Conditioning Regimen Intensity, n (%)

MAC

1 (25%)

3 (60%)

RIC

3 (75%)

2 (40%)

GVHD prophylaxis PTCy+CNI + MM, n (%)

4 (100%)

5 (100%)

Conclusions: Early T-cell chimerism dynamics may be a good predictor of GF after unmanipulated haplo-HSCT. Assessment of chimerism in leukocyte lineages improves anticipation of GF and allows early therapeutic interventions.

In our experience, timely and individualized second allo-HSCT after GF may improve outcome after primary or secondary GF.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P516 PREDICTING MEASURABLE RESIDUAL DISEASE FOR ACUTE LYMPHOBLASTIC LEUKEMIA PATIENTS POSTTRANSPLANTATION

Yue-Wen Wang 1, Guo-Mei Fu1, Lan-Ping Xu1, Yu Wang1, Yi-Fei Cheng1, Yuan-Yuan Zhang1, Xiao-Hui Zhang1, Yan-Rong Liu1, Kai-Yan Liu1, Xiao-Jun Huang1, Ying-Jun Chang1

1Peking University People’s Hospital and Peking University Institute of Hematology, Beijing, China

Background: Posttranspantation Measurable residual disease (MRD) is a powerful prognostic factor for relapse and is related to inferior transplant outcomes in acute lymphoblastic leukemia (ALL). Positive MRD after transplantation has more exact prognostic value in determining occurence than pre-transplantation MRD. However, prediction of positive posttransplantation MRD has not been reported in patients with ALL. This study indicated the predicting factors for positive posttransplantation MRD in patients with ALL.

Methods: We retrospectively analyzed 1683 ALL patients who received hematopoietic stem cell transplantation (HSCT) at a median age of 25.

Results: Both in all patients and in pediatric or adult, HLA-matched sibling donor transplantation or haploidentical HSCT subgroups, T-ALL or B-ALL, positive pre-HSCT MRD was a risk factor for post-HSCT MRD positivity (P < 0.001 for all). Disease status was also a risk factor for post-HSCT MRD positivity in total patients and in the pediatric, haploidentical SCT subgroups, or B-ALL (P=0.035; P =0.003; P =0.003, respectively). A risk score for post-HSCT MRD positivity was built using the disease status and pre-HSCT MRD status. The cumulative incidence of post-HSCT MRD positivity was 12.3%, 25.1%, and 38.8% for subjects with scores of 0, 1, and 2-3, respectively (P<0.001). The risk score was also correlated with the cumulative incidence of post-HSCT MRD positivity and relapse as well as leukemia-free survival and overall survival in multivariate analysis.

Conclusions: In our study, positive pre-MRD status and disease status were identified as two independent risk factors for post-HSCT MRD positivity in ALL patients.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P517 CYTOKINE PROFILES ASSOCIATED WITH GRAFT VERSUS HOST DISEASE AND RECENT THYMIC EMIGRANT T CELL RECONSTITUTION AFTER HEMATOPOIETIC CELL TRANSPLANTATION

Devin McAvoy 1, Kinga Hosszu1, Matthew Thomsen1, Evangelos Ntrivalas1, Elizabeth Klein1, Katina Singh1, Esther Vidal1, Kevin Curran1, Maria Cancio1, Andromachi Scaradavou1, Joseph Oved1, Miguel-Angel Perales1, Andrew Harris1, Andrew Kung1, Jaap Jan Boelens1

1Memorial Sloan Kettering Cancer Center, New York, United States

Background: Recent thymic emigrants (RTEs) are immature naïve T cells originating in the thymus which are critical to immune reconstitution after hematopoietic cell transplantation (HCT). Previous research suggests an association between graft versus host disease (GVHD) and delayed RTE reconstitution. This study investigates cytokine profiles associated with both GVHD and RTE reconstitution after HCT, aiming to identify molecular mechanisms that may clarify the relationship between the two outcomes.

Methods: Immune monitoring (from day +14 to year +2) was performed on pediatric and young adult HCT patients transplanted between July 2020 and January 2023. PBMCs and serum were cryopreserved and later analyzed by spectral flow cytometry and Olink Proximity Extension Assay, respectively. RTEs were phenotypically distinguished from naïve CD4 + T cells by the expression of CD62L and CD31. Cytokine levels at day +15 were correlated with RTE reconstitution and GVHD incidence using Spearman’s Rank test and Gene Set Enrichment Analysis (GSEA).

Results: A total of 113 HCT patients were monitored, with a median age at HCT of 12.7 years (range: 1 month to 28 years). Transplant types included 46 T-replete bone marrow (BM; 41%), 43 T-cell deplete peripheral blood stem cells (TCD; 38%), and 24 cord blood (CB; 21%). There were 49 cases of acute GVHD grade 2-4 (43%; 54% in CB, 39% in TCD, 41% in BM), 13 relapses (11%), and 1 transplant related mortality (<1%).

Before the onset of GVHD, the upregulation of cytokines associated with inflammation (CCL11, TREM1, IL18) and modulation of T cell activation (SIT1, TRANCE, TNFRSF9, DNER, CD6) was observed in comparison to patients who did not develop GVHD (Table 1). Early post-HCT (prior to day 30) RTE numbers are initially attributed to the graft, exhibiting a rapid decline in all patients. However, in patients experiencing grade 0 or 1 GVHD, a replenishment of RTEs from the thymus compensates for the diminishing early RTEs, resulting in sustained RTE levels until day 60, followed by a progressive increase in RTE frequency from ~day 60 post-HCT (Figure 1, blue line). Conversely, patients with active GVHD demonstrate impaired RTE reconstitution until day 180 (Figure 1, red line). Notably, a significant divergence in RTE dynamics between the two groups becomes apparent starting from day 90 (p=0.013; Figure 1).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (57)

Figure 1. RTE values over first 6 months post-HCT in patients grouped by GVHD grade.

Cytokine

Correlation with GVHD

Function

SIT1

0.674

T Cell activation

CCL11

0.470

Cell migration

TRANCE

0.421

T Cell activation

TNFRSF9

0.368

T Cell activation

TREM1

0.361

Amplifies TLR-induced inflammatory pathways

DNER

0.345

T Cell activation

CD6

0.334

T Cell activation/proliferation

IL18

0.327

Increase inflammatory and autoimmune responses

TNFB

0.327

Cytotoxic

Conclusions: Our investigation unveils distinctive cytokine profiles linked to graft-versus-host disease (GVHD) and the reconstitution of recent thymic emigrants (RTE) following hematopoietic cell transplantation (HCT). These identified cytokines play pivotal roles in the modulation of T cell activation and inflammation, processes intricately involved in the initiation and severity of acute GVHD. Furthermore, their influence may extend to impeding the timely reconstitution of RTEs. Larger cohorts are needed for further analysis, and to map RTE reconstitution and identify predictors (e.g., donor characteristics, conditioning regimens, HCT complications).

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P518 FAVOURABLE IMPACT OF POST-TRANSPLANT MINIMAL DISEASE NEGATIVITY ASSESSED BY FLOW CYTOMETRY ON SURVIVAL IN PATIENTS WITH MYELODYSPLASTIC SYNDROMES

Evgeny Klyuchnikov1, Anita Badbaran1, Tetiana Perekhrestenko2, Normann Steiner1, Radwan Massoud 1, Petra Freiberger1, Christine Wolschke1, Francis Ayuk1, Ulrike Bacher3, Nicolaus Kröger1

1University Cancer Center of Hamburg, Hamburg, Germany, 2Shupyk National Healthcare University, Kyiv, Ukraine, 3Bern University Hospital, Bern, Switzerland

Background: Follow-up investigation in patients (pts) with MDS and CMML under therapies mainly relies on hematologic parameters as well morphology, whereas the role of MRD is less clear. The “different from normal” approach following ELN recommendations may provide an innovative MRD option for these pts. In the present study, we focused on the prognostic impact of innovative post-transplant flow MRD monitoring on the outcomes of MDS and CMML pts.

Methods: 116 pts (male, n=70; median, 62 y, 35-79) with MDS (low/intermediate, n=36; high,/very high, n=50; R-IPSS), MDS/MPN (n=17), CMML (intermediate-1/-2, n=11; high, n=4; CIPSS) and available post-transplant day +30 and/or +100 flow MRD data (“different from normal”; ELN recommendations), who received allografts during 2016-2023 years at the Department of Stem Cell Transplantation at University Medical Centre Hamburg were included.

Results: Majority of pts had de novo MDS (n=91, 78%), received matched allografts (n=97, 81%) after RIC regimen (n=77, 66%) with ATG as GvHD prophylaxis (n=105, 91%). Post-transplant flow MRD positivity at any single day ( + 30 and +100) of measurement was considered as prognostically unfavourable. There were 47 MRDneg (n=76, 66%) and 40 MRDpos (n=40, 34%) pts.

During a median follow up of 30 months (10-167), there were 30 mortalities, 22 relapses and 16 NRM events. The 3-year OS and DFS were better in post-transplant MRDneg comparing to MRDpos pts: 75% (62-85%) vs 55% (36-72%, p=0.08); and 61% (41-78%) vs 44% (28-62%, p=0.005), respectively. This was due to a higher relapses in the MRDpos group: 32% (19-49%) vs 11% (5-25%, p=0.004). The median time to relapse was 8 months (1-55). The difference in NRM was not significant. Higher patient age (>60 y), elevated blasts in bone marrow or peripheral blood at allograft were associated with significantly higher relapses and significantly lower DFS. In multivariate analysis, post-transplant MRD negativity in the first 100 days and younger (≤60 y) age had an independent favourable impact on relapses (HR 0.36, 0.15-0.85, p=0.02; HR 0.25, 0.08-0.82, p=0.021), DFS (HR 0.31, 0.15-0.65, p=0.002; HR 0.33, 0.14-0.79, p=0.013) and OS (0.43, 0.20-0.97, p=0.041; HR 0.31, 0.12-0.84, p=0.021), respectively.

Regarding MDS, of 30 post-transplant MRDpos pts, 13 (43%) developed relapses with 9 deaths and four pts died due to NRM. Of 56 post-transplant MRDneg pts, seven developed relapses (13%) with four deaths whereas seven pts died due to NRM. Regarding MDS/MPN overlap, of five post-transplant MRDpos pts, two developed relapses (40%) with one death and one died due to NRM. Of 12 post-transplant MRDneg pts, one patient died due to NRM and one developed relapse (8%) and was alive at the last follow up. Regarding CMML pts, of five post-transplant MRDpos pts, there were no relapses whereas one pt died due to NRM. Of eight MRDneg pts, there were no relapses, whereas two pts died due to NRM.

Conclusions: Post-transplant flow MRD monitoring following the “different from normal approach” can improve relapse prediction in MDS and MDS/MPN patients and contribute to define a population, which may benefit from early post-transplant interventions (e.g. early tapering of immunosuppression, DLIs, hypomethylating agents).

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P519 FLOW-CYTOMETRIC AND TRANSCRIPTIONAL CHARACTERIZATION OF RECENT THYMIC EMIGRANTS AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION (HSCT) IN CHILDREN WITH ACUTE LYMPHOBLASTIC LEUKEMIA (ALL)

Silvia Nucera1,2, Francesca Limido1,2,3, Marco Maria Sindoni 1,2, Cristina Bugarin1, Grazia Fazio1, Andrea Biondi1,2,3, Adriana Balduzzi2,3, Giuseppe Gaipa1

1Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 2University of Milano Bicocca, Monza, Italy, 3Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy

Background: Recent thymic emigrants (RTEs) are naïve CD4 + T cells reflecting thymic activity. Our work aims at characterizing thymic activity during post-HSCT immune reconstitution in children with acute lymphoblastic leukemia (ALL).

Methods: We prospectively analyzed peripheral blood samples from 16 patients undergoing HSCT for ALL applying EuroFlow SCID/RTE flow-cytometric antibody panel. For library preparation we used NuUniversal Plus RNA-Seq with NuQuant 96 (Tecan). We used NextSeq2000 (Illumina) for RNA-seq. Data analysis was performed using Galaxy (https://usegalaxy.eu/) following published workflows. Significantly upregulated or downregulated DEGs were analyzed for pathway enrichment and GSEA using EnrichR (https://maayanlab.cloud/Enrichr/). TCR repertoire analysis was performed using MiXCR software 4.3.2. (MiLaboratories Inc) and immunarch 1.0.0 (ImmunoMind Team).

Results: We analyzed by flow-cytometry 139 samples from 16 patients with a median follow-up of 7.5 months (1-22). We observed a significant delay in the appearance of RTEs in the first 6 months in patients receiving ATLG (p=0.015) (n=8 MUD, n=4 MMD, n=4 MSD). We did not observe differences in RTEs reconstitution kinetics between patients receiving graft from PBSC (n=9) vs BM (n=7). For MUD, we found no significant differences after infusion of frozen (n=4) or fresh grafts (n=4). For MSD and MMD, the RTEs counts at 3 months post-HSCT are significantly higher whit pediatric donors (p= 0.0084). We performed RNA-seq from FACS-sorted RTEs at different timepoints post-HSCT (n=6 patients): RTEs at 1-2 months (n=5) show upregulation of TNFalpha pathway and inflammatory response compared to 4-6 months (n=4). We also compared gene expression profile of paired RTEs and non-naïve CD4 + T cells (n=3 pairs). In RTEs, we observed down-regulation of both inflammatory pathways (TNFalpha or IL2/STAT5) and G2/M checkpoint/mitotic spindle compared to non-naïve CD4 T cells, suggesting more quiescent status. By contrast Notch signaling and Wnt/Beta catenin were upregulated in RTEs. TCR repertoire analysis shows a significant increase in Gini-Simpson diversity index from 1 month to >4 months especially for TCR-alpha, while TCR-beta was more polyclonal. RTEs and non-naïve CD4 T cells display similar degree of polyclonality at later timepoints post-HSCT.

Conclusions: We here show that the EuroFlow SCID/RTE panel can be exploited to monitor thymic regeneration since 1 month post-HSCT. Through RNA-seq analysis we show that RTEs evolve towards a less pro-inflammatory phenotype in the months after HSCT. The comparison with matched non-naïve CD4 + T cells suggests that RTEs are not directly involved in inflammatory response. The meaning of increased Notch signaling remains to be established as nothing is known about Notch signaling in RTEs. Interestingly, we observe upregulation of Wnt/Beta catenin pathway which has been associated with reduced GvHD and maintenance of GvL effect in mouse models. Finally, the analysis of TCR repertoire of RTEs shows that in the first 1-2 months after HSCT RTEs are oligoclonal but evolve to a more polyclonal pattern after few months. Overall, our data show that thymic reconstitution after HSCT is a gradual and dynamic process with a higher degree of complexity than previously hypothesized and that RTEs can represent a target population for therapeutic approaches uncoupling GvHD and GvL effect.

Clinical Trial Registry: Not applicable.

Disclosure: The authors disclose no potential conflict of interest.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P520 LOW PERCENTAGES OF EARLY B-CELL PRECURSORS IN THE BONE MARROW MAY PREDICT THE DEVELOPMENT OF CGVHD FOLLOWING PEDIATRIC HSCT

Elisa Christine Peen 1, Klaus Gottlob Müller1, Claus Henrik Nielsen1, Katrine Kielsen1, Hanne Vibeke Marquart1

1Copenhagen University Hospital Rigshospitalet, Copenhagen, Denmark

Background: Allogeneic HSCT is challenged by chronic GvHD, which contributes substantially to transplant-related morbidity and mortality. Delayed B-cell recovery and impaired B-cell immunity in peripheral blood have been reported in patients with cGvHD, but the mechanisms are unclear, and B-cell development in the bone marrow is rarely addressed.

In this study, we investigated the B-cell reconstitution in the bone marrow as a predictor for cGvHD after pediatric HSCT.

Methods: We included 76 children undergoing HSCT for ALL (n=52) or AML (n=24) from 2010-2021 with a median age of 8.3 years (0.5-17.4). All patients received myeloablative conditioning based on TBI (n=33) or chemotherapy alone (n=43). Patients received BM (n=72) or PB (n=4) grafts from MSD (n=17) or MUD (n=59). GvHD prophylaxis consisted of CyA alone (n=14) or combined with methotrexate and ATG (n=62).

Fresh bone marrow samples were analyzed for specific B-cell subsets using flow cytometry at day 30, 60, 90, 180, 270, 365, 450 and 540 post-HSCT.

Results: The B-cell compartment mainly comprised naïve B-cell precursors stage I (BCPI) and BCPII subsets during the first months post-HSCT and was gradually replaced by more mature subsets, such as BCPIII, mature B-cells, and plasmablasts. Patients with AML diagnosis had higher proportions of plasmablasts from day +30 to +365(all p<0.03), while TBI-based conditioning was associated with less mature B cells and plasmablasts at day +365 (both p<0.05). Recipient age, donor age, donor- and graft type were not associated with B-cell subset distribution.

In patients with aGvHD grade II-IV (n=28, median onset day +17 (7-74), development of the B-cell lineage appeared compromised in bone marrow samples, with reduced levels of all B-cell subsets at +60 days(all p<0.03), but no later time points. This was confirmed in logistic regression models, where aGvHD grade II-IV was associated with fewer B cells at months +2 (CD19: OR=0.95, p=0.003; BPCI OR=0.8, p=0.01; BCPII: OR=0.9, p=0.003). No associations were found between aGvHD and recipient age, diagnosis, TBI, ATG, donor- or graft-type(all p>0.05).

Thirteen children developed cGvHD (extensive cGvHD: n=12) with a median onset on day +218 (121-394). Patients who developed cGvHD had a considerably delayed B-cell reconstitution with a low proportion of total B-cells, BCPI, and BCPII subsets among mononuclear cells from day +60 to +180 compared to patients who did not develop cGvHD (all p<0.05, Figure). BCPI proportions remained significantly reduced until day +365 (1% vs. 3%, p=0.04). In a Cox regression model, patients with a low proportion of total B cells, BCPI, and BCPII at day +60 and +90 had a significantly increased risk of developing cGvHD compared to patients with faster B-cell recovery (HR 4.7-8.9, all p<0.05, Table).

Development of cGvHD was not associated with any transplantation-related factors (Table).

Variables

Risk of cGvHD

HR (95% CI)

P-value

Risk factors

Age >12 years

1.4 (0.6 – 5.4)

0.3

AML diagnosis

0.9 (0.3 – 3.1)

0.9

TBI-based conditioning

0.5 (0.2 – 1.7)

0.3

ATG as part of conditioning

1.9 (0.4 – 8.4)

0.4

MUD donor

1.7 (0.4 – 7.7)

0.5

PB graft

0.6 (0.1 – 4.3)

0.6

Female donor to male recipient

0.4 (0.1 – 2.9)

0.4

Prior aGvHD (grade II-IV)

1.4 (0.5 – 4.6)

0.4

B-cell precursor levels in BM at month + 2

CD19 (<10% of MNC)

4.7 (1.0 – 22.1)

0.05

BCPI (<2% of MNC)

8.9 (1.1 – 70.1)

0.04

BCPII (<1% of MNC)

5.3 (1.1-25.0)

0.04

B-cell precursor levels in BM at month + 3

CD19(<25% of MNC)

6.2 (1.3-29.0)

0.02

BCPI (<5% of MNC)

8.3 (1.0 – 65.4)

0.04

BCPII (<10% of MNC)

3.1 (0.8 – 11.9)

0.09

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (58)

Conclusions: Our findings indicate that development of cGvHD is preceded by a delayed B-cell reconstitution demonstrated by fewer B-cell precursors in the bone marrow already from day +60. This suggests that information on B-cell precursor levels from routine MRD-analysis by flowcytometry may be implemented as a clinical tool to guide tapering of immune suppression to avoid chronic alloreactivity.

Disclosure: None to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P521 MEASURABLE RESIDUAL DISEASE (MRD) AND T-CELL CHIMERISM ARE PROGNOSTIC BIOMARKERS IN ACUTE MYELOID LEUKEMIA (AML) PATIENTS UNDERGOING ALLOGENEIC STEM CELLS TRANSPLANTATION (ASCT)

Elisa Meddi 1, Raffaele Palmieri1, Giovanni Marsili2, Federico Moretti1, Flavia Mallegni1, Alfonso Piciocchi2, Luca Maurillo1, Maria Ilaria Del Principe1, Giovangiacinto Paterno1, Raffaella Cerretti1, Gottardo De Angelis1, Benedetta Mariotti1, Maria Irno Consalvo1, Mariadomenica Divona1, Tiziana Ottone1, Sara Gargiulo1, Giulia Colafranceschi1, Maria Teresa Voso1, Adriano Venditti1, Francesco Buccisano1

1University of Tor Vergata, Rome, Italy, 2GIMEMA, Rome, Italy

Background: Allogeneic stem cells transplantation (ASCT) remains the best curative option for many patients with acute myeloid leukemia (AML). While early determination of Measurable Residual Disease (MRD) is increasingly used to drive transplant selection, the modality of MRD monitoring in the post-ASCT setting and its possible preemptive role to treat impending relapse are still debated. Furthermore, modifications of the T-cell chimerism status may decrease the graft-versus-leukemia hampering its curative effect. In our analysis, we investigated the relationship between these two variables to better predict the outcome of AML patients undergoing ASCT.

Methods: We evaluated the prognostic impact on overall survival (OS) and relapse-free survival (RFS) of any MRD positivity up to 12 months post-transplant in 55 patients who underwent ASCT for high-risk AML in CR1 (32 patients [58%] of which 29 MRDpos and 3 MRDneg), AML in CR2 (17 patients [31%] of wich 12 MRDneg and 5 MRDpos) or active disease (6 patients [11%]). MRD monitoring was performed through multiparametric flow cytometry (MFC) or quantitative polymerase chain reaction (qPCR); the threshold used to define for MRD positivity in MFC was 0.035%, while PCR positivity was defined as >2% in NPM1 mutated and CBF-AML. Either myeloablative or reduced intensity conditioning regimens were adopted according to thiotepa, busulfan, fludarabine (TBF) regimen.

Results: MRD positivity at any timepoint in the post-transplant period was associated with a reduction in both OS [median OS of 2,4 years (y) for MRDpos; median OS not reached for MRDneg, p=0.01] and RFS [median RFS of 2,4 y for MRDpos, median RFS ot reached for MRDneg; p=0.026]. Moreover, MRD positivity between 6 and 12 months after ASCT converted into a shorter OS and a higher likelihood of relapse [median RFS MRDpos 1,9 y, median RFS not reached for MRDneg; p=0.023]. Similarly, mixed donor chimerism negatively affected both OS and RFS at any time point [for RFS, HR 4,73; p=0.003]. In multivariate analysis, MRD positivity and the presence of mixed chimerism showed an independent prognostic value on RFS [HR 4,73; p=0.003]. In our study, 6 MRD-positive patients were eligible for donor lymphocyte infusion (DLI). Of these, only one showed concomitant loss of chimerism and eventually relapsed. The remaining 5 patients treated with DLI never lost full-donor chimerism despite developing GVHD. Type of transplant, conditioning regimen or source of hematopoietic stem cells did not show a statistically significant association.

Conclusions: In our analysis, post-transplant MRD positivity both in the early post-transplant phase and at later timepoints significantly influenced DFS and OS in high-risk AML receiving ASCT. The prognostic impact of MRD status was even more relevant when combined with T-cell chimerism results. These results are in line with those of larger published series (Loke et al, Blood Advances 2023) and underline the importance of a close monitoring of these biomarkers in the post-transplant phase to find a window of opportunity of timely treatment of impending relapse.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P522 THE NUMBER OF CENTRAL MEMORY CD8 + T CELLS INFUSED AND THEIR EARLY RECOVERY AFTER ALLOGENEIC HSCT WAS ASSOCIATED WITH A DECREASED RISK OF DISEASE RELAPSE

Valle Gómez García de Soria 1,2,3, Paula Díaz-Fernández1,2,3, Ana Marcos-Jiménez1,2, Nuria Montes-Casado1,2, Javier Sevilla-Montero1,2, Itxaso Portero-Sainz1,2, Yaiza Pérez-García1,2, Ángela Figuera Álvarez1,2,3, Rafael de la Cámara1,2, Cecilia Muñoz-Calleja1,2,3

1Hospital Universitario de la Princesa, Madrid, Spain, 2Instituto de Investigación Sanitaria Princesa, Madrid, Spain, 3Universidad Autónoma de Madrid, Madrid, Spain

Background: Relapse after allogeneic hematopoietic stem cell transplantation (allo-HSCT) is certain to be determined by immune reconstitution (IR). Nevertheless, there is insufficient data regarding how the quality of IR is related to the occurrence of relapse and the extent of the relapse-free period.

Aim: To identify lymphocyte subpopulations during the IR that could be used as biomarkers to anticipate the relapse in alloHSCT patients.

Methods: This is a single-institution prospective longitudinal study on 108 patients who received a first alloHSCT between January 2011 and March 2022. T cell maturation stages were identified by multiparametric flow cytometry in the donor’s apheresis and in patient’s peripheral blood samples at days +30, +60, +90, +180 and +365 posttransplant. Thus, percentages and absolute numbers of naïve T cells (TN), stem cell-like memory T cells (TSCM), central memory T cells (TCM), effector memory T cells (TEM), and CD45RA+ effector memory T cells (TEMRA) were quantified. Relevant demographic and clinical variables for the transplant outcome were annotated: donor gender/age/relationship with the recipient, HLA matching, underlying disease, conditioning regimen, graft-versus-host disease (GvHD) prophylaxis, status at transplant, graft source, infused CD34+ and CD3+ cells, among others.

Two sets of logistic regression models were designed that included the aforementioned T cell subpopulations, demographics and clinical data: a first one aimed at predicting the occurrence of relapse in the first 2 years posttransplant; the second one was designed to predict the extent of the relapse-free period in relapsing patients.

Results: A multiple logistic regression analysis showed that total CD8 + TCM cells infused into the patient was lower in patients who relapsed in the first 2 years compared to those who maintained a complete remission by that time (OR=0.31, p=0.095). At day +30 after the transplant, a lower percentage of CD8 + TCM over total CD8 + T lymphocytes (CD8 + TCM/CD8 + ) was found in patients who subsequently relapsed (OR=0.028, p=0.095). These models also considered recipient’s age, disease status at transplant, conditioning regimen, GvHD prophylaxis and the occurrence of either acute or chronic GvHD before relapse, as variables that had shown an influence on probability of relapse.

To investigate whether CD8 + TCM could impact the relapse, we focused on those patients who relapsed at any time posttransplant (n= 41) and established new predictive models with the extent of the relapse-free period expressed on days as the dependent variable. We included the same clinical variables considered for the previous models, together with the CD8 + TCM/CD8 + , the percentage of CD8 + TSCM over the total CD8+ cells and donor’s age. Interestingly, a higher proportion of CD8 + TCM/CD8 + , both in the apheresis and at day +30, was associated with a delayed relapse (OR=1.53, p<0.0001 and OR=2.19, p=0.09, respectively).

Conclusions: CD8 + TCM cells in the infused inoculum and at day +30 post-HSCT in peripheral blood seem to play an important role in the maintenance of complete remission. Therefore, this subset could be a predictive biomarker of relapse and a therapeutic objective. Further validation in a larger cohort is needed to verify this finding.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P523 CONTRASTING GENE EXPRESSION PROFILES AND SIGNALING PATHWAYS BETWEEN CIRCULATING DONOR-AND HOST-DERIVED MONOCYTES DURING TOLERANCE IN MIXED CHIMERISM FOLLOWING UNRELATED DONOR CADAVERIC BMT AND LUNG TRANSPLANT

Paul Szabolcs 1, Evelyn Garchar1, Dhivyaa Rajasundaram1, Xiaohua Chen1

1UPMC Children’s Hospital of Pittsburgh, Pittsburgh, United States

Background: Primary immunodeficiency (PID) patients may develop pulmonary complications, and most are ineligible for either BOLT or BMT due to futility. A prospective trial in PID with end-stage lung disease (NCT01852370) tests our hypothesis that persistent engraftment of cadaveric lung donor vertebral body (VB) marrow stem cells could restore immunity and result in lifelong tolerance. We reported previously that a 14-year-old female (IL7R- SCID) underwent BMT 4 months after BOLT from a 1/8 HLA-allele matched donor. The marrow suspension from VB was CD3 + /CD19+depleted and cryopreserved. Initially, she engrafted with 100% donor cells. Myeloid chimerism settled between 7-14% during years 2-5 while T-cells remained >95% donor. T/B cells, T cell receptor (TCR) and B cell receptor (BCR) repertoires, and signal joint TREC exceeded pre-BMT values by 3-6m, data previously reported. She was off immunosuppression by 1yr post-BMT and serial lung biopsies detected no rejection. Circulating donor T-cells 2 years post-BMT were hyporesponsive to host DCs while vigorously responding to 3rd party APCs. Treg depletion or IL-10R blocking did not lead to rebound alloreactivity. TCRbImmunoSEQ® revealed the gradual disappearance of host-reactive clones. Tolerant T-cells showed distinct gene expression profiles with down-regulated signaling pathways related to alloreactivity.

Methods: Here, we assessed gene expression profiles of FACS sorted and >95% pure donor and host monocytes at 2 years post-BMT from a mixed chimerism state of 11% donor CD14+ monocytes. Bulk RNAseq analysis was performed using CLC Genomic Workbench®v23 and IPA® software.

Results: Compared to donor-derived monocytes, host monocytes showed activation of distinct pathways: Macrophage classical activation, PD1-PDL1, Fatty acid activation, Immunogenic cell death, TGF-β, Ferroptosis, p53, Necroptosis, and autophagy, while donor monocytes upregulated LTβR, TNFR2, AMPK, Phagosome formation and CREB pathways.

Conclusions: This is the first case to demonstrate successful engraftment, immune competence, and acquisition of long-term tolerance from deceased organ donor VB marrow. Tolerance was characterized by clonal deletion of alloreactive T-cells, along with altered signaling pathways in circulating host monocytes interacting with tolerant donor T cells.

Clinical Trial Registry: NCT01852370.

Disclosure: None of the authors report any relevant COI.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P524 MEASURABLE RESIDUAL DISEASE AND CHIMERISM ANALYSIS INTERPLAY: IMPACT IN PROGNOSIS OF ACUTE MYELOID LEUKEMIA PATIENTS UNDERGOING ALLOGENIC STEM CELL TRANSPLANTATION

Manuel Jorge Fernandez-Villalobos1, Ignacio Gomez-Centurion1,2, Rebeca Bailen 1,2, Paula Fernandez-Caldas1,2, Lucia Castilla1, Ana Pérez-Corral1,2, Carolina Martinez-Laperche1,2, Mi Kwon1,2,3

1Hospital Universitario Gregorio Marañon, Madrid, Spain, 2Instituto de Investigación Sanitaria Gregorio Marañón (IiSGM), Madrid, Spain, 3Universidad Complutense de Madrid, Madrid, Spain

Background: Measurable residual disease and chimerism analysis are commonly used in acute myeloid leukemia to identify patients at higher risk of relapse.

Methods: AML patients in remission or aplasia who underwent allo-HSCT between 2009 and 2021 in our centre were included in our study. MRD was analyzed both by RT-PCR in bone marrow aspirates(BM) and/or peripheral blood(PB) (NMP1, RUNX1-RUNX1T1, CBFB-MYH11, KMT2A-MLLT3, WT1) and by flow cytometry(FCM) in BM at pre-HSCT (within 30 days), at day +30 and +90 post-SCT. MRD positivity was defined as above cut-off level MRD detection by either FCM or RT-PCR in a given timepoint. Whole BM cellularity and selected CD 34+ lineage chimerism was analyzed by STR at days +30 and +90 post-HSCT. IBM SPSS Statistics v.26 was used for data analysis.

Results: Data from 127 patients was analyzed. Median follow-up time was 49 months (IQR 14-73). Baseline characteristics in Table 1. Pre-HSCT MRD status was positive in 61 patients(48%) and negative in 66 patients(52%). No difference in 3-year-OS (69,7 vs 67,2%) or 3-year-EFS (60,7 vs 60,6%) between MRD- and MRD+ patients at pre-SCT time point was found.

On the other hand, statistical differences were found based on MRD status at day +30 in 3-year-OS (71.7 vs 50%) and 3-year-EFS (67.9 vs 40 %) between MRD- and MRD+ patients respectively. Similar findings were observed regarding MRD at day +90 showing 3-year-OS (77.9 vs 47.6%) and 3-year-EFS (72.6 vs 42.9%) between MRD- and MRD + , respectively. Cumulative incidence of relapse (CIR) was also predicted by MRD at +30(30 vs 27%) and at +90(39 vs 24%).

Mixed chimerism (MC) at day+30 predicted worse 3-year-OS (73.3 vs 54%) and 3-year-EFS (69.3 vs 45.5%) compared to complete chimerism (CC), but no difference was found in terms of CIR. Interplay of both chimerism analysis and MRD at day+30 was analyzed. No difference was found between MRD+ and MRD- if CC was achieved at day +30 in terms of 3-year-OS, 3-year-EFS and CIR (60 vs 75; 53 vs 71; 13.3 vs 17.4%, respectively) but statistical differences were found between MRD+ and MRD- in the setting of MC (20 vs 64; 0 vs 58; 80 vs 12.2 %, respectively). Mixed chimerism at +90 was by itself predicitive for OS, EFS and CIR.

Pre-HSCT MRD+ patients showed a trend to faster immunosupresison withdrawal compared to MRD-(81 vs 68 days). Comparisons reached statistical significance between day+30 MRD+ vs MRD- (79 vs 55 days). PreSCT MRD or day +30 MRD status did not have an impact in terms of acute GVHD grades II-IV or moderate to severe chronic GVHD.

Conclusions: MRD positivity early after HCT, either at day +30 or at day +90, had an impact in AML patients undergoing SCT. MRD positivity at day+30 had a much greater impact in those who do not achieve complete chimerism at day +30, highlighting a very poor prognosis subset of patients that represent a therapeutic challenge.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P525 THE INFLUENCE OF GRAFT IMMUNE COMPOSITION ON IMMUNE RECOVERY AND TRANSPLANT OUTCOME: A SINGLE CENTER EXPERIENCE

Stefania Leone 1, Serena Marotta1, Maria Celentano1, Mariangela Pedata1, Cristina Luise1, Angela Carobene1, Ilaria Migliaccio1, Alfonso Fiumarella2, Aldo Leone2, Mario Toriello3, Daniela Graziano1, Mirella Alberti1, Simona Maria Muggianu Muggianu1, Mafalda Caputo1, Assunta Viola1, Roberta Penta3, Claudio Falco1, Antonio Meles1, Alessandra Picardi1,4

1AORN Cardarelli, Naples, Italy, 2University Federico II of Naples, Naples, Italy, 3AORN Santobono Pausilipon, Naples, Italy, 4Tor Vergata University, Rome, Italy

Background: The success of a Hematopoietic Stem Cell Transplantation (HSCT) rely on the generation of an efficient immune cell recovery (IR), able to both control disease and to limit infectious complications. Several pre and post-transplant factors are known to influence IR, but the heterogeneity of the cohorts and of monitoring strategies limits their application in clinical practice. As well, graft immune composition (GC) is associated to HSCT outcomes. However, the relationship between GC and IR is still unconsidered. Therefore, this study aim to investigate if the type and amount of graft cell subsets infused are able to affect IR and, consequently, transplant outcomes.

Methods: We have evaluated retrospectively GC and IR of 109 allogeneic HSCT performed at AORN Cardarelli of Naples, from June 2019 to August 2023. GC data were available for 79 patients; CD34 + , CD3 + , CD4 + , CD8 + , CD56 + , CD20+ cell subsets have been identified by flow cytometry in the graft, allowing their quantification pro/kg for each patient. Immunological monitoring of CD20, CD4 and CD56 lymphocytes has been performed by flow cytometry at 6 months and 1-year post HSCT, respectively, in 54 and 33 patients, to evaluate IR. Graft and IR characteristics of patients are in the table.

Variable

Graft composition, median (IQR)

CD34 + , cells x 106/kg

4,69 (2,79-6,44)

CD3 + , cells x 107/kg

15,24 (3,89-25,82)

CD4 + , cells x 107/kg

9,81 (2,57-17,39)

CD8 + , cells x 107/kg

5,28 (1,46-8,37)

CD56 + , cells x 107/kg

3,58 (0.87-6.89)

CD20 + , cells x 107/kg

3,41 (0.87-6.29)

Immune Recovery, median (IQR)

CD20 at 6 months, cells/µl

63 (13-150)

CD4 at 6 months, cells/µl

207 (102-295)

CD56 at 6 months, cells/µl

221 (152-320)

CD20 at 1 year, cells/µl

191 (109-430)

CD4 at 1 year, cells/µl

287 (201-471)

CD56 at 1 year, cells/µl

195 (126-346)

Results: Analyzing the relationship between the cell dose pro-kg of graft immune subsets and the absolute count of CD20, CD4 and CD56 lymphocytes at 6 months and 1 year, no direct correlation has been found (p>0.05). To distinguish subjects with a higher IR, we have categorized patients in two groups, using as thresholds the 50th percentile of the absolute numbers of each cell type at 1 year or the 75th percentile at 6 months. The group with higher CD20 IR received a significant increased quantity of CD34+ (median: 5,78 vs 4,4 x106/kg, p=0.024) than patients with lower CD20 IR, while there was no significant differences in terms of graft cell dose sub-population in patients with higher CD4 and CD56 IR (p>0.05). In addition, MAC regimen, GVHD prophylaxis with PTCY or ATG, and aGVHD occurrence significantly affected CD4 IR (respectively: p=0.038; p=0.002; 0.011; p=0.016), while only aGVHD showed a significant impact on CD20 IR (p=0.032). With a median follow-up of 13.8 months, a significantly better OS and lower 1-year NRM were observed in patients with higher CD20 and CD4 IR, while only CD4 IR influence relapse rate. Regarding GC impact on clinical outcome, CD8 dose of the graft significantly increased 1-year NRM.

Conclusions: This preliminary analysis shows that GC in terms of CD34+ cell dose makes more consistent CD20 IR while it seems not to be directly related to IR of CD4 and CD56. As several pre and post-transplant factors influence CD20 and CD4 IR, it is possible that the relationship between GC and IR is conditioned by these ones and, thus, needs further investigation. We confirmed the IR effect on prognosis in terms of OS and NRM, as well as revealed the negative impact of higher CD8 graft cell dose on 1-year NRM.

Disclosure: nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P526 RELAPSE MONITORING BY NPM1-PCR AND ITS PROGNOSTIC VALUE BEFORE AND AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Oliver Kriege 1, Johannes Barucha2,1, Markus Radsak3,1, Jonas Wißkirchen1, Pascal Wölfinger1, Beate Hauptrock1, Eva-Maria Wagner-Drouet1

1University Medical Center of Johannes Gutenberg-University Mainz, Mainz, Germany, 2General Hospital Esslingen, Esslingen, Germany, 3Donau-Isar-Klinikum, Deggendorf, Germany

Background: Monitoring of response and early prediction of relapse are crucial factors for successful treatment of patients with acute myeloid leukemia. Serial evaluation of measurable residual disease (MRD) using mutated Nucleophosmin1 (NMP1) levels has shown great prognostic value to reach and sustain a complete remission. Data about the prognostic impact of a previous allogeneic hematopoietic stem cell transplantation (HSCT) on NPM1-MRD kinetics and prediction of relapse are rare.

Methods: Between 2013 and 2017, 88 patients were diagnosed with a NPM1-A positive AML at our center, 63 Patients underwent allogeneic HSCT in the later course. In all patients NPM1 levels were analyzed within the clinical routine using bone marrow (BM) and peripheral blood (PB) by quantitative real time PCR. After a complete remission, monitoring was performed every 3 months by BM or every 6 weeks by PB for 2 years, after allogeneic HSCT every 3 months by BM for one year and then ongoing every 3 months by PB. Median follow up was 30 months (1-169 month). Relapse occurred in 41 Patients (41 molecular and 39 hematologic relapse), 12 patients developed a second relapse.

We analyzed 379 samples of our MRD-Monitoring retrospectively to evaluate prospective cutoff values for patients before and after allogeneic HSCT.

Results: A cutoff at 1,7 NPM1mut/ABL*100 could be defined to predict a future hematologic relapse with 100% sensitivity and 87,7% specificity (pre vs post HSCT n.s., pooled BM/PB results). For patients without HSCT (n=12), hematological relapse was seen 41 days after the first NPM1 level above the defined ratio, whereas hematological relapse was present after 53 days in patients post-HSCT (n=11) (ns, p=0,909). Median time to relapse (molecular or hematological) was 9 months (range 0,5-99 months). In 19 patients, relapse was recognized as hematologic relapse without early MRD positivity before allogeneic HSCT, no MRD-relapse was missed after. Two patients developed a NPM1 negative relapse, three cases showed molecular relapse without BM relapse but extra-medullary disease. 3-year Overall survival was 75% in patients with molecular relapse (n=16) and 32% for molecular + hematologic relapse (n=18) (p=0,037).

Conclusions: Monitoring of NPM1 levels provide a powerful tool to predict relapse pre- and post-HSCT. There is no significant change within NPM1 cutoff to predict hematologic relapse or time between first NPM1 level above cutoff and hematologic relapse. The calculated ratio in our patients is in line with previous reports from other laboratories. A proportion of patients can be salvaged by early cellular therapies (allogeneic HSCT or donor lymphocytes). Nevertheless, with the current recommended monitoring interval of 4-6 weeks (PB) or three months (BM) during the first 2 years, 18 of 39 relapses in our cohort are missed and only diagnosed as hematologic relapse. Highlighting the importance of successful early relapse therapy in around 50% of the patients, leading to a significant survival advantage, more frequent and prolonged monitoring might be reasonable. Confounding factors for our study are mixed analyses from BM and PB samples, small patient numbers and different monitoring intervals pre- and post-HSCT.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P527 GUIDING POST-TRANSPLANT RELAPSE TREATMENT IN MYELOID MALIGNANCIES WITH QPCR MONITORING OF CHIMERISM DYNAMIC FROM PERIPHERAL WHOLE BLOOD – SINGLE CENTRE EXPERIENCE

Ana Bošković1, Tadeja Dovč Drnovšek2, Primož Rožman2, Klara Šlajpah1, Njetočka Gredelj Šimec1, Matjaž Sever1, Polona Novak 1

1UMC Ljubljana, Ljubljana, Slovenia, 2Blood Transfusion Centre of Slovenia, Ljubljana, Slovenia

Background: Numbers of older patients receiving allogeneic stem cell transplantation (alloSCT) is increasing despite higher treatment related mortality. Biology of disease in this patient population is different: majority has MDS related changes and slow proliferative disease. The most efficient prevention of relapse is GVL effect which is in most cases accompanied by GVHD. Achieving 100% donor chimerism after alloSCT annihilates positive MRD before alloSCT. But due to immune escape such as HLA loss patients can relapse despite full donor chimerism. For patients with clonal hematopoesis every increase in recipient chimerism is important and dictates treatment intervention. Patients with high risk MDS and AML with MDS related changes often lack molecular markers for MRD monitoring and NGS is the only diagnostic mean for MRD detection. However, due to high turnaround time and cost NGS is not widely available and validated method for MRD monitoring after alloSCT. DNA chimerism monitoring from peripheral blood using qPCR can serve as relapse prediction for patient with no available molecular MRD marker and can even guide treatment especially in patients with clonal hematopoesis. Compared to NGS the test is fast, readily available and inexpensive.

Methods: We analysed 175 patients transplanted at UMC Ljubljana, Slovenia since 2019 when we started with qPCR chimerism monitoring using KMRtype, KMRtrack and KMRengine Analysis Software (GenDx, Netherlands). 36/175 (21 %) patients relapsed. 13/36 patients were therapy driven with qPCR chimerism. We analysed disease characteristics and treatment of relapsed patients.

Results: All 13 patients had unfavourable disease characteristics with expected high relapse risk (Table 1). Two patients received upfront first alloSCT, one received azactidine before alloSCT, one patient received azacitidine-venetoclax and 9/13 patients received intensive chemotherapy (IC) before first alloSCT. Median time to relapse after 1st alloSCT was 13 months (r., 1-30). For relapse treatment 3/13 patients received azacitidine, all died due to progressive disease. 3/13 patients received azacitidine-DLI, 1 responded, 1 died due to progression, 1 progressed, but received IC and second alloSCT. 3/13 patients received azacitidine-venetoclax, 1 responded (Patient 1), 1 died due progression, 1 progressed and had 2nd alloSCT. One patient with HR PMF received DLI but progressed and received 2nd alloSCT (Patient 2). One patient received immediate IC and 2nd alloSCT. One patient received IC for 2nd alloSCT but died due to sepsis in aplasia. One patient relapsed 1 month after 1st alloSCT; immunosuppression was reduced and she developed grade IV GVHD but died due to relapse. Two patients had also concomitant aGVHD grade III and relapse after 1st alloSCT, both with mutated p53 and complex karyotype. One patient had severe cGVHD after 2nd alloSCT. No nonrelapse mortality after 2nd alloSCT and DLI was observed. After a median follow up of 30 months (r., 8-54), median OS was 16 months (r. 4-52) and OS of 45 % at 4 years was observed.

Patient characteristics and treatment (n=13)

N or Median (Range, %)

Age at 1st alloSCT

55 (r., 29-67)

Male/female

5/8

Disease

AML

7 (54 %)

MDS with excess blasts

5 (38 %)

PMF

1 (8 %)

Disease risk

Complex karyotype

6 (46 %)

NGS (unfavourable: ASXL1, RAS, SRSF2…)

5 (38 %)

Primary resistant AML

2 (15 %)

Therapy before first alloSCT

Azacitidine

1 (8 %)

Azacitidine+venetoclax

1 (8 %)

DAC

3 (23 %)

DA

3 (23 %)

FLAG

3 (23 %)

Conditioning regimens for first alloSCT

Cy-Bu

6 (46 %)

TEC-RIC

1 (8 %)

TBF

5 (38 %)

Flu-Bu

1 (8 %)

Donor for first alloSCT

MUD

9 (69 %)

MMUD

1 (8 %)

Haploidentical

3 (23 %)

Relapse after 1st alloSCT (months)

13 (r., 1-30)

Relapse treatment after first alloSCT

DLI

4 (31 %)

2nd alloSCT

4 (31 %)

Azacitidine

3 (23 %)

Azacitidine + venetoclax

3 (23 %)

FLAG

3 (23 %)

CLARA

1 (8 %)

Conditioning regimens for second alloSCT

TEC-RIC

1 (25 %)

Treo-Flu

1 (25 %)

TBF

2 (50 %)

Donor for second alloSCT

MUD

3 (75 %)

Haploidentical

1 (25 %)

aGVHD (gr. III-IV) after first alloSCT

3 (23 %)

cGVHD (gr. III) after second alloSCT

1 (8 %)

OS

16 (r., 4-52)

Conclusions: For patients with high risk myeloid malignancies relapsing after alloSCT, whole blood DNA chimerism guided treatment is an option due to fast turnaround time and availability, especially in patients with slow proliferative clonal hematopoiesis and no molecular MRD marker.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P528 THE COMBINATION OF DISEASE BIOLOGY WITH PRE-HSCT MRD STATUS COULD BETTER STRATIFY RELAPSE RISK AFTER HSCT AMONG ADULT AML PATIENTS IN FIRST COMPLETE REMISSION

Margherita Ursi1,2, Francesco Barbato1,2, Francesco De Felice1,2, Enrico Maffini2, Marcello Roberto 1,2, Gianluca Storci2, Salvatore Nicola Bertuccio2, Daria Messelodi2, Serena De Matteis2, Noemi Laprovitera2, Maria Naddeo2, Irene Salamon2, Francesco Iannotta2, Elisa Dan2, Luca Zazzeroni1, Barbara Sinigaglia2, Enrica Tomassini2, Massimiliano Bonafè1,2, Mario Arpinati2, Francesca Bonifazi2

1University of Bologna, Bologna, Italy, 2IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy

Background: Allogenic stem cell transplantation (HSCT) is the most effective treatment for Acute Myeloid Leukemia (AML), however relapse remains the major cause of failure and it carries a poor prognosis. A better stratification of the individual relapse risk is essential to give each patient the best chance of survival.

Methods: The aim was to analyze the relative contribution of disease biology and minimal residual disease (MRD) at the time of HSCT on subsequent relapse. We included patients in first complete morphologic remission (CR1) with a MRD assessment on bone marrow samples within 30 days before transplant. Disease biology was assessed throught cytogenetics/molecular biology and characterized according to ELN 2017 risk stratification.

MRD was measured through quantitative RT-PCR for patients with NPM mutations (threshold 0.01%) or fusion genes (RUNX1-RUNXT1; CBFb-MYH 11) and with multiparameteric flow cytometry (threshold 0.1%) for the others.

Relapse-free-survival (RFS) was the primary endpoint.

Cumulative incidence of relapse (CIR) and non relapse mortality (NRM), considered as competitive events, were secondary endpoints.

Results:

CHARACTERISTICS

ALL PATIENTS

N=59

MRD positive

N=31

MRD negative

N=28

P*

Age median (range)

57 (18-72)

56 (30-72)

59 (18-71)

0.73

Risk ELN 2017, n (%)

-Favourable

15 (25)

9 (29)

6 (21)

0.45

-Intermediate

25 (42)

11 (35,5)

14 (50)

-Adverse

18 (31)

11 (35,5)

7 (25)

-N.V.

1 (2)

1 (4)

Complex karyotype and/or TP53 mut, n(%)

-Yes

4 (7)

3 (10)

1 (4)

0.35

-No

55 (93)

28 (90)

27 (96)

Chemotherapy before HSCT, n(%)

-Yes

50 (85)

26 (84)

24 (86)

0.84

-No

9 (15)

5 (16)

4 (14)

Conditioning, n(%)

-MAC

35 (59)

19 (61)

16 (57)

0.75

-RIC

24 (41)

12 (39)

12 (43)

Donor matching, n(%)

-MUD (8/8)

35 (59)

20 (64)

15 (53,5)

0.59

-MMUD (7/8)

16 (27)

8 (26)

8 (28,5)

-APLO

8 (14)

3 (10)

5 (18)

Donor source, n(%)

-PBSC

57 (97)

30 (97)

27 (96)

0.94

-BM

2 (3)

1 (3)

1 (4)

HCT-CI n(%)

-0-2

32 (54)

17 (55)

15 (53,5)

0.92

->2

27 (46)

14 (45)

13 (46,5)

Tab.1, *X2 or Mann-Whitney as appropriate.

We retrospectively analyzed 59 AML patients who underwent HSCT at Bone Marrow Transplant Programme in Bologna, between 2019 and 2023.

Among these, 28 were MRD negative (MRD-) and 31 MRD positive (MRD + ) before HSCT.

Cytogenetics and molecular characteristics according to ELN 2017 were distribuited as follows: 9 had favourable, 11 intermediate and 11 adverse risk within MRD+ group; 6 had favourable, 14 intermediate, 7 adverse risk (1 unknown) within MRD- group.

Clinical and transplant characteristics did not differ significantly between the two groups (Table 1).

At a median follow-up of 540 days (range 36-1623), 6 patients relapsed (10%), of whom 5 MRD + and 1 MRD – at HSCT; 4 out of 6 had ELN adverse risk (3 MRD + , 1 MRD-) and 2 intermediate (both MRD + ). MRD + patients showed a trend to inferior RFS (3-year RFS 69% MRD+ vs 88% MRD-) although not statistically significant (p=0.17). Similarly patients with ELN adverse risk showed inferior RFS compared to ELN favourable-intermediate chategories, although not statistically significant (p=0.11).

Integrating ELN cathegory and MRD before transplant allowed us to segragate patients in two main risk groups: a first one including ELN favourable risk MRD +/- and ELN intermediate risk/MRD- versus a second one including ELN intermediate risk/MRD+ and ELN adverse risk MRD + /-.

Patients included in the second group showed a significantly inferior RFS respect to those of the first group with a 3-year RFS of 58% (95% C.I. 19%-83%) vs 88% (95% C.I. 67%-96%) (p=0.03, median 1107 days vs not reached).

Consistently, CIR was significantly higher in the first group, 3-year CIR 35% (95% CI 0.07%-0.67%) vs 0% (p=0.001) while NRM was similar (p=0.7).

Conclusions: The combination of disease risk by molecular and/or cytogenetics alterations with MRD status before HSCT could better refine the risk of relapse after transplant than MRD or genetics alone, especially in the ELN-intermediate cathegory.

This might help to tailor strategies aimed at reducing relapse in higher risk group, as early tapering of immunosuppression, and design of prospective clinical trials.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P529 IMPACT OF ABSOLUTE LYMPHOCYTE COUNT RECOVERY AT DIFFERENT TIME-POINTS ON CLINICAL OUTCOMES AFTER THE ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Nour Ben Abdeljelil 1, Hana Ben Hammamia1, Ines Jemaa1, Rihab Ouerghi1, Insaf Ben Yaiche1, Ines Turki1, Lamia Torjemane1, Sabrine Mekni1, Rimmel Kanoun1, Dorra Belloumi1, Saloua Ladeb1, Tarek Ben othman1

1Service d’Hématologie et de Greffe, Centre National de Greffe de Moelle Osseuse, Tunis, Tunisia

Background: Absolute lymphocyte count (ALC) recovery after allogeneic hematopoietic stem cell transplantation (allo-HSCT) seems to be associated with transplant outcomes. The objective of the study was to determine the impact of ALC recovery at different time-points on clinical outcomes.

Methods: We retrospectively evaluated outcomes of ALC at different time-points of patients who underwent a first allo-HSCT from MSD from 2015 to 2022, excluding patients who died or whose developed graft failure before day 30 and patients underwent multiple allo-HSCT. Conditioning regimen consisted on TBI-based or Busulfan iv-based for malignant disease and cyclophosphamide (CY) with or without antithymocyte globulin (ATG) with or without fludarabine (FLU) for aplastic anemia (AA). GVHD prophylaxis consisted on cyclosporine +/-short course of methotrexate +/- ATG. Lymphocyte recovery was defined as ALC ≥ 300/µL for D + 30, ≥ 500/µL for D + 100 and ≥ 750/µL for D + 180.

Results: A total of 190 patients were included, with median age of 27 years (range, 7 – 55 years). Diseases were ALL (n =63, 45%), AML (n =64, 46%), MSD (n =3), HL/NHL (n = 4), AA (n = 49, 26%) and others (n =8). Conditioning regimens consisted of TBI-based (n = 30, 16%), busulfan iv-based (n=111, 58%), CY + /- ATG + /- FLU (n = 49, 26%). The median follow-up was 24 months (50 days - 103 months). The median ALC was 1147/µL (range, 40 - 7940) on D + 30, 1438/µL (range, 150 - 6850) on D + 100 and 1950/µL (range, 390 – 12 330) on D + 180, representing 87%, 90% and 94% of recovered patients, respectively.

In multivariate analysis, factors associated with ALC recovery on D + 30 were PBSC and CMV reactivation, aGVHD on D + 100 and PBSC on D + 180.

In multivariate analysis, ALC <300/µl on D + 30 (OR: 3.7; 95%[CI]: 1.25 – 10.93; p=0.018) and aGVHD (OR: 6.6; 95%[CI]: 3.1 – 13.9; p<10-3) were independent prognostic factors for CMV reactivation.

Malignant disease (OR: 8; 95% [CI]: 2.7 – 23.4; p<10-3), donor–recipient sex mismatch (Female to male) (OR: 2.3; 95% [CI]: 1 – 5.2; p=0.04), ALC <500/µl on D + 100 (OR:4.4; 95% [CI]: 1.1 – 18.1; p=0.038) and CMV reactivation (OR: 11.6; 95% [CI]: 5 – 28; p<10-3) were significantly associated with aGvHD.

Adult’s patients (OR: 7; 95% [CI]: 2.2 – 21.5; p=0.001), malignant disease (OR: 8.9; 95% [CI]: 2.2 – 35.6; p=0.002), ALC <750/µl on D + 180 (OR: 8.3; 95% [CI]: 1.4 – 49; p=0.02) and aGVHD (OR: 10; 95% [CI]: 3.9 – 25.6; p= p<10-3) were significantly associated with cGVHD.

The only predictive factor of relapse in patients with malignant disease was ALC <750/µl on D + 180 (OR: 24.5; 95%[CI]: 2.3– 264.8; p=0.008).

ALC ≥750/µL on D + 180 was associated with better OS (OR: 0.2; 95% [CI]: 0.5 – 0.8; p=0.02) and EFS (and (OR: 0.1; 95% [CI]: 0.02 – 0.5; p=0.006), while CMV reactivation was the only predictive of non-relapse mortality (OR: 5.2; 95% [CI]: 1.2 – 21.7; p=0.02).

Conclusions: Low lymphocyte counts on D + 30 and D + 100 was predictive of CMV reactivation and aGVHD, respectively, while ALC recovery on D + 180 was predictive of better survival may be related to lower cGVHD and relapse.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P530 PHENOTYPES OF BONE MARROW MONOCYTES IN HEMATOPOIETIC CELL TRANSPLANTATION FOR ACUTE MYELOID LEUKEMIA: A DESCRIPTIVE PILOT STUDY

Gül Yavuz Ermiş1, Klara Dalva1, Ekin Kırcalı2, Şenay İpek1, Nihal Okul1, Güldane Cengiz Seval1, Pervin Topçuoğlu1, Selami Koçak Toprak1, Meltem Kurt Yüksel 1

1Ankara University/Hematology, Ankara, Turkey, 2Atatürk Chest Diseases and Chest Surgery Trainingand Research Hospital/ Hematology, Ankara, Turkey

Background: In healthy individuals, 0-3 % of BM aspiration consists of monocytes, more than 90% of are the classical type. Phenotypically, monocytes are classified in three different subsets according to the clusters of differentiation, they express: Classical M01 (CD14 bright/ CD16 -), intermediate M02 (CD14 bright/CD16 + ) and non-classical M03 (CD14 dim/CD16 +) by flow cytometry. In this series of acute myeloid leukemia (AML) patients, we aimed to investigate the relation of monocyte subsets and clinical outcomes of allotransplant recipients.

Methods: We retrospectively investigated 38 AML patients who underwent allogenic stem cell transplantation (ASCT) in complete remission at Ankara University Hematology Department. Pretransplantation and the 28th day peripheral blood absolute monocyte numbers (AMNs) are categorised in three subgroups (<0.2, 0.2-0.95 and >0.95 X 109 /l) by total blood count and bone marrow (BM) monocyte percents and monocyte subsets by flow cytometry. Monoblasts are excluded in monocyte subsets. TRM is the first 100 days mortality related with ASCT. Stastistical analysis is performed with SPSS v.26 using Kaplan Meier, Mann Whitney U and Kruskall Wallis methods and used a significance level of 5%.

Results: The demographics and details about transplants, flow cytometric analysis of pre and post- transplant BM and AMNs are shown on Table. The median overall survival after diagnosis and ASCT are 42.5(30.5- 45.7) and 17.8(1.7-33.8) months respectively. Twoyears disease free survival (DFS) after diagnosis is 80%, however DFS after ASCT in 18 months is 75%. Pretransplant AMNs and 28th day M01 subsets in de novo AML is significantly higher than secondary AML (p= 0.048). Three (7.8%) cases relapsed at 180th day and 14 (26.3%) cases suffered from grade 2-4 acute graft versus host disease (aGVHD), no differences both pre and posttransplant 28th days percentage, subsets of monocytes, AMNs. Primary engraftman failure (PEF) was found in three (7.8%) cases, 28th day monocyte percentage M01 subset (p=0.005) and 28th days AMNs (p=0.09) are lower, M02 (p=0.025) and M03 (p=0.005) subsets was higher than in PEF group. TRM is detected in four (10.5 %) cases, 100th, 180th and 365th day mortality is 10.5%, 26.3%, 36.8 %, in all groups 28th day BM monocyte percentage was higher(p<0.05). According to AMNs categories the pretransplantation BM monocyte percentages differ. (p=0.013).

Full size table

.

Conclusions: According the literature most of the monocytes circulating in the first 7-10 days after transplantation (during pancytopenia) are the M03 however through the days 12th -28th are M01 subtype.

In this pilot study there were no differences in the percentage BM monocytes, monocyte subgroups, AMNs, grade 2-4 aGVHD development, relapse and TRM. However in three patients who have PEF, both AMNs and M01 subgroup were low whereas M02 and M03 subgroups were high on the 28th day. BM monocyte percentage and subtypes may play a critical role in foreseeing PEF, but not aGVHD, relapse and TRM. Prospective studies are needed to verify this hypothesis.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P531 AVAILABLE METHODS FOR MONITORING AML NPM1 + PATIENTS POST ALLOGENEIC TRANSPLANTATION CANNOT PREDICT OUTCOME

Apostolia Papalexandri 1, Eleni Gavriilaki2, Vassiliki Kanava1, Panayiotis Dolgyras1, Fotini Kika1, Tasoula Touloumenidou1, Aggeliki Paleta1, Georgia Konstantinidou1, Panayiota Zerva1, Lamprini Vachtsetzi1, Christos Demosthenous1, Zoi Boussiou1, Anna Vardi1, Maria Papathanasiou1, Ioannis Batsis1, Anastasia Athanasiadou1, Anastasia Marvaki1, Chrisavgi Lalayianni1, Ioanna Sakellari1

1G. Papanikolaou Hospital, Thessaloniki, Greece, 2Aristotle University of Thessaloniki, Thessaloniki, Greece

Background: Chimerism and minimal residual disease (MRD) by real-time PCR (qPCR) are suggested to be prognostic for post-transplantation relapse in patients with NPM1 mutated AML(AML NPM1 + ). According to the ELN, the most sensitive, well documented technique should be used. Chimerism is usually performed by Fragment Analysis of Short Tandem Repeats (FA-STR) and the sensitivity does not exceed 1%. Monitoring of NPM1 mutation by qPCR although sensitive, returns false positive results, setting a relatively high threshold for therapeutic intervention. In the context of allogeneic transplantation(allo-HCT), such doubtful positive results could lead either to misinterpretation and unnecessary treatment, or ignoring of imminent relapse. The aim of this study is to investigate and compare the predictive value of both approaches post transplantation.

Methods: Thirty-nine patients with AML NPM1+ who underwent allo-HCT were included. All patients harboured NPM1 A, B or D mutation, measurable by qPCR and had at least one sample measured by both methods. Chimerism was performed by FA-STR. For both, bone marrow was used.

Results: Among 39 patients(10 female, 29 male), 15 were transplanted in CR1, 25 in CR2 and 4 in active disease. Median follow-up after HSCT was 26 months(2-66). Most of the patients(25/39, 62%) received graft from a Volunteer Unrelated Donor (VUD), 9/39 from identical sibling and 5/39 from haplo-identical relatives. Median age at HSCT was 48(22-65) years. Sixteen out of 39 patients harboured FLT3-ITD. Overall, 278 samples were studied. Of these, qPCR post induction was available for 26/39 and pre-transplantation qPCR for 33/39 patients. Post transplantation, 219 samples were studied, 183 by both methods simultaneously, median 4/patient. Median disease free survival(DFS) was 44 months(95%CI: 26-62). Median and mean survival(OS) was 66 and 49 months(95%CI:39-58). Post-induction NPM1-qPCR and pre-transplantation NPM1-qPCR didn’t influence DFS and OS significantly. However, patients with pre-transplantation NPM1-qPCR<10-3 showed a better outcome {mean OS 46 months (95%CI: 36-55) vs 35 months (95% CI: 21-48) for patients with qPCR >=10-3}, figure 1. NPM1-qPCR on day +100 was available in 31 patients: only 9/31 (29%) were qPCR negative compared to 17/30 (57%) patients with FA-STR negativity (complete chimerism, CC). MRD positivity on day+100 was not correlated to the outcome, regardless the method used. On +6months, qPCR negativity increased (21/31,67%). However, patients continuously MRD negative after +100day (17/39) vs patients who had at least one positive result by either method (22/39) had no difference in DFS and OS. Eighty-two samples showed discrepancy: there were more positive qPCR samples in patients with CC by FA-STR (47/82) compared to negative qPCR samples with mixed chimerism(35/82). A significant amount of patients (11/18) with qPCR + /FA-STR-, CC remained in remission, without any therapeutic interventions. They were mostly patients with low level positivity, NPM1qPCR less than minus 3.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (59)

Conclusions: Among AML NPM1 patients who undergo allo-HCT, monitoring of MRD by qPCR or FA-STR could not predict outcome. Lower levels of residual disease pre transplantation may be beneficial, however, low level positivity of qPCR post transplantation does not correlate to relapse. Our results reflect the scepticism on the available methods for measuring NPM1 mutant transcripts.

Disclosure: No conflict of interest.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P532 DONOR LYMPHOCYTE INFUSION, A SINGLE-CENTER RETROSPECTIVE SAFETY ANALYSIS

Birgitte Strand Bergland 1, Anders Eivind Myhre1, Camilla Dao Vo1, Ingerid Weum Abrahamsen1, Tobias Gedde-Dahl1,2, Tor Henrik Anderson Tvedt1

1Oslo University Hospital, Oslo, Norway, 2Institute of Clinical Medicine, University of Oslo, Oslo, Norway

Background: Donor lymphocyte infusion (DLI) is increasingly employed post-allogeneic stem cell transplant (HSCT) for high-risk cases. Prophylactically given to very high-risk patients such as acute myeloid leukemia with complex karyotypes, TP53-mutation or after second HSCT and preemptively for minimal residual disease (MRD) or decreasing donor chimerism. The current study is a single-center retrospective analysis of patients receiving DLI focusing on overall survival, treatment related complications and risk of graft-versus-host-disease (GVHD).

Methods: Single-center registry data analysis of all patients receiving DLI at Oslo University Hospital between September 2018 and March 2023.

Results: Fifty patients received DLI during the study period, 33 males and 17 females with a median age of 63.5 years (19-74). Treatment was initiated preemptively for MRD in 10 patients, decreasing donor chimerism in 15 patients, prophylactically for high-risk disease in 14 patients while 11 patients received DLI as treatment for overt disease relapse. Median time from HSCT to DLI was 217.5 days (101-1956). The median donor chimerism at the first DLI-infusion was 98.5% (10-99%). The median number of infusions was two, while 20, 15, 11 and 5 patients received 1, 2, 3 or 4 infusions respectively. Six patients had previously known GVHD, none had active GVHD at the time of infusion.

Ten patients died during the observation period. After a median follow up time of 371.5 days (range: 28-1584) cumulative survival rates indicate that 86% (95%CI 0.71-0.93) of patients were alive. There was one case of treatment related mortality (TRM) secondary to GVHD, while one patient died due to post-transplant lymphoproliferative disease. Eight died due to disease relapse, four having received DLI for overt relapse and four prophylactically for high-risk disease. We found no single variable, continuous or dichotomous, significantly associated with better overall survival. Age at the time of infusion was the only variable significantly associated with relapse related morality both in the univariate analysis (p=0.05, provided 10-year increments; 95%CI 1.00-2.30) and multivariate analysis (p=0.037, provided 10 year-increments; 95%CI 1.03-2.62).

Twenty-two patients developed GVHD whereof 10 grade III-IV requiring systemic immunosuppressive therapy. The cumulative incidence of GVHD and grade III-IV GVHD at one year follow-up was 47.5% (95%CI 0.34-0.64) and 24.9% (95%CI 0.13-0.45) respectively, and 56.4% (95%CI 0.40-0.74) and 37.6% (95%CI 0.21-0.62) at two year follow-up. Higher donor chimerism prior to DLI was significantly associated with increased risk of grade III-IV GVHD (p=0.025, HR 1.42, 95%CI 1.04-1.94) in the univariate analysis but not in the multivariate analysis. Lastly, time from HSCT to DLI was associated with increased risk of grade III-IV GVHD in the multivariate analysis (p=0.027, provided 100-day increments: HR 1.12, 95%CI 1.01-1.24).

Conclusions: Patients receiving DLI typically have a high risk of disease relapse. Our center experience confirms that the risk following DLI is low. While 56.4% of patients experienced GVHD at two-year follow-up, 37.6% required systemic immunosuppressive therapy for GVHD and only one case of death was attributed to GVHD.

Disclosure: Birgitte Strand Bergland: Nothing to declare.

Anders Eivind Myhre: Consultant/advisor: Takeda, Bristol Myers Squibb, Astra Zeneca, Immedica. Speaking fees: Novartis, Takeda, Unimedic.

Camilla Dao Vo: Nothing to declare.

Ingerid Weum Abrahamsen: Nothing to declare.

Tobias Gedde-Dahl: Nothing to declare.

Tor Henrik Anderson Tvedt: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P533 WT1 MONITORING AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: A NEW IDEA OF DYNAMIC ASSESSMENT THROUGH THE PARAMETER “DELTA-WT1 THRESHOLD”. A SINGLE-CENTRE RETROSPECTIVE STUDY

Beatrice Manghisi 1, Paola Perfetti1, Marilena Fedele1, Elisabetta Terruzzi1, Andrea Aroldi1, Sonia Palamini2, Martina Venegoni2, Carlo Gambacorti Passerini3, Matteo Parma1

1IRCCS San Gerardo dei Tintori, Monza, Italy, 2Tettamanti Center, IRCCS San Gerardo dei Tintori, Monza, Italy, 3Università di Milano-Bicocca, Monza, Italy

Background: The evaluation of minimal residual disease (MRD) allows early detection and treatment of relapse in many haematological malignancies. Acute myeloid leukemias (AML) and myelodysplastic syndromes (MDS) often lack molecular MRD markers, making it difficult to perform a proper post-transplantation monitoring.

WT1 gene, over-expressed in approximately 80% of AML and 50% of MDS, has been recently proposed by ELN as a MRD marker in the absence of other molecular options. Previous reported experiences in this field provide discordant results about informative time-points, positivity thresholds and post-HSCT follow-up.

Methods: This single-centre study aims to evaluate the utility of WT1 as a molecular MRD marker in the post-HSCT setting. In our Hematology Unit, all the patients with a transplant-eligible AML or MDS lacking MRD markers are screened at the onset for WT1 expression on bone marrow by real-time quantitative PCR; patients with a positive result according to ELN thresholds (WT1 ≥ 250 copies/104 ABL) undergo serial WT1 monitoring after HSCT.

Since June 2020 to June 2023, 46 patients were screened for WT1 at diagnosis, 33 resulting positive and 13 negative; HSCT was performed in 29 and 12 patients in the WT1-positive and negative group respectively.

Results: Comparing clinical and biological characteristics at disease onset between WT1-positive and negative groups, we observed that MDS was significantly more represented in the WT1-negative group (53.85% vs 15.15%, p = 0.021), while AML was more frequent in the WT1-positive one. Consistently with this result, median marrow blast count was significantly higher in the WT1-positive group (50% vs 13%, p = 0.0088). Interestingly, ASXL1 mutation was more frequent in the WT1-negative group (69.23% vs 9.31%, p = 0.0001). Six post-HSCT relapses were observed in the WT1-positive (20.69%) and three in the WT1-negative (25%) group. In the WT1-positive group, the serial monitoring showed that progression-free survival (PFS) was significantly superior for patients displaying WT1 levels persistently < 250 copies in the first 3 months after HSCT, compared to patients who had at least a WT1 result ≥ 250 copies in the same period (median PFS “not reached” vs 6.05 months, p = 0.0018).

We introduced the new concept of “dynamic assessment” by evaluating the variation of WT1 levels in two consecutive measures (“Delta-WT1 threshold”). Interestingly, we observed that Delta-WT1 ≥ 100, compared to Delta-WT1 < 100, even under the threshold of 250 copies, was associated to a significantly higher number of relapses, (66.67% Vs 7.14% p = 0.0139) and lower median PFS (“not reached” Vs 18.73 months, p = 0.0246).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (60)

Conclusions: Our study confirms the different WT1 overexpression in AML and MDS, probably ascribable to a higher “disease burden” in AML. The correlation between ASXL1 mutation and WT1 negativity could be explained by the prevalence of MDS in this subgroup.

At last, our results support the utility of the serial WT1 monitoring after HSCT, particularly in the first months, with a relevant element of novelty represented by the role of a dynamic WT1 assessment in predicting relapse, as we showed in our study through a new parameter named “Delta-WT1 threshold”.

Disclosure: nothing to disclose.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P534 CHIMERISM DYNAMICS POST ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A TOUGH NUT TO CRACK

Fatma SE Ebeid1, Sara Makkeyah1, Safa Matbouly1, Heba G.A. Ali1, Nayera HK Elsherif 1

1Ain Shams University, Pediatric Hematology/Oncology & BMT Unit, Cairo, Egypt

Background: The dynamics of chimerism evolution determines the success of allogeneic hematopoietic stem cell transplantation (HSCT). The intensity of the conditioning, the T-cell content and the GVHD prophylaxis, influence the degree of chimerism after SCT.

Methods: Our Objective is to describe our 2-years’ experience in the management of the evolution of mixed chimerism post HSCT in children with malignant and nonmalignant at Ain Shams University in Cairo, Egypt, with the available resources.

The study includes 19 allogeneic HSCT, all from HLA matched related donors and all received myeloablative conditioning regimens which differed according to the disease and initially cyclosporine (CSA) and methotrexate as GVHD prophylaxis. Chimerism analysis from peripheral blood (PB) was performed by FISH for the sex chromosomes or VNTR on days +28, +60, +90, +180, +365 and once a year thereafter, Moreover, chimerism was analyzed in PB every 2 weeks in case of mixed chimerism until complete chimerism (CC) was achieved. There is no available lineage specific chimerism nor quantitative assessment of chimerism. Results of chimerism follow-up were censored once upon relapse or rejection.

Results: Of the 19 patients, nine developed mixed chimerism with the following diagnosis severe aplastic anemia (1), β-thalassemia major (2), ALL (1), AML (1), HLH (1), SCID (2), and Schwachman diamond (1). The patient with HLH was a twelve-year-old boy who had early mixed chimerism that reverted complete at day+90 with the shifting of CSA to sirolimus and remained till date (D + 400). Two β-thalassemia patients developed mixed chimerism at day + 90 and day+ 325 post-transplant and remained non transfused with a stable hemoglobin and a HbA% of 87-97% till date (day+362 and day+481) without intervention. The girl with aplastic anemia and the patient with Schwachman diamond developed mixed chimerism at day +468 and day+28 respectively. Both reverted completely upon intensification of immunosuppressives. Two SCID patients had mixed chimerism at day+90 and immunosuppressives withdrawn; one girl had BCGitis denoting immune reconstitution with normal TRECs and KRECs and remained till date (day+270), unlike the boy who developed several CMV reactivations and died 5 months post-transplant. One 8 years old girl with ALL developed mixed chimerism at day +90, CSA was ceased yet 2 weeks later developed 26% blast in BM. She received 2 cycles of FLAG with reversion to negative MRD and complete chimerism and remained to date Day+500. One AML boy who had mixed chimerism at day +60 with a rising MRD, he had repeated courses of donor lymphocyte infusion at escalating doses which succeeded initially at reverting chimerism to complete and MRD to negative at day +159 before he developed mixed chimerism again at day+246 and a bone marrow relapse and did not respond to salvage chemotherapy.

Conclusions: With the limited resources and the absence of more sophisticated and sensitive analysis of chimerism, we were still able to successfully intervene when a decreasing donor chimerism or persistent MC is detected.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P535 ANALYSIS OF THE EXTENDED T-LYMPHOCYTES PHENOTYPE REVEALS DIFFERENCES IN THE EARLY IMMUNE RECONSTITUTION AMONG PEDIATRIC PATIENTS RECEIVING MATCHED-UNRELATED DONOR OR HAPLOIDENTICAL HSCT

Alessandro Di Gangi 1,2, Giorgio Costagliola3, Annalisa Legitimo4, Eva Parolo3, Elisa Costa4, Chiara Lardone4, Gabriella Casazza3, Mariacristina Menconi3

1Sant’Anna School of Advanced Studies, Pisa, Italy, 2Fondazione Pisana per la Scienza ONLUS, San Giuliano Terme, Italy, 3U.O. Oncoematologia Pediatrica, Azienda Ospedaliero Universitaria Pisana, Pisa, Italy, 4University of Pisa, Pisa, Italy

Background: Immune reconstitution (IR) after hematopoietic stem cell transplantation (HSCT) has been extensively investigated however, limited evidence exists regarding the kinetics of distinct subpopulations identified through advanced immunophenotyping in pediatric patients undergoing haploidentical (HAPLO) HSCT with post-transplant cyclophosphamide (PTCY) versus those undergoing matched-unrelated donor (MUD) HSCT.

Methods: We included patients treated with HSCT from MUD or HAPLO donors at our department from 2020 to 2023 for both malignant and non-malignant diseases. All patients underwent myeloablative conditioning with the exception of those affected by severe aplastic anemia, while all patients received Rituximab before the graft infusion. Both peripheral stem cells (PBSC) and bone marrow grafts (BM) were considered. Extended T-cell immunophenotyping was performed at 3 months after HSCT. Blood was collected in EDTA tubes and immediately processed. CD3+CD4+ (helper) and CD3+CD8+ (cytotoxic) lymphocytes were also analyzed for the expression of CD45RA, CD62L, and CD31 to identify naïve (CD45RA+CD62L+), central memory (CM, CD45RACD62L+), effector memory (EM, CD45RACD62L), terminal differentiated (TEMRA, CD45RA+CD62L), and recent thymic emigrants (RTE, CD45RA+CD62L+CD31+). Regulatory T-cells (Treg) were identified as a CD4+CD25+/++CD127low/− cells while CD45RA expression estimated their naïve counterpart. CD16/56+ identifies NK cells. CD4-CD8- were defined as double negative (DNT). Non-parametric or t-test were applied as appropriate. Results are expressed as means and standard errors (SE) or median and inter-quartile range (IQR).

Results: We enrolled 14 patients who underwent HSCT from MUD (n=10) or HAPLO (n=5) donors, median age was 10 years old IQR=13.5, similar among groups (p>.05). One second MUD transplant was included. Patients were affected by acute leukemias (n=8), myelodysplastic syndromes (n=1), severe aplastic anemia (n=2), neuroblastoma (n=1) or transfusion-dependent thalassemia (n=2). BM grafts were less represented (n=4) than PBSC (n=11), with similar rates among groups. Graft source was not related to disease type. CD3+, T-cytotoxic, NK-cells, DNT lymphocytes were similar in HAPLO and MUD cohorts while T-helper, Treg and naïve counterparts were slightly more represented in HAPLO (table 1. RTE were dramatically increased in HAPLO. Other subgroups did not show any relevant difference. CD4+/CD8+ ratio were increased in HAPLO but not statistically significant (mean=1.1 SE=0.6 and mean=0.8 SE=0.3 respectively, p>.05). The ratio between CD4+naïve and the global CD4+memory population (considering both EM and CM) was higher in HAPLO platforms than in MUD (HAPLO=0.2 SE=0.2, MUD=0.1 SE=0.03, p=.03) while the specular ratio on CD8+ cells did not show any difference (HAPLO=0.8 SE=0.2 and MUD=0.5 SE=0.2, p>.05). Similarly, the ratio among CD4+ and CD8+ naïve cells were higher in HAPLO settings but without statistical significance (HAPLO=0.6 SE=0.3, MUD=0.3 SE=0.2, p=.12).

Variable

Mean

SE

P-value

Variable

Mean

SE

P-value

CD3+

HAPLO

67.6%

14.2

>0.05

CD4+

naïve

HAPLO

14.4%

5.6

0.02

MUD

58.1%

9.8

MUD

4.0%

3.1

CD4+

HAPLO

20.7%

5.9

>0.05

CD4+

EM

HAPLO

22.9%

6.5

>0.05

MUD

14.8%

2.2

MUD

46.6%

8.6

CD8+

HAPLO

44.9%

14.1

>0.05

CD4+

CM

HAPLO

61.9%

2.7

>0.05

MUD

40.6%

8.8

MUD

47.5%

8.6

DNT

HAPLO

2.2%

1.3

>0.05

CD4+

TEMRA

HAPLO

0.8%

0.3

>0.05

MUD

3.3%

2.7

MUD

1.9%

1.2

NK

HAPLO

31.0%

13.9

>0.05

CD8+

naïve

HAPLO

34.9%

8.9

>0.05

MUD

40.2%

7.7

MUD

18.5%

7.3

Treg

HAPLO

12.2%

2.9

0.07

CD8+

EM

HAPLO

10.2%

4.6

>0.05

MUD

6.2%

1.5

MUD

28.3%

7.1

Treg

naïve

HAPLO

8.8%

3.7

0.007

CD8+

CM

HAPLO

29.1%

7.2

>0.05

MUD

0.5%

0,3

MUD

33.1%

7.8

RTE

HAPLO

7.9%

2.9

0.004

CD8+

TEMRA

HAPLO

22.6%

10.2

>0.05

MUD

0.3%

0.1

MUD

20.1%

5.6

Conclusions: Despite the limitations deriving from the small sample size and the heterogeneity of the cohort, taken together these observations suggest a more rapid IR in HAPLO-PTCY than in MUD which is expressed by a more pronounced recovery of the CD4/CD8 ratio and an increased thymic output, highlighted by the levels of naïve T-cells and RTE. Clinical implications are under investigation and larger cohorts are needed to better elucidate the kinetic of IR.

Disclosure: Nothing to declare.

17: Minimal Residual Disease, Tolerance, Chimerism and Immune Reconstitution

P536 CLINICAL FACTORS ASSOCIATED WITH IMMUNOGLOBULINS AFTER ALLO-SCT

Alberto Blanco Sánchez1, José María Sánchez Pina 1, Adolfo Sáez Marín1, Guillermo Ramos Moreno1, Esther Parra Virto1, Andrea Tamayo Soto1, Lucía Alba Medina1, Reyes Más Babio1, María Calbacho Robles1, Joaquín Martínez-López1

1Hospital Universitario 12 de Octubre, Madrid, Spain

Background: Allogeneic hematopoietic stem cell transplantation constitutes the curative treatment for many patients with various hematologic disorders. It is a complex procedure with potential serious complications, especially infections. Immune reconstitution occurs progressively and slowly, with some patients even maintaining a state of immunosuppression for life. Few studies have explored the risk factors for hypogammaglobulinemia following this procedure, and there is limited evidence in the era of post-transplant cyclophosphamide platform (PT-Cy). In our study, we aim to analyze the risk factors for hypogammaglobulinemia in patients who received PT-Cy.

Methods: This is a single-center retrospective study in which we analyzed the immunoglobulin levels of patients undergoing a first allogeneic transplant who received PT-Cy. Other variables such as age, underlying disease or development of graft versus host disease (GvHD) were evaluated. The data were analyzed using SPSS (version 25).

Results: Sixty-six cases undergoing allogeneic transplantation between 2015 and 2022 were studied. These patients received GvHD prophylaxis with cyclosporine, mycophenolate, and post-transplant cyclophosphamide. The median age was 45 years. The primary transplant indication was acute myeloid leukemia (25 patients, 37.9%), followed by non-Hodgkin lymphoma (10 patients, 15.2%), and lymphoblastic leukemia (8 patients, 12.1%). A statistically significant association was found between pre-transplant immunoglobulin levels and those detected at one year. Additionally, patients with non Hodgkin lymphoma and those who experienced grade II-IV acute graft-versus-host disease presented significantly lower immunoglobulin levels. No differences were found regarding age, sex, conditioning regimen or HLA source.

Variable

Median, range

Absolute/frequency

Mean Ig, SD

p

Age:

45.5 years (13.2)

1076 (486.4)

0.77

Sex

Female

33 (50%)

1016 (519)

0.3

Underlying condition

AML

25 (37.9%)

1148.6 (381)

0.037

NHL

10 (15.2%)

633.6 (221.6)

ALL

8 (12.1%)

1228.25 (648.1)

HL

7 (10.6%)

1145.3 (458.4)

Other

15 (24.2%)

1155.3 (711.1)

Previous Ig level

1233.6 (520.3)

1076 (486.4)

0.02

Conditioning

Myeloablative

27 (40.1%)

1171.3 (541.9)

0.24

Reduced intensity

39 (59.9%)

1041.5 (397.6)

GvHD II-IV

17 (25.8%)

866 (402.2)

0.037

GvHD I or none

49 (74.2%)

1149.4 (495.3)

HLA source

Haplo

32 (48.5%)

1076.5 (430.4)

0.18

Related donor

27 (41%)

1046 (472)

Unrelated donor 10/10

7 (10.5%)

1193.1 (788)

Conclusions: The data from our series align with those shown in previous studies (although all of them were conducted before the introduction of PT-Cy), demonstrating higher rates of hypogammaglobulinemia in patients with lymphoproliferative syndrome, significant GVHD, and low pre-transplant Ig levels. The limited sample size could explain the absence of an association with HLA donor source. The identification of these factors can help define high-risk infection groups, optimizing preventive strategies such as the administration of intravenous immunoglobulins, or the vaccination strategy.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P537 REAL-WORLD OUTCOMES OF UPFRONT AUTOLOGOUS HEMATOPOIETIC STEM CELL TRANSPLANTATION IN NEWLY DIAGNOSED MULTIPLE MYELOMA PATIENTS WITH DEL (17P)

Curtis Marcoux 1, Oren Pasvolsky2, Denái R. Milton2, Hina Khan3, Mark R. Tanner2, Amna Ahmed4, Qaiser Bashir2, Samer Srour2, Neeraj Saini2, Paul Lin2, Jeremy Ramdial2, Yago Nieto2, Guilin Tang2, Yosra Aljawai2, Hans C. Lee2, Krina K. Patel2, Partow Kebriaei2, Sheeba K. Thomas2, Donna M. Weber2, Robert Z. Orlowski2, Elizabeth J. Shpall2, Richard E. Champlin2, Muzaffar H. Qazilbash2

1Dalhousie University, Halifax, Canada, 2The University of Texas MD Anderson Cancer Center, Houston, United States, 3McGovern Medical School, The University of Texas Health Sciences Center at Houston, Houston, United States, 4Jinnah Sindh Medical University, Karachi, Pakistan

Background: With the use of novel agents such as immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), and anti-CD38 monoclonal antibodies, median survival of multiple myeloma (MM) now exceeds 10 years. However, there remains considerable heterogeneity in outcomes, influenced by clinical and cytogenetic factors, with deletion of the short arm of chromosome 17 (17p13.1) being one of the most powerful predictors of poor outcomes. This study explores real-world outcomes in newly diagnosed MM patients with del(17p), who underwent upfront autologous hematopoietic stem cell transplantation (auto-SCT) at our institution.

Methods: In this single-center retrospective study, we included newly diagnosed MM patients with del(17p) who received an upfront auto-SCT between 2008 and 2018. Primary endpoints were progression-free survival (PFS) and overall survival (OS), and secondary endpoints being hematological response and minimal residual disease (MRD) status post-auto-SCT.

Results: A total of 115 patients with del(17p) were included. Median age at auto-SCT was 62 years (range 34-83) and 63 (55%) were male. Concurrent high-risk cytogenetics abnormalities included t(4;14), t(14;16), and 1q21+ in 13%, 7%, and 22% of patients, respectively. Induction regimens included bortezomib, lenalidomide, and dexamethasone (VRd) (n=40, 35%), cyclophosphamide, bortezomib and dexamethasone (CyBorD) (n=25, 22%), bortezomib and dexamethasone (Vd) (n=19, 17%) and carfilzomib, lenalidomide and dexamethasone (KRd) (n=15, 13%). Melphalan was the most frequently used conditioning regimen (75%), and 70% received post-transplant maintenance therapy. After induction, 10% and 51% achieved ≥ CR and ≥ VGPR, respectively, with 26% (29/112) reaching MRD negative ≥ VGPR. Overall, 22% and 42% of patients achieved ≥ CR at Day 100 and at best post-transplant response evaluation, respectively, while 69% and 83% achieved ≥ VGPR at the same timepoints, respectively. A MRD negative ≥ VGPR was seen in 55% (48/87) of patients post-transplant. Median follow-up was 31.4 months (range 3.1-199.1). Median PFS and OS for the entire cohort was 19.9 and 71.5 months, respectively, and 5-year OS was 53%. Concurrent del(17p) and t(4;14) were associated with significantly worse outcomes, with median PFS and OS of 11.5 and 22.4 months, respectively. In multivariable analysis (MVA) for PFS, female sex was associated with worse PFS (hazard ratio [95% CI]: 2.74 [1.66-4.50], p < 0.001), while MRD negative response post-transplant (0.35 [0.18-0.69], p=0.002) and the use of post-transplant maintenance therapy (0.49 [0.29-0.82], p=0.007) were associated with improved PFS. In MVA for OS, female sex (2.31 [1.24-4.29], p=0.008) and presence of t(4;14) (2.63 [1.13-6.10], p=0.025) were associated with worse OS while KPS of ≥ 90 (0.49 [0.25-0.97], p=0.041) was associated with improved OS.

Conclusions: This study affirms del(17p) as a high-risk cytogenetic abnormality with unfavourable outcomes despite modern therapies. The co-occurrence of del(17p) and t(4;14) was associated with particularly poor outcomes. These high-risk patients may benefit from the incorporation of newer treatment modalities including bispecific antibodies and chimeric antigen receptor T (CAR-T) cells early in the course of treatment.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P538 IMPACT OF DARA-VTD INDUCTION THERAPY ON HEMATOPOIETIC STEM CELL COLLECTION AND ENGRAFTMENT IN MULTIPLE MYELOMA PATIENTS ELIGIBLE FOR ASCT: RESULTS OF THE REAL-LIFE PRIMULA STUDY

Vanda Strafella1, Immacolata Attolico 2, Francesco Tarantini1,2, Paola Carluccio2, Paola Curci2, Nicola Sgherza2, Rita Rizzi1,2, Angelo Ostuni2, Gabriele Buda3, Maria Livia Del Giudice3, Viviana Beatrice Valli4, Giuseppe Mele5, Candida Rosaria Germano6, Angela Maria Quinto7, Giulia Palazzo8, Massimiliano Arangio Febbo9, Lorella Melillo10, Nicola Di Renzo11, Francesco Albano1,2, Pellegrino Musto1,2

1Aldo Moro University School of Medicine, Bari, Italy, 2AOUC Policlinico, Bari, Italy, 3University of Pisa, Pisa, Italy, 4ASST Ovest Milanese Legnano Hospital, Legnano, Italy, 5Ospedale Antonio Perrino, Brindisi, Italy, 6Ospedale Monsignor R. Dimiccoli, Barletta, Italy, 7IRCCS - Istituto Tumori “Giovanni Paolo II”, Bari, Italy, 8San Giuseppe Moscati Hospital, Taranto, Italy, 9Veneto Institute of Oncology IOV-IRCCS, Padova, Italy, 10Ospedali Riuniti, Foggia, Italy, 11Vito Fazzi Hospital, Lecce, Italy

Background: Induction therapy with daratumumab/bortezomib/thalidomide/dexamethasone (D-VTd) is currently a standard of care in transplant eligible patients with newly diagnosed Multiple Myeloma (MM). Some data, however, would suggest that daratumumab could influence stem cell mobilization and engraftment after autologous stem cell tranplantation (ASCT). The multicenter, observational, retrospective, “real life” PRIMULA study (ImPact of induction theRapy with D-VTd on collection of hemopoietic stem cells and engraftment In patients with MULtiple myeloma eligible to Autologous transplantation) aims to identify the impact of daratumumab on peripheral blood hematopoietic stem cell (PBSC) mobilization, collection, and post-transplant engraftment.

Methods: One hundred five MM patients from 10 italian centers were enrolled from February 2022 to July 2023. Patients characteristics are shown in the table. After 4-6 cycles of D-VTd induction (Dara-group), stem-cell mobilization was performed with G-CSF only or chemotherapy (cyclophosphamide 2-4g/m²) plus G-CSF. Plerixafor was administered as preemptive or salvage therapy. Patients underwent ASCT after conditioning with melphalan (100-200mg/m²). Data were compared with an historical cohort of 43 patients treated with VTd (non-Dara-group).

CHARACTERISTICS

D-VTd

Age at diagnosis (years)

(N=105)

Median (range)

61 (36-69)

Sex

(N=105)

Male/ Female

62 (59%) /43 (41%)

Paraprotein

(N=105)

IgG/ IgA/ Kappa or lambda/ Non secretory-other

41(39%) / 19(18%) / 23(22%) / 22(21%)

ISS stage

(N=90)

I/II/III

46(51%) / 24(27%) / 20(22%)

R-ISS stage

(N=76)

I/II/III

28(37%) / 35(46%) / 13(17%)

Creatinine (mg/dL)

(N=105)

Median (range)

0,89 (0,3-8)

Bone Marrow plasma cells (%)

(N=105)

Median (range)

50 (10-98)

Extramedullary disease (EMD)

(N=105)

Yes/ No-not evaluated

14(13%) / 91(87%)

Osteolytic lesions

(N=105)

Yes/No

82(78%) / 23(22%)

Results: In the Dara-group, a lower dose of CD34+ was collected (median:6.81x106/kg vs 8.67x106/kg in the non-Dara-group; p<0.0001) (Figure) and plerixafor was administered in a higher number of patients (69/101 evaluable vs 10/43; p<0.0001). Fifty of 105 (47%) patients in Dara-group and 11/43 (26%) in non-Dara-group had a number of apheresis ≥2 (p=0.009).

Seventeen patients (16%) in the Dara-group required a second mobilization attempt vs 1 (2%) in the non-Dara-group (p=0.024). Among the 17 patients in the Dara-group who underwent a second mobilization procedure, 3 (18%) had an additional mobilization failure, while 14 patients (82%) completed the mobilization. “Proven poor mobilizers” were 7 (7%) in the Dara-group and 0 in the non-Dara-group (p=0.1). Ninety-one of 105 (87%) patients in the Dara-group have currently performed their first transplant. At the first ASCT, a lower median number of CD34+ was reinfused in the Dara-group (4x106/kg vs 4.5x106/kg; p=0.0079). Median time to neutrophil and platelet engraftment in the Dara-group was longer than in the non-Dara-group (11 days vs 10 days, p<0.0001 and 13 days vs 11 days, p<0.0001, respectively) (Figure). No correlation was observed between different doses of G-CSF and cyclophosphamide administered and CD34+ yield (p=0.6 and 0.2 respectively).

Among variables such as age, sex, FISH alterations, ISS, R-ISS, bone marrow plasma cells at diagnosis, response to induction therapy, number of cycles of D-VTd, prior radiotherapy, osteolytic lesions on CT or PET/CT, focal lesions on MRI, extramedullary disease, only the male sex had a statistically significant positive relationship with the success of mobilization (multivariate analysis: p=0.007, univariate analysis: p=0.001).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (61)

Conclusions: PRIMULA study evaluates impact of Daratumumab on CD34+ mobilization, harvest and autologous stem cell transplant in one of the largest, real-life cohort of patients reported in literature. Our preliminary data (we plan to expand to patients from 19 italian centers) confirms that first line Daratumumab negatively impacts on CD34+ yield, number of apheretic procedures, plerixafor use, number of mobilization attempts and time to engraftment, without worsening transplant outcome. However, this could have implications when planning a double ASCT strategy.

Clinical Trial Registry: ID RSO 249.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P539 SAFETY AND EARLY EFFICACY OF TANDEM AUTOLOGOUS STEM-CELL TRANSPLANTATION AFTER DARATUMUMAB VTD INDUCTION IN NEWLY DIAGNOSED MULTIPLE MYELOMA

Carmine Liberatore1, Elena Rossi2, Lara Malerba3, Francesca Fioritoni1, Francesca Di Landro2, Francesca Fazio4, Silvia Ferraro5, Giusy Antolino6, Laura De Padua7, Ugo Coppetelli8, Velia Bongarzoni9, Stefano Pulini1, Patrizia Chiusolo2, Doriana Vaddinelli1, Annalisa Natale 1, Giuseppe Visani3, Valerio De Stefano2, Mauro Di Ianni1

1Santo Spirito Civil Hospital, Pescara, Italy, 2Catholic University, Fondazione Policlinico Universitario ‘A Gemelli’ IRCCS, Rome, Italy, 3Hematology and Hematopoietic Stem Cell Transplant Center, AORMN, Pesaro, Italy, 4Azienda Ospedaliera Policlinico Umberto I, Sapienza University of Rome, Rome, Italy, 5Hematology Unit, S. Eugenio Hospital, Rome, Italy, 6Hematology, Sant’Andrea University Hospital, Sapienza University of Rome, Rome, Italy, 7Hematology Unit, Fabrizio Spaziani Hospital, Frosinone, Italy, 8Hematology and Transplant Unit, Santa Maria Goretti Hospital, AUSL, Latina, Italy, 9San Giovanni Addolorata Hospital Complex, Rome, Italy

Background: The introduction of anti-CD38 monoclonal antibodies in frontline treatment of newly diagnosed multiple myeloma (NDMM) significantly improved response rates and survival, nonetheless high-risk patients still have dismal outcomes. Whereas CASSIOPEIA trial included Daratumumab/Bortezomib/Thalidomide/Dexamethasone (Dara-VTd) induction followed by single autologous stem-cell transplantation (ASCT) and consolidation, previous studies clearly showed a survival advantage with tandem ASCT in HR-NDMM and several ongoing trials still include tandem ASCT after anti-CD38-based quadruplet induction.

Methods: We report a retrospective collaborative multicenter analysis of HR-NDMM who underwent tandem ASCT following Dara-VTd induction as per clinical practice. Criteria for definition of HR-NDMM included either the presence of high-risk FISH abnormalities as del17p13, t(4;14), t(14;16), gain/ampl1q21, advanced disease stage (R-ISS stage 3, R2-ISS stage 3-4), extramedullary disease (EMD) or less than complete response (CR) after I ASCT.

Results: From 1st December 2021 to 30th September 2023, 40 HR-NDMM consecutively received Dara-VTd induction followed by tandem ASCT at our institutions. Median age at diagnosis was 52 years (range: 32-70), 15% were R ISS 3 and 35% were R2-ISS 3-4. Twenty-two patients (55%) had high-risk FISH abnormalities and 3 patients (8%) had EMD. After a median of 4 Dara-VTd cycle (range: 4-6) overall response rate (ORR) was 100%, with 40% VGPR and 31% CR/sCR (Figure 1). Following stem-cell mobilization with cyclophosphamide and G-CSF, patients underwent leukapheresis and collected a median total amount of 10,3 x106CD34+cells/kg (range: 6,5-34); 33% required Plerixafor. After a median of 207 days from start of induction (range: 168-310), 40 patients underwent ASCT. Median number of infused CD34+cells was 4,6 x106/kg (range: 2,6-8). Patients obtained stable neutrophils and platelets engraftments after a median of 12 days (range: 9-14) and 15 days (range: 7-21), respectively. Transplantation characteristics and toxicities are reported in Table 1. After I ASCT, response rates deepened, with 38% VGPR and 55% CR/sCR. After a median of 129 days from I ASCT (range: 82-242), 40 patients underwent tandem ASCT because of high-risk FISH abnormalities (n=17; 42%), advanced disease stage (n=3, 8%), EMD (n=2, 5%), less than CR after I ASCT (n=12, 30%) or other (n=6, 15%). Median number of infused CD34+cells was 4,6 x106/kg (range: 2,1-8,5). Neutrophils and platelets engraftments were obtained after a median of 11 days (range: 7-14) and 14 days (range: 7-22), respectively. Transplantation characteristics and toxicities are reported in Table 1. Response rates further improved after tandem ASCT, with 25% VGPR and 68% CR/sCR. Twenty patients (50%) also received 2 cycles of Dara-VTd consolidation, without relevant toxicities, whereas 30 patients (75%) already started maintenance. At last disease assessment, one patient had disease progression and ORR was 98% (VGPR 20% and CR/sCR 76%). After a median follow up of 573 days (range: 350-745), all patients were alive.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (62)

Table 1. Characteristics of autologous stem-cell transplantations (ASCT)

I ASCT II ASCT

Days from start of induction to ASCT: median (range)

207 (168-310)

342 (279-453)

Conditioning regimen: n (%)

• Melphalan 200 mg/m2

37 (92%)

7 (92%)

• Melphalan 140 mg/m2

3 (8%)

3 (8%)

Number of infused CD34+cells x106/kg: median (range)

4,6 (2,6-8)

4,6 (2,1-8,5)

Days to engraftments: median (range)

• Neutrophils

12 (9-14)

11 (7-14)

• Platelets

15 (7-21)

14 (7-22)

Bacterial adverse events: n (%)

• Febrile neutropenia (grade 3)

12 (30%)

7 (18%)

• Sepsis (grade 3)

2 (5%)

4 (10%)

• Pneumonia (grade 3)

0 (0%)

4 (10%)

• Soft tissue infection (grade 2)

1 (2%)

0 (0%)

• Urinary tract infection (grade 2)

1 (2%)

1 (2%)

Viral adverse events: n (%)

• CMV viremia (grade 2)

4 (10%)

3 (7%)

• CMV viremia (grade 3)

0 (0%)

1 (2%)

• HHV6 viremia (grade 2)

0 (0%)

2 (5%)

• Respiratory syncytial virus infection (grade 2)

0 (0%)

1 (2%)

Fungal adverse events: n (%)

• Probable IFI (grade 3)

0 (0%)

1 (2%)

• Candidemia (grade 3)

0 (0%)

1 (2%)

Other adverse events: n (%)

• Atrial fibrillation (grade 3)

2 (5%)

2 (5%)

• Pulmonary edema (grade 3)

0 (0%)

1 (2%)

• Deep vein thrombosis (grade 2)

0 (0%)

1 (2%)

Late hematological adverse events: n (%)

• Neutropenia (grade 3)

0 (0%)

1 (2%)

• Platelet count decreased (grade 3)

0 (0%)

1 (2%)

Conclusions: For the first time to our knowledge and in real-life setting, tandem ASCT proved feasible in HR-NDMM who received Dara-VTd induction. Transplantation outcomes were favorable, with both limited hematological and non-hematological toxicities. Although longer follow-up is required to further elucidate its benefit, tandem ASCT proved effective to increasing the depth of response in HR-MM.

Clinical Trial Registry: NA.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P540 RESPONSE AND SURVIVAL IMPROVEMENT WITH AUTOLOGOUS STEM CELL TRANSPLANTATION IN PATIENTS WITH NEWLY DIAGNOSED MULTIPLE MYELOMA: DIFFERENT INDUCTION THERAPY IMPACT OVER THE LAST 30 YEARS

José Miguel Mateos Pérez 1,2, Francesc Fernández-Avilés3,2, Carlos Fernández de Larrea1,2, Maria Teresa Cibeira1,2, Natalia Tovar1,2, Carmen Martínez3,2, Luis Gerardo Rodrígez-Lobato1,2, Maria Suárez-Lledó3,2, Maria Queralt Salas3,2, Montserrat Rovira3,2, Joan Bladé1,2, Laura Rosiñol1,3,2

1Amyloidosis and Multiple Myeloma Unit, Barcelona, Spain, 2Hospital Clínic of Barcelona, IDIBAPS, University of Barcelona, Barcelona, Spain, 3Hematopoietic Transplantation Unit, Barcelona, Spain

Background: Autologous stem cell transplantation (ASCT) is the standard of care in younger patients with newly diagnosed multiple myeloma (NDMM). Several induction regimens have been used with different outcomes after ASCT. However, the impact of ASCT on response improvement is not well known.

Methods: Patients with NDMM who underwent ASCT at the Hospital Clinic of Barcelona were retrospectively analyzed to assess the impact of ASCT on overall response rates (ORR) and survival according to the different induction regimen. Patients refractory to induction therapy or those who received more than one regimen before ASCT were excluded from the study. Statistical and survival analysis were performed with STATAv16.1.

Results: From January 1990 to June 2022, 350 patients with NDMM underwent ASCT. Median age at diagnosis was 56.5 years (IQR, 49.5-62.3) and 54% were male. One hundred forty-nine patients (42.6%) received induction therapy with chemotherapy (QT), 40 (11.4%) with bortezomib-based (PI) and 155 (44.3%) with immunomodulatory-based therapy (IMiD) (Table 1). The most frequent conditioning regimen was melphalan (300, 85.7%), followed by busulfan plus melphalan (29, 8.3%) and melphalan plus total body irradiation (20, 5.7%).

In the overall series, the ORR before ASCT was 88.2% (19.1% CR, 25.4% VGPR, 43.7% PR) and increased up to 94% after transplant (43.4% CR, 24.6% VGPR, 26.0% PR). The median increase in CR was 24.3%, being 19.7% in the QT group, 31.4% in the PI group and 26.2% in the IMID group, without significant differences between groups (p=0.216). After excluding the patients who were in CR at the time of ASCT, 144 patients (50.9%) deepened the response at day 100 post-ASCT, with no significant differences between those treated with QT, PI or IMIDs (46.5% vs. 70.0% vs. 50.4%) (p=0.068). Response rates before and after ASCT by treatment group are detailed in Table 1.

The median progression free survival (PFS) since diagnosis in the overall series was 45.8 months, being significantly longer in the IMiD group compared with the PI and QT groups (52.8 vs 40.7 vs 39.1 months, p=0.0019). The overall survival (OS) in the overall series was 123.5 months. Patients treated with IMID-based regimens had a significantly longer OS compared with patients treated with PI or QT (median not reached vs 121.3 v 86.7 months, p=0.0001). PFS and OS by treatment groups are shown in Figure 1.

PRE-ASCT

RESPONSE

POST-ASCT RESPONSE (Day 100)

Chemotherapy (QT)

(N=149)

CR

20 (13,4%)

49 (33,1%)

VGPR

14 (9,4%)

24 (16,2%)

PR

86 (57,7%)

61 (41,2%)

MR

18 (12,1%)

5 (3,4%)

Stable disease

4 (2,7%)

1 (0,8%)

Progression

7 (4,7%)

7 (4,7%)

Early death

-

1 (0,6%)

Bortezomib-based (PI)

(N=40)

VD/VCD

CR

10 (25,0%)

22 (56,4%)

VGPR

11 (27,5%)

8 (20,5%)

PR

14 (35,0%)

8 (20,5%)

MR

3 (7,5%)

-

Progression

2 (5,0%)

-

Early death

-

1 (2,6%)

IMID-based (IMiD)

CR

36 (23,2%)

76 (49,4%)

(N=155)

VGPR

63 (40,7%)

52 (33,8%)

TD

PR

49 (31,6%)

20 (13,0%)

VTD

MR

3 (1,9%)

1 (0,6%)

VRD

Stable disease

-

1 (0,6%)

KRD

Progression

4 (2,6%)

4 (2,6%)

Table 1. Grade of response before and after ASCT by treatment group. CR: complete response; VGPR: very good partial response; PR: partial response; MR: minimal response. ASCT: autologous stem cell transplantation.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (63)

Conclusions: ASCT improves the CR rate in about one-fourth of the patients. The increase in CR rate was similar in patients treated with novel drugs compared with those treated with conventional chemotherapy. Response was deepened in half of the patients who were not in CR before ASCT irrespective of the type of induction regimen. A significant survival benefit has been observed over the last 30 years with the introduction of the new drugs.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P541 OUTCOMES OF FRAIL PATIENTS RECEIVING HIGH-DOSE CHEMOTHERAPY/AUTOLOGOUS STEM CELL TRANSPLANTATION FOR MULTIPLE MYELOMA

Stephanie Yohay 1, Temitope Oloyede2, Binod Dhakal1, Anita D’Souza1, Ayesha Aijaz3, Meera Mohan1, Ravi Narra1, Marcelo Pasquini1,2, Mehdi Hamadani1,2, Ciara Louise Freeman4, Othman Salim Akhtar1,2

1Medical College of Wisconsin, Milwaukee, United States, 2Center for International Blood and Marrow Transplantation, Milwaukee, United States, 3We Care Scan Center, Srinagar, Kashmir, India, 4Moffitt Cancer Center, Tampa, United States

Background: Multiple studies have demonstrated comparable outcomes in older adults with multiple myeloma (MM) receiving high-dose chemotherapy and autologous hematopoietic cell transplantation (autoHCT). Despite these reassuring data, access to autoHCT remains poor in this subgroup, often due to physician perception of frailty. Geriatric screening tools are superior to physician judgement in identifying frail patients at risk for poor outcomes. In patients with non-transplant eligible MM, a simplified frailty index (SFI, Facon et al, Leukemia 2020) predicts survival but data is limited in the transplant-eligible setting. In this prospective observational cohort study, we examine the association of an adapted version of the SFI with survival outcomes in patients with MM receiving frontline autoHCT.

Methods: We used data made publicly available by the Center for International Blood and Marrow Transplantation Research (CIBMTR) post-publication (Munshi et al, 2020). We included all patients in the database receiving melphalan/autoHCT for MM within 12 months of diagnosis between 2013 and 2017. We used an adapted version of the SFI which combines an evaluation of age (76-80 years, 1 point, >80 years, 2 points), eastern cooperative oncology group performance status (ECOG PS 1, 1 point, ECOG PS ≥2, 2 points) and comorbidity for which we used the hematopoietic cell transplantation specific comorbidity index (HCT-CI) in lieu of the Charlson comorbidity index (score of ≥2, 1 point). Patients were classified as frail (score ≥2) or non-frail (score 0-1). The primary outcome was overall survival (OS). Secondary outcomes included non-relapse mortality (NRM), progression-free survival (PFS) and relapse rate.

Results: Overall, 15,818 patients were included with a median follow up of 25 months (range, 0.4 - 25 months). Using the adapted SFI, 4950 (31.3%) patients were classified as frail; n=1 patient was classified as frail because of age >80 years alone. A higher proportion of patients in the frail group had DSS/ISS stage III at diagnosis (frail vs non frail, 60.7% vs 51.7%, p<0.01) and high-risk cytogenetics (29.0% vs 26.8%, p<0.01). The NRM at day 100 was higher in frail patients compared to non-frail (1% vs 0.5%, p<0.01) but there was no difference in probability of relapse at 24 months (30% vs 29.7%, p=0.29). Frail patients had inferior PFS (24-months, 66.6% vs 68.7% p<0.01) and OS (24-months, 86.9% vs 90.5%, 95% CI, p<.01). Of the 12,904 patients who received full-dose melphalan at 200 mg/m2, frail patients (n=3513, 27.2%) had higher NRM at day-100 (0.7% vs 0.4%, p<.01) and inferior OS at 24-months (83.4% vs 89.1%, p<0.01). Melphalan dose was reduced to 140 mg/m2 in 28.9% of frail patients versus 13.5% in the non-frail group, p<0.01. Among patients who received melphalan 140 mg/m2 (n=2900), NRM at day 100 was significantly higher in frail patients (1.7% vs 1.0%, p<0.01) and OS was lower (24-month, 83.4% vs 89.1%, p<0.01).

Conclusions: In this large cohort of patients with MM receiving frontline autoHCT, around 1/3rd were classified as frail by the adapted-SFI with increased NRM and inferior OS seen in these patients. Despite this, rates of NRM were low (<2%) in all frailty subgroups.

Table 1: Baseline characteristics and outcomes in patients with MM receiving upfront autoHCT

Characteristics

Frail (n=4950)

Non-frail (n=10,868)

P-Value

Total (n=15818)

Follow-up - median (range)

24.7 (0.9-25.0)

25.0 (0.4-25.0)

25.0 (0.4-25.0)

Median age at HCT, years (min-max)

63.2 (20.2-83.2)

60.9 (25.2-80.8)

<.01a

61.6 (20.2-83.2)

Male no. (%):

2702 (54.6)

6363 (58.5)

<.01b

9065 (57.3)

ECOG PS ≥1, no. (%):

4895 (98.9)

2365 (21.8)

<.01b

7260 (45.9)

HCT-CI ≥2, no. (%):

4924 (99.4)

4556 (41.9)

<.01b

9480 (60)

DDS/ISS stage III at diagnosis, no. (%)

3006 (60.7)

5616 (51.7)

<.01b

8622 (54.5)

High-risk Cytogenetics, no. (%):

1435 (29.0)

2911 (26.8)

<.01b

3400 (21.5)

MEL dose, no. (%):

MEL at 140mg/m2

1431 (28.9)

1469 (13.5)

<.01b

2900 (18.3)

MEL at 200mg/m2

3513 (71.0)

9391 (86.4)

12904 (81.6)

Disease Status prior to SCT, sCR/CR, no. (%)

808 (16.3)

1685 (15.5)

0.07b

2493 (15.8)

NRM at day 100 - est. (95% CI):

1.0% (0.7-1.3)

0.5% (0.4-0.7)

<.01c

0.7% (0.6-0.8)

Relapse at 24-months - est. (95% CI)

30.0% (28.5-31.5)

29.7% (28.7-30.7)

0.29c

29.8% (29.0-30.6)

PFS at 24 months - est. (95% CI)

66.6% (65.1-68.1)

68.7% (67.7-69.7)

<.01d

68.1% (67.2-68.9)

OS at 24 months - est. (95% CI)

86.9% (85.8-87.9)

90.5% (89.8-91.1)

<.01d

89.4% (88.8-89.9)

  1. a Fisher exact test via Mon b Pearson chi-square test, cGray test, d Log-rank test
  2. Abbreviations: HCT-CI, hematopoietic stem cell transplantation comorbidity index; DSS: Durie Salmon Staging; ISS, International Staging System; MEL, melphalan.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P542 BASELINE INVESTIGATIONS AND AUTOLOGOUS STEM CELL TRANSPLANT OUTCOMES: A RETROSPECTIVE REVIEW OF ECHOCARDIOGRAPHY AND PULMONARY FUNCTION TESTS

Yousif Badri 1, Amany Ihab Mohamed1, Gordon Cook1, Roger Owen1, Simon Bulley1, Sylvia Feyler1, Christopher Parrish1, Frances Seymour1

1Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom

Background: Echocardiography (Echo) and Pulmonary Function Tests (PFT) are established baseline tests prior to autologous stem cell transplantation (ASCT), mandated within most clinical trials, and undertaken as a standard of care in most centres. It is unclear whether abnormalities identified have value in assessing ASCT prognosis or for patient selection.

Methods: We conducted a retrospective review of adult patients who underwent ASCT for plasma cell disorders between 2020 and 2022 at a single centre. Results of Echo, PFT and other baseline pre-ASCT investigations, admission, discharge and outcome data were collected from the electronic patient record. Statistical analyses were conducted using bivariate analyses and independent samples t-tests.

Results: 102 (63 male 39 female) adult patients (median age 60.5) were eligible for analysis. The majority (95%) had a background of multilple myeloma (MM). Patients received 1-9 cycles of induction chemotherapy (median 4), most commonly bortezomib-thalidomide-dexamethasone (40.2%) and botezomib-cyclophosphamide-dexamethasone (34.3%). Responses to induction therapy were: CR 16 (15.7%), VGPR 58 (56.9%), PR 27 (26.5%), MR 1 (1%). 88.2% of ASCTs were the patient’s first high-dose procedure. Median stem cell dose infused was 3.05x106 CD34+/kg (range 1.95-12.1).

Baseline echo revealed 2% of patients were in atrial fibrillation, with 3.9% being in each of sinus bradycardia and sinus tachycardia at the time of imaging. Left ventricular ejection fraction was >55% in 77 patients (not quantified in 20). At least mild diastolic dysfunction was noted in 40.2%. Abnormalities of one or more cardiac valves were noted in 28 patients (one valve in 21 patients, 5 in 2, 3 in 1 and all 4 valves in patient); these were almost entirely regurgitant lesions (1 patient had moderate aortic stenosis). A composite cardiac risk score was devised incorporating LFEV<55%, diastolic abnormalities, and valve abnormalities). PFTs revealed a median TLC pred: 93.5% (range 51.6-120%), FEV1 pred 94.9% (54-139) and DLCOc pred 79% (41-115).

Patients with at least 1 valvular abnormality had a significantly shorter time to first antibiotics (p=0.001). The composite cardiac score was also predictive of the likelihood of positive blood cultures at the time of starting antibiotics (p=0.015). LVEF was significantly correlated with performance status as assessed by the treating clinician prior to Echo (p=0.005).

FEV1 pred significantly correlated with the likelihood of prolonged neutropenia (neutrophils <0.5; p=0.007) and readmission before D100 (p=0.04). TLC pred was predictive of obtaining positive blood cultures during admission. DLCOc pred was found to fall with increasing cycles of induction chemotherapy (p=0.001).

Conclusions: Retrospective evaluation of echo and PFTs from patients deemed fit for ASCT reveal a range of qualitative and quantitative abnormalities of cardiorespiratory function, many of which are correlated with increased rates of autograft-associated complications. However, within the range of echo and PFT results deemed acceptable for ASCT, the traditionally employed parameters (e.g. LVEF, DLCOa) do not appear to be predictive of autograft outcomes, which may suggest interpretation of pre-ASCT can be improved to refine patient selection.

Disclosure: None.

27: Multiple Myeloma

P543 IMPROVED SURVIVAL OF MULTIPLE MYELOMA PATIENTS IN RELAPSE AFTER AUTOLOGOUS STEM CELL FOR THOSE WHO PREVIOUSLY ACHIEVED COMPLETE REMISSION AFTER TRANSPLANT

Nour Moukalled 1, Ammar Zahreddine1, Iman Abou Dalle1, Jean El Cheikh1, Ali Bazarbachi1

1American University of Beirut, Beirut, Lebanon

Background: Autologous stem cell transplant (ASCT) remains a standard of care for patients with multiple myeloma. Unfortunately, most patient will relapse after transplant. The disease status at transplant has been suggested to play a pivotal role in predicting post-ASCT outcomes including first and second progression free survival (PFS1 and PFS2 respectively); as well as overall survival (OS). However, the impact of achieving complete remission (CR) or better after ASCT on the survival from post-transplant relapse remains largely unknown.

Methods: This is a retrospective single center analysis of consecutive patients with multiple myeloma who received their first ASCT between 2010 and 2020. We analyzed data according to whether patients achieved or not CR at day 30 to 60 post ASCT.

Results: We identified a total of 183 patients of whom 122 had documented progression post ASCT. The median age for this group was 55.9 (34.9-74.9). High risk cytogenetics was seen in 18.7% of the total cohort. The most common type of induction regimen used was bortezomib-lenalidomide-dexamethasone (VRD) in around 72% of patients. Only 2% received a quadruplet-based induction. Disease status at transplant was partial response (PR) in 35.4%, very good partial response (VGPR) in 58.3% and CR in 6.3%. Most patients (91.7%) received full dose melphalan (200 mg/m2) conditioning. Complete remission post-transplant was achieved in 51 patients (41.8%) (CR group) compared to 71 patients failing to achieve CR (no-CR group). No statistically significant difference was noted when comparing baseline patient and disease characteristics between CR and non-CR groups except for gender where 92.3% of non-CR group were males as compared to 59.1% in the CR group (p value 0.008). The median follow up was 60 months. During this follow up, a total of 48 patients died including 15 and 33 of the CR and non-CR respectively (p value 0.001). The median year of transplant was 2014 and 2015 in the CR and non-CR groups respectively (p value 0.1). The PFS1 and PFS2 were respectively in the CR and non-CR groups, 90 versus 58 months (p value 0.001), and 25 versus 14 months (p value 0.04). Importantly, the overall survival from the time of post-transplant relapse was significantly better in the CR group (median not reached) as compared to around 22 months in the no-CR group (HR 0.41; p value <0.001). On multivariate analysis, the effect of achieving CR after transplant was maintained when correcting for age at transplant, type of myeloma, cytogenetic risk group, melphalan dose and time to transplant.

Conclusions: Achieving CR or better post-ASCT is associated with improved PFS2 and improved OS from the time of post-transplant relapse indicating that these patients have a more sensitive disease that would respond better to subsequent treatments. These data are specifically significant with the currently expanding treatment landscape for patients with multiple myeloma and suggest an important endpoint in trials as well as an additional factor to take into consideration when deciding on treatment sequencing in the future.

Disclosure: None.

27: Multiple Myeloma

P544 NON-CRYOPRESERVED PERIPHERAL BLOOD STEM CELL AUTOLOGOUS TRANSPLANTATION FOR MULTIPLE MYELOMA AS A SAFE ALTERNATIVE IN COUNTRIES WITH LOW RESOURCES: A NINE-YEARS OF BICENTRIC EXPERIENCE

Siham Ahchouch 1, Othman Doghmi1, Selim Jennane1, El Mehdi Mahtat1, Sara Bougar2, Saadia Zafad3, Hicham El Maaroufi1, Kamal Doghmi1

1Military Hospital of Instruction Mohammed V, Rabat, Morocco, 2Tissue and Stem Cells Bank, Rabat, Morocco, 3Al Madina Private Clinic, Casablanca, Morocco

Background: Autologous hematopoietic stem cell transplants(AHSCT)are the standard of care for newly diagnosed and fit young patients with Multiple Myeloma(MM). Although cryopreservation is routinely used in developed countries, AHSCT can be performed using non-cryopreserved stem cells. The objective of this study is to evaluate the safety and toxicity of autologous transplantation of non-frozen peripheral blood stem cells(PBSCT)in Moroccan patients with multiple myeloma(MM).

Methods: We conducted a retrospective bicentric study with all newly diagnosed MM patients who underwent PBSCT without cryopreservation in the clinical hematology department of the Mohammed V military hospital and at the Al Madina Clinic, between January 1,2015 and December 15, 2023. In our context, AHSCT is not proposed to patients >70years old, those with a cardiac ejection fraction less than 50%, or to patients with liver failure, progressive viral hepatitis. Incomplete files were removed from the study. PBSC were mobilized with lenograstim alone or in association with plerixafor depending on the number of CD34+ cells circulating in peripheral blood counted on the fourth day after the start of mobilization. The stem cells collected were stored for 24-48hours in a blood bank refrigerator at a temperature of 4°C. After standard conditioning with high-dose Melphalan, peripheral blood stem cells were reinjected 24h after conditioning.

Results: During the study period,112patients with multiple myeloma were proposed for a PBSCT. Mobilization failure was observed in ten patients, one patient had a Covid-19 infection and another persistent thrombocytopenia. The remaining 100patients underwent AHSCT using non-Cryopreserved PBSC. The median age was 55,5years (range 31-67)with a sex ratio M/F of 1,4. Fifteen(15%)patients had light chain myeloma, eleven (11%)a MM with IgA, and one patient with IgM isotype. Nine patients had a poor prognostic cytogenetic profile and twenty-four had an ISS of 3. Thirteen patients received at least two lines of treatment, and four of them underwent a double autograft. The median time from diagnosis to autograft was 5 months(range 2-115). At the time of transplantation, more than half of the included patients responded to treatment:eleven of them were in complete remission,24 and 65 were in partial response and in very good partial response, respectively. Eighty-two(82%)patients had low-risk HCT-IC. Only two patients received Melphalan conditioning regimens at a dose of140mg/m2(creatinine clearance<30 mg/ml/min). Five patients received lenograstim mobilization in association with plerixafor. In 71 cases, a single session of cytapheresis was required to obtain a median richness of 4.06x106 CD34/kg(range:2,0-12.2). The time required for neutrophil and platelet recovery was 11days(range:7-18) and11days(range: 9-19)respectively. In post-transplant phase, the most frequent non-hematological gradeIII-IV toxicities were mucositis(4%), vomiting(5%)and diarrhea(10%). Parenteral nutrition was necessary to eleven of them. Febrile neutropenia occurred in 55 cases, with 37 documented infections(gastro-intestinal tract including C. Difficile Infection, pulmonary tract, blood culture, urinary and cellulitis). Two patients had neurological complications, seven(7%)a thromboembolic events, one an acute pancreatitis and another a reactivation of viral hepatitis B revealed by membranoproliferative glomerolonephritis. Fifty-two(52%)patients were transfused with median of 2(range0-8)packed red blood cells(PRBC) and 93(93%)with a median of 2(range:0-15) platelet concentrates(standard platelet concentrate or apheresis platelet concentrate). The median duration of hospitalization was 19,5days(range11-48). Two patients died within 100days after transplantation, with transplant-related mortality(TRM)of 2%.

Conclusions: Non-cryopreserved autologous grafts are simple, safe, efficient, and can lead to short- and long-term results comparable to those of their homologue with cryopreservation. This method may be used to perform autologous transplants in more patients with low-cost production, especially in developing countries.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P545 UP-FRONT HIGH DOSE MELPHALAN AND AUTOLOGOUS STEM CELL TRANSPLANTATION FOR NEWLY DIAGNOSED MULTIPLE MYELOMA - SINGLE CENTER REAL-WORLD ANALYSIS OF 165 CASES

Nobuhiro Tsukada 1, Taku Kikuchi1, Kodai Kunisada1, Yuki Oda1, Moe Yogo1, Tomomi Takei1, Kota Sato1, Mizuki Ogura1, Yu Abe1, Kenshi Suzuki1, Tadao Ishida1

1Japanese Red Cross Medical Center, Tokyo, Japan

Background: DETERMINATION study has shown that VRd induction followed by up-front ASCT and maintenance therapy until PD lead to median PFS of 67.5 months. Therefore, up-front ASCT is considered as standard care even in the era of novel agents. We conducted a retrospective analysis on the outcomes of patients received triplet induction treatment followed by up-front ASCT in a real-world setting.

Methods: Total of 165 patients received ASCT between Nov 2016 and May 2023 at Japanese Red Cross Medical Center. Patient characteristics, treatment response before and after ASCT, PFS, and OS were retrospectively analyzed.

Results: Median age was 57.5 (36-70) and 106 patients were male. Myeloma subtypes were IgG 85, IgA 30, IgD 2, IgM 1, BJ 46, and non-secretory 1. R-ISS 1, 2, and 3 were 33, 94, and 29, respectively. R2-ISS 1, 2, 3, and 4 were 34, 48, 60, and 23%. 93% of patients received VRD-based induction treatment. Among 153 evaluable patients, 146 (95%) patients received either consolidation and/or maintenance. Response before ASCT and best response at post-transplant were ≥CR 38% and ≥VGPR 77%, and ≥CR 76% and ≥VGPR 90%, respectively. 106 of 150 patients (70%) obtained MRD-negativity by multiparameter FCM (<10-5) after ASCT. Median PFS of all cases were not reached. 5-year estimated PFS and OS were 58% and 81%, respectively. PFS of patients with high-risk cytogenetics was significantly worse than others (p<0.001, 5-yr PFS 30% vs. 65%). PFS of patients with R2-ISS 4 was significantly worse than others (p<0.001, 5-year PFS 27% vs. 62%). PFS of patients obtained MRD-negativity was significantly better than others. OS of patients obtained MRD-negativity was also significantly better than others.

Conclusions: Treatment outcome is improved by triplet induction treatment, especially combination of PI and IMiDs. Post ASCT consolidation/maintenance.

may lead to improved response. MRD-negativity was correlated with extended PFS.

Disclosure: NT has received personal fees from Janssen and Sanofi.

TK has received personal fees from Janssen, Takeda, and Sanofi. TI received honoraria from Ono, Takeda, Celgene/Bristol-Myers Squibb, and Janssen.

KS has received honoraria from Takeda, Celgene, Ono, Amgen, Novartis, Sanofi, BMS, AbbVie, and Janssen; consultancy fees from Takeda, Amgen, Janssen, and Celgene; and research funding from BMS, Celgene, and Amgen.

The remaining authors declare no competing financial interests.

27: Multiple Myeloma

P546 SETTING UP A HEMATOPOIETIC STEM CELL TRANSPLANT PROGRAM IN BAHRAIN -EXPERIENCE FROM BAHRAIN ONCOLOGY CENTER

Shruti Prem Sudha1, Hazem Afify 1, Nabil Abdelfattah1, Volkan Kahraman1, Salih Aksu1, Aly Rashed1, Cigdem Ozturk1

1Bahrain Oncology Center, Muharraq, Bahrain

Background: Whereas hematopoietic stem cell transplant (HSCT) is widely accessible in developed countries, the significant infrastructure, and health care provider training that are required to provide such a service have prohibited it from being widely adopted in smaller countries. In this abstract we present a brief summary of the experience in developing an HSCT program in the country of Bahrain which has a population of approximately 1.5 million people.

Methods: The stem cell transplant (SCT) program in Bahrain Oncology Centre was initiated in December 2018 as a publicly funded program. The occurrence of the COVID-19 pandemic soon after the establishment of the transplant program was one of the challenges faced by our team which affected the intake of patients in the initial phases of the pandemic. From the period December 2018 a total of 82 autologous and 6 allogenic transplants have been performed. For the purpose of this abstract we report on the first 68 autologous transplant patients who have completed at least one-year follow-up post-transplant.

Results: Autologous transplants were performed for the following diagnoses: multiple myeloma (29) non-Hodgkin’s lymphoma (27), Hodgkin’s lymphoma (20), and germ cell tumor (7). In the autologous transplant cohort, the median age of patients was 51 years (range 22-69 years) and males comprised 63%. The median CD34 dose was 4.7x106 cells/kg (range 3-16 x106 cells /kg). The median time to neutrophil engraftment was 10 days (range 8-14 days) and median time to platelet engraftment was 12 days (range 9-30 days). Transplant related mortality (TRM) was 2.9%. The cause of death was gastro-intestinal sepsis in one patient and diffuse alveolar hemorrhage/pneumonia in the second patient.

At a median follow up of 23 months (range 1-48 months), 14 patients had relapsed (21.5%). Two patients died due to TRM and an additional 9 patients died during follow-up (13.2%) from complications of progressive disease. The 2-year overall survival was 90.5%, (95% CI 83.1-97.9%), while the 2-year progression free survival was 89.2% (95% CI 75.2-94.4%) in the entire cohort.

We also observed that Brentuximab based regimen was superior to ICE as salvage therapy prior to transplant for Hodgkin lymphoma patients.

Among NHL patients, worse outcomes were seen for secondary CNS lymphoma patients with no long term survivors in this sub-group.

Our overall clinical outcomes compare favorably with international standards. The median cost of autologous transplant at our institution was approximately $10,000 USD which is cheaper than that reported in many centers.

Conclusions: Our center is the only center currently offering HSCT in Bahrain, and we are currently a member of the EBMT registry. With the cessation of the pandemic our allogeneic transplant program is also expanding to include more indications and patients. This new program has delivered steady progress from 2019 to 2023 and is delivering transplantation services in numbers not previously delivered to Bahraini patients proving that it is possible to develop hematopoietic stem cell transplant programs in smaller countries.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P547 SALVAGE SECOND AUTOLOGOUS STEM CELL TRANSPLANTS FOR PATIENTS WITH MYELOMA: A 24-YEAR RETROSPECTIVE AUDIT IN A NATIONAL TERTIARY REFERRAL CENTRE (1999-2022)

Micheal Brennan 1, Patrick Hayden1, James Fey1, Catherine Ronayne1, Orla Fallon1, Greg Lee1, Nicola Gardiner1, Ezzat El Hassadi2, Meegahage Perera3, Helen Enright4, Johnny McHugh4, Philip Murphy5, Patrick Thornton5, John Quinn5, Jeremy Sargent5, Mary McCloy6, Peter O’Gorman7, Denis O’Keeffe8, Hilary O’Leary8, Mark Gurney1

1St. James’ Hospital, Dublin, Ireland, 2Waterford University Hospital, Waterford, Ireland, 3Midlands University Hospital, Tullamore, Dublin, Ireland, 4Tallaght University Hospital, Dublin, Ireland, 5Beaumont Hospital, Dublin, Ireland, 6Our Lady Of Lourdes Hospital, Drogheda, Ireland, 7Mater Hospital, Dublin, Ireland, 8Limerick University Hospital, Limerick, Ireland

Background: Upfront autologous stem cell transplantation (ASCT) remains the standard of care in transplant-eligible patients with multiple myeloma (MM). Salvage transplant following relapse has been shown to be an effective form of consolidation after re-induction therapy in multiple studies [1].

Methods: We conducted a retrospective analysis of the autologous stem cell transplant database for patients with MM at St. James’s Hospital (SJH), a national tertiary referral centre, and identified all patients who had had a second salvage ASCT between 1999 and 2022. Data included patient demographics, time to second transplant, and survival outcomes.

Results: Between 1999 and 2022 inclusive, 891 patients underwent ASCT for Myeloma at SJH, 810 upfront and 81 as a second salvage transplant. Activity has increased significantly over time. There were 7 salvage ASCT between 1999 and 2004, 12 (2005-2010), 25 (2011-2016) and 37 (2017-2022). The median age was 56 years (range 39-70 years) and 55 (68%) were male. The breakdown of subtypes was as follows: IgG 43 (53%), IgA (17%), Light Chain (19%) and Other (21%).

Data on re-induction regimens was available on 58/81 (72%) patients. The most common regimens used for re-induction were Rd (19%), CyBorD (17%) and RVd (34%). Rd and Vd were commonly used between 2005 and 2016. A total of 27 of 29 (93%) patients transplanted between 2017 and 2022 have received PI-IMiD combinations for re-induction (RVd, KRd, IRd). Conditioning was with single agent high dose melphalan in all cases. 58 (71%) patients received a dose of 200mg/m2 and 23 (29%) 140mg/m2. There was no transplant-related mortality within the first 100 days. Engraftment was satisfactory in all cases. The median time between the first and second transplant was 55 (range 16-156) months. Median overall survival was 56 months.

Conclusions: The use of salvage second autologous transplants in patients with MM increased significantly at our centre over the last two decades though has plateaued. The pattern of patient referrals has been relatively conservative with a median time between autologous transplants of 55 months overall and 85 months in the most recent cohort (2017-2022). It is likely that the selection of lower risk patients who were more likely to have a durable response to repeated treatment with high dose melphalan and the more recent availability of novel therapies such as anti-CD38 antibodies and bispecific antibodies such as Teclistamab and Talquetamab are factors in the superior OS in patients transplanted since 2015.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (64)

Disclosure: Nothing to declare.

27: Multiple Myeloma

P548 NOVEL DRUGS INCLUDING MONOCLONAL ANTIBODIES AND/OR DONOR LYMPHOCYTE INFUSIONS CONFERRED LONG TERM SURVIVAL TO MULTIPLE MYELOMA PATIENTS RELAPSED AFTER ALLOGENEIC STEM CELL TRANSPLANTATION

Chiara Nozzoli 1, Martina Pucillo2, Massimo Martino3, Luisa Giaccone4, Alessandro Rambaldi5, Edoardo Benedetti6, Domenico Russo7, Nicola Mordini8, Silvia Mangiacavalli9, Pietro Enrico Pioltelli10, Paola Carluccio11, Piero Galieni12, Marco Ladetto13, Simona Sica14, Miriam Isola15, Maria De Martino15, Elena Oldani5, Eliana Degrande16, Elisabetta Antonioli17, Renato Fanin2, Riccardo Saccardi1, Fabio Ciceri18, Francesca Patriarca2

1Cell Therapy and Tranfusion Medicine, Careggi University Hospital, Florence, Italy, 2Azienda Sanitaria Universitaria Friuli Centrale, DAME, University of Udine, Udine, Italy, 3Stem Cell Transplant and Cellular Therapies Unit, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy, 4Stem Cell Transplant Center, AOU Citta’ della Salute e della Scienza, Turin, Italy, 5Hematology and Bone Marrow Transplantation Unit, ASST Papa Giovanni XXIII, Bergamo, Italy, 6UO Hematology, University of Pisa, Pisa, Italy, 7University of Brescia, Brescia, Italy, 8Hematology, Azienda Ospedaliera S Croce e Carlo, Cuneo, Italy, 9Fondazione IRCCS Policlinico San Matteo, Pavia, Italy, 10Ospedale San Gerardo, Monza, Italy, 11Hematology and Stem Cell Transplantation Unit, AOUC Policlinico, Bari, Italy, 12U.O.C. Ematologia e Terapia Cellulare, Ospedale Mazzoni, Ascoli Piceno, Italy, 13Division of Hematology, Azienda Ospedaliera SS. Antonio e Biagio e Cesare Arrigo, Alessandria, Italy, 14Universita’ Cattolica del Sacro Cuore, Roma, Italy, 15Institute of Statistics, DAME, University of Udine, Udine, Italy, 16GITMO Trial office, Milano, Italy, 17Careggi University Hospital, Florence, Italy, 18Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Milan, Italy

Background: Even if allogeneic stem cell transplantation (allo-SCT) is curative for a minority of patients with multiple myeloma (MM), the patients who have relapsed after allo-SCT can experience long term survival supported by novel drugs and donor lymphocyte infusions (DLI).

Methods: We evaluated 242 multiple myeloma (MM) pts reported to the Gruppo Italiano Trapianto Midollo Osseo (GITMO) registry who underwent allogeneic stem cell transplantation (allo-SCTs) between 2009 and 2018 in order to identify predictors for long term outcome in the whole population and for prolonged OS after relapse in the subgroup of relapsed pts.

Results:

Median age at transplant was 54 years (range 29-77). ISS stage 3 was detectable in 52/121 evaluable pts (21%). High risk FISH was present in 53 out of 117 evaluable pts (45%). Allo-SCT was performed after more than 2 lines of treatments in 92 pts (38%). Myeloablative conditioning was administered to 141 allo-SCTs (58%). Stem cell source was peripheral blood for 210 allo-SCTs (87%). Donors were: 98 HLA-identical sibling (40%), 135 unrelated (55%) and 9 haploidentical (4%). Patients characteristics are reported in table1.

The cumulative incidence of TRM was 10% at one year and 28% at five years from Allo-SCT. Grade 2-4 acute GVHD incidence was 22.6% and moderate or severe chronic GVHD occurred in 35.4 % of the pts. The median OS from transplantation was 39.4 months for whole patient population and 38.5 month for the relapsed one.

Median progression free survival (PFS) of the whole population were 19 months from allo-SCT. In the multivariate model, > 2 lines of therapy before allo-SCT (p=0.012), older age (p=0.015), acute GVHD (p=0.002) and high risk FISH (p=0.017), were significant factors associated with reduced OS. Relapse after allo-SCT occurred in 118 (49%) pts at a median of 14.3 months (IQR 7.2-26.9). Thirty-nine pts (25%) were observed without treatment or received chemotherapy or radiotherapy, 9 pts (8%) received at least one salvage treatment including immunomodulating agents, 43 pts (36%) were treated with at least one salvage therapy including proteasome inhibitors, 37 pts (31%) received at least one salvage treatment including monoclonal antibodies (33 daratumumab, 1 elotuzumab,1isatuximab, 2 belantamab). Median OS of relapsed pts was 38.5 months from allo-SCT and 20.2 months from relapse. OS (Fig 1) was significant longer in pts who had received at least 3 salvage treatment lines (p<0.001) including monoclonal antibodies (p=0.007) and/or donor lymphocyte infusions (DLI) (p=0.019); in multivariate model only number of therapeutic lines after relapse reached a statistically significance (p=0.027;HR 0.32; CI 0.12,0.88).

Table 1 : Baseline characteristics

Patient variable

Total

N=242

Median age at transplantation, y (range)

54 (29-77)

Transplant period, n (%)

• 2005-2009

35 (14)

• 2010-2014

128 (53)

• 2015-2020

77 (32)

• unknown

2 (1)

Conditioning, n (%)

• myeloablative

141 (58)

• non-myeloablative

97 (40)

• unknown

4 (2)

Stem cell source, n (%)

• bone marrow

28 (12)

• peripheral blood

210 (87)

• cord blood

4 (1)

Donor HLA matching, n (%)

• identical sibling

98 (40)

• MUD-match

124 (51)

• Mismatch

11 (5)

• MUD-mismatch

9 (4)

ISS stage, n (%)

• 1

43 (18)

• 2

39 (16)

• 3

52 (21)

• unknown

108 (45)

Pre-transplantation therapeutical lines, n (%)

• 1-2

93 (38)

• >2

92 (38)

• unknown

57 (24)

Disease status at transplantation, n (%)

• CR-VGPR

96 (40)

• PR

83 (34)

• SD-PD

54 (22)

• unknown

9 (4)

FISH characterization, n (%)

• High risk

53 (45)

• Unknown

125 (52)

DLI therapy, n (%)

• Yes

28 (12)

• No

206 (85)

• Unknown

8 (3)

Fig 1: Overall survival from relapse therapy based

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (65)

Conclusions: Our results suggest that several treatment lines, including monoclonal antibodies and/or DLI administered for MM relapse after allo-SCT, can act in synergy for a long term disease control.

Clinical Trial Registry:

GITMO-NEW_ALLO_MM.

Disclosure: No conflict of interest to declare.

27: Multiple Myeloma

P549 HAPLOIDENTICAL ALLOGENEIC CELL TRANSPLANTATION IN RELAPSED/REFRACTORY MULTIPLE MYELOMA

Julia Frimmel 1, Anke Morgner2, Claudia Brogsitter3, Karolin Trautmann-Grill3, Desiree Kunadt3, Raphael Teipel3, Christoph Röllig3, Mathias Hänel2, Johannes Schetelig3,4, Friedrich Stölzel1, Martin Bornhäuser3,5,6

1Universitätsklinikum Schleswig-Holstein, Kiel, Germany, 2Klinikum Chemnitz, Chemnitz, Germany, 3Universitätsklinikum Dresden, Dresden, Germany, 4DKMS Clinical Trials Unit, Dresden, Germany, 5National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany, 6German Cancer Consortium (DKTK) partner site Dresden, Dresden, Germany

Background: Although emerging therapies and substances for the treatment of patients with multiple myeloma (MM) have improved therapeutic options, long-term disease control in patients with advanced stage and/or relapsed/refractory MM remains challenging. While the effect of natural killer cell alloreactivity in haploidentical allogeneic hematopoietic cell transplantation (alloHCT) applying post-transplantation cyclophosphamide (PTCy) as graft-versus-host disease (GvHD) prophylaxis is considered a standard treatment option in several hematologic neoplasms, it is a controversial treatment option for patients with MM. In this retrospective analysis, we evaluated all consecutive patients with MM undergoing haploidentical alloHCT using PTCy in our institution.

Methods: With a median follow-up of 34.5 months (range 2 – 69 months) a total of seven patients with relapsed/refractory (r/r) MM underwent haploidentical alloHCT. All patients were heavily pre-treated (mean 6.5 previous lines) with proteasome inhibitors, anti-CD38 antibody, immunomodulatory drugs and had at least one prior autologous HCT. All patients received a chemotherapy-based reduced intensity conditioning regimen, in three cases combined with radioimmunotherapy using Re-188-CD66-AK, and PTCy-based GvHD prophylaxis in combination tacrolimus and mycophenloate mofetil.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (66)

Results: All patients had stable engraftment with complete donor chimerism with n = 4 of the seven patients achieving a complete remission (CR) and n = 3 achieving a very good partial remission (VGPR). AlloHCT resulted in a median progression free survival (PFS) of 24.5 months (range 2 – 55 months) and a median overall survival (OS) of 48 (range 2 – 89 months) for all patients with one transplantation-associated death due to sepsis two months after transplantation. Acute GvHD grade II–IV was observed in n = 2 patients while n = 4 patients developed mild to moderate chronic GvHD in the further course with no GvHD-associated deaths at last follow-up.

Conclusions: Haploidentical alloHCT with individual pre-treatment approaches and conditioning regimens induces disease control in heavily pretreated thus selected patients with r/r MM.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P550 ANTI-HLA ANTIBODIES AND INCIDENCE OF PLATELET TRANSFUSION IN PATIENTS WITH MULTIPLE MYELOMA AFTER AUTO-HSCT

Elena Kuzmich1, Irina Pavlova1, Ivan Kostroma1, Sergey Gritsaev 1

1Russian Research Institute of Hematology and Transfusiology, St. Petersburg, Russian Federation

Background: A number of patients with multiple myeloma (MM) who have undergone auto-HSCT require platelet transfusion. The effectiveness of transfusion depends on various factors, including the presence of anti-HLA antibodies in the patient. The aim of our study was to investigate the effect of pre-existing anti-HLA antibodies on the incidence of platelet transfusion in patients with MM after auto-HSCT.

Methods: The study included 42 patients. The median age of patients was 55 (41-66) years. Gender distribution: 19 males and 23 females. The median time from diagnosis to determination of anti-HLA antibodies was 10 months, during this period all patients received bortezomib-based therapy. Detection of pre-existing anti-HLA antibodies was carried out before the conditioning regime. The Luminex technology and the screening kit for anti-HLA antibodies for class I and for class II (Immucor Transplant Diagnostics, Inc. USA) were used. The result was evaluated as positive at MFI of more than 500 units. Before auto-HSCT, the following conditioning regimens were used: melphalan at a dose of 200 and 140 mg/m2 (n=38; 90.5%), melphalan in combination with carfilzomib (n=3; 7.1%), and melphalan in combination with thiotepa (n=1; 2.4%).

Results: The incidence of pre-existing anti-HLA antibodies in patients with multiple myeloma who received bortezomib-based therapy was 7.1%. The frequency of antibodies to HLA antigens of class I and II in patients of the examined cohort had no significant differences (4.8% vs. 2.4%, p= 0.6). All allosensitized patients were female, and the reason for allosensitization was pregnancy (number of 2 to 3). The established degree of sensitization was medium (MFI in the range of 500 – 3000). The median day (spread) of platelet recovery in allosensitized patients was D + 14 (D + 12 to D + 15). The median day (spread) of platelet recovery in the patients without allosensitization was D + 13 (D + 7 to D + 17). The median (spread) of the number of transfusions in allosensitized patients was 3 (2–3) during the first 10 days after auto-HSCT. The median (spread) of the number of transfusions in patients without allosensitization was 2 (0–5) during the first 10 days after auto-HSCT.

A repeat examination performed 14-21 days after auto-HSCT did not reveal a change in the level of anti-HLA antibodies in sensitized patients.

Conclusions: In our study, we did not find a significant effect of pre-existing anti-HLA antibodies on the number of platelet transfusions in patients with MM after auto-HSCT. This fact can be explained by the rather low degree of allosensitization in our patients received bortezomib-based therapy.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P551 AUTOLOGOUS PERIPHERAL BLOOD STEM CELL HARVESTING AND UTILIZATION IN MULTIPLE MYELOMA PATIENTS IN LUHS KAUNAS CLINICS, LITHUANIA 2015-2022

Titas Tiskevicius 1,2, Domas Vaitiekus1,2, Rolandas Gerbutavicius1,2, Milda Rudzianskiene1,2, Ruta Dambrauskiene1,2, Migle Kulboke1,2, Ignas Gaidamavicius1,2, Diana Remeikiene1,2, Birute Sabaniene1, Ieva Stakaitiene1, Egidija Kukarskyte2, Jonas Surkus1,2, Ruta Leksiene1,2, Elona Juozaityte1,2, Dietger Niederwieser3

1Hospital of Lithuanian University of Health Sciences Kaunas Clinics, Kaunas, Lithuania, 2Lithuanian University of Health Sciences, Kaunas, Lithuania, 3University of Leipzig, Leipzig, Germany

Background: The optimal number of CD34+ cells needed for repetitive autologous hematopoietic stem cell transplantation (auto-HSCT) in multiple myeloma (MM) patients remains unclear. As effective novel therapies for MM emerge, the use of salvage autologous hematopoietic stem cell transplantation (auto-HSCT) for relapse/refractory MM is questionable.

Methods: We conducted a retrospective single center study using the clinical and laboratory databases. The study included 158 MM patients, 384 apheresis, 536 products and 211 auto-HSCT in the hospital of Lithuanian University of Health Sciences Kaunas Clinics from 2015 to 2022. Peripheral blood hematopoietic stem cells (PBHSC) were harvested after mobilization Cyclo+G-CSF (10 µg/kg) or an additional mobilizing agent such as plerixafor (0.24 mg/kg) if needed. PBHSC harvesting was done via apheresis. In our center the goal is to collect enough CD34+ cells for 3 auto-HSCT, which would equate to at least 10 x106 CD34 + /kg. After each apheresis session, CD34+ counts were calculated. If the goal was achieved or if further mobilization was not possible, the harvesting was stopped. We calculated collected cells and divided patients in to 2 groups: A – if the goal is reached, B – goal is not reached. Descriptive statistics were performed on the variables of interest. We report frequencies and percentages as well as mean and ranges where applicable.

Results: In total 158 MM patients were mobilized via 384 apheresis, that harvested 536 products, and transplanted at least 1 time. For 100 (63.3%) of the patients the CD34+ goal was achieved (A group), for 58 (36.7%) was not (B group). Mean collected CD34+ cell count for both groups were 12.45 x106/kg (range 2.0 x106 – 31.4 x106 /kg). To reach the CD34+ cell goal, 3 (3%) patients needed only 1 apheresis, most of them 67 (67%) needed 2 procedures, 28 (28%) – 3 apheresis procedures, and 2 (2%) patients needed 4 procedures. In comparison, in the B group more than half of the patients 35 (60.3%) needed 3 or more apheresis procedures. There is a correlation between CD34+ yield and number of apheresis procedures: patients yielding higher CD34+ counts required fewer apheresis procedures compared to those yielding lower CD34+ counts (p<0.01). Out of the patients that reached the targeted CD34+ goal (A group) transplanted in tandem 37 (37%) patients and 5 (5%) received as salvage auto-HSCT. Out of the B group 6 (10.2%) patients received tandem auto-HSCT, but no one received auto-HSCT as salvage.

Conclusions: Our analysis evaluated the use of stored cryopreserved PBHSC for a salvage auto-HSCT in patients with MM. We showed that we could collect enough PBHSC for third auto-HSCT in most patients and usually they required 2 or 3 apheresis procedures. A third auto-HSCT was performed in just 5% of the included cohort. The increased survival rates offered by new cellular and immunotherapies, alongside the decreasing frequency of conducting salvage auto-HSCT warrant reconsideration of PBHSC collection goals.

Disclosure: Nothing to declare.

27: Multiple Myeloma

P552 AUTOLOGOUS PERIPHERAL BLOOD STEM CELL TRANSPLANTATION IN MULTIPLE MYELOMA AT DHARMAIS HOSPITAL – INDONESIAN NATIONAL CANCER CENTER

Resti Mulya Sari 1, Dwi Wahyunianto Hadisantoso1, Edel Herbitya1, Lyana Setiawan1, Hubertus Hosti Hayuanta1, Muhammad Raya Kurniawan1, Yanto Ciputra1, Annisa Annisa1, I Gusti Ngurah Agastya1, Della Manik Worowerdi Cintakaweni1

1Dharmais National Cancer Center Indonesia, West Jakarta, Indonesia

Background: Since the 1990s, patients who qualify for transplantation have received autologous stem-cell transplantation (ASCT) as the standard treatment for multiple myeloma (MM). ASCT is a challenge in itself in Indonesia. This study reports three cases of MM who underwent Autologous Peripheral Blood Stem Cell Transplantation (PBSCT) in a developing country with limited facilities and costs.

Methods: The chemomobilization regimen consisted of Vinorelbin 25 mg/m2 on day one and Cyclophosphamide 1,500 mg/m2 on day 2, followed by granulocyte colony-stimulating factor (G-CSF) 10 mcg/kg body weight/day. The conditioning regimen used melphalan 200 mg/m2.

Results: We describe a successful autologous PBSCT for three MM patients. One patient was treated with an induction regimen of Thalidomide/Dexamethasone, and another two patients were treated with induction regimens of VCD (Bortezomib, Cyclophosphamide, and Dexamethasone). The source hematopoietic stem cells were harvested from peripheral blood stem cells after chemomobilization. An autologous bone marrow transplant was carried out once remission was reached. The complications after ASCT were febrile neutropenia, anemia, thrombocytopenia, nausea, vomiting, and skin darkness. The engraftment on day +11, day +10, and day +12, respectively. The patient’s condition improved under treatment in the isolation room, and the patients were discharged. Maintenance therapy with lenalidomide used to our patients after ASCT.

Conclusions: Implementing high-quality and safe ASCT can be done in Indonesia with solid support from hospitals and the government. A highly skilled multidisciplinary team is vital to the transplant’s success.

Disclosure: Nothing to declare.

28: Myelodysplastic Syndromes

P553 OUTCOMES OF HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM HAPLOIDENTICAL DONORS IN PATIENTS WITH MYELODYSPLASTIC SYNDROMES – A SINGLE-CENTER RETROSPECTIVE ANALYSIS

Marketa Stastna Markova 1, Ludmila Novakova1, Mariana Koubova1, Barbora Cemusova1, Veronika Valkova1, Antonin Vitek1, Petr Cetkovsky1, Jan Vydra1

1Institute of Haematology and Blood Transfusion, Praha 2, Czech Republic

Background: Allogeneic hematopoietic cell transplantation remains the only curative treatment for Myelodysplastic Syndromes (MDS). The introduction of post-transplant cyclophosphamide-based GvHD prophylaxis (ptCy) has expanded access to this procedure for a broader patient population lacking an HLA-matched donor. However, post-transplantation relapse and transplant-related mortality continue to be significant challenges. Consequently, the question of donor type superiority has arisen, especially in patients diagnosed with MDS.

Methods: A single-center retrospective analysis was conducted on MDS patients who underwent HSCT between 2014 and 2023. Patients without an available HLA-matched donor were transplanted from haploidentical related donors. The study evaluated 117 patients, including 12 with Matched Sibling Donors (MSD), 77 with Matched or One Mismatch Unrelated Donors (MUD), and 28 with Haploidentical Donors (HID). All groups received identical conditioning regimens. Myeloablative regimens included Busulfan and Fludarabine, while Reduced Intensity Conditioning (RIC) involved Fludarabine, Cyclophosphamide, Melphalan, or Fludarabine and Treosulfan. Approximately half of the transplants in each group were RIC. GVHD prophylaxis was based on ptCy for all HID and 31 of the MUD transplants, and ATG was used in the remaining MUD transplants. MSD patients received only a calcineurin inhibitor and mycophenolate mofetil.

Results: One-year overall survival rates for MSD, MUD, and HID groups were 82%, 83%, and 93%, respectively, and three-year survival rates were 71%, 72%, and 73%, with no significant difference between the groups. In high-grade MDS patients (EB1, EB2), one-year survival rates were 66% (MSD), 80% (MUD), and 81% (HID), and three-year rates were 44%, 71%, and 69%, respectively, showing a trend towards statistical significance (p=0.08). In high-risk and very high-risk groups according to the IPSS, a significant advantage was observed in the HID group with one-year survival rates of 94% compared to 69% in MSD + MUD, and three-year rates of 80% versus 61%. No difference in acute and chronic GVHD incidence was observed across the groups.

Conclusions: Our findings suggest that haploidentical donor transplantation using ptCy leads to similar results as MSD and MUD transplantation in patients with myelodysplastic syndromes. Haploidentical donor transplantation could have an OS superiority in High and Very high risk MDS patients according to IPSS.

Disclosure: I have nothing to disclose.

28: Myelodysplastic Syndromes

P554 CHALLENGES AND REALITIES OF HEMATOPOIETIC CELL TRANSPLANTATION OF MYELODYSPLASTIC SYNDROMES PATIENTS IN LATIN AMERICA: A SURVEY

Fernando Barroso Duarte 1, Rodolfo Daniel de Almeida Soares2, Abrahão Elias Hallack Neto3, Anderson João Simione4, Talyta Ellen de Jesus dos Santos Sousa5, Erika Oliveira de Miranda Coelho6, Vaneuza Araujo Moreira Funke7, Nelson Hamerschlak8, Rodolfo Froes Calixto9, Maria Claudia Rodrigues Moreira10, Alicia Enrico11, Marco Aurelio Salvino12, Eduardo José de Alencar Paton13, Mariana Stevenazzi14, Neysimelia Costa Villela15, Carmem Bonfim7, Gisele Loth16, Breno Moreno Gusmão17, Maria Cristina Martins de Almeida Macedo18, Isabella Araújo Duarte19, Vergílio Antônio Rensi Colturato4

1Hospital Universitário Walter Cantídio, Fortaleza, Brazil, 2Natal Hospital Center, Natal, Brazil, 3Federal University of Juiz de Fora, Juiz de Fora, Brazil, 4Amaral Carvalho Hospital, Jaú, Brazil, 5Federal University of Ceara, Fortaleza, Brazil, 6Saint Joana Hospital, Recife, Brazil, 7Federal University of Parana, Curitiba, Brazil, 8Hospital Israelita Albert Einstein, São Paulo, Brazil, 9Real Hospital Português de Beneficência in Pernambuco, Recife, Brazil, 10CHN - Complexo Hospitalar de Niterói, Niterói, Brazil, 11Italian Hospital La Plata, La Plata, Argentina, 12Federal University of Bahia, Salvador, Brazil, 13ONCOBIO Health Services, Nova Lima, Brazil, 14Integral Medical Service, Montevideo, Uruguay, 15Barretos Children’s Cancer Hospital, Barretos, Brazil, 16Hospital de Clínicas da Universidade Federal do Paraná, Curitiba, Brazil, 17Hospital São José, São Paulo, Brazil, 18Intituto Brasileiro de Controle do Câncer, São Paulo, Brazil, 19Centro Universitário Christus - UNICHRISTUS, Fortaleza, Brazil

Background: Hematopoietic Cell Transplantation (HCT) is the only curative therapy available for Myelodysplastic Syndromes (MDS) patients. In Latin America (LA), although there is a specific registry of transplants in MDS, there is no information on the percentage of patients who are referred to HCT centres, those who indeed underwent the procedure or not. The general scenario of LA comprises a higher rate of autologous HCT (60%). In 2018, 127 teams from 14 countries reported activity data into the 2018 WBMT GTA. From all 5642 transplants performed in this period, only 170 (9%) involved MDS/MNP, showing the difficulty to transplant this group of patients. The study aimed to understand the journey of MDS patients referred to HCT centers of LA and key points in this process.

Methods: A questionnaire was directed to members of the Latin American Registry for Myelodysplastic Syndrome through Google Forms, from 2 to 13 May 2022.

Results: A total of 18 physicians that represent 28 centers of adult and pediatrics HCT answered the questionnaire regarding pre and post HCT features. In most centers, less than 5 patients were attended in the last 2 years and less than 30% used geriatric scores in the HCT decision. Transfusion dependence was reported by 89.2% of the centers, and alloimmunization in 22.3%. Data from 162 patients were entered. There was a predominance of high-risk R-IPSS stratification. About 25% of patients were referred for transplantation without an R-IPSS score. Contraindication rate was 14.2% (n=23), with comorbidities and disease refractoriness being the main causes.

Conclusions: The results obtained from this survey bring relevant findings, which raise many questions about the referral of MDS patients to HCT, including the small number of adult and pediatric patients admitted for pre-HCT evaluation. Unfortunately, there is no comprehensive epidemiological data regarding the incidence of MDS in Latin America. It is known that MDS is a disease of older patients, increasing its incidence after the age of 60, but we have reports that, due to exposure to genotoxic agents, cases are not uncommon in younger people and even children in our region. In Brazil, 4383 deaths from MDS were reported in the period from 2014 to 2018, based on the DATA SUS. Despite this data, we can only question whether we have really assessed the adequate number of patients before HSCT, as 38.9% of the centers have assessed less than 5 patients in two years. Even allo HCT is increasing in LA, it is still a small number in comparison to the US and Europe. For example, 30 in LA, 227 in USA/Canada and 181 in Europe in 2016. Since not all centers responded to the survey, the study presents some limitations in capturing the real-world picture. Nevertheless, data obtained is a starting point for understanding the reality of the HSCT in LA and to make decisions aiming to overcome the barriers and promote access to the procedure with quality and safety.

Disclosure: Authors declare they have no conflict of interest.

28: Myelodysplastic Syndromes

P555 OUTCOMES OF RELAPSE POST HEMATOPOIETIC CELL TRANSPLANTATION IN MYELODYSPLASTIC SYNDROMES FROM THE LATIN AMERICAN REGISTER

Fernando Duarte 1, Talyta Ellen de Jesus dos Santos Sousa1, Vaneuza Araújo Moreira Funke2, Nelson Hamerschlak3, Neysimélia Costa Villela4, Maria Cristina Martins de Almeida Macedo5, Afonso Celso Vigorito6, Rodolfo Daniel de Almeida Soares7, Alessandra Paz8, Lilian Diaz9, Mariana Stevenazzi9, Abrahão Elias Hallack Neto10, Gustavo Bettarello11, Breno Moreno Gusmão12, Marco Aurélio Salvino13, Rodolfo Froes Calixto14, Maria Cláudia Rodrigues Moreira15, Gustavo Machado Teixeira16, Cinthya Corrêa Silva3, Eduardo José de Alencar Paton17, Vanderson Rocha18, Alicia Enrico19, Carmem Bonfim20, Ricardo Chiattone21, Anderson João Simioni22, Celso Arrais23, Erika Oliveira de Miranda Coelho24, Vergílio Antônio Rensi Colturato22

1Federal University of Ceara, Fortaleza, Brazil, 2Federal University of Parana, Curitiba, Brazil, 3Hospital Israelita Albert Einstein, São Paulo, Brazil, 4Barretos Children’s Cancer Hospital, Barretos, Brazil, 5Intituto Brasileiro de Controle do Câncer, São Paulo, Brazil, 6State University of Campinas - UNICAMP, Campinas, Brazil, 7Natal Hospital Center, Natal, Brazil, 8Clinical Hospital of Porto Alegre, Porto Alegre, Brazil, 9Center TPH-SMI Integral Medical Service, Montevideo, Uruguay, 10Federal University of Juiz de Fora, Juiz de Fora, Brazil, 11Unidade de Transplante de Medula Óssea Pietro Albuquerque, Brasília, Brazil, 12Hospital São José, São Paulo, Brazil, 13University Hospital Prof. Edgard Santos, Salvador, Brazil, 14Real Hospital Português of Beneficência in Pernambuco, Recife, Brazil, 15National Institute of Cancer-INCA, Rio de Janeiro, Brazil, 16Clinical Hospital of Minas Gerais Federal University, Belo Horizonte, Brazil, 17ONCOBIO Health Services, Nova Lima, Brazil, 18Clinical Hospital of Medicine of São Paulo, São Paulo, Brazil, 19Italian Hospital La Plata, La Plata, Argentina, 20Hospital Pequeno Principe, Curitiba, Brazil, 21Hospital Samaritano of São Paulo, São Paulo, Brazil, 22Amaral Carvalho Hospital, Jaú, Brazil, 23Hospital Sírio Libanês, São Paulo, Brazil, 24Hospital Santa Joana, Recife, Brazil

Background: Relapse after Hematopoietic cell transplantation (HCT) in Myelodysplastic Syndromes (MDS) is a challenge due to the lack of standard protocols. The objective was to analyse outcomes and risk factors of overall and relapse free survival of patients underwent to Hematopoietic Cell Transplantation (HCT).

Methods: A retrospective registry of 400 MDS patients from the Latin American Registry was performed from 1988 to May 2023 from the transplant registry of 32 centers in Latin America. Statistics were performed using SPSSv.23.1, considering a significant p<0.05.

Results: The mean age was 45,54 years. Most patients were ≤ 50 years (50,50 %), about 27,25 % were between 50 and 61 and 22,25 % were > 60 years. There was a predominance of males (58 %). Patients were classified as intermediate (n=88; 22%), high risk (n=72; 18%), low risk (n=38; 9,5%), very high risk (n=24;6%) and very low risk (n=2; 0,5%) in the Prognosis Scoring System (IPSS-R). Myeloablative conditioning (MAC) was performed in 287 patients (71,75%). Donor types were related (65%), non-related (22,75%) and haploidentical (12,25%). The main cell source was bone marrow (51,50%). Complications post-HSCT were observed in 314 patients (78,5%). The most frequent were infections (n= 253; 80,57%), acute graft versus host disease (GVHD) (n=145; 46,18%) and chronic GVHD (n=113; 35,99%). The frequency of death was 39,5% (n=158). The 5-years overall survival rate was 54.5%. It was lower in High/very high-risk patients (p=0,013). Regarding relapse, the median RFS was 1.9. It was observed a trend to lower RFS for patients older than 60y along 20years (p=0,05). For patients with at least 5 years of HCT, those older than 60y had more chance of relapse than those with 51-6 years (p=0,04). In the univariate model, age was associated with risk of relapse, with a 2.52 times higher risk of recurrence for patients aged 60 years or older, compared to those aged up to 50 years. Patients classified as “very high” risk R-IPSS have a 27.66 times higher risk of recurrence than those with low or intermediate risk. Myeloablative regimen had 63% less risk of relapse than those who underwent reduced intensity/nonmyeloablative. Higher RFS was associated with chronic GVHD for 5-y-RFS (p=0,027).

Conclusions: The study showed that relapse occurred more frequently in older patients in shorter and longer periods after HCT, suggesting a clonal evolution probably due to the senescence process. Myeloablative regimen was associated with lower relapse, which was confirmed in the multivariate model. Another favourable factor was the chronic GVHD, which is related in literature to increase GVL effect, reducing relapse rates. The higher death rate in patients with relapse shows the difficulty to treat these patients due to lack of therapeutic options, even with DLI and a second HCT approach. The present study includes patients from a long period, with different realities. Some of them underwent HCT without an adequate stratification or even were not stratified. The outcomes provide the scenario to prospective studies in order to better comprehend the prognostic factors and overcome challenges in the prevention and management of relapse in MDS in LA.

Disclosure: The authors declare they have no conflicts of interest.

28: Myelodysplastic Syndromes

P556 OUTCOME OF HEMATOPOIETIC STEM CELL TRANSPLANTATION OF IN CHILDREN WITH A NPM1 MUTATION AND MYELODYSPLASTIC SYNDROME WITH EXCESS BLASTS

Ayami Yoshimi 1, Miriam Erlacher1, Peter Noellke1, Senthilkumar Ramamoorthy1, Gudrun Göhring2, Shlomit Barzilai-Birenboim3, Ivana Bodova4, Jochen Buechner5, Albert Catala6, Valérie De Haas7, Barbara De Moerloose8, Michael Dworzak9, Henrik Hasle10, Kirsi Jahnukainen11, Krisztian Kallay12, Marko Kavcic13, Paula Kjollerstrom14, Franco Locatelli15, Riccardo Masetti16, Sophia Polychronopoulou17, Markus Schmugge18, Owen Smith19, Jan Stary20, Dominik Turkiewicz21, Marek Ussowicz22, Marcin Wlodarski23, Christian Thiede24, Brigitte Strahm1, Charlotte Niemeyer1

1University of Freiburg, Freiburg, Germany, 2Hannover Medical School, Hannover, Germany, 3Tel Aviv University, Tel Aviv, Israel, 4National Institute of Children’s Diseases, Bratislava, Slovakia, 5Oslo University Hospital, Oslo, Norway, 6Hospital Sant Joan de Deu, Barcelona, Spain, 7Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands, 8Ghent University Hospital, Gent, Belgium, 9St. Anna Children’s Cancer Research Institute, Vienna, Austria, 10Aarhus University Hospital, Aarhus, Denmark, 11Helsinki University Hospital, Hus, Finland, 12National Institute of Hematology and Infectious Diseases, Budapest, Hungary, 13University Medical Centre, Ljubljana, Slovenia, 14Centro Hospitalar Universitário de Lisboa Central, Lisbon, Portugal, 15Sapienza University of Rome, Rome, Italy, 16IRCCS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, 17Aghia Sophia Children’s Hospital, Athens, Greece, 18University Children’s Hospital, Zurich, Switzerland, 19Our Lady’s Children’s Hospital, Dublin,, Ireland, 20University Hospital Motol, Prague, Czech Republic, 21Skåne University Hospital, Lund, Sweden, 22Wroclaw Medical University, Wroclaw, Poland, 23St. Jude Children’s Research Hospital, Memphis, United States, 24University Hospital Carl Gustav Carus, Dresden, Germany

Background: Nucleophosmin 1 (NPM1) mutations are noted in 30% of adults with AML, and less frequently in 5-8% of pediatric patients with AML. They are associated with normal karyotype, presence of FLT3-ITD and favorable outcome. There is limited data on the prevalence and clinical significance of NPM1 mutations in young with myelodysplastic syndrome with excess blasts (MDS-EB). Here we report the outcome of allogeneic hematopoietic stem cell transplantation (HSCT) in pediatric patients with MDS-EB and NPM1 mutation.

Methods: Among 235 patients (<18 years) diagnosed between 07/1998 and 06/2021 with MDS-EB, a NPM1 mutation was retrospectively identified in 14 patients (6%). Most patients received HSCT (226, 96%). After excluding patients with a predisposition syndrome (54) and insufficient genetic or HSCT data (9), outcome of HSCT of patients with a NPM1 mutation was compared to those of patients without a mutation (13 NPM mt vs.150 NPM1WT).

Results: The median age of the 13 patients with NPM1 mutation was 12.6 years (5-17 years). The bone marrow (BM) blast count was 5-9% in six patients and 10-19% in seven patients. All patients had 1-3 additional somatic mutations including PTPN11 (5), NRAS (6) and WT1 (3), while UBTF-TD or FLT3-ITD were absent. Four patients had intensive chemotherapy prior to HSCT followed by complete remission (1), blast persistence (2) or aplasia without blasts (1); one patient received less intensive therapy with thioguanine. The donor was a matched sibling (2), 9-10/10 matched unrelated donor (9), or another unrelated donor (2). The stem cell source was BM (8), peripheral blood (4) or cord blood (1). Nine patients received conditioning with busulfan (Bu), cyclophosphamide and melphalan, two patients were given other Bu-based regimens, and two patients received treosulfan, fludarabine and thiotepa. All patients engrafted. Four patients developed II-IV acute graft-versus-host disease (GVHD), and three patients limited (1) or extensive chronic GVHD (2). One patient relapsed, and three succumbed to non-relapse mortality (NRM). The 5-year-event-free survival following HSCT was 67% (95%CI 40-94%), and not significantly different from that of patients without a NPM1 mutation (57%, 95%CI 49-65%, p=n.s.). In addition, there was no difference between the NPM1mt and NPM1wt groups with respect to the cumulative incidence of relapse (6% in NPM1mt vs. 28% NPM1WT, p=0.13) or NRM (24% in NPM1mt vs. 14% in NPM1WT, p=n.s.).

Conclusions: The incidence of NPM1 mutations in pediatric patients with MDS-EB was 6%. There was no significant difference in outcome of HSCT between the NPM mt and NPM1WT groups. The small number of NPM1 mutated pediatric patients in this cohort does not allow further conclusions on the role and indication of AML therapy or HSCT in young NPM1mt patients with a low blast count. While current classifications categorize patients with NPM1 mutated neoplasia as AML irrespective of blast count (WHO) or in the presence of ≥10% blast percentage (ICC), the standard of care for pediatric patients with NPM1mt disease and low blast count still needs to be defined, particularly in the presence of emerging targeted therapies.

Disclosure: Nothing to declare.

28: Myelodysplastic Syndromes

P557 SYSTEMATIC LITERATURE REVIEW ON THE SAFETY, EFFICACY, AND EFFECTIVENESS OF EATG VERSUS COMPARATORS FOR MDS

Erin Sheffels1, Kevin Kallmes1, Keith Kallmes1, Barbara Możejko-Pastewka2, Raj Gokani 2, Judith Hey-Hadavi1, Andres Quintero2

1Nested Knowledge, St Paul, United States, 2Pfizer, Inc., New York City, United States

Background: Evidence on aplastic anemia and anti-thymocyte globulin (ATG) demonstrates that equine ATG (eATG) and rabbit ATG (rATG) have different effects on the immune system. We performed a systematic literature review to compare clinical outcomes for eATG and rATG in patients with low-risk myelodysplastic syndrome (MDS).

Methods: To identify comparative clinical studies reporting eATG or ATG from unspecified sources, we performed a PRISMA-compliant systematic review of clinical literature using the PubMed database. No date restrictions were used. Patient data collected included age, sex, and MDS risk. Outcomes included incidence of blood-transfusion independence, remission, complete or partial response to treatment, anemia, thrombocytopenia, malignant transformation, other disease progression, mortality, infection, event-free survival, and cumulative adverse events. Due to study heterogeneity, inferential statistics were not performed.

Results: From 423 identified records, this analysis included nine studies (670 patients). Eight studies reported eATG and one did not specify the source of ATG. Comparators included rATG, cyclosporine A and best supportive care. For complete or partial response, overall survival, or adverse events, most studies found no significant difference between eATG and the comparator. For complete or partial response, three studies demonstrated eATG’s superiority over the comparator, and for transfusion independence, one study demonstrated eATG’s superiority to rATG. Across all studies and outcomes, there was no evidence of eATG being inferior to rATG.

Conclusions: The body of evidence on MDS and ATG indicates that eATG is non-inferior to rATG and other comparators, and may further be superior, particularly for transfusion independence. Limitations of our review include low study quality and high heterogeneity in comparators and patient risk categories. More randomized controlled studies are needed to confirm the relative efficacy, effectiveness, and safety of eATG and other treatments in low-risk MDS.

Disclosure: Conflicts of interest: Barbara Możejko-Pastewka, Raj Gokani, Judith Hey-Hadavi, and Andres Quintero are employees of Pfizer Inc. Kevin Kallmes is employed by and holds equity in Nested Knowledge, Inc; he holds equity in and serves on the board of Superior Medical Experts, Inc., and Piraeus Medical, Inc.

22: Myeloproliferative Neoplasm

P558 TCRALPHABETA/CD19 DEPLETED ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN COMBINATION WITH POST-TRANSPLANT RUXOLITINIB FOR MYELOFIBROSIS

Flores Weverling1, Yousra van der Leest1, Frances Verheij1, Iris Brinkman1, Anna van Rhenen1, Lotte van der Wagen1, Laura Daenen1, Kasper Westinga2, Henk-Jan Prins2, Lisette van de Corput3, Reinier Raymakers1, Anke Janssen1, Jurgen Kuball1, Moniek de Witte 1

1University Medical Center, Utrecht, Netherlands, 2Cell Therapy Facility, University Medical Center, Utrecht, Netherlands, 3Central Diagnostic Laboratory, University Medical Center, Utrecht, Netherlands

Background: Allogeneic hematopoietic stemcell transplantation (allo-HCT) is frequently complicated by Graft-versus-Host Disease (GVHD) in patients with myelofibrosis (MF). Consequently, GVHD free, relapse free survival (GRFS) remains suboptimal. TCRalphabeta/CD19-depletion allo-HCT results in low incidences of aGVHD and cGVHD, but experience in MF is limited.

Methods: This retrospective study includes 30 patients with primary or secondary MF who received an allo-HCT between June 2017 and April 2022 in the UMC Utrecht. Data-cut off was set at 1 October 2023. Written informed consent was obtained in accordance with JACIE guidelines. Stem cells were derived from peripheral blood of matched related donors or matched unrelated donors (10/10 or 9/10). Patients received a myeloablative conditioning consisting of ATG (Thymoglobulin®), fludarabine and busulfan. Ex vivo graft engineering was performed with anti-alphabeta T cell receptor (TCR) and CD19 antibodies, in combination with magnetic microbeads using the automated CliniMACS device (Miltenyi Biotec ®). Initially, mycophenolate mofetil (MMF) was administered for 28D as single-agent GVHD prophylaxis (RUXminus). Due to high transplant-related mortality (TRM), post-transplant ruxolitinib (RUX) (5mg 2dd for 14D) was added to MMF (RUXplus). From 2021 minimal residual disease (MRD) was assessed by performing digital droplet PCR for the relevant driver mutation. One or more pre-emptive DLIs were administered once the MRD exceeded > 0.1% at two consecutive timepoints.

Results: Median age at transplant was 66 years (range 33-72). 20 patients (66%) received post-transplant RUX (RUXplus). The cumulative incidence (CI) of grade 2-4 and 3-4 aGVHD at D100 was 17% and 10% in RUXplus and 50% and 30% in RUXminus. The 2Y CI of extensive cGVHD extensive was 0% in RUXplus and 10% in RUXminus. One patient in the RUXminus group had a secondary rejection and 1 patient in the RUXplus group had a poor graft function. The 2Y CI of relapse was 15% in RUXplus and 23% in RUXminus. The 2Y NRM was 19% in RUXplus and 32% in RUXminus. All relapses occurred before the implementation of MRD follow-up. Three (of 5) patients with a relapse are still in CR after a therapeutic DLI (n=2) or 2nd allo-SCT (n=1). Within the RUXplus group, 5 patients became MRD positive. Three from 5 patients received 2 pre-emptive DLIs (1x10E6 and 5x10E6 CD3 cells/kg) after which they became MRD negative; the other 2 patients have not been evaluated yet. These outcomes translated in a 2Y overall survival of 85% in the RUXplus versus 50% in RUXminus group; 2Y event free survival of 70% versus 40% and 2Y GRFS of 65% versus 20%.

Conclusions: We have developed an allo-HCT platform where we can administer a myeloablative conditioning to facilitate engraftment and combine early ATG, alphabeta TCR/CD19 depletion with a short course of post transplant RUX to limit severe GVHD in patients with MF. The 2Y OS, EFS and GRFS are favorable and with systematic analysis of MRD and pre-emptive DLI outcomes may improve even further.

Disclosure: J.K. reports grants Novartis, and Miltenyi Biotech and is inventor on patents dealing with γδ T-cell–related aspects.

22: Myeloproliferative Neoplasm

P559 MINIMAL RESIDUAL DISEASE MONITORING OF DRIVER MUTATION BY DIGITAL DROPLET PCR IS PREDICTIVE OF RELAPSE AFTER ALLOGENEIC STEM CELL TRANSPLATION IN MYELOFIBROSIS

Maria Chiara Finazzi 1,2, Roberta Stavola1, Francesca Valsecchi2, Alessia Civini2, Chiara Pavoni2, Matteo Raviglione2, Anna Grassi2, Alessandra Algarotti2, Maria Caterina Micò2, Federico Lussana1,2, Benedetta Rambaldi2, Gianluca Cavallaro2, Giuliana Rizzuto2, Orietta Spinelli2, Alessandro Rambaldi1,2, Silvia Salmoiraghi2

1Università degli Studi di Milano, Milano, Italy, 2ASST Papa Giovanni XXIII, Bergamo, Italy

Background: Relapse is one of the leading causes of transplant failure in myelofibrosis (MF). Post-transplant molecular monitoring of the driver mutation is recommended, but data are lacking about the preferred monitoring method and how to clinically interpret low positivity during follow-up.

Methods: We studied 21 patients transplanted for MF at the Bergamo Bone Marrow Transplant Unit. The median age at transplant was 61 years (42-67). All patients underwent a reduced-intensity conditioning regimen. Five patients (24%) underwent splenectomy before transplant. Minimal residual disease (MRD) monitoring was studied by digital droplet PCR (ddPCR) in patients carrying either JAK2V617F (n=15) or MPL (n=6) mutation. MF patients with CALR mutation are not evaluable by ddPCR at this time. A total of 114 post-transplant samples were studied. Post-transplant MRD monitoring by dd PCR was also performed for the following mutations, identified by Next Generation Sequencing technology: ASXL1, U2AF1, IDH1, PTPN11 and SF3B1.

Results: The JAK2V617F mutation was present at transplant with a median VAF of 78% (18.1-94.3), while the median VAF of the MPL mutation was 78.2% (23-85). An additional high-risk mutation (HRM) was detected by NGS in 11 (52%) patients. According to the Mutation-Enhanced International Prognostic Scoring System 70 plus (MIPPS70 + ), patients were allocated to the low (10%)-intermediate (25%)-high (55%)-very high (2%) risk group. After a median follow-up of 2.6 years (range 0.6-10.9), the 3-year Overall Survival (OS) and Relapse Free Survival (RFS) were 83% and 74% respectively, while the 3-year Cumulative Incidence of Relapse (CIR) and Non-Relapse Mortality (NRM) were 22% and 5%, respectively. Four patients experienced hematological relapse during follow-up, after a median time of 18 months (2-24).

MRD positivity of the driver mutation was detected in 65%, 62%, 67%, 35%, 28%, 29% of patients respectively at day +30, +60, +90, +180, +360, +720 after transplant. The median time to reach an MRD negative status was 64 days (range 28-1091) after transplant, while the median time to develop MRD positivity after the achievement of first negativity was 94 days (59-871).

An MRD-positive status at day +30, day +90 and at any time point >+180 days after transplant was significantly associated with a higher relapse risk (p<0.0001). The 3-year CIR for patients with MRD positivity at day +90 was 32% versus 0% for patients with MRD negativity (Figure 1).

By univariable analysis, MIPPS70plus, presence of HRM mutations, VAF of the driver mutation, unfavorable karyotype, pre-transplant peripheral blasts ≥2% or pre-transplant splenectomy were not associated with the development of MRD positivity at day +90 or day >+180 after transplant. MRD positivity at any time point did not impact OS.

Six patients were also followed up for an additional mutation (ASXL1, U2AF1, IDH1, PTPN11 and SF3B1). The MRD monitoring of these mutations had a similar trend compared to the driver mutation.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (67)

Conclusions: Post-transplant molecular monitoring of a driver mutation can anticipate hematological relapse in MF, thus allowing the starting of pre-emptive therapy to reduce the relapse risk. ddPCR is a reliable method for MRD monitoring after transplant.

Clinical Trial Registry: Not available.

Disclosure: Nothing to declare.

22: Myeloproliferative Neoplasm

P560 EVALUATION AND IMPLICATIONS OF PORTAL AND PULMONARY HYPERTENSION IN MYELOFIBROSIS PRIOR TO TRANSPLANT: A PRACTICE-BASED SURVEY OF THE CHRONIC MALIGNANCIES WORKING PARTY OF THE EBMT

Giorgia Battipaglia 1, Nicola Polverelli2, Joe Tuffnell3, Patrizia Chiusolo4, Marie Robin5, Massimiliano Gambella6, Annoek Broers7, Elisa Sala8, Jakob Passweg9, Sabine Furst10, Henrik Sengeloev11, Remy Dulery12, Moniek de Witte13, Ibrahim Yakoub-Agha14, Maria Chiara Finazzi15, Claudia Wehr16, Arnon Nagler17, Deborah Richardson18, Wolfgang Bethge19, Andrew Clark20, Joanna Drozd-Sokolowska21, Kavita Raj22, Tomasz Czerw23, Juan Carlos Hernández-Boluda24, Donal P. McLornan22

1Federico II University of Naples, Hematology Division, Naples, Italy, 2Unit of Bone Marrow Transplantation, Division of Hematology, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy, 3EBMT Leiden Study Unit, Leiden, Netherlands, 4Università Cattolica Sacro Cuore, Rome, Italy, 5Saint-Louis Hospital, BMT Unit, Paris, France, 6IRCCS Ospedale Policlinico San Martino, Genova, Italy, 7Erasmus MC Cancer Institute, Rotterdam, Netherlands, 8Klinik fuer Innere Medzin III, Ulm, Germany, 9University Hospital of Basel, Basel, Switzerland, 10Programme de Transplantation et Therapie cellulaire de Marseille, Marseille, France, 11Rigshospitalet, Copenaghen, Denmark, 12Hopital Saint Antoine, Paris, France, 13University Medical Center of Utrecht, Utrecht, Netherlands, 14CHU de Lille, Lille, France, 15ASST Papa Giovanni XXIII, Bergamo, Italy, 16Hematology, Oncology and Stem Cell Transplantation, Medical Center, University of Freiburg, Faculty of Medicine, Freiburg, Germany, 17Chaim Sheba Medical Center, Tel-Hashomer, Israel, 18Southampton General Hospital, Southampton, United Kingdom, 19Universitaet Tuebingen, Tuebingen, Germany, 20Glasgow Royal Infirmary, Glasgow, United Kingdom, 21Central Clinical Hospital, The Medical University of Warsaw, Warsaw, Poland, 22University College London Hospital NHS Trust, London, United Kingdom, 23Maria-Sklodowska-Curie National Research Institute of Oncology, Gliwice Branch, Poland, 24Hospital Clinico Universitario, Valencia, Spain

Background: No specific guidance to approach transplant candidates with myelofibrosis (MF)-specific comorbidities exists and there is scarce data on transplant centres policies in this setting.

Methods: An electronic survey to evaluate how MF-specific comorbidities are approached and whether they affect decision to transplant was launched to EBMT centres performing ≥5 MF transplants/year (n=63).

Results: A total of 41 centres (65%) answered. A) pre-transplant evaluation: Radiological assessment of hepato-splenomegaly is often performed prior (88% and 78% for liver and spleen, respectively, mainly relying on CT scan [51%], ultrasound imaging [41%], or both [29%]) to transplant. Screening for portal hypertension (PH) is routinely performed in 54% centres, never in 12%, and guided by clinical manifestations in the remaining. Involvement of hepatologists/gastroenterologists is always/very often considered in patients with signs of PH prior to transplant. Only 7% centres involve hepatologists in all cases, while pre-existing liver function tests abnormalities (n=7), imaging abnormalities (n=8) or signs of PH (n=9) are indications for referral in the remaining centres.

Of note, most centres (61%) do not perform routine screening for gastroesophageal varices; this is systematically considered or guided by clinical manifestations in 7% and 32% centres, respectively. B) Transplant decision-making: Radiological evidence of PH does not routinely represent a formal contraindication for allo-HCT in most cases (78%). Presence of gastroesophageal varices in 34% centres is always (n=6) or occasionally (n=8) considered a formal contraindication to allo-HCT. A prior history of portal vein thrombosis never (78%) or occasionally (15%) represents a formal contraindication. Three centres would not proceed to transplant in such cases. However, in cases complicated with cavernomas, only 44% centres would proceed with transplant, 32% would consider it a formal contraindication and 20% would prioritize clinical consequences of prior thrombosis and cavernoma to guide transplant decisions. Need for transjugular intrahepatic portosystemic shunt (TIPSS) represents a formal contraindication to transplant for 17% centres while 20% would balance disease severity and clinical sequelae of thrombosis. The remaining 63% centres would proceed regardless of prior TIPSS. Less importance is given to non-portal splanchnic vein thrombosis (SVT), with all but one centre proceeding to transplant regardless of prior SVT. Pulmonary hypertension prior to transplant represents a contraindication for transplant in 7 centres while 30 would proceed only after a careful pneumologist evaluation. The remaining centres consider disease severity and benefit-risk ratio. C) post-transplant monitoring: Radiological assessment of hepato-splenomegaly is generally performed after transplant (71% of centres), but at heterogeneous intervals. Post-transplant monitoring of SVT is performed through ultrasound imaging in 78% of cases (n=32, alone [n=11], coupled with CT scan [n=16] or with magnetic resonance imaging [n=3] or with both [n=3]). Specialists are routinely involved for post-transplant monitoring of PH in 34% centres, while in 61% this is based on the presence of overt symptoms.

Conclusions: Our survey highlights a considerable heterogeneity across responding centres in approaching MF-specific comorbidities prior to transplant, with variable influence on transplant decision-making and high variability in post-transplant evaluations. These results suggest that harmonization guidelines are needed to best individualize therapy in this highly comorbid patient population.

Disclosure: No COI to disclose.

22: Myeloproliferative Neoplasm

P561 IMPACT OF NUMBER OF CD34 + CELLS INFUSED ON OVERALL SURVIVAL IN MYELOFIBROSIS PATIENTS AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Filippo Frioni 1, Sabrina Giammarco2, Elisabetta Metafuni2, Maria Assunta Limongiello2, Nicola Piccirillo2, John Donald Marra1, Luca Di Marino1, Luciana Teofili2, Andrea Bacigalupo1, Simona Sica2, Patrizia Chiusolo2

1Università Cattolica del Sacro Cuore, Rome, Italy, 2Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy

Background: Despite the increase of allogeneic hematopoietic stem cell transplantation (HSCT) performed for Myelofibrosis (MF), predictors of survival after transplantation are largely yet to be identified.

Methods: We conducted a monocentric study to determine the impact of various clinical (age, splenomegaly, constitutional symptoms, gender, previous treatment with Ruxolitinib, cytoreductive therapy, cardiovascular risk factors, and a diagnosis of secondary MF), biochemical (anemia, leukocytosis, thrombocytopenia, peripheral blood myeloblasts ≥2% of white blood cell count, and LDH), histological (bone marrow fibrosis at the time of transplant), cytogenetic, molecular, and transplant specific (hematopoietic cell transplantation specific comorbidity index, donor, stem cell source, number of CD34+ cells received at the time of transplantation, number of CD3+ cells received at the time of transplantation, conditioning regimen, Graft versus host disease (GvHD) prophylaxis scheme, and GvHD occurrence) variables on the overall survival of 96 consecutive patients subjected to allogeneic HSCT for MF. Cox regression proportional hazard was used to analyze determinants of overall survival.

Results: The median age at the time of transplant was 54 years (range 29-73), 43 (44%) patients were female and 53 (55%) patients were male. The diagnosis was primary MF in 41 (43%) patients and secondary MF in 55 (57%) patients (19 post-Polycythemia Vera and 36 post-Essential Thrombocythemia), the most common driver mutation was JAK2V617F mutation (68 patients, 71%), grade 3 bone marrow fibrosis was present in 31 (32%) patients at the time of transplant, 70 (73%) patients were previously treated with Ruxolitinib and a radiologically evident splenomegaly was detectable in 91 (95%) patients before transplant. 67 out of 96 (70%) patients harbored an unfavorable cytogenetic, 67 (70%) patients had at least 1 detrimental mutation. Reduced intensity conditioning regimen was largely preferred with only two days of busulfan, and GvHD prophylaxis consisted of a post-transplant cyclophosphamide-based regimen in 79 (82%) patients. 57 (59%) patients underwent an HLA-identical HSCT (38 from matched unrelated donor and 23 from related donor); stem cell source was peripheral blood in 74 (77%) patients, bone marrow in 21 (22%) patients and cord blood in 1 (1%) patient. A total of 17 (19%) patients experienced a grade ≥2 GvHD. The median number of infused CD34+ cells was 5.4 x 10^6/Kg, range 2.3-12 x 10^6/Kg. This study found that both molecular risk and transplant-specific risk were associated with lower overall survival. SRSF2 mutation (p 0.065, HR 2.437), TP53 mutation (p 0.06, HR 3.96), age below 57 years (p 0.03, HR 2.81), thrombocytopenia (p 0.03, HR 2.18), and less than 5 x 10^6/Kg of CD34+ cells (p 0.009, HR 3.86) were significant factors in the univariable analysis. On multivariable analysis, only TP53 mutation (p 0.03, HR 5), and the number of CD34+ cells received (p 0.003, HR 21) remained statistically significant.

Conclusions: This study shows the impact of the number of CD34+ cells infused at time of transplantation on overall survival in MF patients after allogeneic HSCT.

Disclosure: Nothing to declare.

22: Myeloproliferative Neoplasm

P562 AVAPRITINIB IN THE POST-ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT SETTING IN PATIENTS WITH ADVANCED SYSTEMIC MASTOCYTOSIS

Deborah Christen 1, Johannes Luebke2, Juliana Schwaab2, Anne Kaiser1, Svetlana Rylova3, Saša Dimitrijević3, Ilda Bidollari3, Jens Peter Panse1, Andreas Reiter2,1

1University Hospital RWTH Aachen, Aachen, Germany, 2University Hospital Mannheim, Mannheim, Germany, 3Blueprint Medicines Corporation, Cambridge, United States

Background: Advanced Systemic Mastocytosis (AdvSM) is a rare clonal hematologic neoplasm associated with poor survival and driven by KIT D816V mutation in most cases. Avapritinib, a tyrosine kinase inhibitor selectively targeting KIT D816V, significantly improved outcomes for patients with AdvSM by inducing deep and durable responses. Use of avapritinib to induce remission before allogeneic hematopoietic stem cell transplantation (alloHSCT) in AdvSM was reported previously. However, little is known about the role of avapritinib in the post-alloHSCT setting of AdvSM. We here describe first experiences with avapritinib in patients with AdvSM post-alloHSCT.

Methods: Charts of patients enrolled in the phase 2 PATHFINDER clinical trial (NCT03580655) of avapritinib in AdvSM were retrospectively reviewed to identify those who received avapritinib post-alloHSCT. Mutation analysis and serum tryptase levels were assessed in peripheral blood. Responses to treatment were assessed centrally using modified International Working Group-Myeloproliferative Neoplasms Research and Treatment and European Competence Network on Mastocytosis (mIWG-MRT-ECNM) criteria.

Results: Three patients were identified; 2 enrolled in PATHFINDER post-alloHSCT, 1 enrolled pre-alloHSCT (Table). All were male, aged 61, 64, and 68 years, with diagnosis of mast cell leukemia with associated hematologic neoplasm. All had normal cytogenetics and Karnofsky performance status was 100% (patient 1) or 90% (patients 2 and 3). Transplant and donor characteristics are described in the table. Patients 1 and 3 started midostaurin 3-4 months post-alloHSCT but discontinued due to disease progression or tolerability, respectively; both then started avapritinib approximately 1-year post-alloHSCT. Patient 1 started avapritinib 200 mg daily and reduced dose to 50 mg due to grade 3 thrombocytopenia; treatment duration was 26 months and best response was complete remission with partial hematologic recovery (CRh). Patient 1 discontinued avapritinib due to angiosarcoma relapse leading to death. Patient 3 started avapritinib 100 mg daily with no dose modifications and achieved complete response; treatment is ongoing (>18 months) with good tolerability. Patient 2 started avapritinib 200 mg daily 3 months post-alloHSCT with no dose modifications; treatment duration was 17 months and best response was partial response (PR). Patient 2 discontinued due to grade 4 COVID-19 pneumonia and E. coli sepsis and died following worsening of E. coli sepsis. Neither death was considered avapritinib-related. Overall, all patients experienced reductions in bone marrow mast cell %, KIT D816V variant allele frequency, and serum tryptase level. The safety profile of avapritinib was favorable with only 1 patient requiring dose modification due to thrombocytopenia, and there were no treatment-related discontinuations. Most adverse events (AEs) related to avapritinib were grade 1/2; grade ≥3 AEs included thrombocytopenia and increased gamma-glutamyl transferase (patient 1). Patient 3, ongoing, had no grade ≥3 events, thrombocytopenia resolved after alloHSCT, and neutrophil counts stabilized at >1,000/µL.

Conclusions: In this retrospective chart review, avapritinib use appears feasible in patients with AdvSM post-alloHSCT, with durable responses and reduction of objective disease biomarkers. Patients should be closely monitored as clinically indicated for thrombocytopenia and/or transplant-related complications. Safety profile was consistent with previously published reports, and no new findings emerged. Further studies are needed to confirm optimal administration of avapritinib post-alloHSCT.

Table

Patient 1

Patient 2

Patient 3

Patient characteristics at alloHSCT

Age, years

68

61

64

Diagnosis

MCL-ET

MCL-MDS

MCL-MDS

Sorror score

5

Response before alloHSCT

PD on midostaurin

PD on midostaurin

CI on avapritinib

AlloHSCT

Type of donor (match)

Unrelated (10/10)

Related (10/10)

Unrelated (10/10)

Conditioning

Fludarabine, Busulfan

Fludarabine, Treosulfan

Fludarabine, TBI

GVHD prophylaxis

CsA, MTX, ATG

CsA, MTX

ATG, Tacrolimus, MMF

Transplant outcome

d + 35 post-alloHSCT, BM MC burden 32%

Chimerism: 88% (d + 35), 100% (d + 1,019)

d + 28 post-alloHSCT, BM MC burden 22%

Chimerism: 95% (d + 28), 100% (d + 546)

DNMT3A, SRSF2, TET2 negative, KIT D816V 17% in BM

BM MC burden <5%

Chimerism: 100% (d + 734)

Acute GVHD, grade

II (GI tract)

Suspected GVHD, DD infectious colitis

I

Other complications

VOD, acute kidney failure, CMV reactivation

Acute kidney failure, grade 2 heart failure, cardiorenal syndrome, ischemic stroke, ascending colon colitis

Paroxysmal atrial fibrillation

Post-alloHSCT treatments and outcomes

Midostaurin dose and response

Started 4 mo post-transplant, 50 mg 1-0-1, duration 8 mo; response = PD

NA

Started 3 mo post-transplant, 25 mg 3-0-3, duration 8 mo: response = PD

Reason for discontinuation of midostaurin

PD, relapse

NA

Toxicity (neutropenia)

Avapritinib daily dose and response per mIWG-MRT-ECNM criteria

200 mg started 1 y post-alloHSCT titrated to 50 mg; duration 2 y, 2 mo; best response = CRh

200 mg started 3 mo post-alloHSCT; duration 1 y, 5 mo; best response = PR

100 mg started 1 y post-alloHSCT; best response = CR

Grade ≥3 treatment related AEs

Thrombocytopenia, gamma GT increased

None

None

Status at last follow-up

Avapritinib discontinued due to relapse of angiosarcoma

Avapritinib discontinued due to E. coli sepsis

Avapritinib ongoing; >1 y, 6 mo

  1. AE, adverse event; alloHSCT, allogeneic hematopoietic stem cell transplantation; ATG, antithymocyte globulin; BM, bone marrow; CI, clinical improvement; CMV, cytomegalovirus; CR, complete response; CRh, complete remission with partial hematologic recovery; CsA, cyclosporine A; d, day; DD, differential diagnosis; ET, essential thrombocytopenia; gamma GT, gamma-glutamyl transferase; GI, gastrointestinal; GVHD, graft versus host disease; MC, mast cell; MCL, mast cell leukemia, MDS, myelodysplastic syndrome; mIWG-MRT-ECNM, modified International Working Group-Myeloproliferative Neoplasms Research and Treatment and European Competence Network on Mastocytosis; mo, month; MTX, methotrexate; MMF, mycophenolate mofetil; NA, not applicable; PB, peripheral blood; PD, progressive disease; PR, partial response; SD, stable disease; TBI, total body irradiation; VAF, variant allele frequency; VOD, veno-occlusive disease; y, year.

Clinical Trial Registry: Clinicaltrials.gov NCT03580655.

Disclosure: DKC - Blueprint Medicines: Consultancy and Honoraria; Pfizer: Consultancy; BeiGene: Consultancy; Astra Zeneca: Honoraria; Amgen: Travel support; Novartis: Consultancy; Janssen: Travel support.

JL - Nothing to declare.

JS - Blueprint: Consultancy and honoraria; Novartis: honoraria.

AK - Blueprint Medicines: Consultancy and Honoraria.

SR - Current employee of Blueprint Medicines Corporation.

SD - Current employee of Blueprint Medicines Corporation.

IB - Current employee of Blueprint Medicines Corporation.

JPP - Consultant, speaker, and/or scientific advisory boards for Alexion, Apellis, AstraZeneca, Blueprint Medicines Corporation, Boehringer Ingelheim, Bristol Myers Squibb, Chugai Pharmaceutical, F. Hoffmann-LaRoche, Novartis, Pfizer, and Sanofi-Pasteur.

AR - Consultant for Blueprint Medicines Corporation; Cogent Therapeutics LLC; Novartis Pharma; grants (institution) from Blueprint Medicines Coporation and Novartis Pharma.

22: Myeloproliferative Neoplasm

P563 ASCIMINIB: A NEW TKI BEFORE AND AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHRONIC MYELOID LEUKEMIA

Iuliia Vlasova 1, Elena Morozova1, Elza Lomaia2, Tamara Chitanava2, Ksenia Tsvirko1, Tatiana Rudakova1, Nikita Volkov1, Tatiana Gindina1, Dmitriy Motorin3, Yulia Alexeeva2, Valeriya Katerina1, Ivan Moiseev1, Alexander Kulagin1

1RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russian Federation, 2Almazov National Medical Research Centre, St. Petersburg, Russian Federation, 3Russian Research Institute of Hematology and Transfusiology, St. Petersburg, Russian Federation

Background: Pre-transplant use of 2nd generation TKIs (nilotinib / dasatinib) does not change the risk associated with allogeneic hemopoietic stem cell transplantation (allo-HSCT) recipients (Niederwieser C.,2021, Masouridi-Levrat S.,2021). Asciminib (ASC), a first-in-class BCR::ABL1 allosteric inhibitor, is a therapeutic option in patients after allo-HSCT (Fiona Fernando et al., 2023). There is yet no data available for patients receiving ASC before allo-HSCT.

Methods: Aim: To present the updated results of pre- and post-transplant ASC in allo-HSCT recipients.

In the Managed Access Program (MAP) by Novartis we reviewed data of 20 patients (pts) with CML, who received pre- and post-HSCT therapy with ASC between September 2021 and January 2023. 12 pts received ASC with median duration of 194 days (61-377) and subsequently underwent allo-HSCT. Baseline characteristics: male 58%; median age 41 years (range 28-59); median duration of CML before ASC 2.8 years (range 0.3-15); 3 pts were in chronic phase (CP) CML, 4 and 5 pts had a history of accelerated phase (AP) and blast crisis (BC), respectively. Nine pts (75%) had BCR::ABL1 mutations, 7 pts (58%) had BCR:ABL1t315i. 4 pts (33%) had additional chromosomal abnormalities. 2 pts (16%) had ASXL1 mutations. 9 pts (75%) received ≥3 TKIs, including ponatinib in 4 pts (33%). CML status before allo-HSCT: 4 pts achieved CHR, 1 – CCyR, 2 and 1 MMR and MR4, respectively, and 4 pts without CHR. All pts received allo-HSCT with reduced dose intensity conditioning regimen and GVHD prophylaxis with PtCy. 14 pts received post-transplant ASC: 8 pts with minimal residual disease (MRD), 6 pts with molecular relapse (MR).

Results: Pre-transplant ASC dose was 40 BID for 5 (41%) pts, 7 (59%) pts started with 200 mg BID. There were no adverse events (AEs) of any grade in 11 pts (92%) and 1 (8%) patient developed grade 3-4 neutropenia and thrombocytopenia and continued treatment at reduced dose of 20 mg BID. No unusual toxicity was observed during conditioning. Me engraftment time was D + 20 (range 18-24). One patient developed gr. 1 VOD with resolution during therapy. Primary and secondary graft failures were recorded in 1 pt each. In the post-transplant period Me level of BCR::ABL1 at the start of ASC therapy was 0.115% (0.003-53%). In 6 (42%) pts the starting dose of ASC was 20 mg BID; the dose was de-escalated due to poor graft function in the early post-transplant period. Only 1 pt (8%) had an AE (thrombocytopenia gr 4). Acute GVHD 3-4 up to 100 days was 21%, and was not significantly associated with ASC. A total of 14 (70%) pts achieved CMR. With a Me follow-up after allo-HSCT of 495 days 13 (65%) pts are alive.

Conclusions: In this small study ASC was effective as bridge-therapy before allo-HSCT in highly pretreated CML pts. Post-transplant ASC was well tolerated and induced improvement in molecular response. Low rate of AEs supports ASC as an attractive post-transplant strategy and potentially delays the need for donor lymphocyte infusion.

Disclosure: Nothing to declare.

22: Myeloproliferative Neoplasm

P564 EXCELLENT LONG TERM SURVIVAL DESPITE LOW RATES OF GVHD IN PATIENTS WITH MYELOFIBROSIS FOLLOWING ALLOGENIC HSCT: A SINGLE CENTRE EXPERIENCE

Mariam Amer 1, Michael Nathan1, Christopher Dalley1, Kim Orchard1, Sara Main1, Amy Creighton1, Jane Lamb1, Helen Snow1, Annie Major1, Sarah Holtby1, Linda Jarvis1, Mathew Lee1, Muhammad Maqbool1, Hwai Jing Hiew1, Fiona Duggan1, Deborah Richardson1

1University Hospital Southampton, Southampton, United Kingdom

Background: A haematopoietic stem cell transplant (HSCT) is the only curative option for patients with myelofibrosis. Since 2014: this has represented 5 % of our yearly transplant activity. We retrospectively analysed the overall survival (OS) and Graft versus host disease free relapse free survival (GFRS)in this challenging cohort.

Methods: We retrospectively analysed the electronic records of patients who received a haematopoietic stem cell transplant, for the period between June 2009 – September 2022. We have locked the follow up data in September 2023. Statistical analysis and Kaplan Meier survival curves were analysed with graph pad prism, version 10.

Results:

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (68)

A total of 34 patients received 37 allogeneic stem cell transplants for myelofibrosis. Mean age of the patients was 57 years; range 41-71 years of age. 10/34 patients had a sibling donor, 19 /34patients had a fully matched unrelated donor, 6/34 patients had one antigen mm unrelated donor. 18/34 patients had primary myelofibrosis; 16/34 patients had secondary Myelofibrosis. For the former, 3/18,8/18and 7/18 were classified as intermediate 1, intermediate 2, high-risk groups respectively. For secondary myelofibrosis 3/16,4/16,6/16,3/16 were classified as low risk, intermediate1, intermediate 2, and high risk respectively. 19/34 patients received fludarabine, melphalan and campath, 14/34 patients received Fludarabine, busulfan and campath conditioning, 1/34 patient received Fludarabine treosulfan and campath. Ciclosporin and methotrexate were used as graft versus host disease (GVHD) prophylaxis. The median OS was not reached (undefined) for the whole cohort, where the GFRS median was 8.9 years. 6/34(17 %) patient’s disease has relapsed, progression free survival was undefined. 1/6 patient has reattained remission by escalating doses of donor lymphocyte infusions, the second had a second stem cell transplant, 2 patients are undergoing treatment in the form of escalating doses of DLI, one patient is awaiting a second transplant, and two patients died of progressive disease.3/34 patients had graft failure- one was treated to resolution with escalating doses of DLI, and two received another stem cell transplant, using the same donor. The Incidence of acute Graft versus host disease (GVHD)Grade 1-2 (16/34), Grade 3- 4 (5/34 patients). While 4 /34, 3/34,3/34 had mild, moderate and severe chronic GVHD respectively. Of the latter group, only one patient required ongoing long-term immunosuppression. Transplant related mortality in our cohort was 17%.5/34 patients had Veno-oclusive disease, which was treated with no significant sequel. 24/34 patients are in complete remission.13 patients have received further therapy; DLI post-transplant, 3/13 received DLIs prophylactically,2/13 for mixed T cell chimerism,2/13 for delayed immune reconstitution, and 6/13 were indicated for loss of myeloid chimerism and relapse. The use of DLI was successful in 3/13 to restore full donor chimerism, and 2 patients are still ongoing treatment.

Conclusions: Our experience highlights excellent outcomes with long term disease control and acceptable transplant related mortality. It highlights the successful use of DLIs in the post-transplant setting.

Disclosure: Nothing to declare.

22: Myeloproliferative Neoplasm

P565 VENETOCLAX PLUS AZACITIDINE VERSUS AZACITIDINE ALONE AS POST-TRANSPLANT MAINTENANCE TREATMENT IN HIGH-RISK AML AND MDS

Yigeng Cao1, Wenwen Guo1, Erlie Jiang 1

1Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin, China

Background: Disease recurrence is the main cause of transplantation failure for high-risk acute myeloid leukemia (AML) and myelodysplastic syndrome (MDS). Exploring innovative approaches to prevent relapse in AML and MDS after Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is in urgent need. Both bcl-2 inhibitor venetoclax (VEN) and azacitidine (AZA) possess significant antitumor activity effects against myeloid neoplasms. The efficacy and safety of AZA alone as maintenance therapy in the posttransplant setting have been reported but the results are controversial. The combination of VEN and AZA has resulted in high response rates in treating AML patients.

A prospective, multicenter, randomized clinical trial has been conducted to compare the efficacy and safety of the combination of VEN and AZA (VEN/AZA) and AZA alone as maintenance treatment in high-risk AML and MDS in the post-transplant setting.

Methods: Patients with AML or MDS who were evaluated as high risk of relapse and underwent allo-HSCT were randomly assigned into VEN/AZA group and AZA group by gender, age, and disease type based on the central randomization system. AZA was administered by subcutaneous injection for 5 days at a dose of 50 mg/m2. VEN was administrated at the dosage of 200 mg daily from day 1 to day 7. VEN/AZA maintenance therapy regimen comprised AZA and VEN used alternately every month. AZA alone group was administrated AZA every 2 months. Maintenance treatment lasted at least 1 year after transplantation unless relapse occurred. The primary endpoint of the study included overall survival (OS), disease-free survival (DFS), and cumulative incidence of relapse (CIR). The second endpoints included adverse events (AEs), nonrelapse mortality (NRM), incidence of acute graft versus host disease (aGVHD), and chronic GVHD (cGVHD).

Results: From June 2022 to June 2023, 24 patients were given the study protocol. 9 patients (6 AML, 3 MDS) received VEN/AZA maintenance and 15 (7 AML, 8 MDS) patients received only AZA maintenance. The median initial time to receive maintenance treatment was 3.67 (IQR, 3.07-5.51) months after transplantation. The median follow-up time was 11.30 (95%CI, 8.18-14.42) months. Only 1 patient relapsed and died 3 months after relapse on the study. The overall survival (OS) and the cumulative incidence of relapse (CIR) at 500 days were 100% vs 75.0% (95% CI, 42.59%-100%, p=0.39) and 0% vs 12.5% (95% CI, 0.48%-44.48%, p=0.39) for VEN/AZA and AZA group, respectively. The most common adverse effects (AEs) were hematological toxicities. Grade 3 or 4 hematological toxicities were observed in 5/9 (55.5%) in the VEN/AZA and 7/15 (46.7%) in the AZA group.

Conclusions: Our results suggested that both VEN/AZA and AZA maintenance therapy were tolerated and the combination of VEN and AZA as maintenance therapy may be feasible and safe in the posttransplant setting to prevent relapse for high-risk AML and MDS patients.

Disclosure: Nothing to declare.

22: Myeloproliferative Neoplasm

P566 CURRENT PRACTICE WHEN TRANSPLANTING PATIENTS WITH MYELOFIBROSIS: A SURVEY ON BEHALF OF THE BRITISH SOCIETY OF BLOOD AND MARROW TRANSPLANTATION AND CELLULAR THERAPY (BSBMTCT)

Emma Kempshall 1, Donal McLornan2, Anjum Khan3, Pramilla Krishnamurthy4, Daniele Avenoso4, Sebastian Francis5, Matthew Collin6

1University Hospital of Wales, Cardiff, United Kingdom, 2University College London, London, United Kingdom, 3Leeds Teaching Hospital, Leeds, United Kingdom, 4Kings College Hospital NHS Foundation Trust, London, United Kingdom, 5Sheffield Teaching Hospital, Sheffield, United Kingdom, 6Newcastle Hospital NHS Foundation Trust, Newcastle upon Tyne, United Kingdom

Background: Whilst several agents offer symptom control in myelofibrosis (MF), allogeneic haematopoietic cell transplantation (allo-HCT) remains the only curative option. As more patients with MF proceed to allo-HCT, many challenges exist when considering the optimum transplant strategy.

Methods: This survey was developed by a group of UK transplant physicians and was then approved by the Clinical Trials Sub Committee (CTSC) of the BSBMTCT (CTCS-2101) before being distributed via their network. It explored pre-, peri- and post-transplant strategies in MF patients planned for allo-HCT.

Results: A total of 16 replies were received from physicians across 12 UK allo-HCT centres. As replies within individual centres varied, they were treated as individual responses.

A total of 13/16 responders used NGS profiling routinely ahead of transplant. The DIPSS-plus score was universally used for risk stratification, with 11/16 also using MIPSS70 and 8/16 MYSEC-PM. 4/16 responders used the MTSS score to predict transplant outcome.

Dose of PBSC requested ranged from 4-10 x10^6/kg.

A total of 13/16 responders used Flu Bu ATG as standard conditioning, with 3/16 using Flu Bu Campath and 1/16 Flu Mel Campath. 2/16 centres were routinely using Treosulfan conditioning. Some varied conditioning regimens based on age or comorbidities.

Dose of Busulfan varied; 6.4mg/kg (n=3), 8mg/kg (n=9) and 9.6mg/kg (n=3). Several centres dosed based on age or comorbidities[Pk1]. Both centres using Treosulfan utilised 30g/m2. A further 5 centres were considering introducing Treosulfan.

Of the 13/16 centres using ATG, 6/13 used GenzymeÔ and 6/13 used ThymoglobulinÔ. Of the 6 Genzyme users, 2/6 used 6mg/kg and 3/6 used 5mg/kg. For Thymoglobulin,4/6 administered a total of 5mg/kg. One of these specified a higher dose (7.5mg/kg) in mismatched transplants. One centre used 3.75mg/kg/day for 3 days (total 11.25mg/kg).

A total of 4/16 centres used Campath. 1/4 used 50mg, 1/4 50mg in MUDs and 30mg in siblings and 1/4 30mg. 1 responder used ATG or Campath.

All centres used a ciclosporin backbone for GvHD prophylaxis, with 4/16 also using Methotrexate, 6/16 MMF and 4/16 using MMF in MUD allos only. 1 centre routinely used post-transplant cyclophosphamide (PTCY) in mismatched donor allo-HCT. Commencement of immunosuppression wean ranged from Day+30 (sib only) through to D140.

Regarding ruxolitinib, 13/16 centres weaned ahead of transplant with 9/16 stopping prior to conditioning, 5/16 during conditioning and 2/16 post engraftment.

Regarding measurable residual disease monitoring, JAK2/CALR burden was routinely measured by 11/16 responders at variable post allo-HCT time points.

[Pk1]This was duplicated info above so deleted the statement earlier.

Conclusions: FB-ATG remains the commonest conditioning regimen used across the UK for MF allo-HCT, although dosing strategies varied. Although treosulfan is currently only used by a few centres, several are considering its introduction. A recent EBMT registry study would support its use (Blood (2023) 142 (Supplement 1): 473).

PBSC dose, use of JAKi, timing of immunosuppression wean and post-transplant MRD monitoring strategies varied significantly, reflecting ongoing challenges with poor engraftment, rates of GVHD and relapse. A prospective randomised controlled trial of allo-HCT strategies in MF including optimal conditioning, the role of JAK inhibition, MRD and DLI strategies is justified.

Clinical Trial Registry: Clinical Trials Sub Committee of the British Society of Blood and Marrow Transplantation and Cellular Therapy (CTCS-2101).

Disclosure: Emma, Kempshall: Medac- Funding (Honoraria).

Anjum, Khan: Medac – Funding (Honoraria).

Donal, McLornan: JAZZ (Honoraria), Novartis (Speaker Fees), AbbVie (Speaker Fees), GSK (Speaker Fees and honoraria).

Pramila, Krishmamurthy: Abbvie (speaker fees), Astellas (speaker fees), Janssen (Conference support and speaker fees), Stemline-Menarini (speaker fees), Jazz (conference support).

22: Myeloproliferative Neoplasm

P567 LONG-TERM OUTCOME OF ALLOGENEIC TRANSPLANTATION IN PRIMARY AND SECONDARY MYELOFIBROSIS

Alzbeta Zavrelova 1,1, Benjamin Visek1, Miriam Lanska1, Pavel Zak1, Petra Lukesova1, Jakub Radocha1

1Charles University, Faculty of Medicine in Hradec Králové, Hradec Kralove, Czech Republic

Background: Allogeneic stem cell transplantation is the only curable option for patients with myelofibrosis and is clearly indicated in eligible patients with higher-risk disease.

Methods: This was a retrospective analysis of all consecutive patients with primary or secondary myelofibrosis transplanted at our institution from Januray 2002 till December 2022. Patients with acute leukemia transformation were not included.

Results: We were able to identify 32 patients (20 men and 12 women) who underwent allogeneic transplantation for myelofibrosis in these 20 years. Their median age was 59 years (43-65 years). All patients were diagnosed with primary or secondary myelofibrosis which evolved from other Ph negative myeloproliferative diseases. 5 patients were transplanted with a matched sibling donor, 1 patient with a mismatched sibling donor 8/10, 26 patients were transplanted from unrelated donors (17 pts matched unrelated donor 10/10 and 9 pts mismatched unrelated donor 9/10). All conditioning regimens were reduced intensity with fludarabine and busulfan 8-10mg/kg (roral or equivalent intravenous dose) and all patients received Thymoglobuline as GVHD prophylaxis. Median overall survival was 114.2 months (overall survival in 1 year was 81%) and the median progression-free survival was not reached. 10 pts (31%) experienced relapse, 3 pts had progression to acute leukemia. The incidence of all grade aGVHD, grade 3-4 aGHVD, and chGVHD was 34%, 13% and 22% respectively. The cause of death was relapse in 9 pts, GVHD in 3 pts, COVID-19 infection in 2 pts, one pt died from sepsis, breast cancer, EBV PTLD and sudden death.

Conclusions: Allogeneic transplantation is a valid option for patients with higher risk of myelofibrosis with very good long-term results.

Disclosure: none conflict of interest.

7: New Drugs and Cell-based Immune Therapies

P568 FIRST-IN-HUMAN PHASE I/II CLINICAL TRIAL OF IG-TREGS FOR GVHD PREVENTION

Memnon Lysandrou1, Dionysia Kefala1, Panagiota Christofi1,2, Maria Liga1, Antonis Miggos3, Charys Papagregoriou4, Elisavet Vlachonikola3, Nikolaos Savvopoulos1, Vassiliki Zacharioudaki 1, Ioanna Vallianou2, Eleutheria Sagiadinou1, Dimitris Tsokanas1, Rodanthy Theodorellou5, Anastasia Papadopoulou2, Evi Triantafyllou1, Ioanna Sakellari2, Paul Costeas4,6, Anastasia Chatzidimitriou3, Evangelia Yannaki2, Alexandros Spyridonidis1

1University General Hospital of Patras/University of Patras, Patras, Greece, 2George Papanikolaou Hospital, Thessaloniki, Greece, 3Institute of Applied Biosciences, Centre for Research and Technology Hellas, Thessaloniki, Greece, 4Center for the Study of Hematological and other Malignancies, Nicosia, Cyprus, 5Pharmassist Ltd., Athens, Greece, 6Karaiskakio Foundation, Nicosia, Cyprus

Background: Our approach for GVHD prevention aims to mimic the paradigm of foeto-maternal tolerance, which represents the best example of allogeneic tolerance in nature. HLA-G is an immunomodulatory molecule expressed in the placenta to protect the foetus from maternal immune attack and which is epigenetically silenced in normal healthy tissues. In preclinical studies, we epigenetically modulated peripheral blood T-cells in a clinical scale GMP-protocol to produce a highly in vitro and in vivo immunosuppressive induced-Treg population enriched in HLA-G expressing T-cells(ATMP termed iG-Tregs). Besides de novo HLA-G expression, iG-Tregs display distinct traits of other established regulatory T-cell populations(e.g. LAG-3, TIGIT, TIM-3). We herein present the study protocol of the first-in-human phase I/II clinical trial of iG-Tregs and the immune-monitoring assessment of the first patient(IG01) included(EUDRACT-2021-006367-26).

Methods: We conduct a phase I/II, two-center, interventional, dose escalation(3 + 3 design, cohorts of 0,1/0,5/1,5x106 iG-Tregs cells/kg) study in adult patients undergoing allo-HCT from an HLA-matched sibling donor, which serves also as the donor for iG-Treg manufacturing. The primary objective is to assess the safety and the maximum tolerable dose of ex vivo generated iG-Tregs and the secondary objective is to assess the clinical efficacy in preventing GVHD. Patient and donor eligibility are evaluated at d + 30 after allo-HCT, donor undergoes 60-120ml unstimulated leukapheresis, and infusion is performed approximately 4 weeks later. Patients with aGVHD≥II any time before infusion are excluded. Product is released after standard QC testing and should contain ≥10% viable HLA-G+CD3+ cells(Table 1). After infusion, the patient undergoes fast pharmacological immunosuppression tapering. Besides clinical assessment, product and patient samples are analysed for their phenotype via spectral flow-cytometry(Aurora, Cytek) and for their T-cell-receptor(TCR) repertoire via Immune-seq(Illumina).

Table 1: Quality control testing of final iG-Tregs product.

Quality Control Test

Method

Criteria

Purpose

Sterility

Cultures for aerobic and anaerobic bacteria & cultures for fungi

Negative

Final Release

Mycoplasm

Biochemical assay

Negative

Final Release

Endotoxin Levels

LAL test

< 5.0 EU/kg

Final Release

Immunophenotype

Flow cytometry

CD3+ ≥ 80%, alive HLA-G+ cells ≥ 10%

Product Manufacturing

Cell Numbers

Cell counting with haemocytometer

≥ 0,1x106/kg

Product Manufacturing

Viability

Trypan blue staining

Viable cells ≥ 50%

Product Manufacturing

Results: IG01 is a 58-year-old female with high-risk AML who underwent an HLA-matched allo-HCT(CMV+/+) from her 51-year-old female sibling. Starting material for iG-Tregs manufacturing was 118ml steady-state leukapheresis collected at d + 33(38,6% CD3+ cells). The final product contained 60x106cells(1,5-fold expansion of in vitro culture), 82,3% CD3+ cells and 13,52% HLAG+CD3+ cells. iG-Tregs infusion(5,6x106 cells; 0,1x106/kg) took place on d + 64 followed by 25%-weekly cyclosporine dose reduction. The patient did not present any infusion-related toxicities and remained GvHD-free, disease-free, without CMV reactivation and no other complications up to last follow-up(d + 201). Integrated molecular and phenotypic monitoring of IG01 revealed an initial rise in HLA-G + T-cells at week 1 post-infusion(% HLA-G+/T-cells, baseline vs 1-week; 3,69% vs 4,88%), retention of 7,2% of iG-Tregs-derived TCR clonotypes at the 4-week timepoint, and increase of naturally occurring CD4+CD25+CD127- Tregs(nTregs) at 8 weeks post iG-Tregs administration(nTreg/T-cells, baseline vs 8-week; 2,06 vs 3,56).

Conclusions: This is the first-in-human phase I/II study assessing iG-Tregs in the context of GvHD. Up to now, one patient has been enrolled while several more are under evaluation for inclusion in the upcoming months. IG01 displayed excellent clinical outcome with initial data hinting towards possible transient persistence of iG-Tregs following administration and complex immunological interplay within the T-cell compartment.

Clinical Trial Registry: EUDRACT-2021-006367-26.

Disclosure: Nothing to declare.

7: New Drugs and Cell-based Immune Therapies

P569 GILTERITINIB WITH VENETOCLAX, ACTINOMYCIN D, AND LOW-DOSE CYTARABINE AS BRIDGING TO ALLOGENEIC STEM CELL TRANSPLANTATION IN RELAPSED OR REFRACTORY FLT3 MUTATED ACUTE MYELOID LEUKEMIA

Andrius Žučenka1,2, Guoda Daukėlaitė1,2, Vilmantė Vaitekėnaitė1,2, Regina Pileckytė1,2, Igoris Trociukas2, Adomas Bukauskas1,2, Rita Čekauskienė 1,2, Laimonas Griškevičius1,2

1Vilnius University, Vilnius, Lithuania, 2Vilnius University Hospital Santaros Klinikos, Vilnius, Lithuania

Background: Allogeneic stem cell transplantation (alloSCT) is essential in improving outcomes of relapsed or refractory acute myeloid leukemia (R/R AML) patients. Though single-agent Gilteritinib is the standard-of-care for R/R FLT3m AML, preliminary data of novel regimens combining second-generation FLT3 inhibitors (FLT3i), Venetoclax and hypomethylating agents demonstrate higher response and bridging to alloSCT rates in transplant-eligible patients. Herein, we present our data on a quadruplet regimen G-ACTIVE as salvage and bridging therapy for R/R FLT3m AML.

Methods: We performed a retrospective, single-center study. The inclusion criteria were: age > 18 years, diagnosis of R/R FLT3m AML, salvage therapy with the quadruplet G-ACTIVE, and eligibility for alloSCT at baseline. All patients gave consent for data collection and treatment which was approved by the Institutional Ethics Committee. The 28-day G-ACTIVE cycle consisted of Venetoclax (ramp-up of 100-200-400 mg on days -2, -1, 0, and 600 mg/d from day 1 and continued individually for a maximum of 28 days), Gilteritinib 80 or 120 mg/d started on any day of Cycle 1, Actinomycin D 12.5 mcg/kg i/v on days 1-3 (or days 1,2 if age > 65 years) and Cytarabine 20 mg/m2 s/c on days 1-10. The number of Venetoclax and Gilteritinib days was individualized based on the patient’s status, age, early bone marrow assessment (Day 10-14), and treatment-emergent toxicity. Responders could receive a second G-ACTIVE cycle based on the alloSCT timing and measurable residual disease (MRD).

We collected data on patients’ characteristics, AML genomics, prior antileukemic therapy, CR+CRp, ORR (CR+CRp+MLFS), MRD negativity (multiparameter flow cytometry and qRT-PCR) rates, percentage of patients bridged to alloSCT, conditioning regimens, Gilteritinib maintenance rates, OS, RFS, EFS, and toxicity after G-ACTIVE.

Results: Thirteen patients (8 female) have been included in the study. The median age was 64 years (34-72) and the median ECOG was 1 (0-2). FLT3-ITD was identified in 77% (10/13) of cases, 15% (2/13) had FLT3-TKD, and 8% (1/13) had FLT3-ITD + FLT3-TKD. The patients had previously received a median of 2 (1-3) prior therapy lines, 53% (7/13) were exposed to Midostaurin, and 8% (1/13) had previously received Venetoclax. The ORR and CR+CRp rates were 100% (13/13) and 77% (10/13), respectively. MRD negativity was confirmed in 46% (6/13). AlloSCT was performed in 85% (11/13) with either MAC (36%, 4/11) or RIC (64%, 7/11). All 11 allotransplanted patients restarted single-agent Gilteritinib as post-alloSCT maintenance. Two patients in MLFS did not proceed to alloSCT due to hemophagocytic lymphohistiocytosis-related death in aplasia (1) and COVID-19 (1). The median OS, RFS, and EFS were not reached. The 24-month OS, RFS, and EFS were 67%, 53%, and 51%, respectively). Detailed protocol treatment and hematological recovery data are summarized in Table 1. The most common non-hematological Grade 3 adverse events were febrile neutropenia (69%, 9/13) and pneumonia (23%, 3/13). One patient died within the first 60 days after G-ACTIVE.

Table 1.

Characteristic

Patients (N=13)

Gilteritinib 80 mg dose during cycle 1 of G-ACTIVE, % (n/N)

31% (4/13)

Gilteritinib 120 mg dose during cycle 1 of G-ACTIVE, % (n/N)

69% (9/13)

Gilteritinib days during cycle 1 of G-ACTIVE, median (min-max)

21 (9-28)

Venetoclax days during cycle 1 of G-ACTIVE, median (min-max)

14 (10-28)

ANC recovery >0.5 x109 day during cycle 1 of G-ACTIVE, median (min-max)

26 (18-57)

PLT recovery >50 x109 day during cycle 1 of G-ACTIVE, median (min-max)

27 (17-37)

ANC recovery >1 x109 day during cycle 1 of G-ACTIVE, median (min-max)

27 (19-59)

PLT recovery >100 x109 day during cycle 1 of G-ACTIVE, median (min-max)

33 (20-40)

Conclusions: The quadruplet G-ACTIVE salvage regimen resulted in promising antileukemic efficacy with a high bridge-to-alloSCT rate in transplant-eligible R/R FLT3m AML patients.

Clinical Trial Registry: Clinical trial number: 2019/2-1088-591. Vilnius Regional Bioethics Committee, https://www.mf.vu.lt/mokslas/vilniaus-regioninis-biomedicininiu-tyrimu-etikos-komitetas.

Disclosure: Žučenka: Abbvie: Consultancy, Honoraria, Other: Travel Expenses; Astellas: Consultancy, Honoraria; Pfizer: Consultancy; Novartis: Consultancy, Honoraria, Other: Travel Expenses; Jannsen: Consultancy, Honoraria, Other: Travel Expenses; Takeda: Other: Travel expenses.

Pileckytė: Abbvie: Consultancy, Honoraria, Other: Travel Expenses.

Trociukas: Takeda: Other: Travel expenses.

Griškevičius: Miltenyi Biomedicine: Membership on an entity’s Board of Directors or advisory committees.

Daukėlaitė, Vaitekėnaitė, Čekauskienė, Bukauskas: nothing to declare.

7: New Drugs and Cell-based Immune Therapies

P570 EFFICACY OF SALVAGE THERAPY INCLUDING NOVEL AGENTS IN ADULT B-CELL ACUTE LYMPHOBLASTIC LEUKAEMIA (B-ALL) PROGRESSING AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: A REAL-WORLD UK STUDY

Alexandros Rampotas 1, Fotios Panitsas2, Maximillian Brodermann1, Sridhar Chaganti3, Bouziana Styliani4, Emma Nicholson5, Samanth Drummond6, Sharon Allen7, Andrew King7, Anne-Louise Latif6, Henry Crosland3, Daniele Avenoso4, Claire Roddie1

1University College London Hospital NHS Foundation Trust, London, United Kingdom, 2Laikon University Hospital, Athens, Greece, 3University Hospitals Birmingham NHS Foundation Trust, Birmingham, United Kingdom, 4King’s College London Hospital NHS Foundation Trust, London, United Kingdom, 5The Royal Marsden NHS Foundation Trust, London, United Kingdom, 6Queen Elizabeth University Hospital, Glascow, United Kingdom, 7Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom

Background: For adult B-ALL, relapse following allogenic stem-cell-transplantation (allo-SCT) confers a dismal prognosis. Historically, intensive salvage therapy was limited to Donor Lymphocyte Infusion (DLI) or second allo-SCT, but more recently, the emergence of novel targeted therapies such as Blinatumomab, Inotuzumab and CAR-T cells have broadened treatment options. This real-world, UK-wide study marks the first attempt to compare eficacy of post-allo-SCT salvage treatment options. Crucially, we explore the value of DLI and second allo-SCT, illustrating the complexity of treatment decision making in this space.

Methods: This retrospective analysis of 123 cases of adult B-ALL post-allo-SCT relapse included relapsed cases between 2010-2022 from 6 UK hospitals. Survival outcomes were estimated using Kaplan-Meier and compared across groups with log-rank tests. Landmark analysis at 2.5 months post-relapse was conducted to address immortal time bias. The landmark time was chosen empirically, taking into account time needed from relapse to DLI or CAR-T delivery. A comparison group with similar baseline characteristics as the CAR-T group was identified using propensity score matching, in order to assess the effect of CAR-T on OS and PFS. The project received approval from the British Society of Blood and Marrow Transplantation, and data handling strictly adhered to the principles of the Declaration of Helsinki.

Results: 92 of 123 patients had sufficient data to be included in the analysis (median follow-up, 24.8 months). 17 patients received CAR-T therapy: 4 received UCART19; 1 received dual-targeting CD19/CD22 CAR-T, and 12 received CD19 CAR-T (Kymriah; Obe-cel). 75 patients received non-CAR-T therapies (Table 1). 24 patients received DLI either as standalone treatment (n=7) or supplementary to another treatment option (n=17). The CAR-T group were younger, with higher-risk disease and a median of 2 prior lines of therapy. Median time from relapse to treatment was 2.8 and 0.32 months for CAR-T and for non-CAR-T, respectively. 18 patients underwent a second allogeneic transplant at a median of 5 months after relapse, consolidating response to salvage CAR-T in 7 cases. Median OS after second alloSCT was 23.6months with prior CAR-T, and 18months if not used as consolidation to CAR-T therapy (p=0.3). Landmark analysis at 2.5months post relapse for CAR-T group (N=17) vs DLI (N=19) vs Other therapies (N=25) demonstrated that both CAR-Ts and DLI have better outcomes over other therapies. Median OS: 28.5months(13-NE) vs 24.7months(6.7-36.3) vs 6.4months(2.8-10.8) and PFS: 14.2months(4.6-NE) vs 6.3months(2.3-19.6) vs 2.5months(1.2-9.4). CAR-Ts PFS was not significantly better than DLI (p=0.2), but were superior to other therapies (p=0.003). When CAR-T cell group was compared with non-CAR-T propensity score-matched group, CAR-T had significantly prolonged OS (median, 31 months(15-NE) vs 8.9 months(5.3-13.5), HR=0.17, p=0.005) and PFS (16.7 months(7-NE) vs 5 months(1-13), HR=0.33 p=0.008).

CAR-T group (N=17)

Non-CAR-T group (N=17) - following propensity matching

DLI group (N=24)

Non-CAR-T total group (N=75)

Treatment received at first relapse

UCART19 4/17 (23.5%)

CD19 CAR-T 12/17 (70.6%)

CD22 CAR-T 1/17 (5.9%)

Blinatumumab 5/17 (29.4%)

TKI 2/17 (11.7%)

Inotuzumab 5/17 (29.4%)

Intensive Chemotherapy (5/17 (29.4%)

Supplementary DLI to other therapy 17/24 (71%)

Blinatumumab 1/17 (6%)

TKI 11/17 (65%)

Inotuzumab 3/17 (18%)

Chemotherapy and TKI 1/17 (6%)

Non-intensive chemotherapy 1/17 (6%)

Standalone DLI 7/24 (29%)

Blinatumumab 11/75 (14.6%)

TKI 24/75 (32%)

Inotuzumab 12/75 (16%)

Intensive Chemotherapy 10/75 (13.3%)

Intensive Chemotherapy and TKI 3/75 (4%)

Non-intensive chemotherapy 9/75 (12%)

Palliative measures only (Intrathecal, radiotherapy) 5/75 (6.7%)

Second allogeneic transplant 1/75 (1.3%)

Age (years) (Median, IQR)

26 (20-42)

28 (21-46)

49 (44-53)

26 (20-42)

Philadelphia Chromosome mutation

2/17 (11.8%)

2/17 (11.8%)

15/24 (62.5%)

28/75 (37.3%)

MLL rearrangement

3/17 (17.6%)

2/17 (11.8%)

1/24 (4.1%)

5/71 (7%)

Risk group

Standard/Intermediate 2/16 (12.5%)

Standard/Intermediate 1/15 (6.7%)

High/very high risk 21/21 (100%)

Standard Intermediate 8/72 (11.1%)

High /Very high risk 14/16 (87.5%)

High /Very high risk 14/15 (93.3%)

Not known 3/24

High/ Very high risk 64/72 (88.9%)

Not known 1/16

Not known 2/17

Not known 3/72

ECOG

0 10/13 (76.9%)

0 9/15 (60%)

0 7/15 (46.7%)

0 29/54 (53.7%)

1 3/13 (23.1%)

1 6/15 (40%)

1 7/15 (46.7%)

1 22/54 (40.7%)

2 0/13

2 0/15

2 1/15 (6.7%)

2 3/54 (5.6%)

Not known 4/17

Not known 2/17

Not known 9/24

Not known 8/75

Number of prior lines of therapy (median, IQR)

2 (1-2)

2 (1-3)

1 (1-1)

1 (1-2)

MRD status at transplant

Negative 13/17 (76.6%)

Negative 9/17 (52.9%)

Negative 9/24 (37.5%)

Negative 30/75 (40%)

Positive 1/17 (5.9%)

Positive 5/17 (29.4%)

Positive 10/24 (41.7%)

Positive 17/75 (22.7%)

Unknown 3/17 (17.6%)

Unknown 3/17 (17.6%)

Unknown 5/24 (20.8%)

Unknown 28/75 (37.3%)

Time from relapse to treatment (months, IQR)

2.8 (2.5-3.8)

0.7 (0.3-0.9)

0.6 (0-1)

0.32 (0.1-1)

Median OS (N=

31 months (95% CI 15.5-NE)

11.9 months (95% CI 5.3-23)

27.2 months (95% CI 9.2-38.8)

7 months (95%CI 4.6-12.1)

Median PFS (N=

16.7 months (95% CI 7.1-NE)

5 months (95% CI 1.4-12.9)

6 months (95% CI 3.8-20.7)

3.9 months (95% CI 3-5)

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (69)

Conclusions: This study highlights the superior outcomes of CAR-T therapy for relapsed B-ALL post allo-SCT, supported by rigorous landmark analysis and propensity matching. The efficacy of 2nd allo-SCT is limited, unless utilized as post-CAR-T consolidation. DLI emerges as a potentially beneficial intervention and its impact should be explored in other datasets to confirm this finding. Encouragingly, our data underscore the importance of early CAR-T referral in relapsed adult B-ALL post-allo-SCT.

Disclosure: Alex Rampotas received conference fees by Gilead. Claire Roddie declares Honoraria: Gilead Sciences Consulting or Advisory Role: Autolus, Kite/Gilead, Bristol Myers Squibb/Celgene Speakers’ Bureau: Novartis Pharmaceuticals UK Ltd, Gilead Sciences Travel, Accommodations, Expenses: Gilead Sciences, Autolus. Anna-Louise Latif declares honoraria from Kite and Novartis. All other authors declare no conflicts of interest.

7: New Drugs and Cell-based Immune Therapies

P571 TREOSULFAN PLUS FLUDARABINE “REDUCED INTENSITY CONDITIONING” WITH POSTTRANSPLANT CYCLOPHOSPHAMIDE IN PATIENTS WITH ACUTE MYELOID LEUKEMIA OLDER THAN 65 YEARS

Vincenzo Federico 1,2, Rosella Matera1, Dalila Salvatore3, Filippo Antonio Canale4, Manuela Merla3, Daniela Valente3, Corine Contento1, Giulia Campagna1, Doriana Vaddinelli5, Annalisa Natale5, Davide Seripa1, Stella Santarone5, Tiziana Grassi2, Francesco Bagordo2, Nicola Di Renzo1, Massimo Martino4, Angelo Michele Carella3

1“Vito Fazzi” Hospital, Lecce, Italy, 2University of Salento, Lecce, Italy, 3Casa Sollievo della Sofferenza Hospital, San Giovanni Rotondo, Italy, 4CTMO Grande Ospedale Metropolitano “Bianchi-Melacrino. Morelli”, Reggio Calabria, Italy, 5“Civile” Hospital, Pescara, Italy

Background: Allogenic stem cell transplantation (allo-HSCT) is the only curative option for intermediate and high-risk adult acute myeloid leukemia (AML). The majority of patients (pts) older than 65y had been excluded from this potentially curative option for decreasing allo-HSCT tolerance, for toxicity related to myeloablative conditioning and for a low probability of finding a HLA-identical donor. We report here the outcome of AML pts with older than 65y who underwent unmanipulated haploidentical transplantation (aplo-HSC) received a reduced intensity conditioning with intravenous 10 g/m2 treosulfan daily, for 3 days plus 30 mg/m2 intravenous fludarabine daily for 5 days (T10F) and post-transplant cyclophosphamide (PT-CY) as prevention of gvhd, with encouraging results in terms of survival and relapse incidence, engraftment and graft-versus-host disase (GvHD) incidence.

Methods: Between Jun 2019 and June 2023, 30 consecutive adult pts aged ≥65y received an aplo-HSCT for AML in four Hematology Transplant Unit. Patients’ median age was 69y (range 65–74). Twenty-four pts (80%) and 6 (20%) experienced this in 1° complete remission (CR) and in ≥ 2° CR respectively. GvHD prophylaxis consisted of post-transplant cyclophosphamide (PT-CY), mycophenolate mofetile and ciclosporin. All patients received peripheral blood stem cells as sours of graft (PBSC) TAB1

Transplant characteristics

KPS ≥ 90%

30 (100%)

Cell dose CD34 + :

Mean ˣ 106/Kg (range)

6.2 (4.6-9.5)

Donor Kinship:

Brother/sister

8 (27%)

Child

10 (33%)

Parent

11 (37%)

Niece

1 (3%)

Time diagnosis to HAPLO-PT-CY months (range)

11 (5-77)

Extrahematological toxicity Grade3-4):

Nausea

8 (26%)

Vomit

6 (20%)

Diarrhea

5 (16%)

Mucositis

8 (26%)

Sepsis

8 (26%)

Invasive Micosys

2 (6%)

PT-Cy (GVHD prophylaxis):

PT-Cy; 50 mg/m2 day+3 + 4

14 (47%)

PT-Cy; 50 mg/m2 day+3 + 5

12 (40%)

PT-Cy; 40 mg/m2 day+3 + 4

4 (13%)

Results: The median time to neutrophil engraftment>0.5ˣ109/l and platelet>20ˣ109/l was 14 days (range 13-21) and 22 days (range 13-48) respectively. All pts except one (3%) achieved full engraftment at day 30. One (3%) patient in CR2 had autologous reconstitution at day 90. Among the 27 (90%) pts alive at day 100, chimerism was full donor in 25 pts (92%), one patient for mixed chimerism received donor lymphocyte infusion. The cumulative incidence of acute GvHD was 25% (grade I-II and III-IV was 10% and 15% respectively) and cumulative incidence chronic GvHD (evaluated in 27 patients) was 35% (grade I-II and III-IV was 20% and 15% respectively). After a median follow-up of 15 months (range 3-50); 18 (60%) pts are alive and sustained leukemia remission with minimal residual desease negativity and 12 (40%) pts died. Only 4 (13%) pts died of progressive disease, while 8 (26%) pts died from transplant related causes (4 pts die for infections, 3 pts from GvHD, and 1 pts from cachesia). The estimate day 100 and 1-year respectively overall survival were 76% and 65% respectively. The TRM was 17% and 26% at day 100 and 1-year respectively. The estimate day 100 and 1-year cumulative incidence of relapse were 4% and 13% respectively.

Conclusions: Our results confirm that reduced intensity conditioning with treosulfan plus fludarabine beafore Aplo-HCT with posttransplant cyclophosphamide should be considered in AML patients older than 65y in absence of related or unrelated matching donor. This reduced intensity conditioning (T10F) would seem to limit relapse and GvHD without increasing TRM. Multicentric and larger studies with similar platforms are needed to confirm our results.

Disclosure: No conflicts to declare.

7: New Drugs and Cell-based Immune Therapies

P572 ORAL IPTACOPAN MONOTHERAPY MAINTAINS EFFICACY AND SAFETY OVER 48 WEEKS IN COMPLEMENT INHIBITOR-NAÏVE PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA (PNH) IN THE PHASE III APPOINT-PNH TRIAL

Antonio M. Risitano 1,2, Bing Han3, Yasutaka Ueda4, Jaroslaw P. Maciejewski5, Rong Fu6, Li Zhang7, Austin Kulasekararaj8,9,10, Alexander Röth11, Lee Ping Chew12, Jun Ho Jang13,14, Lily Wong Lee Lee15, Jens Panse16,17, Eng-Soo Yap18, Luana Marano1,2, Flore Sicre de Fontbrune19,20, Chen Yang3, Hui Liu6, Roochi Trikha8,9,10, Navin Mahajan21, Tomasz Lawniczek21, Zhixin Wang22, Christine Thorburn23, Shujie Li22, Marion Dahlke21, Régis Peffault de Latour19,20

1AORN Moscati, Avellino, Italy, 2University of Naples Federico II, Naples, Italy, 3Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China, 4Osaka University Graduate School of Medicine, Suita, Japan, 5Taussig Cancer Institute, Cleveland Clinic, Cleveland, United States, 6Tianjin Medical University General Hospital, Tianjin, China, 7Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin, China, 8King’s College Hospital NHS, London, United Kingdom, 9National Institute for Health and Care Research and Wellcome King’s Research Facility, London, United Kingdom, 10King’s College London, London, United Kingdom, 11West German Cancer Center, University Hospital Essen, University Duisburg-Essen, Essen, Germany, 12Hospital Umum, Sarawak, Kuching, Malaysia, 13Samsung Medical Center, Seoul, Korea, Republic of, 14Sungkyunkwan University School of Medicine, Seoul, Korea, Republic of, 15Queen Elizabeth Hospital, Kota Kinabalu, Malaysia, 16University Hospital RWTH Aachen, Aachen, Germany, 17Center for Integrated Oncology (CIO), Aachen Bonn Cologne Düsseldorf, Germany, 18National University Cancer Institute, Singapore, Singapore, 19French Référence Center for Aplastic Anemia and Paroxysmal Nocturnal Hemoglobinuria, Paris, France, 20Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France, 21Novartis Pharma AG, Basel, Switzerland, 22China Novartis Institutes for BioMedical Research Co Ltd, Shanghai, China, 23Novartis Pharmaceuticals UK Limited, London, United Kingdom

Background: Iptacopan (first-in-class, oral factor B inhibitor) demonstrated efficacy and safety in the 24-week core treatment period of the single-arm, open-label, multicenter, Phase III APPOINT-PNH trial (NCT04820530) in complement inhibitor-naïve patients with hemolytic PNH. We report 48-week efficacy, safety and tolerability data from APPOINT-PNH (completed 18/04/2023).

Methods: Complement inhibitor-naïve adult PNH patients with mean hemoglobin <10 g/dL and lactate dehydrogenase (LDH) >1.5 × upper limit of normal (ULN) received iptacopan monotherapy 200 mg twice daily for 48 weeks (24-week treatment period and optional 24-week extension period).

Results: All 40 enrolled patients completed APPOINT-PNH. At Week 48, the percentage of patients achieving a hemoglobin increase from baseline ≥2 g/dL (irrespective of transfusions) was 97.4% (Table); this was maintained from Week 24 (94.9%). The percentage of patients achieving hemoglobin ≥12 g/dL (irrespective of transfusions) was 79.5% at Week 48, which was numerically higher than at Week 24 (66.7%). The mean hemoglobin level reached 12.56 (standard deviation [SD] 1.49) g/dL at Week 24 (mean change from baseline: +4.41 [SD 1.40] g/dL) and 13.24 (SD 1.80) g/dL at Week 48 (mean change from baseline: +5.09 [SD 2.01] g/dL), showing further improvement after Week 24. One patient required red blood cell transfusions between Weeks 2 and 48; the estimated proportion of patients achieving transfusion avoidance was 97.5% (95% confidence interval [CI] 92.5, 100.0). Fatigue improvements during the core treatment period were maintained across the extension period; mean change from baseline in Functional Assessment of Chronic Illness Therapy (FACIT) – Fatigue score was +12.1 (SD 9.93) at Week 24 and +10.4 (SD 10.14) at Week 48. LDH reductions were sustained across the extension period; at Week 48, median LDH level was 261.5 (interquartile range: 206.0–334.0) U/L and median LDH change from baseline was −1241.5 (interquartile range: −1795.0 to −925.0) U/L. The percentage of patients with LDH ≤1.5 × ULN was 95% at Week 24 and 85% at Week 48. At Week 48, the mean absolute reticulocyte count (ARC) was 79.51 (SD 42.60) × 109/L, demonstrating maintained improvement. In the extension period, two patients experienced a clinical breakthrough hemolysis (BTH) event; the adjusted annualized clinical BTH rate was 0.05 (95% CI 0.01, 0.17). Potential complement-amplifying conditions were identified for both patients; both BTH events resolved without iptacopan discontinuation (one during APPOINT-PNH, one after the trial ended). No patients experienced a major adverse vascular event. Throughout APPOINT-PNH, headache (30.0% of patients), COVID-19 (22.5%), upper respiratory tract infection (17.5%) and diarrhea (15.0%) were the most common treatment-emergent adverse events (TEAEs); eight patients (20.0%) experienced at least one serious TEAE. There were no treatment discontinuations or deaths.

Table: Summary of efficacy parameters at Week 24 and Week 48 in the APPOINT-PNH trial

Week 24

Week 48

Endpoints

Proportion of patients

Summary statistic

Proportion of patients

Summary statistic

n/M*

Percentage (%)

n/M*

Percentage (%)

Hemoglobin level increase from baseline of ≥2 g/dL

37/39

94.9

38/39

97.4

Hemoglobin of ≥12 g/dL

26/39

66.7

31/39

79.5

n/N

Percentage (%)

n/N

Percentage (%)

LDH ≤1.5 × ULN

38/40

95.0

34/40

85.0§

M/N

Change from baseline

M/N

Change from baseline

Mean (SD) change from baseline in hemoglobin level (g/dL)

39/40

+4.41 (1.40)

39/40

+5.09 (2.01)

Mean (SD) change from baseline** in FACIT-Fatigue score

37/40

+12.1 (9.93)

39/40

+10.4 (10.14)

Median (IQR) change from baseline†† in LDH level (U/L)

39/40

−1271.0 (−1830.0 to −920.0)

40/40

−1241.5 (−1795.0 to −925.0)§

Mean (SD) change from baseline‡‡ in ARC (109/L)

39/40

−87.00 (65.16)

39/40

−76.55 (50.15)

n/M*

Estimated proportion§§

(% [95% CI]) from Week 2 to Week 24

n/M*

Estimated proportion§§

(% [95% CI]) from Week 2 to Week 48

Transfusion avoidance

40/40

97.6 (92.5, 100)

39/40

97.5 (92.5, 100.0)

n/N¶¶

Adjusted annualized rate

(% [95% CI])

n/N¶¶

Adjusted annualized rate

(% [95% CI])

Rate of clinical BTH***

0/40

0 (0.00, 0.17)

2/40

0.05 (0.01, 0.17)

Rate of MAVEs

0/40

0 (0.00, 0.17)

0/40

0.00 (0.00, 0.09)

  1. *n=number of patients who met the criteria, M=number of patients with evaluable/non-missing data; Includes post-transfusion data; n=number of patients, N=total number of patients; §Variability in LDH level at Week 48 was mainly driven by one outlier; M=number of patients with evaluable/non-missing data, N=total number of patients; Mean (SD) baseline hemoglobin level was 8.16 (1.09) g/dL; **Mean (SD) baseline FACIT-Fatigue score was 32.8 (10.17); ††Median (IQR) baseline LDH level was 1581.5 (1144.5 to 2054.5) U/L; ‡‡Mean (SD) baseline ARC was 154.33 (63.67) × 109/L; §§Estimated proportions reflect the probability of meeting the endpoint criteria among the study population. Estimated proportions were computed using simple proportion, based on observed data, and the 95% CI was obtained using the bootstrap method; Patients not receiving or not requiring transfusions in the specified time period; ¶¶n=number of patients with at least one event, N=total number of patients; ***Events that met the protocol-specified criteria for clinical BTH ARC, absolute reticulocyte count; BTH, breakthrough hemolysis; CI, confidence interval; FACIT-Fatigue, Functional Assessment of Chronic Illness Therapy – Fatigue; IQR, interquartile range; LDH, lactate dehydrogenase; MAVE, major adverse vascular event; SD, standard deviation; ULN, upper limit of normal

Conclusions: Improvements from the 24-week core treatment period were sustained throughout the extension period to the end of APPOINT-PNH, demonstrated by most patients maintaining clinically meaningful hemoglobin increases, normal/near-normal hemoglobin levels, good intravascular hemolysis control, transfusion avoidance, and improvements in fatigue and ARC. Iptacopan was well tolerated, with a safety profile consistent with 24-week data. Together, results suggest oral iptacopan monotherapy could become a practice-changing, convenient outpatient treatment for hemolytic PNH patients.

Clinical Trial Registry: NCT04820530 (https://clinicaltrials.gov/study/NCT04820530).

Disclosure: Antonio Risitano reports consultancy, honoraria and research funding for F. Hoffmann-La Roche Ltd and Alexion, AstraZeneca Rare Disease; and consultancy and honoraria for Novartis. Régis Peffault de Latour reports consultancy, honoraria and/or research funding for Alexion, Amgen, F. Hoffman-La Roche Ltd, Merck Sharp and Dohme, Novartis Pharma AG, Pfizer, Samsung and Sobi.

7: New Drugs and Cell-based Immune Therapies

P573 T CELLS DIRECTED AGAINST THE METASTATIC DRIVER CHONDROMODULIN-1 IN EWING SARCOMA: COMPARATIVE ENGINEERING WITH CRISPR/CAS9 VS. RETROVIRAL GENE TRANSFER FOR ADOPTIVE TRANSFER

Uwe Thiel 1, Kristina von Heyking1, Busheng Xue1, Hendrik Gaßmann1, Melanie Thiede1, Kilian Schober2, Josef Mautner3, Julia Hauer4, Jürgen Ruland5, Dirk H. Busch6, Stefan E.G. Burdach7

1Technical Universtity Munich, Paediatric Haematology and Oncology Children’s Cancer Research Center, Munich, Germany, 2Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, 81674 Munich, Germany, Munich, Germany, 3Helmholtz Centre Munich, Munich, Germany, 4TUM School of Medicine, Children’s Hospital Munich Schwabing, Technical University of Munich, Munich, Germany, 5Institute of Chemistry and Pathobiochemistry, TUM School of Medicine, Technical University of Munich, Munich, Germany, 6Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, Munich, Germany, 7Translational Pediatric Cancer Research-Institute of Pathology, School of Medicine, Technical University of Munich, Munich, Germany

Background: We assess orthotopic T cell receptor replacement using CRISPR/Cas9 based non-viral TCR transfection in comparison to randomly inserting retroviral transduction against Ewing sarcoma (EwS) overexpressed CHM1 in a peptide/ HLA-A*02:01 restricted manner.

Methods: Firstly, we confirmed the feasibility of orthotopic replacement of the endogenous TCR by CRISPR/Cas9 with a TCR targeting our canonical metastatic driver chondromodulin-1 (CHM1). CRISPR/Cas9 engineered T cell products specifically recognized and killed HLA-A*02:01+ EwS cell lines. We then compared T cells engineered with either CRISPR/Cas9 or randomly inserting retroviral gene transfer.

Results: The efficiency of retroviral transduction was stronger compared to CRISPR/Cas9 gene editing. Both engineered T cell products specifically recognized tumor cells and elicited cytotoxicity against EwS. Transduction rates varied, however, in both technique, depending on donor source. Especially electroporation within CRISPR/Cas9 mediated orthotopic TCR replacement was associated with significant initial cell damage. Donor choice and less traumatic transduction techniques may be crucial to render non-viral CRISPR/Cas9 mediated transduction more robust. Despite lower transduction rates in the non-viral transduction setting, cytotoxicity was stronger and prolonged in contrast to retrovirally transduced T cells.

Conclusions: Non-viral CRISPR/Cas9 mediated orthotopic TCR transfection yields excellent preclinical in vitro EwS cell line specific recognition an lysis with long-lasting cytotoxicity compared to retroviral transduction with random TCR transduction. This technique may be more adequate for GMP conform tumor specific T cell generation for future immunotherapeutic clinical translation.

Disclosure: The authors declare no conflict of interest. Funding: We are grateful to the China Scholarship Council (#201908210290) and the Cura Placida Children’s Cancer Research Foundation (#06082021/#5110378) for their financial support to B.X., as well as to the St. Baldrick’s Foundation (Martha’s BEST Grant for all #663113) and the ‘Du musst kämpfen’ Wohltätigkeitsinitiative (Access to Systems Biology-Based Individualized Cell Therapies for Adolescents with Refractory Pediatric Cancer, AdoRe, #200310stb) for their grants to S.E.G.B. U.T. is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—Project number 501830041, the “Zukunft Gesundheit e.V.” and the Dr. Sepp and Hanne Sturm Memorial Foundation. U.T. and H.G. are funded by the Robert Pfleger Foundation and U.T., H.G., and S.E.G.B. by the Wilhelm Sander-Foundation (U.T. 2018.072.1. and 2021.007.1.; S.E.G.B. 2018.072.1). D.H.B. and K.S. received funding for the OTR development from SFB-TRR 338/1 2021—452881907 (project A01) and SFB 1054/3—210592381 (project B09). J.R. was supported by research grants from the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation-Project-ID 360372040—SFB 1335), and the European Research Council (ERC) under the European Union’s Horizon 2020 research and innovation programme (grant agreement number 834154). H.G. was also funded by the KKF program of TUM School of Medicine.

7: New Drugs and Cell-based Immune Therapies

P574 UTILITY OF ROMIPLOSTIM IN PAEDIATRIC ALLOGENEIC STEM CELL TRANSPLANT ASSOCIATED POOR GRAFT FUNCTION, IMMUNE MEDIATED CYTOPENIA AND IMMUNE MEDIATED GRAFT FAILURE

Srividhya Senthil 1, Abdul Moothedath2, Abbey Forster1, Ramya Hanasoge-Nataraj1, Madeleine Powys1,1, Omima Mustafa1,1, Robert Wynn1,1

1Royal Manchester Children’s Hospital, Manchester, United Kingdom, 2Royal Manchester Children’s Hoapital, Manchester, United Kingdom

Background: Poor graft function (PGF) and Immune related cytopenia (IMC) cause significant morbidity and mortality post allogeneic stem cell transplant (SCT). Conventional treatment of PGF and IMC are CD34 selected Stem-cell-boost (SCB) and Immunosuppressive treatment (IST) respectively. We have previously reported a relationship between IMC and graft failure (GF), and that IMC might presage GF, a forme fruste. There is lack of data on utility of romiplostim in paediatric SCT. We report our significant experience of romiplostim in PGF and IMC in paediatric allogeneic SCT. Of note, we highlight its probable utility in preventing GF.

Methods: This is a retrospective study of paediatric allogeneic SCT recipients who received romiplostim for treatment of cytopenia. The case records were analysed to get the patient, donor and transplant characteristics, aetiology of cytopenia and dose, duration and the response to romiplostim.

Results: The cohort included mainly non-malignant patients with only one JMML patient who was more than 2 years post SCT. The median age was 33 months (range 11-87 months). Of the 9 cases, 2 had ITP, 2 had PGF, 4 had IMC and 1 had Secondary GF. The contributory factors implicated in cytopenia aetiology, included alloreactivity (n=4), Adenoviremia (n=2), CMV (n=3), myelosuppressive drugs (n=5), low stem cell dose (n=1), ABO incompatibility (n=1) and gram-negative sepsis (n=1). Romiplostim was commenced from 70 to 774 days post-transplant at a weekly dose of either 2.5, 5, 10 mcg/kg depending on the depth of cytopenia and doses were escalated to a maximum of 10mcg/kg to achieve desired response. The Overall Response (OR) defined as achievement of platelet count >100*109/L, absolute neutrophil count >1*109/L and Hb>80g/L without transfusion support, was achieved in 8 of the 9 patients, giving an Overall Response Rate (ORR) of 89%. The median time to achieve OR was 42 days (range 21-83 days) and that to achieve platelet and neutrophil response were 42 and 39 days respectively. Failure of response was seen in 1 with secondary GF who underwent a second allogeneic SCT after 2 doses.

Full size table

.

Importantly, Romiplostim averted the need for a planned second allogeneic SCT in one and planned SCB in 2. It reduced the need for prolonged IST in 6 patients.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (70)

Conclusions: This is the largest paediatric SCT based study on romiplostim highlighting not only its role in PGF buts also in IMC and GF. Romiplostim is a safe, effective and cost-efficient alternative to SCB, second allogeneic transplants, prolonged IST and its ensuing complications in these settings. We recommend its promotion over other toxic approaches in these settings and a larger survey of its use and prospective studies with longer follow up.

Clinical Trial Registry: not applicable.

Disclosure: The authors report no conflict of interest.

7: New Drugs and Cell-based Immune Therapies

P575 RAG2>-/-ΓC-/- MICE HUMANIZED WITH CD34+ HEMATOPOIETIC STEM CELLS ARE A SUITABLE TOOL TO SCREEN FOR FUNCTIONAL EWING SARCOMA-SPECIFIC T CELLS IN VIVO

Uwe Thiel 1, Sabine Heim1, Hendrik Gaßmann1, Kristina von Heyking1, Sebastian J. Schober1, Melanie Thiede1, Markus Niemeyer2, Dirk Busch3, Ropert Oostendorp4, Irene Esposito5, Julia Hauer1, Günther HS Richter6

1TUM School of Medicine, Children’s Hospital Munich Schwabing, Technical University of Munich, Munich, Germany, 2Tumor- und Brust-Zentrum Ostschweiz, St. Gallen, Switzerland, 3Institute for Medical Microbiology, Immunology and Hygiene, School of Medicine, Technical University of Munich, Munich, Germany, 4Technical University of Munich, Munich, Germany, 5Institute of Pathology, Medical Faculty, Heinrich-Heine University and University Hospital of Duesseldorf, Düsseldorf, Germany, 6Charité Universitätsmedizin, Berlin, Germany

Background: In this study we implement a humanized mouse system to scan for allorestricted CD8 + T cells against Ewing’s sarcoma (EwS) associated antigens.

Methods: Rag2-/-γc-/- mice were irradiated (3.5 Gy) at birth and transplanted into the liver with 1.2x105 HLA-A*0201- human cord blood-derived hematopoietic stem cells. At the age of ≥ 6 weeks, mice were i.v. vaccinated with EwS peptides pulsed on HLA-A*0201+ dendritic cells (DC) or received either no humanization, no DC vaccination or DCs vaccination without peptide load. After three vaccinations with 2x106 DCs, all mice received 2x106 A673 EwS cells s.c. At day 17 mice were analyzed for lymphatic organs, tumor burden and antigen-specific allorestricted HLA-A*0201- CD8+T cells.

Results: Mice regularly showed human CD4 + /CD8 + /CD4 + CD8 + T cell reconstitution in thymi and T and B cell reconstitution in spleens. Two mice showed lymph nodes with presence of human T cells. 65% of humanized mice showed peripheral reconstitution with >10% human CD45+cells. Tumor size between study group mice vs. respective controls was significant (p <0.05). The degree of peripheral reconstitution was not associated with tumor size. EwS peptide multimer stained T cells were detected in lymph nodes of the only study group mouse without tumor burden. Respective T cells specifically recognized peptide-pulsed target cells and HLA-A2+/peptide+ EwS cells.

Conclusions: Immunization of humanized mice with peptide-antigen loaded DC induced an allorestricted T cell response capable to suppress EwS growth. This tool allows screening for antigens presented by different tumor and other disease entities to identify functional antigen-specific T cell receptors for future adoptive transfer.

Disclosure: The authors declare no conflict of interest. SJS and UT were funded by the Wilhelm Sander-Foundation (2018.072.1. and 2021.007.1.) and the Cura Placida Children’s Cancer Research Foundation (20210823). SJS and HG were supported by the KKF clinician-scientist program of the School of Medicine, Technical University of Munich, and furthermore SJS was funded by the BZKF Young Scientist Fellowship. UT is funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation)—Project number 501830041, the “Zukunft Gesundheit e.V.” and the Dr. Sepp and Hanne Sturm Memorial Foundation. UT and HG are funded by the Robert Pfleger Foundation.

7: New Drugs and Cell-based Immune Therapies

P576 REAL-WORLD EFFECTIVENES OF VENETOCLAX COMBINED WITH IPOMETILATING-AGENTS AS A BRIDGE TO ALLOGENEIC TRANSPLANTATION IN ACUTE MIELOID LEUKEMIA: RETROSPECTIVE AND MULTICENTRIC STUDY OF APULIAN HEMATOLOGICAL NETWORK “REP05”

Vincenzo Federico 1,2, Rosella Matera1, Domenico Pastore3, Giuseppe Tarantini4, Lorella Melillo5, Angelo Michele Carella6, Attilio Guarini7, Caterina Buquicchio4, Lucia Ciuffreda5, Mariachiara Abbenante6, Paola Carluccio8, Crescenza Pasciolla7, Mariapaolo Fina1, Alessandro Spina3, Marina Urbano3, Daniela Carlino1, Mariangela Lecciso1, Michelina Dargenio1, Davide Seripa1, Pellegrino Musto8,9, Giorgina Specchia10, Nicola Di Renzo1

1“Vito Fazzi” Hospital, Lecce, Italy, 2University of Salento, Lecce, Italy, 3“A. Perrino” Hospital, Brindisi, Italy, 4“Mons. Dimiccoli” Hospital, Barletta, Italy, 5“Riuniti” Hospital, Foggia, Italy, 6Casa Sollievo della sofferenza“ Hospital”, San Giovanni Rotondo, Italy, 7“IRCCS Oncologico” Hospital, Bari, Italy, 8“Policlinico” Hospital, Bari, Italy, 9University School of Medicine, Bari, Italy, 10Rete Ematologica Pugliese, Bari, Italy

Background: The combination of venetoclax (VEN) and hipometilatig agents (HMA) represents a practice-changing innovation in the treatment of acute myeloid leukemia (AML). Nowadays VEN and HMA agent are the standard of care for both relapsed/refractory (R/R) AML and for newly diagnosed (ND) AML patients (pts) unfit for intensive chemotherapy. The role of allo-HSCT in pts that improvement of their performance status after this combination is not still fully investigated. We report here the outcome of patients with ND or R/R-AML who underwent to allo-HSCT after VEN-HMA agent in a real life experience.

Methods: From May 2018 to December 2022, a total of 192 pts were treated with VEN-HMA within of Apulian Hematological Network. Fifty (26%), 24 (40%) ND- and 26 (60%) R/R-AML, median age 63 years (range: 23-74) after improvement of their performance status and obtaining complete remission underwent to allo-HSCT. Allo-HSCT was performed after a median of 4 cycles (range: 3-6) of VEN at median dose of 100 mg/daily (range: 100-400) with decitabine 20 mg/m2 days 1-5 of each 28-day cycle in 32 patients (64%) or azacitidine 75 mg/m2 days 1-7 of each 28-day cycle in 18 (36%) pts. In all cases graft source was mobilized PBSC; 34 pts (68%) received thiotepa, busulfan and fludarabine as mieloablative conditioning regimen and 16 pts (32%) fludarabine with treosulfan/busulfan as reduced intensity conditioning regimen. Twentynine pts (58%) received an haplo-donor and PT-CY 50 mg/Kg day +3 and +5 as GvHD prophylaxis combined with ciclosporin and micofenolate mofetile.

Results: The median number of CD34+blood cells infused was 6.8 ˣ 106/Kg (range: 4.2-8.2). The median time to neutrophil engraftment > 0.5 ˣ 109 /l and platelet > 20 ˣ 109 /l was 14 days (range: 13- 18) and 19 (range: 14-26) respectively. All but one of the patients achieved full engraftment. One patient with ND-AML had primary engraftment failure. The cumulative incidence of aGvHD was 26% and 13% and cGVHD (evaluated in 38 patients) was 35%, and 13% for grade I-II, III-IV respectively. After a median follow-up of 15 months (range: 3-58), 38 (76%) pts were alive in complete remission. Seven patients (14%) died of progressive disease, while 5 (10%) pts died from transplant related causes (3 pts die for infections, 2 pts from GvHD). At time to analysis the median overall survival it’s not achieved and the estimate 1-year and 2-years overall survival was 82% and 64% respectively. The estimate 1-year cumulative incidence of transplant related mortality and relapse were 17% and 14% respectively. No statistically significant difference in estimate median OS was observed when allo-HSCT was performed after VEN-HMA in ND-AML vs R/R AML (p=0.6).

Conclusions: Our results albeit obtained in a small series of patients shown how the VEN-HMA combination should be regarded as a good bridge to allo-HSCT both in ND-AML and R/R AML. Multicentric and larger studies with similar platforms are needed to confirm our results.

Disclosure: no conflicts of interest to declare.

7: New Drugs and Cell-based Immune Therapies

P577 T CELL RECEPTOR IDENTIFICATION AGAINST EWING SARCOMA ASSOCIATED DICKKOPF 2 (DKK2) DERIVED ANTIGEN USING SINGLE-CELL RNA SEQUENCING

Sophia Laube 1, Kristina von Heyking1, Melanie Thiede1, Hendrik Gassmann1, Sebastian Johannes Schober1, Jennifer Eck1, Hannah Hüls1, Carolin Prexler1, Sabrina Wagner2, Elvira D’Ippolito2, Julia Hauer1, Stefan E G. Burdach3, Dirk H. Busch2, Uwe Thiel1

1Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany, 2Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany, 3Institute of Pathology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany

Background: We explored Dickkopf 2 (DKK2), overexpressed in Ewing sarcoma (EwS), as a target for preclinical adoptive T cell approaches to pave the way for new therapies to treat advanced EwS.

Methods: In silico prediction and T2 HLA-A*02:01 binding assays were used to identify HLA-A*02:01 + /DKK2 derived antigens for priming of HLA-A*02:01- CD8 + T cells from healthy donors using peptide-pulsed HLA-A*02:01+ dendritic cells. T cells were selected with specific multimers and expanded to conduct functional tests. Clones with specific recognition of HLA-A*02:01 + /peptide+ EwS cell lines were analysed using polymerase chain reaction (PCR) and single-cell RNA sequencing (scRNAseq) to identify the DNA sequence of the T cell receptor (TCR).

Results: We identified a T cell clone and its respective TCR specifically targeting the selected DKK2 nonamer. The T cell clone recognised and attacked HLA-A*02:01+ EwS cell lines in Interferon-γ and Granzyme B ELISpot assays. Moreover, it lysed these cell lines dose-dependently in an xCELLigence assay. By comparing different methods to identify the specific TCR sequence, we were able to demonstrate that scRNAseq is clearly superior to the conventional method using PCR and gel electrophoresis. ScRNAseq could reliably and quickly identify the DNA sequence of the specific TCR, especially after prolonged cultivation of the T cells and with low cell numbers.

Conclusions: The herein identified DKK2 peptide is a promising target for adoptive immunotherapy in advanced EwS. ScRNAseq greatly facilitates TCR identification compared to PCR and gel electrophoresis. The identified TCR will be expressed in HLA-A*02:01 + T cells using orthotopic CRISPR/Cas9 or random retroviral transduction and subsequently investigated in further experiments in vitro and in vivo.

Disclosure: The authors declare no conflict of interest. UT was funded by the Wilhelm Sander-Foundation (2018.072.1. and 2021.007.1.) and the Cura Placida Children’s Cancer Research Foundation (20210823). UT is currently funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - Project number 501830041, the “Zukunft Gesundheit e.V.”, the Dr. Sepp and Hanne Sturm Memorial Foundation and the Robert Pfleger Foundation.

7: New Drugs and Cell-based Immune Therapies

P578 TARGETED CD8+ T CELL THERAPY IN ADVANCED EWING SARCOMA: EXPLORING LIPI, GPR64, PAX7, AND CHM1 AS TARGETS FOR ADOPTIVE TRANSFER APPROACHES

Hannah Hüls 1, Kristina von Heyking1, Melanie Thiede1, Hendrik Gassmann1, Sebastian Johannes Schober1, Jennifer Eck1, Sophia Laube1, Carolin Prexler1, Sabrina Wagner2, Elvira D’Ippolito2, Julia Hauer1, Stefan E G. Burdach3, Dirk H. Busch2, Uwe Thiel1

1Kinderklinik München Schwabing, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany, 2Institute of Medical Microbiology, Immunology and Hygiene, Technische Universität München, Munich, Germany, 3Institute of Pathology, Klinikum rechts der Isar, TUM School of Medicine and Health, Technical University of Munich, Munich, Germany

Background: To explore new options for therapies treating advanced Ewing Sarcoma (EwS), we examined preclinical T cell based adoptive immunotherapeutic approaches against EwS-associated antigens LIPI, GPR64, PAX7, and CHM1.

Methods: In silico derived nonamers of the respective peptides were evaluated in T2-HLA-A*02:01-binding assays. Selected peptides were pulsed with HLA-A*02:01+ dendritic cells to prime HLA-A*02:01- CD8 + T cells. Following a two-week co-cultivation period, proliferating T cells were multimer-stained, sorted, and subsequently expanded through a single-cell dilution process. The selected clone populations were evaluated for HLA-A*02:01/peptide specificity in ELISpot assays. Selected EwS-specific candidates were further expanded, and the DNA sequences of the T cell receptors (TCRs) were analysed using gel electrophoresis and polymerase chain reaction (PCR). Additionally, single-cell RNA sequencing (scRNAseq) was employed for a comprehensive analysis of T cells.

Results: LIPI antigen-pulsed T cells exhibited limited EwS-specific recognition, targeting only peptide-pulsed T2 cells as a positive control and failing to recognize EwS cell lines. PAX7 and GPR64 specific T cells demonstrated the ability to target EwS cell lines in vitro, although a complete TCR sequence could not be identified. Both CHM1 and GPR64 specific T cell receptors confirmed EwS specificity and were re-examined via scRNAseq. In the case of CHM1, a new TCR was successfully identified using both PCR and scRNAseq.

Conclusions: PAX7, GPR64, and CHM1 emerge as promising targets for adoptive immunotherapy in advanced EwS. PAX7 is also overexpressed in synovial sarcoma and alveolar rhabdomyosarcoma, and GPR64 in nephroblastoma, rendering these antigens particularly interesting putative T cell targets. The identified CHM1 TCR sequence will be further investigated in additional in vitro experiments and a mouse model. Future research will focus on revisiting PAX7 and GPR64, with an emphasis on scRNAseq, as this technique simplifies TCR identification, particularly after extended cell cultivation—a challenge encountered, particularly with PAX7 cells. The findings underscore the potential of targeted T cell therapy in addressing advanced EwS but emphasise the need for continued optimization of T cell manufacturing.

Disclosure: The authors declare no conflict of interest. UT was funded by the Wilhelm Sander-Foundation (2018.072.1. and 2021.007.1.) and the Cura Placida Children’s Cancer Research Foundation (20210823). UT is currently funded by the Deutsche Forschungsgemeinschaft (DFG, German Research Foundation) - Project number 501830041, the “Zukunft Gesundheit e.V.”, the Dr. Sepp and Hanne Sturm Memorial Foundation and the Robert Pfleger Foundation.

7: New Drugs and Cell-based Immune Therapies

P579 COMPARATIVE EFFECTIVENESS OF IPTACOPAN VERSUS C5 INHIBITORS IN COMPLEMENT INHIBITOR-NAÏVE PATIENTS WITH PAROXYSMAL NOCTURNAL HEMOGLOBINURIA FROM THE PHASE III APPOINT-PNH TRIAL AND REAL-WORLD APPEX COHORT

Matthew Holt 1, Richard J. Kelly1, Jilles M. Fermont2, Georgina Bermann2, Ariel Chernofsky2, Jens Haenig2, Marion Dahlke2, Régis Peffault de Latour3,4

1St James’s University Hospital, Leeds, United Kingdom, 2Novartis Pharma AG, Basel, Switzerland, 3French Référence Center for Aplastic Anemia and Paroxysmal Nocturnal Hemoglobinuria, Paris, France, 4Assistance Publique Hôpitaux de Paris, Université Paris Cité, Paris, France

Background: Paroxysmal nocturnal hemoglobinuria (PNH) is a rare disease characterized by hemolysis and consequent anemia, bone marrow failure and thrombosis. C5 inhibitors (C5i) eculizumab and ravulizumab are approved treatments for PNH that prevent intravascular hemolysis. However, up to 82% and 39% of patients treated with the aforementioned C5i remain anemic and transfusion dependent, respectively. Results from the single-arm Phase III APPOINT-PNH trial (NCT04820530) investigating monotherapy with the oral factor B inhibitor iptacopan in adult complement inhibitor-naïve patients demonstrated clinically significant hemoglobin improvements in the absence of red blood cell transfusions (RBCTs). However, there are no data comparing the hematological response of iptacopan with C5i in this population. We estimated the comparative effectiveness of iptacopan versus C5i in adult C5i-naïve PNH patients.

Methods: This retrospective study included patients in APPOINT-PNH who received iptacopan monotherapy and the real-world APPEX cohort who received routine C5i treatment at PNH reference hospitals in France and the United Kingdom (NCT05842486). Hematological response was defined as the proportion of patients who would have achieved a ≥2 g/dL hemoglobin increase from baseline in the absence of RBCTs; hemoglobin ≥12 g/dL in the absence of RBCTs; transfusion avoidance and change from baseline lactate dehydrogenase levels and reticulocyte count. The analysis was adjusted for confounding using a propensity score to construct a weighted prediction of treatment outcomes that would have been observed had APPOINT-PNH participants received C5i. Estimated differences between treatments were derived using the orthogonalized score form of the efficient influence function and cross-fitting. Confidence bounds for differences accounting for multiple imputations in APPOINT-PNH were obtained using Rubin’s combination rules.

Results: Baseline measurements were available for 85/92 patients from APPEX. The overall analytical cohort comprised 125 patients (APPOINT-PNH, n=40; APPEX, n=85). Baseline covariates between APPOINT-PNH and APPEX were well balanced, with all standardized mean differences within ±0.10 after weighting (Figure). Differences in the proportion of patients (95% CI) achieving a ≥2 g/dL hemoglobin increase, hemoglobin levels ≥12 g/dL and transfusion independence were 68.2 (40.9, 95.6), 53.4 (31.4, 75.3) and 38.8 (15.1, 62.5), respectively, all in favor of iptacopan versus C5i. The ratio of percent change from baseline in lactate dehydrogenase levels was 0.51 (0.40, 0.67), and the difference in change from baseline in reticulocyte count was −75.5 (−106.9, −44.2), both in favor of iptacopan (Table).

Table. Comparative effectiveness of iptacopan and C5i for hematological endpoints in APPOINT-PNH and APPEX

Endpoint

Estimate

Difference in treatment effect

(iptacopan versus C5i)

Response as increase from baseline in hemoglobin ≥2 g/dL* in the absence of RBCTs

Difference in proportions, % (95% CI)

68.2 (40.9, 95.6)§,

Response as having hemoglobin levels ≥12 g/dL* in the absence of RBCTs

Difference in proportions, % (95% CI)

53.4 (31.4, 75.3)§,¶

Transfusion avoidance

Difference in proportions, % (95% CI)

38.8 (15.1, 62.5)§,**

Percent change from baseline in lactate dehydrogenase levels

Ratio of percent levels to baseline (95% CI)

0.51 (0.40, 0.67)§,††

Change from baseline in reticulocyte count‡‡

Difference in change from baseline, 109/L (95% CI)

−75.5 (−106.9, −44.2)§,§§

  1. *Endpoint for C5i included measurements between Day 100 and Day 200 (mean of all available measurements); Endpoint for C5i included measurements between Day 15 and Day 200. Transfusion avoidance was assessed between Day 14 and Day 168 in APPOINT-PNH; Derived using the orthogonalized score form of the efficient influence function and cross-fitting; §In favor of iptacopan; The estimated proportion (95% CI) in APPOINT-PNH was 92.2% (82.5, 100). The weighted estimate (95% CI) in APPEX was 27.9% (15.0, 42.0); The estimated proportion (95% CI) in APPOINT-PNH was 62.8% (47.5, 77.5). The weighted estimate (95% CI) in APPEX was 11.5% (3.7, 21.1); **The estimated proportion in APPOINT-PNH was 97.6% (92.5, 100). The estimated proportion in APPEX was 59.0% (53.7, 73.3); ††The adjusted mean (95% CI) percentage change from baseline in lactate dehydrogenase levels was −83.55% (−84.90, −82.08) in APPOINT-PNH. The estimated mean (95% CI) percentage change from baseline in lactate dehydrogenase levels was −70.7% (−75.1, −66.1) in APPEX. Therefore, the mean percentage of baseline lactate dehydrogenase levels was approximately 16% for iptacopan and approximately 29% for C5i, corresponding to a ratio slightly over 50% in favor of iptacopan; ‡‡Endpoint for C5i included measurements between Day 1 and Day 200; §§The mean (95% CI) change from baseline in reticulocyte count was −82.48 × 109/L (−89.33, −75.62) in APPOINT-PNH. The estimated mean (95% CI) change from baseline in reticulocyte count was −2.5 (−21.2, 13.7) in APPEX C5i, C5 inhibitor; CI, confidence interval; RBCT, red blood cell transfusion

Conclusions: This analysis provides information on hematological endpoints on C5i treatment, showing C5i leads to hematological improvements but to a much lesser extent than iptacopan monotherapy did in APPOINT-PNH. Limitations of APPEX include the irregular frequency of measurements in real-world clinical practice, which differs from the scheduled measurement regime in APPOINT-PNH. There was no adjustment for regional differences, with a larger proportion of Asian patients enrolled in APPOINT-PNH compared with APPEX, in which patients resided in Europe. However, ethnicity does not influence the pharmacokinetics of iptacopan or C5i; no differences in efficacy or safety have been reported for C5i in Asian patients. The results indicate that C5i-naïve patients with PNH may experience greater improvements in hematological response with iptacopan compared with C5i.

Clinical Trial Registry: NCT05842486 (APPEX; https://clinicaltrials.gov/study/NCT05842486) and NCT04820530 (APPOINT-PNH; https://classic.clinicaltrials.gov/ct2/show/NCT04820530).

Disclosure: Matthew Holt reports research funding from Sobi and support with congress travel from Alexion. Richard J Kelly reports consultancy, research funding, speaker fees and/or advisory board attendance for Alexion, Florio, Novartis Pharma AG, Otsuka, Sobi and Roche. Jilles M Fermont, Ariel Chernofsky, Jens Haenig and Marion Dahlke are employees of and equity holders in Novartis Pharma AG. Georgina Bermann is an equity holder and was an employee of Novartis Pharma AG when the study was performed. Régis Peffault de Latour reports consultancy, honoraria and/or research funding for Alexion, Amgen, F. Hoffman-La Roche, Merck Sharp and Dohme, Novartis Pharma AG, Pfizer, Samsung and Sobi.

7: New Drugs and Cell-based Immune Therapies

P580 TREATMENT WITH BLAST-MODULATORY RESPONSE MODIFIERS INDUCED SPECIFIC IMMUNE RESPONSES AND LED TO DISEASE STABILIZATION IN A PATIENT WITH REFRACTORY AML AFTER SECOND ALLOGENEIC SCT

Philipp Anand1, Giuliano Fillipini Velazquez 2, Joudi Abdulmajid1, Xiaojia Feng1, Klaus Hirschbühl2, Anwesha Sinha1, Helga Schmetzer1, Christoph Schmid2

1Ludwig-Maximilian-University, Muinch, Germany, 2Augsburg University Hospital and Medical Faculty, Augsburg, Germany

Background: Novel treatment strategies are urgently needed for the treatment of relapsed/refractory AML. The combination of the blast-modulating response modifiers GM-CSF and prostaglandin (PG)E1 (‘Kit-M’) has been shown to convert myeloid leukaemic blasts ex vivo into dendritic cells of leukaemic origin (DCleu), presenting individual patients` leukaemic antigens in a costimulatory context to immune cells. Innate and adaptive immune cells are specifically directed against blasts and develop memory cells. Similar effects could be shown in a rat leukaemia model.

Methods: A 65-year-old patient with secondary AML (FLT3-ITD + /BCR/ABL1 + ), had received multiple lines of therapy including conventional chemotherapy, various TKI, a matched unrelated and a haploidentical allogeneic stem cell transplantation (SCT). Early after the second SCT he had developed hematologic and extramedullary (pleural effusions) relapse, that was refractory to 4 cycles of salvage treatment with azacitidine. Detection of HLA loss precluded treatment with donor lymphocyte infusions.

After extensive discussion with the patient and obtaining written informed consent and permittance by the health care provider, we initiated an individual salvage therapy with Kit-M (PGE-1 given i.v. over 2 hours q12 hours, GM-CSF as continuous infusion) Treatment was divided into two phases, the first being a ramp-up phase consisting of 4 weeks of treatment with increasing doses of PGE-1 (week1: 20µg d1-3, 40µg d4-5; week 2: 40µg d1-5; week 3: 40µg d1-5; week 4: 80µg d1-5). GM-CSF dose was 75µg/m2 throughout the entire treatment. After the ramp-up phase, treatment was continued at the highest PGE-1 dose in a one-week on, one week-off schedule. Overall treatment duration was 4 months.

Results: KIT-M treatment was well tolerated in all PGE-1 doses. No serious adverse events, and in particular no signs of stimulation of the leukemia by GM-CSF were observed. The most important clinical finding was the stabilisation of peripheral blast counts and extramedullary manifestation of the leukaemia over a period of 4 months without further antileukaemic tretament. In addition, Kit-M treatment led to a transient reduction in the frequency of RBC transfusions (25% less transfusions) as compared to the prior treatment line. After 4 months of therapy the patient developed progressive leukaemia and finally died.

As compared to the status before Kit-M treatment, immune-monitoring of the patient’s blood showed increased (mature) DCleu, downregulated expression of checkpoint markers (CTLA4, PD1) on blasts and T-cells and induction of (potentially leukaemia-specific) immune cells: IFNy-producing and degranulating (e.g. non-naïve, memory T-cells, NK-cells and downregulated regulatory T-cells). An increase and stabilisation of immunostimulatory effects were observed during treatment, whereas a slight decrease was measured during treatment interruptions. After 4 months, we observed a decrease in (potentially leukaemia-specific) immune cell (non-naïve, memory T-cells), while others remained at elevated levels (degranulating T-cells, NK-cells).

Conclusions: Kit M treatment of a highly therapy/SCT refractory patient was well tolerated, induced specific immune responses and led to disease stabilisation over four months without further systemic therapy.

Disclosure: Modiblast Pharma GmbH (Oberhaching, Germany) holds the European Patent 15 801 987.7-1118 and US Patent 15-517627 ‘Use of immunomodulatory effective compositions for the immunotherapeutic treatment of patients suffering from myeloid leukemias’, with whom H.M.S. is involved with

8: Non-haematopoietic Stem Cells and Regenerative Medicine

P581 REDUCED PRESENCE OF MESENCHYMAL STEM CELLS IN BONE MARROW ASPIRATES OF FEMALE PATIENTS PERSISTS AFTER ALLOGENEIC STEM CELL TRANSPLANTATION

Judith Schaffrath 1, Ole Vollstädt2, Jana Lützkendorf1, Cornelia Baum1, Sabine Edemir1, Kirstin Lauer1, Lutz P. Müller1

1Martin-Luther-University Halle-Wittenberg, Halle (Saale), Germany, 2Sana HANSE-Klinikum, Wismar, Germany

Background: Mesenchymal stem cells (MSC) are an essential component of the bone marrow (BM) niche, which is highly relevant for maintaining the hematopoietic stem cell population and controlling their differentiation. Therefore, the fibroblast-like cells within the BM stroma play a crucial role in the regulation of both healthy hematopoiesis as well as the development and maintenance of hematopoietic neoplasms. After allogeneic stem cell transplantation (alloSCT), MSC originate from the recipient and a damaged niche contributes to poor graft function after alloSCT.

For in vitro analyses, human BM-MSCs are mainly derived from plastic adherent BM mononuclear cells (MNCs) as colony-forming unit–fibroblast (CFU-F) and are defined according to consensus criteria.

Methods: In a retrospective single center analysis, we analyzed MSC presence in BM aspirates from patients who had undergone a routine BM biopsy during the course of their treatment. BM MNCs were isolated by density gradient separation and subsequently cultivated to obtain total MSC. Results were correlated with the clinical data of the patients, including diagnosis, treatment and outcome. Both univariate analysis and multiple regression were performed.

Results: We analyzed a total of 1292 BM biopsies from 737 patients with a median age of 61 years (range 15 – 91) which were obtained between 2003 and 2021, including patients with various hematological malignancies and nonmalignant diseases as well as healthy individuals. Patients were either treatment naïve (n=297; 23%) or had already received different treatments, including chemotherapy (75.0 %), radiation therapy (33.4 %), autologous (autoSCT, 7.0 %) and / or allogeneic SCT (42.0 %).

The multivariate analysis of all samples showed a significantly reduced presence of MSCs in patients with female sex (p<0.001), older age (> 60 y, p=0.005), after radiation therapy (p<0.001) and after autoSCT (p<0.001). Patient-based multivariate analysis of one biopsy per patient (the last sample obtained) additionally showed a significant correlation between previous alloSCT and reduced presence of MSCs (p=0.02). Although not significant, there was also a difference in CFU-F count in samples with CFU-F presence, with a lower number of CFU-Fs in samples from female patients.

Considering only therapy-naïve patients and thus excluding influence of treatment, female sex remained a significant factor for reduced presence of MSCs (p=0.033). This difference was detectable in both younger and older patients (≤ and > 60 years) and remained noticeable after chemotherapy, radiation, autoSCT and alloSCT. This significant difference in samples from female patients after alloSCT persisted in multivariate analysis.

Conclusions: In our large cohort, there was a significantly reduced presence of MSCs in bone marrow aspirates of female patients. Given the gender differences in the incidence and outcome of myeloid neoplasms and the importance of the bone marrow niche for allogeneic stem cell engraftment, further studies should follow to confirm these differences and explore the underlying biology.

Disclosure: Nothing to declare.

8: Non-haematopoietic Stem Cells and Regenerative Medicine

P582 EFFICACY AND SAFETY OF A CORD BLOOD-DERIVED PLATELET CONCENTRATE FOR THE TREATMENT OF OCULAR SURFACE DISEASES: RESULTS FROM A SINGLE-CENTER PROSPECTIVE STUDY

Camilla Delponte1, Laura Mazzucco2, Maria Rosa Astori3, Lucia Brunello3, Sara Butera3, Paolo Rivela3, Valeria Balbo2, Davide Dealberti4, Riccardo Dondolin5, Monia Lunghi5, Francesca Pollis2, Marco Ladetto3, Francesco Zallio 3

1Università degli Studi del Piemonte Orientale “Amedeo Avogadro” (UPO), Alessandria, Italy, 2Transfusion and Regeneration Medicine, SS Antonio & Biagio and C. Arrigo Hospital, Alessandria, Italy, 3SS Antonio & Biagio and C. Arrigo Hospital, Alessandria, Italy, 4SS Antonio & Biagio and C. Arrigo Hospital, Alesandria, Italy, 5Division of Hematology, AOU Maggiore della Carità, Novara, Italy

Background: Ocular surface diseases, such as chronic Graft Versus Host Disease (cGVHD) and Sjögren Syndrome, cause severe patient distress and have an adverse impact on long-term quality of life.

As conventional therapies have proven unsatisfactory, several ongoing studies are aiming at investigating the potential therapeutic effect of topical blood-derived products including umbilical cord blood serum (CBS).

The rationale of using CBS instead of autologous platelet lysate is based on higher concentrations of growth factors and anti-inflammatory effects, suggesting greater efficacy in managing chronic dry-eye inflammation.

We conducted a single center prospective study to assess the efficacy and safety of a lysed platelet concentrate derived from CBS in patients suffering from ocular cGVHD after allogenic transplant and Sjögren Syndrome.

Primary objectives were 1)the evaluation of change in corneal damage in patients with moderate/severe keratoconjunctivitis (via Oxford score) and 2)the evaluation of improvement in the extent of damage to the corneal epithelium and post-treatment lacrimation (through Schirmer test and BUT test). Secondary objective is the evaluation of change in dry-eye symptoms, pain and photophobia affecting ADLs (via OSDI score and VAS.).

Methods: Twenty-five cGVHD and nine Sjögren syndrome patients with moderate/severe keratoconjunctivitis were enrolled in the trial (CPCB19 v.1.0 - 08.04.2019).

The treatment lasted 4 months with a dosage of one drop per eye 3-6 times a day.

Upon enrollment and at 1-2-4 months of treatment, we assessed both parameters of ocular surface and symptoms.

Results: Thirty-one of 34 patients completed the study. After 120 days of treatment, comparing the VAS score, we registered substantial improvements, especially in patients’ quality of life: at T0 the mean value was 6.53 (range 2-10), while at T4 it was 2.4 (range 0-8). The OSDI score is consistent with these results, going from a mean value of 68.9 at T0 to 25.63 at T4.

Slighter objective differences were measured: Schirmer test registered a mean value of 3.94 at T0 and 4.6 at T4; S-BUT went from a mean value of 5.11 at T0 to 8.42 at T4.

Moreover, Oxford scheme detected an objective improvement in the corneal damage: all patients enrolled had at least a grade 2 at T0, while, at T4, most of them reached a grade 0 and only 5 had a grade 1.

Focusing on the 25 patients with cGVHD, at T0, according to NIH score, 4 had a score 1 ocular cGVHD, 20 had a score2, and 1 had a score 3; at T4, 2 had a grade 0, 19 had a score 1, and 2 had a score 2 (2 patients didn’t complete the study). Therefore, 78.26% registered an improvement of the NIH score and 8.7% reached a score 0.

At T0 10 patients were under systemic immunosuppression, while at T4 four patients were still under calcineurin-inhibitors, 4 under Ruxolitinib but 6 stopped corticosteroids.

Conclusions: Our results suggest that heterologous CBS-based eye drops represent a promising therapeutic approach in the healing of severe dry-eye in keratoconjuntivitis sicca and patients’ subjective symptoms relief. These are worthwhile of investigation in a multicenter study.

Clinical Trial Registry: CPCB19 v.1.0 - 08.04.2019.

Disclosure: Nothing to declare.

8: Non-haematopoietic Stem Cells and Regenerative Medicine

P583 IMPACT OF DIMENSIONALITY AND EXTRACELLULAR MATRIX ON MESENCHYMAL STEM CELL PHENOTYPE

Jeong Jun Kim1, Ji Hye Lee1, Hee Hoon Yoon1, Jun Eun Park 2

1HLB Cell Co Ltd, Hwasung, Korea, Republic of, 2Korea University Medical School, Seoul, Korea, Republic of

Background: Umbilical cord blood-derived mesenchymal stem cells (UCB-MSCs) possess self-renewal, multi-differentiation, and tissue regenerative capabilities, making them a promising candidate for various disease treatments. However, the role of the extracellular matrix (ECM) in the microenvironment remains poorly understood. In this study, we aimed to assess the impact of the ECM on phenotypic changes based on cell dimensionality and ECM types.

Methods: To analyze phenotypic differences, UCB-MSCs were cultured in low attachment microwells for 24 hours to form spheroids. These spheroids were then embedded in either no ECM, type I collagen, or Matrigel, and cultured for 48 hours with or without IFN-gamma. The harvested cells were compared to those of the monolayer.

Results: Depending on cell dimensionality and types of ECM, UCB-MSCs exhibited significant variations in the expression of PD-L1, HLA-DR, and IDO, indicating distinct phenotypes.

Conclusions: The phenotype of UCB-MSCs is believed to be strongly influenced by filament rearrangement and antigenicity depending on cellular dimensionality and ECM type.

Disclosure: No conflict of interest in this abstract.

14: Non-infectious Early Complications

P584 PROTECTING HLA-A AND HLA-B ANTIGENS THROUGH EPLETS ANALYSIS IN THE SETTING OF PLATELET TRANSFUSIONS IN HAPLOIDENTICAL HEMATOPOIETIC STEM CELL ALLOGRAFT CANDIDATES

Magalie Joris1, Amandine Charbonnier1, Delphine Lebon1, Judith Desoutter1, Etienne Paubelle1, Nicolas Guillaume 1

1University Medical Centre, Amiens, France

Background: HLA haploidentical and mismatched donors introduce the complication of donor-specific HLA antibodies (DSA) in Hematopoïetic Stem Cell Transplantation (HSCT). Platelet transfusion is the main risk factor for the prevalence of class I anti-HLA antibodies in haploidentical allograft candidates, providing evidence for guiding the assessment of anti-HLA antibodies to select HLA-mismatched transplant candidates. In this study, we aimed to extend the opportunity of selecting identified haploidentical stem cell donors for HSCT and avoid the occurrence of DSA before the graft. For this purpose, we wanted to know whether a restrictive list of protective class I HLA antigens deduced by eplets (epitopes) carried by potential haploidentical donors can be a feasible strategy for finding a sufficient number of platelet components (PCs) while preserving the chances that the recipient can be transfused in a timely manner.

Methods: We selected 121 recipients, candidates for HSCT. For each recipient, we identified haploidentical siblings and relatives by HLA genotyping. Of these 121 recipients, 114 donor/recipient pairings could be analyzed for class I HLA-A, -B, and –C alleles for the eplet mismatch survey. For each pair, recipient HLA-A, -B, -C and sibling/relative HLA-A, -B, high resolution typing was entered in the HLA Eplet Mismatch calculator on the HLA Eplet Registry (https://www.epregistry.com.br/). The calculator returned a list of confirmed mismatched HLA eplets in recipient versus donor direction. We selected “confirmed” antibody-verified eplets to their exposition level (intermediate and high) attributable to their location on the HLA molecule. Then, we defined a list of HLA antigens sharing eplets carried by these donors, becoming “unacceptable antigens” for upcoming platelet transfusions. Using the virtual Panel Reactive antibody (vPRA) calculator on https://www.etrl.org/hoofdform.aspx, the Eurotransplant Reference laboratory website, we defined the theoretical availability of single-donor HLA-matched platelets by determining the percentage of platelet donors expected to have one or more unacceptable antigens in our European population. Moreover, we determined the actual availability of single-donor HLA-matched platelets in the database of our regional transfusion center (EFS, Etablissement Français du Sang).

Results: The average number of mismatched eplets was 5.8 (± 3.3). From these eplets, we defined the number of HLA antigens carrying them. The mean involved HLA-A and -B antigens was 38.5 (± 2) (range from 0 to 22 HLA-A antigens and from 0 to 49 HLA-B antigens). The vPRA was 80 ± 2.29% (75.25 to 84.32) (i.e. mean ± standard error of the mean). In other words, our vPRA values range from 9.6% to 99.9% which means that, at the eplet level, between 90.4% to 0.1% of PCs are theoretically available for our recipient. The increase of vPRA is inversely proportional to the number of available PCs in real life. We defined a threshold value of vPRA at 94.9% that would predict the real availability of 10 PCs. With this strategy, in 68% of cases in our cohort, the PCs selection for transfusion seemed feasible.

Conclusions: Epitopic protection of HLA molecules is possible in real life, but proves to be very restrictive in terms of choice of PC.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P585 CLINICAL SIGNIFICANCE OF CYTOKINE RELEASE SYNDROME FOLLOWING HLA-MISMATCHED HEMATOPOIETIC STEM CELL TRANSPLANTATION: A RETROSPECTIVE MONOCENTRIC STUDY ON 250 TRANSPLANTS

Filippo Frioni 1, Eugenio Galli2, Sabrina Giammarco2, Elisabetta Metafuni2, Federica Sorà2, Maria Assunta Limongiello2, Roberto Maggi1, John Donald Marra1, Luca Di Marino1, Andrea Mattozzi1, Andrea Bacigalupo1, Simona Sica2, Patrizia Chiusolo2

1Università Cattolica del Sacro Cuore, Rome, Italy, 2Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy

Background: Cytokine release syndrome (CRS) has been described after allogeneic hematopoietic stem cell transplantation (HSCT), mostly after haploidentical HSCT, in T-cell repleted transplants, after peripheral blood HSCT, and in post-transplant Cyclophosphamide (ptCy) based platforms: we focused on the clinical significance of CRS following all HLA-mismatched transplants, regardless of stem cell source and GvHD prophylaxis scheme.

Methods: The identification of CRS was conducted retrospectively on patients receiving allogeneic HSCT from an haploidentical or single mismatch unrelated donor, experiencing a microbiologically negative fever after graft infusion. CRS was defined and graded basing on the revised American Society for Transplantation and Cellular Therapy criteria. Cox regression proportional hazard was used to assess the impact of CRS on clinical outcomes.

Results: 250 patients were included, 173 (69%) patients receiving haploidentical and 77 (31%) single mismatch HSCT. The median age was 61 years (range 19-75), most common diagnosis were AML (117 patients, 47%), MDS (44 patients, 18%), Myelofibrosis (43 patients, 17%), and ALL (33 patients, 13%). A myeloablative conditioning regimen was administered to 107 (43%) patients, while 143 (57%) patients underwent a reduced intensity conditioning regimen. ptCy based GvHD prophylaxis was administered in 203 patients (81%). Stem cell source was bone marrow in 125 patients (50%) and peripheral blood in the other 50% of patients; 127 (51%) patients experienced acute Graft versus Host Disease (GvHD) of any grade and 177 (71%) patients a chronic GvHD. CRS was present in 117 patients (47%); more commonly of grade 1 (81% of all CRS). CRS occurred more frequently after haploidentical HSCT (p=0.037, HR 1.19), and among all haploidentical transplants more frequently after peripheral blood graft (p=0.014 HR 1.79); considering only single mismatch unrelated donor, we found no difference according to stem cell source (p 0.5). When applying ptCy based GvHD prophylaxis, the totality of CRS fully resolved after the second administration. CRS occurrence correlates with lower 2-years non-relapse mortality (NRM) (p 0.037): 1-year NRM was 17% in CRS patients versus 26% in non-CRS patients while the 2-years NRM was 18% versus 31% in CRS and non-CRS patients respectively. The cumulative incidence of 2-year NRM was lower in CRS patients (17%) versus non-CRS patients (29%, p 0.044). CRS also correlates with longer relapse free survival (RFS) (p <0.001): 1-year RFS 88% versus 73% and 2-years RFS 85% versus 66% in CRS and non-CRS patients respectively. The 2-year cumulative incidence of relapse was higher in non-CRS patients (18%) versus CRS patients (10%), despite not reaching statistical significance (p 0.08). When looking at acute and chronic GvHD occurrence we found no difference between the two groups (p 0.104 and 0.752 for acute and chronic GvHD occurrence).

Conclusions: Mismatch-driven CRS correlates with longer RFS, with lower NRM after allogeneic HCST, and does not increase acute or chronic GvHD occurrence. We explained the lower NRM with the faster engraftment reached after peripheral blood graft. We also think that ptCy can explain that CRS (starting before ptCy administration and fully resolved immediately after) does not correlate with higher incidence of GvHD.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P586 INVESTIGATING VASCULAR ENDOTHELIAL DYSFUNCTION BY SYNDECAN-1 FOLLOWING PEDIATRIC HSCT

Sarah Weischendorff 1, Denise Elbæk Horan1, Katrine Kielsen1, Maria Ebbesen Sørum1, Marianne Ifversen1, Pär Ingemar Johansson1, Klaus Mûller1,2

1Rigshopitalet, Copenhagen University Hospital, Copenhagen OE, Denmark, 2University of Copenhagen, Copenhagen, Denmark

Background: Allogeneic HSCT is often complicated by acute non-infectious toxicities including sinusoidal obstruction syndrome (SOS), engraftment syndrome (ES) and capillary leak syndrome (CLS), all of which are believed to arise from endothelial dysfunction. However, our understanding of the pathophysiological mechanisms remains incomplete, and diagnoses continue to rely on overlapping clinical criteria. Biochemical evaluation of endothelial damage might help identify pathogenetically homogenous entities and enable early risk assessment.

Syndecan-1 is expressed on endothelial cells as part of the glycocalyx, important for vascular health. Elevated circulating syndecan-1 reflects endothelial glycocalyx degradation, which is associated with increased vascular permeability, unregulated vasodilation, and microthrombosis, offering plausible pathophysiological explanations for the clinical manifestations. We aimed to investigate the kinetics of plasma syndecan-1 and its relation to treatment-related complications during pediatric HSCT.

Methods: We prospectively included 113 children (1-18 years) undergoing myeloablative HSCT for malignant (n=69) or benign (n=44) diseases. Donors were HLA-matched sibling donors (n=33) or matched unrelated donors (n=80). Stem cell source was mainly bone marrow (n=108) and conditioning was based on TBI (n=27), busulfan (n=50) or other chemotherapy regimens (n=36).

Patients were screened for endothelial dysfunction-related complications including SOS (pediatric EBMT criteria), ES (Spitzer criteria) and CLS (weight gain above 3% from baseline combined with a positive fluid balance despite diuretic administration). Plasma syndecan-1 levels were measured weekly by ELISA during the early phase of HSCT. Data were analyzed using mixed model repeated measures analyses for time and group comparisons and Spearman’s rank-order correlation.

Results: During HSCT, plasma syndecan-1 significantly increased from a median level of 71 (IQR 49-124) ng/mL before conditioning, peaking at 131 (91-178) ng/mL on graft infusion day. Levels remained elevated for the first four weeks post-HSCT (all p<0.0001), gradually decreasing to baseline on week 13.

Thirty-two patients fulfilled the criteria for severe SOS (defined as grade 3-4), whereas CLS and ES were diagnosed in 15 and 13 patients, respectively.

Patients with severe SOS had consistently higher post-HSCT syndecan-1 levels than patients with no/mild SOS (Figure 1), and this association remained significant after adjustment for age, sex, and busulfan-based conditioning.

No differences in syndecan-1 levels were observed for patients with CLS or ES compared to those without, but patients with >5% weight gain (n=41) had significantly elevated syndecan-1 levels on days +7 (123 ng/mL vs. 97 ng/mL, p=0.041) and +14 (110 vs. 89, p=0.032). Additionally, syndecan-1 levels on days +7 and +14 correlated positively with the cumulated days of diuretic treatment within the first three months post-HSCT (rs=0.25, p=0.017 on both days), and with time to platelet recovery ≥50x109 cells/L (rs=0.25-0.39, p<0.01).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (71)

Conclusions: A sustained increase in syndecan-1 was observed in patients with severe SOS, while syndecan-1 levels were unrelated to CLS and ES, as based on clinical criteria. However, a weight-gain >5% during the early post-transplant period was associated with increased syndecan-1 levels, as was need for diuretic treatment and prolonged thrombocytopenia, possibly reflecting increased vascular permeability and increased platelet consumption. If further validated, syndecan-1 holds promise as a biomarker for endothelial dysfunction-related complications, guiding diagnosis and treatment interventions.

Clinical Trial Registry: NA.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P587 ASSESSING THE IMPACT OF FLUID OVERLOAD ON OUTCOMES AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Ana Belen Bocanegra 1, David Jaimovich2, Irene Solano1, Javier Martinez-Costa1, Jorge Verdejo1, Maria Valdenebro1, Carlos De Miguel1, Guiomar Bautista1, Sangeeta Hingorani3, Rafael Francisco Duarte1

1Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Spain, 2Favaloro University Hospital, Buenos Aires, Argentina, 3University of Washington/Division of Nephrology Seattle Children’s Hospital, Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, United States

Background: Fluid overload (FO) is common after allogeneic HCT and may associate with various other organ and transplant complications. Although largely unexplored as a hazard in HCT, a group has recently reported that FO may be an independent risk factor for patient outcomes in this setting (https://doi.org/10.1016/j.bbmt.2017.08.021.).

Methods: We conducted a retrospective study of consecutive allogeneic HCT recipients from 2009 to 2022 at our institution to determine the prevalence of FO, according to the criteria defined by Rondon et al, and its association with patient outcomes. We documented maximal grade of FO between date of admission and day +30 or patient discharge, whichever occurred first, with particular focus on FO beyond grades 0 and 1 (2: symptomatic fluid retention requiring ongoing diuretic therapy, weight gain ≥10% to <20%; 3: weight gain ≥20% unresponsive to diuretic therapy and possible organ dysfunction; 4: progressive dysfunction of more than one organ system or requiring intensive care).

Results: A total of 313 allo-HCT patients were included: 181 men (58%); median age 49 years (16-71); 61% acute leukemia, 19% chronic lymphoid and 16% chronic myeloid malignancies, 4% other indications; 39% matched siblings, 38% cord blood, 18% haploidentical and 14% unrelated donors; 51% myeloablative conditioning). One hundred and twenty-five patients (40%) had FO as defined above: 74 grade 1 (24%) and 51 grade ≥2 (16%). In keeping with weight increase, grade ≥2 FO cases had a median +1512 mL (236-3800 mL) positive fluid balance. Grade ≥2 FO was more common in HCT from alternative donors (p<0.001), in patients who develop acute kidney injury (29% grade ≥2; p<0.001), SOS/VOD (13%; p<0.001) or TA-TMA (11%; p=0.001), and those requiring total parenteral nutrition (29%; p=0.007) or admission to ICU (12%; p<0.001), but did not associate with other patient and HCT factors such as sex, diagnosis, comorbidity score, conditioning intensity, baseline pre-HCT creatinine, or acute GVHD (data not shown). Patients with grade ≥2 FO had significantly lower overall survival (OS) than those with no FO or grade 1, both early at day +100 (61% vs 93%; p<0.001) and later at one year after HCT (43% vs 69%; p<0.001), respectively (Figure 1). Multivariate analysis showed that the impact of FO on OS was independent from other complications and factors associated with FO and with transplant outcome (Table 1).

Table 1: Multivariate Analysis (Cox Regression) of Factors with an Impact on Overall Survival (OS) at Days + 100 and + 365 after Allogeneic HCT

OS at Day +100

OS at Day +365

OR

95% CI

P

OR

95% CI

P

Fluid Overload (grade ≥ 2)

0,304

0,153 - 0,605

0,001

0,547

0,343 - 0,873

0,011

Acute Kidney Injury (grade ≥2)

0,162

0,072 - 0,364

<0,001

0,420

0,277 - 0,636

<0,001

Age (≥49 years old)

0,435

0,219 – 0,865

0,018

0,623

0,423 – 0,917

0,017

Thrombotic Microangiopathy

-

-

-

0,506

0,310 – 0,823

0,006

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (72)

Conclusions: FO ≥ grade 2 is relatively frequent in the first 30 days during the initial admission for allogeneic HCT, and associates with lower OS early at day +100 and at one year. Although FO associates with other patient and transplant characteristics and complications, the impact on FO on OS is independent from these other concomitant factors. These findings warrant further investigation, including potential early intervention, on FO as a risk factor in allogeneic HCT.

Disclosure: No interest to declare.

14: Non-infectious Early Complications

P588 BEYOND PAIN MANAGEMENT: ASSESSING THE NEGATIVE IMPACT OF INTENSE OPIOID USE ON ALLOHCT SURVIVAL

Tommy Alfaro Moya 1, Igor Novitzky Basso1, Shiyi Chen1, Mats Remberger2, Refik Saskin3, Jonas Mattsson1

1Princess Margaret Cancer Centre, Toronto, Canada, 2Uppsala University Hospital, Uppsala, Sweden, 3Institute for Clinical Evaluative Sciences, Toronto, Canada

Background: Hematological malignancies and allogeneic stem cell transplantation (alloHCT) are conditions that are commonly associated with pain for which opioids are commonly prescribed. We hypothesized that opioids negatively affect the outcomes of patients undergoing alloHCT for hematological malignancies.

Methods: We merged our local transplant database with the anonymized pharmacy fill records from the Institute for Clinical Evaluative Sciences (ICES, Toronto, ON). We analyzed the outcomes of 681 patients who underwent alloHCT at Princess Margaret Cancer Centre from January 2010 to December 2019. The cohort comprised 51 intense opioid users (IOU) with an opioid prescription for >30 days within 1 year post alloHCT, and 630 control patients. To mitigate survivorship bias, the survival analysis start point in the non-IOU cohort was the median opioid initiation time in the IOU group, 95 days.

Results: The two-year overall survival (OS) was 53% (95% CI 49.2-57%) for non-IOU and 29.4% (19.2-45%) for IOU (HR 1.77, 1.26-2.48, p<0.001), with relapse being the primary cause of death in the IOU group and infection in the non-IOU group. Median survival was 1055 days (95% CI 712-1738) for non-IOU and 208 days for the IOU (95% CI 151-415). The non-relapse mortality (NRM) at 1 year was 37.3% (95% CI 25.9%-53.5%) for IOU and 27.5% (95% CI 24.2%-31.2%) for non-IOU groups (p not significant), and IOU was independently associated with higher NRM, along with age, donor type, and Hematopoietic cell transplant specific comorbidity index (HCT-C) scores. After 2 years, 31.4% (20.8-47.4) of IOU patients experienced relapse, and 16.4% (13.7-19.5) of the non-IOU relapsed, p=0.005, other factors such as disease stage, conditioning regimen, cytogenetics, and Disease Risk Index (DRI) were significant predictors. Relapse-free survival (RFS) analysis indicated that IOU was associated with a higher risk of events (death or relapse) in both univariate and multivariate analyses, along with factors like age, donor type, cytogenetics, and HCT-CI scores. In terms of acute graft versus host disease (aGVHD), and chronic graft versus host disease (cGVHD), IOU did not significantly impact the risk in multivariate analyses. However, high HCT-CI scores were associated with an increased risk of aGVHD, and donor type significantly influenced cGVHD risk. Graft versus host disease relapse-free survival (GRFS) analysis showed that IOU was associated with a higher risk of adverse events in the univariate analysis, but this association was not maintained in the multivariate analysis. Donor type and DRI were identified as significant predictors of adverse events in GRFS.

Conclusions: This study, the largest of its kind, reveals a negative association between intense opioid use and overall survival following alloHCT. The prolonged use of opioids was also linked to worse NRM and increased relapse rates. While causality cannot be definitively established due to the study’s retrospective nature, the findings raise concerns about the potential impact of opioids on immune function and disease control in alloHCT recipients. The results suggest a need for re-evaluating pain management strategies in alloHCT patients, considering alternative approaches to reduce toxicity while ensuring effective pain control.

Disclosure: The authors have no conflicts of interest to disclose.

14: Non-infectious Early Complications

P589 VENO-OCCLUSIVE DISEASE/SINUSOIDAL OBSTRUCTION SYNDROME (VOD/SOS)-ASSOCIATED MORBIDITY IN CHILDREN POST HEMATOPOIETIC STEM CELL TRANSPLANTATION - A HINT FOR AN ADEQUATE PROPHYLAXIS?

Zofia Szmit 1, Zuzanna Gamrot1, Anna Król1, Jowita Frączkiewicz1, Monika Mielcarek-Siedziuk1, Karolina Liszka1, Igor Olejnik1, Tomasz Jarmoliński1, Selim Corbacioglu2, Krzysztof Kałwak1

1Wroclaw Medical University, Wrocław, Poland, 2Medical University in Regensburg, Regensburg, Germany

Background: The introduction of sensitive criteria for early diagnosis of veno-occlusive disease/sinusoidal obstruction syndrome (VOD/SOS) such as the pediatric EBMT criteria made preemptive intervention for VOD/SOS possible and led to a significant improvement of overall VOD/SOS-related survival of children post-HSCT. However, the data on the impact of such early intervention on VOD/SOS-related morbidity is scarce.

Methods: Retrospective chart analysis included all 551 pediatric patients who underwent (allo/auto) HSCT in the Department of Pediatric Hematology, Oncology, and BMT in Wrocław, Poland, between November 2015 and December 2022. In the study cohort, 43 (8%) patients developed VOD/SOS post-HSCT. Using propensity score matching with the nearest neighbor method, we selected 43 patients serving as a control group. To assess VOD/SOS-related morbidity, the analysis included the timing of hematopoietic recovery, the occurrence of bleeding/thrombotic episodes, the use of blood products and human albumin, the incidence of acute kidney injury, respiratory disorder, and the need for supportive care during the first 60 days post-HSCT.

Results: The median number of weight-adjusted transfused blood products, including platelets (PLT), red blood cells (RBC), and plasma (FFP) was significantly higher in patients with VOD/SOS compared to the control group [PLT 13 vs 5; RBC 5 vs 2; FFP 0 vs 0; p<0.001]. Patients with VOD/SOS had relevantly higher demand for human albumin replacement (median 60 days cumulative dose/kg: 2.2g vs 0g; p<0.001) and antithrombin III concentrate (ATIII, median weight-adjusted doses: 5 vs 0; p<0.001). The platelet recovery was significantly prolonged in children with VOD/SOS (median: 47.5 days vs 16 days; p<0.001). In the VOD/SOS group, six patients developed hepatorenal syndrome, compared to none in the control group. Similarly, among VOD/SOS patients, six presented with bleeding/thrombotic episodes, 9 patients developed acute renal impairment, and 3 cases needed admission to the ICU compared to zero such cases in the control group. Patients who suffered from VOD/SOS had significantly higher INR levels on day 30 post-transplant in comparison to the control group and lower ATIII levels (INR 1.2 vs 1.09; p<0.001; ATIII 88 vs 102; p<0,001).

The overall survival and transplant-related mortality were comparable in both groups (OS 0.85); however, the length of hospitalization was relevantly longer among VOD/SOS patients (36 days vs 29 days; p<0.001).

Conclusions: Recent advances in VOD/SOS diagnosis and treatment have improved the survival of VOD/SOS patients, similar to OS in the overall HSCT cohort. However, the VOD/SOS-related morbidity remains a challenge. Delayed hematopoietic recovery in VOD/SOS patients makes them more susceptible to opportunistic infections. The use of blood products, human albumin, risk for developing life-threatening conditions such as severe bleeding episodes, acute kidney failure, or respiratory failure is significantly higher in VOD/SOS patients. Along with prolonged hospitalization, VOD/SOS not only exacerbates post-HSCT recovery but is also a noticeable economic burden. Despite preemptive treatment of VOD with defibrotide, the abovementioned results may justify the need for adequate VOD/SOS prevention. Well-defined VOD/SOS prophylaxis guidelines are strongly recommended, particularly in high-risk pediatric HSCT recipients.

Disclosure: Selim Corbacioglu is an Advisory Board Member of Jazz Pharmaceuticals.

Other authors have nothing to declare.

14: Non-infectious Early Complications

P590 OUTCOME AND COMPLEMENT SYSTEM INVOLVEMENT IN PATIENTS WITH THROMBOTIC MICROANGIOPATHY AFTER ALLOGENEIC STEM CELL TRANSPLANTATION

Jacopo Mariotti 1, Massimo Cugno2, Luigi Porcaro3, Daniela Taurino1, Barbara Sarina1, Chiara de philippis1, Daniele Mannina1, Armando Santoro1, Gianluigi Ardissino4, Stefania Bramanti1

1IRCCS Humanitas Research Hospital, Rozzano (milan), Italy, 2Center for HUS Prevention, Control and Management at Pediatric Nephrology, Dialysis and Transplant Unit, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy, 3Medical Genetics Laboratory, Fondazione IRCCS Ca’ Granda Ospedale Maggiore Policlinico, Milan, Italy, 4Università degli Studi di Milano, Milan, Italy

Background: Transplant-associated thrombotic microangiopathy (TA-TMA) is a rare, but often lethal complication, occurring after allogeneic hematopoietic stem cell transplantation (allo-SCT). Incidence of TA-TMA has a huge variability due to the use of several different diagnostic criteria by hematologic centers. The EBMT and IBMDR have recently published a consensus to harmonize the diagnosis of TA-TMA (Schoettler et al, 2023). The panel adopted a modified version of the Jodele criteria (developed in a pediatric population; Jodele et al 2015) and established that TA-TMA is diagnosed if ≥ 4 of 7 features (comprising: anemia, thrombocytopenia, hypertension, increase of LDH, schistocytes, soluble C5b-9, proteinuria) are present. These new criteria aim to facilitate early identification of TA-TMA and underline the role of complement activation for the diagnosis and prognosis; however, very limited results have been published in the adult setting.

The main objective of the study was to analyze EBMT criteria in a group of adult patients with suspected TA-TMA to identify those at higher risk of mortality. The other objective was to study whether abnormalities in complement system and in genes encoding for its proteins were more frequent in patients with confirmed TA-TMA.

Methods: Sixteen out of 80 adult patients receiving an allo-SCT for a hematologic malignancy at our Institution had a suspected diagnosis of post-transplant TA-TMA. These 16 patients were evaluated for ADAMTS13, soluble C5b-9 (sC5b-9) complex and complement genetic studies on blood samples obtained at time of TA-TMA suspicion. ADAMTS13 activity plasma levels were measured with a fluorescence resonance energy transfer assay. SC5b-9 plasma levels were determined by ELISA. Complement genetic studies were performed by next-generation sequencing and multiplex ligation–dependent probe amplification.

Results: Considering the 16 patients with suspected TA-TMA, one was excluded because of ADAMTS13<3%, thus affected with thrombotic thrombocytopenic purpura. Patients’ characteristics are summarized in Table I. Median of post-transplant days for suspecting TA-TMA was 80 days (IQ range 57-95 days). Five of 15 patients had <4 criteria, therefore TA-TMA was ruled out. In this subgroup of 5 patients, one (20%) had a mutation of CFHR3 encoding gene, none had signs of complement activation and one (20%) subsequently died for non-relapse causes. Ten patients had 4 or more EBMT criteria: five had 4 criteria, 2 had 5, 2 had 6 and one had 7. This group of 10 patients had signs of complement activation, i.e increased levels of sC5b-9 (median 527 ng/ml, IQ range 323-821 ng/ml [normal range 139-463 ng/ml]) and 5 (50%) were found to have mutations of genes involved in the regulation of complement cascade. Six out of these 10 patients (60%) died of non-relapse causes, and in particular, of the 5 with very high levels of sC5b-9 (median 1287 ng/ml, IQ 696-2193 ng/ml) 4 died.

Conclusions: Our preliminary data show that adoption of EBMT criteria in post-transplant patients with suspected TA-TMA successfully identifies a subset of patients at higher risk of mortality. Complement activation, together with abnormalities in genes encoding for complement regulatory proteins are more commonly found in these patients.

Clinical Trial Registry: Funded by the European Union - Next Generation EU - NRRP M6C2 - Investment 2.1 Enhancement and strengthening of biomedical research in the NHS - Project code PNRR-MAD-2022-12376816.

Disclosure: The authors have no conflict of interest to disclose.

14: Non-infectious Early Complications

P591 BONE REMODELLING ARE SIGNIFICANTLY IMPAIRED AFTER PEDIATRIC HSCT AND INFLUENCED BY DIAGNOSIS, CONDITIONING REGIMEN AND ACUTE GRAFT-VERSUS-HOST DISEASE

Katrine Kielsen 1, Kathrine Fogelstrøm1, Anne Nissen1, Marianne Ifversen1, Bolette Hartmann2, Klaus Müller1,2

1Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark, 2University of Copenhagen, Copenhagen, Denmark

Background: The successful outcome of pediatric HSCT is challenged by multiple late effects, including increased risk of bone loss with a reported prevalence of osteoporosis and osteopenia in 21% and 26% of children and adolescent at 5 years post-HSCT. This indicates that patients undergoing pediatric HSCT are at risk of developing osteoporosis much earlier than expected as part of normal age-related bone loss. However, the mechanisms behind this early occurring loss of bone mass after HSCT is incompletely understood.

C-terminal telopeptide of type I collagen (CTX) and N-terminal propeptide of type I procollagen (P1 NP) are the recommended biomarkers for assessment of osteoclast and osteoblast activity, respectively, in clinical studies. In this study, we investigated changes in bone resorption and bone formation by measuring CTX and P1 NP during pediatric allogeneic HSCT.

Methods: We prospectively included 102 patients undergoing HSCT from 2015-2022 in Denmark. Median recipient age was 9.5 years (1.2-17.9), and 61 were boys. Diagnoses included acute leukemias (n=41), other malignancies (n=22), PID (n=14) and other benign disorders (n=25). Conditioning regimens were myeloablative and based on TBI (n=22) or high-dose chemotherapy alone (n=80). Donors were either MSD (n=29) or MUD (n=73), and grafts were either BM (n=97) or PB (n=5).

Plasma levels of CTX and P1 NP were measured by automated chemiluminescence immunoassay before conditioning, at day of HSCT before graft infusion, and at day +7, +14, +21, +30, +60, +90, +180 and +365 post-HSCT.

Results: Pre-conditioning levels of P1 NP were reduced compared to age- and gender-matched healthy controls (corresponding to 33.9% of median reference value), while CTX were within the normal level [1]. During the first month of HSCT, CTX tended to increase and peaked at day +21 (2.2 vs. 1.7 ng/mL, P=0.0056). This pattern appeared to be mirrored by P1 NP, which decreased drastically from day 0 (149 vs. 407 ng/mL, P<0.0001) to day +30 compared with pre-conditioning levels (all P<0.05).

We next addressed if transplant-related factors influence bone remodeling markers early post-HSCT and found a tendency to reduced CTX and P1 NP associated with malignant diagnosis, TBI, a high toxicity conditioning regimen, aGvHD grade II-IV, treatment with steroids, and a high level of systemic inflammation (by C-reactive protein) (Table). Especially aGvHD was also associated with a significantly reduced ratio of P1 NP/CTX from day +14 to +30 (all P<0.05, Figure), which was further enhanced in patients treated with high doses of steroids.

From day +60 to +365 post-HSCT, CTX levels normalized, while P1 NP increased significantly at day +180 and +365 compared to pre-conditioning levels (day 180: 407 vs. 638 ng/mL, P<0.0001).

CTX levels [ng/ml]

day +21 (median)

P-value

P1 NP levels [ng/ml]

day +21 (median)

P-value

Diagnosis

(Malignant vs. benign)

2.0 vs. 2.7

0.0012

165 vs. 320

0.0014

TBI

(Yes vs. No)

1.6 vs. 2.4

0.021

165 vs. 261

0.16

Conditioning regimen

(High vs. reduced toxicity)

1.9 vs. 2.5

0.0019

206 vs. 333

0.0005

Donor type

(MSD vs. MUD)

2.5 vs. 2.1

0.73

313 vs. 272

0.87

Acute GvHD

(Grade 0-I vs. II-IV)

2.2 vs. 2.3

0.93

273 vs. 127

0.0009

Treatment with steroids

(Yes vs. No)

2.1 vs. 2.5

0.17

106 vs. 373

0.0015

Maximum CRP within 1 month post-HSCT

(<20 mg/dL vs. 20-60 mg/dL vs. >60 mg/dL)

2.0 vs. 2.2 vs. 1.7

0.0078

366 vs. 369 vs. 194

<0.0001

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (73)

Conclusions: Children undergoing HSCT have significantly reduced bone formation prior to conditioning compared to healthy controls. During the first month post-HSCT, the bone formation decreased even further, while bone resorption increased, indicating a significantly impaired bone remodeling. Several factors, such as conditioning regimens with high toxicity and aGvHD seemed to increase the risk of bone loss. Taken together, impaired bone remodeling before and in the early toxic phase after HSCT may be important contributing causes for early occurring osteoporosis after pediatric HSCT.

Clinical Trial Registry: NA.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P592 IDIOPATHIC UPPER GASTROINTESTINAL BLEEDING AFTER ALLOGENEIC STEM CELL TRANSPLANTATION: AN UNPRECEDENTED COMPLICATION

Juan Eirís 1, Marina Pérez-Bravo2, Nuria Rausell1, Carla Satorres1, Marta Villalba1, Pedro Chorão1, Juan Montoro1,3, Pedro Asensi1, Pablo Granados1, David Martínez-Campuzano1, Alberto Louro1, Marta Henriques4, Ana Facal5, Mª Consejo Ortí-Verdet6, Miguel Sanz1, Javier de la Rubia1,3, Jaime Sanz1, Aitana Balaguer-Roselló1

1Hospital Universitari i Politècnic La Fe, Valencia, Spain, 2Hospital Arnau de Vilanova, Valencia, Spain, 3Universidad Católica de Valencia, Valencia, Spain, 4Hospital de São João, Porto, Portugal, 5Hospital General de Castellón, Castellón, Spain, 6Hospital Clínico Universitario de Valencia, Valencia, Spain

Background: Bleeding complications are common events after allogeneic stem cell transplantation (alloHSCT), mostly due to moderate to severe thrombocytopenia, drug toxicity or infections.

We aimed to describe the incidence, characteristics and outcomes of patients developing idiopathic upper gastrointestinal bleeding (IGIB) after alloHSCT with a post-transplant cyclophosphamide (PT-Cy)-based graft-versus-host disease (GvHD) prophylaxis and to identify related risk factors.

Methods: All consecutive adult patients who underwent an alloHSCT in our institution with PT-Cy were included. GvHD prophylaxis regimens also included mycophenolate mofetil (MMF) and either sirolimus or tacrolimus. Upper gastrointestinal bleeding was defined as any clinically significant episode of hematemesis and/or melena, graded 2 or more following the NCI-CTCAE. Clinical characteristics, gastroscopic and histologic findings, management, and outcomes of patients developing upper gastrointestinal bleeding were retrospectively analyzed. Bleeding episodes were classified as IGIB if no other causes were detected.

Results: A total of 526 patients received an alloHSCT with PT-Cy from January 2017 to September 2023. Most of the patients received thiotepa, busulfan and fludarabine (TBF) as conditioning regimen (413, 78%) and a PT-Cy, sirolimus and MMF GvHD prophylaxis (481, 91%). Median follow-up of surviving patients was 28 months (3-80). Thirty-five upper gastrointestinal bleeding events were identified in 34 patients (6.3%). Recognized causes were GvHD (n=1), cytomegalovirus disease (n=1), disseminated adenoviral disease (n=1) and severe thrombocytopenia before myeloid engraftment (n=4). Patients with no identifiable cause (n=28) were considered to have IGIB; their clinical characteristics, management and outcomes are described in Table 1. Typical macroscopic and microscopic findings are displayed in Figure 1.

Patients who developed IGIB were older (median age 59 versus 53 years, p = 0.014), and no other statistically significant differences were detected in baseline patient, donor, and transplant characteristics. Cumulative incidence of IGIB was 5.3% (confidence interval 95%, 3.4-7) at 5 months, occurring at a median of 64 days (range 23-125) from alloHSCT. All patients except one required hospitalization, and another one died due to IGIB. Every patient was taking sirolimus at IGIB diagnosis. No differences in 2-year overall survival (65 vs 69%, p=0.3), 1-year transplant-related mortality (15% vs 13%, p=0.9) and 1-year disease free survival (64% vs 81%, p=0.2) cumulative incidence were observed when comparing IGIB with non-IGIB patients.

Risk factors for IGIB were age 60 years (p=0.013), history of alcohol abuse (p=0.014), diagnosis of myelofibrosis or myelomonocytic chronic leukemia (p=0.003), and advanced disease state (p=0.017).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (74)

Figure 1: Most frequent endoscopic and histologic findings at IGIB diagnosis. A, B: Diffuse bleeding predominantly observed at the gastric antrum; C: Mild inflammatory infiltrate with lymphocyte predominance and edema of the lamina propia (x20, hematoxilin-eosin). D: Normal antral mucosa (x10, hematoxilin-eosin).

IGIB (n=28)

non-IGIB (n=498)

p

Age, median (range)

59 (30-69)

53 (15-72)

0.014

Sex, n (%)

Male

21 (75)

297 (60)

0.11

HCT-CI, median (range)

3 (0-7)

2 (0-9)

0.78

Related medical priors, n (%)

Diabetes mellitus

4 (14)

28 (6)

0.082

Dyslipidemia

5 (18)

56 (11)

0.355

Smoking habit

12 (43)

168 (34)

0.315

Alcohol abuse

3 (11)

18 (4)

0.094

Liver disease

2 (7)

46 (9)

1

Peptic ulcer disease

0 (0)

3 (1)

1

Disease, n (%)

0.32

Acute myeloid leukemia

12 (43)

236 (47)

Acute lymphoid leukemia

4 (14)

81 (16)

Myelodisplastic neoplasms and myelomonocytic chronic leukemia

7 (25)

55 (11)

Chronic myeoloproliferative neoplasms

2 (7)

25 (5)

Lymphoproliferative disease

2 (7)

82 (16)

Aplastic anemia

1 (4)

16 (3)

Others

0 (0)

3 (1)

Donor, n (%)

Haploidentical donor

9 (32)

144 (29)

0.91

Matched sibling donor

10 (36)

175 (35)

Matched unrelated donor

9 (32)

179 (36)

Graft source, n (%)

Bone marrow

4 (14)

38 (8)

0.268

Peripheral blood

24 (86)

460 (92)

Conditioning regimen, n (%)

TBF

25 (89)

386 (78)

0.73

BU-FLU

0 (0)

52 (10)

TBI-FLU

0 (0)

22 (4)

FLAG-IDA-containing

0 (0)

9 (2)

Others

3 (11)

20 (5)

Conditioning regimen intensity, n (%)

Reduced intensity

18 (64)

303 (61)

0.843

Myeloablative

10 (36)

194 (39)

GvHD prophylaxis, n (%)

PT-CY + MMF + sirolimus

28 (100)

453 (91)

0.15

PT-CY + MMF + tacrolimus

0 (0)

45 (9)

Engraftment, median (range)

Days to myeloid engraftment >1x109/L

19 (13-47)

17 (6-57)

0.23

Days to platelet engraftment >20 x109/L

39 (14-263)

29 (6-248)

0.11

Days to IGIB episode, median (range)

64 (30-125)

Laboratory data

Hemoglobin descent (g/dL), median (range)

2.3 (0.2-4.4)

Platelet count (x109/L), median (range)

17.5 (3-86)

Platelet count prior to IGIB (x109/L), median (range)

31 (5-218)

Urea (mg/dL), median (range)

36.5 (11-83)

Reactive C protein (mg/L), median (range)

21 (2.8-250)

Coagulopathy, n (%)

0 (0)

Sirolimus overdose (previous two weeks), n (%)

9 (35)

Radiology data, n (%)

Splenomegaly

3 (12)

Portal hypertension features

3 (12)

Management during hospitalization

Days of hospitalization, median (range)

12 (0-51)

Red blood cell transfusions, median (range)

10.5 (2-54)

Platelet transfusions, median (range)

8.5 (0-87)

Fresh frozen plasma transfusions, median (range)

0 (0-6)

Start of total parenteral nutrition (TPN), n (%)

15 (58)

Days of TPN if started, median (range)

10 (5-26)

Intensive care unit admission, n (%)

3 (12)

Withdrawal of sirolimus, n (%)

17 (65)

Evolution

Reoccurrence of bleeding, n (%)

10 (38)

Patients with sirolimus at reoccurrence, n (%)

8 (80)

Deaths caused by bleeding, n (%)

1 (3)

Table 1: Patients’ baseline characteristics. Statistically significant p values are highlighted in bold.

Conclusions: IGIB is a severe but rarely fatal complication after alloHSCT. Scarce data are available in this setting, especially using a PT-Cy-based platform with sirolimus and MMF. Pathogenesis may be multifactorial, with patient and disease-specific characteristics, chemotherapy-related insult, and mTOR inhibitor antiangiogenic effects as possible culprits. Taking a conservative approach to manage IGIB seems safe, though more research is needed.

Disclosure: Authors have no conflicts of interest to declare.

14: Non-infectious Early Complications

P593 ACUTE KIDNEY INJURY AND CRONIC KIDNEY DISEASE AFTER ALLOGENEIC HCT. A SINGLE-CENTER EXPERIENCE

Ana Belén Bocanegra 1, Jaimovich David2, Irene Solano3, Javier Martinez-Costa3, Jorge Verdejo3, Maria Valdenebro3, Ana Muñoz Sanchez3, Carlos De Miguel3, Guiomar Bautista3, Sangeeta Hingorani4, Rafael Francisco Duarte3

1Hospital Puerta de Hierro Majadahonda, Majadahonda, Spain, 2Favaloro University Hospital, Buenos Aires, Argentina, 3Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Spain, 4University of Washington/Division of Nephrology Seattle Children’s Hospital, Division of Clinical Research, Fred Hutchinson Cancer Center, Seattle, United States

Background: Acute kidney injury (AKI) and chronic kidney disease (CKD) are common complications of allogeneic hematopoietic cell transplantation (allo-HCT) that associate with other patient and transplant characteristics and complications, may increase morbidity and impact patient outcomes.

Methods: We performed a retrospective analysis of consecutive allogeneic HCT recipients at our institution from 2009 to 2022 to examine AKI and CKD and their correlation with patient characteristics and outcomes. AKI episodes, according to KDIGO criteria, were registered during the initial admission for allo-HCT, from date admission to date of discharge, or up to day +30 in patients discharged earlier. CKD according to the decrease in the estimated glomerular filtration rate (eGFR) was registered up until one year after allo-HCT.

Results: Three-hundred consecutive allo-HCT recipients were included: median age of 49 (16-71), 57.3% males (Table 1). Pre-transplant median (IQR) creatinine (Cr) was 0.7 (0.21–1.45) mg/dl and eGFR was 106 (50.3–170) ml/min per 1.73 m2. Overall, 131 patients (43.7%) presented at least one episode of AKI: 44 stage 1 (14.7%) and 87 stage = or >2 (29%). The median time from hospital admission to first AKI episode was 26 days (2-187). Out of all patients with AKI, 32 had 2 AKI episodes (24.6%) and 10 had 3 AKI episodes (7.7%), 33 required ICU admission (25.2%), including 25 that required renal replacement therapy (19.1%). AKI stage = or >2 was more common in patients admitted to ICU (80.1% vs 21.9%; p<0.001), in those with SOS/VOD (71.0% vs 22.9%; p<0.001), TA-TMA (52.9% vs 25.9%; p=0.002), fungal infections (75% vs 26.5%; p<0.001) and BK virus nephritis/cystitis (72.2% vs 26.2%; p<0.001), but did not associate with other factors such as sex, underlying diagnosis, comorbidity score, conditioning intensity, baseline Cr, acute or chronic GVHD, parenteral nutrition, use of calcineurin inhibitors (CyA/tacrolimus), or the use of other potentially nephrotoxic drugs. Eighty-two of 300 patients (27.3%) had CKD during the first year after HCT: 32 had stage 1-2 (10.6%), 48 had stage 3-4 (16%), and only two patients (0.7%) ended up in dialysis. CKD was more common in patients who presented AKI (n=45, 34.3% of AKI cases and 54.9% of CKD cases) than in those without prior AKI (n=37, 21.9% of AKI cases and 45.1% of CKD cases; p=0.019). Median follow-up was 18.9 months (0–158.5). Survival was lower in patients with AKI stage = or >2 than in those with AKI Stages 1 or 0 at day +100 (65% vs 88% vs 97%) and one year (42% vs 62% vs 77%), respectively (p<0.001). CKD did not have an impact on survival.

Table 1. Characteristics of allogeneic HCT.

Characteristics

All patients (n=300)

AKI = or >2 (n=87)

p-value*

Donor, n (%)

<0.001

HLA identical sibling

36 (27.5)

15 (17.2)

Unrelated donor

17 (13)

12 (13.8)

Cord blood

46 (35)

33 (37.9)

Haploidentical donor

32 (24.4)

27 (31)

Conditioning regimen, n (%)

0.510

Myeloablative

153 (51)

47 (54.1)

Non-myeloablative

147 (49)

40 (45.9)

SORROR, n (%)

0.615

140 (46.7)

37 (42.5)

1-2

67 (22.3)

26 (29.9)

= or > 3

57 (19)

13 (14.9)

missing values

36 (12)

12 (64)

Days of admission median (range)

36 (8-256)

54 (24-256)

0.000

  1. *Between No AKI and AKI Stage = or >2
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (75)

Conclusions: AKI is frequent after allo-HCT, and moderate-severe stage stage = or >2 AKI associate with other infective and non-infective transplant complications and associates with poorer patient outcomes. CKD is also frequent after allo-HCT, and although it often associates with prior AKI, it can also happen de novo in a high percentage of patients. Prevention, early recognition, and interventions to preserve renal function and prevent AKI and CKD need to be considered in the management of allo-HCT recipients.

Disclosure: No disclosures to declare.

14: Non-infectious Early Complications

P594 SINUSOIDAL OBSTRUCTION SYNDROME/VENO-OCCLUSIVE DISEASE: A RETROSPECTIVE STUDY OF THE SPANISH HEMATOLOGY ASSOCIATION-HEMATOPOIETIC STEM CELL TRANSPLANTATION GROUP (GETH)

Cristina Blázquez Goñi 1, Francisco Manuel Martin Dominguez1, Silvia García Canale1, Cristina Díaz de Heredia2, Melissa Panesso Romero2, Maribel Benítez Carabante2, María Luz Uría2, David Bueno3, Annalisa Paviglianiti4, Pilar Palomo Moraleda5, Ana Isabel Gallardo Morillo6, Antonia Pascual Martínez6, Marina Acera Gómez7, Alexandra Regueiro8, Carlos Vallejo Llamas9, Gillen Oarbeascoa10, Diana Campos11, Leslie González Pinedo12, Melissa Torres12, Julia Marsal Ricomà13, Marta González Vicent14, María José Jiménez Lorenzo15, Estefanía García Torres16, Mónica López Duarte17, Andrés Sánchez Salinas18, Pedro González Sierra19, José Luis López Lorenzo20, Amaya Zabalza21, Sara Redondo Velao22, Oriana López-Godino23, Alejandro Luna de Abia24, Cristina Beléndez10, Beatriz Aguado25, María Teresa Artola Urain26, Patricia Jiménez Guerrero27, Agustín Nieto Vázquez28, Francisca Almagro Torres29, José Antonio Sanz30, Beatriz de Rueda31, Rocío Picón González32, José Antonio Pérez Simón1

1University Hospital Virgen del Rocio, Instituto de Biomedicina de Sevilla (IBIS / CISC), Seville, Spain, 2University Hospital Vall de Hebrón, Barcelona, Spain, 3University Hospital La Paz, Madrid, Spain, 4University Hospital ICO Duran i Reynals, Barcelona, Spain, 5University Hospital Central de Asturias, Oviedo, Spain, 6University Hospital Central de Málaga, Málaga, Spain, 7University Hospital Salamanca, Salamanca, Spain, 8University Hospital Santiago, Santiago de Compostela, Spain, 9University Hospital Clínico de Santiago, Santiago de Compostela, Spain, 10University Hospital Gregorio Marañón, Madrid, Spain, 11University Hospital Clínico de Valencia, Valencia, Spain, 12University Hospital Doctor Negrín, Las Palmas de Gran Canaria, Spain, 13University Hospital Sant Joan de Deu, Barcelona, Spain, 14University Hospital Niño Jesús, Madrid, Spain, 15University Hospital Germans Trias, Barcelona, Spain, 16University Hospital Reina Sofía, Córdoba, Spain, 17University Hospital Marqués de Valdecilla, Santander, Spain, 18University Hospital Virgen de la Arrixaca, Murcia, Spain, 19University Hospital Virgen de las Nieves, Granada, Spain, 20University Hospital Fundación Jiménez Díaz, Madrid, Spain, 21University Hospital Navarra, Pamplona, Spain, 22University Hospital Sant Pau, Barcelona, Spain, 23University Hospital Morales Meseguer, Murcia, Spain, 24University Hospital Ramón y Cajal, Madrid, Spain, 25University Hospital La Princesa, Madrid, Spain, 26University Hospital Donosti, San Sebastián, Spain, 27University Hospital Jerez, Jerez de la Frontera, Spain, 28University Hospital Vigo, Vigo, Spain, 29University Hospital Jaén, Jaén, Spain, 30University Hospital Son Espases, Mallorca, Spain, 31University Hospital Miguel Servet, Zaragoza, Spain, 32Faculty of Sciences, Cádiz University, Cádiz, Spain

Background: Veno-occlusive disease (SOS/VOD), is a potentially life-threatening complication that may develop after hematopoietic cell transplantation (HCT).

The aims were to evaluate the incidence of SOS/VOD in a large cohort of children and adults transplanted in centres across Spain by applying EBMT, Baltimore and Seattle criteria and to analyse the risk factors underlying this complication.

Methods: This is a retrospective, multicenter and observational study performed in 30 spanish centers between January 2019 and December 2021, to analyze the main uses of defibrotide in autologous or allogeneic HCT. EBMT, Seattle and Baltimore criteria were retrospectively applied for diagnoses of SOS/VOD and severity grading. Overall survival (OS) and event-free survival were calculated according to the Kaplan-Meier method. The risk of death for causes unrelated to malignant progression, defined as nonrelapse mortality (NRM), and that of developing SOS/VOD were calculated as cumulative incidences to adjust the analysis for competing risks.

Results: A total of 300 patients were included, 183 male (61%) and 117 female (39%), with a median age of 29 years; most frequent diagnoses were ALL and AML (52.2%). 272 patients underwent Allogenic transplantation (90.66%) and 28 Autologous (9.33%).

The median time of SOS/VOD occurrence was 35 days after HSCT (range, 4 to 742). 187 patients were treated with defibrotide for a median time of 19 days (6- 84); defibrotide was used as prophylaxis in 113 patients (23%). The overall mortality directly related to SOS/VOD was 48.8% at 3 months post-HCT (in children: moderate 9.3%, severe 14.8% and very severe 46.3% (p=0.03): in adults was 19%, 25% and 42% for moderate, severe and very severe VOD (p=0.027)) and 21.1% at 1-year post-HCT.

Using standard definitions including EBMT criteria, around 45% (n = 49/108) of adult cases were classified as late-onset VOD but with D100 survival (75% vs 54%, p=0.015) compared to classical VOD. Mortality D100 in those who develop VOD in classic and late-onset were GVHD (25% vs 31%), infection (56% vs 52%), cardiac (9% vs 0%) and pulmonary (15% vs 0%).

Depending on bilirubin levels, mortality D100 in adults was 44% (bilirubin < 2 mg/dL), 53% (bilirubin 2-5 mg/dL) and 65% (bilirubin > 5 mg/dL) (p=0.20); in children was 10% (bilirubin < 2 mg/dL), 8.5% (bilirubin 2-5 mg/dL) and 56% (bilirubin > 5 mg/dL) (p=0.0012).

Prognostic factors implicated in mortality D100 in adult population include bilirubin levels < 5. No other parameters were found statistically significant in mortality.

Survival D100 in adults who discontinued defibrotide due to VOD resolution was 78% vs 28% in those who discontinued for other reasons; in children, survival D100 was 100 % vs 50%.

Defibrotide toxicity analyses showed that the most frequent adverse events were haemorrhage (10.6%), gastrointestinal bleeding (6.4%) and coagulopathy (5.6%).

Conclusions: Overall survival on D100 was higher in late onset adult patients than classic adults, and also in patients with defibrotide discontinuation due to VOD resolution.

Defibrotide was safe, consistent with previous studies.

Disclosure: This work was supported with a fund by Jazz Pharmaceuticals.

14: Non-infectious Early Complications

P595 SAFETY AND BENEFIT OF LIVER BIOPSY AFTER ALLOGENIC STEM CELL TRANSPLANTATION: A RETROSPECTIVE SINGLE-CENTER STUDY

Marie Maulini 1, Aurélie Bornand1, Federico Simonetta1, Anne-Claire Mamez1, Chiara Bernardi1, Federica Giannotti1, Yan Beauverd1, Thomas Longval1, Maria Mappoura1, Sarah Noetzlin1, Laetitia Dubouchet1, Sarah Perdikis-Prati1, Evgenia Laspa1, Cuong-An Do1, Thien-An Tran1, Carmen De Ramon Ortiz1, Amandine Pradier1, Sarah Morin1, Stavroula Masouridi-Levrat1, Simon Maulini2, Laura Rubbia-Brandt1, Laurent Spahr1, Yves Chalandon1

1University Hospital of Geneva (HUG), Genève, Switzerland, 2Ecole polytechnique fédérale de Lausanne (EPFL), Lausanne, Switzerland

Background: Liver complications after allogeneic hematopoietic stem cell transplantation (alloHSCT) are frequent and causes can be diverse. The three most common aetiologies are graft-versus-host disease (GVHD), drug -induced toxicity and infectious hepatitis. Another cause of hepatic complication after alloHSCT is sinusoidal obstruction syndrome/veno-occlusive disease (SOS/VOD). Liver tests alone are not sufficient to differentiate these pathologies, and gold standard for diagnosis remains histology. However, most transplant centers are reluctant to perform liver biopsy because of the potential complications of this procedure.

Methods: The purpose of this article is to demonstrate the safety and benefit of liver biopsy (using either the transjugular or percutaneous route) in patients with liver complications after alloHSCT. We extracted from our database a total of 145 liver biopsies performed in 109 patients who underwent alloHSCT between 2011 and 2021 at the University Hospital of Geneva (HUG). To assess safety, we looked for adverse events following percutaneous or transjugular liver biopsy defined as pain, bleedings, infections, transfer to ICU and death. To assess benefit, we analysed pre- and post-biopsy diagnosis and compared them. We also extracted treatment modalities adapted to the histological results and determined if the biopsy led to any change in clinical management.

Results: The median age at time of liver biopsy was 51 years old (range 18-75 years old); 75% of the patients were men. The median time between transplant and liver biopsy was 333 days. The most represented diseases were acute leukemias, non-Hodgkin lymphomas and myelodysplastic neoplasms. 38.62 % of patients received a myeloablative conditioning, 60 % a reduced-intensity conditioning and 1.38 % a sequential conditioning. There was an identical proportion of HLA-identical sibling donors as matched-unrelated donors (37.24 % each), 14.48 % of haplo-identical donors and 11.04 % of mismatched donors. 65.52 % of biopsies were transjugular, and 33.10 % percutaneous (1.38 % unknown).

Regarding complications, there were 5 cases (3.44 %) of pain, 4 (2.75 %) of bleeding (3 after transjugular biopsy, 1 after percutaneous biopsy), 1 (0.68 %) of infection and 3 (2.06 %) of transfer to ICU. There were no deaths related to the procedure.

Regarding benefit, post-biopsy diagnosis was different from pre-biopsy diagnosis in 82 cases (56.56 %), it was identical in 48 cases (33.10 %) and remained undetermined in 15 cases (10.34 %). Liver biopsy led to a change in clinical management in 86 cases (59.72 %). It did not lead to any change in 57 cases (38.89 %). The management remained unknown in 2 cases (1.39 %).

Conclusions: Our study suggests that liver biopsy in alloHSCT patients is safe (very few significant adverse events) and useful, leading to changes in diagnosis and clinical management in a substantial number of cases.

Disclosure: Y. Chalandon has received consulting fees for advisory board from MSD, Novartis, Incyte, BMS, Pfizer, Abbvie, Roche, Jazz, Gilead, Amgen, Astra-Zeneca, Servier; Travel support from MSD, Roche, Gilead, Amgen, Incyte, Abbvie, Janssen, Astra-Zeneca, Jazz, Sanofi all via the institution. S. Masouridi-Levrat has received travel support from Gilead, BeiGene, Jazz and Sanofi all via the institution.

14: Non-infectious Early Complications

P596 INVESTIGATION OF LABORATORY PARAMETERS BEFORE THE ONSET OF HEPATIC SINUSOIDAL OBSTRUCTION SYNDROME IN CHILDREN AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION

Bernd Gruhn 1, Lorena Johann1

1Jena University Hospital, Jena, Germany

Background: Hepatic sinusoidal obstruction syndrome (SOS) is a serious complication following hematopoietic stem cell transplantation (HSCT). Early diagnosis and treatment with defibrotide can improve patient outcome. The aim of our study was to detect laboratory parameters following HSCT that can predict the occurrence of SOS.

Methods: We analyzed 182 children who underwent HSCT for the first time and without defibrotide prophylaxis. Our study population consisted of 126 patients who received allogeneic HSCT (69.2%) and 56 patients who underwent autologous HSCT (30.8%). The diagnosis of SOS was based on the pediatric criteria of European Society for Blood and Marrow Transplantation. We investigated the following 15 laboratory parameters after HSCT before the onset of SOS: international normalized ratio (INR), activated partial thromboplastin time (aPTT), reptilase time, factor VIII, factor XIII, von Willebrand factor (vWF), von Willebrand factor activity (vWF activity), fibrinogen, antithrombin III, protein C, protein S, D-dimer, alanine aminotransferase (ALT), bilirubin, and ferritin. The last laboratory values within one week before the onset of SOS were analyzed in the SOS patient group. In the non-SOS patient group, we investigated the parameters for four weeks after transplantation and calculated the median. We selected cut-off values for the continuous parameters based on receiver operating characteristic curves analyses and existing reference values. We performed Mann-Whitney U test, univariate and multivariate analyses to find significant associations between the values of laboratory parameters and the occurrence of SOS. The probability of developing SOS was calculated using the significant independent variables.

Results: The overall incidence of SOS was 14.8% at a median time of 13 days after HSCT. SOS developed in 24 of 126 allogeneic (19.1%) and in 3 of 56 autologous (5.4%) HSCT patients. Only 3 out of 27 patients (11.1%) who suffered from very severe SOS died within 100 days after HSCT. In the univariate analysis, we found significant associations between the onset of SOS and the following parameters: INR ≥ 1.3, aPTT ≥ 40 sec, reptilase time ≥ 18.3 sec, factor VIII ≤ 80%, antithrombin III ≤ 75%, protein C ≤ 48%, D-dimer ≥ 315 µg/L, bilirubin ≥ 9 µmol/L, and ferritin ≥ 3100 µg/L. In multivariate analysis, we confirmed INR ≥ 1.3 [odds ratio (OR) = 8.104, P = .006], aPTT ≥ 40 sec (OR = 10.174, P = .001), protein C ≤ 48% (OR = 5.215, P = .014), and ferritin ≥ 3100 µg/L (OR = 7.472, P = .004) as independent risk factors after HSCT before SOS. If three of the four significant cut-off values were present, the probability of developing SOS was more than 70%. The probability of SOS was 96%, if all four laboratory parameters were changed according to the cut-off values.

Conclusions: The laboratory parameters INR, aPTT, protein C, and ferritin were very useful to predict the occurrence of SOS. These four parameters can enable earlier diagnosis and treatment leading to an improved outcome. In addition, this is the first report on a significant association between SOS and high values of INR and aPTT after HSCT before SOS.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P597 OUTCOME OF ALLOGENEIC HCT RECIPIENTS COMPARED TO OTHER PATIENTS WITH HEMATOLOGICAL MALIGNANCIES ADMITTED TO THE INTENSIVE CARE UNIT. A SINGLE CENTRE STUDY

Ana Belen Bocanegra 1, Marta Perez Calle1, Jorge Verdejo1, Alicia Segura1, Ana María Bellón1, Rosalia Alonso1, Ana Amaro Harpigny1, Patricia Enciso1, Inmaculada Tendero1, Maria Esther Martinez-Muñoz1, Carlos De Miguel1, Guiomar Bautista1, Ines Lipperheide1, Daniel Ballesteros1, Rafael Francisco Duarte1

1Hospital Universitario Puerta de Hierro Majadahonda, Majadahonda, Spain

Background: Critically ill patients with hematological malignancies (HM) often require admission to the intensive care unit (ICU). Their outcomes maybe poor, and their overall eligibility for admission to ICU, in particular for those after allogeneic transplant (allo-HCT), remains a clinical decision based on hematology and intensive medicine criteria as well as on the center’s clinical experience. Here, we aim to assess the clinical outcomes of our HM patients admitted to the ICU, with a particular focus on allo-HCT recipients and other characteristics that may associate with short-term ICU mortality and longer-term survival.

Methods: We have retrospectively analyzed all patients with HM, including allo-HCT recipients, who were admitted to the ICU at our center from 2009 to 2022. Key clinical outcomes were ICU mortality, and 100 days and 1 year mortality after ICU admission. Mann-Whitney Test and Fisher’s exact test were used to compare continuous and categorical variables, respectively. The Kaplan-Meier method was employed to estimate the probability of survival. Hazard ratios for ICU mortality were estimated using both univariable and multivariable Cox regression. Variables with a significant p-value in the univariable analysis were included in the multivariable analysis. Statistical analysis was performed on STATA 16 software.

Results:

Table 1. Characteristics of ICU Admission and Treatment.

Characteristics

All patients

(n=191)

Non-AlloHCT

(n=110)

AlloHSCT

(n= 81)

p-value

Reason for ICU admission, n (%)

Respiratory failure

72 (37.7)

32 (29)

40 (49.4)

0.006

Distributive Shock (sepsis)

49 (25.7)

33 (30)

16 (19.8)

0.132

Other shock (cardiogenic, …)

17 (8.9)

11 (10)

6 (7.4)

0.614

Acute kidney injury

5 (2.6)

2 (1.8)

3 (3.7)

0.652

Neurologic cause

5 (2.6)

4 (3.7)

1 (1.2)

0.397

Bleeding

6 (3.1)

4 (3.7)

2 (2.5)

1.000

Others

37 (19.4)

24 (21.8)

13 (16)

0.358

ICU treatment, n (%)

Vasoactive agents

116 (60.7)

68 (61.8)

48 (59.3)

0.639

Invasive mechanical ventilation

81 (42.4)

38 (34.6)

43 (53)

0.016

Non-invasive mechanical ventilation

2 (1)

2 (1.8)

0.661

High-Flow nasal cannula

39 (20.4)

20 (18.2)

19 (23.5)

0.476

Renal replacement therapy

54 (28.3)

23 (20.9)

31 (38.3)

0.014

APACHE II score at admission, median (range)

15 (3-39)

14.5 (3-39)

15 (4-37)

0.492

Time from alloHCT to admission to ICU in days, median (range)

-

-

113 (-1-3351)

-

Time from hospitalization to admission to ICU in days, median (range)

11 (0-88)

5 (0-68)

19 (0-88)

<0.001

Days in ICU, median (range)

5 (0-127)

4.5 (0-83)

6 (0-127)

0.033

A total of 191 patients were studied [median age 56 years (16-84); 118 men (62%); 91 acute leukemia (48%), 51 lymphoma (27%), 24 plasma cell discrasias (12%), 16 CLL (8%) and 9 MDS/MPN (5%)], including 81 allo-HCT recipients (42%) from matched unrelated (38, 47%), related (28, 34%) or haploidentical (15, 19%) donors. Median time from hospitalization to ICU admission was longer in allo-HCT recipients (19 vs 5 days, p<0.001). Respiratory failure, the commonest cause of admission to ICU (38%), was more common in allo-HCT recipients (49% vs 29%, p=0.006; Table 1), a higher percentage of whom required invasive mechanical ventilation (53% vs 35%, p=0.016). They also required more renal replacement therapy (38% vs 21%, p=0.014). Of note, APACHE II score at ICU admission was comparable between groups. The rate of ICU mortality was 30.4 % overall, 37% and 25.5% for patients with and without allo-HCT, respectively (p=0.111). Longer-term overall survival for all cases was 50.8% at day 100 and 37.7% at one year since ICU admission, lower for allo-HCT recipients (38.8% vs 60% at day 100 and 24.7% vs 47.3% at one year, respectively; p=0.001). The primary hematological diagnosis, severe neutropenia, [RD1] disease status at ICU admission, arterial pH and Br in fist 24h at ICU admission, use of renal replacement therapies and mechanical ventilation were found to be independent risk factors of ICU mortality in multivariate analysis.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (76)

Conclusions: Clinical outcome of patients with HM admitted to ICU remains unfavorable. Nevertheless, nearly 70% survive and are discharged from ICU, with a 38% probability of longer-term survival at one year. Allo-HCT recipients require more invasive mechanical ventilation and renal replacement, although their probability of survival and discharge from ICU, at 63%, is not statistically different from that of other HM patients. They have, however, poorer longer-term survival at one year. Further investigation is warranted.

Disclosure: No conflict of interes to declare.

14: Non-infectious Early Complications

P598 ADVERSE PROGNOSTIC IMPACT OF PRETRANSPLANT HEPCIDIN AND FERRITIN ON OUTCOMES FOLLOWING HEMATOPOIETIC STEM CELL TRANSPLANTATION

Michelle Pirotte 1, Marianne Fillet2, Laurence Seidel3, Evelyne Willems1, Sophie Servais1, Frédéric Baron1, Yves Beguin1

1University Hospital of Liège, Liège, Belgium, 2Laboratory for the Analysis of Medicines, University of Liège, Liège, Belgium, 3University Hospital of Liège and University of Liege, Liège, Belgium

Background: Elevated pre-transplant ferritin levels have consistently shown associations with reduced overall survival (OS) and progression-free survival (PFS) as well as an elevated susceptibility to infections. Hepcidin, the pivotal regulator of iron metabolism, has received limited attention after allogeneic hematopoietic stem cell transplantation (allo-HSCT), with an uncertain impact on survival and infections. We explore the association between pretransplant ferritin and hepcidin with OS, PFS, non-relapse mortality (NRM), infections, engraftent, and graft-versus-host disease (GVHD) in 502 patients.

Methods: 502 patients who underwent allo-HSCT at the University Hospital of Liège between 1999 and 2012 and had a minimal follow-up of 5 years were included. Pretransplant serum hepcidin (measured by mass spectrometry) and ferritin levels were investigated using univariate (UV) and multivariate (MV) linear regression. Patients were grouped into quartiles based their values. OS, PFS, NRM, engraftment, infections, and GVHD were estimated using the Kaplan-Meier method, compared between subgroups (log-rank test), and analyzed in multivariate Cox models.

Results: Median pretransplant hepcidin levels was 25.13 ng/mL, and ferritin 1,083 ng/mL, with a median ferritin peak of 3,419 ng/mL within the first 100 days post-transplantation. High ferritin, CRP, pretransplant red blood cell (RBC) transfusions, HCT-CI, performance status (PS) and low neutrophils were independently associated with elevated pretransplant hepcidin (R² = 0.44, p <0.0001). Pre-transplant ferritin levels were positively associated with pre-transplant hepcidin, transferrin saturation, alanine aminotransferase, creatinine, pretransplant RBC and platelet transfusions, PS, and negatively correlated with hemoglobin levels and influenced by diagnostic type (R²=0.67, p<0.0001). OS and PFS were significantly lower in the highest hepcidin (p<0.0001 & p=0.0006) and the ferritin (p<0.0001 & p=0.0003) groups compared to groups with lower values. Predictive power of ferritin surpasses that of hepcidin on survival outcomes in UV analysis. In MV analysis, pretransplant ferritin was significantly correlated with decreased survival, unlike hepcidin. Elevated pretransplant ferritin was also associated with an increased risk of post-transplant infections in UV but not in MV analysis, yielding to transferrin saturation (Tsat), while hepcidin did not have a significant effect on infections. Ferritin, but not hepcidin, correlated with poor engraftment in UV but not in MV analysis. No significant association was observed between the two parameters and GVHD.

.

Table 1 A. Multivariate analysis of 10-years OS:

Significant parameters

P value

Hazard Ratio

95% confidence interval

Pretransplant serum ferritin

0.01

1.17

1.03

1.33

Pretransplant total bilirubin

0.01

1.45

1.09

1.92

Disease risk index score

0.0004

1.43

1.17

1.74

Performance status

0.001

1.58

1.20

2.08

Age

0.002

1.02

1.01

1.03

Table 1 B. Multivariate analysis of 1-year PFS:

Significant parameters

P value

Hazard Ratio

95% confidence interval

Pretransplant serum ferritin

0.02

1.19

1.03

1.37

Pretransplant total bilirubin

0.02

1.46

1.06

2.02

Myelodysplastic syndrome (vs acute leukemia)

0.002

2.07

1.31

3.26

Disease risk index score

0.004

1.40

1.11

1.77

Number of treatment lines

0.006

1.19

1.05

1.35

Sex match (other vs F donor to M recipient)

0.007

0.56

0.37

0.85

Mycotic prophylaxis (other vs fluconazole)

0.03

1.43

1.04

1.98

Table 1 C. Multivariate analysis of time to first infection:

Significant parameters

P value

Hazard Ratio

95% confidence interval

Pretransplant saturation of transferrin

0.03

1.25

0.50

0.96

Non myeloablative vs myeloablative conditioning

<0.0001

0.17

0.12

0.25

Sex recipient (F vs M)

0.045

1.30

1.01

1.68

HLA-mismatched unrelated vs HLA identical sibling donor

<0.0001

1.91

1.38

2.63

Use of antibiotic prophylaxis

0.03

0.69

0.50

0.96

Conclusions: The significance of pretransplant hepcidin and ferritin levels is complex. These levels not only serve as indicators of iron overload resulting from prior transfusions but are also influenced by various factors reflecting patient comorbidities (inflammation, renal insufficiency, hepatic cytolysis, anemia, and overall comorbidities) and the underlying hematologic disease. Elevated pretransplant serum ferritin levels are associated with adverse outcomes, including reduced OS, PFS and NRM as well as an elevated risk of infections and poor engraftment. However, it largely loses its predictive power in multivariate analysis.

Higher pretransplant serum ferritin levels exhibit a stronger correlation with adverse post-transplant outcomes compared to elevated hepcidin. This study emphasizes the intricate nature of iron metabolism and its influence on allo-HSCT outcomes, underscoring the importance of considering multiple biological and clinical variables in transplantation prognosis.

Disclosure: I don’t have any disclosure.

14: Non-infectious Early Complications

P599 IMPACT OF GRANULOCYTE-COLONY STIMULATING FACTOR (G-CSF) ON CLINICAL OUTCOMES IN ALLOGENEIC STEM CELL TRANSPLANTATION: INSIGHTS FROM A SINGLE-CENTER EXPERIENCE AT PRINCESS MARGARET CANCER CENTRE

Ahmed Alnughmush1,2, Ayman Sayyed1,2, Mohammed Kawari 2, Mats Remberger3, Carol Chen1, Caden Chiarello1, Ivan Pasic1,2, Igor Novitzky-Basso1,2, Arjun Datt Law1,2, Wilson Lam1,2, Dennis (Dong Hwan) Kim1,2, Fotios V. Michelis1,2, Armin Gerbitz1,2, Auro Viswabandya1,2, Rajat Kumar1,2, Jonas Mattsson1,2,4

1Hans Messner Allogeneic Transplant Program, Division of Medical Oncology and Hematology. Princess Margaret Cancer Centre, University Health Network, Toronto, Canada, 2University of Toronto, Toronto, Canada, 3Uppsala University Hospital, Uppsala University, Uppsala, Sweden, 4Gloria and Seymour Epstein Chair in Cell Therapy and Transplantation, University of Toronto, Toronto, Canada

Background: Despite decades of post-allogeneic stem cell transplantation growth factor utilization, its role remains undefined, sparking ongoing debates and research. The theoretical impacts of growth factors were challenged in numerous studies. The practice of using G-CSF post BMT varies widely between transplant physicians and centers.

Methods: We conducted a retrospective analysis of 509 patients undergoing allogeneic hematopoietic cell transplantation between May 1, 2019, and May 31, 2022, at Princess Margaret Cancer Centre, University Health Network, Toronto, Canada. G-CSF was routinely administered to most patients on day+7 pre-May 1, 2021; thereafter, its use became discretionary.

The primary objective is to determine the incidence and timing of neutrophil and platelet engraftment in patients with or without G-CSF support. Secondary endpoints include overall survival, non-relapse mortality, duration of hospital stay, GvHD incidence, and engraftment syndrome occurrence.

Results: Patient demographics and baseline characteristics are summarized in Table 1.

Within our cohort, recipients of post-transplant G-CSF exhibited accelerated ANC engraftment compared to those without G-CSF administration (median: 16 vs. 18 days, respectively, P<0.001). It is crucial to underscore that individuals who received G-CSF after May 2021 experienced a prolonged period to ANC and platelet engraftment. However, this subgroup is inherently biased, primarily comprising patients with underlying issues such as graft failure, rendering their extended engraftment unrelated to G-CSF use.

Among those on G-CSF, a notable delay in platelet engraftment was observed (Median: 21 vs. 17 days for those not on G-CSF). Despite G-CSF hastening ANC engraftment, this did not manifest in clinically significant outcomes, as evidenced by comparable initial lengths of stay and rates of length of stay at day 100 between the group receiving G-CSF before May 2021 and those who did not receive it (median LOS 30 vs. 30 days, P=0.15 & 33 vs. 33 days, P=0.27, respectively).

No significant differences were observed in 1-year Overall Survival (OS) and 1-year Non-Relapse Mortality (NRM) among the groups. In our cohort, G-CSF usage did not increase the risk of Graft-versus-Host Disease (GVHD), and the 1-year GRFS for those receiving G-CSF before May 2021 was 60.6%, compared to 59% for those not receiving G-GCSF.

An intriguing observation in our study is that while the risk of engraftment syndrome remained consistent whether G-CSF was administered or not, there was a significantly higher risk of developing engraftment syndrome among those receiving it for a prolonged period (median 11 vs. 9 days, P=0.02). Moreover, in our cohort, engraftment syndrome contributed to a significant prolongation in length of stay (median 36 vs. 31 days for those who did not develop engraftment syndrome, P<0.001). Multivariate analysis for engraftment syndrome identified HLA-Mismatch (OR 1.72, 95% CI 1.03-2.88, P=0.038) and the number of G-CSF days (OR 1.04, 95% CI 1.00 - 1.09, P=0.038) as factors increasing the risk of engraftment syndrome.

Characteristics

All Patients (N= 509)

Age 58 (18-76)

G-CSF before May 2021 (N=298)

Age 58 (18-76)

G-CSF after May 2021

(N=94)

Age 59 (18-74)

No G-CSF (N= 117)

Age 54 (18-73)

p-value

AML

231 (45.4)

147 (49.3)

35 (37.2)

49 (41.9)

0.18

ALL & lymphoma

85 (16.7)

42 (14.1)

21 (22.4)

22 (18.8)

MPAL

12 (2.4)

11 (3.7)

1 (1.1)

MDS & CMML

98 (19.3)

55 (18.5)

21 (22.3)

22 (18.8)

CML/CLL

19 (3.7)

10 (3.3)

4 (4.2)

5 (4.3)

MF

33 (6.5)

20 (6.7)

7 (7.4)

6 (5.1)

Other malignant

4 (0.8)

2 (0.7)

1 (1.1)

1 (0.9)

Non-Malignant

27 (5.3)

11 (3.7)

4 (4.3)

12 (10.3)

Source: BM/PBSC

22/487

12/286

4/90

6/111

0.88

CD34 dose

6.9 (0.3-13.7)

7.1 (0.3-13.7)

6.3 (1.2-10.2)

6.6 (1.3-11.3)

0.008

MRD

113 (22.2)

58 (19.4)

17 (18.1)

38 (32.5)

0.46

MUD

239 (47.0)

137 (46.0)

45 (47.9)

57 (48.7)

Haplo

108 (21.2)

69 (23.1)

26 (27.7)

13 (11.1)

MM URD

49 (9.6)

34 (11.4)

6 (6.4)

9 (7.7)

Conditioning:

RIC/MAC

301/208

182/116

62/32

57/60

0.02

GVHD prophy: ATG

79 (15.5)

19 (6.4)

5 (5.3)

55 (47.0)

<0.001

GVHD prophy: Other

15 (2.9)

3 (1.0)

4 (4.3)

8 (6.8)

GVHD prophy: PTCy

77 (15.1)

55 (18.5)

12 (12.8)

10 (8.5)

GVHD prophy: PTCy+ATG

338 (66.4)

221 (74.2)

73 (77.7)

44 (37.6)

Conclusions: Although the administration of G-CSF post BMT led to a numerically faster neutrophil engraftment, it did not translate into clinically significant outcomes. The risk of engraftment syndrome (ES) necessitates careful consideration, and its association warrants further investigation in future studies.

Disclosure: Author Name: Jonas Mattsson.

Speaking & consultation honoraria: Merck, Jazz Pharmaceuticals, Takeda, Gilead, Mallinckrodt, Sanofi, Novartis.

14: Non-infectious Early Complications

P600 VALIDATION OF EBMT 2023 VOD CRITERIA: IMPACT ON EARLY DIAGNOSIS AND SENSITIVITY/ESPECIFICITY OF PROBABLE VOD CATEGORY

Monica Cabrero 1,2, Maria Cortes-Rodriguez3,2, Almudena Cabero1,2, Estefania Perez-Lopez1,2, Ana Africa Martin-Lopez1,2, Monica Baile1,2, Alejandro Avendaño1,2, Ana Garcia-Bacelar1,2, Lorena Hernandez-Medina2, Marina Acera2, Lourdes Vazquez1,2, Fermin Sanchez-Guijo1,2, Lucia Lopez-Corral1,2

1Salamanca University Hospital, Salamanca, Spain, 2Institute of Biomedical Research of Salamanca (IBSAL), Salamanca, Spain, 3University of Salamanca, Salamanca, Spain

Background: Veno-occlusive disease (VOD) is a potentially severe complication affecting around 5-10% of patients receiving an allogeneic stem cell transplantation (AlloSCT) with over 80% mortality without targeted therapy. Early diagnosis and treatment improve VOD outcomes. Classic Seattle/Baltimore and EBMT16 criteriaunderestimate anicteric SOS/VOD and ultrasound (US) findings are only partially considered. To fill these gaps, an EBMT consensus published in 2023 re-defined VOD criteria including a category of “probable VOD”. In the present study, we focus on validating these EBMT23 criteria both in VOD patients and in a group of AlloSCT recipients.

Methods: We designed a retrospective study of patients diagnosed with VOD at our center in the last 10 years to evaluate impact of EBMT23 criteria in in two distinct groups: patients diagnosed with VOD and recipients of allogeneic stem cell transplantation (AlloSCT)In a 2nd step, we retrospectively review all consecutive patients receiving an AlloSCT in our center in 2023 with >100 days of follow-up to calculate specificity, sensitivity, and positive/negative predictive values of probable VOD category.

Results: To analyse impact of EBMT23 criteria, we reviewed 33 patients diagnosed with VOD (31 AlloSCT, 1 CAR-T and 1 autologous SCT). The median time to diagnosis was 17 days (3-125) and 13 (39%) were late-onset VOD; 15 cases were anicteric at diagnosis, while 6 (18%) remained anicteric throughout the entire VOD episode, with significant differences between classical and late-onset (95 vs 61%; p=0.008). Incidence of diagnostic items are shown in table 1. Ultrasound was performed in all patients, being compatible with VOD in 30 (91%) and necessary for diagnosis in 11 (33%), 3 (15%) in classical and 8 (62%) in late-onset VOD (p=0.009). Ultrasound findings are described in table 1.

All patients but 1 met probable VOD criteria, allowing an earlier diagnosis in 14 (42%) cases with a median of 2 days (1-7), 1.5 days (1-7) in classical and 3 days (1-5) in late-onset VOD. Twenty-four patients (73%) qualified for EBMT23 clinical VOD.

To explore our second objective, we retrospectively reviewed 61 patients receiving an AlloSCT from January to September 2023. From them, 7 met criteria for probable VOD. Two of them progressed to very severe VOD and one remained as mild probable VOD, while 4 had an alternative diagnosis of infection proved by positive cultures or radiological findings. From the 3 patients diagnosed as VOD probable, only 1 qualified for clinical VOD. We did not prove any VOD diagnosis among patients who did not fulfil probable VOD criteria.

Based on this data, sensitivity and specificity for VOD probable are 100% (95% CI: 83-100) and 93.1% (85.7 – 100). Positive and negative predictive value are 42.86% and 100% respectively.

Table 1

CLINICAL VARIABLE

N (%)

Classical (n=20) vs late-onset (n=13) VOD

Days to onset from VOD diagnosis (median, range)

Weight gain over 5%

28 (85%)

18 (90%) vs 10 (77%)

NS

-1 (-15, 3)

Painful hepatomegaly

25 (77%)

18 (90%) vs 11 (85%)

NS

0 (-7, 4)

Ascites

31 (94%)

18 (90%) vs 13 (100%)

NS

0 (-11, 7)

Bb >2 at diagnosis

18 (55%)

13 (65%) vs 5 (39%)

NS

NA

Bb >2 during VOD episode

26 (79%)

19 (95%) vs 8 (61%)

p=0.008

0 (-4, 9)

VOD-related ultrasound data

30 (91%)

17 (85%) vs 13 (100%)

NA

Hepatomegaly

27 (82%)

16 (80%) vs 11 (85%)

Gallbladder wall thickening

24 (72%)

14 (70%) vs 10 (77%)

Reversal portal flow

8 (24%)

4 (20%) vs 4 (31%)

High hepatic art. RI

9 (27%)

6 (30% vs 3 (23%)

Abnormal suprahepatic flow

16 (49%)

8 (40%) vs 8 (62%)

Conclusions:

  • New probable VOD category emerges as a useful tool for early VOD diagnosis, while the clinical VOD category still underdiagnose anicteric VOD.

  • Probable VOD criteria show high sensitivity and specificity, with almost half of patients meeting probable VOD criteria finally diagnosed with VOD.

  • A meticulous differential diagnosis including infections, drug toxicity and GVHD is mandatory in these patients.

.

Disclosure: First author receives grants, speaker honoraria and travel support from Jazz pharmaceuticals.

14: Non-infectious Early Complications

P601 EFFICACY AND SAFETY OF NETUPITANT/PALONOSETRON COMBINATION (NEPA) AND LOW DOSE OF DEXAMETHASONE IN PREVENTING NAUSEA AND VOMITING IN PATIENTS UNDERGOING ALLOGENEIC STEM CELL TRANSPLANTATION

Alessandro Spina 1, Marina Aurora Urbano1, Claudia Schifone1, Francesca Merchionne1, Maria Laura Di Noi1, Gianluca Guaragna1, Erminia Rinaldi1, Giovanni Quintana1, Giuseppe Mele1, Domenico Pastore1

1Hematology and Transplant Unit, Brindisi, Italy

Background: Chemotherapy-induced nausea and vomiting (CINV) can be associated with reduced quality of life and meaningful complications in allogeneic stem cell transplantation (AlloSCT). Currently, a three-drug combination containing a neurokinin-1 receptor antagonist (NK1-RA), a 5-hydroxytryptamine-3(5-HT3)-RA, and corticosteroids in recommended as CINV prophylaxis regimen. Netupitant-palonosetron (NEPA) has been approved for highly and moderately emetogenic chemotherapy, but experience in alloSCT setting is limited. Furthermore dexamethasone (12-20 mg daily) is reommended by the antiemetic guidelines but its administration may be associated with a wide range of side effects, including serious infections in patients undergoing alloSCT. We retrospectively analyzed the efficacy and safety of NEPA and low dose of dexamethasone in alloSCT.

Methods: We analyzed 93 adult patients (acute myeloid leukemia=44, acute lymphoblastic leukemia=20, myelodysplastic syndrome=6 non-Hodgkin lymphoma=15, Hodgkin lymphoma= 3, Myelofibrosis= 5) who underwent alloSCT from matched related donor (n=37), matched unrelated donor (n=16), and haploidentical (n=40) donor. The median age was 50 years (median 18-72); the conditioning regimen was myeloablative (n=59) or reduced intensity(n=34). Stem cell source was peripheral blood stme cells in 95% of the patients. NEPA (300 mg of netupitant plus 0,5 mg of palonosetron) and low dose of dexamethasone (4 mg) was administered every other day of conditioning regimen, starting from the first day of conditioning regimen, with a maximum of 4 total doses. The observation period started with initiation of conditioning regimen and continued for 48 h after the last dose of chemotherapy.

Results: Response rate were 80% (75/93) for complete response (no emesis, no rescue medication), 78% (73/93) for complete control (complete response an no more than mild nausea); moreover, the percentage of patients that did not suffer any emetic episodes were 90% (84/93) and of patients that did not require a rescue therapy for controlling CINV were 88% (80/93). Moderate and severe episodes of nausea were reported in 13 patients (14%). Regarding the safety profiles, one patient presented headache, one patient experienced grade 1 abdominal pain and two had grade 1 constipation.

Conclusions: NEPA and low dose of dexamethasone, administered every other day, show to be very effective in preventing CINV in patients undergoing alloSCT with a good tolerability profile.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P602 CYTOKINE RELEASE SYNDROME AFTER HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION AND ITS IMPACT ON OVERALL SURVIVAL

Isabella Silva Pimentel Pittol 1, Camila Carminatti Isoppo2, André Dias Américo1, Germano Glauber de Medeiros Lima1, Eurides Leite da Rosa3, João Antônio Gonçalves Garreta Prats1, Hegta Tainá Rodrigues Figueroa1, Juliana Matos Pessoa1, Phillip Scheinberg1, Fauze Lutfe Ayoub1, Fabio Rodrigues Kerbauy1

1Beneficiencia Portuguesa de São Paulo, São Paulo, Brazil, 2Privado, Campinas, Brazil, 3Hospital 9 de Julho, São Paulo, Brazil

Background: Haploidentical hematopoietic stem cell transplantion (HSCT) significantly broadened the pool of patients with hematological malignancies for curative therapy. Symptoms such as fever, vasodilation, and end-organ damage characterize cytokine release syndrome (CRS). Present estimates indicate an incidence rate of 50% among haploidentical grafts, but the implications and outcomes of CRS still lack conclusive understanding. Within our institution, haploidentical donors rank as the second most prevalent donor type for allogeneic transplants. In this context, we share our institution’s experience with CRS following haploidentical transplantation.

Methods: We undertook a retrospective analysis at A Beneficência Portuguesa de Sao Paulo (BP) encompassing all haploidentical transplants conducted from November 2016 to December 2022. Our study involved the retrieval of data related to CRS occurrence, CRS grading based on the Common Terminology Criteria for Adverse Events, overall survival rates, causes of death, and the incidence of documented infections, encompassing both bacterial and fungal infections.

Results: During the study’s observation period, 103 haplo [ps1] transplants were conducted. All patients received standard graft-versus-host disease prophylaxis, involving post-transplant cyclophosphamide on days +3 and +4, followed by calcineurin inhibitors (CNI) and mycophenolate mofetil (MMF) starting at day +5. Hematopoietic stem cells were infused, sourced from either peripheral blood (91.3%) or bone marrow (8.7%). The median CD34+ hematopoietic stem cell (HSC) infusion per patient was 5.11*106/Kg (95% CI%, 4.52-6.15), with no significant difference between patients experiencing grade 2-4 CRS and those with grades 0-1 (5.46*106/Kg, 95% CI, 5.1-5.8 vs 5.57*106/Kg, 95% CI, 4.7-6.4, p = 0.8).

The median age of patients was 47 years (IQR 30-61), and a majority (61/103) underwent a reduced intensity conditioning regimen (RIC) for the treatment of mostly malignant diseases. CRS of any grade occurred in 85.4% of patients (95% CI, 77-91), with 92% of them receiving only antipyretics and supportive measures without specific therapies. CRS grade 2-4 (21.4%) independently correlated with death (HR = 3.2, 95% CI, 1.7-5.57, p < 0.001) after accounting for confounders such as age, HCT-CI, conditioning regimen and disease status (refer to complete multivariable analysis in Table 1). Survival rates were notably lower in this context, as illustrated in Figure 1, with 1-year estimates at 52% (95% CI, 44.5-68.3) for CRS grades 0-1 versus 34.7% (95% CI, 19.3-62.5) for higher grades.

Furthermore, severe CRS in the sample was not associated with documented bacterial infections (OR = 0.9, 95% CI, 0.3-3.14) or invasive fungal infections (OR = 2.28, 95% CI, 0.6-7.7).

Multivariate analysis of CRS grade 2 or greater, impact on death

HR (95% CI)

p value

Univariate analysis

CRS grade 2 or greater

2.87 (1.58-5.24)

< 0.001

Multivariate analysis

CRS grade 2 or greater

3.52 (1.87-6.62)

< 0.001

Age (as continuous value)

1.02 (1.0-1.04)

0.003

HCT-CI

0.97 (0.77-1.21)

0.79

Myeloablative Conditioning

1.97 (1.05-3.68)

0.03

Transplant not in complete remission

1.74 (0.99-3.06)

0.05

  1. HR = hazard ratio; 95% CI = confidence interval 95%.
  2. CRS, cytokine release syndrome; HCT-CI, hematopoietic cell transplantation comorbidity index
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (77)

Conclusions: Patients grappling with high-grade CRS confront a less favorable prognosis in our cohort independently of comorbidities and disease status. [ps1] The effectiveness of tocilizumab or dexamethasone in addressing haplo-CRS remains uncertain, lacking definitive support from randomized trials. Exploring interventions to mitigate haplo-CRS, such as the early initiation of CNI and MMF (adopted at our center as of February 2023), emerges as a potential avenue for improving outcomes to mitigate morbidity from more severe forms of CRS.

Disclosure: João Prats received speaker fees from Knight Pharmaceutical and MSD. André Américo received speaker fees from Janssen, Knight Pharmaceutical and MSD. Eurides Rosa received speaker fees from Aztra Zeneca and MSD. Fauze Ayub receites speaker fees from Janssen. Phillip Scheinberg has done scientific presentations for Novartis, Roche, Alexion, Janssen, AstraZeneca; Grants/Research Support: Alnylam, Pfizer; has received grants or research support from Alnylam, Pfizer; has received consultancy fees from Roche, Alexion, Pfizer, BioCryst, Novartis, Astellas; and has been a speaker for Novartis, Pfizer, Alexion. Isabella Pittol, Hegta Figueiroa, Fábio Kerbauy, Juliana Pessoa, Germano Lima and Camila Isoppo have no conflicts of interest to declare.

14: Non-infectious Early Complications

P603 EXPLORING THE SAFETY PROFILE OF NORETHINDRONE IN POST-MENARCHAL STEM CELL TRANSPLANT PATIENTS: ADDRESSING VOD CONCERNS AND CAUTIONARY CONSIDERATIONS

Mahvish Rahim1, Jodi Skiles1, Devin Dinora1, Jessica Harrison1, Ryanne Green1, Allie Carter1, April Rahrig 1

1Riley Hospital for Children, Indianapolis, United States

Background: Norethindrone (NE) can be used to manage menstrual bleeding in the setting of prolonged thrombocytopenia during stem cell transplant (SCT), however NE use has previously been associated with the development of veno-occlusive disease (VOD).

Methods: This is a retrospective study in which we reviewed all patients who underwent either allogenic or autologous SCT at Riley Hospital for Children between 2006 and 2022. Female patients who were started on NE for menstrual suppression at any point from start of preparative regimen to Day +30 post SCT were included in the detailed chart review. All patients who were assigned female sex at birth (AFAB) are reported as female in this study.

Results: During our study time-period, there were n=467 patients transplanted, of which 181 were female (38.8%). The incidence of VOD diagnosis in the entire cohort was 15.6% (73/467). The incidence of VOD diagnosis amongst male patients was 12.2% (35 of 286). Within the females who received NE, 4 of 18 (22.2%) were diagnosed with VOD compared to 34 of 163 (20.9%) females who did not receive NE (p=0.893). The median age of patients who received NE and were diagnosed with VOD (n=4, median age 21.5 years) was significantly older than subjects who received NE but were not diagnosed with VOD (n=14, median age 15.5 years, p=0.0003). Neither the starting dose nor the maximum dose of NE was statistically significant for increased incidence of diagnosis with VOD (p=0.525; p=0.431 respectively). All four of the patients who were diagnosed with VOD with NE exposure were classified per EBMT criteria as very severe VOD both at diagnosis as well as at the maximum severity of the disease.

Conclusions: Overall, we find that that there is no significant difference in the incidence of VOD diagnosis in female patients who received NE during SCT compared to female patients who did not receive NE. However, based on our findings it would be reasonable to avoid NE use in older female patients during the immediate peri-transplant period. To our knowledge, this is the first such investigation of NE as a potential risk factor for VOD since the Hagglund et.al publication in 1998. Although our patient cohort is small, our findings are not consistent with previously published data on VOD risk associated with NE use during SCT. NE use in post-menarchal female patients during the acute peri-transplant period may be safe.

Disclosure: Nothing to Declare.

14: Non-infectious Early Complications

P604 POST-TRANSPLANT DE NOVO ANTI-HLA DONOR SPECIFIC ANTIBODIES MAY CONTRIBUTE TO POOR GRAFT FUNCTION AFTER HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION

Xinyu Ji 1, Luxin Yang1,2, Xiaoyu Lai1,2, Yishan Ye1,2, Yibo Wu1,2, Shipei Xiang1, Yi Luo1,2, Lizhen Liu1,2

1The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China, 2Liangzhu Laboratory, Hangzhou, China

Background: Anti-HLA donor-specific antibodies (DSAs) are currently an important challenge in haploidentical hematopoietic stem cell transplantation (haplo-HSCT). While pre-formed DSAs have been associated with graft failure and poor graft function (PGF), newly developing (de novo) DSAs are rarely reported. De novo DSAs were early and more reported in solid organ transplantation, which were associated with higher incidence of rejection and worse survival. In haplo-HSCT. previous research showed that most de novo DSAs were detected 30 days after HSCT, and most mean fluorescence intensity (MFI) of which were below 2000. The effect of de novo DSAs on haplo-HSCT outcome is still unclear.

Methods: We present a case of a haplo-HSCT patient who developed de novo DSAs only 16 days after transplantation with the highest MFI of 7406.23, which caused poor graft function (PGF).

Results: A 39-year-old female with acute myelogenous leukemia received haplo-HSCT in the First Affiliated Hospital, Zhejiang University School of Medicine. DSA was tested negative before transplantation. Until day 16 after transplantation, her hematopoiesis had not recovered, with a white blood cell (WBC) count of 0.07×109/L and a platelet count of 3×109/L in peripheral blood. DSA was retested on day 16, which showed anti-HLA DRB1*09:01 DSA at a high MFI of 7406.23 and anti-HLA DQB1*03:03 DSA at MFI of 2164.04. Based on above findings of de novo DSAs, the patient received two plasma exchanges on day 21 and day 24, and intravenous immunoglobulin (40g/d) on day 25 to 26). After desensitization, the highest DSA MFI decreased to 1243.78. Neutrophil engraftment was achieved on day 27, while the platelet count remained below 20×109/L. A donor cell engraftment exam based on short tandem repeats (STR) on day 34 showed that 99.58% of donor-type cells. The minimal residual disease (MRD) and FLT3-ITD mutation were both negative. Then the patient was considered to suffer primary PGF. Recom binan t human throm bopoi etin injection was used from day 6, which was replaced by Eltrombopag from day 42. Mesenchymal stem cells (MSC, 1.0×106/kg) were infused once a week from day 63 for a total of four times. On day 105, 34mL of cryopreserved peripheral blood donor stem cells (1.0×106/kg CD34+cells and 3.76×107/kg CD3+ cells) were infused. On day 130, a WBC count of 2.77×109/L in patient’s peripheral blood was reached. On day 150, the bone marrow morphology showed a successful engraftment. In the following 2 years, the patient’s blood cell counts maintains normal. The most recent exam, 24 months after HSCT, showed a bone marrow remission in morphology and 99% donor-type cells in bone marrow.

Conclusions: Although de novo DSAs were not common after HSCT, it should be concerned when patients got PGF. Early DSA test and desensitization therapy could help achieve a successful engraftment.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P605 IMPACT OF EARLY CYCLOSPORINE INITIATION ON CYTOKINE RELEASE SYNDROME AND TRANSPLANT OUTCOMES IN PEDIATRIC T CELL REPLETE PERIPHERAL BLOOD HAPLO-IDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANT

Veerendra Patil 1, Pavan Kumar Boyella1, Pallavi Ladda1, Rakesh Pinninti1, Rohan Tewani1, Senthil Rajappa1

1Basavatarakam Indo-American Cancer Hospital and Research Institute, Hyderabad, India

Background: Haploidentical hematopoietic stem cell transplant (Haplo-HSCT) has gained popularity in recent past because of its comparable outcomes and ease of donor availability in the absence of HLA matched sibling donor. The preferred source of grafts in low and middle income countries like India is peripheral blood (PB) due to various logistic issues. Meanwhile the PB graft is shown to cause higher grade cytokine release syndrome (CRS) in haplo-HSCT with reported bad impact on transplant outcomes (Abboud 2021, Jayakumar 2020). But, with modifying the schedule of immunosuppression with respect to post transplant cyclophosphamide (PTCY), the frequency and severity of CRS were seen to decrease (Ruggeri 2020).

Methods: ALL patients less than 18 years with hematologic malignancy who underwent T cell replete haplo-HSCT with PB grafts at a single center in India were retrospectively analyzed. All patients received myeloablative conditioning with either 12 Gy TBI with Cyclophosphamide or thiotepa (10mg/Kg), treosulfan (36-42g/m2) and fludarabine (150 mg/m2) regime. Graft versus host disease (GVHD) prophylaxis included cyclosporine from day -1, Cyclophosphamide 50mg/Kg/day on D + 3 and D + 4 and oral Mycofenolate mofitil from D + 5 post HSCT. Patients with fever >38 degree post stem cell infusion till day +7 with negative infection workup and infusion reactions were considered as CRS. The grading of CRS was as per ASTCT consensus (Lee 2018). The data was analyzed for incidence and severity of CRS and GVHD, overall survival (OS), relapse free survival (RFS) and GHVD/relapse free survival (GRFS).

Results: A total of 27 patients with hematological malignancy were analyzed which included acute lymphoblastic leukemia (18/27), acute myeloid leukemia (7/27), mixed phenotypic leukemia (1/27) and juvenile myelomonocytic leukemia (2/27). The disease status before HSCT was CR-1, CR-2, CR-3 and active disease in 50%, 25%, 10.7% and 14.3% of patients respectively. A grade I CRS was noted in 60.7% of patients (17/27) and none had grade II or more. All patients engrafted with >95% donor chimerism at 100 days. The incidence of grade II-IV acute GVHD, grade III-IV acute GVHD and moderate to severe chronic GVHD were 28.6%, 7.1% and 7.6% respectively. The 3 year estimates of OS, RFS and GRFS were 78%, 82.5% and 74% respectively. The incidence and severity of acute or chronic GVHD was not influenced by CRS.

Conclusions: Our results suggest that initiation of cyclosporine before PB graft infusion would help in mitigating the severity of CRS and probably helps in increasing the OS and GRFS in pediatric haplo-HSCT. Peripheral blood graft is a valid option in haplo-HSCT in children with malignancy. Larger prospective studies would help in validating our outcomes.

Disclosure: I declare no conflict of interest.

14: Non-infectious Early Complications

P606 USE OF GRANULOCYTE COLONY-STIMULATING FACTORS IS FEASIBLE IN ALL DONOR SETTING WITHOUT INCREASING THE RISK OF ENGRAFTMENT SYNDROME

Maria Luisa Giannattasio 1, Linda Piccolo1, Andrea Cacace1, Davide Pio Abagnale1, Giuseppe Gaeta1, Francesco Grimaldi1, Simona Avilia1, Mara Memoli1, Lucia Ammirati2, Valentina Maglione2, Catello Califano2, Claudia Andretta3, Ermanno Badi3, Pasqualino Correale3, Patrizia Ricci4, Domenica Borzacchiello4, Marco Picardi1, Antonio Feliciello4, Antonio Leonardi3, Fabrizio Pane1, Giorgia Battipaglia1

1Federico II University of Naples, Naples, Italy, 2Onco-Hematology Unit, “A. Tortora” Hospital, Pagani, Italy, 3Federico II University of Naples, Apheresis Center, Naples, Italy, 4Federico II University of Naples, Manipulation Laboratory, Naples, Italy

Background: Granulocyte colony-stimulating factor (G-CSF) may favor a more rapid neutrophil engraftment after allogeneic hematopoietic stem cell transplantation (allo-HSCT), allowing shorter time of hospitalization. However, concerns for an increased risk of engraftment syndrome prompt in some cases to limits its use only to patients with high risk of graft failure.

Methods: We report a single-center experience with the use of G-CSF in patients undergoing allo-HSCT for hematologic malignancies from july 2020 to august 2023. When graft-versus-host disease (GVHD) prophylaxis incorporated either methotrexate or post-transplant cyclophosphamide, G-CSF was started the day after the last dose of these drugs was administered. Otherwise, it was started at neutropenia onset.

Results: Forty patients (15 males, 25 females) with a median age at transplant of 52 (range 25-71) years, underwent allo-HSCT for various hematological malignancies (AML [n=29], ALL [n=7], MDS [n=2], lymphoproliferative disease [n=2]). Peripheral blood was more frequently used as stem cell source (63%). Donors were HLA identical siblings for 14, unrelated for 18 (10/10 n=12, 9/10 n=6) and haploidentical for 8 patients. Thirty-three patients were transplanted in CR (CR1, n=24; CR2, n=8; >CR2, n=1), 7 with active disease. Conditioning regimen was myeloablative in 19 (48%), reduced-intensity in 14 (35%) and sequential in 7 (17%) patients. GVHD prophylaxis consisted in cyclosporine A alone (n=2) or with either methotrexate (n=13) or mycophenolate mofetil (n=25). Addition of antityhmocyte globulin or post-transplant cyclophosphamide was recorded in 27 and 12 patients, respectively.

Median duration of G-CSF treatment was 9 (range 1-20) days. All patients achieved neutrophil engraftment in a median time of 14 (range 9-23) days. All but one patient experienced platelet engraftment in a median time of 17 (range 10-44) days. Median duration of hospitalisation was 30 (range 20-56) days. No cases of engraftment syndrome were observed. Acute GVHD of all grades occurred in 9 patients, including 4 grade 3-4. Chronic GVHD occurred in 2 patients, one being extensive. Eight patients experienced disease relapse. Death occurred in 8 patients, including 3 due to GVHD, 4 due to disease relapse and 1 due to unknown causes. With a median follow-up of 21 (range 4-40) months, 1-year GRFS, PFS and OS were 57±8%, 70±8% and 79±7%.

Conclusions: In our experience, G-CSF is feasible in all donors setting and does not result in increased incidence of engraftment syndrome.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P607 THE UTILITY OF EXTRACORPOREAL ULTRASONOGRAPHY SCORE (HOKUS-10) IN PEDIATRIC SOS/VOD

Atsushi Narita 1, Hideki Muramatsu1, Ryo Maemura1, Daiki Yamashita1, Daichi Sajiki1, Yusuke Tsumura1, Ayako Yamamori1, Manabu Wakamatsu1, Kotaro Narita1, Shinsuke Kataoka1, Yoshiyuki Takahashi1

1Nagoya University Graduate School of Medicine, Nagoya, Japan

Background: Sinusoidal Obstruction Syndrome/Veno-Occlusive Disease (SOS/VOD) is a critical complication affecting 7-27% of pediatric patients undergoing hematopoietic stem cell transplantation. The Hokkaido ultrasonography (US)-based scoring system (HokUS-10), a diagnostic tool for SOS/VOD, shows promise for pre-symptomatic assessment in adults (Biol. Blood Marrow Transplant. 2018;24:1896–1900.). While potentially transformative for early detection methods, its effectiveness in children remains to be conclusively established.

Methods: A retrospective review of 118 patients undergoing hematopoietic cell transplantation at our department between 2020 and 2022 was conducted. Diagnosis and severity classification of SOS/VOD in pediatric cases were adhered to the European Society for Blood and Marrow Transplantation (EBMT) criteria. When SOS/VOD was suspected or diagnosed, US was performed. The HokUS-10 scoring system comprises 10 parameters, including hepatomegaly, gallbladder wall thickening, ascites, portal vein (PV) and paraumbilical vein (PUV) dilatation, and abnormal blood flow signals in the PV and PUV. The HokUS-10 cut-off for the diagnosis of SOS/VOD was set at 5 (maximum score, 13).

Results: Among the observed patients, 8 patients (7%) were diagnosed with SOS/VOD. Their median age at transplantation was 5 years (range: 3-11 years). Underlying diseases were neuroblastoma (n = 6), acute lymphocytic leukemia (n = 1), and chronic active EB virus infection (n = 1); seven patients had allogeneic transplants and one had an autologous peripheral blood stem cell transplant. Ascites effusion was present in 5 patients, and jaundice was present in 3 patients. The median duration from HSCT to the onset of SOS/VOD was 24.5 days (range: 17-217 days), and all patients were recieved defibrotide within 3 days of the diagnosis of SOS/VOD. The median score of the HokUS-10 in the SOS/VOD group was 5 (range, 3 to 8) at diagnosis. Four of the six patients had a positive HokUS-10 score (≥5 points), and all patients had severe/most severe disease. Positivity rates were notable for right lobe anterior-posterior diameter (5/6), ascites (6/6), and PUV diameter (4/6), while PV main diameter, PV main blood flow direction, and PV mean blood flow velocity were negative in all cases. We conducted additional analysis to examine the relationship between the initial HokUS-10 total score and its evolution following SOS/VOD treatment. Our findings revealed a significant improvement in scores at follow-up compared to those at the time of diagnosis (P = 0.035). Early intervention demonstrated efficacy in resolving SOS/VOD in all patients, achieving remission within a median period of 19 days (range: 5-36 days) from the initiation of defibrotide.

Conclusions: Our study indicated the efficacy of HokuUS-10 in improving diagnostic accuracy in pediatric patients with SOS/VOD. Further studies with a large number of pediatric patients are needed to validate more optimal evaluation criteria specific to children.

Clinical Trial Registry: not applicavle.

Disclosure: no disclosures.

14: Non-infectious Early Complications

P608 RETROSPECTIVE REVIEW OF TRANSPLANT AND TRANSFUSION OUTCOMES IN PATIENTS WITH PRE-TRANSPLANT RED CELL ALLOANTIBODIES

Samantha Drummond 1, Jennifer Laird2, David Irvine3

1Queen Elizabeth University Hosptial, Glasgow, United Kingdom, 2Scottish National Blood Transfusion Service, Glasgow, United Kingdom, 3Queen Elizabeth University Hospital, Glasgow, United Kingdom

Background: Red cell alloantibodies may form following a sensitising event such as transfusion or pregnancy. Patients who have formed an alloantibody are at increased risk of forming further antibodies. In the haematopoietic stem cell transplant setting, the incidence of red cell alloimmunisation is variable. Patients with non-ABO red cell antibodies are at higher risk of haemolytic anaemia post-transplant. In addition, patients with red cell alloantibodies may experience delays in transfusion due to difficulties in identifying compatible units.

The aim of this retrospective review is to determine the impact of pre-transplant red cell alloantibodies on allogenic haematopoietic stem cell transplant outcomes. The objectives are to identify whether the presence of red cell antibodies affects time to engraftment, transfusion requirements and mortality.

Methods: Electronic admission records (Trakcare (Intersystems) and Clinical Portal (Orion Health) were reviewed to identify patients admitted for an allogenic haematopoietic stem cell transplant between 1st January 2020 and December 31st2022 at the Queen Elizabeth University Hospital, Glasgow. Electronic patient records were used to determine clinical outcomes.

Results: Over the two-year period, 147 allogenic stem cell transplants were performed in 145 patients. The average age of transplant recipients was 46.7 years and 54% were male. Patient characteristics are summarised in table 1. Almost all patients (99%) required blood product support during admission.

Thirteen patients (9%) had an alloantibody identified. Of these patients, 46% (6 patients) had more than one alloantibody detected. A greater proportion of patients with alloantibodies experienced a transfusion reaction (23% vs 4%). In those without alloantibodies, the average number of packed red cell transfusions was 6.49 (0-76) and platelet transfusions 8.52 (0-153). In those with red cell antibodies this was 5.84 (0-18) and 4.46 (1-11) respectively. There were no instances of autoimmune haemolytic anaemia in either group. Fifteen percent (2 patients) of those with red cell alloantibodies were transfusion dependent at D100 compared to 23% (31 patients) without alloantibodies. The length of inpatient stay was similar in each group (33.2 and 33.7 days). A post-transplant neutrophil count of >0.5 was achieved on average at 15.29 days in those without red cell alloantibodies and in those with red cell alloantibodies at 12.1 days.

Patient characteristics

All recipients (n=147)

No alloantibody detected (n=134)

Allo-antibody detected (n=13)

Age (years)

46.7

46.5

50.4

Male (%)

54

57

30

ABO mismatch (%)

No

60

57

77

Major

24

26

8

Minor

13

13

8

Bidirectional

3

2

8

Stem cell source (%) PBSC

99

99

100

Bone marrow

1

1

Diagnosis (%)

AML

39

40

31

CML

4

4

ALL

17

19

Myelofibrosis

5

5

8

Other

15

11

46

Aplastic anaemia

6

7

Myelodysplastic syndrome

13

13

15

Alive (%)

71

69

77

Length of follow up (days)

847

848

823

Conclusions: In this retrospective review, there was no negative impact of red cell alloantibodies on engraftment, transfusion dependence at D100 or mortality. Patients with red cell alloantibodies appear more likely to experience a transfusion reaction. However, the small numbers in this review limited the opportunity for subgroup analysis. No patients experienced post-transplant autoimmune haemolytic anaemia during the follow-up period. Further work is required to determine whether specific alloantibodies are associated with greater risk of autoimmune haemolytic anaemia and adverse transplant outcomes.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P609 TWO GLOBAL PHASE 3 TRIALS OF THE EFFICACY AND SAFETY OF RAVULIZUMAB IN ADULT AND PEDIATRIC PATIENTS WITH THROMBOTIC MICROANGIOPATHY AFTER HEMATOPOIETIC STEM CELL TRANSPLANT

Carolyn Hahn 1, Elsa Konig1, Jonathan Monteleone1, Edward Wang1

1Alexion, AstraZeneca Rare Disease, Boston, United States

Background: Hematopoietic stem cell transplant-associated thrombotic microangiopathy (HSCT-TMA) is a post-transplant condition affecting 10%–35% of HSCT recipients and is associated with high morbidity and mortality. HSCT-TMA primarily manifests as an endotheliopathy that is mediated by complement system overactivation and elevated terminal complement activity. Ravulizumab is a long-acting complement C5 inhibitor which provides immediate, complete and sustained inhibition of the terminal complement pathway. Ravulizumab is approved for the treatment of patients with atypical hemolytic uremic syndrome (aHUS), among other indications, and has demonstrated efficacy in resolving complement-mediated TMA.

Methods: Two global phase 3 trials are in progress to assess the efficacy and safety of ravulizumab in patients with TMA presenting within 12 months of HSCT. NCT04543591 is a randomized, double-blind, placebo-controlled trial in adults and adolescents aged ≥ 12 years and NCT04557735 is a single-arm, open-label trial in pediatric patients aged ≥ 28 days to < 18 years. In both trials, HSCT-TMA is defined according to diagnostic criteria similar to the modified Jodele criteria published recently by a global panel of experts (Schoettler ML et al. Transplant Cell Ther 2023;29:151–63). For each trial, an open-label dose confirmation analysis was performed once ≥ 10 patients completed the Day 21 visit. For the adult/adolescent trial, this analysis was performed as a separate open-label stage (Stage 1). Ninety-four adult/adolescent patients are planned to be randomized to receive the confirmed ravulizumab dose or placebo in Stage 2. When ~50% of the planned Stage 2 patients have completed or withdrawn from the 26-week treatment period, a sample size re-estimation analysis will be performed. The pediatric trial will include 40 pediatric patients with HSCT-TMA treated with ravulizumab.

Results: The primary endpoint of both trials is TMA response, which is a composite measure of hematologic and renal parameters. The 26-week treatment period will then be followed by a 26-week follow-up period off ravulizumab. Overall survival will also be assessed at various time points.

Conclusions: These phase 3 trials assessing the efficacy and safety of ravulizumab in adults and pediatric patients with HSCT-TMA are part of a scientifically robust clinical development program aiming to provide substantial evidence to inform clinical practice and improve understanding on how to recognize, diagnose, treat, and manage these patients.

Clinical Trial Registry: NCT04543591, https://clinicaltrials.gov/.

Disclosure: CH, EK, JM and EW are employees of Alexion, AstraZeneca Rare Disease and are AstraZeneca shareholders.

14: Non-infectious Early Complications

P610 RITUXIMAB, BORTEZOMIB, PLASMA EXCHANGE COMBINED WITH IVIG FOR DESENSITIZATION DURING HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN PATIENTS WITH A POSITIVE DONOR-SPECIFIC ANTI-HLA ANTIBODY

Fang Liu 1, Cunbang Wang1, Yecheng Li1, Min Chen1, Jialin Duan1, Xinyu Wei1, Yongli Li1, Jing Wu1, Xiaofei Shen1, Ying Zhou1, Jinwei Li1, Yaoling Fu1, Lingling Yu1

1BOE Hospital, Chengdu, China

Background: Haploidentical stem cell transplantation (haplo-SCT) is increasing overtime for many patients(pts) in China in recent years. However, the presence of Donor specific HLA antibodies (DSA) in recipients increases the rist of engraftment failure. So far, no standardized desensitization method has been established.

Methods: From Feb 2022 to Sep 2023, 12 pts had positive DSA received desensitization before a haplo-SCT. The DSA level was determined by using the LUMINEX technique. The values were expressed in mean fluorescence intensity (MFI). DSA were considered positive if MFI value was ≥ 500 and strong positive if the MFI value was ≥ 10000. The desensitization treatment included Rituximab 375 mg/m2 D1, 8, 15, 22 Bortezomib 1.3 mg/m2 D1, 4, 7, 11 and Plasma-Exchange followed by high dose intravenous polyvalent immunoglobulins(IVIg) (Figure). DSA level was reevaluated during desensitization, before starting the conditioning regimen and the day before stem cell infusion.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (78)

Results: The median age of the pts was 37 years (range, 14-50), 10 were females and 2 males. Diagnosis was acute myeloid leukemia in 5 pts, myelodysplastic syndrome in 2 pts, acute lymphocytic leukemia in 3 pts and severe aplastic anemia in 2 pts. 10 donors were from direct family members and 2 from collateral relatives. Donor-recipient sex match: F/F:4; M/F:6; M/M:2. The median MFI value before desensitization was 8600 (range, 2000-22000). Most pt (7) had DSAs against HLA class I antigens.10/12 pts decreased DSA levels to less than 5000 MFI and 2 pts still more than 5000 MFI, median MFI value after desensitization was 2800 (range, <500-12000). 11 pts engrafted but one pt experienced primary engraft failure due to persistent strong positive DSA above 10000 MFI. All the 11 engrafted pts did not rebound with a persistent full chimerism.

Conclusions: Our study shows that desensitization with Rituximab, Bortezomib, plasma exchange and IVIg combined regimen can effectively decrease DSA and improve engraftment after haplo-SCT. Further and larger scale studies are needed to better explore the best methods of successful desensitization.

Clinical Trial Registry: no.

Disclosure: no.

14: Non-infectious Early Complications

P611 PREDICTIVE FACTORS OF GRAFT FAILURE IN ADULT AND PAEDIATRIC PATIENTS WITH MALIGNANT AND NON-MALIGNANT DISEASE UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANT (HSCT)

Pietro Merli 1, Régis Peffault de Latour2,3, Emmanuel Monnet4, Malin Löfqvist4, Franco Locatelli1

1IRCCS, Bambino Gesù Children’s Hospital, Catholic University of the Sacred Heart, Rome, Italy, 2French Reference Center for Aplastic Anemia and PNH Hematology-Bone Marrow Transplantation, Hôpital Saint-Louis AP-HP, Paris, France, 3Université Paris Diderot, Paris, France, 4Swedish Orphan Biovitrum (Sobi), Basel, Switzerland

Background: This exploratory clinical study enrolled adult and paediatric patients undergoing HSCT for non-malignant or malignant conditions to investigate the utility of circulating chemokine C-X-C motif ligand 9 (CXCL9), a specific biomarker of interferon gamma (IFNγ) activity, in predicting graft failure (GF). The association of specific risk-factor profiles with GF was also investigated.

Methods: This prospective, single arm, non-interventional, observational study included patients receiving allogeneic HSCT who had pre-defined risk factors for GF (NCT04494061). All investigated outcomes were exploratory. Data from hospital records and blood samples were collected 7 days before HSCT, at HSCT, at days 1, 3, 5, 9, 13, 17, 21, 28, 31 and 37 post-HSCT, and at the time of suspected GF up to day 100. Serum CXCL9 levels were analysed alongside standard laboratory assessments measuring engraftment. Descriptive statistics are presented.

Results: Overall, 86/106 enrolled patients received HSCT before the study was prematurely terminated for non-safety-related reasons. The patient population was predominantly male (60.5%) with a median (range) age of 19 (2–73) years. A similar proportion of adult (≥18 years, 52.3%) and paediatric (<18 years, 47.7%) patients participated. Overall, 36.0% of patients had underlying non-malignant disease and 64.0% had malignant disease, of which aplastic anaemia (48.4%) and acute myeloid leukaemia (32.7%) were the most common, respectively. GF occurred in 5/86 (5.8%) patients. No cases of secondary GF were observed, which may be due to low patient numbers and short follow-up. Most patients experiencing GF had underlying malignant disease (4/5; 80.0%). Peak CXCL9 levels were elevated post-transplant in all patients who experienced GF (median, 1487 ng/L; range, 734–5753 ng/L) and in some patients who did not experience GF (median, 416 ng/L; range, 40–6628 ng/L; Figure). Early study termination and low patient numbers prevent strong conclusions on the predictive characteristics of CXCL9, but the available data suggest that CXCL9 alone is not sufficient to accurately predict GF. A high rate of GF was observed with T-cell depleted or double umbilical cord blood (UCB) grafts (21.1% [4/19]) and mismatched unrelated donors (MMUD; 16.0% [4/25]). Unexpectedly, a haploidentical donor was associated with a comparatively low rate of GF (2.5% [1/ 40]). The highest GF rates were observed in patients with a combination of known risk factors for GF, particularly those with both MMUD and T-cell depleted or double UCB transplants (GF rate, 60.0% [3/5]; Table). However, these results should be interpreted with caution due to low patient numbers. GF risk was similar in adult and paediatric patients.

Single risk factors

GF risk factor

Patients with GF, n

Patients without GF, n

GF rate, %

Haploidentical donor (T-depleted AND T-replete)

N=40

1

39

2.5

RIC

N=41

2

39

4.9

PBSC

N=45

3

42

6.7

MMUD OR haploidentical donor

N=65

5

60

7.7

MAC

N=32

3

29

9.4

T-cell depleted graft

N=17

2

15

11.8

CXCL9 ≥734 pg/mLa

N=34

5

29

14.7

MMUD

N=25

4

21

16.0

CXCL9 ≥1010 pg/mLa

N=22

4

18

18.2

T-cell depleted OR double UCB graft

N=19

4

15

21.1

UCB

N=7

2

5

28.6

Risk factor combinations

Risk factor combination

Patients with GF, n

Patients without GF, n

GF rate, %

MMUD OR haploidentical donor AND T-cell depleted OR double UCB graft

N=19

4

15

21.1

MMUD AND T-cell depleted OR double UCB graft

N=5

3

2

60.0

  1. aCXCL9 values used were the highest CXCL9 values observed post-HSCT for individual patients.
  2. CXCL9, chemokine C-X-C motif ligand 9; GF, graft failure; MAC, myeloablative conditioning; MMUD, mismatched unrelated donor; PBSC, peripheral blood stem cell; RIC, reduced intensity conditioning; UCB, umbilical cord blood.
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (79)

Conclusions: GF rates in this prospective, observational study of high-risk patients undergoing HSCT were lower than expected, possibly due to clinical improvements in managing transplantation and limited follow-up duration. Elevated IFNγ activity was evident during GF, but the limited data collected did not support that elevated IFNγ activity is a robust predictor of GF risk. However, consistent with prior literature, our findings provide preliminary evidence that patients with a combination of risk factors for GF may be at higher risk of GF and could benefit from preventative treatment approaches, such as IFNγ blockade.

Clinical Trial Registry: ClinicalTrials.gov NCT04494061.

Disclosure: Pietro Merli is a consultant for Sobi and Jazz Pharmaceutical. Régis Peffault de Latour is a consultant to Alexion, Amgen, MSD, Novartis and Pfizer, and has received research grants from Alexion, Amgen, Novartis and Pfizer. Emmanuel Monnet and Malin Löfqvist are employees and shareholders of Swedish Orphan Biovitrum (Sobi). Franco Locatelli discloses honoraria from Amgen, Novartis and Sobi, and consulting fees from Amgen. This study was funded by Sobi.

14: Non-infectious Early Complications

P612 EARLY LOW DOSE CYCLOSPORINE AND MYCOPHENOLATE REDUCE THE RISK OF CYTOKINE RELEASE SYNDROME AFTER HLA-HAPLOIDENTICAL PERIPHERAL BLOOD STEM CELL TRANSPLANTATION WITH PTCY BASED GVHD PROPHYLAXIS

Jan Vydra 1, Ludmila Nováková1, Veronika Válková1, Markéta Šťastná Marková1, Mariana Koubová1, Anna Dobrovolná1, Barbora Čemusová1, Antonín Vítek1, Petr Cetkovský1

1Institute of Hematology and Blood Transfusion, Prague, Czech Republic

Background: T cell replete allogeneic hematopoietic stem cell transplantation (HSCT) from haploidentical donors (HAPLO) using PBSC grafts is associated with a significant risk of severe cytokine release syndrome (CRS) during first days after graft infusion. In order to decrease the risk of CRS, we use a modified GvHD prophylaxis protocol with low-dose cyclosporine at 1 mg/kg from day 0 increased to 3 mg/kg on day 6, mycophenolate 15 mg/kg BID from day 0 and cyclophosphamide 50 mg/kg on days +3 and +5. We report the incidence of CRS and transplant outcomes in a cohort of patients who received haplo HSCT at our center using this approach.

Methods: Patients who received haplo HSCT at Institute of Hematology and Blood Transfusion in Prague after 2017 were included in the analysis. Patient and donor characteristics and main outcomes were retrieved from our transplant database. Patient charts, notes and electronic medical records were reviewed for signs and symptoms of CRS. Summary statistics, Kaplan-Meier and cummulative incidence estimates were calculated for main transplant outcomes.

Results: 96 patients were included in the analysis. Patient characteristics are summarised in Table 1. Forty four (45.8%) patients experienced possible CRS grade I - at least one day of fever ≥ 38°C between days 0 and 5 after HSCT. Patients with fever received supportive care with antipyretics and empirical antibiotic therapy. Importantly, no patient developed grade ≥ 2 CRS and no patient needed specific therapy for CRS (e.g. tocilizumab or dexamethasone). OS at two years was 71.9% (95%CI 63% - 83%), NRM 9%, relapse rate 20%. The incidence of acute GVHD grade 2-4 was 35%, 50% of patients developed chronic GVHD, which was mild in 33% and moderate/severe in 17%.

Table 1 - Patient characteristics

Age – median, range

47.5 [18.0 - 68.0]

Diagnosis

Acute myeloid leukaemia

38 (39.6%)

Myelodysplastic syndrome

21 (21.9%)

Acute lymphoblastic leukaemia

6 (6.25%)

Lymphoma and CLL

15 (15.6%)

Other

16 (16.6%)

CMV serology

neg.

30 (31.2%)

pos.

66 (68.7%)

Graft Source: PBPC

96 (100%)

Conditioning regimen:

Myeloablative (Bu/Flu, Flu/12Gy TBI)

55 (57.3%)

Reduced intensity (Flu/Mel, Flu/Cy/2Gy TBI)

41 (42.7%)

CD34 cell dose (106/kg) – median, range

6.46 [2.24 - 16.7]

CD3 cell dose (106/kg) – median, range

268 [67.1 - 673]

Conclusions: Early low dose cyclosporine with mycophenolate mofetil are effective in preventing CRS after T cell replete haplo-HSCT using PBSC grafts. Main limitation of the study is that is is retrospective and based on a chart review. Prospective randomized comparison with standard ptCy/CsA/MMF schedule is needed to confirm these results.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P613 INCIDENCE, RISK FACTORS, AND OUTCOMES OF TRANSPLANT-ASSOCIATED THROMBOTIC MICROANGIOPATHY IN PEDIATRIC PATIENTS AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION

Kyung-Nam Koh 1, Su Hyun Yun1, Eun Seok Choi1, Sung Han Kang1, Hyery Kim1, Ho Joon Im1

1Asan Medical Center Children’s Hospital, Seoul, Korea, Republic of

Background: The aim of this study was to examine the frequency and survival rates of transplant-associated thrombotic microangiopathy (TA-TMA) in pediatric patients who have undergone allogeneic hematopoietic cell transplantation (HCT). Additionally, the study aimed to identify risk factors for TA-TMA, which could help in starting treatments early and possibly improve the outcomes for these patients.

Methods: A total of 173 consecutive pediatric and adolescent patients who underwent HCT at Asan Medical Center Children’s Hospital from May 2018 to May 2023 were eligible to participate in this prospective study. Regression analyses were performed to identify risk variables of TA-TMA. The predictive power of a time-dependent receiver operating characteristic curve of possible laboratory variables calculated. Gray’s competing risk analysis applied to calculate cumulative incidence, non-relapse mortality (NRM) of TA-TMA. Overall survival (OS) was measured by the Kaplan–Meier method.

Results: The cumulative incidence of TA-TMA 1-year (yr) and 3-yr following HCT is 6.5% (95% CI), OS rate 1-yr and 3-yr following HCT was found to be 63.6% in the group with TMA, compared with 98.8% and 83.5% in the group without TMA (p = .074) and the NRM rate 1-yr and 3-yr following HCT was found to be 18.1 % compared with 6.1% (p = .165). As a clinical risk factors, SOS (unadjusted HR, 5.80, p = .048), IPA (unadjusted HR, 7.60, p = .002) were associated with TMA occurrence. Laboratory risk variables tested from d0 to d182 following allogeneic HCT, BNP elevation ≥ 400 pg/mL (unadjusted HR, 10.70, p = .001), LDH ≥ 2ULN (unadjusted HR, p = .009), Cystatin-C GFR decreased ≥ 1/2 × baseline (unadjusted HR, 8.41, p = .002) and rUPCR ≥ 1mg/mg (unadjusted HR, p = .005) were associated with TMA occurrence. SOS (adjusted HR (aHR), p = .002), IPA (aHR, 51.7, p = .008), BNP elevation ≥ 400 pg/mL (aHR, 34.7, p = .033) and Cystatin-C GFR decreased ≥ 1/2 × baseline (aHR, 23.5, p = .048) were associated with TMA occurrence in multivariable analysis. An increase in BNP levels above 400 pg/mL was observed within the interquartile (IQR) range of -95d to +8d, a decrease in cystatin-C GFR occur in range of -120d to 9d, a LDH more than two-fold was observed between -70d and -8d and elevation of rUPCR by more than 1 mg/mg occurs between the periods -74d and -14d from the diagnosis of TMA.

Conclusions: In conclusion, our data show a poor clinical outcome of TA-TMA following allogeneic HCT, represented by OS and NRM. SOS, IPA as clinical variables, BNP ≥ 400 pg/mL, and Cystatin-C GFR decreased ≥ 1/2 × baseline from d0 to d182, as laboratory risk factors found to have significant association with TMA occurrence. In order to identify prognostic biomarkers and risk factors which might be employed to improve the clinical outcome of TA-TMA in a standardized and consistent manner, a multicenter study needs to be performed.

Disclosure: I have no conflict of interest to disclose.

14: Non-infectious Early Complications

P614 STEM CELL MOBILIZATION EFFECTS ON HEART SIZES AND FUNCTION

Domas Vaitiekus 1, Ignas Gaidamavicius1, Audrone Vaitiekiene1, Migle Kulboke1, Monika Bieseviciene1, Benas Kireilis1, Ruta Dambrauskiene1, Milda Rudzianskiene1, Antanas Jankauskas1, Elona Juozaityte1, Dietger Niederwieser1, Jolanta Justina Vaskelyte1, Gintare Sakalyte1, Rolandas Gerbutavicius1

1Lithuanian University of Health Sciences, Kaunas, Lithuania

Background: Hematopoietic stem cell transplantation (HSCT) can cause cardiac toxicity of different grade. In this study we aimed to evaluate the impact of mobilization procedure for autologous HSCT process on left and right ventricle sizes and systolic function.

Methods: Data of 35 patients undergoing autologous HSCT at the Department of Oncology and Hematology in the Hospital of Lithuanian University of Health Sciences Kaunas Clinics from October 2021 till Feb 2023 were analyzed. Bioethics approvement for prospective study was obtained (No BE-2-96). HSCT was performed for various reasons – mostly multiple myeloma, also mantle cell lymphoma, Hodgkin lymphoma, diffuse large B cell lymphoma, Ewing sarcoma. All patients underwent hematopoietic stem cell mobilization with chemotherapy and filgrastim 10 µg/kg/d. Echocardiography was performed before enrolling to the transplantation process and after mobilization before the conditioning regimen. Left ventricular end diastolic diameter (LVEDD), LVEDD index according to body surface area (BSA) (LVEDDi), left ventricular ejection fraction (LV EF), left ventricular global longitudinal strain (GLS), right ventricle (RV) end diastolic diameter (RVEDD, tricuspid annular systolic velocity (S’) using tissue doppler and tricuspid annular plane systolic excursion (TAPSE) were measured before and after mobilization and compared.

Statistical analysis was performed using SPSS statistics 20. Qualitative data is presented as an absolute value (N) and percentage (%), quantitative parameters are given as average (m ± standard deviation). We used paired- samples T test to compare quantitative parameters. Statistically significant difference was considered when p<0.05.

Results: Out of 35 patients there were 18 men (51,4%) and 17 women (48,6%). Mean age was 57,4±13,15 years, ranging from 18 to 74. 26 (74,3%) patients had multiple myeloma, 4 (11,4%) mantle cell lymphoma, 2 (5,7%) Hodgkin ‘s lymphoma, 2 (5,7%) PCNS diffuse large B cell lymphoma, 1 (2,9%) Ewing sarcoma.

Statistically significant difference was observed in the change of RV function. S‘ and TAPSE slightly decreased. Changes of echocardiographic parameters are presented in Table 1.

Echocardiographic values

Before mobilization

After mobilization

p

LVEDD, mm

46,03±3,426

45,93±4,548

0,888

LVEDDi, mm

24,82±2,699

24,83±2,514

0,967

LV EF, %

60,72±7,241

61,38±6,844

0,667

GLS, %

-17,57±3,266

-18,07±3,402

0,309

RVEDD, mm

35,66±3,85

34,49±4,395

0,07

S‘, cm/s

14,04±2,716

12,67±3,125

0,007

TAPSE, mm

22,26±2,944

20,91±3,425

0,019

Table 1. The change of echocardiographic parameters before and after mobilization procedure.

Conclusions: Mobilization procedure in patients undergoing autologous HSCT is associated with reduced RV systolic function. S‘ and TAPSE might be used for early cardiotoxicity evaluation in HSCT patients.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P615 EASIX SCORE AS INDEPENDENT PREDICTOR FOR OVERALL SURVIVAL IN PATIENTS WITH ACUTE LEUKEMIA AND MDS UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION

Penka Ganeva1, Andriyana Bankova 1, Georgi Vasilev1, Krasen Venkov1, Kameliya Milcheva1, Victoria Yankova1, Margarita Guenova1, Georgi Mihaylov1

1National Specialized Hospital for Hematological Diseases, Sofia, Bulgaria

Background: Prognostic models are crucial for the assessment of hematopoietic cell transplantation (HCT) recipients, as they help transplant physicians to make treatment decisions by using objective rather than subjective tools. Recently, the endothelial activation and stress index (EASIX), which assesses endothelial dysfunction based on LDH, creatinine and platelet count, was shown to independently predict clinical outcomes in both recipients of autologous and allogeneic HCT. Here, we report on the predictive value of EASIX in allo-HCT recipients and compare survival models and survival predictions based on two different cut-off values.

Methods: Clinical and laboratory data were retrospectively collected from patients with AML, MDS and ALL who underwent allo-HCT from 01/2021 - 06/2023. EASIX was calculated for all patients before start of the conditioning by standardized formula: LDH × Creatinine / platelet count. Receiver operating characteristic (ROC) curve and Cox regression were applied to assess the prognostic potential of EASIX by the area under the curve (AUC) and Akaike Information Criterion (AIC) was used to establish an optimal cut-off. Multivariate analysis (MVA) using Cox regression and аccelerated failure time models were used to assess the effect of EASIX on overall survival (OS) while adjusting for the confounding effect of other covariates of interest.

Results: A total of 65 patients were included in the analysis. Median age was 42 years, majority of patients were male (67.7%), and underwent allo-HCT to treat myeloid disease (44/65, 67,7%). In total, 43 patients (66%) were transplanted in CR ≥ 1, whereas 22 (34%) had active/stable disease at time of allo-HCT. Conditioning intensity was MAC in 34, RIC in 18 and FLAMSA-RIC in 13 of the patients. Donors were MRD (n=15), MUD (n=22), haploidentical (n=26) and MMUD (n=2). At timepoint of final analysis, 58.5% (n=38) of the patients were still alive vs 41.5% (n=27) who died with median time from allo-HCT to dead 18 months. A ROC curve analysis was applied with an estimation of the AUC for EASIX 0.72, p=0.003 (Fig. 1). Based on Youden`s Index two potential cut-off values were identified as optimal: (1) log2-EASIX=0.021 with 81% sensitivity and 60% specificity and (2) log2-EASIX=1.7 with 37% sensitivity and >90% specificity, respectively. By using AIC for between-model comparison we found that cut-off for log2-EASIX=1.7 better predicts survival. Patients with log2-EASIX ≥ 1.7 had significantly lower OS (p= 0.005). Nevertheless, further determination of optimal cut-off values for the EASIX might necessitate larger patient cohort for better validation. In MVA adjusted for gender, donor type, disease status, conditioning intensity and EASIX, we found that EASIX was strong independent predictor for OS [HR (95%Cl): 1.407 (1.167, 1,696); p < 0.001].

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (80)

Conclusions: Our results highlight the importance of EASIX as independent predictor for OS in patients with acute leukemia and MDS undergoing allo-HCT. However, further optimization of cut-off values is necessary in order to improve both sensitivity and specificity of the score.

Disclosure: No conflict of interest.

14: Non-infectious Early Complications

P616 EARLY CICLOSPORINE TO EVEROLIMUS CONVERSION POST HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR PATIENT WITH CALCINEURIN INHIBITORS INDUCED TOXICITIES

Benjamin Bouchacourt 1, Raynier Devillier1,2, Didier Blaise1,2, Thomas Pagliardini1, Sabine Furst1, Samia Harbi1, Faezeh Legrand1, Charlotte Nykolyszyn1, Federico Pagnussat1, Pierre Jean Weiller1, Claude Lemarie1,3, Boris Calmels1,3, Christian Chabannon1,3,2

1Paoli Calmettes Institute, Marseille, France, 2Aix Marseille Univ, CNRS, INSERM, CRCM, Marseille, France, 3CIC Biothérapies : INSERM CBT-1409, Marseille, France

Background: Calcineurin inhibitor (CNI) based graft versus host disease (GVHD) prophylaxis is the main standard in hematopoietic stem cell transplantation (HSCT). However, CNIs are responsible for substantial side effects such as kidney failure, thrombotic microangiopathy (TMA) or neurotoxicity. In this study, we aimed to evaluate 8 patients who transitioned early from Ciclosporine (CSA) to Everolimus, owing to nephrotoxicity or due to transplant-associated thrombotic microangiopathy (TA-TMA).

Methods: This retrospective observational study selected consecutive patient ≥ 18 years old who were converted from CSA to Everolimus before day 100 post HSCT, at Paoli Calmettes Institute (Marseille) in 2022. CSA imputability for kidney injury was assessed after eliminating other causes, including pre renal and tubular injury. TA-TMA were diagnosed using O-TMA criteria by Cho et al. At day one after CSA was stopped, patients received IV corticotherapy 1 mg/kg, and Everolimus 1 mg BID (0.5 mg BID in case of Voriconazole co-medication) was started when residual CSA blood concentrations went under 100 ng/ml. Corticotherapy was stopped after achieving the Everolimus target trough level of 6-10 ug/L.

Results: Eight patient met the inclusion criteria. All patient received a peripheral blood stem cell transplant, and a GVHD prophylaxis with CSA, Mycophenolate Mofetil and post transplant Cyclophosphamide. Donor was an HLA haploidentical related donor for 6 of them and an HLA missmatch 9/10 unrelated donor for the 2 remaining.

The median interval between HSCT and Everolimus initiation was 35 days (range 17-72). The reasons for CSA conversion was nephrotoxicity for 6 patient, and TA-TMA for 2 patient (One with acute kidney failure). Median serum level creatinine at transitioning was 2.1 times baseline (range 0.9-4.3). All patients achieved a complete response after CSA discontinuation and Everolimus initiation, with a median time of 7.5 days (range 2-34).

Almost all patient (7/8) have suffered of significant GVHD after the replacement. Six patients (75%) presented ≥ G2 acute GVHD and 4 (50%) developed moderate chronic GVHD, respectively with a median time of 59.5 (range 10-119), and 168 (range 119-208) days since CSA to Everolimus converting. Five patients suffered from severe Everolimus imputable toxicity, 3 diarrhea grade 3, and 2 gastrointestinal hemorrhage (gastro-intestinal GVH was excluded), leading to Everolimus replacement by another immunosuppressive drug.

With a median follow-up of 18 months since HSCT, 2 patients relapsed (1 plasma cell leukemia and 1 MDS/MPN unclassifiable). One patient died from carbapenemase septic shock.

Full size table

.

Conclusions: CSA to Everolimus conversion is an effective strategy for CNI induced complication, but the incidence of GVHD and gastrointestinal toxicity is much too high, therefore we do not recommend this approach. Other promising options must be explored, such as CNI to Ruxolitinib transitioning.

Disclosure: No disclosure.

14: Non-infectious Early Complications

P617 IMMUNE-MEDIATED CYTOPENIA IN PEDIATRIC PATIENTS FOLLOWING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: ON BEHALF OF THE RBC DISORDER WORKING PARTY OF THE KOREAN SOCIETY OF HEMATOLOGY

Hyoung Soo Choi 1, Jeong-A Park2, Hee Won Chueh3, Hee-Jo Baek4, Hoon Kook4

1Seoul National University College of Medicine, Seoul National University Bundang Hospital, Seongnam, Korea, Republic of, 2Inha University College of Medicine, Inha University Hospital, Incheon, Korea, Republic of, 3Inje University Haeundae Paik Hospital, Busan, Korea, Republic of, 4Chonnam National University Medical School, Chonnam National University Hwasun Hospital, Gwangju, Korea, Republic of

Background: Immune-mediated cytopenia (IMC) is a significant complication that can occur after allogeneic hematopoietic stem cell transplantation (HSCT), posing significant risk of morbidity and mortality. It encompasses a spectrum of conditions including autoimmune hemolytic anemia (AIHA), thrombocytopenia (ITP), and neutropenia, either alone or in combination (e.g., as Evans syndrome). The pathogenesis of IMC is not well-understood, and its management can be very challenging. This study aims to present a case series of IMC following allogeneic HSCT in pediatric and adolescent patients.

Methods: This retrospective study, conducted by the RBC Disorder Working Party of the Korean Society of Hematology, included cases of patients aged under 18 who underwent allogeneic HSCT at four transplantation centers in Korea from 2000 to 2022. Medical records were analyzed to assess clinical characteristics and treatment outcomes.

Results: Among the 332 cases of allogeneic HSCT, 6 cases (1.8%) developed IMC. It was observed in 2 patients with severe aplastic anemia (SAA), 2 with acute lymphoblastic leukemia, 1 with T-lymphoblastic lymphoma (T-LBL), and 1 with therapy-related myelodysplastic syndrome. The median age at transplant for the 6 patients was 7.1 years, with a range of 0.8 to 17.5 years. The IMC diagnosed included 3 cases of AIHA and 3 cases of Evans syndrome. The median time from HSCT to the development of IMC was 59 days, ranging from 21 to 106 days. The stem cell sources were peripheral blood stem cells in 4 patients (2 with HLA- matched sibling, 1 with HLA-haploidentical mother, and 1 with HLA-matched unrelated donor). Additionally, 1 patient received unrelated HLA-matched bone marrow, and 1 received umbilical cord blood with a 4/6 locus HLA-match. Two AIHA patients (1 SAA and 1 T-LBL) responded to the first line therapy of steroid and intravenous immunoglobulin and achieved complete response. The remaining 4 patients, consisting of 1 with AIHA and 3 with Evans syndrome, required secondary or subsequent treatments, including rituximab and sirolimus. Among them, 2 patients showed a complete response, and 2 showed a partial response.

Conclusions: IMC is a rare complication that occurs following allogeneic HSCT. While it can be managed with immunosuppressive agents for the prevention and treatment of GVHD, there are cases where it does not respond well to various treatment options, making it a challenging condition. Close monitoring, early recognition, and tailored therapeutic interventions are necessary, considering the identification of risk factors. Continued efforts are required to implement effective treatment strategies, including the utilization of sirolimus and other novel agents for refractory cases.

Disclosure: Nothing to declair.

14: Non-infectious Early Complications

P618 ENGRAFTMENT SYNDROME IN PATIENTS UNDERGOING AUTOLOGOUS STEM CELL TRANSPLANTATION FOR MULTIPLE MYELOMA: A SINGLE-CENTER DATA ANALYSIS OF CLINICAL FEATURES AND RISK FACTORS

Shanshan Liu 1, Luxin Yang2, Lizhen Liu2, Yan Gao1, Xianqi Feng1, He Huang2, Yi Luo2

1The Affiliated Hospital of Qingdao University, Shandong, China, 2The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China

Background: Engraftment Syndrome (ES) was reported to appear in 31~59% of patients who underwent allo or auto hematopoietic stem cell transplantation. With the increasing use of ASCT in multiple myeloma treatment, ES has become more prevalent among ASCT recipients. Although awareness of ES has grown, ongoing discussions persist regarding its diagnostic criteria, pathogenesis, and high-risk factors. This study aims to better improving diagnostic criteria and analyze potential risk factors contributing to ES.

Methods: We collected data from 131 consecutive multiple myeloma patients undergoing ASCT at the First Affiliated Hospital of Zhejiang University from January 2019 to July 30, 2022.

Results: A total of 46 (36.5%) patients were diagnosed ES. Nearly all patients diagnosed with ES exhibited fever, diarrhea, nausea, vomiting, and weight gain exceeding 2.5%. Among them, 30 patients (65.2%) displayed skin rashes, while 4 patients (8.7%) exhibited lung infiltrative changes, including 2 (4.3%) with hypoxemia. Additionally, 12 patients (26.1%) presented with hepatic dysfunction, 13 patients (28.3%) displayed renal dysfunction, and 2 patients (4.3%) demonstrated encephalopathy symptoms.

The management of ES depending on the patient’s symptoms, either prednisone at 15-20 mg/day or dexamethasone at 5-10 mg qd or qod was prescribed. Patients with CD34 reinfusion counts exceeding 10 × 10^6/kg avoided granulocyte colony-stimulating factor (G-CSF) application. Infection-prone patients were given broad-spectrum antibiotics. Among the 126 patients, none required ICU admission or advanced life support.

In the single-factor analysis, no difference was found in age, gender diagnostic subtypes, prior cyclophosphamide (CTX) application, disease status during collection and transplantation and treatment lines. And Application of daratumumab and carfilzomib, mobilization with and higher CD34 reinfusion count were not associated with higher risk of ES. The duration of neutrophil engraftment, onset of fever, CRP levels, and the presence of microbiological evidence of infection were also undifferentiated. Otherwise the combination of bortezomib in the preconditioning regimen (27 cases, 58.7% vs 30 cases, 37.5%, P=0.034) was identified as high-risk factor. The factor also showed statistically significant differences in the multivariate analysis.

Conclusions: From this data, the combination of bortezomib in the preconditioning regimen were identified as independent high-risk factors for ES.

Disclosure: The authors declare that they have no competing interests.

14: Non-infectious Early Complications

P619 DONOR SPECIFIC ANTI-HLA ANTIBODIES IN HAPLOIDENTICAL PERIPHERAL STEM CELL TRANSPLANTATION: SINGLE CENTER EXPERIENCE

Alessandro Spina 1, Marina Aurora Urbano1, Claudia Schifone1, Maria Laura Di Noi1, Francesca Merchionne1, Maria Rosaria Coppi1, Maria Patrizia D’Errico1, Maria Antonietta Miccoli2, Domenico Pastore1

1Hematology and Transplant Unit, Brindisi, Italy, 2Division of Transfusion Unit, Brindisi, Italy

Background: In the setting of haploidentical stem cells transplantation, the presence of antibodies directed against donor-specific HLA allele(s) or antigen(s) (DSA) represents a barrier for stem cell engraftment, leading to primary graft failure, delayed engraftment or poor graft function. We report an evaluation of donor specific anti-HLA antibodies testing and desensitization strategies employed in patients who were candidate to haploidentical stem cell transplantation between September 2018 and June 2023.

Methods: The anti-HLA antibody search was performed using the Luminex bead assays and expressed as mean fluorescence intensity (MFI); a MFI>1000 was considered as positive. If the patient had DSAs a desensitization strategy was employed with rituximab 375 mg/m2 (day −15), 2 single volume plasmaphereses (days −9 and −8), intravenous immunoglobulins 400 mg/kg (day −7). The conditioning regimen consisted of thiotepa, fludarabine and busulphan; the latter was infused over 3 days in myeloablative conditioning and 2 days in reduced intensity conditioning. Stem cell source was peripheral blood stem cells in all patients. GvHD prophylaxis consisted of cyclosporine, mycophenolate mofetil and cyclophosphamide.

Results: Eight of 50 patients (16%) who underwent haploidentical stem cell transplantation, (3 females and 5 males), median age 48 years (range 19-70) showed donor specific anti-HLA antibodies: 6 for class I HLA antigens (median MFI 4100) and 2 for both class I HLA and II (median MFI 1500). The conditioning regimes was myeloablative (3 patients) and reduced intensity (5 patients). The median infused CD34 and CD3 was 5,6 x 10e6/kg (range 3,1- 6,4) and 202 x 10e6/kg (range 111-284), respectively. All eight patients obtained a donor neutrophil engraftment (median 19 days, range 21-29 days); 7/8 patients obtained platelet engraftment (median 33 days, range 17-100 days) One patient died within 30 days after HSCT. Five patients obtained DSAs clearance within 30 days; a full donor chimerism was observed in 6/8 patients within 60 days after HSCT.

Conclusions: The densitization strategy used offers a reasonable option in patients with DSA positive that need to haploidentical stem cell transplantation and who lack another available donor. Prospective multicenter studies are needed to better define and validate consensus strategies on DSA management.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P620 EARLY ONSET OF EPSTEIN-BARR VIRUS POSITIVE MULTIPLE MYELOMA TYPE OF POSTTRANSPLANT LYMPHOPROLIFERATIVE DISORDER – A CASE OF INFREQUENT COMPLICATION AFTER ALLOGENIC PERIPHERAL BLOOD STEAM CELL TRANSPLANTATION

Kamila Kruczkowska-Tarantowicz1, Piotr Rzepecki1

1Military Institute of Medicine - National Research Institute, Warsaw, Poland

Background: Plasmocytoma-like/multiple myeloma posttransplant lymphoproliferative disorder (PTLD) is uncommon but serious implication after allogenic steam cell transplantation. According to the literature survival is definitely worse than in other forms of PTLDs.

Methods: In March 2022 a 39-year-old female without any prior history of disease was admitted to the Department of Haematology with fever, general weakness and life-threatining anaemia. Physical examination showed paleness, peteachie but no organomegaly or lymphadenopathy, also in ultrasonography, was founded. Blood smear revealed monocytosis and immature myeloid cells. Bone marrow aspirate was riched-cell with 4% blasts and over 40% of monocytic cells. The patient was diagnosed with high-risk chronic myelomonocitic leukaemia – myelodysplastic type (CMML-MD), and hypomethylating-agent treatment with intention to allogenic transplantaion was performed.

Cytogenetics showed normal kariotype and no bcr-abl mutation was detected. Due to the lack of family donor, unrelated donor with the same blood group was urgently machted.

After second course of azacitidine transformation to acute myelomonocytic leukaemie occured. Intensive treatment with induction regimen followed by allogenic peripheral blood steam cell transplantation was needed. According to the agressive clinical course, augumented conditionig regimen with melphalan, treosulfan, fludarabine, antithymocitc globuline and methotrexate, cyclosporin as immunosupresion was applied. Besides grade 3 mucositis there were no other complications during direct post-transplant term.

At day +41 the patient presented on emergency with fever, diarrhoea and peripheral lymphadenopathy.

No sings of graft-versus-host disease were detected. Laboratory tests showed pancytopenia, acute/severe kidney impairment with creatinine level at 406,6umol/l, and Epstein-Barr Virus quantitaive plasma level at almost 300 000 copies per mililter.

Serum elecrophoresis and immunofixation proved monoclonal IgG kappa and trace of IgM lambda proteins. In bone marrow aspirate immunophenotyping clonal plasmacytic cells were confirmed. Computed tomography scans indicated submandibular, cervical lymphadenopathy and splenomegaly with no osteolitic leasions. We performed trephine biopsy and surgical lymph node biopsy. Immunosupressive treatment was tapered, subsequently chemotherapy regimen based on bortezomib and anthracycline was given. Althoug two doses of rituximab the EBV-DNA quantitative plasma level arose to over 4 millions per mililiter. Unfortunately general status of the patient deteriorated rapidly with death at day +54 after alloPBSCT.

Results: Post-mortem obtained results of threphine biopsy showed hypocellular preparates with intestinal infiltrations of plasmatic cells, in immunohistochemistry positive for CD138, kappa chain and negative for CD20, with histopatological diagnosis of monomorfic plasmocytosis in PTLD course. However necrotic lymph node with clusters of large cells in immunohistochemistry study positive for CD20, CD79a, EBV-LMP1 was in constance with monomrphic post-transplant lymphoproliferative disease, EBV + , diffuse large B-cell lymphoma type.

Conclusions: Available data suggest that monomorphic multiple myeloma PTLD tends to behave in manner more similar to lymphomas with mass leasions rather than bone marrow impairment. In this case two related histopatology results indicate highly-agrresive type of disease and unfavourable clinical course.

Clinical Trial Registry: No.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P621 THE ROLE OF DARATUMUMAB IN POST-TRANSPLANT COMPLICATIONS: A PROSPECTIVE STUDY ON PRCA AND AIHA

Sabrina Giammarco 1, Maria Assunta Limongiello1, Luca Di Marino2, Elisabetta Metafuni1, Maggi Roberto2, Federica Sorá2, Eugenio Galli1, Patrizia Chiusolo2, Simona Sica2

1Fondazione Policlinico Universitario A. Gemelli, Rome, Italy, 2Universitá Cattolica del Sacro Cuore, Roma, Italy

Background: Pure red cell aplasia (PRCA) and autoimmune haemolytic anemia (AIHA) post-hemapoietic stem cell transplantation (HSCT) are an unmet medical need with no established standard of care, significantly affecting patient’s quality of life and posing a challenge for clinicians. The anti-CD38 IgG-kappa Daratumumab appears to be a safe and efficace treatment compared to prior strategies.

Methods: Our study is a prospective monocentric investigation assessing the use of daratumumab in this complication following allo-HSCT. Specifically, we present 5 patients with median age 65 years: 4 diagnosed with PRCA and 1 with AIHA post-HSCT. All patients, except one, had prior rituximab exposure and plasmapheresis and erythopoietin.

Patients were evaluated at 30, 60, 90, and 365 days post-transplantation, with reassessment of complete blood count and bone marrow aspirate. Another time point was set at 30 days from daratumumab start and at the end of therapy (not reached by 2 patients). The primary endpoint is transfusion independence, with discontinuation of erythropoietin as a secondary endpoint. Before daratumomab treatment, we ruled out infectious causes of ineffective erythropoiesis such EBV and CMV infection, and confirmed reduced erythroid precursors in bone marrow in 4; all of them had full donor chimerism.

Results: The patients enrolled required at least 1 unit of RBC/week. All patients responded positively to anti-CD38 antibody exposure, improving reticulocyte count. Almost all side effects that patients exhibited occurred after the first administration and were of mild severity; these included fever and skin rash. 3 patients were able to discontinue the use of erythropoietin and transfusions, with a median of the last transfusion occurring 30 days (20-74) from the initiation of daratumomab therapy. Two patients failed to achieve transfusion indipendence: one, still on treatment, received only 2 daratumomab cycles, suggesting a need for extended exposure. The 2nd patient developed sepsis 10 days after the 5th daratumomab administration, leading to ICU admission and death 15 days later. While daratumomab’s role in sepsis is uncertain, it is noteworthy that, except for a mild rash, the patient generally tolerated the treatment well without important hematologic toxicity. Although all the patients had reticulocytopenia, one was demonstrated to have AIHA anti-D with non responsive treatment to rituximab and plasmapheresis.

Patients number

5

Disease

PRCA 4 (80%)

AHIA 1 (20%)

Gender n (%)

Male 2 (40%)

Female 3 (60%)

Median age at transplant

Years (range)

65 years (45-68)

Hematological disease n (%)

AML

2 (40%)

MDS

1 (20%)

SAA

1 (20%)

LH

1 (20%)

Donor Type n (%)

MUD 8/8

1 (20%)

MUD 7/8

2 (60%)

SIBLING

1 (20%)

Conditioning regimen n (%)

TBF2

2 (40%)

TBF1

2 (40%)

Baltimora

1 (20%)

Median Hb value at 3 months from HSCT (range, g/dl)

6.9 (6.4-7.4)

Median reticulocytes value at 3 months

(range, %)

0.2 (0-0.5)

Median Hb value pre-daratumumab (range, g/dl)

7.4 (6.5-7.7)

Median reticulocytes value pre-daratumumab (range, %)

0.4 (0 – 1.6)

Mean days after-HSCT by daratumomab start

165 (112 – 259)

Median number of daratumumab

5 (2 – 8)

Median Hb value post-daratumumab at 30 days (range, g/dl)

8.3 (6.7 – 10.7)

Median reticulocytes value post-daratumumab at 30 days(range, %)

1.9 (0.3 – 5.8)

Conclusions: Poor graft function remains a management challenge for clinicians and has a significant impact on the patient’s quality of life. Currently, therapeutic options appear ineffective, making it difficult to address the diverse needs of post-transplant patients. Although daratumomab cannot be considered a one-size-fits-all solution, it certainly prompts further reflection on immunomodulation. Immune dysregulation in the form of a graft-versus-bone marrow response offers a unifying explanation for the intricacies of the bone marrow microenvironment. It is suspected to also act on the patient’s CD27-memory cells, which, at least in PRCA, seem to play an important role. It is noteworthy that these cells lose CD20-positivity, and this might partly explain the inefficacy of previous strategies. The number of patients does not allow for efficacy evaluation; however, similar to previous case reports, our center’s experience strengthens the role of daratumomab use.

Disclosure: No disclosure to declare.

14: Non-infectious Early Complications

P622 TRANSPLANT ASSOCIATED THROMBOTIC MICROANGIOPATHY IS AN IMPORTANT CAUSE OF HIGH EARLY MORTALITY IN ALLOGENIC BONE MARROW TRANSPLANT

Pawan Kumar Singh 1, Isha Gambhir1, Ravi Shanker1, Rasika Setia1, Anil Handoo1

1BLK-Max Superspeciality Hospital, New Delhi, India

Background: Allogenic bone marrow transplant is done to cure many hematological disorders. In early post transplant phase the complication of Thrombotic Microangiopathy (TMA) is seen most commonly with Calcineurin Inhibitors. But Transplant Associated-Thrombotic Microangiopathy (TA-TMA) is far less common and it is associated with very high mortality and the usual treatment like Therapeutic Plasma Exchange, Steroid or IVIg are not effective. Based on the pathogenesis the approved therapies are targeted on the complement pathway.

Methods: One hundred ninety three consecutive patients, who underwent allogenic BMT were enrolled in this study from October 2018 till October 2023.

Results: A total of 193 patients who underwent allogenic BMT were enrolled with a median age 22 years (range 0.5 - 64 years) out of which 134 (69%) males were included. Majority patients had Acute leukemia (45%) followed by hemoglobinopathies (14%) and then Aplastic anemia (13%) and rest had various indications. Out of these 193 patients, 26 (14%) developed Thrombotic Microangiopathy and in 21 patients the cause was Calcineurin inhibitor (Cyclosporine or Tacrolimus). Five patients (2.6%) developed Transplant Associated-Thrombotic Microangiopathy (TA-TMA) based of City of Hope Criteria. The median day of onset of TA-TMA was day +20 (range 11 – 139). Out of these 5 patients 3 were Haplo BMT, 1 was Matched Unrelated Donor Transplant and 1 was Matched Sibling Donor transplant. All patients who developed TA-TMA died and the median days from the onset to death were 6 days (range 2 – 30 days). Most patients were managed with supportive treatment in the form of IVIg, steroids and Rituximab. Anti-complement therapies were not given because of unavailability of drug in the country. In this study, the risk factors found to be associated with the development of TA-TMA were Haplo-identical donor, use of Total Body Irradiation in conditioning and CMV reactivation.

Conclusions: Transplant Associated-Thrombotic Microangiopathy (TA-TMA) is a life threatening less frequent complication of allogenic BMT and is associated with very high mortality in the absence of treatment with Anti-complement therapies. Early recognition of this complication and early administration of anti-complement therpay is the key factor in the management.

Disclosure: None.

14: Non-infectious Early Complications

P623 IMMUNE DYSREGULATION AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION BEYOND GVHD: A COMPREHENSIVE PEDIATRIC CASE REPORT - FROM DIAGNOSTICS TO TAILORED BTK INHIBITION BY IBRUTINIB

Peter Švec 1, Maria Füssiová1, Jaroslava Adamčáková1, Ivana Boďová1, Júlia Horáková1, Dominika Dóczyová1, Miroslava Pozdechová1, Tomáš Sýkora1, Alexandra Kolenova1

1National Institute of Children´s Diseases and Comenius University, Bratislava, Slovakia

Background: Immune dysregulation after allogeneic hematopoietic cell transplantation (allo HCT) presenting with prolonged systemic inflammation, non-infectious fevers and aberrant oligoclonal B cell development - without alloreactivity fulfilling clinical criteria of acute or chronic GvHD - is scarcely described and probably underreported in literature. We aim to contribute a small piece of information to stimulate further research efforts.

Methods: We provide an instructive pediatric case report with comprehensive diagnostic work-up and tailoring therapy.

Results: 15yo boy with T-ALL in CR1, pre-transplant history of disseminated aspergillosis (lungs, CNS, liver), underwent 10/10 MUD HCT after myeloablative (TBI 12Gy VP16) conditioning and serotherapy (rATG), with MTX and cyclosporin A (CsA) GvHD prophylaxis. Transplant complications included peri-transplant COVID-19, acute skin GvHD gr II responsive to steroids, CMV reactivation, repeated catheter-related blood stream infections, bacterial endocarditis, drug-induced acute renal failure and secondary poor graft function.

Daily fevers of unknown origin up to 39°C started at D + 64 and continued beyond D + 180 - accompanied by fatigue and increased systemic inflammation (CRP 4-10x UNL). The patient remained without clinical signs typical for acute or chronic GvHD (except for moderately decreased appetite). Despite extensive investigations no active infection was proven (repeated cultures and PCR panels directed against bacterial, viral, fungal and parasitic agents were performed, imaging (incl. chest CT, head MRI), testing of cerebrospinal fluid and bronchoalveolar lavage). Significant oligoclonal hypergammaglobulinemia developed (IgG 31 g/L) with autoantibodies detected (ANA, pANCA). T cell immune reconstitution was slow (CD4> 100 / µL reached at D + 180). Long Covid-19 / MIS-C and uncovered continued endocarditis were considered not likely with this particular clinical course and tests performed. The long-term stable clinical picture with little dynamics favored immune dysregulation over infection.

Immunosuppressive strategies besides continued calcineurin inhibitor (CsA then tacrolimus) included initially corticosteroids (partial response, severe side effects) and then ruxolitinib (no effect). Response was noted after high dosed anti-IL1 treatment (anakinra 100mg q12h). However, 8-week IL-1 cytokine inhibition did not cease the immune dysregulation and fevers returned during tapering attempt. Due to aberrant B cell activation Bruton kinase inhibition (ibrutinib), also targeting T cell pathways (ITK, JAK3) was selected as a further line of therapy and led to cessation of fevers, improved appetite, and general performance.

Conclusions: Immune dysregulation after alloHCT may present with chronic systemic inflammation, B cell deregulation, significant impact on quality of life and carry long-term risks. Standard immunosuppressive strategies used in GvHD therapy may not be effective. BTK and IL-1 signaling inhibition were observed to be beneficial. Improved knowledge about driving mechanisms, the role of alloimmunity and autoinflammation, precise definitions and treatment guidelines are needed.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P624 A RARE CASE OF MULTIPLY RELAPSED POST-ALLOGENEIC STEM CELL TRANSPLANT IMMUNE THROMBOTIC THROMBOCYTOPENIC PURPURA IN A YOUNG PATIENT WITH GATA-2 POSITIVE MYELODYSPLASTIC SYNDROME

Angela Dăscălescu 1, Antohe Ion2, Elena Dolachi3, Roxana Dumitru4

1University Of Medicine and Pharmacy “Gr. T. Popa”, Iași, Romania, 2University of Medicine and Pharmacy “Gr. T. Popa”, Iași, Romania, 3Regional Oncology Institute, Iași, Romania, 4Regional Oncology Institute, Iași, Romania

Background: Transplantation-associated thrombotic microangiopathy (TA-TMA) is an infrequent yet severe complication of hematopoietic stem cell transplantation (HSCT), presenting an incidence ranging from 0.5% to 76%. Despite treatment efforts, the mortality rate for TA-TMA remains high, ranging from 60% to 90%.

Classic idiopathic TTP is associated with a deficiency in ADAMTS13 activity, a metalloproteinase responsible for cleaving ultra-large von Willebrand factor multimers. On the other hand, TA-TMA has a more complex, multifactorial etiology involving endothelial damage induced by intensive conditioning therapy, irradiation, immunosuppressants, infection, and graft-versus-host disease. This explains the lack of efficacy of plasma exchange, a conventional treatment for idiopathic TTP aimed at restoring ADAMTS-13 activity, in the case of TA-TMA.

Methods: We here present the case of a 25-year-old male patient who received an allogeneic stem cell transplant from a 9/10 matched unrelated donor for a myelodisplastic syndrome with GATA-2 germline mutation.

Results: The patient developed features of TA-TMA on day +21 posttranplant (progressive thrombocytopenia, evidence of microangiopathic hemolytic anemia, neurologic dysfunction, acute kidney injury), but whose subsequent evaluation revealed a level of ADAMTS13 activity below 1%. Given this finding, he received immune PTT- directed therapy with plasma exchange therapy (TPE), Caplacizumab, steroids and Rituximab, with prompt hematologic and neurologic recovery. Subsequently, being diagnosed with poor graft function, he received a CD34 purified cell boost from the same donor on day +204, which prompted another relapse of the immune PTT. He developed a total of three PTT relapses (day +42, +220 and +284), all of which were treated with TPE, Caplacizumab and Rituximab. Upon the last relapse weekly Cyclophosphamide was added to his immunosuppressive therapy.

Conclusions: We here highlight the physiopathologic differences between the two PTT forms and we hypothesize on the existence of a donor-derived factor prompting repeated immune TTP relapses in this patient.

Disclosure: Nothing to declare.

14: Non-infectious Early Complications

P625 CENTRAL NERVOUS SYSTEM TOXICITY WITH EXCLUSIVE INVOLVEMENT OF THE BRAIN WHITE MATTER LIKELY RELATED TO TOXIC/IMMUNOLOGIC CAUSES. A PRESENTATION OF 4 SIMILAR CASES

Valentina Sangiorgio1, Elena Agostani 1, Marilena Fedele2, Paola Perfetti2, Cristina Capraro2, Elisabetta Terruzzi2, Andrea Aroldi2, Carlo Gambacorti Passerini1, Matteo Parma2

1Università di Milano-Bicocca, Monza, Italy, 2IRCCS San Gerardo dei Tintori, Monza, Italy

Background: Central nervous system (CNS) complications are an emerging cause of morbidity and mortality after allogenic hematopoietic stem cell transplantation (HSCT) and their clinical management can be highly challenging. CNS complications can occur secondarily to a range of causes including infections, toxic effects, metabolic, vascular, immune-mediated processes and as a direct disease-related neurotoxicity. We present the clinical history of four patients (pts) undergoing an allo-HSCT at our Department between July and September 2023, all experiencing early neurological complications after the transplantation procedure.

Methods: 4 pts (2 males and 2 females) aged between 45 and 65 underwent HSCT for hematologic malignancies (donor source: 2 sibling, 1 MUD, 1 haplo). Conditioning regimen was ablative for 1 pts (Thiotepa Busulfan Fludarabine) and of reduced intensity (Treosulfan-Fludarabine) for 3 pts. Anti Thimocyte Globulin (ATG) was administered in 3 pts, Pt-CTX in 1. GvHD prophylaxis included Cyclosporin in 2 pts and Tacrolimus in 2 pts.

Results: Between day +13 and +42 after HSCT all patients developed neurological symptoms including a progressive altered mental status and ideomotor slowing; cortical blindness was seen in two cases. The electroencephalogram showed non-specific alterations. Brain magnetic resonance imaging (MRI) in all cases revealed a moderate FLAIR sequence hyperintensity in the cerebral white matter of the corona radiate with a symmetrical distribution, mostly consistent with a toxic/immunological damage (Figure 1). Cerebrospinal fluid examination yielded negative results for malignancy and infections including bacteria, virus (Herpes, BK-JC, EBV, WNV), protozoa and prions. Peripheral blood sequencing tests for viruses were similarly negative. A post-transplant thrombotic microangiopathy was ruled out by the mean of conventional blood tests. In all patients, calcineurin inhibitors were discontinued (assuming a possible contribution to the CNS damage) with no benefit. A treatment with high-dose steroid (1g of intravenous -IV-methylprednisolone for 3 days) along with 5-day infusion of IV immunoglobulins (0.4mg/Kg daily) was performed in the hypothesis of an immune-mediated process. 1 pts showed a progressive improvement with a complete neurological recovery within 30 days; 3 pts had a transient improvement followed by an irreversible worsening leading them to coma and death. Subsequent brain MRI documented a progressive extension of the white matter alterations. Postmortem examinations (PME) were performed and the histological examinations are still ongoing.

Conclusions: Metabolic, infectious, immunologic and drug-related toxicities explain a large percentage of CNS complications. The ones we described are not dependent on metabolic or infectious complications, neither related to calcineurin inhibitors. Fludarabine is the only drug shared by all pts, instead ATG was used in 3 pts (included the only pts who had a positive outcome). Fludarabine is a potential cause of severe encephalopathy with white matter symmetric involvement, in the setting of HSCT conditioning. An immune-mediated contribution can be hypothesized based on the response to steroid and immunoglobulin we observed in one patient. In our cases, it can be hypothesized that a drug related damage (due to Fludarabine) was the initial trigger, followed by a secondary immunologic aggression, uncontrolled in 3/4 pts. We are waiting for the PME, which hopefully will provide further insights.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (81)

Disclosure: No conflict of interest.

14: Non-infectious Early Complications

P626 EVALUATING AN UPDATED ANTIEMETIC PROTOCOL IN HAEMATOPOIETIC STEM CELL TRANSPLANTATION

Katie Robertson1, Muhammad Saif 1, Ann Griffiths1, Thomas Sanders1, Daniel Monnery1

1Clatterbridge Cancer Centre, Liverpool, United Kingdom

Background: Haematopoietic stem cell transplantation (HSCT) is a vital treatment option for patients with a number of haematological and non-haematological disorders. however, this treatment is associated with with significant toxicities. A common side effect of HSCT is nausea and vomiting caused by use of high dose conditioning chemotherapy. These symptoms are often difficult to manage with first line anti-emetic therapy including ondansetron, steroids and NK1 antagonists.

In our trust, an antiemetic protocol has been developed, reflecting recommendations in MASCC/ESMO guidelines, including the addition of olanzapine as a second line intervention.

The aim of this study is to evaluate the effectiveness of the updated protocol in the management of nausea and vomiting post-stem cell transplant.

Methods: In July 2023, the new protocol was launched and a prospective evaluation of its efficacy was undertaken. Patients receiving stem cell transplant at our centre reported the effectiveness of each line of therapy within the protocol in managing nausea, as quantified using the IPOS scale. Initial results are presented in this analysis whilst data collection is ongoing.

Results: At the time of current analysis, data on 36 patients were available. Of these, 11 (31%) had good nausea control with ondansetron, steroid and NK1 antagonists at the time of chemotherapy, followed by ondansetron alone via continuous subcutaneous infusion (CSCI). Of the patients that progressed to a combination of ondansetron and cyclizine, 6 (33%) had good control, and 12 (67%) had poor control. In the group that progressed to a combination of ondansetron and levomepromazine, 3 (43%) reported good control, and 4 (57%) had poor control. 3 of the patients with poor control on ondansetron and cyclizine, and 1 patient with levomepromazine and ondansetron, were switched to olanzapine. Of this group, 1 (33%) had good control, and 2 (67%) had poor control with the use of olanzapine via CSCI.

Conclusions: The majority of patients in this cohort required combinations of antiemetics beyond ondansetron alone. The early results of this study do not support the use of olanzapine monotherapy in preference to combination therapy in this population.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P627 ECULIZUMAB TREATMENT OF HEMATOPOIETIC STEM CELL TRANSPLANTATION-ASSOCIATED THROMBOTIC MICROANGIOPATHY: SINGLE CENTER EXPERIENCE

Tatiana Rudakova1, Julia Vlasova1, Olesya Paina1, Olga Slesarchuk1, Marina Gorodnova1, Tatyana Schegoleva1, Oleg Goloshchapov1, Tatyana Bykova 1, Elena Morozova1, Lyudmila Zubarovskaya1, Ivan Moiseev1, Alexander Kulagin1

1Pavlov University, St. Petersburg, Russian Federation

Background: Hematopoietic stem cell transplantation-associated thrombotic microangiopathy (TA-TMA) is a severe and potentially life-threatening complication. Complement inhibition may be a target to prevent endothelial injury and dismal outcomes of patients with TA-TMA.

Methods: We analyzed outcomes of 8 adult and 6 pediatric patients with TA-TMA treated with eculizumab in 2015-2023. Median age was 27 (1-62). Indications for allo-HSCT were acute leukemia (n=6), myelodysplastic syndrome (n=2), osteopetrosis (n=2), dyskeratosis congenita (n=1), Schwachmann-Diamond syndrome (n=1), Hodgkin’s lymphoma (n=1), primary myelofibrosis (n=1). Allo-HSCT was performed from matched unrelated (n=6), matched related (n=3) and haploidentical donor (n=5). Bone marrow was the graft source in a half of cases. Busulfan-based conditioning was used in 6 (43 %) cases with total dose of 8-16 mg/kg. Graft-versus-host disease (GVHD) prophylaxis included calcineurin inhibitors in 12 of 14 cases, 5 of 14 were ATG-based. TA-TMA was diagnosed with the use of International Working Group of the European Group for Blood and Marrow Transplantation criteria (2007) and with TMA Harmonization Panel Consensus Recommended Diagnostic Criteria (2023). Single eculizumab dose was 600 mg given weekly. Response to treatment was evaluated by monitoring renal function, blood pressure, proteinuria, thrombocytopenia, anemia, transfusion requirements, presence of schistocytes, and lactate dehydrogenase (LDH). Complete response (CR) to eculizumab was defined as resolution multiple organ failure, red cells and platelet transfusions, normalization of LDH. Patients with no response (NR) remained dependent on transfusions, did not recover vital organ function after last eculizumab dose, or died with active TA-TMA. Partial response (PR) was assigned to those who did not meet CR or NR criteria.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (82)

Results: TA-TMA was diagnosed during the first 100 days after transplantation in 10 of 14 (92%) subjects, at a median of 34 days (13-62). Four patients had late TA-TMA, all of them had prolonged therapy for GVHD. Median follow-up time from the onset of TA-TMA to the last contact was 156 (10-2823) days. Median time of eculizumab therapy start was 5 (0-30) days after the onset of TA-TMA. Eight of 14 treated patients (57%) achieved CR, 3 (21%) had PR, and 3 (21%) had NR. Cumulative incidence of LDH≤1,5 upper limit of normal achieving after eculizumab therapy was 73% (95% CI, 34-91) with median time of 17 days (1-198). Cumulative incidence of platelet level ≥ 50x109/l was 77% (95% CI, 21-97) with median time of platelet normalization 57 days (6-384). The median number of eculizumab doses was 2 (range, 1-6). Eculizumab was well tolerated without any significant side effects. One-year overall survival was 56 % (95% CI, 35-90). A total of 8 subjects died, 2 of them had active laboratory signs of TMA at death. Other causes of death were relapse (n=2), infection (n=2), GVHD (n=1), VOD (n=1).

Conclusions: Eculizumab is a well-tolerated promising therapy of TA-TMA. More studies are needed to establish timing and dosing regimen in TA-TMA.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P628 IMMUNE THROMBOCYTOPENIA FOLLOWING ALLOGENEIC STEM CELL TRANSPLANT : RESULTS FROM A FRENCH MULTICENTRIC RETROSPECTIVE STUDY OF 35 CASES

Antoine Gondé 1,2, Aude Legal3, Flore Sicre de Fontbrune4, Mathieu Puyade5, Maud d’Aveni6, Yves Béguin7, Carmen Botella Garcia8, Éolia Brissot9, Étienne Daguindau10, Bertrand Godeau1, Marc Michel1, Thibault Comont3

1Hôpital Henri-Mondor, Créteil, France, 2Université Paris-Cité, Paris, France, 3IUCT Oncopole, Toulouse, France, 4Hôpital Saint-Louis, Paris, France, 5CHU de Poitiers, Poitiers, France, 6CHRU de Nancy, Nancy, France, 7CHU de Liège, Liège, Belgium, 8CHU de Bordeaux, Bordeaux, France, 9Hôpital Saint-Antoine, Paris, France, 10CHU de Besançon, Besançon, France

Background: Immune thrombocytopenia (ITP) is a rare but potentially severe complication of allogeneic hematopoietic stem cell transplant (HSCT). Its accurate diagnosis is often challenging, and the factors associated with its onset are poorly understood. There is a lack of recent published data regarding the precise characteristics and outcome of patients diagnosed with ITP after HSCT.

Methods: This multicentric retrospective study included adult patients from the French Society of Bone Marrow Transplant database, who had undergone HSCT between 2008 and 2023 and obtained a full platelet count recovery following transplant (> 100 x 109/L) and who subsequently developed ITP within 10 years following HSCT. Patients were required to have had bone marrow evaluation compatible with the diagnosis of ITP.

Results: Thirty-five patients (including 11 women) were included. The median age at the time of transplant was 55 years [IQR 48–65]. Thirty-four (97%) patients were transplanted for hematological malignancies, with a predominance of acute leukemias (n = 18, 51%), and 1 case of aplastic anemia. Transplants were predominantly HLA-compatible (10/10, geno- or pheno-identical, n = 25/35, 71%), with only 5 haplo-identical transplants. Twenty-eight patients (80%) had reduced-intensity conditioning (Table 1). Eighteen patients (51%) developed stage 2–4 acute GVHD, and 14 (40%) chronic GVHD.

Transplant characteristics

n = 35

Median age at transplant (IQR) — years

55 (48–65)

Transplant indication — no. (%)

Myeloid neoplasm

27 (77%)

Lymphoid neoplasm

7 (20%)

Other

1 (3%)

Conditioning intensity — no. (%)

Reduced intensity

28 (80%)

Myelo-ablative

7 (20%)

HLA compatibility — no. (%)

10/10

25 (71%)

9/10

3 (9%)

5/10 (haplo-identical)

5 (14%)

Immune thrombocytopenia characteristics

n = 35

Median time to diagnosis after transplant day 0 (IQR) — months

10.1 (6.1–20.7)

Khellaf bleeding score — no. (%)

5 (14%)

> 8

9 (25%)

Life-threatening bleeding — no. (%)

6 (17%)

Death at initial onset — no. (%)

3 (9%)

Diagnosis of ITP was made after a median of 10.1 months post-transplant [6.1–20.7] (Table 1). Patients were asymptomatic in 31% of cases, whereas 25% of patients had severe bleeding (Khellaf score > 8), with life-threatening bleeding in 6 cases. The median nadir of platelet count was 5 x 109/L [0–13.5].

At last evaluation before ITP onset, 88% of patients showed complete donor chimerism, and 16 (46%) were still under immunosuppressive therapy. Bone marrow evaluation showed megakaryocytes in normal or increased numbers in 70% of cases. 16/29 (55%) of tested patients had active viral replication in peripheral blood, mainly EBV (n = 13, medial viral load 3.1 log).

Regarding treatment, 27/35 (77%) patients required more than 1 line of therapy, with a median of 4 lines [2–5]. Twenty-nine patients (83%) received corticosteroids, 29 received intravenous immunoglobulins, with overall response rates (ORR) of 52% and 59%, respectively. As second- or third-line treatments, 40% of patients received eltrombopag, 49% romiplostim (respective response rates of 43% and 53%). Rituximab was administered to 46% of patients, with an ORR of 50%. Among the 8 patients who received immunosuppressants for ITP, 6 (75%) obtained a response.

After a median duration of follow-up of 30 months [9.5–80.5], 27 (80%) patients were in complete remission, whereas 7 (20%) had refractory ITP with a platelet count < 30 x109/L. Median time to best response was 2.2 months [0.6–6.9]. Three patients died from intracranial bleeding at initial onset of ITP.

Conclusions: This multicentric retrospective cohort of post-HSCT ITP highlights its heterogeneous and sometimes life-threatening clinical presentation, along with its recurrent refractoriness to usual therapies of ITP. This study is ongoing and still recruiting. A detailed statistical analysis with follow in order to identify possible predictive and prognostic markers of ITP after HSCT.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P629 AT-HOME AUTOLOGOUS HEMATOPOIETIC CELL TRANSPLANT FOR ADULTS WITH HEMATOLOGICAL MALIGNANCIES. HOW FRAILTY SYNDROME IMPACTS AND EVOLVES DURING HCT PROCEDURE

Juan Ortiz 1, Maria Teresa Solano2, Cristina Galleago2, Nuria Ballestar2, Anna Serrahima2, Noemi de llobet2, Raquel Salinas2, Alexandra Patricia Martinez2, María Suárez-Lledó2, Beatriz Merchán2, Paola Charry2, Joan Cid2, Miquel Lozano2, Laura Rosiñol2, Carmen Martinez2, Montserrat Rovira2, Enric Carreras2, Francesc Fernandez2

1hospital universitario galdakao-usansolo, Galdakao, Spain, 2Clinical Institute of Hematology and Oncology (ICMHO), Hospital Clínic de Barcelona, Barcelona, Spain

Background: Frailty in patients undergoing autologous hematopoietic cell transplantation (auto-HCT) has been associated in previous studies with prolonged hospitalization and increased toxicity. At-Home auto-HCT is being done at our institution since November 2000. Since April 2021, the frailty state of patients is evaluated routinely in adults undergoing HCT using the HCT Frailty Scale. Considering the higher transplant toxicity associated to frail adult undergoing auto-HCT, this study investigates the impact and dynamics of frailty in outpatient auto-HCT.

Methods: This study includes the 80 consecutive adults with Lymphoma and multiple myeloma (MM) undergoing at-home auto-HCT at our institution between June 2021 and June 2023. They were evaluated for frailty at first consultation (pre-apheresis), at HCT admission, and at day +100. According to the HCT Frailty Scale, patients were classified into fit, pre-frail and frail categories. All patients provided informed consent. Prospective data was analyzed in December 2023.

Results: The median age was 58 years (range: 19-69), 41 (51.2%) patients were males, and MM was the most prevalent indication for auto-HCT 45 (56.3%). At first consultation, 24 (30.0%) adults were classified as fit, 48 (60.0%) as pre-frail, and 8 (10.0%) as frail. Frail patients were more likely to be older (OR 1.16 P=0.077), and a KPS < 90% (OR 27, p=0.012), with a higher number of comorbidities (HCT-CI>3: OR 11.9, p=0.035). However, the underlying diagnosis did not impact the incidence of frailty at the first consultation (Or 0.99, p=0.999) (Multivariate binary logistic regression analysis). Transplant methodology did not differ based on the frailty condition of patients. As per protocol, conditioning regimen and stem cell infusion were always administered at hospital. Patients were discharged on day +1. No patient received prophylactic G-CSF. Patients with MM received prophylaxis for engraftment syndrome with decreasing doses of corticosteroids. Transplant results were evaluated according to the frail stages determined at first consultation in all patients and according to their baseline diagnosis. Transplant-related mortality was 0. As described in Table 1, pre-frail and frail adults had comparable transplant results as in fit patients. The median duration of the outpatient HCT was 15 days (range: 13-22). Four (5.0%) patients required HCT readmission, mostly due to febrile neutropenia, with no differences according to frail stages.

Trends on frailty were evaluated in the subsample of 50 patients who were evaluated for frailty at the three pre-defined timepoints. Fit, pre-frail and frail incidences did not differ between patients with LPD and MM at any timepoint (p>0.05). Statistically significant variations were observed in the frailty stages depending on the timepoints when frailty was evaluated.

TABLE 1. Results According to HCT Frailty Scale

Full size table

Conclusions: The results support that at-home auto-HCT is safe and can successfully be performed in fit, pre-frail, and frail adults with an experienced and multidisciplinary team. Frailty syndrome was not associated with worse transplant outcomes. Although conclusions are limited by the reduced sample size, the observed differences on frailty incidences during patients follow-up support that frailty is dynamic, and hence, potentially amendable with specific interventions.

Disclosure: nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P630 FRAILTY ASSESSMENT IN ADULTS UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION OUTCOMES. PROSPECTIVE STUDY ON BEHALF OF THE GRUPO ESPAÑOL DE TRASPLANTE HEMATOPOYÉTICO Y TERAPIA CELULAR (GETH-TC)

Maria Queralt Salas 1,2, Maria Teresa Solano1,2, Mónica Baile-González3,2, Marina Acera-Gómez3,2, Laura Fox4,2, Maria del Mar Pérez-Artigas4,2, Ana Santamaría5,2, María del Carmen Quintela-González5,2, Andrés Sánchez-Salinas6,2, Joaquina M. Salmerón-Camacho6,2, Verónica Illana-Álvaro7,2, Zahra Abdallahi-Lefdil7,2, Javier Cornago-Navascues78,2, Laura Pardo8,2, Sara Fernández-Luis9,2, Leddy Patricia Vega-Suárez9,2, Sara Villar10,2, Patricia Beorlegui-Murillo10,2, Albert Esquirol11,2, Isabel Izquierdo-García12,2, Sonia Rodríguez González13,2, Alberto Mussetti13,2, Esperanza Lavilla14,2, Javier López-Marin15,2, Ángel Cedillo2, Leyre Bento16,2, Anna Sureda13,2

1Hospital Clínic de Barcelona, Barcelona, Spain, 2Grupo Español de Trasplante de Progenitores Hematopoyéticos y Terapia Celular, Madrid, Spain, 3Complejo Asistencial Universitario de Salamanca/IBSAL, Salamanca, Spain, 4Hospital Universitari Vall d’Hebron, Barcelona, Spain, 5Hospital Álvaro Cunqueiro, Vigo, Spain, 6H.C.U. Virgen de la Arrixaca, Murcia, Spain, 7Hospital Universitario de la Princesa, Madrid, Spain, 8Hospital Universitario Fundación Jiménez Díaz, Madrid, Spain, 9Hosptal Universitario Marqués de Valdecilla, Santander, Spain, 10Clínica Universidad de Navarra, Pamplona, Spain, 11Hospital de la Santa Creu i Sant Pau, Barcelona, Spain, 12Hospital Universitario Miguel Servet, Zaragoza, Spain, 13Institut Català d’Oncologia - Hospital Duran i Reynals, L’Hospitalet de Llobregat, Spain, 14Hospital Universitario Lucus Augusti, Lugo, Spain, 15Hospital Universitario de Alicante, Alicante, Spain, 16Hospital Universitario Son Espases, Mallorca, Spain

Background: Since February 2022, fifteen institutions members of the GETH-TC are participating in a multicenter, observational and prospective study with the purpose of investigating the state of frailty of adult patient candidates to allo-HCT and evaluate the effect of frailty in transplant outcomes.

Methods: All patients consulted for allo-HCT are eligible to be included in the study after providing informed consent. Patients are classified into fit, pre-frail and frail categories using HCT Frailty Scale (Salas et al. BMT 2023). Frailty syndrome is measured longitudinally at first consultation, HCT admission, and on days +100, +180, and 1 year. Frailty is evaluated clinical practice and utilizing existing resources. Patient´s cognition is additionally explored using Mini-Cog. The median time to complete the evaluation is 10 minutes. These results are not used to determine HCT eligibility or to design the HCT process. Prospective data was updated in December 2023.

Results: The 378 consecutive adult candidates for allo-HCT evaluated for frailty at first consultation and HCT admission between February 2022 and September 2023 were included in the study. Median patients’ age was 56 (range, 18-76); 66.1% were males, and myeloid malignancies were the prevalent baseline diagnosis (74.6%). Frail patients were more likely to have a KPS<90% (OR 2.5, p=0.01) and an abnormal Mini-Cog test (<3) (OR 4.42, P=0.004). No association was observed between frailty and older age (>60 years) (p=0.395), female sex (p=0.998), and comorbidities (HCT-CI>3) (p=0.925) (multivariate binary regression analysis).

Sixty-six (17.5%) patients went through a pre-transplant rehabilitation (pre-hab) program. With this information, the dynamics of frailty of these patients was compared with that of patients who did not join a pre-hab program. As described in Table 1, At HCT admission, the distribution of patients across the frailty categories was different in paatients who underwent pre-hab and those who did not (p=0.005).

Lastly, the power of the HCT Frailty Scale to predict OS and NRM was evaluated in the subsample of 342 patients that had a minimum follow-up of 100 days among survivors. Considering the redistribution of patients after pre-hab, the statistical analysis was conducted based on data obtained at HCT admission. The probability of OS at 1-year of patients classified as fit, pre-frail and frail categories was, respectively, 86.2%, 72.9%, and 51.9% (p<0.001). Furthermore, the multivariate regression analysis reported at Table 1 confirmed that frail patients had a lower OS (HR 3.67, p<0.001) and higher NRM (HR 2.87, p=0.041) irrespective of age, comorbidities and KPS.

Full size table

.

Conclusions: This study validates the applicability of the HCT Frailty Scale at HCT institutions that are part of a health care system different from that where it was first implemented. The HCT Frailty Scale classifies adult candidates for allo-HCT in three categories of frailty that have predictive power over transplant outcomes. Frailty is independent of age, dynamic, and can be improved through pre-hab programs with the positive result of improving OS.

Clinical Trial Registry: No applicable.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P631 LATE EFFECTS AFTER PEDIATRIC ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Raquel García Ruiz1, Sara Fernández-Luis1, Miriam Sánchez Escamilla1, María Terán Díaz1, Juan Manuel Cerezo Martín1, Juan José Dominguéz-García1, Arancha Bermúdez Rodríguez1, Enrique M. Ocio San Miguel1, Mónica López Duarte 1

1Hospital Universitario Marqués de Valdecilla (IDIVAL), Santander, Spain

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a curative treatment for pediatric patients with malignant and non-malignant diseases. Given the significant improvement on the procedure, there is a growing number of long-term survivors after pediatric HSCT, what has led to a substantially increased burden of late effects (LE) including chronic complications and organ impairments.

The aim of this study is to assess LE since first HSCT and to evaluate factors that impact on its development.

Methods: Retrospective study was made on patients <18 years-old who underwent HSCT in our institution between 1980-2020 and had at least 24 months of follow-up.

Endocrine, pulmonary, cardiac, neurological, oral and ocular effects, cardiovascular risk factors (CVRF), infectious complications and secondary neoplasm were collected as LE. Cumulative incidence (CI) values are shown based on the number of patients without missing data.

Results: During the study period, 142 pediatric patients underwent HSCT, 67 patients died before 24 months and follow-up was lost in 6 patients. Sixty-nine patients were finally analysed. The median follow-up was 110.7 months (range 25.9-482.7), the median age at transplant was 13.5 years old (range 1.1-17.6) and the most frequent indication was acute lymphoblastic leukaemia (28 patients, 40.6%). Patients and HSCT characteristics are shown in Table-1.

Table-1. Patients and HSCT characteristics.

Total (n = 69)

Age, median (min-max)

13.5 (1.1-17.6)

Sex, n (%)

Male

35 (50.7)

Female

34 (49.3)

Underlying diagnosis, n (%)

Acute leukemia

38 (55.1)

Bone marrow failure

18 (26.1)

MPN/MDS

8 (11.6)

Non-malignant

3 (4.3)

NHL

2 (2.9)

Malignant disease status at HSCT, n (%)

Complete response 1

27 (56.3)

Complete response 2

15 (31.3)

Active disease or partial response

6 (12.5)

Donor type, n (%)

Matched related donor

41 (59.4)

Haploidentical related donor

5 (7.2)

Matched unrelated donor

19 (27.5)

Mismatch unrelated donor

4 (5.8)

Stem cell source, n (%)

Bone marrow

60 (87.0)

Peripheral blood

5 (7.2)

Umbilical cord blood

4 (5.8)

Conditioning regimen, n (%)

Based in TBI

41 (59.4)

Based in Busulfan

12 (17.4)

Cyclophosphamide ± fludarabine

13 (18.8)

Others*

3 (4.3)

GvHD prophylaxis, n (%)

Calcineurin inhibitor + MTX/MMF

38 (57.6)

MTX

9 (13.6)

Cyclophosphamide ± corticosteroid

12 (18.2)

Others†

3 (4.5)

No prophylaxis

4 (6.1)

Lymphoid depletion, n (%)

No

52 (75.4)

ATGs

15 (21.7)

Alemtuzumab

2 (2.9)

  1. Abbreviations: ATGs, Anti-T-cell globulins; GvHD, graft versus host disease; MMF, mycophenolate mofetil; MTX, methotrexate; MPN, myeloproliferative neoplasm; MDS, myelodysplastic síndromes; NHL, non-Hodgkin lymphomas; TBI, total body irradiation.
  2. *Others conditionig regimen: 2 patients treosulfan, fludarabine and thiotepa; 1 patient daunorubicin, tioguanine and cytarabine.
  3. †Other GvHD prophylaxis: unknown.

Fifty-eight patients (84.1%) developed at least one LE and 30 patients (43.5%) developed ≥3. The CI at 5 and 10 years was 65.7% and 79.5% respectively. Median onset of the first LE was 26.9 months (range 2.1-362.4).

Most frequent LE were endocrine and ocular, followed by CVRF, oral and infectious. CI of endocrine, CVRF and ocular events at 5 and 10-year raised from 38% to 50%, 14% to 25% and 23% to 28% respectively. Oral and infectious complications had similar 5 and 10-year CI (15% to 17% and 14% to 18%).

Some particularly severe LE, such as pulmonary, neurological and cardiac complications had a CI at 10 years of 13%, 3% and 2% respectively. Twelve patients developed secondary malignancies, with a 10-year CI of 12%, being 3 of them hematological.

Patients who developed chronic graft-versus-host-disease (cGvHD) had a higher frequency of delayed growth (OR 3.5, IC95% 1.0-12.1, p=0.048), diabetes mellitus (OR 4.5, IC95% 1.0-19.8, p=0.045), pulmonary LE (OR 24.2, IC95% 2.8-210.2, p=0.004), dry eye (OR 6, IC95% 1.1-34.3, p=0.044) and recurrent infections (OR 8.8, IC95% 1.7-46.5, p=0,010). Xerostomia and dental caries were all detected in patients who developed cGvHD. Two patients with cGvHD required a pulmonary transplantation.

Patients who received total body irradiation had more frequency of cataracts (OR 12.6, IC95% 1.5-105.3, p=0.020) and gonadal dysfunction (OR 4.0, IC95% 1.2-13.8, p=0.028).

Higher age was a protective factor for delayed growth (OR 0.76, IC95% 0,65-0.89, p=0.001).

Conclusions: In our series, a high proportion of patients suffered from LE (84.1%) and 43,5% had ≥3 LE. Endocrine, CVRF and ocular effects increased over the time and the development of second neoplasms was important.

Future efforts should be focused on reducing development of LE and an early detection. Development of validated and individualised follow-up programmes should be designed.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P632 OUTCOMES AND RISK FACTORS FOR SURVIVAL OF LUNG TRANSPLANTATION AFTER PRIOR ALLOGENEIC HAEMATOPOIETIC STEM CELL TRANSPLANTATION: A STUDY FROM THE EBMT TRANSPLANT COMPLICATIONS WORKING PARTY

Saskia Bos1, Juan Montoro 2, Christophe Peczynski3, Pascale Ambron3, Manuela Badoglio3, Robin Vos4, Tobias Gedde-Dahl5, Mahmoud Aljurf6, Werner Rabitsch7, David Ma8, Harshita Rajasekariah8, Jaime Sanz2, Johannes Schetelig9, Anna Paola Iori10, Christian Koenecke11, Mar Bellido12, Juergen Finke13, Montserrat Rovira14, Angela Figuera15, Rabah Redjoul16, Ivan Moiseev17, Olaf Penack18, Hélène Schoemans19, Zina Peric20

1Newcastle University and Freeman Hospital, Newcastle upon Tyne, United Kingdom, 2University Hospital La Fe, Valencia, Spain, 3EBMT Transplant Complications Working Party, Sorbonne University, Hôpital Saint Antoine, Paris, France, 4University Hospitals Leuven, Leuven, Belgium, 5Oslo University Hospital, Oslo, Norway, 6King Faisal Specialist Hospital & Research Centre, Riyadh, Saudi Arabia, 7Medizinische Universitaet Wien, Vienna, Austria, 8St. Vincent’s Hospital, Sydney, Australia, 9University Hospital Dresden, Dresden, Germany, 10Policlinico Umberto I, Rome, Italy, 11Hannover Medical School, Hannover, Germany, 12University Medical Center Groningen, Groningen, Netherlands, 13University of Freiburg, Freiburg, Germany, 14University of Barcelona, Barcelona, Spain, 15Hospital de la Princesa, Madrid, Spain, 16Hôpital Henri Mondor, Créteil, France, 17Pavlov First Saint Petersburg State Medical University, Saint Petersburg, Russian Federation, 18Universitätsmedizin Berlin, Berlin, Germany, 19University Hospitals Leuven and KU Leuven, Leuven, Belgium, 20University Hospital Zagreb, Zagreb, Croatia

Background: Pulmonary chronic graft-versus-host disease (cGvHD) is a detrimental late pulmonary complication after allogeneic haematopoietic stem cell transplantation (alloHSCT) with significant morbidity and mortality and limited treatment options. Lung transplantation has the potential to provide survival benefit but is rarely performed due to concerns about poor outcomes, and specific alloHSCT-related risk factors have not been well studied.

Methods: The EBMT database was searched for adult patients who received a lung transplant for pulmonary cGvHD after undergoing at least one alloHSCT between 2000 and 2019 for this retrospective multicentre study. Thirty European and non-European centres participated. Clinical data before and after lung transplantation were reviewed, with specific emphasis on alloHSCT-related factors.

Results: Seventy-one patients from 14 countries were included, 59% of whom were male. Main indications for alloHSCT were acute (62%) and chronic (14%) leukaemia, followed by myelodysplastic/myeloproliferative disorders (10%) at a median age of 30 years (IQR 24-40). 14% had received a previous autologous or allogeneic HSCT. Identical siblings were the most common donors (49%), with peripheral blood grafts being the main source of stem cells (86%). Median time from alloHSCT to diagnosis of pulmonary cGvHD was 13 months (IQR 8-22). Main indication for lung transplantation was bronchiolitis obliterans syndrome (79%), at a median of 57 months (IQR 34-78) after alloHSCT. Most patients underwent bilateral lung transplantion (93%) and 79% of patients were on immunosuppressive therapy at the time of transplant. Overall survival after lung transplantation was 87% (CI% 80-95) at 1 year and 71% (CI% 61-83) at 5 years. Univariate analyses identified unrelated stem cell donor (p=0.007), immunosuppressive treatment (p=0.042), specifically corticosteroids (p=0.006), and extrapulmonary cGvHD (p=0.040) at the time of lung transplantation as significant risk factors for poorer survival. Main causes of death were infection in 20%, followed by respiratory failure (17%), secondary malignancy (13%) and haematological relapse (10%). Haematological relapse occurred in 4 patients (8%) at a median time of 16 months post-lung transplantation.

Conclusions: Lung transplantation is a valid treatment option for selected patients with end-stage pulmonary cGvHD, with long-term outcomes comparable to global registry data of patients transplanted for other indications. Patients with extrapulmonary cGvHD and those receiving corticosteroids at the time of transplantation demonstrated worse survival, indicating that active extrapulmonary disease should be optimised pretransplant to improve the chances of a good long-term outcome.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P633 DEPRESSION IN SURVIVORS OF HEMATOPOIETIC STEM CELL TRANSPLANTATION COMPARED WITH A MATCHED GENERAL POPULATION COMPARISON SAMPLE: THE MOSA STUDY

Bianca Wauben 1, M.W.M. van der Poel1, M. van Greevenbroek2, S. Koehler2, H.C. Schouten1, N. van Yperen2

1Maastricht University Medical Center, Maastricht, Netherlands, 2University Maastricht, Maastricht, Netherlands

Background: Psychological symptoms such as anxiety, cognitive problems, and depression are a prevalent complication among hematopoietic stem cell transplantation (HCT) survivors. Although psychological distress generally improves over time, a significant subgroup of HCT survivors experiences persistent depressive symptoms many years after transplant.

Aim:

(1) To quantify the prevalence and incidence rates of depressive symptoms and depression in patients treated with hematopoietic stem cell transplantation who survived at least 2 years, and to compare these with a matched reference group representing the general population.

(2) To identify potential predictors (cardiovascular, lifestyle, socioeconomic, and psychological factors) of differences between the HCT survivor group and the reference group.

Methods: Within the single-centre Maastricht Observational study of late effects after Stem cell trAnsplantation (MOSA) a validated Dutch version of the 9-item Patient Health Questionnaire (PHQ-9) was used to assess prevalent depressive symptoms at baseline. Pre-defined cut-off score of ≥10 was used as a dichotomous scoring system for defining clinically relevant depressive symptoms. Incident depressive symptoms were defined as no depressive symptoms at baseline, and presence of depressive symptoms (PHQ-9≥10) on at least one follow-up moment. A total of 121 HCT survivors were group-matched on a 1:4 ratio by age, sex, and level of education to the reference group selected from a large population based cohort resembling the general population. Cox survival analyses adjusted for different sets of covariates were used to study differences in risk of incident depressive symptoms between HCT survivors and the reference group.

Results: The mean age at time of transplant was 50.5 (SD 11.09) years, and the mean number of years of survivorship was 8.61 (SD 5.48) years. The majority of HCT survivors were treated with autologous HCT (62%, N = 75). Most HCT survivors were diagnosed with lymphoid malignancies (35%, N = 42), multiple myeloma (24%, N = 29), and acute leukemia (22%, N = 27). Table 1 The total duration of follow up in HCT survivors was 5.2 (SD 1.6) years vs. 8.6 (SD 2.3) years in reference group.

The prevalence of depression in HCT survivors at baseline (study participation) was 6.9%, and 3.4% in the reference group (p = .678). After adjustment for covariates (age, sex, educational level, cholesterol, kidney function, blood pressure, cardiovascular disease, type 2 diabetes status/glucose tolerance status, BMI, medication (glucose lowering, lipid modifying, and blood pressure lowering), smoking status, and alcohol use) were 2.3 times more likely to develop depressive symptoms compared to the general population (95%CI = 1.32 – 3.85, p = .003).

Table 1 Transplantation characteristics

Transplantation characteristics

HCT total % (N)

Auto HCT % (N)

Allo % (N)

Type of transplant

121

62% (75)

38% (46)

Sex/men

56.2% (68)

57.3% (43)

54.3% (25)

Mean age at time of transplant (years)

50.5 (SD 11.1)

51.4 (SD 10.6)

49.1 (SD 11.9)

Years after transplant (survivorship)

8.6 (SD 5.5)

8.9 (SD 5.4)

8.0 (SD 5.6)

Disease category

Lymphoma

34.7% (42)

48.0% (36)

13.0% (6)

Multiple myleoma

24.0% (29)

36.0% (27)

4.3% (2)

Acute leukemia

22.3% (27)

5.3% (4)

50.0% (23)

Conclusions: This study shows that a subgroup of HCT survivors may be more susceptible to depression as a long-term effect of being treated with HCT compared to a matched general population sample. Awareness and surveillance of depressive symptoms in HCT survivors is important to enable early detection to improve follow up care.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P634 PREGNANCY OUTCOMES IN WOMEN FOLLOWING HAEMATOPOIETIC STEM CELL TRANSPLANT FOR HAEMATOLOGICAL CONDITIONS

Michele Robinson1, Melanie Davies1, Anna L. David1, Panagiotis Kottaridis 1

1University College London Hospital, London, United Kingdom

Background: Improved survival rates for childhood haematological conditions have resulted in larger numbers of long-term survivors facing late effects, including gonadal dysfunction, infertility and pregnancy complications. We investigated pregnancy outcomes amongst female patients who have undergone HSCT for malignant and benign conditions.

Methods: Patients who conceived following HSCT were identified from the late effects clinic at the Reproductive Medicine unit, University College London Hospital (2000-2023). Information on patient demographics, medical history and pregnancy outcome was collected from electronic patient records.

Results: There were 64 pregnancies amongst 36 women were recorded. The age of the women at HSCT was wide (6 months to 32 years). 72% (n=26) had HSCT for malignant conditions (12 lymphoma, 10 leukaemia, 2 myelodysplasia, 1 Ewing Sarcoma with bone marrow metastases and 1 neuroblastoma), and the remaining 10 patients (28%) for aplastic anaemia (n=5), congenital immunodeficiency syndromes (n=3) and beta thalassaemia major (n=2). 66% (n=24) received an allograft transplant (6 sibling and 18 matched unrelated donor), of which 70% (n=14) underwent total body irradiation (TBI). 22% (n=8) received an autograft transplant, of whom 50% (n=4) underwent TBI. 11% (n=4) were lacking transplant details.

Most pregnancies (83%, n=53) were conceived spontaneously with 17% (n=11) achieved using assisted reproductive techniques (ART), 9 using donor oocytes. The mean age of women achieving spontaneous pregnancy was 26.5 years, compared with 34.8 years for those who achieved pregnancy using assisted conception. 88% (n=57) were singleton pregnancies, 8% (n=5) twins and 1 ectopic.

Pregnancy outcomes were missing in 7 cases. 8% (n=4) of pregnancies were terminated, one for foetal anomaly at 14 weeks gestation, one on medical advice as conception was too soon following HSCT, and two for patient choice.

Nearly one-quarter (23%, n=15) pregnancies resulted in miscarriage, with half (53%, n=8) before 12 weeks and 47% (n=7) between 14 - 21 week’s gestation. 56% pregnancies resulted in livebirth (n=36), with 61% (n=22) of live-births delivered at term (37-43 weeks gestation), with a mean birth weight of 3.35kg (range 2.49-4.89kg). Of preterm births, one (3%) was moderate to late preterm (34-37 weeks), [DA2] 19% (n=7[DA3]) were very preterm (28-34 weeks[DA4]) and 11% (n=4) were extreme preterm (<28 weeks). The mean birth weight for preterm infants was 1.39kg (range 0.78 – 3.2kg). One patient experienced a neonatal death at 2 weeks of age, due to preterm prelabour rupture of membranes (PPROM) at 21 weeks and delivery at 23 weeks gestation.

Conclusions: Spontaneous pregnancies can occur following HSCT and TBI, despite high rates of premature ovarian insufficiency.

A significantly increased risk for second trimester miscarriage and preterm birth was noted. This is likely to be due the effects of TBI resulting on reduced uterine elasticity, and radiation induced vascular damage causing utero-placental insufficiency. Maternal health factors, such as treatment induced nephrotoxicity or cardiomyopathy, may also contribute to poorer outcomes. Normal birth weights were observed in full term pregnancies.

Specialist obstetric care is required for this population, with consideration of cervical length, surveillance and need for cerclage to minimise risk of preterm birth.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P635 SAFETY AND EFFICACY OF IMMUNE CHECKPOINT INHIBITORS FOR SOLID ORGAN MALIGNANCIES AFTER ALLOGENEIC HSCT: A RETROSPECTIVE, MULTICENTRIC STUDY FROM THE EBMT TRANSPLANT COMPLICATIONS WORKING PARTY

Jan Brijs 1, Christophe Peczynski2,3, William Boreland2,3, Angela Cuoghi4, Johan Maertens1, Mohamad Mohty5, Nicolaus Kröger6, Philipp Nakov7, Annoek E.C. Broers8, Matthias Eder9, Concepcion Herrera Arroyo10, Martin Kaufmann11, Ron Ram12, Michel Schaap13, Olaf Penack3,14, Christian Könecke3,9, Ivan Moiseev3,15, Helene Schoemans1,3,16, Zinaida Peric3,17

1University Hospitals Leuven, Leuven, Belgium, 2EBMT Paris Study Office / CEREST-TC, INSERM UMR-S 938, Sorbonne University, Hôpital Saint Antoine, Paris, France, 3EBMT Transplant Complications Working Party (TCWP), Paris, France, 4Azienda Ospedaliero-Universitaria di Modena, Modena, Italy, 5Hôpital Saint Antoine, Paris, France, 6University Medical Centre Hamburg, Hamburg, Germany, 7University Hospital Schleswig-Holstein, Kiel, Kiel University, Kiel, Germany, 8Erasmus MC Cancer Institute, Rotterdam, Netherlands, 9Hannover Medical School, Hannover, Germany, 10Hospital Universitario Reina Sofia, Cordoba, Spain, 11Robert Bosch Krankenhaus, Stuttgart, Germany, 12Tel Aviv Sourasky Medical Centre, Tel Aviv, Israel, 13Radboud University Medical Centre, Nijmegen, Netherlands, 14Charité - Universitätsmedizin, Berlin, Germany, 15First Pavlov State Medical University of St Petersburg, St Petersburg, Russian Federation, 16KU Leuven - University of Leuven, Leuven, Belgium, 17University Hospital Centre Rijeka and School of Medicine, University of Rijeka, Rijeka, Croatia

Background: Immune checkpoint inhibitors (ICI) have shown significant and sustainable response rates in hemato-oncology, but are also known for their immune-related adverse events (irAE). Their use after allogeneic hematopoietic stem cell transplantation (alloHSCT) has been associated with an increased risk of developing graft-versus-host disease (GvHD) in Hodgkin lymphoma. In this study, we used the EBMT database to gather a cohort of patients to understand their potential toxicity when treating solid organ malignancies (SOM) after alloHSCT.

Methods: In this multicentric, observational, retrospective cohort study, the EBMT database was screened to identify all adult patients having undergone an alloHSCT for a haematological malignancy, who had been treated with an ICI (defined as anti-CTLA-4 and/or anti-PD-1/PD-L1 therapy) for a SOM after alloHSCT. Patients were excluded if they had received cord blood, more than one alloHSCT or DLI before ICI, ex-vivo T cell depletion or ICI before transplantation. EBMT centres were also invited to report any additional patients meeting these criteria. All participating centres provided detailed information regarding ICI therapy and outcome. Acute and chronic GvHD were graded according to the Glucksberg criteria and the NIH criteria respectively, and response to ICI was based on the RECIST criteria.

Results: Twenty patients (7 female (35%) and 13 male (65%) patients) from 11 EBMT centres were identified (patient characteristics: table 1). The most frequent SOMs treated with ICI were lung carcinoma and melanoma. Median time between alloHSCT and diagnosis of the SOM was 5.6 years (range: -0.8-19.6 years), with two patients diagnosed with a SOM before alloHSCT. The most frequently used ICIs were pembrolizumab and nivolumab. Median time between transplantation and first ICI administration was 6.1 years (range: 0.4-23.4 years). At the time of ICI administration, only two patients (10%) had active chronic GvHD.

After ICI administration, only one patient developed treatment-emergent new onset grade III acute GvHD, 6 days after administration, leading to discontinuation of ICI treatment. Another patient developed treatment-emergent (progressive/recurrent) severe chronic GvHD, 324 days after administration, without ICI treatment discontinuation. In addition, six patients developed one or more irAE (table 1), leading to discontinuation of ICI treatment.

The best response to ICI was complete response in 1 patient, partial response in 7 patients, stable disease in 1 patient and progressive disease in 8 patients (missing n=3). The dismal overall survival is shown in figure 1, with 15 patients dying by the end of the follow-up period with the majority succumbing to a secondary malignancy (11/15).

Initial haematological disease treated with alloHSCT

N (%)

Solid organ malignancy treated with ICI

N (%)

GvHD after ICI

N

Non-Hodgkin lymphoma (NHL)

6 (30)

Lung carcinoma

7 (35)

Acute GvHD (new onset)

1

Myelodysplastic syndrome (MDS)

3 (15)

Melanoma

6 (30)

Chronic GvHD (progressive/recurrent)

1

Myeloproliferative neoplasm (MPN)

3 (15)

Renal cell carcinoma

1 (5)

Hodgkin lymphoma (HL)

2 (10)

Bladder carcinoma

1 (5)

Acute myeloid leukemia (AML)

2 (10)

Colon carcinoma

1 (5)

Types of irAE

N

Chronic lymphoid leukemia (CLL)

2 (10)

Sarcoma

1 (5)

Lung

4

Multiple myeloma (MM)

1 (5)

Merkel cell carcinoma

1 (5)

Gastro-intestinal

3

MDS/MPN overlap syndromes

1 (5)

Pleural mesothelioma

1 (5)

Liver

2

Laryngeal carcinoma

1 (5)

Endocrinopathy

2

Skin

1

GvHD after alloHSCT

N (%)

Other

2

Acute GvHD

9 (45)

Types of ICI used

N (%)

Chronic GvHD

9 (45)

Pembrolizumab

10 (50)

Nivolumab

7 (35)

Ipilimumab + nivolumab

2 (10)

Avelumab

1 (5)

Table 1: Patient characteristics

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (83)

Figure 1: Overall survival, in months, after ICI administration.

Conclusions: Based on this case series, we see a low frequency of GvHD following ICI use after alloHSCT and a comparable frequency of irAE compared to other patients treated with ICI, with about one third of patients discontinuing treatment due to adverse events.

Considering the dismal prognosis of these patients, we recommend to further monitor the administration of ICI through real-life data registries and to develop prospective trials to identify new therapeutic strategies to improve the prognosis of patients needing treatment for SOM after alloHSCT.

Clinical Trial Registry: S64876.

Disclosure: Jan Brijs has no conflicts of interest to declare.

Christophe Peczynski has no conflicts of interest to declare.

William Boreland has no conflicts of interest to declare.

Angela Cuoghi has no conflicts of interest to declare.

Johan Maertens has no conflicts of interest to declare.

Mohamad Mohty has no conflicts of interest to declare.

Nicolaus Kröger has no conflicts of interest to declare.

Philipp Nakov has no conflicts of interest to declare.

Annoek E.C. Broers has no conflicts of interest to declare.

Matthias Eder has no conflicts of interest to declare.

Concepcion Herrera Arroyo has no conflicts of interest to declare.

Martin Kaufmann has no conflicts of interest to declare.

Ron Ram has no conflicts of interest to declare.

Michel Schaap has no conflicts of interest to declare.

Olaf Penack has no conflicts of interest to declare.

Christian Könecke has no conflicts of interest to declare.

Ivan Moiseev has no conflicts of interest to declare.

Hélène Schoemans reports having received personal fees from Incyte, Janssen, Novartis, Sanofi and from the Belgian Hematological Society (BHS), as well as research grants from Novartis and the BHS, all paid to her institution. She has also received non-financial support (travel grants) from Gilead, Pfizer, the EBMT (European Society for Blood and Marrow transplantation) and the CIBMTR (Center for International Bone Marrow Transplantation Research).

Zinaida Peric has no conflicts of interest to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P636 POST-TRANSPLANT CYCLOPHOSPHAMIDE IS ASSOCIATED WITH REDUCED RATE OF SECONDARY MALIGNANCIES POST ALLOGENEIC STEM CELL TRANSPLANT

Nihar Desai 1, Mariana Pinto Pereira1, Mats Remberger2, Rajat Kumar1, Dennis Kim1, Auro Viswabandya1, Arjun Law1, Wilson Lam1, Ivan Pasic1, Armin Gerbitz1, Igor Novitzky-Basso1, Jonas Mattsson1, Fotios Michelis1

1Princess Margaret Cancer Centre, Toronto, Canada, 2Uppsala University and and KFUE, Uppsala University Hospital, Uppsala, Sweden

Background: Advancements in allogeneic hematopoietic cell transplantation (allo-HCT) over the past decade have significantly enhanced patient outcomes with more long-term survivors. However, these survivors face an increased risk of secondary malignancies (SM) post-transplant.

Methods: Patients were aged 18-75 years at allo-HCT, transplanted at our center from October 2015 to May 2021. Data collected included patient and transplant characteristics, the instances and locations of SM, and treatment and outcomes of patients with SM. Baseline variables were summarized using descriptive statistics. Cumulative incidence (CumI) of SM was determined using the competing risk method with death as the competing event. Overall Survival (OS) was estimated using the Kaplan Meir method. A p-value of <0.05 was considered statistically significant.

Results: Among 884 patients post allo-HCT, 73 (8.3%) developed SM, including 19 (26%) with non-metastatic squamous cell carcinoma (SCC) and basal cell carcinoma (BCC) of the skin (Table 1). The median age at diagnosis of SM was 62 years (range, 53-68). The median time from allo-HCT to development of SM was 19.3 months (range, 3.1 – 30.5), and 26.6 months (range, 11.6 - 40.7 months) after excluding patients with post-transplant lymphoproliferative disorder (PTLD). The median follow up post-transplant was 32.9 months (range, 8.3 – 51.8).

The CumI of SM for the entire cohort was 6.8% at 3 years and 9.4% at 5 years. After excluding non-metastatic skin BCC and SCC, these figures were 5.4% at 3 years and 6.7% at 5 years. Acute leukemia was the most common indication for transplant (59%). Most patients (76.3%) received reduced intensity conditioning (RIC), and 637 (72%) received dual T-cell depletion using Anti-Thymocyte Globulin (ATG) with Post-Transplant Cyclophosphamide (PTCy) as GVHD prophylaxis. 721 patients (81%) received total body irradiation, with 621 (70%) receiving ≤200 cGy.

The use of PTCy was associated with a reduced risk of SM in both univariate (HR 0.56; p=0.025) and multivariable analysis (HR 0.55; p=0.02). Patients with any history of smoking before allo-HCT had a higher likelihood of developing SM on univariate (HR 1.89 p=0.009) and multivariable analysis (HR 1.73; p=0.02). The median follow up after diagnosis of SM was 17 months (range, 6-47). The OS after diagnosis of SM was 53% (37.8-66.2) at 3 years and 49.9% (34.5-63.5) at 5 years for the 54 patients with SM excluding skin SCC/BCC.

Ten patients with SM subsequently developed an additional SM at a median of 22.5 months (range, 8-44.7) after the first SM, and 39.1 months (range 24.7-51.5) after allo-HCT. Half of them had PTLD as their first SM. Nine of these patients are alive at the time of last follow up.

Table 1: Secondary malignancies post allogeneic stem cell transplant

Type of Malignancy

n

Months to SM (median, IQR)

Treatment for SM

Surgery

Chemotherapy/Immunotherapy

Palliation

Hematological malignancies

• t-AML

• Multiple Myeloma

• SMM

• T-ALL

• PTLD

39

4

2

3

1

29

12 (3.1 – 30.4)

15.9 (12.1 – 27.1)

21.9 (15.7 – 29.2)

30.5 (17.8 – 46.6)

22.5 (-)

3.2 (2.4 – 6.4)

1

2

1

24

3

Non- hematological malignancies

34

21.9 (11.6 – 40.7)

28

6

2

Skin Cancers

• BCC

• SCC

• Melanoma

20

6

13

1

23.2 (12.4 - 40.5)

29.2 (16.2 – 43.8)

22.2 (12.3 – 36.8)34.3 (-)

6

12

1

1

Renal cell carcinoma

2

5.5 (4.3 – 7)

1

1

Lung cancer

2

42 (34.8 – 49.8)

1

2

Carcinoma endometrium

3

30.9 (22.8 – 42.5)

2

2

CUP

1

27.1 (-)

1

Thymoma

1

21.9 (-)

1

Thyroid cancer

1

47.2 (-)

1

Carcinoma prostate

3

44.5 (34.8 – 56.8)

3

Tonsillar carcinoma

1

45.9 (-)

1

Total

73

19.3 (3.1-30.5)

28

34

5

  1. BCC: Basal cell carcinoma; CUP: carcinoma of unknown primary; PTLD: Post-transplant lymphoproliferative disorder; SMM: Smoldering multiple myeloma; t-AML: therapy related acute myeloid leukemia; T-ALL: T-cell acute lymphoblastic leukemia.

Conclusions: The use of PTCy results in a reduced incidence of SM following allo-HCT. This may be related to the significant reduction in GVHD with PTCy use. Additionally, patients with a history of smoking pre-transplant have a higher likelihood of developing SM. These findings warrant confirmation in a larger cohort. Moreover, the survival of allo-HCT patients with SM is favorable, warranting routine long-term monitoring and follow up.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P637 MOBILE HEALTH IN THE MANAGEMENT OF ALLOGENIC STEM CELL TRANSPLANT RECIPIENTS (MY-MEDULA STUDY): PRELIMINARY RESULTS OF THE CLINICAL TRIAL

Sara Redondo 1, Anna De Dios1, Marina Vallve1, Jordi Real1, Merce Triquell1, Olga Aso1, Albert Esquirol1, Me Moreno-Martinez1, Mireia Riba1, Julia Ruiz1, Eva Tobajas1, Jorge Sierra1, Javier Briones1, Rodrigo Martino1, Mar Gomis-Pastor1, Irene Garcia-Cadenas1

1Hospital de la Sant Creu i Sant Pau, IIB-Sant Pau and José Carreras Leukemia Research Institutes, Universitat Autónoma de Barcelona, Barcelona, Spain

Background: Advances in allo-HCT have led to an increasing number of transplant survivors. However, post-transplant survival is challenged by several complications.

MHealth has proven to be an effective strategy improving chronic illness care without adding costs. MY-Medula is a new mHealth tool designed to improve the post-HCT recovery, offering personalized care by an interdisciplinary team.

Having demonstrated the feasibility, and the degree of satisfaction by patients and professionals in a previous pilot study, this analysis describes the preliminary usability results within the ongoing clinical trial (CT).

Methods: Prospective, randomized single-center CT carried out in a tertiary university hospital by an interdisciplinary team (IIBSP-EME-2019-44; approved by the local ethics commitee).

MY-Medula has several funcions, including a medication diary, self-monitoring of vital signs, physical exercise, food intake tracker, mood log and adverse effects register. In addition, it includes a bidirectional patient-health professional real time messaging service. Early intervention is provided based on the individualized evaluation of patient’s records.

We assessed usability through App records, employing the PROs (patient-reported outcomes), in the intervention group (IG). Additionally, we evaluated treatment adherence using ARMSe questionnaire at baseline, and 6 months, the degree of empowerment with CAMBADOS at baseline across the entire cohort, and satisfaction among professionals.

Results: Between August 2022 and November 2023, 75 recipients were offered to participate in the CT within 1month post-transplant discharge; 6 (8%) refused due to challenges with the technology. Consequently, 69 participants were included: 38 (55%) in the control group (CG), and 31(45%) in the IG (Table 1). The CAMBADOS questionnaire obtained a median empowerment score at baseline of 67.8 (considered positive >50), with only one patient below this score in the CG.

At data cutoff-November 2023, with a median follow-up of 277 days (range, 6-479), a total of 929 text messages were received, most frequently related to filling prescriptions (34%), symptom description (25.3%) and doubts about medication (13.2%), and a total of 1782 self-monitoring records were created and reviewed by the nursing and/or the medical team. Most of them were related to blood pressure-heart rate (48%), and weight (25%). The evolution of the different records and messaging decreased over time, being 55.6% and 100% in the first trimester, and 14.8% and 37% in the third. Interestingly, no cases of low adherence to medication were identified in the 63% (n=41) of the patients who fulfilled the ARMSe questionnaire over time.

Regarding healthcare professionals, 12 team members responded to the usability and satifaction survey. Usability was assessed through 6 questions, resulting in an average rating of 4.1 out of 5. Despite 4 (33%) individuals feel overloaded by the platform workflow, all participants would recommend the tool to other healthcare professionals.

Variables

Control Group N=38

Intervention Group N=31

p

Age, median [range]

54 [19-68]

57 [47-64]

0.28

Sex, male

18 (47.4%)

17 (54.8%)

0.62

Underlying disease

-Acute myeloid leukemia

19 (50%)

11 (35.5%)

0.33

- Acute lymphoblastic leukemia

7 (18.4%)

6 (19.4%)

1.00

Complete response

26 (68.4%)

23 (74.2%)

0.79

Source, Peripheral blood

36 (94.7%)

30 (96.8%)

1.00

Donor Type

-Related HLA identical

12 (31.6%)

6 (19.4%)

0.28

-Unrelated identical

14 (36.8%)

20 (64.5%)

0.03

Haploidentical

7 (18.4%)

4 (12.9%)

0.74

-Unrelated mismatch-

4 (10.5%)

1 (3.2%)

0.37

Conditioning regimen

-Myeloablative

21 (55.3%)

12 (38.7%)

0.23

CD34+ cells infused (x 106/kg), median [range]

6.0 [4.4-6.8]

6.0 [0.8-8.1]

0.12

GVHD prophylaxis

-Sirolimus-Tacrolimus

0 (0%)

1 (3.2%)

0.45

-PTCy

36 (94.7%)

28 (90.3%)

0.65

Conclusions: The introduction of mHealth as a complementary tool in the follow-up of allo-HCT recipients is feasible with less than 10% of adult patients refusing it due to barriers with digital technology. Despite encouraging rates of usability during the first 3 months, sustained patient engagement remains a challenge. Conversely, healthcare professionals, though managing additional tasks, express satisfaction with the new tool.

Disclosure: The study has the collaboration of the pharmaceutical company Amgen S.A. which assumed the cost of the technological development and those derived from the implementation of the tool. Amgen had no influence on the study design, data collection or data analysis.

15: Non-infectious Late Effects, Quality of Life and Fertility

P638 HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR PATIENTS FROM AREAS OF CONFLICT: THE KING HUSSEIN CANCER CENTER TRANSPLANTATION PROGRAM EXPERIENCE

Salwa Saadeh 1,2, Haya Hamarsha1, Razan Odeh3, Mohammad Makoseh1,2, Zaid Abdel Rahamn1

1King Hussein Cancer Center, Amman, Jordan, 2University of Jordan, Amman, Jordan, 3An-Najah National University, Nablus, Palestine

Background: Hematopoietic stem cell transplantation (HSCT) is a potentially curative modality for a number of hematological diseases. However, it is a complex treatment modality that requires the presence of a rigorous infrastructure and program. Also, it is a costly and lengthy treatment, limiting access to many patients, more so for patients originating from areas of conflict. King Hussein Cancer Center (KHCC) has a well-establish HSCT program, that serves as the only program in Jordan, and one of the few in the area. It has been serving Jordanian and non-Jordanian patients since its establishment in 2003, with increasing numbers of patients originating form areas of conflict within the MENA region during the past 16 years. The aim of this study is to describe the role of the BMT program at KHCC in managing patients from areas of conflict.

Methods: This is a descriptive study about BMT procedures in patients from areas of conflict in the MENA region, namely Palestine, Iraq, Syria, Libya and Yemen. We obtained data from the KHCC data registry for patients treated and/or transplanted at KHCC. Collected data included number of patients, type of transplant, nationality, survival and source of financial coverage. IRB approval was obtained. Simple descriptive statistics were applied.

Results: Over a period of 16 years (2007-2022), a total of 45,966 new patients were treated at KHCC, 80% Jordanian, 17.2% non-Jordanians from areas of conflict, and only 2.2% from other countries. During the same time-period, 2,404 HSCTs were performed, 78.7% for Jordanians, 17.5% for patient from areas of conflict and only 3.8% from other countries. There was significant fluctuation in the rates of BMT procedures done per country per year, most noticeable for patients form Palestine, mainly related to unsteady referral rates depending on financial coverage availability. The main source of financial coverage was through the country-of-origin referring governmental organizations (65.7%), or The King Hussein Cancer Foundation Good Will Fund (11.9%) and the Jordanian Ministry of Health expenditures (17%). For allogeneic transplant patients, survival rates were not statistically different; 1- and 2-year OS were 76.4% and 70.1% versus 80.0% and 73.0% for patients form Jordan and from areas of conflict, respectively, p=0.54 (Table 1). Outcome data, however, are difficult to interpret due a significant number of patients from areas of conflict being censored due to loss of follow-up, a total of 52.5% of patients. Median follow-up for patients lost to follow-up in months varied per country of origin; 29.5, 37, 14 and 15 months for patients from Palestine, Iraq, Syria and Libya, respectively. This limits the ability to diagnose or follow patients for longer term outcomes such as cGVHD, and late organ dysfunction.

Conclusions: Patients living in, or originating from, areas of conflict who undergo HSCT are less likely to receive the expected longitudinal care needed for follow-up, and long-term outcomes are difficult to interpret. Although HSCT at KHCC contributes to delivery of adequate treatment to these patients, this remains limited, mainly due to lack of financial coverage of this costly treatment.

Nationality

One year OS Rate (95% C.I)

Two years OS Rate (95% C.I)

Jordanian

76.4% (73.9%, 78.8%)

70.1% (67.4%, 72.7%)

Areas of conflict

80.0(74.7%, 84.8%)

73.0% (66.9%, 78.7%)

Iraqi

87.0% (70.5%, 97.3%)

87.0% (70.5%, 97.3%)

Libyan

84.7% (75.9%, 91.8%)

76.8% (64.8%, 86.9%)

Palestinian

76.8% (68.4%, 84.3%)

69.1% (59.8%, 77.7%)

Syrian

76.7% (58.6%, 90.8%)

64.9% (43.7%, 83.4%)

Yemeni

74.0% (45.6%, 94.4%)

74.0% (45.6%, 94.4%)

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P639 AUTOIMMUNE NEUTROPENIA AFTER ALLOGENIC BONE MARROW TRANSPLANTATION DUE TO ANTIBODIES SPECIFICALLY TARGETING THE HNA-1A ANTIGEN IN PEDIATRIC PATIENTS

Maria Luz Uria Oficialdegui 1, Enric Casanovas2, Carme Canals2, Nuria Nogues2, Marta Rodriguez2, Cecilia Gonzalez2, Melissa Panesso1, Laura Alonso1, Lucia Garcia3, Monica Linares2, M. Isabel Benitez-Carabante1, Cristina Diaz-de-Heredia1

1Hospital Universitari Vall d´Hebron, Barcelona, Spain, 2Banc de Sang i Teixits, Barcelona, Spain, 3Hospital Universitari Son Espases, Palma de Mallorca, Spain

Background: Autoimmune neutropenia (AIN), a rare complication following hematopoietic stem cell transplantation (HSCT) results from the development of antibodies targeting Human Neutrophil allo-Antigens (HNAs) in donor-derived neutrophils, leading to an increased risk of infections and potential graft failure (GF). HNAs are usually associated with neonatal alloimmune neutropenia and transfusion-related acute lung injury. We report the first two documented cases of AIN post-HSCT due to antibodies specifically targeting the HNA-1a antigen.

Methods: Retrospective analysis of clinical and laboratory patient records. Antibody granulocyte-immunofluorescence test and LABScreenä Multi assay (Luminex® xMAP® technology) were used for screening HLA and HNA antibody and to identify anti-HNA-1a specificity. HNA-1 genotyping of patient and donor was performed.

Results: The underlying disease, conditioning intensity, post-HSCT course, diagnosis timeline, and evolution was different in both cases.

Patient 1: 6-years-old boy, with X-linked adrenoleukodystrophy underwent a bone marrow matched unrelated donor (MUD) transplant, following a myeloablative conditioning regimen (busulfan/fludarabine) and Thymoglobulin. GVHD-prophylaxis: cyclosporine/methotrexate. Due to the absence of neutrophil engraftment on day +28, with successful engraftment of other cell lines, an investigation of granulocytic-antibodies was conducted. Direct test confirmed the presence of antibodies attached to the surface of patient’s granulocytes. Specific antibodies against HNA-1a antigen (IgM>>IgG), were identified in serum. Patient’s HNA-1a genotyping was negative, while the donor tested positive. Screening for HNA-antibodies in the patient’s pre-HSCT serum sample was negative. Patient received treatment with steroids, rituximab, high-dose Intravenous Immunoglobulin (IVIG), and sirolimus. Finally neutrophil engraftment was successful on day +54 after-HSCT. Unfortunately, severe pancytopenia and secondary GF occurred, requiring a second HSCT 6 months after the first transplant. Second donor tested negative for HNA-1a. After the second transplant, neutrophil counts reached normal levels on day +15, achieving full donor chimerism. Subsequent follow-up revealed a gradual decline in IgM levels and the stability of anti-HNA-1a IgG antibodies. Unfortunately, the patient died due to neurological progression of his underlying disease.

Patient 2: 18-years-old girl, with Dyskeratosis Congenita underwent a CD34+ selected peripheral blood MUD transplant, following a reduced-intensity conditioning regimen (fludarabine/cyclophosphamide/TBI) and Thymoglobulin. GVHD-prophylaxis: cyclosporine/MMF. Successful engraftment on day +11. Post-HSCT course was challenging, marked by several non-hematological complications. Two years post-transplant she was diagnosed with immune thrombocytopenia and started Thrombopoietin receptor agonist(AR-TPO) treatment, leading to a stabilized platelet count (>100x10^9/L). One year later, persistent and severe neutropenia was observed with recurrent urinary tract infections. Direct test was highly positive due antibodies attached to the surface of patient’s granulocytes, along with HNA-1a specific antibodies (IgM/IgG) in the serum. Patient’s HNA-1a genotyping was negative, while the donor tested positive. No HNA-antibodies were present in pre-HSCT serum sample. Patient received G-CSF treatment, alongside rituximab and high-dose IVIG after AIN diagnosis. Currently, she exhibits complete donor-chimerism and is under close monitoring.

Conclusions: AIN post-HSCT is a rare and challenging complication. Our cases illustrate the development of anti-HNA-1a antibodies against donor-specific neutrophil antigens, highlighting the need for careful monitoring. Further research is necessary to better understand the pathogenesis of this condition, the potential relation with graft failure and its prevention and treatment.

Disclosure: none.

15: Non-infectious Late Effects, Quality of Life and Fertility

P640 IONA - INTERDISCIPLINARY ONCOLOGICAL FOLLOW-UP CLINIC IN VIENNA

Alexandra Böhm 1, Sabine Burger1, Anna Pucher2, Sabina Ziomek2, Arno Hraschan2, Sophie Lindermann2, Barbara Schröder-Aranyosy2, Felix Keil1

1Hanusch Hospital, Vienna, Austria, 2Gesundheitszentrum Mariahilf, Vienna, Austria

Background: Cancer in childhood and adolescence can be cured in many cases nowadays. In Vienna, patients are treated either in the St. Anna Children’s Hospital or at the University Clinic for Pediatrics and Adolescent Medicine (Neuro-Oncology). Short-term follow-up is usually performed in the children’s hospitals. Afterwards patients had to organize the necessary examinations themselves for many years. This so-called transition was not successful in many cases, as the detection and treatment of therapy and disease-associated secondary diseases requires appropriate expertise.

Methods: “IONA - Interdisciplinary Oncological Follow-Up Clinic” is now offering age-appropriate, medical and psychosocial long-term follow-up since the beginning of 2020. A team of one experienced hemato/oncologist, two case-managers, two clinical psychologists and a social worker are offering the so-called “survivors” a specialized care, including medical check-ups tailored to the treatment and advices on risk factors in order to identify and treat possible late effects early on. In addition, there is psychosocial care and, if necessary, neuropsychological diagnostics. The health center where IONA is located also offers an optimal multi-professional network for this group of patients.

Results: More than 500 survivors have been transitioned since IONA started in early 2020, 37 of them underwent allogeneic stem cell transplantation (SCT), 26 of them autologous SCT. The majority of them suffered from CNS tumor, acute leukemia or lymphoma and were assigned directly by the hospitals, which guarantees continuous care. There is also regular exchange between the attending physicians as well as the psychologists, social workers and administrative employees. During the COVID pandemic, video and audio telephony were also used to hold joint conversations.

Conclusions: IONA offers interdisciplinary, standardized long-term follow-up care for patients from the age of 18 who had a hemato/oncologic disease in their childhood, adolescence or young adulthood and who have completed their medical therapy and short-term follow-up care at the responsible clinic. IONA accompanies patients in their transition process through close cooperation with the referring hospitals.

Disclosure: No conflict of interest.

15: Non-infectious Late Effects, Quality of Life and Fertility

P641 RAISING FOLLOW-UP AFTER CELL THERAPY TO THE NEXT LEVEL: A TRANS-REGIONAL, TRANS-SECTORAL, INTERDISCIPLINARY RESEARCH PROJECT

Katharina Egger-Heidrich 1, Martin Schneider1, Gabriele Müller1, Roman Schmädig1, Franziska Schmidt1, Lynn Leppla2,3, Sabina De Geest3, Alexandra Teynor4, Markus Wolfien1,5, Martin Sedlmayr1,5, Mathias Hänel6, Anke Morgner6, Vladan Vucinic7, Jochen Schmitt1, Johannes Schetelig1, Uwe Platzbecker7, Martin Bornhäuser1, Jan Moritz Middeke1

1University Hospital Carl Gustav Carus, Dresden, Germany, 2University of Freiburg, Faculty of Medicine, Freiburg, Germany, 3Institute of Nursing Science, University of Basel, Basel, Switzerland, 4Institute for agile Software Development, University of Applied Sciences Augsburg, Augsburg, Germany, 5Center for Scalable Data Analytics and Artificial Intelligence (ScaDS.AI), Dresden/Leipzig, Dresden, Germany, 6Klinikum Chemnitz, Chemnitz, Germany, 7University Hospital Leipzig, Leipzig, Germany

Background: Allogeneic stem cell transplantation (alloSCT) and CAR-T-Cell therapies (CAR-T) led to a significant increase of survival in hematologic malignancies during the last decade. Both procedures are associated with substantial toxicities, which go beyond the inpatient stay. Rehospitalisations are common, especially within the first year after cell therapy. A close follow-up under supervision of the cell therapy center is therefore mandatory. This is challenging as many patients live far away from the center and, in view of rising patient numbers, stresses hospital resources. The study presented here, named “Cross-sectoral care of patients with hematological diseases following innovative cell therapySPIZ”, aims to reduce the mortality and hospital readmission rate of patients treated with cell therapy.

Methods: As a multi-disciplinary team of hematologists, nursing and public health specialists, we developed a digitally supported follow-up-program for alloSCT and CAR-T patients after discharge from hospital. Efficacy will be evaluated in a prospective, 1:1 (SPIZ versus standard of care) randomized clinical trial (RCT) that will be conducted in all three cell therapy centers in Saxony. The combined primary endpoint will be rehospitalization and mortality. The cooperation with the region’s largest statutory health insurance allows an additional health economic evaluation.

Results: All patients (n=302) will receive regular outpatient clinic visits as indicated by their general condition and received therapy. Patients within the interventional arm (IA; n=151) are provided with a case manager who organizes all outpatient clinic visits, examinations, home visits by nurses. In particular during early follow up, IA patients receive regular home visits by a nurse specialist for hematology/cell transplantation checking the general condition, vital parameters, medication adherence, and obtain laboratory examples. Additionally, all IA patients are asked to document their vital signs and symptoms into a follow-up app (SMILe) which is read out by the case manager every working day. Furthermore, to consult psycho-oncologists, social workers and hematology specialists, video consultations are provided to all IA patients. Finally, all IA patients are discussed regularly in panel discussions involving all stakeholders (center physicians, oncologists in practice, nurses, psycho-oncologists, social workers). The RCT is running from from January 1st, 2024 to May 31th, 2026.

Conclusions: We aim to improve outcome after cell therapy by an intensive multimodal, interdisciplinary care program including digital tools. The described program is in general transferable to other regions and indications. After positive evaluation of this RCT, permanent implementation of this follow-up programme is planned.

Disclosure: Development and Implementation of this study is funded by the innovation committee of the Joint Federal Committee Germany (Funding number: 01NVF22108). Development and evaluation of the study is supported by the AOK plus, the largest statutory health insurance in the region.

15: Non-infectious Late Effects, Quality of Life and Fertility

P642 SEVERE POLYAUTOIMMUNITY AFTER ALLOGENIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: POTENTIAL ROLE OF THE GENETIC BACKGROUND AND CHIMERISM LOSS

Giorgio Costagliola1, Alessandro Di Gangi1, Eva Parolo 1, Chiara Lardone1, Sayla Bernasconi1, Sofia D’Elios1, Nina Tyutyusheva1, Gabriella Casazza1, Mariacristina Menconi1

1Azienda Ospedaliero Universitaria Pisana, Pisa, Italy

Background: Autoimmune diseases following hematopoietic stem cell transplantation (HSCT) have been largely described both after autologous and allogenic HSCT, and consist primarily in autoimmune cytopenias and thyroid diseases, but secondary rheumatological diseases and systemic autoimmune diseases have also been reported.

Methods: We describe the case of a patient who underwent HSCT for acute ALL and further developed multiple autoimmune disorders in the years following HSCT.

Results: The patient is a female who was diagnosed with pro-B acute ALL MLL/AF4 positive with t(4;11) rearrangement at 2 moths of age. She was treated according to the Interfant ’06 protocol and, at the age of 10 months, she underwent allogenic HSCT (matched unrelated donor, 10/10) from cord blood. The conditioning regimen consisted in Busulfan, Thiotepa and Fludarabine and the GvHD prophylaxis was performed with anti-thymocyte globulin, cyclosporine and metilprednisolone. The patient developed only grade I acute cutaneous GvHD treated with corticosteroids, and did not experience relevant infections. At the first evaluation, she achieved full-donor chimerism, that was maintained during the first 8 years of follow-up. During the first months after HSCT, our patient developed chronic cutaneous GvHD (grade I), treated with topical corticosteroids, followed by the development of vitiligo and lichen-like lesions.

In the first year after HSCT she was also diagnosed with Graves-Basedow disease and received treatment with metimazole for the following 5 years. Ten years after HSCT, she was diagnosed with antinuclear antibody (ANA) positive polyarticular juvenile idiopathic arthritis (JIA). Concomitantly, reduced basal cortisol levels in association with positivity of anti-hypophysis autoantibodies were detected. Therefore, the patient underwent ACTH stimulation testing, that confirmed reduced pituitary function, and brain and hypophysis MRI, that showed elements consistent with the diagnosis of autoimmune hypophysitis. Post-HSCT chimerism was also repeated, showing a marked reduction compared to previous controls (50% donor chimerism). Therefore, the patient underwent treatment with methotrexate, followed by the introduction of etanercept after the first disease relapse, and replacement therapy with hydrocortisone.

Given the extended autoimmune phenotype of our patient and the history of early-onset ALL, targeted whole exome sequencing was performed on germline DNA extracted by cutaneous fibroblasts. The analysis is currently ongoing.

Conclusions: This case offers several interesting points for discussion since it provides, to our knowledge, the most extended autoimmune spectrum observed after HSCT in a single patient and the only reported case of autoimmune hypophysithis after HSCT. Moreover, although genetic testing is currently ongoing, it highlights the need for searching for underlying causes responsible both for the early development of malignancies and immune dysregulation in this category of patients, as the knowledge in the field of inborn errors of immunity with immune dysregulation is rapidly expanding. Finally, the chimerism reduction observed in this patient opens interesting physiopathological and clinical perspectives. As other cases of late-onset autoimmunity after HSCT have been reported after the reduction of chimerism, the collection of data from this rare subset of patients could provide new fundamental insights in the pathogenesis of autoimmunity following allogenic HSCT, and potentially lead to personalized screening and prevention strategies.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

15: Non-infectious Late Effects, Quality of Life and Fertility

P643 A CASE REPORT OF A REFRACTORY EVANS SYNDROME AFTER ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION

Viktoria Yankova 1, Krasen Venkov1, Andriyana Bankova1, Kameliya Milcheva1, Penka Ganeva1, Jonka Lazarova1, Georgi Mihaylov1, Galya Kondeva1

1Specialized Hospital for Active Treatment of Hematological Diseases, Sofia, Bulgaria

Background: Autoimmune cytopenias (AIC) post allogeneic hematopoietic cell transplantation (allo-HCT) are exceedingly rare complications. Risk factors identified for AIC following allo-HCT include donor mismatch, major ABO incompatibility, CMV reactivation, presence of GvHD and myeloablative conditioning (MAC). No standardized treatment for post allo-HCT AIC exists, with most therapeutic knowledge derived from primary AIC cases.

Methods: Here, we report on a patient with Evans syndrome following allo-HCT, who was successfully treated with third line treatment with Sirolimus.

Results: A 44-year-old male patient was diagnosed with intermediate-risk acute myeloid leukemia (AML) according to European LeukemiaNet (ELN) guidelines. The patient underwent allo-HCT in first complete remission (MRD negative) in January 2022 from MUD (10/10) male donor, major ABO incompatible, following MAC (Cy/TBI). From April to June 2022 the patient experienced multiple CMV reactivations, initially treated with Vangancyclovir. Due to severe pancytopenia CMV-treatment was changed to Foscarnet, which led to acute renal injury and therefore Foscarnet was also discontinued. Tapering and cessation of prophylactic immunosuppression with calcineurin inhibitor 120 days post allo-HCT in the absence of GvHD led to effective CMV control. Following pneumonia caused by parainfluenza virus, the patient was readmitted in August 2022 with clinical and laboratory findings of autoimmune hemolytic anemia (AIHA; Hb < 70 g/l; Reticulocytes – 67 G/L.; Haptoglobin < 0.08 g/L.; LDH – 700 U/L; Positive direct Coombs – IgG 4 + , complement 4 + ). Bone marrow analysis showed normocellular BM with full donor chimerism. Therapy with intravenous immunoglobulin was ineffective. Furthermore, a decrease in platelet count to 40 G/L was documented and given the absence of other causes for thrombocytopenia and the preserved regenerative potential of the bone marrow the condition was interpreted as immune thrombocytopenia. This, in combination with AIHA, led to the diagnosis of Evans syndrome. Therapy with Prednisolone and Rituximab resulted only in partial response and triggered CMV reactivation, requiring Cidofovir treatment, which again led to acute kidney injury and pancytopenia. A recurrence of AIHA in November 2022 was treated with Bortezomib. Haptoglobin and LDH levels return to normal, however hemoglobin did not improve significantly. Secondary poor graft function was evident in January 2023, marked by low neutrophil (Neu < 1 G/l) and platelet counts (Plt < 50 G/l), hypo-/aplastic bone marrow, and preserved donor chimerism 92%. The patient received T-cell repleted peripheral blood stem cell (PBSC) boost. By day +32 following the infusion of the PBSC boost, the patient showed improved reticulocyte, neutrophil, and platelet levels, with restored bone marrow cellularity and full donor chimerism. However, another AIHA spike occurred, featuring absolute B-lymphopenia in peripheral blood and increased T-cell infiltration in bone marrow aspirate. Third-line therapy with Sirolimus resulted in normalization of hemoglobin levels within the first month.

Conclusions: Our case demonstrates the diagnostic and treatment challenges opposed by a rare complication of Evans syndrome in allo-HCT recipient, who was successfully treated with Sirolimus. The severity, resistance to standard treatments, and high mortality rate of AIC necessitate better understanding of this state and implementation of universal treatment concept.

Disclosure: Nothing to declare.

18: Paediatric Issues

P644 PLASMA REG3Α AND CITRULLINE LEVELS PREDICT GASTROINTESTINAL AGVHD BEFORE CLINICAL ONSET IN PEDIATRIC HSCT

Nakisa Kamari-Kany1, Sarah Weischendorff 1,2, Christian Enevold1, Marianne Ifversen1, Katrine Kielsen1, Klaus Müller1,2

1Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark, 2University of Copenhagen, Copenhagen, Denmark

Background: Allogeneic HSCT remains challenged by the occurrence of aGvHD with gastrointestinal (GI) involvement standing out as a major contributor to transplant-related morbidity and mortality. The antimicrobial peptide Regenerating islet-derived 3α (Reg3α), primarily produced by Paneth cells, protects against invasive pathogens. Elevated circulating Reg3α levels have been demonstrated in patients with GI aGvHD, yet detailed insights into Reg3α kinetics and its associations with intestinal barrier integrity are needed.

Accordingly, we conducted a comprehensive study of Reg3α kinetics during pediatric HSCT, alongside measuring plasma citrulline, reflecting intestinal barrier integrity.

Methods: We included 117 children (1-18 years) undergoing myeloablative, matched-donor HSCT between 2010-2020 in Denmark. Patient and transplant characteristics are specified in Table 1.

Plasma levels of Reg3α were measured by ELISA before conditioning and at day 0, +7, +14, +21 and +30 post-transplant. Samples from 17 healthy young adults were included for comparison. Citrulline levels were measured in 75 of the patients at corresponding time points using a Waters Acquity™ Ultra-Performance Liquid Chromatography system. Reg3α and citrulline levels are reported as median (IQR).

Table 1: Patient and transplant characteristics

Sex, no. (%)

Male

68 (58.1)

Age at transplantation (years), median (range)

Recipients

8.9 (1.1-17.9)

Donors

24.1 (1.5-56.0)

Disease at transplantation, no. (%)

Malignant

72 (61.5)

Benign

45 (38.5)

Donor type, no. (%)

Matched sibling donor

33 (28.2)

Matched unrelated donor

84 (71.8)

Stem cell source, no. (%)

Bone marrow

112 (95.7)

Peripheral blood

5 (4.3)

Conditioning regimen, no. (%)

TBI (12 GY) + CY/VP16

23 (19.7)

BU + CY ± MEL

32 (27.4)

Other BU-based regimens

19 (16.2)

Other conditioning regimens

43 (36.8)

Results: Pre-conditioning Reg3α levels were comparable to healthy controls (19.8 (14.0-32.5) ng/mL vs. 17.5 (11.7-21.8) ng/mL). Reg3α increased after conditioning to a maximum of 92.8 (53-176.1) ng/mL at day +0 (P<0.001), followed by a subsequent decline, however remaining consistently higher than pre-conditioning levels (all P<0.05).

Thirty-eight patients (32.5%) developed aGvHD grade II-IV (MAGIC criteria) and showed elevated levels of Reg3α at day +14 and +21 (P<0.01), an observation that remained significant after adjusting for diagnosis, donor type, TBI and busulfan-based conditioning.

Patients developing GI aGvHD (n=16, 13.7%) had significantly increased Reg3α levels as early as day +7 and continuing through all subsequent time points, compared to patients with solely extraintestinal aGvHD manifestations and those without aGvHD, persisting in multivariable analysis. Patients experiencing lower GI aGvHD involvement (n=7) exhibited higher Reg3α levels than patients with isolated upper GI aGvHD (n=9) during days 0-14 (at day 0: 209.6 (180.2-287.2) ng/mL vs. 77.8 (52.4-116.3) ng/mL, P=0.03).

Citrulline levels decreased from a pre-conditioning level of 21.2 µmol/mL, reaching nadir of 7.5 µmol/mL at day +7 (P<0.001). Citrulline and Reg3α levels were consistently, negatively correlated at all time points, strongest at day +14 (rs=-0.5, P<0.001). Moreover, citrulline levels were reduced even prior to conditioning in patients who developed GI GvHD compared to those without aGvHD and those with solely extraintestinal involvement. This decrease persisted throughout the first two weeks post-HSCT (all P<0.05).

Conclusions: Our study reveals a significant early increase in Reg3α levels following HSCT in pediatric patients developing GI aGvHD already prior to clinical manifestations. Importantly, we extend previous observations by establishing a direct link between elevated Reg3α levels and a compromised intestinal barrier integrity. Kinetics of citrulline levels indicate that patients developing GI GvHD exhibit significantly reduced intestinal barrier integrity even before the initiation of conditioning therapy. These findings strongly support a combined use of Reg3α and citrulline as prognostic biomarkers for GI aGvHD, potentially informing pre-emptive intensification of immunosuppression in high-risk patients.

Disclosure: Nothing to declare.

18: Paediatric Issues

P645 THE OUTCOME OF HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS WITH CHEMOREFRACTORY ACUTE MYELOID LEUKEMIA

Maria Ilushina1, Larisa Shelikhova1, Daria Shasheleva1, Yuliya Skvortsova1, Sergey Blagov1, Dmitriy Balashov1, Galina Novichkova1, Alexei Maschan 1, Michael Maschan1

1Dmitry Rogachev Federal Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation

Background: Children with AML who fail to achieve remission on high-dose chemotherapy have no chance of survival without allo-HSCT. However, even with the most intensive preparative regimens, including total body irradiation, the reported survival rate in this cohort varies from 9% to 44%. We report here the results of a retrospective outcome research in a cohort of children with chemorefractory AML, who received allogenic HSCT.

Methods: Sixty-nine consecutive pediatric patients with AML in active disease ((induction failure, n=32, refractory relapse, n=37, 30 female/39 male, median age 9,4 years) underwent allo-HSCT between Feb 2012 and Jan 2020, the median follow-up time for survivors was 5,47 (2-9) years. Forty-four pts received haploidentical graft, 12 graft from MRD and 8 graft from matched unrelated donor (MUD).

Chemorefractory disease was defined as persistence of morphologic leukemia (blast count >5%) after at least two courses of intensive chemotherapy, bone marrow burden before cytoreduction were >15%(n=42), <15% (n=27). 33 patients received a cytoreductive chemotherapy, the conditioning regimen was initiated four days upon completion of the cytoreduction.

Sixty-five pts received treosulfan-based preparative regimen, while TBI-based regimen was used in 4 pts., either melphalan (n=39), thiotepa (n=29) or vepesid (n=11) were added as a second agent.

Four regimens of GvHD prophylaxis were used: regimen 1 (n=19): horse anti-thymocyte globulin 50 mg/kg + Tacro/MTX; regimen 2 (n=11): thymoglobulin 5mg/kg, rituximab 200 mg/m2 and bortezomib on day +2, + 5 (n=9); regimen 3 (n=36): tocilizumab 8 mg/kg and post-transplant bortezomib (n=33), additionally 24 pts received abatacept at 10 mg/kg on day+2, + 7, + 14, + 28; regimen 4: post-transplantation cyclophosphamide(n=3).

TCRαβ + /CD19 + -depletion of SCT with CliniMACS technology was implemented in 55 cases. Twenty-one patients received post-transplant therapy by courses of hypomethylating agents; 48 received modified DLI.

Results: 47 (83%) patients engrafted and achieved complete remission by day +30. Three pts died before engraftment due to infection, in 9 cases leukemia persisted, all of them died due to refractory AML. The median time to ANC and platelets recovery was 14 days (10-49).

The CI of aGvHD grade 2-4 in TCR ab depletion group was 36% (95% 26-51%) compare in unmanipulated graft 64% (95% 43-94%)(p=0,019), chGVHD 17% in whole group. Among all patients, TRM was CI 8% (95% 3,3-18%), CI of relapse was 54% (95%=43-67%). The cumulative risk of relapse after HSCT in the high (more than 15%) and low (less than 15%) BM burden groups was 69 (95%=56-84%) and 31% (95%17-55%), respectively with p=0.002.

NK-cell recovery at day +30 was significantly associated with decreased incidence of relapse, CI of relapse was 77% (95%=61-96%). In patients with peripheral blood NK recovery < median, and 44% (95% 29-67%) in those with NK-cell number >median (p=0,013). At a median follow-up of 5,5 years, event-free survival was 37% and overall survival was 42%.

Conclusions: Our data suggest that allogenic HSCT provides a reasonable chance of cure in a cohort of children with chemorefractory AML and creates a solid basis for further improvement. Good early recovery of NK cells and lower BM burden at the moment of HSCT are associated with significantly improved survival and decreased relapse incidence.

Disclosure: M. Maschan - Miltenyi Biotec - Honoraria.

D. Balashov - Octafarm Lecturers fee.

18: Paediatric Issues

P646 OUTCOMES AFTER HEMATOPOIETIC CELL TRANSPLANTATION FOR HURLER SYNDROME AFTER IMPLEMENTATION NEWBORN SCREENING IN US AND EUROPE

Jaap Jan Boelens 1, Caroline Lindemans2, Peter van Hasselt3, Klaas Koop3, Maria I. Cancio1, Paul J. Orchard4, Troy C. Lund4

1Memorial Sloan Kettering Cancer Center, New York, United States, 2Prinses Maxima Centrum, Utrecht, Netherlands, 3UMC Utrecht, Utrecht, Netherlands, 4University of Minnesota, Minneapolis, United States

Background: Hurler syndrome (HS), the most severe phenotype in the spectrum of Mucopolysaccharidosis type I, is caused by a severe deficiency of the lysosomal enzyme alpha-L-iduronidase (IDUA). HS is clinically characterized by a progressive and ultimately fatal multi-system deterioration with involvement of the central nervous system. At present, hematopoietic cell transplantation (HCT) is the only treatment able to prevent central nervous system deterioration and therefore considered the treatment of choice in HS. In large intercontinental cohorts, predictors for outcomes (short and long-term) were found to be age at HCT and enzyme levels after HCT. Newborn screening (NBS) and early HCT could therefore potentially impact outcomes. In several States in the US and Europe NBS has been implemented over the last couple years. We were interested in the age at HCT and outcome of HS patients diagnosed by NBS.

Methods: All HS patients transplanted in 3 centers in states with NBS were included; Minneapolis, New York (United States) and Utrecht (The Netherlands). The main outcomes of interest were (engrafted)-survival, Graft-versus-Host Disease, auto-immune cytopenia (AIC), sinusoidal obstructive syndrome (SOS) and donor chimerism. Because of the recent implementation of NBS for HS, long-term outcomes are unevaluable.

Results: 14 patients with concordant HS genotype and phenotype were included; all were identified by genotype matching the HS clinical phenotype. One patient received a non-carrier matched sibling donor and 13 an unrelated cord blood donor (five 8/8, five 7/8 and three 6/8 match). All patients received a busulfan-based myeloablative conditioning (cumulative Bu exposure of 90mg*h/L) combined with either Fludarabine 160mg/m2 (n=13), or Clofarabine (120mg/m2)/Fludarabine (40mg/m2) (n=1). In 7 patients rituximab (day -10, 28) and in 2 cyclophosphamide (50mg/kg) was added as AIC prophylaxis. Patients had a median age of 3.5 (2.4-6) months at HCT. All patients (100%) are engrafted and alive at a median follow-up of 2.9 (0.3-4.8) years. One patient (7%), who was enrolled on an expanded CB trial, experienced a late graft failure (at 1 year), but was successfully retransplanted (and now 3 years after 2nd HCT). Two patients (7%) experienced moderate/severe acute GvHD (1 grade 1 and 1 grade 3) and one patient experienced SOS (7%). No chronic GvHD, no auto-immune cytopenia events or CNS deterioration were noted. At the latest follow-up timepoint, all patients are full donor chimeric (>95%) and have IDUA levels in the normal range.

Conclusions: NBS facilitates curative HCT for HS patients very early in life. HCT for HS in the first months of life is safe and effective. Longer-term follow-up is needed to study the impact on residual disease burden.

Disclosure: JJB has honoraria, consulting fees (including DMC work): Bluerock, Sanofi, Sobi, Omeros, SmartImmune, Advanced Clinical and Immusoft (not related to the topic). TCL has honoraria, consulting fees and/or research support from Denali, Orchard Therapeutics, Sanofi Genzyme, PJO has received honoraria, consulting fees, and/or research support from Denali, JCR, Orchard Therapeutics, Regenxbio, Sanofi Genzyme.

18: Paediatric Issues

P647 ADDITION OF DINUTUXIMAB-BETA ON DAY -1 TO IMPROVE GRAFT-VERSUS-NEUROBLASTOMA EFFECTS OF HAPLOIDENTICAL STEM CELL TRANSPLANTATION IN RELAPSED HIGH-RISK NEUROBLASTOMA

Christina Lämmle 1, Michaela Döring1, Johannes H. Schulte1, Tim Flaadt1, Florian Heubach1, Peter Lang1,2,3, Christian M. Seitz1,2,3

1University Children´s Hospital Tuebingen, Tübingen, Germany, 2Excellence cluster iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, Tuebingen, Germany, 3German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Partner site Tuebingen, Tuebingen, Germany

Background: Despite intensive treatment strategies the outcome of patients with relapsed high-risk neuroblastoma remains poor. We have recently published improved survival rates after haploidentical stem cell transplantation (haploSCT) followed by six cycles of dinutuximab-beta after immune reconstitution. We hypothesized that early administration of dinutuximab-beta, as part of the conditioning regimen, might further improve graft-versus-neuroblastoma effects by activating NK- and gamma/delta-T-cells infused with the TCR-alpha/beta depleted graft, particularly for patients with macroscopic residual disease prior to haploSCT.

Methods: Between 2019 and 2023, 16 patients with relapsed high-risk neuroblastoma (rHR-NB) underwent haploSCT utilizing TCRab/CD19 depleted graft. Patients were randomized for day -1 administration of a single dose of dinutuximab-beta (10 mg/m2) vs. standard protocol according to our registry. Remission status before haploSCT in the dinutuximab-beta were CR in 4/16 and PR in 12/16 patients, equally distributed in both groups. Primary aims of the pilot study were to demonstrate feasibility of day -1 dinutuximab-beta administration and to analyze the effects on acute toxicities, engraftment, and immune reconstitution as well as inflammation associated complications, particularly the induction of GvHD.

Results: Overall dinutuximab-beta administration on day -1 was well tolerated. No severe adverse events occurred under treatment. Moderate and self-limiting monitored side effects were tachycardia in all and fever in 6/8 patients. Dinutuximab-beta associated pain was well tolerable under supportive analgetic medication. At day 1 we found elevated CRP-levels (3.19 [0.6-11.6] vs. 0.82 [0.2-3.5]) in the dinutuximab beta group. Standardized functional ex vivo testing on day 0 revealed sufficient dinutuximab-beta serum-levels as measured by CDC and ADCC-induction against target cell lines. All patients engrafted with a slightly delayed neutrophile take (11.5 days [10-14] vs. 10.5 days [4-13]) and platelet recovery (13.5 days [7-31] vs. 8.5 days [6-14]) in dinutuximab group.

No significant differences in early immune reconstitution, as measured for CD3/TcRgd, CD3/TcRab, CD3/CD4, CD3/CD8 positive T cells, CD19 positive B cells and CD16/CD56 positive NK cells, were found. No patients had severe viral or fungal infections. One patient in the dinutuximab-beta group showed CMV reactivation. In the dinutuximab-beta group five patients (62,5%) experienced acute skin GvHD (stadium I-II /overall grade 1), rapidly resolved under topic therapy. One patient developed gut GvHD (stadium III, overall grade 3) responsive to systemic steroid plus ruxolitinib and tacrolimus. In contrast no GvHD was monitored in control group. No transplant related mortality was observed. Of note, 6 patients of the dinutuximab-beta group with PR prior to transplantation achieved CR after haploSCT. The pilot study was not powered to measure general effects of the intervention on survival parameters.

Conclusions: Administration of dinutuximab-beta on day -1 is feasible. Acute side effects were mild. We found no impairment of engraftment and immune reconstitution. Our data suggests the induction of systemic immune activation by dinutuximab-beta, resulting in an increased risk for mild to moderate aGvHD, this requires further investigation. This pilot study might serve as a prerequisite to further evaluate optimized dosing schedules of dinutuximab-beta in the context of haploSCT, to provide treatment options to rHR-NB patients with insufficient remission status.

Disclosure: Nothing to declare.

18: Paediatric Issues

P648 HHV-6 INFECTION MAY IMPAIR IMMUNE RECONSTITUTION FOLLOWING PEDIATRIC HAPLO-HSCT

Katrine Kielsen 1, Eva Kannik Haastrup1, Lisbeth Pernille Andersen1, Tania Masmas1, Marianne Ifversen1

1Rigshospitalet, Copenhagen University Hospital, Copenhagen, Denmark

Background: Haplo-identical HSCT is challenged by a delayed T and B-cell recovery hampering the patients’ immunity towards infections and malignant clones. Especially viral infections constitute a major concern and are thought to further impair the immune reconstitution (IR).

In this study, we investigated the impact of early viral infections (CMV, EBV, adenovirus and HHV-6) and IR on clinical outcomes after pediatric haplo-HSCT.

Methods: We prospectively included 44 consecutive children undergoing allogeneic HSCT with a haplo-identical donor from 2012-2023 in a single center in Denmark (Table). Haplo-HSCT was used as first (n=29) or second HSCT (n=15). Ex-vivo depletion was performed on PB grafts from parental donors by TCRαβ-cell depletion with the CliniMACS Plus (n=16, 2012-2015) or TCRαβ/CD19-cell depletion with the CliniMACS Prodigy (n=32, 2016-2023).

T- and B-cell counts and viral reactivation were measured regularly post-HSCT.

Transplant characteristics

Values (n=44)

HSCT no., n. (%)

1. HSCT

29 (66%)

2. HSCT

15 (34%)

Recipient age, median (range)

8.1 years (0.3-17.8)

Diagnosis, n. (%)

ALL

12 (27%)

AML

7 (16%)

Other malignancies

8 (18%)

PID

12 (27%)

Other benign disorders

4 (9%)

Conditioning regimens, n. (%)

TBI + VP16

3 (7%)

Clo-Mel-TT

14 (32%)

Flu-Treo-TT

11 (25%)

Flu-Mel-TT

8 (18%)

Other chemotherapy-based regimens

8 (18%)

In vitro T-cell depletion, n. (%)

ATG

42 (95%)

Campath

2 (5%)

In vitro B-cell depletion, n. (%)

Rituximab

44 (100%)

Cellular composition of grafts, median (range)

CD34 count

12.7 (7.3-24.0)

CD3 count

22.5 (9.1-125.0)

Prophylactic immune suppression with MMF, n. (%)

16 (36%)

Results: Median follow-up time was 6.7 years (0.5-11.5) with a 5-year cumulative incidence of non-relapse mortality of 16.0% (95%CI: 4.8-33.0) and a cumulative incidence of relapse of 24.5% (95%CI: 9.7-43.0), projected into a 5-year overall and disease-free survival of 69.0% (95%CI: 56.3-84.7) and 59.5% (95%CI: 42.9-82.5). Ten patients (23%) received DLI post-transplant.

All patients but 4 (9%) engrafted with a median time to neutrophil and platelet recovery of 11 (range 9-30) and 12 (range 10-99) days, respectively. Three of the four patients experiencing graft failure were rescued with a second haplo-HSCT using the same donor or the other parent. CD4 and CD8 T cells counts above 200/µL were reached after one year for 86.9% and 76.9% of patients, respectively, and B cell recovery of 100/µL were reached for 87.6%. Cumulative incidence of grade II-IV acute GvHD and chronic GvHD were 42.1% (95%CI: 23.5-56.2) and 8.0% (95%CI: 0-18.3), respectively. The severe GvHD-free, disease-free survival was 48.3% (95%CI: 29.3-62.1).

Overall, 25 patients developed a viral reactivation (56.8%, 5 had ≥2 infections) at median 27 days post-HSCT (range: 0-121), including CMV (n=9), ADV (n=12) and HHV6 (n=10). Viral reactivation post-HSCT tended to be associated with occurrence of aGvHD grade II-IV (OR=3.5, P=0.07). Furthermore, patients with HHV-6 infection tended to have a slower recovery of CD4 + T cells and B cells (Figure), although not reaching statistical significance. Viral infection was not associated with survival or relapse.

A rapid IR (CD4 cells >200/µL or B cells >100/µL within 180 days in patients without relapse, death or HSCT at this time point) tended to be associated with less HHV-6 infection (OR=0.17 for HHV-6 if B cells >100/µL within 180 days, P=0.058), but was not significantly associated with development of aGvHD or survival.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (84)

Conclusions: Overall, clinical outcomes were acceptable in our cohort consisting of many patients with high-risk disease. Recovery of T and B cells were reached within one year for most of the patients. None of our patients experienced EBV reactivation, and reactivation of CMV and ADV represented a minor challenge. However, viral reactivation with HHV-6 remained a concern post-transplant and may be associated with a higher occurrence of GvHD and delayed IR.

Clinical Trial Registry: NA.

Disclosure: Nothing to declare.

18: Paediatric Issues

P649 FINAL HEIGHT AND IMPACT OF GROWTH HORMONE TREATMENT IN CHILDREN RECEIVING TBI-BASED CONDITIONING REGIMEN BEFORE HSCT FOR ACUTE LYMPHOBLASTIC LEUKEMIA

Charlotte Calvo1,2, Coline Mornet3, Caroline Storey3, Carine Halfon-Domenech4, Cécile Pochon5, Marie-Dominique Tabone6, Guy Leverger6, André Baruchel1, Catherine Paillard7, Charlotte Jubert8, Marilyne Poirée9, Dominique Plantaz10, Justyna Kanold11, Virginie Gandemer12, Anne Sirvent13, Sandrine Thouvenin14, Marlène Pasquet15, Julie Berbis16, Pascal Auquier17, Jean-Hugues Dalle 1,2, Gérard Michel18, Paul Saultier19

1Robert Debré Hospital, GHU APHP Nord Université Paris Cité, Paris, France, 2INSERM UMR-976, Institut de Recherche Saint-Louis (IRSL), Paris, France, 3Robert Debré Hospital, Paris Cité University, AP-HP, Paris, France, 4Institut d’Hémato-Oncologie Pédiatrique (IHOP), Lyon, France, 5Hôpital d’Enfants de Brabois, Vandoeuvre Les Nancy, France, 6Armand Trousseau Hospital, Sorbonne University, AP-HP, Paris, France, 7University Hospital of Strasbourg, Strasbourg, France, 8University Hospital of Bordeaux, Bordeaux, France, 9University Hospital L’Archet, Nice, France, 10University Hospital of Grenoble, Grenoble, France, 11University Hospital of Clermont-Ferrand, Clermont-Ferrand, France, 12University Hospital of Rennes, Rennes, France, 13University Hospital of Montpellier, Montpellier, France, 14University Hospital St Etienne, St Etienne, France, 15Toulouse University Hospital, Toulouse, France, 16Aix Marseille University, UR3279 CERESS, APHM Marseille, Marseille, France, 17Aix Marseille University, UR3279 CERESS, APHM, Marseille, France, 18Aix Marseille Univ, APHM, CERESS, La Timone Children’s Hospital, Marseille, France, 19Aix Marseille Univ, APHM, INSERM, INRAe, C2VN, La Timone Children’s Hospital, Marseille, France

Background: Growth impairment is a well-known long-term sequela for acute lymphoblastic leukemia (ALL) children patients undergoing hematopoietic stem cell transplantation (HSCT) after total body irradiation (TBI)-based conditioning regimen. However, studies reporting final height (FH) and effect of growth hormone treatment (GHTx) are limited by small size cohorts, heterogeneous diseases or conditioning regimens, or too short follow-up. Here, we describe FH, impact of GHTx and risk factors for growth impairment among childhood ALL survivors treated with TBI and HSCT from the LEA (Leucémie de l’Enfant et de l’Adolescent) French cohort.

Methods: French patients participating in the LEA program who were treated for an ALL with HSCT after TBI-based conditioning before puberty (i.e., <9 and <11 years old for girls and boys, respectively) were included if they had reached adulthood. Patients who had received additional irradiation on the central nervous system or were carriers of a genetic disease were excluded. Height was expressed as standard deviation (SD) scores according to French pediatric references for sex and age. Growth was evaluated at different time points: leukemia diagnosis, HSCT, beginning of puberty and at adulthood. Growth Hormone (GH) deficiency and GHTx were reported. Occurrence of metabolic syndrome and secondary malignancies were reported as well as risk factors that could influence growth impairment.

Results: One hundred and eighty-five patients have been included in the study, 63% were men. Mean age (±SEM) at first HSCT and at last follow-up visit were 6.6±0.2 and 24.6±5.7 years, respectively. All patient received TBI-based conditioning regimen before their first HSCT; 80% received allogenic HSCT versus 20% autologous, and nine patients (5%) received ≥2 HSCT.

Final mean height was 153±1cm for women and 163±1cm for men. Most of the height growth defect occurred during puberty, irrespective of the age at transplant: mean FH was -1.9 SD for both male and female patients. GH deficiency was suspected in 72 patients based on clinical criteria and biologically confirmed in 43 of them (23%). Thirty-eight patients received GHTx. In these patients, FH was -2.2SD (153±2cm and 160±2cm for female and male patients, respectively) and height growth SD score decline from transplant to adulthood was -1.9 SD for girls and -2.4 SD for boys, suggesting a better efficacy of GHTx in girls. There was no increase in secondary malignancies or meningioma or metabolic syndrome in patients treated with GHTx. Multivariate analysis (Table) identified four significant determinants of HSCT-related growth alteration: male sex (linear regression coefficients ß=-0.56; p=0.001), younger age at HSCT (ß=0.61 for patients >6.6 years old at HSCT, p=1.10-6), ≥2 HSCT (ß=-0.93; p=0.003) and patients taller at HSCT (ß=-0.24 per each additional SD unit; p=0.0003).

Risk factors influencing growth impairment

Univariate analysis

Multivariate analysis

n

Mean ± SEM or correlation coefficient

ß

p-value

ß

p-value

Gender

Female

69

-1.96 ±0.12

1

0.007

1

Male

116

-2.37 ±0,09

-0.41

-0.49

0.001

Age at first transplant

≤6,63

93

-2.46 ±0.11

1

0.001

1

6,64+

92

-1.98 ±0.09

0.48

0.7

1.12E-06

Number of HSCT

1

176

-2.17 ±0.07

0.007

1

≥2

9

-3.1 ±0.39

-0.93

-0.93

0.003

Relapse

No

76

-2.25 ±0.11

1

0.737

Yes

109

-2.2 ±0.1

0.05

Time of HSCT

<Jun-1993

47

-2.18 ±0.15

1

0.99

Jun-1993 - Jan-2000

46

-2.24 ±0.18

-0.06

Jan-2000 - Dec-2005

46

-2.23 ±0.13

-0.06

>Dec-2005

46

-2.23 ±0.12

-0.05

Height at first HSCT (SD)

185

-0.232

-0.2

0.001

-0.24

0.0003

Conclusions: These results confirmed that TBI and HSCT induce a severe growth impairment in pre-pubertal patients treated for ALL. Our data show that boys are more affected than girls. GHTx was not associated with an increased risk of secondary malignancies or metabolic syndrome and appears to be effective in limiting the final growth defect.

Clinical Trial Registry: clinicaltrials.gov identifier: NCT 01756599.

Disclosure: Nothing to declare.

18: Paediatric Issues

P650 ENGRAFTMENT SYNDROME AND ACUTE GVHD AFTER HAPLOIDENTICAL POST-TRANSPLANT CYCLOPHOSPHAMIDE: A PAEDIATRIC MULTICENTRIC EXPERIENCE

Maura Faraci 1, Francesco Saglio2, Francesca Baudi1, Filomena Pierri1, Francesca Bagnasco1, Stefano Giardino1, Francesca Gottardi3, Davide Leardini3, Franca Fagioli2,4, Riccardo Masetti3

1IRCCS Istituto Giannina Gaslini, Genova, Italy, 2Pediatric Oncohematology Regina Margherita Children Hospital AOU Città della Salute e della Scienza di Torino, Turin, Italy, 3Pediatric Hematology Oncology IRCCS Azienda Ospedaliero Universitaria di Bologna, Bologna, Italy, 4University of Turin, Turin, Italy

Background: Post-transplant cyclophosphamide (PT-CY) is an effective Graft-versus-Host Disease (GvHD) prophylaxis in patients undergoing haploidentical hemopoietic stem cells transplantation (HSCT), nevertheless information about the incidence of engraftment syndrome (ES) occurred with this platform is lacking.

Methods: During the period from 2012 to 2023, 85 paediatric patients with acute lymphoblastic leukaemia (AL) received 101 haploidentical HSCTs with PT-CY in 3 Italian paediatric transplant units.

We analysed the incidence of ES and acute GvHD in relation to the day of start of calcineurin inhibitor (CNI) therapy: ≤ or > day 0.

Results: 39 patients were affected by B-cell ALL (B-ALL), 40 by acute myeloid leukemia (AML), 4 by T-cell acute lymphoblastic leukemia (T-ALL) and 2 by early T-cell precursor (ETP) acute lymphoblastic leukemia/lymphoma. The conditioning regimen was busulfan-based in 39 HSCTs, treosulfan-based in 6, thiotepa with fludarabine in 1, TBI based in 55 transplants. The source of HSCT was bone marrow (BM) in 85 and peripheral blood stem cell (PBSC) in 16. The median count of mononuclear cells engrafted was 6.32x108/kg and that CD34+ was 7x106/kg. In addition to PY-CY, all patients received as GvHD prophylaxis a CNI drug plus mycophenolate mofetil (MMF) and granulocyte-colony stimulating factor (G-CSF). In 50 HSCTs the CNI was cyclosporin (CsA), it was started ≤ day 0 (0; -1) in 20 patients, and it was started > day 0 in 79 patients ( + 5 in 77; +6 in 1; and +1 in 1). In 49 HSCTs the CNI was tacrolimus (Tac), it was started in all cases at day +5. In 2 transplants, GvHD prophylaxis and G-CSF were not administered since early development of multi-organ failure and subsequent death.

ES developed after 21 (20.7%) out of 101 transplants and it was not evaluable in 3 patients due to rejection and early onset of complications. The incidence of ES was significantly associated with the infusion of PBSC rather than BM cells (p = 0.005) and with the count of mononuclear cells engrafted (p = 0.002). The timing of start of CNI (≤ or > day 0) did not correlate with the incidence of ES.

The earlier start of CNI (≤ day 0) was statistically associated with higher incidence of a-GvHD (p < 0,001), that occurred in 38 HSCTs.

The overall survival (OS) in our cohort was 74.1%. Seven patients (31.8%) died for transplant-related complications and 15 (68.1%) for relapse or progression of the AL. Among the 21 patients who developed ES, 4 (14.2%) died for TR complications.

Conclusions: ES is a severe complication of HSCT, its incidence appears to be associated with use of PBSC and with hight count of mononuclear cells engrafted, but the timing of initiation of CNI does not seem to affect its incidence, while an earlier initiation of CNI appears to be associated with development of a-GVHD.

Disclosure: None conflict of interest.

18: Paediatric Issues

P651 PULMONARY COMPLICATIONS IN CHILDREN FOLLOWING HAEMATOPOIETIC STEM CELL TRANSPLANT

Hannah Walker 1, Diane Hanna1, Theresa Cole1, Gabrielle Haeusler1, Shivanthan Shanthikumar1, Melanie Neeland2

1The Royal Children’s Hospital, Melbourne, Australia, 2Murdoch Children’s Research Institute, Melbourne, Australia

Background: As outcomes have improved following hematopoietic stem cell transplant (HSCT) a greater focus has been placed on survivorship, and in particular the detection and management of HSCT related complications. Pulmonary complications represent a common and serious group of post-HSCT complications, yet Australian data regarding pulmonary complications in children post-HSCT is lacking.

Methods: Retrospective single-centre analysis of pulmonary complications in children who received an allogeneic HSCT at The Royal Children’s Hospital (Melbourne) between 2016 - 2022. The primary objectives of this study were to describe the incidence and outcomes of children who develop pulmonary complications post-HSCT.

Results: 222 episodes of allogeneic HSCT were identified, these occurred in 203 individual patients. The median age at the time of HSCT was 6 years, median follow-up 3 years, and 59% (132/222) of children had a malignant indication for HSCT. The most common malignant indications for transplant were B-ALL (41/132) and AML (43/132). The most common non-malignant indications for transplant were immunodeficiency (44/90) and Aplastic anaemia (11/90). Pulmonary complications occurred in 55% (112/203) of patients post HSCT, with the majority occurring within the first 100 days post HSCT (63%). The most common presenting symptoms of a child’s pulmonary complication were fever (58.8%), cough (43%) and hypoxia (37%). The most frequent investigations performed at presentation of symptoms were chest x-ray (71%) and Computed topography scan (52%). In terms of more invasive strategies; bronchoalveolar lavage was performed in 47.4% of children and lung biopsy in 7% of episodes of pulmonary complication. Children were diagnosed with infectious (56 %,63/112), non-infectious (28 %, 31/112) or both (16%,18/112) pulmonary complications. Frequently (20%, 22/112) children experienced more than one pulmonary complication post HCT. Non-infectious complications included bronchiolitis obliterans syndrome 13% (15/112) and idiopathic pneumonia syndrome 4% (5/112). Within this total cohort 30% (62/203) of children required paediatric intensive care unit (PICU) admission. Of this group, 48% (30/62) were admitted to PICU due to a pulmonary complication and of the two children who required ECMO, both received this due to a pulmonary complication. The overall mortality was 18% (38/203), 29% of these deaths were in patients post a second HSCT (11/38). Pulmonary complications directly contributed to mortality in 18 children but pulmonary complications occurred in 31 of the 38 children who died post HSCT. This is reflected in Figure 1; showing a Kaplan Meier curve illustrating the difference in survival between children who developed a pulmonary complication post HCT compared to those who did not. This survival curve showed a hazard ratio of 4.68 (95% CI 2.4-8.9) p <0.01.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (85)

Conclusions: This study shows pulmonary complications occur very commonly in children post HSCT and are frequently severe in nature, requiring high level support in an intensive care unit. This study also showed a significant increased risk of death post allo-HSCT in the children who developed a pulmonary complication, highlighted in Figure 1. This highlights the urgent need for improved strategies for the investigation, diagnosis and management of these pulmonary complications.

Clinical Trial Registry: N/A.

Disclosure: Nothing to declare.

18: Paediatric Issues

P652 EXCELLENT SURVIVAL USING T-REPLETE UMBILICAL CORD BLOOD TRANSPLANT IN HIGH RISK PAEDIATRIC MYELOID LEUKAEMIA, INCLUDING IN REFRACTORY DISEASE

Kate Davies 1, Rob Wynn1

1Royal Manchester Children’s Hospital, Manchester University NHS Foundation Trust, Manchester, United Kingdom

Background: Haematopoietic stem cell transplant offers a potential cure for paediatric patients with high risk myeloid malignancies. Recent multi-centre studies have shown improved outcomes using T-replete cord blood (TRCB) when compared to other stem cell sources, particularly in patients with detectable MRD pre-transplant. We report on nearly 10 years of survival outcomes from a large paediatric transplant centre.

Methods: Hospital records were reviewed for paediatric patients undergoing TRCB transplant between January 2014 and September 2023 at Royal Manchester Children’s Hospital. Data was collected on patient demographics, previous lines of treatment, transplant conditioning, survival, GVHD and transplant-related mortality.

Results: 54 patients underwent TRCB transplant during the period studied. Of these, 67% were flow MRD positive at the start of conditioning. 35% of patients had previously undergone HSCT.

80% of patients received an HLA-mismatched transplant. 44% of patients received myeloablative conditioning and 56% reduced intensity conditioning.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (86)

Overall survival was 59% across all patients, 44% in the MRD positive group and 89% in the MRD negative group. MRD positivity is associated with a statistically significant decrease in overall survival. There was no significant difference in survival between patients receiving HLA-matched and mismatched transplants. 36% of deaths were due to transplant-related mortality and 64% due to relapsed or refractory disease.

65% of patients developed acute GVHD of any grade, though only 43% of patients required treatment with systemic immunosuppression. Only 3 patients in the study (5%) developed chronic GVHD.

Conclusions: We report on a large, homogeneous cohort of patients with very high-risk myeloid disease, as evidenced by the significant proportion of patients who had undergone previous HSCT and the high rates of MRD positivity pre-transplant. Historical outcomes for such patients have been poor. The use of mismatched TRCB as a donor source allows us to maximise the graft-versus-leukaemia effect in this difficult to treat population, contributing to improvements in overall and event-free survival.

Though a significant proportion of patients developed acute GVHD, the majority did not require systemic immunosuppression, and only a small number developed chronic GVHD.

None of the patients that relapsed were salvageable with further lines of therapy. This suggests that, at present, TRCB transplant is the optimal treatment strategy for high-risk myeloid leukaemia.

This patient cohort includes patients treated on the GRANS trial which utilises granulocyte infusions alongside mismatched TRCB. Detailed results from this study will be reported separately.

Disclosure: Nothing to declare.

18: Paediatric Issues

P653 TCRΑΒ + /CD19 + -DEPLETION IN HEMATOPOIETIC STEM CELLS TRANSPLANTATION FROM MATCHED UNRELATED AND HAPLOIDENTICAL DONORS IN CHILDREN WITH HIGH-RISK ACUTE MYELOBLASTIC LEUKEMIA

Larisa Shelikhova 1, Olga Molostova1, Maria Ilyushina1, Yuliya Skvortsova1, Irina Shipitsina1, Rimma Khismatullina1, Sergey Blagov1, Svetlana Kozlovskaya1, Irina Kalinina1, Dina Baidildina1, Elena Gytovskaya1, Dmitriy Balashov1, Alexey Kazachenok1, Alexander Popov1, Ekaterina Mikhailova1, Julia Olshanskaya1, Dmitriy Litvinov1, Galina Novichkova1, Alexey Maschan1, Michael Maschan1

1Dmitry Rogachev Federal Research Centre of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation

Background: Among children with AML with high-risk or poor response to therapy alloHSCT provides best chance of LFS. Choice of donor and overall strategy of preparative regimen and GvHD prevention remain an area of active research. Depletion of ab T cells was developed to improve the outcomes of HSCT by decreasing the incidence of GVHD while maintaining the anti-leukemia effects and infection control. We report here the results of a retrospective outcome research in a cohort of children with high-risk AML, who received ab T cell-depleted HSCT.

Methods: A total of 216 pediatric patients with AML underwent first allogeneic HSCT between May 2012 and February 2022. 171 pts received haploidentical graft, 45 a graft from MUD. Disease status at transplant was a CR 1 in 112 pts, >CR1 in 41 pts and AD in 63 pts. 202 pts received treosulfan-based preparative regimen, while TBI-based regimen was used in 14 pts.

Three regimens of GvHD prophylaxis were used: regimen 1 (n=30) included horse ATG at 50 mg/kg and CsA + /-Mtx, regimen 2 (n=56): thymoglobulin 5mg/kg, rituximab 200 mg/m2 on d-1 and bortezomib on day +2, +5 and regimen 3 (n=130): rituximab 200 mg/m2 and tocilizumab at 8 mg/kg on day -1, abatacept at 10 mg/kg on day +2, +7, +14, +28 and post-transplant bortezomib on d + 2, +5. TCRαβ + /CD19+ depletion of HSCT with CliniMACS technology was implemented in all cases.

Results: Three patients died before engraftment due to bacterial infection, 4 had disease progression at day +30. Primary engraftment was achieved in 190 (94%) patients with median time to engraftment of 13 days. Five patients failed to engraft and required rescue with a second HSCT from the alternative donors. The CI of TRM among all patients was 5,6%, 4% for haploidentical and 11% for MUD groups, p = 0,076. The direct causes of non-relapse death included bacterial and viral infections. The CI of aGvHD grade II-IV was 19%, grade 3-4 was 4,6% and chronic GvHD – 12%. The CI of aGvHD 2-4 was 51% with regimen 1, 11% with regimen 2 and 15,7% with regimen 3, p < 0,0001. The CI of relapse at 6 years was 35% in the entire cohort. A pre-HSCT AD status was associated with increased risk of relapse in the UA (p < 0,0001). In CR group serotherapy also associated with high relapse rate(p=0,08), compare with AD group (p=0,5) Pts with CR, EFS and OS at 6-years were 57% and 69% for the whole cohort, EFS in AD group was 38% as compared to 61% in CR1 and 73% in >CR1 groups (p=0,000). EFS with serotherapy was 65%, without 49%, p=0,04 without effect on OS. The 6-years GRFS in whole group was 57%. Median time of follow-up for survivors was 6 years.

Conclusions: We confirm that the depletion of TCR-alpha/beta and CD19 lymphocytes from the graft ensures high engraftment rate, low burden of GvHD and acceptable TRM in pediatric high-risk AML patients. AD status prior HSCT and absence of serotherapy in CR group are associated with high relapse rate and low survival.

Disclosure: M. Maschan: Miltenyi Biotec Honoraria.

D. Balashov: Ocrafarm: Other Lectures fee.

18: Paediatric Issues

P654 IMPLEMENTATION OF CYP3 A5 GENOTYPING TO INDIVIDUALISE TACROLIMUS DOSE IN PAEDIATRIC PATIENTS UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANTATION

Iván López Torija 1, Laura Gras Martín1, Pablo Escribano Sanz1, Pau Riera Armengol1, Carla Miñarro Chacón1, Sara Bernal Noguera1, Montserrat Torrent Español1, Esther Rojas Rodriguez1, Susana Boronat Guerrero1, Edurne Fernández de Gamarra Martínez1

1Hospital Santa Creu i Sant Pau, Barcelona, Spain

Background: Tacrolimus is a widely used drug for the prophylaxis of graft-versus-host disease. CYP3 A5 is the most involved enzyme in its metabolism. The Clinical Pharmacogenetics Implementation Consortium (CPIC) recommends genotyping CYP3 A5 before starting treatments and increasing the starting dose in extensive and intermediate metabolizers. The aim of this study is to describe the implementation of CYP3 A5 genotyping in paediatric patients undergoing haematopoietic stem cell transplantation (HSCT) to individualise the initial dose of tacrolimus and shorten the time to therapeutic doses in all cases.

Methods: A total of 17 patients, 14 of them diagnosed with haematological malignancies, 2 patients with non-malignant haematological pathology and one immunodeficiency, undergoing allogeneic HSCT from March 2022 to November 2023, were included.

CYP3 A5 genotyping is added to pre-transplant testing. The Genetics Service determines the CYP3 A5 loss-of-function alleles (*3,*6 and *7) in peripheral blood by real-time PCR using Taqman® probes. According to the recommendations of the CPIC and the Pharmacy Service, tacrolimus was started at 1.5 times the standard dose (0.03 mg/kg/day intravenous) in patients with extensive and intermediate metabolism. The standard dose is ideal for slow metabolizers, which is the predominant genotype in Caucasian population. In all cases, the route of administration was changed to oral when the clinical situation allowed it, calculating the equivalent dose according to bioavailability and interactions. Tacrolimus levels were determined at least twice weekly during the intravenous administration period and weekly during the oral phase for the first 3 months.

Results: Of the 17 patients genotyped, 11 of them (65%) had the *3/*3 genotype (poor metabolizers). Five patients (29%) had the *1/*3 genotype, and one patient (6%) had the *1/*7 genotype (intermediate metabolizers).

In intermediate metabolizers who started treatment at a higher dose instead of standard dose, therapeutic range concentrations were reached between day +1 and +3 from baseline, although in 4/5 patients the dose was subsequently reduced to 0.03 mg/kg/day.

In poor metabolizers (genotype *3/*3), who started with a standard regimen, concentrations in therapeutic range were reached on day +7 from the start, with the maintenance dose being 0.03 mg/kg/day in 8/11 patients and a lower dose in the rest of the patients.

Regarding oral treatment, there was great variability in the doses required to maintain concentrations in therapeutic range, due to the influence of factors such as oral absorption and interactions with azoles. We do observe a greater number of intermediate metaboliser patients requiring doses higher than standard doses.

Conclusions: CYP3 A5 genotyping enables individualizing the initial tacrolimus dose in paediatric patients undergoing haematopoietic stem cell transplantation and optimizing the time needed to obtain concentrations in the therapeutic range.

Besides that, we confirm the need to maintain pharmacokinetics as a standard care practice in this type of patients to confirm adequate exposure to drugs with a narrow therapeutic range and high inter and intra-individual variability.

The main limitation of the study was the sample size. More experience is needed to stablish solid conclusions.

Disclosure: No disclosures.

18: Paediatric Issues

P655 >EPIDEMIOLOGY OF GUT COLONIZATION AND INFECTION BY CARBAPENEM-RESISTANT GRAM-NEGATIVE BACILLI (CR-GNB) IN PEDIATRIC HEMATO-ONCOLOGICAL AND TRANSPLANTED PATIENTS

Adriana Balduzzi 1,2, Iacopo Bellani2, Bianca Monti2, Marta Adavastro2, Francesca Limido2, Sonia Bonanomi1, Arianna De Buglio2, Marianna Rossi1, Francesca Vendemini1, Sergio Foresti1, Andrea Biondi1,2, Paolo Bonfanti1,2, Marco Guglielmo Migliorino1, Sergio Malandrin1, Francesca Iannuzzi1

1Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 2Milano-Bicocca University, Milano, Italy

Background: Pediatric patients affected with hematological malignancies and undergoing HSCT are at high risk of being colonized by Carbapenem-resistant Gram-negative bacilli (CR-GNB), due to high exposure to multiple antibiotics, frequent and prolonged hospital admission, inflammation, apoptosis and discontinuity of the mucosal barrier, caused by chemotherapy and radiotherapy, potentially promoting bacterial translocation and sepsis.

Methods: The aim of this retrospective study is to assess epidemiology of gut colonization by CR-GNB, besides febrile episodes and rate of infection by CR-GNB.

All consecutive admitted patients, having rectal swab for CR-GNB, have been enrolled and clinical data on febrile episodes and blood cultures for patients with at least one positive swab have been analyzed from the first positivity up to the detection of three negative consecutive samples.

Results: 52 (3.8%) out of 1347 pediatric patients enrolled were detected positive for one or more CR-GNB at least once, 69% of whom were undergoing transplant, with 9 patients detected positive prior to HSCT. 29% of the CR-GNB positive patients had referred from East European Countries.

The most frequent CR-GNB was Escherichia coli, then Pseudomonas aeruginosa and Klebsiella pneumoniae; the most frequent mechanisms of resistance were metallo-beta-lactamases type in the 79% of the cases, with VIM detected in 59%, and KPC in the 7%.

In the 52 patients observed throughout the positivity period, the same CR-GNB was detected in the blood cultures in only 8 (8%) of the 99 febrile episodes recorded.

Conclusions: Our center epidemiology, with most frequent metallo-beta-lactamases resistance mechanism, differs from the national Italian pattern, where KPC is the most frequent enzyme of resistance, possibly due to the high number of patients coming from Eastern Europe. The colonization by CR-GNB does not necessarily translates into an infection and most febrile episodes in colonized patients could not be attributed to the CR-GNB, which supports the concept of sparing anti-microbial agents selected according to the pattern of the isolates unless sepsis criteria are encountered.

Disclosure: Nothing to declare.

18: Paediatric Issues

P656 ALPHA/BETA-DEPLETION AND EARLY TIMING OF SEROTHERAPY PROVIDES RELIABLE ENGRAFTMENT AND LOW TRANSPLANT RELATED MORBIDITY IN CHILDREN UNDERGOING HEMATOPOIETIC STEM CELL TRANSPLANTATION FOR MUCOPOLYSACCHARIDOSIS

Annika Ewert 1, Anna Zychlinsky Scharff1, Rita Beier1, Britta Maecker-Kolhoff1, Isolde Schridde1, Kirsten Mischke1, Karl Walter Sykora1, Martin Sauer1

1Hannover Medical School, Hannover, Germany

Background: HSCT is the current standard of care for children with MPSIH, and an important treatment option for patients with MPS1H/S, MPS-IS, MPS II, MPS IV, and MPS VII. In 2005, the EBMT released guidelines suggesting a donor-type hierarchy as follows: Non-carrier-HLA-matched family donor (MFD), matched unrelated cord blood (UCB), and matched unrelated donor (MUD). UCB has become established as a graft source with good engraftment rates at an acceptable rate of GVHD. Although the risk of graft failure after UCB-HSCT has been reduced in recent years, the trend of negative outcomes has shifted from autologous reconstitution to aplastic graft failure. Here, we report outcomes using an alternative approach: a standardized myeloablative regimen, ab-depleted peripheral blood stem cells (PBSC) from MUDs, and early timing serotherapy in the conditioning process.

Methods: We performed a mono‐centric, retrospective study examining 24 patients with Mucopolysaccaridosis who underwent HSCT at Hannover Medical School between 2003 and 2022. Mean age at HSCT was 1.96 years (range 1.41–2.56). 41.7% of the patients were female. We collected clinical data from patients’ medical records, including donor type, conditioning regimen, timing and dose of ATG, and chimerism on follow-up. The median clinical follow up time was 4.25 years (range 0.52- 10.14). The median follow-up for most recent chimerism was 1.77 years after transplantation (range 0.40- 8.00).

Results: Of 24 patients analyzed in this retrospective cohort, 5/24 received an ab-depleted MUD graft with early (defined as more than 7 days prior to transplantation) ATG (Grafalon at a dose of 10 mg/kg BW per dose on three consecutive days). In this ab-Group, the median number of CD34 positive cells was 17.76 x 10^6/kg BW (range 9.85-29.62), the median number of a/b positive cells in the grafts was 3.25x10^4/kg BW (range 2.18-4.26). 18/24 patients were transplanted with alternative T cell depletion methods (ATDM-Group, mainly CD34 positive-selected PBSCs). Median number of CD34 positive cells was 16.40x10^6/kg BW (range 8.93-31.93). One patient received an ab-depleted MUD graft with conventionally timed serotherapy. Donor-derived chimerism in the ab-Group settled at a median rate of 100% (range 98.04-100.0) between 30 and 360 days after HSCT. No graft failure occurred. Patients of the ATDM-Group reached median chimerism rates of 96.6% (range 82.65-100.0). Five ATDM-Group patients required donor lymphocyte infusions (DLI) in order to halt autologous recovery, while none of the patients in the ab-depletion group received DLI. Time to neutrophil engraftment was shorter in the ab-Group (median of 12 days after ab-depletion vs 16 days in the ATDM-Group). In the ab-depletion group, one patient was diagnosed with stage II aGVHD (16.7%). In the ATDM group 4/18 children were diagnosed with aGVHD (22.2%, 2 patients stage I, one patient stage II, one patient stage III).

Conclusions: We show first evidence that an ab-depleted MUD PBS-graft with early ATG-timing in combination with a myeloablative conditioning regimen can provide stable donor-derived long-term engraftment with a low risk of GVHD. Transplant-associated morbidity, often triggered and aggravated by GVHD in this patient cohort, was low.

Disclosure: Nothing to declare.

18: Paediatric Issues

P657 CARDIORESPIRATORY FITNESS, PHYSICAL PERFORMANCE, AND METABOLIC SYNDROME IN ADULT SURVIVORS OF PAEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANTATION

Anne Nissen 1, Tina Gerbek1, Kathrine Fogelstrøm1, Peter Schmidt-Andersen1, Kaspar Sørensen1, Abigail L. Mackey2,3, Martin Kaj Fridh1, Klaus Müller1,3,4

1Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark, 2Institute of Sports Medicine Copenhagen, Copenhagen University Hospital, Bispebjerg & Frederiksberg, Copenhagen, Denmark, 3University of Copenhagen, Copenhagen, Denmark, 4Institute for Inflammation Research, Copenhagen University Hospital, Rigshospitalet, Copenhagen, Denmark

Background: Long-term survivors of paediatric allogeneic hematopoietic stem cell transplantation (HSCT) are at high risk of developing treatment-related late effects, including impaired cardiorespiratory fitness and physical performance as well as metabolic syndrome (MetS). This high incidence of MetS has been ascribed to the transplant and the conditioning regimen, but it remains unknown to what extent, reduced cardiorespiratory fitness and physical performance may account for the high risk of MetS among the survivors.

Methods: This cross-sectional, follow-up study included 90 survivors of paediatric HSCT (median age 30.3 years, 46 men) with a median time from HSCT to follow-up examination of 20.2 years (range: 5.9;37.0 years), and 32 healthy, age- and sex-matched controls (median age 29.0 years). Cardiorespiratory fitness was evaluated by a cardiopulmonary exercise test on an electronically braked cycle ergometer. We examined physical performance through sit-to-stand 30 s and 60 s, handgrip strength, timed-up-and-go, walking pace, and 6-minute-walk tests.

We assessed blood pressure and waist circumference, while fasting plasma samples were examined for HDL-cholesterol levels, triglycerides, and glucose concentrations. MetS was defined according to the definition proposed by the American Heart Association.

Multiple regression analyses corrected for age and sex were applied for categorial covariates and the physical outcomes, and multiple regression analysis corrected for sex were applied for continuous outcomes. A p-value below 0.05 was considered significant.

Results: HSCT survivors demonstrated significantly lower performance levels in all aspects of cardiorespiratory fitness and physical performance outcomes compared to healthy, age- and sex-matched controls, resembling levels observed in the elderly population.

HSCT-variables including malignant or benign diagnoses, total body irradiation, acute graft versus host disease (GvHD), chronic GvHD and donor match showed no significant associations to outcomes from any of the physical tests.

MetS was present in 28% of survivors and 76% fulfilled one or more criteria for MetS. MetS associated negatively with VO2 peak (mean difference (MD) -4.6 ml/kg/min, CI95 [-8.7;-0.5], p=0.03), the 6-minute walk test (MD -55.5 m, CI95 [-102.8;-8.2], p=0.02), walking pace (-0.3 m/s, CI95 [-0.5;-0.02], p=0.03) and the timed-up-and-go test (MD 0.9 s, CI95 [0.3;1.4], p=0.003), yet survivors without MetS also exhibited low levels of fitness and physical performance when compared to healthy controls.

Conclusions: In this study, we demonstrate that long-term survivors of paediatric HSCT are at risk of having a severely reduced cardiorespiratory fitness and impaired physical performance as compared with healthy, age- and sex-matched controls, irrespective of HSCT-related variables. Further, we present evidence indicating that the high incidence of MetS in these survivors may be partially ascribed to reduced physical performance. Overall, this data underlines the importance of monitoring physical performance status in the follow-up care of all survivors of paediatric HSCT, and the potential of reducing the risk of MetS by physical training should be further investigated.

Disclosure: Nothing to declare.

18: Paediatric Issues

P658 HEMATOPOIETIC TRANSPLANTATION WITH CD45RA+ DEPLETION AND ADOPTIVE THERAPY WITH CD45RO- T-LYMPHOCYTES AND NK CELLS IN PEDIATRIC PATIENTS DIAGNOSED WITH HEMATOLOGICAL MALIGNANCIES AND NON-MALIGNANT DISEASES

Mercedes Gasior Kabat 1, David Bueno1, Luisa Sisinni1, Yasmina Mozo1, Raquel De Paz1, Dolores Corral1, Mikel Fernandez Artazcoz1, Ana Belen Romero1, Antonio Marcos1, Victor Jimenez-Yuste1, Antonio Pérez-Martínez1

1Hospital Universitario La Paz, Madrid, Spain

Background: We describe implementation of clinical program of pediatric HSCT with naïve T-cells depletion/CD45RA-/RO+ T-cells adoptive therapy and NK cells in University Hospital La Paz, Madrid, Spain.

Methods: Results structured in 3 stages: “group 1” HSCT with CD45RA-depleted graft, “group 2” CD45RA-depleted HSCT and non-prophylactic adoptive therapy with CD45RA-/RO+ T-cells, “group 3” CD45RA-depleted HSCT, adoptive therapy with prophylactic CD45RA-/RO+ T-cells and NK-cells on day +7.

Fifty-one pediatric patients received HSCT with CD34+ enrichment/CD45RA+ depletion from February 2015-September 2021. Fifty-six HSCT collected, source mobilized peripheral blood in all cases. Leukapheresis product processed using CliniMACS device (Miltenyi Biotec).

Results:

Group 1

Group 2

Group 3

p

HSCT procedures

15

11

30

Patients

14

11

26

Median age (%), range

8 (0,5-15)

12 (4-15)

9 (1-15)

0,099

Diagnosis, n (%)

0,074

AML

3 (20)

2 (18,2)

7 (23,2)

B-ALL

2 (13,3)

3 (27,3)

14 (46,7)

T-ALL

3 (13,3)

1 (9,1)

5 (16,7)

Mixed-phemotype leukemia

1 (6,7)

1 (9,1)

Aplastic Anaemia

1 (6,7)

2 (18,2)

1 (3,3)

Immunodeficiency

5 (33,3)

1 (9,1)

MDS

1 (9,1)

2 (6,7)

Amegakaryocytic thrombocytopenia

1 (6,7)

Hodgkin lymphoma

1 (3,3)

Disease status, n (%)

0,791

(malignacies)

CR1-CR2

5 (62,5)

6 (75)

21 (72,4)

>CR2

2 (25)

2 /25)

7 (24,1)

Refractory

1 (12,5)

1 (3,4)

Previous HSCT, n (%)

3 (20)

2 (18,2)

11 (36,7)

0,353

Donor, n (%)

0,113

Haploidentical

13 (86,7)

10 (90,9)

26 (86,7)

MRD

1 (9,1)

4 (13,3)

MUD

2 (13,3)

Fifty-six HSCT performed in 51 patients, median age 9 years (IQR 7,5). Forty-nine haploidentical donors (87,5%), 1 mismatch non-related (2%), 4 identical related (7%), 2 identical non-related (3,5%). Diagnosis: 19 ALL-B (34%), 8 T-ALL (14,3%), 12 AML (21,4%), 2 mixed phenotype leukemia (3,6%), 3 MDS (5,3%), 1 Hodgkin lymphoma (1,8%), 4 severe aplastic anaemia (7%), 6 immunodeficiencies (10,8%), 1 amegakaryocytic thrombocytopenia (1,8%). Patients received 2 cellular products in first 2 stages of the protocol and 3 in group 3. First product median 6,41x106/Kg CD34+ (IQR 3,05x106/Kg). Second product median 1x104/Kg (IQR 4,95x104) CD3+CD45RA+ and 5x107/Kg (IQR 3,44x107/Kg) CD45RO+. Median CD45RA+ depletion log10 4,75 (IQR 2,45). Third product: CD3+ depletion/CD56+ enrichment, median 1.7×107/kg (IQR 1,84×107/kg) CD3-CD56+ and 2,4×103/kg (IQR 9,06×103/kg) of CD3+CD56-.

Neutrophil engraftment was median 10 days (IQR 1), 20 x109/L platelets median 13 (IQR 6), 50 x109/L median 16 (IQR 8,75). Two (3,57%) primary graft failure (GF). Four (7,14%) secondary GF, median 38,5 days (IQR 38,75).

Incidence aGvHD≥grade II-IV at 1 year 49,4% (CI95% 35-63,8%) group 1, 50,9% (CI95% 34,5-67,3%) group 2, 49,7% (IC95% 40,3-59,1%) group 3 (p=0,877). Chronic GvHD incidence: 16,7% (CI95% 1,5-31,9%) group 1, 46% (CI95% 28,7-63,3%) group 2 and 29,7% (CI95% 20,3-39,1%) group 3 (p=0,373).

CMV reactivation: 6 (40%) in group 1, group 2 in 5 (45,4%), group 3, 9 (30%), no significant differences. HHV-6 encephalitis in 1 (6,5%) and 2 (18,2%) in groups 1 and 2 respectively, no cases in group 3.

Two-year relapse incidence 43% (CI95% 26-60%) group 1, 20% (CI95% 2,1-37,9%) group 2, 9,1% (CI95% 2,9-15,3%) group 3 (p=0,029). Two-year TRM significantly lower in group 3 (p=0,001). Two-year-OS and significantly higher in group 3: 86,5% (CI95% 80,2-92,8) (p=<0,001), and 2-year EFS significantly higher in group 3 (p=<0,001): 20% (CI95% 9,7-30,3%) group 1, 18,2% (CI95% 6,6-29,8%) group 2 and 78,6% (CI95% 70,8-86,4%) group 3. One-year GRFS 6,7% (CI95% 0,3-13,1%), 18,2% (CI95% 6,6-29,8%), 52,7% (Ic95% 43,5-61,9%) groups 1, 2, 3 respectively. One-year GRFS significantly higher in group 3 (p=0,002). CD4+ cells reached >0,1 x103/mcL in groups 1 and 3, 1 month after HSCT, continued increasing progressively.

Conclusions: CD45RA-depleted HSCT showed fast engraftment and low GF. Acute/chronic GvHD higher compared to other groups using same platform. At beginning, high rate of HHV-6 encephalitis, no cases in group 3 after NK cell infusion implemented.

Relapse-incidence decreased significantly as protocol evolved. TRM was significantly lower in last stage of protocol; OS, EFS, GRFS significantly higher in group 3.

Disclosure: Nothing to declare.

18: Paediatric Issues

P659 PSYCHOLOGICAL IMPACT OF HEMATOPOIETIC STEM CELL DONATION ON PEDIATRIC SIBLING DONORS

Sara Mostafa Makkeyah 1, Nihal Hussien Aly1, Eslam Elsayed Elhawary2, Sally Abdelmonsif Mohammed1, Salwa Amin Abdelhamid1

1Ain Shams University, Cairo, Egypt, 2Tanta University, Tanta, Egypt

Background: Pediatric sibling donors of hematopoietic stem cells (HSC) are at increased risk of psychological difficulties. The psychological requirements of young sibling donors are frequently disregarded, and less is known about the psychological dangers of donation which sometimes outweigh the physical concerns. We aimed to evaluate the possible psychological impacts of HSC donation in pediatric sibling donors of patients undergoing HSCT, and to compare them to a group of non-donor siblings.

Methods: To explore the different aspects of mental health, we used the following five questionnaires: the Child Anxiety Related Emotional Disorders (SCARED) for anxiety, the Children’s Depression Inventory (CDI) to assess depression, the Rosenberg self-esteem scale (RSES) which measures self-esteem, the Revised Child Impact of Event Scale (CRIES-13) for the assessment of post-traumatic stress disorder, and the Decision Regret scale (DRS) which evaluates distress or regret following a healthcare related decision. All questionnaires were administered following the completion of the patient’s admission for HSCT.

Results: We compared twenty-two sibling donors (mean age 12.5±4.5 years, 16 males and 6 females) to fifteen healthy non-donor siblings (mean ± SD of age was 11.3±4.0 years, 9 males and 6 females). Almost half of the patients (10/22) were transplanted for thalassemia. Other diagnoses included relapsed/refractory acute leukemia (18%), severe aplastic anemia in four patients (18%), two patients had primary immunodeficiency (9%), and one patient with Fanconi anemia. None of the participants regretted the decision of HSC donation to their siblings. Forty-one percent of donor siblings suffered from anxiety disorders with mean total anxiety score of 21.6±9.3 as compared to 15.6±8.5 in the control group (p= 0.026). On analysis of different anxiety subdomains, donors had higher scores in general anxiety (5.0±3.0 vs 3.1±2.7, p= 0.031) and in panic anxiety (3.8±3.6 for donors vs 1.9±2.6 for controls, p= 0.0448), while other domains were comparable between the two groups (separation anxiety 5.5 ±2.9 vs 5.2±2.7, p= 0.394; social anxiety 6.3±4.0 vs 4.9±3.0, p= 0.120; significant school anxiety 1.0±1.0 vs 0.5±0.8, p= 0.058). Total anxiety score did not correlate with age at study entry (r=0.044, p=0.845) or age at HSC donation (r=0.02, p=0.929). Nine percent of donors showed severe depression and 13.6% had low self-esteem (defined as Rosenberg score of less than 15). Although donor siblings scored higher in the avoidance subdomains of the CRIES questionnaire evaluating post-traumatic stress (6.2±3.7 vs 3.1±2.3, p= 0.003), they had lower scores in the intrusion subdomain (6.5±3.1 vs 8.9±3.7, p=0.0218), and collectively, the total CRIES scores were comparable between both groups (19.8±8.7 vs 18.1±9.3 for donors and controls respectively, p= 0.281). Both groups showed comparable scores on the Rosenberg self-esteem questionnaire (donors 17.9±3.1 versus controls 19.1±4.9, p= 0.178).

Conclusions: Pediatric sibling donors have higher frequency of anxiety and increased risk in some post-traumatic stress domains. None of the donors showed regret for donating HSC. It is imperative to prioritize the provision of sufficient psychosocial assistance to young sibling donors and their mental health.

Disclosure: Nothing to declare.

18: Paediatric Issues

P660 AUTOLOGOUS STEM CELL TRANSPLANTATION IN HIGH-RISK NEUROBLASTOMA: A LONG-TERM FOLLOW-UP

Ana Pinto1, Isabelina Ferreira 1, Gilda Teixeira1, Ana Sofia Jorge1, Pedro Sousa1, Maria João Gutierrez1, Bárbara Marques2, Ana Forjaz Lacerda1, Fernando Leal-da-Costa1, Nuno Miranda1

1IPOFG, Lisboa, Portugal, 2INSA, Lisboa, Portugal

Background: Neuroblastoma is the most common extracranial solid tumour of childhood. It arises from the developing sympathetic nervous system from neural crest cells, usually resulting in tumours in the adrenal glands or the sympathetic ganglia. It is the most common solid malignancy in the first year of life with a median age at diagnosis of 18 months and 90% of cases are diagnosed in the first decade of life.

The clinical presentation is highly variable, ranging from an asymptomatic incidental mass to a symptomatic widely disseminated disease. The clinical and biological heterogeneity leads to differences in outcome from spontaneous regression to metastasis, inexorable progression and death despite intensive therapy.

Methods: A retrospective analysis was performed on patients with high-risk neuroblastoma, INSS stage 2-3 with MYCN amplification or stage 4, after autologous SCT between 1994/10/13 and 2020/03/16. Response to previous treatment, MYCN status, pre-transplant NSE (above or normal range) was collected. Conditioning regimen was mainly busulfan + melphalan. Post-transplant therapy included radiation, dinutuximab beta, 13-cis-retinoic acid. Overall survival was calculated according to Kaplan Meier, using log-rank tests for group comparison, with a significance level of 5%.

Results: 68 patients were included, median follow-up 6.8 years (0.3– 25.3 years); 73.5% males, median age 3.9 years (16 months – 27 years), 85.3% in stage 4, 29.4% with MYCN amplification (20/65), 50% had elevated pre-transplant NSE (24/ 48); prior to HDCT 51.5% were in complete response and 48.5% in partial response. When data was censored, 39.7% were alive (1 toxic death). The 5-year and 10-year OS were 36% (95%CI 24-42) and 34% (95%CI 22-40). There was no significant difference in the OS between children transplanted before and after 2010 (p = 0.24), response to previous therapy (p = 0.66), MYCN status (p = 0.79) and NSE value (p = 0.81).

Conclusions: Although autologous SCT have improved the prognosis of this patients, the outcome is still unsatisfactory. The main cause of failure after transplantation is tumor progression % The 5-year and 10 years OS were respectively 35.7% and 33.5%, very similar to previous reported data. The majority of events occurred in the first 5 years after transplantation with only one toxic dead.

In this study, we could not demonstrate a statistically significant difference between the overall survival of patients transplanted after complete or parti’al remission. In this study, there was no significant difference in the survival of transplanted patients based on MYCN status but the big majority of NMYC amplified had been submitted to high dose chemotherapy.

When we compare the results from 1994 until 2010 and from 2010 until 2021 there was no significant difference.

The pre-transplant NSE status also had no significant impact in overall survival.

Our data favours that transplant should be pursued even with partial response, but better strategies are in need.

Disclosure: The authors declare no conflit of interests.

18: Paediatric Issues

P661 OUTCOMES OF ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN WITH DIAMOND-BLACKFAN ANEMIA – REPORT OF THE POLISH PEDIATRIC STUDY GROUP FOR HEMATOPOIETIC STEM CELL TRANSPLANTATION

Anna Pieczonka 1, Olga Zajac-Spychala1, Krzysztof Kałwak2, Marek Ussowicz2, Jolanta Goździk3, Katarzyna Drabko4, Jan Styczynski5, Agnieszka Sobkowiak-Sobierajska1, Katarzyna Derwich1, Jacek Wachowiak1

1Poznan University of Medical Sciences, Poznan, Poland, 2Wrocław Medical University, Wrocław, Poland, 3Jagiellonian University Medical Colege, Kraków, Poland, 4Medical University of Lublin, Lublin, Poland, 5Colegium Medicum, Nicolaus Copernicus University Torun, Bydgoszcz, Poland

Background: Diamond-Blackfan anemia (DBA)) is a rare congenital pure red cell aplasia related to mutations in the genes encoding ribosomal proteins of small and large ribosomal subunits. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only one curative treatment option, which is indicated in case of non-response to steroids, steroid dependency at a dose ≥0.3 mg/kg/day, transfusion dependency, alloimmunization to RBC, progressive pancytopenia, or clonal evolution to MDS/AML. The aim of the study was assessment of the outcomes of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in DBA patients.

Methods: Between 1998-2021, twenty nine (12 girls, 41.4%; 17 boys, 58.6%) patients with DBA were transplanted in the Polish Pediatric Study Group for Hematopoietic Stem Cell Transplantation centres. Median age at HSCT was 2 years (range, 1-14 years). Nineteen patients (65.5%) were transplanted from a matched unrelated donor (MUD); 7 children (24.2%) from a matched sibling (MSD) and 3 (10.3%) from a haploidentical donor. The stem cell source was bone marrow in 50.3% of the patients (n=14) and peripheral blood in 51.7% of them (n=15). The preparative regimen was busulfan/busilvex-based in 15 patients (51.7%) and treosulfan-based in 14 (50.3%).

Results: Initial engraftment was achieved in all but three (10.3%) patients. One patient (3.4%) experienced secondary graft failure. For two children (6.9%) more than 1 transplantation was performed. The one-year cumulative incidence of aGvHD II-IV was 29.8% The one-year cumulative incidence of cGvHD was 11.9%, being limited in n=3 and extensive in n=1. Five (17.2%) patients died during follow-up (11-390 days after HSCT median 82 days) due to infections (n=4) or bleeding into the central nervous system (n=1); two of them were transplanted from haploidentical donor in 1999-2000. Twenty four (82.7%) patients are alive with median follow-up of 10 years (range 0.95-21 years). Five-year overall survival and event-free survival were 82% and 78%, respectively. Five-year overall survival for patients prepared for HSCT with busulfan were 85% and 80% for children prepared with treosulfan. Five-year overall survival were 100% and 84% in children who received cells from MSD and from MUD, respectively.

Conclusions: This data shows that nowadays both allo-HSCT from MSD and MUD is efficient and safe for DBA patients.

Disclosure: Nothing to declare.

18: Paediatric Issues

P662 OUTCOMES OF ALLO-HSCT FROM MISMATCHED DONORS WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE IN CHILDREN WITH NON- MALIGNANT DISEASES

Tatyana Bykova 1, Anna Osipova1, Olga Slesarchuk1, Oleg Goloshchapov1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1

1RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russian Federation

Background: Allogeneic hematopoietic cell transplantation (allo-HSCT) is a potentially curative therapy for a variety nonmalignant diseases (NMD), either inherited or acquired. A fully human leukocyte antigen (HLA)-matched sibling (MSD) is considered the “gold standard” graft source for patients undergoing allo-HSCT. Alternative donor strategies such as mismatched unrelated donors (MMUD) or haploidentical donors (haplo) associated with increased risk graft failure and graft-versus-host-disease (GVHD), major factors affecting survival probability and quality of life after HSCT. Post-transplant cyclophosphamide (PTCy) is effective GVHD prophylaxis option for adult patients (pts) with malignant diseases, but there are limited data in children with NMD.

Methods: Between September 2011 and October 2023, 55 pediatric patients with NMD received allo-HSCT from HLA-mismatched donors with PTCy-based GVHD prophylaxis. Diagnosis were: severe aplastic anemia(SAA) – 16 (29%), inborn bone marrow failure syndromes (IBMFS) – 2 (4%), hemoglobinopathies – 7 (13%), primary.

immunodeficiency (PID) – 22 (40%), inborn metabolic disorders (IMD) – 8 (14%). Median time from diagnosis to transplant was 9 (0.5-123) and 2 month (0.5-3) for the whole cohort and SCID patients, respectively. Conditioning regimen (CR) were: MAC – 11 (20%), RTC – 27 (49%), RIC – 16 (29%), one SCID pt. received HSCT without CR. GVHD prophylaxis was based on 50 mg/kg PTCy on day +3, + 4 in combination with calcineurin inhibitors +/- MMF (n=27), mTOR inhibitors +/- MMF (n=24); ATG was added in 15 pts., ruxolitinib in 8 pts., other agents in 3 pts.

Results: A total of 42 pts (82%) (MMUD – 9/12 (75%), haplo – 37/43 (86%), p=0.39) achieved primary engraftment; 4 pts died before engraftment due to infectious complications; 3 pts after haplo-HSCT experienced secondary graft failure (GF). Median time to neutrophil recovery was 24 (13-39): for MMUD – 23 (13-32), for haplo – 24 (17-39), p=0.75. Median time to platelet recovery was 25 days (11-109): for MMUD – 17 (11-37), for haplo – 26 (12-109), p=0.04. Eight pts were retransplanted (4 pts from the same donor, 4 pts from another donor) resulted in engraftment in 7 pts. The cumulative incidence (CI) of acute GVHD (aGVHD) II-IV and III-IV was 18% and 6%, respectively, with no significant difference between MMUD and Haplo. Three pts developed chronic GVHD with CI of 10%, severe in all cases (MMUD 2, Haplo 1). With a median follow-up of 24 months (3-119), the 3-year probability of overall survival was 78% (83%, 77% in MMUD and haplo groups respectively, p=0.54). GVHD-free/failure-free survival was 54% (55%, 53% for MMUD and haplo groups, respectively, p=0.83).

Conclusions: Allo-HSCT from mismatched unrelated and haplo donors with PTCy-based GVHD prophylaxis is associated with promising outcomes in children with different NMD with acceptable rate of graft failure, severe acute and chronic GvHD.

Disclosure: Nothing to declare.

18: Paediatric Issues

P663 CARDIOVASCULAR RISK FACTORS AND SUBCLINICAL ORGAN DAMAGE ARE COMMON AND INCREASING OVER TIME IN CHILDREN AFTER HEMATOPOIETIC STEM CELL TRANSPLANTATION

Damaris Werner 1, Jeannine von der Born1, Elena Lehmann1, Nima Memaran1, Britta Maecker-Kolhoff1, Martin Sauer1, Anette Melk1, Rita Beier1

1Medizinische Hochschule Hannover, Hannover, Germany

Background: Advances in hematopoietic stem cell transplantation (HSCT) result in an increasing number of long-term survivors with cardiovascular (CV) morbidity and mortality becoming a relevant disease burden. While children and young adults rarely present with clinically apparent CV disease (CVD), CV risk factors and subclinical organ damage might be predictive for the development of manifest CVD.

Methods: We enrolled 81 children after allogeneic HSCT at in a prospective longitudinal study assessing annually anthropometric data, laboratory parameters, office and 24h ambulatory blood pressure monitoring (ABPM), aortic pulse wave velocity (PWV), and carotid intima media thickness (IMT). 29 of the patients were included immediately before HSCT while 52 had already undergone HSCT at study inclusion. We obtained a total of 29 observations before and 283 after transplantation. For this analysis, we included data obtained post HSCT only. Z-scores adjusted for sex, age or height were calculated for anthropometric assessment, blood pressure (BP), ABPM, PWV, and IMT.

Results: The baseline characteristics of the patients are depicted in Table 1. We repeatedly (at least twice) detected classical CV risk factors (elevated waist circumference (WC), elevated BMI or arterial hypertension (aHTN)) in 18% of our patients. Subclinical organ damage (elevated PWV or elevated IMT) was identified in as many as 42%. Especially elevated IMT was very common with 37% of children repeatedly exhibiting values above the 95. percentile. 29% of patients presented with at least one of the assessed CV risk factors.

In univariate analysis, younger age at HSCT and longer follow up after HSCT correlated with higher IMT and higher systolic BP (in both office measurements and ABPM). Meanwhile, higher physical activity (self-reported in hours per week) correlated with lower systolic BP, lower PWV, and lower IMT. In our cohort, patients with non-malignant diseases had higher systolic BP, higher PWV, and higher IMT. Higher systolic BP was also observed in children after chemotherapy-based conditioning regimens, busulfan exposure, and with chronic GVHD.

The association between higher systolic BP and younger age at HSCT as well as chronic GVHD was confirmed in multivariable analysis. Time after HSCT did not reach significance. A correlation between higher IMT and younger age at HSCT and longer time (in this case since study inclusion) was also confirmed.

Patient characteristics

Number (%) or average ± SD (range)

Sex (m/f)

46 (57%) / 35 (43%)

Age at HSCT (years)

10.5 ± 4.5 (0.8 – 18.1)

Age at study inclusion (years)

13.1 ± 4,5 (6.0 – 25.6)

Follow-up time since HSCT (years)

5.9 ± 4.4 (0.2 – 16.7)

Follow-up time since study inclusion (years)

3.3 ± 2.6 (0 – 9.6)

Number of visits

3.9 ± 2.0 (1 – 8)

Underlying disease

Malignant

56 (69%)

• ALL or lymphoma / AML /MDS

• 34 (42%) / 11 (14%) / 11 (14%)

Non-Malignant

25 (31%)

• Hematological / immunological / other

• 13 (16%) / 7 (8%) / 5 (6%)

HSCT characteristics

Conditioning (chemo-based / TBI > 4 Gy)

59 73%) / 22 (27%)

• Busulfan exposition

• - 14 (17%)

Graft origin (MRD / MUD / Haplo)

24 (30%) / 56 (69%) / 1 (1%)

Complications

aGvHD Grade III/IV

3 (4%)

cGvHD

12 (15%)

VOD

16 (20%)

Conclusions: CV risk factors and subclinical organ damage are common in survivors of pediatric HSCT. Known risks factors such as obesity and arterial hypertension are common and may offer targets for intervention while transplant-specific risk factors such as certain underlying diseases, busulfan exposure and chronic GvHD may be helpful in identifying patients with a need for intensified follow-up. Importantly, IMT and possibly SBP seem to aggravate over time. This stresses the importance of a tailored follow up program for HSCT survivors. In this context, special attention might be given to the transition from pediatric to adult follow-up care.

Disclosure: Nothing to declare.

18: Paediatric Issues

P664 POSITIVE PRE-TRANSPLANT RESPIRATORY VIRAL PCR IS ASSOCIATED WITH INCREASED DAY 100 TRANSPLANT-RELATED MORTALITY IN PEDIATRIC HSCT RECIPIENTS

Jane Trainor 1, Benjamin Hanisch1, Kelly Lyons Snelling1, Robert Podolsky1, David Alex Jacobsohn1

1Children’s National Hospital, Washington, United States

Background: Hematopoietic Stem Cell Transplant (HSCT) is a curative procedure for many life-threatening hematological indications. Respiratory viral infections (RVI) cause substantial morbidity and mortality in pediatric recipients. Prior studies have found that patients receiving myeloablative conditioning with RVI lower respiratory disease have increased mortality and fewer days alive and out of the hospital. The impact of mild/asymptomatic RVI is unclear.

To reduce transplant morbidity, a pre-transplant RVI screening program was started at Children’s National Hospital which included symptom screening and Respiratory viral (RV) PCR testing for each HSCT patient within one week of and on day of HSCT admission. RV PCR information was incorporated into the decision to proceed with transplantation. The objective of this study was to evaluate the impact of RVI on HSCT outcomes.

Methods: A retrospective review of pre-transplant RV PCR, symptom screening, and clinical outcomes was done for all patients receiving allogeneic HSCT from 7/1/2016- 3/31/2023. Prior studies have classified viral infections into high- or low-risk categories for post-transplant complications. We analyzed the impact of positive RV results on outcomes within 100 days post-HSCT (mortality, grade 3/4 GVHD, adenoviremia, transplant-associated thrombotic microangiopathy (TA-TMA), need for positive pressure ventilation, intubation, bronchoalveolar lavage, and ICU transfer). We used logistic regression for all analyses and the Holm method to adjust for multiple testing. Exclusion criteria included missing pre-transplant RV PCR as well as SCID (more likely to be RV positive and have poor outcomes) to prevent bias.

Results: 170 patients were eligible to be included. Covariates examined were age (median 9.7, range 0.3-28.6 years), gender (69 female, 101 male), malignancy (62), T-cell depletion (18), myeloablative conditioning (111), serotherapy (78), stem cell source (98 bone marrow, 18 cord, 54 PBSC), donor type (103 matched, 67 mismatched), and CMV concordance (96). Indications for transplant were hemoglobinopathies, leukemias, metabolic disorders, aplastic anemia, and HLH. Of the 170, 41 tested positive for RVs (33 low-risk, 8 high-risk). Outcomes were initially analyzed separately by low-risk and high-risk viruses. Given the small number of high-risk patients, statistical analysis was performed by looking at all positive RVs together. There were too few patients with RV symptoms to analyze separately. If patients were intentionally delayed based on an RV PCR result, the RV PCR results used in this study were the last pre-transplant one before admission.

Within the first 100 days post-transplant, a positive pre-HSCT RVI was significantly associated with increased mortality (odds ratio (OR) = 5.57, p = 0.04 after adjusting for multiple testing) and requirement for ICU transfer (OR = 3.45, p = 0.006). All other outcomes did not differ significantly based on pre-HSCT RV PCR positivity. In a sensitivity analysis, the statistical significance of the two associations did not change when potential confounding covariates were included in analyses. See table for all results.

Respiratory Virus Negative (% (95%CI)

Respiratory Virus Positive (% (95%CI)

Odds Ratio

P value

ICU Transfer

21.6 (15.5-29.4)%

48.8 (34.0-63.7)%

3.45

0.006

Day 100 transplant-related mortality

3.0 (1.1-7.7)%

14.6 (6.7-29.0)%

5.57

0.04

Positive pressure ventilation

17.2 (11.7-24.5)%

31.7 (19.4-47.3)%

2.24

NS*

Intubation

6.7 (3.5-12.4)%

19.5 (10.1-34.4)%

3.37

NS

Broncho-alveolar Lavage

8.2 (4.6-14.2)%

17.1 (8.4-31.7)%

2.30

NS

TA-TMA

5.2 (2.5-10.6)%

17.1 (8.4-31.7)%

3.74

NS

Adenoviremia

28.4 (21.4-36.6)%

29.3 (17.4-44.8)%

1.05

NS

Severe (grade 3-4) acute GVHD

20.9 (14.8-28.6)%

41.5 (27.6-56.9)%

1.72

NS

Conclusions: Routine pre-transplant viral testing should be performed to increase the safety of HSCTs. Pre-transplant viral testing may allow providers to know when to monitor certain patients more closely post-transplant, and when to potentially delay elective transplant until the patient has cleared their virus.

Clinical Trial Registry: N/A.

Disclosure: Nothing to declare.

18: Paediatric Issues

P665 HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PEDIATRIC PATIENTS WITH MYELODYSPLASTIC SYNDROME

Anna Osipova1, Tatiana Bykova 1, Olga Slesarchuk1, Olesya Paina1, Oleg Goloshchapov1, Tatiana Gindina1, Elena Semenova1, Alexander Kulagin1, Ludmila Zubarovskaya1

1RM Gorbacheva Research Institute, Pavlov University, Saint Petersburg, Russian Federation

Background: Childhood MDS is a heterogeneous group of hematopoietic stem cell clonal diseases, characterized by ineffective hematopoiesis with a high risk of transformation to acute leukemia. Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is the only curative therapy for children with myelodysplastic syndrome. Allo-HSCT from a haploidentical donor is relevant for patients without HLA-compatible related and unrelated donor, but current data are very limited.

Methods: Twelve patients who underwent 16 haploidentical transplants between 2008-2023 were included in the analysis. The median age was 8 years (1-18 years), gender – 7 male, 5 female. There were the following types of MDS: refractory cytopenia of childhood – 1 (8%); MDS with excess blasts 6 pts (50%), myelodysplasia-related acute myeloid leukemia 5 (42%). Cytogenetic findings included a normal karyotype – 4 pts (31%), monosomy 7 – 5 pts (42%) and trisomy 8 – 3 (25%). The median time from diagnosis to allo-HSCT was 9 months. Myeloablative conditioning regimens (MAC) were used in 8 pts (67%): Busulfan-based – 5 pts (62,5%), Treosulfan-based – 3 pts (37,5%); reduced-intensity conditioning (RIC) in 4 pts (33%). MAC consisted of Busulfan (Bu) 16 mg/kg + Cyclophosphamide 120 mg/kg. RIC included Fludarabine (Flu) 150 mg/m2 + Melphalan 140 mg/m2, Flu 150-180 mg/m2 + Bu 8mg/kg. The graft source was a bone marrow in 8 pts (67%), PBSC in 2 pts (16,5%) and combination of BM and PBSC in 2 pts (16,5%). GVHD prophylaxis included PTCy in 7 pts (58%), ATG – 5 pts (42%).

Results: The engraftment was diagnosed in 8 pts (67%) with median time of 18 days (14-23). Acute GVHD grade 1-2 was diagnosed in 5 pts (62,5%), mild chronic GVHD – in 3 (37,5%). Transplant related mortality was 39%. Relapses were in 3 patients (25%). 3-year overall survival (OS) and EFS were 62 % and 49%, respectively. OS after PTCY-based GVHD prophylaxis and ATG-based GVHD prophylaxis was 80% and 40% (p=0,154) respectively. Second allo-HSCT was performed in 4 children (for primary graft failure - 3, secondary graft failure-1). OS in patients transplanted within 15 months of diagnosis was 87.5%, while all patients transplanted later died of disease or transplant complications (p = 0.002).

Conclusions: Haplo-HSCT is feasible and effective therapy with more success before 15 months after diagnosis in pediatric MDS for patients lacking matched related or unrelated donor.

Disclosure: Nothing to declare.

18: Paediatric Issues

P666 COMPARISONS OF PROGNOSIS OF ALLO-HSCT AFTER CAR-T CELL THERAPY OR CHEMOTHERAPY IN PAEDIATRIC PATIENTS OF REFRACTORY/RELAPSED B-CELL ACUTE LYMPHOBLASTIC LEUKEMIA : A MULTICENTER STUDY CCCG-ALL-2015

Bohan Li 1, Jing Chen2, Chengjuan Luo2, Shaoyan Hu1

1Children’s Hospital of Soochow University, Suzhou, China, 2Shanghai Children’s Medical Center, Shang Hai, China

Background: Refractory/relapsed (R/R) B-cell acute lymphoblastic leukemia(B-ALL) is a leading cause of morbidity and mortality in children. R/R B-ALL patients often undergo consolidation allogeneic hematopoietic stem cell transplantation (allo-HSCT) to maintain long-term remission following chimeric antigen receptor (CAR) T cell therapy or chemotherapy. However, the effectiveness and safety of allo-HSCT after CAR-T therapy remain controversial. The objective of this study is to investigate the comparative prognosis between allo-HSCT followed by CAR-T therapy and chemotherapy for B-ALL in pediatric patients.

Methods: We analyzed the outcomes for 269 R/R B-ALL paediatric patients who received allo-HSCT after CAR-T therapy (CAR-T-allo-HSCT group, n=142) or with chemotherapy (Chemotherapy-allo-HSCT group, n=127) in a multi-institutional clinical study with ALL diagnosed in 20 hospitals from January 2015 to December 2022. Prior to allo-HSCT, all patients treated using the Chinese Children’s Cancer Group ALL-2015 (CCCG-ALL-2015) protocol registered with the Chinese Clinical Trial Registry (ChiCTR-IPR-14005706).

Chemotherapy(n=127)

CAR-T(n=142)

n

%

n

%

P value

Age, y

≥7

71

55.9

78

54.9

0.90

Median(range)

7.7(0.7-17.9)

7.9(1.3-17.8)

0.99

Patient sex

0.44

Female

40

68.5

55

38.7

Male

87

31.5

87

61.3

Disease risk index

0.40

Low

24

18.9

36

25.4

Intermediate

87

68.5

92

64.8

High

16

12.6

14

9.8

Disease status at HSCT

0.001

0.001

NR

8

6.3

2

1.5

PR

7

5.5

CR

112

88.2

140

98.5

<0.001

CR1

42

37.5

19

13.6

CR2

65

58.0

110

78.6

≥CR3

5

4.5

11

7.9

Results: The CAR-T-allo-HSCT group had more patients in second CR compared to the chemotherapy-allo-HSCT group (78.6% vs.58.0%; p<0.001) and more with negative MRD(86.9% vs 66.2%; p<0.001). The CAR-T-allo-HSCT cohort had a higher incidence of moderate/severe chronic graft-versus-host disease (mod/sev cGVHD,19.09% [95% CI: 12.15-26.03%] vs. 7.33% [95%CI: 25.2-26.0%]; p=0.016). In CR1 subset, the CAR-T-allo-HSCT group had lower overall survival(OS,41.30% [95% CI: 21.60-78.90%] vs. 84.80% [95%CI: 74.20-96.90%]; p=0.006), grade II-IV acute GVHD mod/sev cGVHD and relapse free survival(GRFS,32.10% [95% CI: 14.60-70.50%] vs. 66.90% [95%CI: 53.60-83.60%]; p=0.02) and higher non-relapse related mortality (NRM, 40.74% [95% CI: 11.82-69.22%] vs. 10.29% [95%CI: 0.50-20.09%]; p=0.03)compared to the chemotherapy group. Furthermore, in the CAR-T-allo-HSCT group, CAR-T treatment and allo-HSCT separated by more than 61 days had a higher GRFS(>61:64.3% [95% CI: 52.74-75.86%] vs <61: 38.3% [95%CI: 25.95-50.65%]; p=0.001), lower incidence of grade II-IV aGVHD(>61: 19.44% [95% CI: 10.28-28.65%] vs <61: 41.27% [95%CI: 29.00-53.54%]; p=0.003)and III-IV aGVHD(>61: 6.94% [95% CI: 1.04-12.84%] vs <61: 26.98% [95%CI: 15.80-37.99%]; p=0.001).

Conclusions: The CAR-T therapy eliminates pre-HSCT MRD, and OS and RFS were similar to to those in the chemotherapy group. But the CAR-T-allo-HSCT cohort had a higher incidence of mod/sev cGVHD. In the CR1 subset CAR-T-allo-HSCT group had a worse prognosis compared to the chemotherapy-allo-HSCT group. The CAR-T treatment and allo-HSCT intervals longer than 61 days would have higher GRFS and lower incidence of II-IVaGVHD and III-IV aGVHD.

Disclosure: Nothing to declare.

18: Paediatric Issues

P667 TCR-ΑΒ DEPLETED HAPLOIDENTICAL TRANSPLANT WITH CD 45 RA- DEPLETED MEMORY T CELL ADD-BACK IN PAEDIATRIC NON-MALIGNANT DISEASES IN A SINGLE PAEDIATRIC CENTER: 10-YEAR RESULTS

Tan Ah Moy 1,2, Michaela Seng1,2, Pham Ngi Thoc Anh1, Vijayakumari K1, Mya Soe Nwe1, Prasad Iyer1,2, Shui Yen Soh1,2, Joyce Lam1,2, Wing Leung1,3

1KK Women’s and Children’s Hospital, Singapore, Singapore, 2Duke-NUS Medical School, Singapore, Singapore, 3University of Hong Kong, Hong Kong, Hong Kong, SAR of China

Background: A major barrier to paediatric haematopoietic stem cell transplant in Southeast Asia has been the difficulty of finding an appropriate HLA matched donor. Haploidentical donor (H-ID) transplantation has allowed us to proceed to a transplant with comparable outcomes to unrelated donor transplant for non-malignant disorders. This is particularly important in a situation where a lack of local donors, significant time for procurement and concerns using cryopreserved donor products during COVID-19 pandemic present as barriers to transplant.

Methods: A review of retrospective data was approved by institutional ethics. GSCF-mobilised peripheral stem cells were divided in a 9:1 or 8:2 ratio for TCR-αβ -depletion and CD45 RA -depletion respectively using the CliniMACS platform. Target CD34+ doses were 8-10x10(6)/kg and a maximum TCRA/BCD3+ cell dose of 2.5x10(4)/Kg, CD45RA-CD3+doses between 1-5x10(6)/kg. Data was censored at patient’s last visit.

A reduced intensity (RIC) conditioning regime TLI 6 Gy, Fludarabine(Fluda)160 mg/m2, Thiotepa (TT)10mg/kg and Melphalan 140mg/m2 was used in most cases. Myeloablative (MAC) conditioning was required using ATG, Busulfan/Treosulfan, Fludarabine, Cyclophosphamide (CPA), TT for Thalassemia and some Inborn Errors of Immunity(IEI) with a high risk of rejection.

Tarcolimus was used as anti- GVHD medication. KIR genotyping and donor specific antibody measurement were done in all to select the better donor. Disease indication, conditioning chemotherapy, graft details, engraftment, viral reactivations, chimerism, GVHD and overall outcome were studied.

Results: Fifty- two patients underwent TCR-αβ depleted H-ID transplant between 2014-2023(June). Of these 28 (54%) were for nonmalignant disorders. Severe aplastic anemia (SAA) /Bone marrow failure syndrome (BMFS) 10, Blood transfusion dependent Thalassemia (Thal)8 and PID10; Chronic granulomatous disease3, CD40 Ligand deficiency2, Interferonopathy1, Chronic Active EBV infection1, SCID2, Wiskott-Aldrich syndrome1.

TRM at 1 year was 1/28(4%). Graft-rejection occurred in 2 patients with SAA =2/28(7%) both salvageable with second H-ID using a different donor. Median neutrophil engraftments 14.5days(10-56) while median platelets engraftment 13.5 days(7-47 days). Viral reactivations were common and occurred in 21/28(72%). The viruses were CMV(10), EBV(8), HHV6(8), adenov(6), BKV(3) while parvovirus, RSV and influenza occurred in one patient each. Most of virus responded well to antiviral agents except one case of adenovirus required Virus-Specific-T cells. Fatal viral infections occurred in one pt with SAA with RSV 6 months post-transplant and one from influenza at 3.5 year post BMT in a BMFD with chest deformity. Acute graft versus host Grade I-IV occurred 8/28(29%); only one had Grade III and the rest were Grade I-II. None had chronic GVHD. The 5yr OS was 90%.

During this period we performed 21 allogeneic transplants for non-malignant disease using MSD 6; MUD13; UCBT2. TRM at 1 year was nil. GR occurred in 2/21 (10%), one from UCBT and one MUD, both salvaged by second transplant. 1 died of severe GVHD at 1.5 year later. The 5yr OS was 95.2%.

Conclusions: The results showed 5yr OS for haplo-ID were comparable with other sources of stem cells despite the frequent viral reactivation. The good outcomes of T-depleted haploidentical allows efficient way of doing transplant in Singapore.

Disclosure: Nothing to declare.

18: Paediatric Issues

P668 AIEOP EXPERT BOARD RECOMMENDATIONS FOR MANAGEMENT OF POST-HCT HEMORRHAGIC CYSTITIS IN PEDIATRIC SETTING

Gianluca Dell’Orso 1, Simone Cesaro2, Marcello Carlucci1, Evelina Olcese1, Adriana Balduzzi3, Francesca Vendemini3, Massimo Catti4, Francesco Saglio4, Francesca Compagno5, Natalia Maximova6, Marco Rabusin6, Maria Cristina Menconi7, Katia Perruccio8, Elena Soncini9, Francesco Paolo Tambaro10, Veronica Tintori11, Daria Pagliara12, Maura Faraci1

1IRCCS- Istituto G. Gaslini, Genova, Italy, 2Azienda Ospedaliera Universitaria Integrata Verona, Verona, Italy, 3University of Milan-Bicocca - Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy, 4A.O.U. Città della Salute e della Scienza-Regina Margherita Children’s Hospital, Turin, Italy, 5Fondazione IRCCS Policlinico San Matteo, Pavia, Italy, 6Institute for Maternal and Child Health IRCCS “Burlo Garofolo”, Trieste, Italy, 7Pediatric Clinic of University of Pisa, Pisa, Italy, 8Santa Maria della Misericordia Hospital, Perugia, Italy, 9Spedali Civili di Brescia, Brescia, Italy, 10Azienda Ospedialiera di Rilievo Nazionale (AORN), Santobono-Pausilipon, Napoli, Italy, 11Meyer Children’s University Hospital, Florence, Italy, 12IRCCS Bambino Gesù Children’s Hospital, Rome, Italy

Background: Evidence about optimal management of hemorrhagic cystitis (HC) in pediatric hematopoietic cell transplantation (HCT) is limited, due to the lack of controlled trials and differences in management strategies reported in literature.

Methods: After a literature review focused on pediatric HSCT and an online survey, a panel of experts belonging to Associazione Italiana di Emato-Oncologia Pediatrica (AIEOP) HCT Working Group (WG) and Infectious Disease and Supportive Care WG held a meeting to define a consensus on the clinical management strategies.

The panel focused on the following main topics: (1) prevention of early-onset HC; (2) detection of BK polyomavirus (BKPyV) DNA by quantitative PCR (qPCR) in urine and plasma; (3) antiviral therapy; (4) supportive treatment; (5) urological management.

Results: In summary, for early-onset HC, high-dose cyclophosphamide requires hyperhydration with intravenous (IV) continuous infusion MESNA, with or without the addition of IV MESNA bolus. Diuretic management according to fluid balance is recommended, also according to specific patient- or transplant-related features. Urinary alkalinisation is not recommended. The prospective surveillance of BKPyV-DNA load on blood and urine by qPCR is not recommended in all patients due to the lack of pre-emptive strategies, but only in case of clinical suspicion for BKPyV-HC and in the clinical follow-up to determine the virological response to the treatment. Low-dose cidofovir (1mg/kg two or three times a week), is the preferred treatment due to safety profile, the reported efficacy and the possibility to avoid the co-administration of oral probenecid. Treatment is recommended until clinical and/or virological response (disappearance of macro-hematuria and at least 1 log10 decline BK viremia, respectively), at least for 4-6 doses, based on a patient-specific schedule. Reduction of immunosuppression is an option to consider very cautiously due to individual risk/benefit ratio of worsening graft-versus-host disease. Pain management is manly based on opiate derivatives that do not interfere with coagulation process. A higher platelet transfusion threshold (50 x 109/L) is recommended except for patients with platelet refractoriness, where a threshold of 20 x 109/L is acceptable. The use of tranexamic acid is not recommended for the risk of intra-bladder coagulation, while the use of factor VII and factor XIII is reported anecdotically as effective. The use of anti-spasmodic is not supported by data of efficacy. Hyperbaric oxygen therapy is an option for refractory or persistent HC, but with safety and logistical issues. It is recommended to share the clinical decisions with a consultant urologist since the first suspicion for HC, as well as performing bladder ultrasound to assess the presence of clots in bladder. For refractory HC with significant pain or urinary obstruction, a three-way catheter is recommended to perform a continuous bladder irrigation; an alternative option is a two-way catheter associated to a percutaneous cystostomy, according to patient’s anatomical features. Urological surgery, e.g. cystectomy, should be limited to life-threatening acute events.

Conclusions: In summary, strong evidence is lacking. The panel agreed the reported recommendations to define common strategies in preventions, diagnosis, and management of HC in pediatric HSCT setting.

Disclosure: None conflict of interest.

18: Paediatric Issues

P669 EXCELLENT SURVIVAL AND LONG TERM OUTCOMES IN INFANTS RECEIVING TOTAL BODY IRRADIATION FOR CONDITIONING PRIOR TO ALLOGENEIC TRANSPLANT

Hannah Lust1, Stephanie Powell1, Karina Danner Koptik1, Erin Kaseda1, Jennifer Schneiderman1, Sonali Chaudhury 1

1Ann & Robert H. Lurie Children’s Hospital, Chicago, United States

Background: The optimal approach to conditioning for allogeneic stem cell transplant (alloHCT) in infants <12mo of age remains controversial, particularly use of total body irradiation (TBI) which while most effective in disease cure, has significant long-term effects. We describe long-term survival and outcomes in patients with infant leukemia who received alloHCT with TBI-based conditioning.

Methods: We performed a retrospective review of 35 patients diagnosed <1 year of age with ALL or AML who underwent alloHCT at our institution between 1992-2022. Neurocognitive data was gathered from most recent neuropsychological testing reports, including measures of intellectual reasoning, processing speed, attention, and visual motor skills.

Results: Baseline characteristics can be found in table 1. Seventy two percent of patients underwent alloHCT in CR1. Most patients had ALL (n=30, 86%), while 5 patients (14%) had AML; 64% of patients were positive for KMT2A rearrangement. All received 12Gy TBI with 83% receiving VP-16/cyclophosphamide conditioning. Other conditioning chemotherapy regimens included thiotepa/cyclophosphamide (n=3, 8%) and busulfan/fludarabine n (n=3, 8%). Three patients received a craniospinal boost. With a median follow up of 16yrs, 5yr OS and RFS was 75% (95% CI 58-87%), and 64% (95% CI 51-82%) respectively. OS and RFS were not impacted by age at alloHCT or donor type, but OS significantly improved in the recent era). Nine deaths occurred with 14% TRM by day 100. Cumulative incidence of relapse after alloHCT was 17%. Two patients experienced VOD. Only 1 patient experienced severe (grade 3) aGVHD; no grade 4 aGVHD or extensive cGVHD was seen. In addition to organ toxicity monitoring, 13 survivors received comprehensive neuropsychological testing at our institution. Excluding 2 patients with complicating comorbidities (preterm birth, stroke), most patients had intact cognitive abilities with 90% demonstrating IQ in the broad average range. Significant deficits (<2 SD below mean) were limited poor visual motor skills in 2 patients. Mild deficits (1.5-2 SD below mean) were noted in 2 patients for processing speed and immediate attention; only one patient demonstrated mild deficits in IQ. Seven patients were diagnosed with non-hematologic secondary malignancies. Growth hormone replacement was used in 42% of survivors, while thyroid hormone replacement was required in 46% of survivors.

Sex, N (%)

M

20 (56)

F

15 (44)

Race/ethnicity, N (%)

White

18 (53)

African American/Black

4 (11)

Hispanic/Latino

13 (36)

Age at SCT, N (%)

<12mo

16 (46)

12-24mo

10 (28)

24-48mo

9 (25)

CNS disease at diagnosis or relapse, N (%)

Y

14 (40)

N

15 (43)

not available/unknown

6 (17)

Disease status at time of alloSCT*

CR1

25 (72)

CR2

9 (26)

Donor, N (%)^

MRD

12 (35)

MUD

11 (31)

MMUD

9 (25)

Haplo

1 (3)

Source, N (%)#

Bone marrow

9 (25)

PBSC

8 (22)

UCB

18 (50)

  1. *disease status unknown for 1 patient
  2. ^donor type unknown for 2 patients
  3. #graft source unknown for 1 patient

Conclusions: We describe excellent outcomes with low relapse rates for patients with infant leukemia receiving TBI-based conditioning alloHCT, with long-term OS approaching 90% in the recent era. The majority of patients demonstrated unimpaired neurocognitive abilities; noted deficits were largely supportable. These long-term outcomes with TBI-based conditioning are acceptable for a high-risk group of patients with significantly improved chances of survival.

Disclosure: Nothing to declare.

18: Paediatric Issues

P670 BCG-VACCINE COMPLICATIONS IN CHILDREN WITH INFANT ACUTE LYMPHOBLASTIC LEUKEMIA

Mikhail Lemeshev1, Alexandra Laberko 1, Larisa Shelikhova1, Veronika Fominykh1, Lili Khachatryan1, Galina Solopova1, Natalia Myakova1, Dmitry Balashov1

1Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology, and Immunology, Moscow, Russian Federation

Background: BCG infection (BCGitis) is a rare complication in infants receiving the Bacillus Calmette-Guérin (BCG) vaccine, which is used in newborns for tuberculosis prevention in highly endemic countries. BCG-vaccine is based on live-attenuated strain of Mycobacterium bovis, which is safe for healthy individuals, but can cause infection in those with impaired immunity – mostly, children with primary immunodeficiencies or acquired immunodeficiency syndrome. BCGitis in infants with acute leukemia is extremely rare complication with only single cases reported in the literature.

Methods: From 2018 to 2023, five cases of BCGitis in infants with acute lymphoblastic leukemia were observed in our center. All of them received BCG-vaccine within the first days of life. Median age at leukemia diagnosis was 1.5 (range 0.5-5.3) months, 4/5 cases had KTM2A rearrangements. Therapy protocols were ALL-baby-2021 in P2-P4, MLL-baby-2006 in P1 and ALL-MB-2015 in P5.

Two patients developed BCGitis after hematopoietic stem cell transplantation (HSCT). P4 received HSCT after 2 courses of chemotherapy (CHT) and blinatumomab and P5 had refractory disease and received 5 courses of CHT and blinatomumab. Conditioning regimens included: fludarabine 150mg/m2, treosulfan 42g/m2, etoposide 60mg/kg in P4 and fludarabine 150mg/m2, treosulfan 36g/m2, cyclofosfamide 50mg/kg in P5. Post-transplant immunosupression contained cyclosporine in both with combination of abatacept/vedolizumab in P5.

Results: BCGitis was observed in 5 patients: P1, P2, P3 during chemo/immunotherapy and P4, P5 after HSCT. P2-P5 had local infection of vaccination site area and P1 had regional disease with axillar lymph node involvement and abscess formation.

Median time from therapy start to BCGitis manifestation was 2.3 months: 1.1, 1.9, and 2.3 months in pre-HSCT cases and 5.1 and 6.5 months in those who had BCGitis post-HSCT (days +3 and +33 after HSCT).

P1 developed regional BCGitis after 2 courses of CHT with normal neutrophil counts and CD3 + 1.03x109/ml. P2 developed BCGitis on blinatumomab (16th day) following induction therapy with normal neutrophil counts and lymphocyte count 0,86 x109/ml. In P3 BCGitis development was associated with hematological recovery after induction therapy and CD3 + 2.02 x109/ml. P4 with BCGitis at day +3 after HSCT had cytopenia, P5 with BCGitis at day +33 had normal neutrophil recovery and 0.62x109/ml of CD3 + .

BCGitis was treated with combinations of amikacin, izoniazid, levofloxacin, ethambutol. In P2-P3 a combination of 2 and in P1, P4-5 a combination of 3 drugs were used, 4 patients also received local therapy with dimexide and rifampicin and P1 also had surgical abscess removement. P1 and P2 with pre-HSCT BCGitis who underwent HSCT and both patients with post-HSCT BCGitis received antimycobacterial therapy up to immune recovery. None of those with BCGitis pre-HSCT and received prophylactic antimycobacterial therapy developed BCGitis after HSCT.

Conclusions: BCGitis in children with infant leukemia is a rare. The cause may be in incomplete clearence of life Mbt.bovis due to young age, followed by intensive immunoablation. Moreover, in some patients (likely P3 and P5) BCGitis may be potentiated by hematologic/immune recovery. There are no guidelines for BCGitis treatment in infants with hematological malignancy, but combination of 2-3 antimycobactrial agents leads to good response.

Disclosure: Nothing to declare.

18: Paediatric Issues

P671 WORKFORCE CONSENSUS RECOMMENDATIONS FOR PAEDIATRIC HSCT CENTRES ON BEHALF OF THE PAEDIATRIC SUB-COMMITTEE OF THE BRITISH SOCIETY OF BLOOD, BONE MARROW TRANSPLANTATION CELLULAR THERAPY (BSBMTCT)

Reem Elfeky 1, Valerie Broderick2, Anna- Marie Ewins3, Elizabeth Rivers1, Wing Roberts4, Mary Slatter4, Kanchan Rao1, Beki James5, Brenda Gibson3

1Great Ormond Street (GOS) Hospital for Children NHS Foundation Trust, University College London GOS Institute of Child Health, and NIHR GOSH BRC, London, United Kingdom, 2Children’s Health Ireland at Crumlin, Dublin, Ireland, United Kingdom, 3The Royal Hospital for Children, Glasgow, United Kingdom, 4Great North Children’s Hospital, Newcastle Upon Tyne, United Kingdom, 5Regional Centre for Paediatric Haematology Leeds Children’s Hospital, Leeds, United Kingdom

Background: Hematopoietic Stem Cell Transplant (HSCT) programmes require to be staffed by a well-trained, experienced and adequately resourced multidisciplinary team. HSCT programmes should be resourced to a level which provides flexibility for development as indications expand and new therapeutic approaches move from experimental to standard practice. The Paediatric BSBMTCT group established a working sub-group to produce recommendations for the workforce requirements for a paediatric stem cell transplant centre (TC) in the UK and Ireland. The workforce included representation from haematological and immunological backgrounds and TCs of different sizes and specialty interest with geographical representation within the UK and Ireland. The agreed remit of the group was to write consensus recommendations for the medical, nursing, data management, HSCT coordination, quality management, pharmacy and allied health professional workforce requirements for the provision of a safe and effective paediatric HSCT service.

Methods: Five steps were taken to establish the recommendations. Firstly, a PubMed/Medline literature search was carried out for publications discussing workforce requirements for a safe and efficient HSCT centre using key words “workforce for “BMT” / “bone marrow transplant”/ “haematopoietic stem cell transplant”/” HSCT, “staff for haematopoietic stem cell transplant”, “staff for BMT”, “staff for bone marrow transplant”, “nursing ratios for haematopoietic stem cell transplant”. Secondly, the working group met fortnightly for 12 months to i) understand and consider staffing within the centres represented and to agree if these were appropriate and safe ii) to drew-up recommendations which were presented at a BSBMTCT meeting with all TCs present. Thirdly, draft recommendations were disseminated to i) establish if TCs agreed with the recommendations in concept or if these required modification / change? ii) establish if TCs meet the recommendations. Fourthly, the workforce group met to discuss proposed changes and revised recommendations following the national BSBMTCT audit meeting when all TCs were present for their input to achieve consensus recommendations. Fifthly, the final version of the recommendations was drafted.

Results: Relevant literature is sparse. Six publications were identified mainly describing physician/patient ratios with one publication on other staff/patient ratios (including co-ordinator, pharmacist, psychosocial) and a further additional publication for social worker benchmarking.

Table 1: Workforce consensus recommendations

Consultant staffing

1. There should be a minimum of four senior doctors experienced in transplant in each centre able to participate in a 24h on-call rota.

2. Consultants with other responsibilities manage approximately 5 HSCT patients per annum. Dedicated HSCT consultants need to manage 10 HSCT per annum.

3. There should be a weekly ward round of all members of the HSCT team.

4. Every TC must have a programme director that is a consultant with a minimum of 2-years’ experience in looking after paediatric HSCT patients.

Middle grade staffing

1. There should be one non-rotating middle grade medical staff member providing continuity for HSCT patients.

2. A combination of clinical experience (minimum 12 months) in addition to taking the EBMT exam or equivalent is a desired outcome for middle grade trainees to demonstrate competency in managing HSCT patients.

3. A consultant should round at least twice weekly with the middle grade medical staff member.

4. A consultant/non-rotating middle grade doctor should be available to review day care patients/clinic patients.

Nursing staff

1. There should be one clinical nurse specialist (CNS) for every 10 HSCT patients per annum.

2. Every unit should invest in transplant specific training of HSCT nurses.

3. There should always be a majority of nurses on any shift who are experienced in paediatric HSCT (including administration of IV chemotherapy, infusion of stem cells) and its complications.

4. There should be at least 1 nurse for every 2 HSCT inpatients.

5. Consideration should be given to establishing advanced nurse specialist (ANP) posts which offer continuity of care and support the middle grade medical team.

Data manager (DM)

1. The DM hours should follow the BSBMT recommendation of 27-hrs /week +2.9 hrs for every 10 patients per year.

2. The DM should be Band 5 or higher.

3. The DM should be given the opportunity of training and attending courses.

HSCT co-ordinator

1. The role of HSCT coordinator should be provided by either administrative staff with significant clinical input, or by CNS/ANP staff with significant administrative support.

Quality manager (QM)

1. There should be at least one QM.

2. The QM should be band 5 or greater.

3. Regardless of the background, QM should be offered training.

Pharmacist

1. There should be one dedicated WTE pharmacist for every 20 new HSCT patients per annum.

2. There should be a specialist pharmacist within and out with normal working hours.

Social worker

1. There should be a dedicated WTE social worker for every 25 new HSCT patients per year.

2. There should be access to a social worker for every new HSCT patient as part of the transplant preparation pathway.

Psychologist

1. HSCT patients should be able to access psychology support pre and peri-transplant when needed and ideally within 2 weeks. They may also have post-transplant needs and all patients should also have access to psychology support within a reasonable timeframe.

Dietician

1. Dedicated dietetic support to allow a daily review of inpatients and attendance at the HSCT clinic.

Physiotherapist

1. Physiotherapy staffing should allow pre transplant assessment, twice weekly assessment of inpatients and attendance at HSCT clinics for post-HSCT assessment.

Occupational therapist and speech & language therapist

1. Dedicated support should be available from each department to allow a regular review of inpatients with needs and attendance at the HSCT clinic.

Play therapist

1. There should be provision for input from play therapy or a teenage activity coordinator for every patient daily during the working week.

Additional services which may be charity or trust funded

1. Music therapy.

2. Complementary therapy.

3. Family support workers.

Conclusions: Workforce consensus recommendations have been made. These will require funding. It is important that there is standardization across all HSCT centres in UK and Ireland and that these standards meet minimum staffing requirements and equity of care across centres for safe and effective management of paediatric recipients of HSCT. Next steps will include presenting these recommendations to national and commissioning committees followed by publication in a peer reviewed journal.

Disclosure: Authors confirm no conflict of interest.

18: Paediatric Issues

P672 HAPLOIDENTICAL HEMATOPOIETIC STEM CELL TRANSPLANTATION WITH A CONTROLLED NUMBER OF CD3 + CELLS AND POST-TRANSPLANT CYCLOPHOSPHAMIDE: EXPERIENCE FROM A SINGLE PEDIATRIC CENTER

Giulia Albrici1, Giulia Baresi1, Stefano Rossi1, Elena Soncini1, Elisa Bertoni1, Marta Comini2, Federica Bolda2, Alessandra Beghin2, Arnalda Lanfranchi2, Fulvio Porta1, Marianna Maffeis 1

1Pediatric Oncohematology Unit and Pediatric Bone Marrow Transplant Center, Children Hospital Brescia, Brescia, Italy, 2Stem Cell Laboratory, Clinical Chemistry Laboratory, ASST Spedali Civili, Brescia, Italy

Background: A haploidentical donor can be identified in almost all pediatric patients allowing the transplant procedure to be performed quickly. GVHD and graft failure are major concerns in haploidentical hematopoietic stem cell transplantation (HSCT). The use of post-transplant cyclophosphamide (PT-Cy) for GVHD prophylaxis is well established in haploidentical HSCT using T-replete grafts. The higher risk and severity of GVHD have limited the use of peripheral blood stem cells (PBSC) in this setting. We previously reported the use of a manipulation technique based on CD34+ positive selection and subsequent addition of a controlled number of T lymphocytes (30x10^ 6/Kg BW recipient) in the MUD setting using PBSC grafts. We observed rapid engraftment and a low incidence of both acute and chronic GVHD. We here describe the same manipulation procedure in combination with PT-Cy in the haploidentical HSCT setting.

Methods: We retrospectively collected data on haploidentical HSCT with PBSC-derived CD34+ selection and controlled CD3+ add-back performed at our center since 2019. PT-Cy was administered on days +3 and +5 in combination with serotherapy (ATG or alemtuzumab) and post-transplant immunosuppression with tacrolimus and MMF. Both, malignant and non-malignant diseases were included. According to EBMT guidelines, different conditioning regimens (myeloablative and reduced intensity) were administered based on the underlying disease.

Results: Characteristics of the 13 patients included are shown in Table 1. HSCT was performed for malignant (6/13) and non-malignant diseases (7/13). Median age at HSCT was 8.5 y (range 0.5-21.9). The median follow-up was 9.9 months (range 2.7-43.3). The median number of CD34+ and CD3+ cells infused was 13.63 x106/Kg BW and 29.95 x106/Kg BW, respectively. Ten patients received MAC while 3/13 received RIC. The median neutrophil engraftment was achieved at day +16 (range 13-28); the median platelet engraftment at day +23 (range 17-59). No primary graft failure was observed; only one patient with MDS developed a secondary graft failure due to persistent CMV viremia requiring a second HSCT. The last available CD3+ chimerism showed complete donor chimerism in all patients alive without relapse. Five patients (38.5%) developed GVHD. Only one patient had grade III acute GVDH involving the skin; another patient developed high grade late-acute/chronic GVHD involving the skin and GI. Both were SCID-patients and were treated successfully with extracorporeal photoapheresis. Viral reactivation was observed in 6 patients, but clinical infection was reported in only 3 of them and resolved with antiviral treatment. Data on immune reconstitution are presented in Figure 1. Two oncologic patients died from disease progression and one patient with CID died from multi-organ failure due to COVID-19 after HSCT. No patients died from GVHD, graft complications or invasive infections.

Full size table
The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (87)

Immune reconstitution in the cohort at day+100 and day+180.

Conclusions: Our data suggest that haploidentical transplantation using CD34+ selection combined with a defined add-back of CD3+ (30×106 kg/BW recipient), PT-Cy and serotherapy as GvHD prophylaxis is feasible and safe in pediatric recipients. Good engraftment, low rates of GvHD and infectious complications were observed. Small sample size and short follow-up are the main limitations of our analysis and further studies are needed.

Disclosure: Nothing to declare.

18: Paediatric Issues

P673 PLASMA AMINO ACIDS AND FECAL CALPROTECTIN USAGE FOR THE PREDICTION OF INTESTINAL INJURY AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION IN CHILDREN

Igne Kairiene 1, Jelena Rascon1, Ramune Vaisnore1

1Vilnius University, Vilnius, Lithuania

Background: Preparative regimen-induced intestinal damage (ID) triggers a systemic inflammatory response and GvHD that may result in multiorgan damage and fatal outcomes. Several mechanisms, often overlapping, such as disruption of mucosal and microbiome integrity, infection and inflammatory response contribute to ID development. Thus, its prediction is crucial to identify patients who need prompt interventions. We aimed to evaluate a biomarker panel potentially available for daily use for ID prediction.

Methods: Sixteen consecutive children who underwent an allogeneic HSCT from an HLA-identical sibling or MUD were enrolled into the study. Blood amino acids (AA), C-reactive protein (CRP), absolute neutrophil count (ANC) and fecal calprotectin (fCLP) were evaluated longitudinally during an early post-transplant period. Samples were collected before conditioning, on the graft infusion day and post-transplant days +7, +10, +14, +21, +28. The ID was evaluated at the same time points using CTAC criteria version 5.

Results: 13 of 16 (81%) patients developed ID grade I-III. The mean level of taurine, urea, Α–aminobutyric acid, ornithine was higher whereas levels of asparagine, glutamic acid, Α-aminoadipic acid, citrulline were lower in the patients with ID. Median fCLP and CRP were higher in the group with ID than without it (160.11 (259.91) vs 77.44 (137.29), p=0.041; 47.55 (89.59) vs 8.80 (34,94), p = 0.005 respectively). Median ANC was lower when ID developed (0.00 (0.40) vs 0.61 (1.84), p=0.009). Lasso penalized regression model has identified the most significant biomarkers for ID prediction (citrulline, aspartate, urea, taurine, ethanolamine, histidine, alanine).

Conclusions: The concentration of fCLP increases while the levels of citrulline decrease in patients with ID. These biomarkers can be used interchangeably as they are negatively associated with each other. Citrulline, urea, aspartate, histidine, and alanine could be valuable as a predictive test before developing ID.

Clinical Trial Registry: -.

Disclosure: Nothing to declare.

18: Paediatric Issues

P674 MULTIPLE MINOR PHLEBOTOMIES IN EPP INVOLVING THE LIVER: IS THERE A WAY TO AVOID NEEDING A LIVER AND HAEMATOPOIETIC STEM CELL TRANSPLANT?

Rita Beier 1, Jasmin Barman-Aksözen2, Franziska van Breemen2, Hagen Ott3, Katharina Becker1, Annika Ewert1, Ulrich Baumann1, Eva Pfister1, Martin Sauer1, Elisabeth Minder2, Britta Maecker-Kolhoff1, Anna-Elisabeth Minder2

1Hannover Medical School, Hannover, Germany, 2Swiss Reference Center for Porphyrias, Institut für Labormedizin, Stadtspital Zurich Triemli, Zürich, Switzerland, 3Kinderkrankenhaus Auf der Bult, Hannover, Germany

Background: In 5% of patients with Erythropoietic protoporphyria (EPP) the accumulation of the toxic heme precursor protoporphyrin IX (PPIX) leads to liver failure, requiring life-saving combined liver and sequential hematopoietic stem cell transplantation (HSCT).

Methods: We are taking care of a German family with two children who are affected by EPP. The siblings developed signs of liver involvement due to EPP at the age of 10.

The older daughter received a cadaveric liver transplant at the age of 11 years after being diagnosed with hepatic fibrosis at the age of 10 years. 6 months later she underwent allogeneic HSCT from a MUD (10/10, Flu/Treo/Mel, alpha/beta depleted graft). After the HSCT, she experienced multiple complications (GvHD III (skin, intestine), VOD, secondary vWJS, ITP, ADV infection, cerebral ADV infection) and subsequently died 9 months after the transplant.

The younger brother was diagnosed with liver involvement, but no liver fibrosis, by biopsy at the age of 10. Erythroid haem biosynthesis is regulated by iron availability. Reduced iron availability through multiple minor phlebotomies (MMPs) has successfully reduced PPIX synthesis and reversed liver damage in adults with EPP. After written informed consent and ethical review was obtained, monthly phlebotomies of 50 ml venous blood were started from April 2022. Laboratory parameters to monitor haematological status, liver disease and porphyrin metabolism were quantified from the residual phlebotomy material and urine samples. In addition, liver ultrasound and elastography were performed, physical examinations were performed, and the patient was interviewed about his well-being and subjective treatment effects.

Results: Erythrocyte PPIX rapidly decreased from initially 37.1 µmol/L (reference: <0.2) to currently 12.2 (November 2023). Also, urinary heptacarboxyporphyrin, a marker for liver damage in EPP, decreased from 1.3 to 0.8 µmol/mol creatinine (reference: <0.8). The elastography and normalized transaminases supported reduction of liver damage. Hemoglobin remained within the age and gender adjusted reference range. Concomitantly, light sensitivity, a disease characteristic of EPP caused by the phototoxic properties of PPIX, and quality of life of the patient improved. On the basis of the current figures, it is highly unlikely that the patient’s liver function will worsen to the point where a liver transplant and a sequential HSCT is needed.

Conclusions: MMPs were well tolerated and seemed to have reversed the toxic effect of the heme precursors. Given the family history this might prevent liver transplantation and HSCT. Light tolerance and quality of life are positively affected by this novel intervention.

Disclosure: Rita Beier: travel grants Neovii, Medac, research grants Medac.

Britta Maecker-Kolhoff: travel grants Medac.

All other authors: no COI.

18: Paediatric Issues

P675 COMPARISON OF BUSULFAN-BASED AND TREOSULFAN-BASED HIGH-DOSE CHEMOTHERAPY AND AUTOLOGOUS STEM CELL TRANSPLANTATION IN HIGH-RISK NEUROBLASTOMA

Kyung-Nam Koh 1, Young Kwon Koh2, Aejin Kang1, Ji Young Kim1, Su Hyun Yun1, Sung Han Kang1, Hyery Kim1, Ho Joon Im1

1Asan Medical Center Children’s Hospital, Seoul, Korea, Republic of, 2Chosun University, Gwangju, Korea, Republic of

Background: Although the combination of tandem high-dose chemotherapy and autologous stem cell transplantation (HDCT/ASCT) has improved survival outcomes in high-risk neuroblastoma patients, it has also presented notable toxicity challenges. The purpose of this study was to evaluate and compare the effectiveness and side effects of busulfan and treosulfan in high-dose chemotherapy used for HDCT/ASCT in patients with high-risk neuroblastoma.

Methods: A cohort of 32 patients diagnosed with high-risk neuroblastoma was identified at Asan Medical Center Children’s Hospital between October 2013 and February 2022. The treatment protocol included 8 to 10 cycles of induction chemotherapy and tandem HDCT/ASCT combined with 131I-MIBG therapy. For the first round of HDCT/ASCT, 20 patients received a busulfan/melphalan regimen between 2014 and 2019, and 12 patients received a treosulfan/melphalan regimen between 2020 and the present. The second HDCT regimen consisted of thiotepa and cyclophosphamide.

Results: The 5-year overall survival (OS) and event-free survival (EFS) rates were 76.4% and 57.0%, respectively. In the initial high-dose chemotherapy and autologous stem cell transplantation (HDCT/ASCT) phase, patients in the busulfan group had a median hospital stay of 25 days (ranging from 21 to 42 days), compared to 23 days (ranging from 21 to 26 days) in the treosulfan group. Neutrophil engraftment occurred around day 10 in both groups, with a range of 8-12 days in the busulfan group and 9-11 days in the treosulfan group. Mucositis was a common side effect, affecting 19 out of 20 patients in the busulfan group and 9 out of 12 in the treosulfan group. Neutropenic fever was similarly prevalent, occurring in 55% of the busulfan group and 58% of the treosulfan group. Notably, pulmonary hypertension was reported in 4 patients (20%) receiving busulfan, but not in any patients receiving treosulfan. Additionally, chronic kidney disease developed in 3 busulfan patients (15%) and hepatic veno-occlusive disease in 2 (10%), whereas none of these complications were observed in the treosulfan group.

Conclusions: This preliminary study suggests that treosulfan may offer a better toxicity profile than busulfan in the initial HDCT/ASCT for high-risk neuroblastoma, with fewer severe side effects observed. However, due to the small patient cohort, further research is needed to confirm these findings and establish treosulfan’s long-term safety and efficacy in this treatment context.

Disclosure: I have no conflict of interest to disclose.

18: Paediatric Issues

P676 USE OF BLINATUMOMAB TO ACHIEVE REMISSION AND CONSOLIDATION WITH HAPLOIDENTICAL TRANSPLANT WITH CYCLOPHOSPHAMIDE FOR THE TREATMENT OF CHILDREN WITH REFRACTORY ACUTE LYMPHOBLASTIC LEUKEMIA

Alberto Olaya Vargas 1, Haydee Salazar-Rosales2, Martin Peréz-Gárcia3, Yadira Melchor-Vidal4, Annecy Herver-Olivares5, Gerardo López- Hernández2, Nideshda Ramírez-Ortiz2, Cesar Galván-Diaz2, Alberto Olaya-Nieto2

1Instituto Nacional de Pediatria/Centro Medico ABC, México, Mexico, 2Instituto Nacional de Pediatria, México, Mexico, 3Hospital Infantil de Oncología Hito, Queretaro, Mexico, 4Centro Médico ABC, México, Mexico, 5Centro Medico Nacional La Raza. IMSS, México, Mexico

Background: ALL is the most common childhood cancer. This type of leukemia is highly sensitive with 80-90% remission rate and long-term disease-free survival, consequently with a high possibility of cure rate. However, those patients that relapse and become refractory to conventional chemotherapy treatment have only around 30-40% possibilities to achieve long term remission. In Mexico, it is estimated that the event-free survival in the first remission is not greater than 65%, so the treatment of children with ALL and in high-risk relapse is a problem due to morbidity and mortality related to the use of High doses of chemotherapy and poor long-term survival.

Methods: A quasi exprimental, Interventional, non-Randomized with single group assignment study was conducted in children of 0-18 years of age with a Lansky score >80 and refractory o high risk relapse ALL preB-CD19 +. As a strategy to achieve remission was used blinatumomab a dose 5 μg/m2 for continous infusion of 48 hours during 7 days, increasing the dose to 15 μg/m2 during 28 days, all patients received triple intrathecal therapy with methotrexate, arabinocide and hydrocortisone, two weeks after each cycle of blinatumomab, All patients received blinatumumab cycles hospitalized, during their stay the toxicity data related to blinatumumab was evaluated every day. The patients had a MRD of < 0.002 Log after 2 cycles, received an bone marrow transplant as consolidation therapy.

Results: A total of 52 patients were included, 32 men (60%) and 20 women (40%), with an average age of 9.2 years (range 3-16), 86% of the patients were in relapse and 14% with refractory ALL. 84% patients achieved complete remission after 2 cycles of blinatumomab, 70% of patients who achieved remission had a response with the first cycle of blinatumumab and 30% achieved complete remission with the 2nd cycle of blinatumumab, 70% of patients who achieved remission received a transplant, 30% continued with chemotherapy. The overall survival for the entire group at 2 years was 48.5%, the main factor that influenced survival was obtaining remission with blinatumumab, which obtained a survival rate of 66%, the patients who underwent transplantation after remission achieved a survival of 83%, 70% of the patients received a haploidentical transplant with post-cyclophosphamide use with a survival of 60%.

Conclusions: The use of 2 cycles of blinatumumab is an efficient and safe strategy to achieve remission in children with relapsed/refractory ALL + 19, consolidation with transplant is an efficient therapy, haploidentical transplant is an easily accessible and efficient strategy to consolidate to this group of patients in middle-income countries.

Disclosure: The authors declare that they have no conflict of interest.

18: Paediatric Issues

P677 HEMORRHAGIC CYSTITIS IN CHILDREN AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: INCIDENCE, TREATMENT, OUTCOME, AND RISK FACTORS

Nur Ayca Celik 1, Talia Ileri2, Elif Ince2, Hasan Fatih Cakmaklı2, Berk Burgu1, Zeynep Ceren Karahan1, Seda Kaynak Sahap1, Ebru Dumlupınar1, Mehmet Ertem2

1Ankara University Faculty of Medicine, Ankara, Turkey, 2Ankara University Faculty of Medicine, Division of Pediatric Hematology, Ankara, Turkey

Background: Hemorrhagic cystitis(HC) is a common complication after allogeneic hematopoietic stem cell transplantation(HSCT) characterized by irritative symptoms of the urinary tract and a higher morbidity and mortality rate.

Methods: This study aims to evaluate the incidence, etiology, risk factors, clinical management, and outcomes of hemorrhagic cystitis(HC) in 293 children with 308 allo-HSCT(MSD:249, MUD:43, haplo:16) in our pediatric transplantation unit between Dec 1997-Jan 2023.

Results: Ten recipients(10/308, 3.2%) developed early-onset HC median 0 days(-6-4) after HSCT. All patients used cyclophosphamide 9 patients for the conditioning regimen and 1 patient for GVHD prophylaxis.

Fifty-three recipients(53/308, 17.2%) developed late-onset HC 3-72 days(median, 27 days) after HSCT and macroscopic hematuria lasted 1-90 days(median, 11 days).

Older recipient and donor age(p<0.001 and p=0.036, respectively), haplo HSCT(OR=4.91, p=0.003), unrelated donor(OR=2.44, p=0.021), acute GVHD(OR=2.61, p=0.002) and CMV reactivation in first 100 days(OR=3.78, p=<0.001) were associated with higher risks of development of HC uni-multivariate analyses.

BK viruria and viremia were demonstrated in 32/41(78%) and 12/37(32%) patients respectively who could be evaluated. Notably, all patients with BK viremia also had BK viruria. Other viruses were rarely associated with HC. Median urine and blood BK load were 9x109 and 2322 copies/ml, respectively. Although BK viremia/viruria was associated with HC grade(p<0.05) viral load was not associated with higher risks of HC grade.

Late-onset patients were divided into two groups severe HC group(Grade 3/4 HC, 15/7, 41.6%) and mild HC group(Grade 1/2 HC, 5/26, 58.4%). Older donor age, haploidentical HSCT, PTCY for GVHD prophylaxis, and CMV reactivation in the first 100 days were associated with severe HC(p=<0.05). This group had longer hematuria duration(8 vs 26 days) and longer hospitalization(56 vs 78 days)(P=<0.05)(Table 1). Urinary system ultrasonography was carried out in 20/22 severe and 18/31 mild HC patients. Bladder mucosa thickening was observed in 84.2% of cases, with a mean thickness of 8.52 mm. Diffuse thickening was more frequent than local thickening(25/32) and these findings were not associated with grade. Upper urinary tract dilatation was more frequent in severe HC(55%) than in mild HC(0%)(p= 0.001).

Mild HC cases mostly improved with hydration. In severe HC patients, bladder irrigation was performed to 17, intravesical hyaluronic acid in 14, intravesical “ankaferd blood stopper” in 2, intravesical cidofovir in 1, intravenous cidofovir in 2, and cystoscopic intervention in 4 patients. Improvement in the clinics of patients receiving intravesical hyaluronic acid was 1(0-56) day.

Patients with HC had longer hospitalization than non-HC patients(p=<0.001). The first 100 days TRM and overall death were observed more frequently in late-onset HC patients(OR=2.23, p=0.196 and OR=2.034, p=0.043, respectively). The overall survival rate was 175.3 months whose developed HC, it was 243.8 months in non-HC patients(p=0.021). The 2-year survival rate was 72.9% vs 88% compared to patients without HC. Two children died in the early post-transplant period without resolution of HC, but HC was not the direct cause of death in any patient.

Table 1. Main demographic and clinical characteristics of the late-onset HC patients

Late-onset HC – (n=255)

Late-onset HC + (n=53)

p-value

Age (years)

Median (Range

9.1 (0.3-21)

13.3 (1.8-18.3)

<0.001

Gender

M vs F

140 vs 115

34 vs 19

0.279

Donor age (years)

Median (Range)

13 (0-55)

21 (0-50)

0.036

Type of donor

MRD vs MUD

MRD vs Haplo

215 vs 31 215 vs 9

34 vs 12

34 vs 7

0.021 0.003

Source of SC

BM vs PBSC or CB

174 vs 81

31 vs 22

0.274

ATG

Yes vs No

135 vs 120

23 vs 30

0.894

Prophylaxis of GVHD

CSA vs CSA + MTX

CSA vs PTCY + CSA + MMF

61 vs 184

61 vs 10

12 vs 32

12 vs 9

0.666

0.001

Acute GVHD

Yes vs No

58 vs 197

23 vs 30

0.002

Acute GVHD grade

Mild vs Severe

44 vs 15

16 vs 7

0.855

CMV reactivation

Yes vs No

48 vs 189

25 vs 26

<0.001

PMN recovery (days)

Median (Range)

16 (2-56)

16 (9-36)

0.585

PLT recovery (days)

Median (Range)

24 (9-112)

24 (10-81)

0.364

Hospital stay (days)

Median (Range)

42 (11-208)

61.5 (28-230)

<0.001

TRM

Yes vs No

9 vs 246

4 vs 49

0.196

OS (Cl)

%88

%72.9

0.021

Conclusions: HC is one of the most important complications of HSCT that affects OS significantly. Hydration, bladder irrigation, and intravesical hyaluronic-acid applications are effective and safe approaches in severe HC group.

Clinical Trial Registry: Clinical Trial registry number: 2022000359-1.

http://etikkurul.medicine.ankara.edu.tr.

Disclosure: Nothing to declare.

18: Paediatric Issues

P678 TRANSPLANT-ASSOCIATED THROMBOTIC MICROANGIOPATHY (TA-TMA) IN CHILDREN TREATED WITH ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION (ALLO-HSCT): ONE CENTER STUDY

Tomasz Jarmoliński 1, Monika Rosa1, Iwona Bil-Lula1, Joanna Korlaga1, Krzysztof Kałwak1, Marek Ussowicz1

1Wroclaw Medical University, Wroclaw, Poland

Background: TA-TMA is a severe complication of HSCT, diagnosed in 16% of pediatric patients in the US. It can lead to multi-organ failure and death. Complement activation is a pathogenetic cornerstone of TA-TMA, as evidenced by increased C5b-9 blood concentration. The study aimed to estimate the frequency of TA-TMA and evaluate the usefulness of measurement of C5b-9 plasma concentration in diagnosing TA-TMA in children treated with allo-HSCT.

Methods: 132 children treated with allo-HSCT between January 2021 and December 2023, were prospectively screened during 100 days after HSCT for defined by Jodele clinical and laboratory TA-TMA criteria. The group consisted of 85 boys and 47 girls aged 0.,3-18yr, 66 with non-malignant and 66 with malignant disease. Blood samples for C5b-9 were collected before conditioning (day -7) and on days 0 (transplantation), and +7, +14, +21, +28, +56, +100 post-allo-HSCT. After centrifugation, plasma was collected and frozen at -80oC. Plasma C5b-9 concentration was measured using the EIA method (MicroVue SC5b-9 Plus Enzyme Immunoassay, Quidel; upper limit of normal range 244 ng/ml).

Results: TMA was diagnosed in 7 patients (5%), including one symptomatic before conditioning. The C5b-9 concentration assay was performed in 100 children. The patient symptomatic before conditioning presented with very high (2180 and 1431ng/ml) C5b-9 plasma concentrations on days -7 and 0, respectively, that decreased to 271 ng/ml after eculizumab administration. He died on day +15 due to TMA and sepsis. Post-HSCT TA-TMA was diagnosed in 6 patients, 5 of whom had elevated C5b-9 levels in the range from 271 to 777ng/ml. Moreover, 41 patients without clinical criteria of TMA had elevated C5b-9 concentrations, usually on day +7, after 1 week since cyclosporine A start. Nine patients died, among them 5 with TMA, 1 with elevated C5b-9 without any other symptoms of TMA, and 3 due to early post-transplant complications without TMA.

Conclusions: The incidence of TA-TMA in the Polish pediatric allo-HSCT population is lower than previously reported in the US nonetheless it seems to be an important and underreported cause of mortality. A high C5b-9 plasma concentration correlates with the clinical course of TA-TMA. This preliminary study highlighted the diagnostic value of C5b-9 measurement emphasizing the need for regular monitoring and prompt report turnaround time.

Disclosure: The authors declare no potential conflicts of interest.

This work was supported by The Saving Kids With Cancer Foundation (research grant FNRD.C200.21.001).

18: Paediatric Issues

P679 DISTANCE MAKES THE CELLS GROW FONDER- PAEDIATRIC ALLOGENIC TRANSPLANT AND CELLULAR THERAPY REFERRAL PROGRAM, THE SOUTH AUSTRALIAN EXPERIENCE

Matthew O’Connor 1,2, Catherine Mitchell1, Laura Chapman2, Karen McCleary2, Adam Nelson2, Richard Mitchell2

1Women’s and Children’s Hospital, Adelaide, Australia, 2Sydney Children’s Hospital, Sydney, Australia

Background: The Women’s and Children’s Hospital in Adelaide, South Australia is the only paediatric oncology centre servicing the centre of Australia- from Darwin in Northern Territory down to Adelaide in South Australia, it services a population of two million people spread over 2.4 million square kilometers (the size of the UK ten times over). 7-10 children per year require cellular therapy or allogenic transplantation from this population and this service is not currently performed in our centre. Thus a robust referral pathway and working relationship has been developed with our main transplant referral centre (Kids Cancer Centre, Sydney Children’s Hospital). This is a model that may be applicable to other non-transplant centres around the world.

Methods: Patients complete all pre transplantation investigations and tissue typing in our centre. Fortnightly teleconferences are held with the primary referral centre to discuss upcoming referrals, current inpatients and children likely to return for ongoing management. Written standardised handovers are completed from medical, nursing, allied health (including child life therapy) and social work for each patient and family. This included a standardised referral form to capture the required information. Commercial flights interstate and significant accommodation costs are met by combination of hospital and charity funding. Consent for transplant is performed by the treating centre initially through telehealth then consolidated in person. Patients and families travel for transplantation just prior to commencement, remain for inpatient and initial outpatient management and then return to our centre around day 100 post transplant for ongoing management. Combined telehealth appointments in the early return period with both local and interstate teams present ensure ongoing handover and thorough assessment of post-HSCT complications.

Results: 39 patients over the last five years have been referred, 37 for allogenic transplantation and two for CAR-T therapy with 27 to our main referral centre and 10 across four other centres. They have undergone transplantation (or product infusion) at referral centres and survivors have returned for long term post transplant management. Families report feeling comforted by knowing the referral pathways are sound and proven.

Donor

MSD

Haplo

MUD

MMUD

11

4

20

2

Product

BM

PBSC

Cord

CAR-T

15

11

11

2

Disease

Malignant

Non-Malignant

26

13

Conclusions: The thorough communication approaches described have enabled safe and effective referral of allogenic transplantation candidates to external centres and subsequent acceptance back for ongoing care when clinically appropriate. Consumer feedback is currently being sought to further improve the patient journey.

Disclosure: Nothing to declare.

18: Paediatric Issues

P680 INCREASING TIM-3 + T CELLS IN PERIPHERAL BLOOD OF PEDIATRIC PATIENTS WITH RELAPSE B-ALL AFTER HAPLOIDENTICAL ALLO-HSCT

Liubov Tsvetkova 1, Olga Epifanovskaya1, Elena Babenko1, Olesya Paina1, Polina Kozhokar’1, Zhemal Rakhmanova1, Olesya Yudinceva1, Anna Osipova1, Sabina Ryabenko1, Elena Dobrovolskaya1, Ivan Moiseev1, Alexandr Kulagin1, Ludmila Zubarovskaya1

1RM Gorbacheva Research Institute, Pavlov University, Saint-Petersburg, Russian Federation

Background: Expression of Immune checkpoints receptors such as cytotoxic T lymphocyte antigen 4 (CTLA-4), programmed death-1 (PD-1), Lymphocyte-activation gene 3 (LAG-3), T cell immunoglobulin mucin-domain-containing-3 (TIM-3) plays an important role in the immune escape of solid tumors and part of hematological malignancies. Prognostic value of TIM-3 expression was demonstrated on bone marrow CD4 + T cells at initial diagnosis for risk relapse of pediatric B-precursor acute lymphoblastic leukemia (B-ALL) [Franziska Blaeschke et al.]. However, role of TIM-3 expression at relapse after allo-HSCT is not unclear in adult and pediatric B-ALL. Aim of this study was determination of expression PD-1, CTLA-4, TIM-3, LAG-3 in peripheral blood in pediatric B-ALL patients with remission and active disease after allo-HSCT.

Methods: Twenty-five children with B-ALL underwent haploidentical allo-HSCT and were enrolled in this prospective study. Median age was 9,4 y.o. (0.89-16.5) at allo-HSCT. Complete remission of disease has been keeping in 11 (44%) patients. The median follow-up after allo-HSCT in this group was 9 months (3-14 months). Relapse of leukemia (n=12) was developed in 12 patients (48%): 7 had bone marrow relapse, 1 had extramedullary disease (testicular), 4 – combined relapse. Persistence MRD was observed in 2 patients. The median time to relapse/MRD detection was 213 days after allo-HSCT (D + 25 - D + 1099). In remission group fresh peripheral blood was collected with median of 42 days after allo-HSCT (D + 30-D + 200). The median time of material collection was 152 days after allo-HSCT (D + 70-D + 1101) in relapsed group, p=0.01. Samples were stained with fluorochrome-conjugated antibodies to PD-1, CTLA-4, TIM-3, LAG-3 by 8-color flow cytometry. We compared the absolute values of expression of checkpoint molecules on lymphocytes, monocytes and CD3 + T-cells.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (88)

Results: Our results revealed that TIM-3 expression significantly increases in patients with relapse/MRD of B-ALL after haplo-HSCT on lymphocytes and T-cells of peripheral blood in comparison with patients in remission of leukemia, p=0.0005 and p=0.025. Whereas we did not observe the differences of expression of other checkpoint molecules (PD-1, CTLA-4, TIM3, LAG3). In addition two patients with extramedullary and combined relapse elevated of TIM-3 absolute value at 3 months before developing of relapse in contrast with remission group.

Conclusions: Our data suggests, that TIM-3 overexpression on lymphocytes and T-cells in peripheral blood correlated with relapse after haplo-HSCT. It may identifite exhaustion subsets of T cells in pediatric B-ALL patients and considered as potential biomarker and target for future immunotherapy with TIM-3 inhibitors. More clinical studies are required for understanding prognostic role of TIM-3 in B-ALL after allo-HSCT.

Disclosure: The study was funded by a grant from Russian Science Foundation № 22-15-00491.

18: Paediatric Issues

P681 IMMUNE-MEDIATED CYTOPENIA AFTER PEDIATRIC HEMATOPOIETIC CELL TRANSPLANTATION – A RETROSPECTIVE ANALYSIS

Mária Füssiová 1, Júlia Horáková1, Ivana Boďová1, Jaroslava Adamčáková1, Dominika Dóczyová1, Miroslava Pozdechová1, Tomáš Sýkora1, Peter Švec1, Alexandra Kolenová1

1National Institute of Children’s Diseases, Bratislava, Slovakia

Background: Immune-mediated cytopenia (IMC) – isolated or combined hemolytic anemia, thrombocytopenia, or neutropenia – with an unclear pathophysiology is a rare, but potentially serious complication of hematopoietic cell transplantation (HCT). Diagnosis of IMC is challenging and requires urgent intensification of immunosuppressive therapy due to posttransplant immune dysregulation. We provide a comprehensive single-center analysis of IMC characteristics and their impact on HCT outcomes.

Methods: The retrospective analysis evaluates the incidence, risk profile, time of onset, response to the treatment, mortality and late effects of IMC in children treated in the HCT unit in Bratislava, Slovakia between 2013 and 2022.

Results: A total of 218 pediatric patients who underwent 243 HCT procedures were included in the study (161 allogeneic and 82 autologous transplantations). Six patients (2.4% of transplantations) with a median age of 6.6 years (range, 2-16) developed post-HCT IMC, all after allogeneic HCT (6/161). Common pretransplant predictors associated with immune cytopenia in our cohort were present as follows: allogeneic HCT and serotherapy (6/6), age less than 10 years and female gender (5/6), non-malignant disease (3/6), AB0 mismatch (2/6), peripheral blood stem cell as a graft source (2/6), pre-HCT autoimmunity presented by hypergammaglobulinemia (1/6). We documented several post-HCT risk factors for IMC: infections and lymphopenia <800 µL (4/6), increase of autologous chimerism (3/6) and GvHD (2/6). Three patients (50%) were diagnosed with warm autoimmune hemolytic anemia (AIHA), in 2 patients (33.3%) immune thrombocytopenia (ITP) was confirmed, and one patient (16.6%) developed Evans syndrome (AIHA, ITP). In the analyzed cohort, the two most common IMC presentations were isolated anemia and thrombocytopenia (83.3%). We identified neither trilineage IMC nor neutropenia. AIHA and ITP in 4/6 children (66.6%) had an acute or subacute course of cytopenia. A prolonged disease course was observed in the patient with combined IMC. The median time from HCT to IMC onset was 102.8 days post-HCT (range, 47-195). Therapies used for IMC treatment included corticosteroids (n=5), intravenous immunoglobulins (n=4), rituximab (n=4), mycophenolate mofetil (n=2), sirolimus (n=1), and daratumumab (n=1). In addition, 4 children had concomitant cyclosporine prophylaxis. Overall, an average of 3 unique agents (range, 1-6) were used for the resolution of IMC, achieved at a median of 49.6 days (range, 13-94) after diagnosis. Five of 6 patients recovered completely (83.3%). One child was a non-responder and died due to very severe intracranial hemorrhage (co-incidence of refractory ITP and SARS-CoV-2 infection). The mortality associated with IMC was 16.7% (1/6). One patient died later in a complete remission of IMC due to fatal peritransplant complications. Lastly, we investigated the late effects and consequences of IMC-directed therapies. Prolonged hypogammaglobulinemia induced by rituximab was noted in 50% of patients. Other side effects in the cohort were non-significant.

Full size table

.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (89)

Conclusions: The incidence of post-HCT IMC in our pediatric analysis is comparable to published data (2.4 vs 1.5-10%). Morbidity and mortality from IMC post-HCT have been reduced by early diagnosis and aggressive interventions involving multiple agents targeting antibody production – however, they remain significant.

Disclosure: Nothing to declare.

18: Paediatric Issues

P682 SEQUENTIAL KIDNEY-STEM CELL TRANSPLANTATION IN PATIENT WITH SCHIMKE IMMUNO-OSSEOUS DYSPLASIA: CASE REPORT

Alexandra Shutova1, Alexandr Bazaev1, Yulia Skvortsova1, Alexandr L. Rumyantsev2, Diana Khalikova2, Galina Novichkova1, Michael Maschan 1, Dmitry Balashov1, Alexey Maschan1

1Dmitry Rogachev National Medical Research Center of Pediatric Hematology, Oncology and Immunology, Moscow, Russian Federation, 2Russian Children Clinical Hospital, Moscow, Russian Federation

Background: Schimke Immuno-osseous Dysplasia (SIOD) is extremely rare disease with T-cell immunodeficiency, renal failure, spondyloepiphyseal dysplasia and short stature. SIOD patients who underwent hematopoietic stem-cell transplantation (HSCT) have high rates of post-HSCT mortality in due to severe toxicity and associated complications. We report the case of a boy with SIOD received sequential kidney-stem cell transplantation from the same haploidentical donor in due to achieve immunotolerance to kidney transplant and avoid lifelong immunosuppression therapy (IST).

Methods: The patient was a 11-years old SIOD boy with kidney failure. Disease presents at the age of 5 with glucocorticoid-resistant nephrotic syndrome. SIOD was diagnosed at the age of 6 by identification homozygous SMARCAL-1 gene mutation. Kidney transplantation from haploidentical donor (mother) was performed at the age of 11 in due to terminal kidney failure. Sequential HSCT from the same haploidentical donor was performed 23 days after kidney transplantation. The conditioning regimen consisted of treosulfan 42 g/m2, fludarabine 150 mg/m2, melphalan 140 mg/m2, rabbit ATG 5 mg/kg and rituximab 375 mg/m2. For GVHD prophylaxis patient received PTCY at dose 50 mg/kg/day at +3 + 4 days and ruxolitinib from +5 day (then switched to sirolimus). Patient had a number of severe transplant-related complications including acute GVHD grade 4 (intestinal/skin with epidermolysis bullosa), capillary leak syndrome, delirium, Coombs-positive hemolysis, CMV infection, bacterial sepsis. As GVHD treatment were used methylprednisolone, etanercept, vedolizumab, abatacept, budesonide. Neutrophil engraftment occurred on +28 day after HSCT. At +30, +60 day after HSCT patient had complete donor chimerism. However, trilineage cytopenia which probably had immune origin with increasing mixed chimerism (maximum 10,5% recipient cells, CD34 49% recipient cells, CD3 52,5% recipient cells) occurred by the +100 day after HSCT, therefore the CD34 selected boost-therapy from the same haploidentical donor with TLI 4 Gy and rituximab 375 mg/m2 as lymphodepletion were performed. For GVHD prophylaxis methylprednisolone, abatacept and MMF were used. Neutrophil and platelet engraftment occurred on +18 and +9 day respectively. Transplant-related complications included acute transient ischemic attacks. At present time ( + 165 day after HSCT/ + 45 day after boost) patient has complete donor chimerism and maintained normal kidney function.

Results: Patient received sequential kidney-stem cell transplantation as a curative option. Post-transplant period was burdened by severe immune, infectious and toxic complications. At present time patient has maintained normal kidney and hematopoiesis function with complete donor chimerism. As IST MMF, abatacept and methylprednisolone 1 mg/kg/day with gradual withdrawal by +190 day after HSCT/ + 70 day after boost are used.

Conclusions: Sequential kidney and HSCT from the same donor have emerged as a promising therapeutic option, potentially offering better quality of life with normal kidney an immune function. Moreover, the good outcome of HSCT is the reason for the development of immune tolerance to kidney graft and an argument for the full withdrawal of IST. However, the optimal approach remains unclear considering limited data and potential transplant-related challenges in SIOD patients.

Disclosure: Nothing to declare.

18: Paediatric Issues

P683 CYCLOSPORINE PLUS METHOTREXATE VERSUS CYCLOSPORINE ALONE FOR GRAFT-VERSUS-HOST DISEASE PROPHYLAXIS IN PEDIATRIC PATIENTS UNDERGOING HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM HLA-IDENTICAL SIBLING

Vincenzo Apolito 1, Valeria Ceolin1, Manuela Spadea1,2, Marta Barone1, Marco Basiricò1, Francesco Saglio1, Franca Fagioli1,2

1Regina Margherita Children’s Hospital, Turin, Italy, 2University of Turin, Turin, Italy

Background: A survey conducted on behalf of the European Society for Blood and Marrow Transplantation (EBMT) in 75 pediatric stem cell transplantation (SCT) centers found that graft-versus-host disease (GVHD) prophylaxis in children undergoing matched sibling donor (MSD) SCT is heterogeneous, as 47% of centers used cyclosporine (CSA) alone and 45% CSA plus a short-term course of methotrexate (CSA/MTX) (Lawitschka et al, Transpl Int 2020). Weiss et al (J Cancer Res Clin Oncol 2015) reported higher incidence of relapse and shorter overall survival (OS) in CSA/MTX vs CSA alone patients, together with similar rates of acute and chronic GvHD. However, patients were not comparable because those in CSA/MTX had been treated much earlier than those in CSA group (1984-1997 vs 1998-2008, respectively). We report on a homogeneous cohort of children who underwent MSD in our institution, comparing patients who received CSA alone versus who received CSA/MTX.

Methods: Data of patients who underwent MSD SCT at Regina Margherita Children’s Hospital (Turin, Italy) between 2013 and 2023 were retrospectively collected. In our institution, the decision to add MTX to CSA was made considering type of disease, comorbidities, age and stem cell source (SCS), after discussion in an interdisciplinary board.

Results: We collected 49 patients (Table1). 21 (42.9%) patients received CSA and 28 CSA/MTX (57.1%). Median age of the whole cohort was 10.9 years (0.5-21.9). Younger patients (<12 years old) received more frequently CSA than CSA/MTX (P=0.0013). Thirty-nine patients (79.6%) had malignancies, without differences between CSA and CSA/MTX (P=0.48). SCS was bone marrow in 43 patients (87.5%) and peripheral blood was used in 6 patients (12.5%), all in CSA/MTX group (P=0.03). Nine patients (18.4%) received anti-thymocite globulin, all in CSA/MTX group (P=0.0063). Median follow-up was 4.5 years (0.1-10.1). Cumulative incidence (CI) of acute GvHD at 100 days was lower in CSA/MTX vs CSA (7.1% [95% confidence interval (CI) 1.9%-27.1%] vs 38.1% [95%CI 22.1%-65.7%], P=0.006, respectively) (Figure1). Also in multivariate analysis the use of CSA/MTX led to lower incidence of aGvHD grade II/IV (HR 0.1, 95%CI 0.01-0.95, P=0.04), while other factors did not show any statistical significant impact. CI of moderate/severe chronic GvHD at 1 and 4 years was 7.6% (95%CI 1.99%-28.68%) and 11.9% (95%CI 4.09%-34.5%) in CSA/MTX vs 16.0% (95%CI 5.62%-45.83%) and 22.3% (95%CI 9.30%-53.49%) in CSA (P=0.38), respectively. Only use of bone marrow as SCS was associated with lower incidence of cGvHD by multivariate analysis (HR 0.02, 95%CI 0.002-0.243, P=0.001). 5-year OS was 80.7% (95%CI 62.97%-98.44%) in CSA/MTX vs 83.3% (95%CI 65.96%-100.00%) in CSA alone (P=0.56). In patients with malignancy, CI of relapse at 1 and 4 years was 15.83% (95%CI 5.6%-44.7%) and 33.3% (95%CI 17.2%-64.3%) in CSA/MTX vs 12.2% (95%CI 3.3%-44.9%) and 21.36% (95%CI 7.6%-60.1%) in CSA (P=0.27), respectively.

CSA/MTX (n.)

CSA alone (n.)

P value

Number of patients

28

21

Sex

Male

21

12

Female

7

9

Median age, y (range)

13.7 (4.1-21.9)

7.6 (0.5-14.9)

Age

0.0013

Younger than 12 years

11

18

Older than 12 years

17

3

Disease type

0.48

Malignant

21

18

Non malignant

7

3

Source of SC

0.03

Peripheral blood

6

Bone marrow

22

21

Use of ATG

0.0063

Yes

9

No

19

21

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (90)

Conclusions: Despite being limited by its retrospective nature, our study showed that the addition of MTX to CSA for GvHD prophylaxis in pediatric patients undergoing MSD SCT reduced incidence of acute GVHD grade II/IV without affecting CI of relapse and OS in a homogeneous population and its use should be implemented in pediatric setting.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

18: Paediatric Issues

P684 NUTRITIONAL DYNAMICS IN PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANTATION: A RETROSPECTIVE MONOCENTRIC STUDY ASSESSING BMI PERCENTILES AND CLINICAL OUTCOMES

Timo Stetter1, Michaela Döring 1, Christian Seitz1, Nora Rieflin1, Thomas Baumgarten1, Léa Thérond1, Pia Glogowski1, Johannes Schulte1, Rupert Handgretinger1, Peter Lang1, Karin M. Cabanillas Stanchi1

1University Children’s Hospital Tübingen, Tübingen, Germany

Background: Children and adolescents undergoing hematopoietic stem cell transplantation (HSCT) face an elevated risk of malnutrition due to increased endocrine activity, a catabolic metabolic state, and inflammatory processes. The relationship between nutritional status prior to pediatric HSCT and overall outcome is not yet well understood.

Methods: In this monocentric, retrospective study, pediatric HSCT patients were screened for nutritional status as reflected by age-adjusted BMI-percentile category and observed transplant-related adverse events on the day of in-patient admission before HSCT, day +30 and day +200 after HSCT. Exclusion criteria comprised gastrointestinal malformations, permanent enteral/parenteral nutrition, and chronic inflammatory bowel disease before HSCT.

Results: In this retrospective study, data from 365 pediatric patients (median age: 9 years; 56% female patients) were examined. On admission, 54% of patients were at normal weight (10th - 75th BMI percentile), 22% were underweight (<10th percentile), and 23% were overweight (>75th percentile). In the median, weight fell by at least 1 BMI category by day +200 after HSCT in 20% of patients, and by 2 or more categories in 22%. In 28% of cases the category remained the same, in 27% of cases the patients increased by one or two categories compared to baseline BMI. There was no difference in the timing of leukocyte and granulocyte engraftment, or the occurrence of a graft reaction between the percentile groups (p>0.05). VOD occurred in 21 patients in the entire cohort, of which 7 occurred within the >75th percentile group (p=0.0413). Similarly, there was a higher incidence of higher grade GvHD (°III-°IV) in this group (p=0.0028). Bacterial infections with mainly Enterobacter cloacae (6.3% of patients), Enterococcus faecium (6.9%), Pseudomonas aeruginosa (5.8%), Klebsiella pneumoniae (4.7%), Citrobacter (4.2%), Staphylococcus aureus (2.4%) and Clostridium difficile (1.6%) were detected in the stool. A focus in one percentile group could not be determined. Viral infections were detected in the stool with mainly adenoviruses (14.5% of patients), rotaviruses (2.9%) and noroviruses (2.1%). Here, there was also no relevant association with the BMI percentile. In the long-term follow-up up to 10 years after HSCT, 115 patients (32%) died. In the alloHSCT group, 35 deaths (12% of alloHSCT patients) fell under TRM. Here, of the 35 deceased, 12 (34% of deceased, 4% of total alloHSCT patients) were ascribed to the >75th-97th percentile group; the median proportion of deceased patients was 9% (1% of alloHSCT group) in the other percentile groups. In the autoHSCT group, a total of 23 patients (41%) died; of these, all were due to progression or recurrence of the underlying disease.

Conclusions: Contrary to expectations, complications and TRM were statistically clustered in the 75th-97th BMI percentiles, highlighting the need for closer monitoring of overweight pediatric HSCT patients. While acknowledging that this study focused on body weight and length, not specific nutritional status, ongoing prospective surveys at the study center aim to address this gap.

Disclosure: All authors declare, that they have no conflicts of interests.

18: Paediatric Issues

P685 INTEGRATING CAR T-CELL THERAPY AND ALLOGENEIC STEM CELL TRANSPLANTATION IN CHILDREN WITH HIGH RISK ACUTE LYMPHOBLASTIC LEUKEMIA. THE EXPERIENCE OF A SINGLE CENTER

Blanca Molina 1, Marta Gonzalez Vicent1, Sara Vinagre1, Blanca Herrero1, David Diaz1, Miguel Angel Diaz1

1Hospital Infantil Universitario Niño Jesus, Madrid, Spain

Background: Treatment with CAR T-cell achieves excellent complete remission rates in children and young adults with relapsed or refractory B cell acute lymphoblastic leukemia (B-ALL), but only half maintain long-term remission. Consolidative hematopoietic stem cell transplantation (HSCT) appears to be a potential strategy to reduce relapse risk in these patients. In the same way relapse following HSCT remains the major cause of transplant failure in B-ALL patients so rescue treatment with CAR-T therapy could be a curative option for some of theme.

We retrospectively analyzed long-term follow-up data of B-ALL patients who relapsed post-transplant and received CAR-T therapy and patients who received CAR-T therapy before a consolidative HSCT in a single center between 2016 and 2023.

Methods: A total of 21 patients (10 male, 11 female) with high-risk B ALL were included with a median age of 9 years (range, 3-20). Twelve of them received CAR-T cell therapy as rescue treatment for relapse after HSCT (median time to relapse 12 months; range 3-27) and 9 received CAR-T cell therapy before HSCT. In 3 of these 9 patients a consolidative HSCT was performed, 2 relapsed after CAR-T and received a HSCT as a rescue treatment and 4 underwent a HSCT due to loss of B cell aplasia. Two patients received HSCT, CAR-T and a second HSCT because of relapse after CAR-T. All patients received 4-1BB-based CAR T-cells. Three patients have TP53 alterations at diagnosis. The median time from CAR-T and HSCT was 142 days (range 30-283).

Results: As complications of CAR-T 16/21 patients presented CRS (7 grade 1, 4 grade 2, 2 grade 3 and 3 grade 4) and 4/21 presented ICANS (2 grade 4). As main complications of HSCT post CAR-T 3/12 patients had endothelial damage, 6/12 developed acute GVHD (only 1 patient grade 3-4) and 2 patients had chronic GVHD. With a median follow-up of 38 months (range 8-94) 14/21 patients are alive with a DFS of 64±11%. MRT of CAR-T therapy was 5±3%. MRT of HSCT was 0%. In the univariate analysis, DFS of the patients who received a CAR-T after a HSCT was 73±13% and 50±9% of those patients who received CAR-T previous to HSCT (p=0,1). Having CRS was related to worse DFS (52±7% vs 100%, p=0,07). All the patients who had p53 mutation died (DFS 0% vs 75±5%, p=0,003). Patients who developed acute GVHD had better outcome (DFS 80±9% vs 0%, p:0,031). In the multivariate analysis the only factor who influenced in DFS was TP53 alterations (TP53 alterations vs No TP53 alterations, HR 8; 95% CI, 1.6 to 41,2; p=0,013).

Conclusions:

  • Combination of CAR-T therapy and HSCT in pediatric patients with high-risk ALL could be a curative option for majority of them.

  • Treatment of children with acute lymphoblastic leukemia and TP53 alterations remains a great challenge.

.

Disclosure: Nothing to declare.

18: Paediatric Issues

P686 ASSESSMENT OF THE ENDOTHELIAL GLYCOCALYX BY VIDEO MICROSCOPY IN CHILDREN WITH ACUTE LEUKEMIA AFTER ALLOGENEIC STEM CELL TRANSPLANTATION

Zarina Khaerova1, Zhemal Rakhmanova1, Olesya Paina1, Timur Vlasov2, Elena Semenova1, Ivan Moiseev 1, Alexander Kulagin1, Ludmila Zubarovskaya1

1RM Gorbacheva Research Institute, Pavlov University, Saint Petersburg, Russian Federation, 2Pavlov University, Saint Petersburg, Russian Federation

Background: Endothelial dysfunction is one of the most common complications after allogeneic hematopoietic stem cell transplantation (allo-HSCT). Among of factors contributing to the development of microvascular dysfunction are impairment of the structure and function of the endothelial glycocalyx that acts as an interface between extracellular matrix and cellular membrane for cell-cell communications. The aim. To evaluate endothelial glycocalyx dysfunction by video microscopy in children with acute leukemia after allo-HSCT.

Methods: Data of 18 children (median age of 13 y.o. (8 – 17 y.o.) who underwent allo-HSCT was analyzed. Diagnosis of ALL have 11 pts. (61%), AML – 7 pts. (39%). Allo-HSCT in remission was performed in 17 pts. (92,8%), 1 pts. (8,2%) had an active disease. Myeloablative conditioning received 11 pts. (61%), reduced intensity conditioning – 7 pts. (39%). Allo-HSCTfrom haploidentical related donor was performed in 17 pts. (92,8%), matched related donor - 1children (8,2%). All participants underwent sublingual videomicroscopy by sidestream darkfield imaging (GlycoCheck version 5.2; Microvascular Health Solutions Inc). A study was conducted to quantify vessel density, perfused border region (PBR, an inverse variable of endothelial glycocalyx size), microvscular health score (MVHS), red blood cell (RBC) filling and blood flow in sublingual microvessels ranging from 5 to 25 μm in diameter. The study was conducted on day 0, +30, +60 after allo-HSCT. Ten healthy children volunteers served as control group (median age of 11 y.o. (7 – 17 y.o.).

Results: The general characteristics of the study population are summarized in Table 1. Children on day 0 of allo-HSCT have more impaired microcirculation with higher PBR high flow compared to the control group (1,54 [1,08-3,28] vs. 1,2 [0,98-1,7]; p = 0,09). These groups have a significant difference in the values MVHS (3,31 [0,85-5,26] vs. 4,2 [2,8 -4,7]; p = 0,02) and vessel density (336,5 [233-664] vs. 423 [335-523]; p = 0,03). There was a trend towards worse PBR 5-9 on day +30 after allo-HSCT compared to day 0 (1,15 [0,94- 1,24] vs. 1,08 [0,88-1,15]; p =0,065) and PBR 5-25 is significantly better on day + 60 after allо- HSCT compared to day 0 (1,74 [1,48; 2,2] vs. 2,17 [1,59;2,73] p =0,04). Patients with ALL have a significant difference in PBR 20-25 (2,78 [1,79-3,74] vs. 2,99 [2,86 - 4,0]; p = 0,05), MVHS values (3,74 [1,4-5,26] vs. 2,73 [0,85-4.69]; p = 0,04) and vascular density (368 [249-664] vs. 283 [233-371] (p = 0,05) compared with children with AML. There was a trend of worse PBR 5-9 in children with serious infectious complications after allo- HSCT (1,1 [0,94-1,2] vs. 1,04 [0,8-1,1]; p = 0,07).

Table 1. General characteristics of the study population

Full size table

.

Conclusions: Children with acute leukemia after allо-HSCT have glycocalyx injury, as confirmed by PBR, vessel density and MVHS compared to those in healthy controls. Stronger damage of microvessels 5-9 μm in diameter on day 30 after HSCT compared to day 0 may be explained by alloreactive endothelial injury. Improved PBR 5-25 by day +60 may confirm that endothelial recovery is happening within this timeframe. Further studies are needed to assess the prognostic value of glycocalyx injury and endothelial dysfunction for the development of vascular complications.

Disclosure: Nothing to declare.

18: Paediatric Issues

P687 INTRODUCTION OF BODY IMPEDENCE ANAYSIS TO MEASURE BODY COMPOSITION CHANGES AND FLUID STATUS OF PAEDIATRIC PATIENTS UNDERGOING HAEMATOPOIETIC STEM CELL TRANSPLANTATION

Rebecca Fisher 1, Alice Brewer1, Belinda Meares1, Sheridan Collins1, Sarah Slack1, Melissa Gabriel1

1The Children’s Hospital at Westmead, Westmead, Australia

Background: Traditional anthropometric measurements (weight, height and body mass index) provide information of limited value in paediatric patients undergoing Haematopoietic Stem Cell Transplant (HSCT). Measuring changes in body composition and fluid status can be clinically useful in informing nutrition and fluid management particularly in the early post HSCT period. DXA is the gold standard for measuring body composition. However, it is expensive, non-portable, emits radiation and requires technical expertise. Bioelectrical impedance analysis (BIA) has commonly been used to assess the body composition of children and adolescents. Bioelectrical impedance analysis (BIA) is a non-invasive, affordable, quick, and tested method to accurately assess body composition and fluid status in clinical practice (Marra et al., 2019).

Methods: The required criteria for a BIA device included suitability for use in children, cost, validation data, ability to measure patients in a supine position, portable, quick to measure and functionality to measure both body composition and fluid status. A QuadScan Body Stat 4000 was chosen using these criteria.

User guidelines and competencies were developed to train staff to use the devise to ensure data was collected safely and reliably.

HSCT patients who were able to lie prone for 10 minutes and able to be weighed prior to testing were deemed appropriate for BIA.

Measurements are performed weekly for inpatients and monthly for outpatients up to 1 year post HSCT, unless more frequent measurements are clinically indicated.

Results: From July 2023 to Mid-December 2023, 14 HSCT patients have had BIA measurements with 84 tests being performed. Results were recorded in the medical records. Results are reviewed weekly by the HSCT team and used to inform clinical decisions. BIA results have been used in conjunction with other clinical information to determine patient’s dosing weight used for medications, parenteral nutrition and diuretic use.

Barriers to using BIA in this paediatric population identified so far include younger patients are often unable to lie still in prone position for the required duration of the measurements and therefore often not suitable to be measured, the logistics required to measure all patients at required time points can be difficult to manage, and the lack of reference ranges available in children.

Conclusions: BIA as part of routine surveillance of patients provides additional information can be used to inform clinical decisions and treatment options. Ongoing monitoring of body composition helps to tailor clinical care including nutrition and fluid management, improving health outcomes. Individual patient’s dosing weights can be better calculated using the BIA data as part of the clinical information. There is currently no validated data in this population for BIA use and therefore individual’s results are compared with their baseline results rather than reference ranges.

Disclosure: Nothing to declare.

18: Paediatric Issues

P688 TRANSPLANTATION OUTCOME OF HIGH RISK ACUTE MYELOID LEUKEMIA AND MYELODYSPLASTIC SYNDROME IN CHILDREN: SINGLE CENTER EXPERIENCE IN KOREA

Young Tak Lim 1, Eu Jeen Yang1, Jun Young Oh1

1Pusan National University Children’s Hospital, Yangsan, Korea, Republic of

Background: In acute myeloid leukemia (AML), primary refractory or relapsed disease, and leukemia with unfavorable genetics are considered high risk AML (hrAML), with allogeneic hematopoietic stem cell transplantation (HSCT) representing the standard treatment. Allogeneic HSCT is also only treatment option in high risk myelodysplastic syndrome(MDS), including refractory anemia with excess blasts (RAEB).

Methods: From January 2011 to December 2022, 37 children (up to age 18 years) with high risk AML and MDS who received allogeneic hematopoietic stem cell transplantation at the Department of Pediatrics, Pusan National University Children’s Hospital were reviewed retrospectively. We analyzed treatment outcomes and cumulative incidence of relapse from allogeneic HSCT according to donor types.

Results: There were 31 hrAML and 6 RAEB patients. The hrAML group was comprised of 7 with second remission after relapse, 7 with complex or unfavorable chromosomal abnormalities, 4 with primary refractory, 3 with delayed response to induction therapy, 2 infants, 2 AML-M7, 2 FLT3-ITD and 4 others who were eligible to be transplanted. Twelve patients had HLA-identical sibling donors (ISD), 14 patients had unrelated matched donors (UMD), 9 patients had haplo-identical donors (HID) and 2 had umbilical cord blood donors. The median age at transplantation of patients with hsAML and MDS was 9.8 years (range, 1.1-17.9 years) with a median follow up of 59 months (range, 2-149). The cumulative rates of grade II-IV acute graft versus host disease (GVHD) were 16.7% in the ISD group, 28.6% in the UMD group and 55.6% in the HID group (p=0.063), with cumulative rates of chronic GVHD at 25.0%, 15.4% and 22.2%, respectively (p=0.897). The estimated 5-yr event-free survival rate (EFS) and overall survival (OS) of the entire cohort were 66.1±8.0% and 75.4±7.1%, respectively. According to donor types, estimated 5-yr EFS and OS were 83.3±10.8% / 83.3±10.8% after ISD, 55.1±13.9% / 64.3±12.8% after UMD, 64.8±16.5% / 77.8±13.9% after IHD (p=0.279 / p=0.382). The 5-year cumulative incidences of relapse in patients who received an allogeneic HSCT was 26.8±7.8% without significant differences between donor types (p=0.157).

Conclusions: Allogeneic HSCT including unrelated matched and haploidentical donor transplantation achieved remarkable results in childhood hrAML and MDS, indepenent of donor type.

Disclosure: I have no discposure of potential conflict.

18: Paediatric Issues

P689 BUILDING A PEDIATRIC HEMATOPOIETIC STEM CELL TRANSPLANT UNIT IN PARAGUAY: LESSON LEARNT

Marta Verna 1, Maria Liz Benitez2, Attilio Maria Rovelli1, Jabibi Noguera2, Marta Canesi1, Valentino Conter1, Susy Thiel Figueredo3, Andrea Biondi1,4

1IRCCS San Gerardo dei Tintori, Monza, Italy, 2HGP Niños de Acosta Ñu, Asuncion, Paraguay, 3Instituto de Investigaciones en Ciencias de la Salud UNA, Asuncion, Paraguay, 4School of Medicine and Surgery, University of Milano Bicocca, Milano, Italy

Background: A Hematopoietic Stem Cell Transplant Unit (HSCTU) can be established at a reduced cost in Middle Income Countries (MICs). Aim of the project was to establish a pediatric HSCTU in Hospital Niños de Acosta Ñu (Asunción, Paraguay) after the successful similar project done in Iraqi Kurdistan.

Methods: We analyzed all consecutive patients transplanted since the beginning of the activity from October 2019 to September 2022 and which variables and challenges in the applied methodology were pivotal to the outcome of the project.

Results: A total number of 14 patients were analyzed [TABLE 1]. Median total nucleated cells (TNC) and CD34+ infused with bone marrow (BM) as source were 3x108/Kg and 3.45x106/Kg; with peripheral blood (PB) as source 2x108/Kg and 7x106/Kg, respectively. All patients engrafted with median time for neutrophil count > 0.5x109/L and platelets > 20x109/L of 22 and 23 days in the BM group; 19 and 25 days in the PB one. 5 patients developed grade II-IV acute GVHD (aGVHD) at a median time of 21 days after stem cell infusion; all recovered except for 3 with gut aGVHD stage 4, who developed multiorgan failure. One patient developed mild chronic GVHD (cGVHD). Only 1 severe infectious complication was observed (pneumonia). Nine patients are alive and disease free at a median follow-up of 14.5 months; 1 patient died of disease progression and 4 of transplant related complications.

The startup methodology was based on specific steps: first exploratory mission, 5 days educational course, creation of a protocol handbook including all Standard Operating Procedures (SOPs), definition of a tree diagram (Jacie model), training outside the country and then on the job from the beginning of the clinical activity.

TABLE 1. PATIENT, DISEASE AND TRANSPLANT CHARACTERISTICS

Patients

Number

14

Age at transplant

Mean (yrs)

11.5

Type of disease (1)

AML

3 (1 in CR1, 2 in CR2) (6)

ALL

7 (all in CR2)

AAS

3

SCD

1

Type of donor (2)

MSD

7

Haploidentical donor

7

Source of stem cells (3)

BM

10

PBSC

4

ABO compatibility

Compatible

9

Major incompatibility

4

Minor incompatibility

1

CMV status

R + /D+

11

R-/D+

1

R + /D-

2

Conditioning regimens (4)

Bu-Flu

1

Bu-Flu-Mel (+/- Cy post)

2

Bu-Flu-TT (+/- Cy post)

7

Bu-rATG

3

Bu-Cy-rATG

1

GVHD prophylaxis (5)

MTX 3 dosis + CSA

4

MTX 4 dosis + CSA + MMF + Cy post

7

CSA + rATG

4

  1. Legenda:
  2. (1) AML acute myeloid leukemia, ALL acute lymphoblastic leukemia, SAA severe aplastic anemia, SCD sickle cell disease
  3. (2) MSD matched sibling donor
  4. (3) BM bone marrow, PBSC peripheral blood stem cell
  5. (4) Bu busulfan, Flu fludarabine, Mel melphalan, TT tiotepa, rATG rabbit anti thymocyte globulin
  6. (5) MTX methotrexate, CSA cyclosporin, MMF mycophenolate mofetil
  7. (6) CR complete remission

Conclusions: Clinical activity started in October 2019 but the program was stopped for a long time due to the SARS-CoV-2 pandemic. The present interim analysis revealed no major problems in the harvest procedure, PBSC apheresis procedure, engraftment of stem cells, management of blood products, infection control. We noted that of the 4 non-leukemic deaths, 3 occurred during grade 4 aGVHD; we should highlight that the hospital started with a precocious approach to haploidentical HSCT, after only 3 matched sibling donor (MSD) transplants, due to the low percentage of siblings in paraguayan families and the impossibility to have access to the international donor bank.

The transplant program in Paraguay was our second project, after the experience in Iraqi Kurdistan; the two settings were deeply different, both in socio-cultural aspects as well as in the HSCT program focus: whereas the Iraqi Kurdistan one was looking at thalassemia patients, the project in Paraguay mainly focused on oncological diseases; anyway the same methodology, time schedule and checklists were applied. The capacity building and twinning program approach have been demonstrated to be replicable and efficacious in different settings.

Disclosure: Nothing to declare.

30: Solid Tumours

P690 HIGH-DOSE CHEMOTHERAPY WITH STEM CELL TRANSPLANTATION IN ADULT PRIMARY MEDIASTINAL GERM CELL TUMORS. A RETROSPECTIVE STUDY OF THE EBMT, CELLULAR THERAPY & IMMUNOBIOLOGY WORKING PARTY

Paolo Pedrazzoli1, Manuela Badoglio2, Myriam Labopin2, Giovanni Rosti1, Matthieu Delaye3, Carsten Bokemeyer4, Christoph Seidel4, Jane Apperley5, Elisabetta Metafuni6, Juergen Finke7, Jean-Henri Bourhis8, Christos Kosmas9, Florent Malard10, Jurgen Kuball11, Christian Chabannon12, Annalisa Ruggeri13, Simona Secondino 1

1Fondazione IRCCS Policlinico San Matteo, Pavia, Italy, 2EBMT Study Office Paris, Paris, France, 3Institut Curie, Paris, France, 4University Medical Center Hamburg-Eppendorf, Hamburg, Germany, 5Imperial College London, London, United Kingdom, 6Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy, 7Albert-Ludwigs University Medical Center, Freiburg, Germany, 8Institut Gustave Roussy, Villejuif, France, 9“Metaxa” Memorial Cancer Hospital, Piraeus, Greece, 10Sorbonne Université, Centre de Recherche Saint-Antoine INSERM, Paris, France, 11University Medical Centre Utrecht, Utrecht, Netherlands, 12Aix-Marseille université, Institut Paoli-Calmettes, Marseille, France, 13IRCCS San Raffaele Hospital, Milan, Italy

Background: Primary mediastinal germ cell tumors (PMGCTs) account for 1%-3% of all germ cell tumors, 80% being non seminoma (NS). NS PMGCTs have a poorer prognosis compared to their gonadal counterparts and, according to the International Germ Cell Cancer Collaborative Group, they are considered by definition a “poor risk” disease. Medical treatment is the same of the gonadal counterpart, overall survival (OS) being around 40% declining to 10-15% in case of non-visceral and/or lung metastases. Patients failing first line CT have a dismal prognosis, with only approximately 5%–10% of cases being cured in the salvage setting, irrespective of the regime used.

Because of the rarity of the disease, small series have been reported so far on high dose chemotherapy (HDC) with autologous stem cell transplantation (ASCT) in PMGCTs. Such studies do not allow any conclusion and the role of HDC in this setting is still a matter of debate.

Methods: The present retrospective study analyzed the EBMT Registry data regarding adult patients with PMGCTs who underwent HDC and ASCT between 2000 and 2018. All centers with eligible patients were requested to provide additional data including details on pretreatment, disease status at Transplant, post-ASCT treatments and histology. Primary outcomes were OS and progression-free survival (PFS), both measured from the date of first ASCT.

Results: A total of 69 patients from 19 EBMT centers (9 countries) met the eligibility criteria of this study: 5 were seminoma and 64 were NS. Median age was 31 years (range 19-71). Median follow-up post-HDC was 8.4 years. 49 patients underwent HDC after one or two lines and 20 in subsequent relapses. The status at HDC was complete remission (CR) in 9 patients, 31 partial response (PR), stable disease (SD) in 6, progressive disease (PD) in 16, (7 missing information). HDC consisted mainly of carboplatin/etoposide doublet for two or three cycles. All patients received PBPC. Four toxic deaths were recorded in the first 30 days from transplantation (MOF and infections) and one case died after six months for cardiac disease. In the entire population, the probability of PFS was 36% at 5 and 10 years, OS was 37% at 5 years and 33% at 10 years.

Patients undergoing HDC after one or two lines had a better PFS and OS compared to those treated in subsequent relapses (5y PFS: 45% vs 15%; 5y OS: 44% vs 20%). CR or PR at HDC in CR or PR was associated with a better PFS and OS compared to SD, PD or relapse (5y PFS: 49% vs 20%, 5y OS: 50% vs 19%).

Conclusions: This, to our knowledge, is the most extensive study of ASCT in PMGCTs patients and the first to thoroughly investigate potential predictors of benefit from this treatment. Our data suggest that HDC and ASCT may well represent a therapeutic option in selected patients with PMGCTs after first relapse or even as front-line program.

Disclosure: Nothing to declare.

30: Solid Tumours

P691 SURVIVAL OUTCOMES IN PANCREATIC CANCER: EVALUATING THE IMPACT OF WIPPLE’S SURGERY AND HAEMATOPOIETIC CELL TRANSPLANTATION

Olle Ringden 1, Johan Permert2, Johan Törlen1, Stephan Mielke1, Behnam Sadeghi1

1Karolinska Institutet, Huddinge, Sweden, 2Karolinska Instititet, Huddinge, Sweden

Background: Pancreatic ductal adenocarcinoma (PDAC), a foremost cause of cancer mortality, exhibits a dismal five-year survival rate below 10%. Haematopoietic cell transplantation (HCT) demonstrates notable anti-cancer efficacy, albeit with risks of severe complications, including graft-versus-host disease(GVHD).

Methods: Two male patients aged 47 and 66 (Patient 1 and 2, respectively), underwent radical Wipple’s surgery and subsequent HCT from HLA-identical sibling donors. The non-myeloablative conditioning regimen included fludarabine(30 mg/m´2/day for 5 days) and cyclophosphamide (60 mg/kg for 2 days). GVHD prophylaxis entailed cyclosporin A and four doses of Methotrexate. A control group of six patients(one female and five males, aged 67-75) underwent Wipple’s surgery but lacked HLA-identical siblings. Their treatment varied from Gemcitabine given to 5 patients and 5-Fluorouracil to one patient, with two patients declining chemotherapies. Another 60-year-old male (patient 3) with PDAC, having undergone Wipple’s surgery and gastrectomy, recieved HCT from a matched unrelated donor. He is a colleague and member of the ethics commitee.

Histological analysis confirmed complete resection (R0/R1) in all cases. Ethical approval was granted by the Karolinska Institutet(88/96,69/99,385/99,2006/858-314), with written informed consent obtained. ClinicalTrials.Gov NCT02207985.

Results: The contol group, lacking HCT, succumbed to progressive PDAC within a median of 3 years(range 1-3 years).

Petient 1 experienced mild acute GVHD of the skin, effectively treated with steroids, and later mild chronic GVHD of the mouth and gut, managed with photopheresis. He remains well 17 years post-HCT. Patient 2 developed grade III gastrointestinal acute GVHD which responded to steroids. He had reactivation by acute GVHD in gastrointestinal tract and skin at three and six months, which responded to steroids. At 10 months he had reactivation of gastrointestinal GVHD and obstructive broncheolitis(OB). He then suffered from OB and frequent pneumonia. He passed away 17 years post-HCT due to OB and pneumonia. Patient 3 experienced graft rejection;post-gastrectomy, he relies on parenteral nutrition, but is otherwise well 12 years post-HCT. Like GVHD, allograft rejection also has an anticancer effect experimentally and clinically(Rubio, MT et al Blood,2003;102:2300-2307;Söderdahl, G et al Transplantation,2003;75:1061-1066).

All three HCT patients remained free of PDAC, confirmed by CT scans and low CA19-9 levels.

Conclusions: HCT may offer a viable adjuvant therapy for PDAC patients post-radical surgery without distant metastasis.

Clinical Trial Registry: ClinicalTrials.Gov NCT02207985.

Disclosure: Nothing to disclose.

30: Solid Tumours

P692 SUCCESSFUL HIGH DOSE CHEMOTHERAPY WITH STEM CELL RESCUE IN DISSEMINATED OVARIAN CHORIOCARCINOMA COMPLICATED WITH CHORIOCARCINOMA SYNDROME

Abdulaziz Bin Mesained 1, Abdulwahab Alharthi1, Walid Hazem1, Omar Alsharif1, Nawaf Alkhayat1, Mohammad Alshahrani1, Hasna Hamzi1, Amal Binhassan1, Yasser Elborai1

1Prince Sultan Military Medical City, Riyadh, Saudi Arabia

Background: Choriocarcinoma is a rare aggressive malignant germ cell tumor. There are 2 subtypes; gestational and non-gestational choriocarcinoma. Both subtypes have significantly different biological activity and prognosis. Non-gestational ovarian choriocarcinoma (NGOC) accounts for <1% of ovarian germ cell tumors and may develop into a rare and fatal complication like choriocarcinoma syndrome which is a hemorrhagic manifestation of metastatic sites. The optimal treatment strategy for metastatic disease remains unclear due to its low incidence and poor prognosis.

Methods: We reported a case of metastatic (NGOC) presented with choriocarcinoma syndrome and successfully managed with aggressive supportive care, surgical excision, high-risk chemotherapy protocol, and followed by high-dose chemotherapy with stem cell rescue.

Results: A 13-year-old girl presented with intermittent abdominal and chest pain. Abdominal examination revealed a diffusely mild tender abdomen. A computed tomography (CT) scan of the chest showed multiple cannon balls in both lungs (max 3.5cm), the abdomen showed a 1cm hypodense lesion of the lower pole of the left kidney and gastric mass in the stomach fundus, while the pelvis showed huge mass raising from right ovary. The serum tumor biomarker showed significantly high β-HCG. GIT endoscopy showed a fungating metastatic site in the stomach complicated by extensive bleeding. The surgeon did maximum debulking with residual mass, histopathology concluded the diagnosis as choriocarcinoma with extensive hemorrhage and necrosis, immunohistochemistry stains showed positive β-HCG, HPL, PLA, pan-CK and negative CD117, OCT4, CD30. The chemotherapy regimen is a of total 6 cycles, 2 cycles of Cisplatin/ Etoposide/ Bleomycin (PEB) and 4 cycles of Cyclophosphamide- PEB (C-PEB). Evaluation after 1st line of chemotherapy showed β-HCG <1.0 IU/L while the image scan showed regression of the primary pelvic lesion, mild regression in the size of extensive pulmonary nodules with three new nodular basal lung areas noted, regression of renal lesions, and the visualized parts of the stomach showed circumferential fundal thickening. Due to an unexpected response to 1st line of chemotherapy, 2nd line regimen of VeIP (Vinblastin 0.11mg/kg/day x 2 days, Ifosfamide 1200 mg/m2/day x 5 days, Cisplatin 20 mg/m2/day x 5 days) was recommended. Evaluation after 4 cycles of VeIP showed almost complete remission with a small residual mass at the tumor bed most probably fibrosis. We consolidated this remission by high dose chemotherapy (Carboplatin 700 mg/m2 and Etoposide 750 mg/m2, day -5, -4, and -3) followed by autologous stem cell rescue. Regular follow-up by β-HCG monthly and images every 3 months in the first year including PET scan showed no significant abnormal metabolic activity. Now, she completed 7 years of follow-up with complete remission and perfect general condition.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (91)

Conclusions: Choriocarcinoma syndrome is a fatal complication of advanced NGOC. Sequential and intensive chemotherapy are key components of the successful management of metastatic choriocarcinoma. After controlling the disease, we recommend consolidating this remission with high-dose chemotherapy followed by autologous stem cell rescue. Due to its rarity and poor prognosis, more studies are needed to provide evidence of optional therapy.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P693 DONOR-SPECIFIC ANTI-HLA ANTIBODIES (DSAS) IN PATIENTS UNDERGOING ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANTATION FROM MISMATCHED DONORS, ON BEHALF OF GITMO AND AIBT

Ursula La Rocca1,2, Roberto Ricci1, Alfonso Piciocchi3, Walter Barberi1, Elena Oldani4, Alida Dominietto5, Raffaella Cerretti6, Alessandra Picardi6, Francesca Bonifazi7, Riccardo Saccardi8, Maura Faraci9, Giovanni Grillo10, Lucia Farina11, Benedetto Bruno12,13, Anna Grassi4, Anna Proia14, Elena Tagliaferri15, Giuseppina De Simone16, Michele Malagola17, Michela Cerno18, Simone Cesaro19, Paolo Bernasconi20, Lucia Prezioso21, Paola Carluccio22, Nicola Mordini23, Matteo Pelosini24, Attilio Olivieri25, Patrizia Chiusolo26, Stella Santarone27, Michele Cimminiello28, Roberto Crocchiolo10, Franco Papola29, Gianni Rombolà30, Nicoletta Sacchi31, Valeria Miotti32,33, Lia Mele34,33, Benedetta Mazzi35,33, Fabio Ciceri36, Massimo Martino37, Anna Paola Iori 1

1Sapienza University of Rome, Rome, Italy, 2Italian National Institute of Health, Rome, Italy, 3GIMEMA Data Center, Rome, Italy, 4Bone Marrow Transplant Unit, ASST Papa Giovanni XXIII, Bergamo, Italy, 5IRCCS Ospedale Policlinico, S. Martino, Genoa, Italy, 6Stem Cell Transplant Unit, Policlinico Tor Vergata, Rome, Italy, 7IRCSS Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy, 8Careggi University Hospital, Florence, Italy, 9UOSD Bone Marrow Transplant Center-IRCCS Istituto G. Gaslini, Genoa, Italy, 10ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy, 11Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy, 12Division of Hematology, AOU Citta’ della Salute e della Scienza di Torino, Torino, Italy, 13University of Torino, Torino, Italy, 14Unity of Hematology and Stem Cell Transplant, Azienda Ospedaliera S. Camillo Forlanini, Rome, Italy, 15Hematology Unit, Fondazione IRCCS Ca’Granda Ospedale Maggiore Policlinico, Milan, Italy, 16Unità Operativa di Trapianto di Cellule Staminali Emopoietiche e Terapie Cellulari, Azienda Ospedaliera di Rilievo Nazionale Santobono-Pausillipon, Naples, Italy, 17ASST-Spedali Civili di Brescia, University of Brescia, Brescia, Italy, Italy, 18Division of Hematology and Stem Cell Transplantation, Azienda Sanitaria Universitaria Friuli Centrale, Piazzale S. Maria della Misericordia 10, Udine, Italy, 19Azienda Ospedaliera Universitaria Integrata, Verona, Italy, 20Division of Hematology, Fondazione IRCCS Policlinico San Matteo, University of Pavia, Pavia, Italy, 21Hematology and Stem Cell Transplant Unit, Ospedale Maggiore, Parma, Italy, 22Unit of Hematology and Stem Cell Transplantation, AOUC Policlinico, Bari, Italy, 23Hematology Division, Azienda Ospedaliera S. Croce e Carle, Cuneo, Italy, 24Clinical and Experimental Medicine, Section of Hematology, University of Pisa, Pisa, Italy, 25University of Ancona, Azienda Ospedaliera Universitaria Ospedali Riuniti di Ancona, Ancona, Italy, 26Universita’ Cattolica del Sacro Cuore, Rome, Italy, 27Bone Marrow Transplant Center, Ospedale Civile, Pescara, Italy, 28Hematology and Bone Marrow Transplantation Unit, S. Carlo Hospital, Potenza, Italy, 29Regional Centre of Immunohaematology and Tissue Typing, S. Salvatore Hospital, L’Aquila, Italy, 30Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy, 31Italian Bone Marrow Donor Registry, E. O. Ospedali Galliera, Genoa, Italy, 32Laboratory of Immunogenetics, Santa Maria della Misericordia University Hospital, Udine, Italy, 33Associazione Italiana di Immunogenetica e Biologia dei Trapianti, AIBT, Italy, 34Laboratorio HLA e Processazione Cellule Staminali Ematopoietiche SC Medicina Trasfusionale, Azienda Ospedaliera S.S. Antonio e Biagio e C. Arrigo, Alessandria, Italy, 35Immuno-hematology and transfusion medicine Unit San Raffaele Scientific Institute, Milano, Italy, 36University Vita-Salute San Raffaele, IRCCS, S. Raffaele Scientific Institute, Milan, Italy, 37Stem Cell Transplant and Cellular Therapies Unit, “Bianchi-Melacrino-Morelli” Hospital, Reggio Calabria, Italy

Background: The Italian Group for Blood and Marrow Transplantation (GITMO) and Italian Society for Immunogenetics and Transplantation Biology (AIBT) conducted a study on antibodies directed against donor-specific HLA alleles/antigens (DSAs) among Italian Transplant Centers (ITC), aimed at describing Italian clinical and laboratory approaches to DSAs. Here, we present an update of the results obtained.

Methods: Retrospective multicentric study, including all patients who received a mismatched (mm) hematopoietic stem cell transplantation (HSCT) from January2014 to June2017. Twenty-six ITC submitted their data regarding anti-HLA antibodies/DSA detection and management.

Results: 804 patients (38%F, 62%M) with a median age of 48 years (0-72) were evaluable. Overall, 355(44%) were screened: 91/355 showed anti-HLA antibodies (25.6%), 23 DSAs (6.5%). Luminex platform was employed in 93.3% of cases. Other methods included Flow Cytometry, ELISA, and CDC assay; in 7.6% of cases a C1q/C3d assay was performed. F gender(p=0.003) and at least 4previous pregnancies (p=0.024) showed a statistically impact on alloimmunization. Eleven of the 23(48%) patients with DSAs underwent desensitization, with various schedules, mainly based on Rituximab, plasmapheresis, intravenous immunoglobulin, platelet (PLT) transfusions. In 7 cases (30%), no desensitization was employed. An alternative donor was selected for 5 patients (22%). All desensitized patients showed neutrophil (ANC) engraftment, PLT engraftment was observed in all but one. All 5 patients for whom an alternative donor was selected experienced full donor engraftment. Among the patients who did not receive desensitization, 2 did not obtain PLT engraftment and 1 showed secondary thrombocytopenia. ANC and PLT engraftment were obtained in 93.6% and 86.6% of the whole population, respectively, and were statistically associated with the absence of anti-HLA antibodies, ABO match, a higher number of infused nucleated cells and lack of a-GvHD. In addition, significant factors for PLTS engraftment were the use of leuco-depleted transfusions (p=0.005), HLA match (p<0.00), younger age of the patient(p=0.021). GF was associated with bone marrow stem cell source (p=0.032), and a lower number of infused CD34 + (p=0.033). A full donor engraftment was associated with not-leucodepleted blood transfusions(p= 0.006), HLA match (p<0.001), the degree of HLA mismatch in related donors(p=0.014), anti-HLA antibodies (p<0.022), donor gender (p=0.091), the number of total nucleated cells(p=0.004) and CD34 + (p= 0.046), stem cell source (p=0.017), conditioning regimen intensity(p=0.009), ABO match (p=0.004). The 48- and 60-months OS resulted in 57.4 % and 56.5%, respectively. In univariate analysis, the following variables showed an impact on OS: a positive result for antibodies directed against both HLA classes(p<0.001); donor and patient(p<0.001)age; the hematological and molecular remission at HSCT (p<0.001 and p=0.013, respectively); HLAmatch(p=0.007);ANC(<0.001)andPLTSengraftment(p=0.001), early(p<0.001)and lateGF(<0.001), grade>II a-GVHD (p=0.017). Multivariate analysis showed that both classes HLA antibodies(p=0.006), patient age(p=0.020), full donor engraftment (p=0.010), and an early GF (p=0.004) maintained a role on OS.

Conclusions: In ITC a uniform policy to manage DSA before a mmHSCT is lacking. Anti-HLA antibodies and DSAs were confirmed risk factors affecting OS. DSAs were managed with different approaches resulting in stable engraftment in 81.9% of patients. Our study supports the clinical relevance of DSAs detection/management in mmHSCT. A standardized approach of DS is warranted.

Clinical Trial Registry: https://clinicaltrials.gov/ct2/show/record/NCT04888286.

Disclosure: No conflicts of interest to disclose.

10: Stem Cell Donor

P694 DETERMINING THE PREDICTIVE IMPACT OF DONOR PARITY ON THE OUTCOMES OF HUMAN LEUKOCYTE ANTIGEN MATCHED HEMATOPOIETIC STEM CELL TRANSPLANTS: A RETROSPECTIVE, SINGLE-CENTER STUDY

Tanaz Bahri1, Maryam Barkhordar 1, Mojtaba Azari1, Hosein Kamranzadeh1, Seied Asadollah Mousavi1, Soroush Rad1, Sahar Tavakoli Shiraji1, Mohammad Vaezi1

1Cell Therapy and Hematopoietic Stem Cell Transplantation Research Center, Research Institute for Oncology, Hematology, and Cell Therapy, Tehran University of Medical Sciences, Tehran, Iran, Islamic Republic of

Background: The importance of Human Leukocyte Antigen (HLA) matching between patient and donor has been extensively studied, but less attention has been paid to the impact of donor characteristics as age, sex and parity on outcome of HLA-identical Sibling allogeneic stem cell transplantation.

Methods: We evaluated the effect of donor parity on transplant outcomes in a large homogeneously treated population that received an non-T-cell depleted allo-SCT from an HLA-identical sibling donors between 2010 and 2021 at our center. All patients were transplanted from a peripheral blood stem cell source following a myeloablative Busulfan-based conditioning regimen and cyclosporine and a short course of methotrexate as GVHD prophylaxis.

Results: A total of 1103 allo-SCT recipients were included, 188 (17.04%) of these patients had transplants from parous female donors, whereas 621 (56.30%) and 294 (26.70%) of these patients received transplants from male and nulliparous female donors, respectively. The donors’ and receivers’ mean ages were 33.51 and 33.69 years, respectively, with a median age of 32 for both groups. Furthermore, as the primary disease, 415 (37.62%) of the recipients had ALL, and 688 (62.38%) had AML. All patients have been followed up for a median of 73.59 (95%CI: 69.78– 75.89) months. Allo-SCT from parous female donors compared to male and nulliparous females was associated with a significantly higher incidence of grade III-IV acute GVHD (55.27% vs. 11.34 and 10.84%) and extensive chronic GVHD (64.32% vs. 15.52 and 13.65%) respectively. The 5-year relapse incidence (RI) for parous female transplant recipients was significantly lower than for male and nulliparous female recipients (21.26% versus 39.24% and 46.51%, P= 0.00). Additional factors associated with higher RI in univariate analysis were recipient age and sex (male vs. female), as well as disease status of second complete remission and above (≥ CR2) before transplant. Allo-SCT from parous female donors was also associated with significantly poorer GRFS of 5 years compared to male and nulliparous females (11.48% vs 41.41% and 36.01%, P= 0.00). On the other hand, the 5-year OS among individuals transplanted from parous female donors was comparable to those from nulliparous females but significantly reduced than recipients of male donors.

Outcome

Donor sex/ parity

Incidence/ Probability (%)

95% CI

P-value

Grade III-IV aGVHD

Parous female

55.27

43.96-69.00

0.00

Nulliparous female

10.84

7.58-16.00

Male

11.34

8.88-14.00

Extensive cGVHD

Parous female

64.32

50.67-82.00

0.00

Nulliparous female

13.65

9.57-19.00

Male

15.52

12.38-19.00

GRFS

Parous female

11.48

7.19-16.87

0.00

Nulliparous female

36.01

30.17-41.87

Male

41.41

37.32-45.43

RI

Parous female

21.26

14.22-31.77

0.00

Nulliparous female

46.51

37.35-57.91

Male

39.24

33.53-45.93

OS

Parous female

49.17

41.57-56.31

0.039

Nulliparous female

48.61

42.48-54.45

Male

56.32

52.12-60.29

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (92)

Conclusions: This study casts light on the influence of donor parity on outcomes following HLA-identical allogeneic HSCT. However, our data showed that the predictive impact of donor parity on higher incidences of grade III-IV aGVHD and extensive cGVHD can be counterbalanced by the benefit of increasing graft-versus-leukemia effect and a lower risk of relapse, resulting in no significant effect on survival, although it led to poorer GRFS. The findings highlight the importance of non-HLA factors in shaping GVHD incidence, relapse rates, and overall survival. This information equips clinicians with valuable insights for making informed decisions about donor choice and effectively managing potential GVHD risks.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P695 POST-TRANSPLANTATION CYCLOPHOSPHAMIDE IMPROVES GRAFT-VERSUS-HOST DISEASE-FREE, RELAPSE-FREE SURVIVAL IN HLA-DPB1 MISMATCHED UNRELATED DONOR ALLOGENEIC TRANSPLANT

Shannon R. McCurdy1, Scott R. Solomon2, Brian C. Shaffer3, Meilun He4, Yung-Tsi Bolon 4, Amanda G. Blouin3, Alla Keyzner5, Francisco A. Socola6, Uroosa Ibrahim5, Jun Zou7, Hana Safah6, Nakhle Saba6, Shahinaz Gadalla8, Miguel-Angel Perales9, Sophie Paczesny10, Steven Marsh11, Effie W. Petersdorf12, Tao Wang13, Ephraim J. Fuchs14, Stephanie J. Lee12

1University of Pennsylvania, Philadelphia, United States, 2The Blood and Marrow Transplant Group of Georgia, Northside Hospital, Atlanta, United States, 3Memorial Sloan Kettering Cancer Center, New York City, United States, 4Center for International Blood and Marrow Transplant Research, Minneapolis, United States, 5Tish Cancer Institute, Icahn School of Medicine at Mount Sinai, New York City, United States, 6Tulane School of Medicine, New Orleans, United States, 7MD Anderson Cancer Center, Houston, United States, 8National Cancer Institute, Rockville, United States, 9Memorial Sloan Kettering Cancer Center, New York, United States, 10Medical University of South Carolina, Charleston, United States, 11Anthony Nolan Research Institute, Royal Free Campus, London, United Kingdom, 12Fred Hutchinson Cancer Center, Seattle, United States, 13Medical College of Wisconsin, Milwaukee, United States, 14Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, United States

Background: Post-transplantation cyclophosphamide (PTCy)-based graft-versus-host disease (GVHD) prophylaxis has been readily adopted in HLA-mismatched unrelated donor (MMUD) hematopoietic cell transplantation (HCT), but whether PTCy improves the outcomes for 8/8 HLA-matched HCT with mismatches (MM) at HLA–DPB1 is unknown.

Methods: In a Center for International Blood and Marrow Transplantation Research (CIBMTR) analysis, we performed Cox’s proportional hazard models to evaluate the association of GVHD-prophylaxis strategy with overall survival (OS), GVHD-free, relapse-free survival (GRFS), treatment-related mortality (TRM), relapse, and GVHD outcomes after HCT from an unrelated donor with one or two MM at HLA-DPB1. Nonpermissive (NP) MM at HLA-DPB1 were defined by the T-cell-epitope groups model. A stepwise backward variable selection strategy with an alpha threshold of p<0.05 for retention was used. To adjust for multiple testing, p-values of ≤ 0.01 were considered significant.

Results: Eligible patients received a first allogeneic HCT from 2015-2020 for a diagnosis of acute leukemia or myelodysplastic syndrome. Recipients underwent either HLA-DPB1 NP MM (n=434) or permissive MM (n=789) HCT with PTCy +/- mycophenolate mofetil and/or a calcineurin inhibitor or HLA-DPB1 NP MM (n=939) or permissive MM (n=1,952) HCT with methotrexate and tacrolimus (MTX/Tac). OS adjusted for recipient age was not significantly different after HCT with MTX/Tac (hazard ratio [HR]: 1.2, 0.99-1.47, p=0.067) when compared with PTCy. However, in multivariate analysis, TRM was significantly greater (HR: 1.74, 1.28-2.38, p=0.0005) and relapse was significantly lower (HR: 0.76, 0.62-0.93, p=0.0082) in the MTX/Tac group. Grade II-IV acute GVHD (HR: 1.45, 1.14-1.83, p=0.002), grade III-IV acute GVHD (HR: 2.41, 1.61-3.59, p<0.0001), chronic GVHD (HR: 3.12, 2.25-4.32, p<0.0001), and moderate/severe chronic GVHD (HR: 4.42, 3.13-6.22, p<0.0001) rates were all higher in the MTX/Tac cohort. PTCy was associated with significant improvements in GRFS when compared to MTX/Tac (HR: 1.68, 1.39-2.04, p<0.0001) with 1-yr estimates for GRFS of 54% [95% confidence interval (CI): 49-58%] and 39% (95% CI: 35-43%), respectively. GRFS was also significantly better for patients receiving either HLA-DPB1 NP MM or permissive MM HCT with PTCy when compared to those receiving HLA-DBP1 NP MM HCT with MTX/Tac (HR: 1.59, 1.33-1.89, P<0.0001) or those receiving HLA-DBP1 permissive MM HCT with MTX/Tac (HR: 1.46, 1.23-1.73, p <0.0001). For recipients who underwent HLA–DPB1 NP MM HCT with PTCy there were no significant differences in OS, LFS, TRM, relapse, grade III-IV acute GVHD, moderate-severe chronic GVHD, or GRFS when compared to 12/12 HLA-matched unrelated donor HCT with PTCy (n=297) or HLA-DPB1 permissive MM HCT with PTCy. However, risk of grade II-IV acute GVHD was lower in patients with 12/12 HCT with PTCy (HR: 0.72, 0.57-0.92, p=0.0074) compared to HLA-DPB1 NP MM HCT with PTCy.

Conclusions: When compared with MTX/Tac-based GVHD prophylaxis, PTCy is associated with improved GRFS, less TRM, and decreased rates of all types of GVHD when HLA-DPB1 MM donors are used. PTCy should be the preferred GVHD-prophylaxis strategy for HLA-DPB1 MM unrelated donor HCT. Moreover, PTCy abrogates the detrimental effect of NP MM at HLA-DBP1 and thus matching at HLA-DPB1 should not be a major determinant when selecting donors.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P696 THE RESCUE TRANSPLANTATION STRATEGY FOR GRAFT FAILURE: DONOR SELECTION AND GVHD PROPHYLAXIS IN THE CURRENT ERA

Mizuki Watanabe 1,2, Junya Kanda1, Kentaro Fukushima3, Kaito Harada4, Naoyuki Uchida5, Makoto Onizuka4, Noriko Doki6, Ken-ichi Matsuoka7, Takahiro Fukuda2, Satoshi Yoshihara8, Toshiro Kawakita9, Shingo Yano10, Masatsugu Tanaka11, Yasushi Onishi12, Fumihiko Ishimaru13, Tatsuo Ichinohe14, Yoshiko Atsuta15, Kimikazu Yakushijin16, Hideki Nakasone17,18

1Kyoto University Hospital, Kyoto, Japan, 2National Cancer Center Hospital, Tokyo, Japan, 3Osaka University Graduate School of Medicine, Osaka, Japan, 4Tokai University School of Medicine, Isehara, Japan, 5Federation of National Public Service Personnel Mutual Aid Associations Toranomon Hospital, Tokyo, Japan, 6Tokyo Metropolitan Cancer and Infectious Diseases Center, Tokyo, Japan, 7Okayama University Hospital, Okayama, Japan, 8Hyogo Medical University Hospital, Nishinomiya, Japan, 9National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan, 10The Jikei University School of Medicine, Tokyo, Japan, 11Kanagawa Cancer Center, Yokohama, Japan, 12Tohoku University Hospital, Sendai, Japan, 13Japanese Red Cross Kanto-Koshinetsu Block Blood Center, Tokyo, Japan, 14Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan, 15Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan, 16Kobe University Hospital, Kobe, Japan, 17Jichi Medical University Saitama Medical Center, Saitama, Japan, 18Jichi Medical University, Shimotsuke, Japan

Background: Second allogeneic hematopoietic stem cell transplantation (2nd allo-HSCT) to rescue graft failure is often challenging. Since this situation usually requires an immediate donor coordination, cord blood units (CBUs) has been most commonly used for its versatility. Meanwhile, recent widespread use of haploidentical donors has widened the possibility of donor choice, which is also expected to be beneficial in the rescue transplantation. Hence, we evaluated the efficacy/safety of transplantation using related peripheral blood (rPBSCT)/related bone marrow (rBMT) with post-transplant cyclophosphamide (PTCy) as a rescue stem cell source.

Methods: Patients aged 16-77 years who had received their 2nd allo-HSCT using cord blood unit (CBU) or human leukocyte antigen (HLA)-mismatched rPB/rBM for graft failure in Japan between 2000 and 2021 were included. Overall survival (OS), neutrophil engraftment and other transplant outcomes were assessed.

Results: A total of 1145 patients were included with a median follow-up period of 4.5 (range, 0.1-18.6) years for survivors. The interval between the 1st and the 2nd allo-HSCT was 42 days (range, 21-2250) and the number of patients receiving their 2nd allo-HSCT in the early (interval between the 1st and 2nd allo-SHCT < 90days) or late (≥ 90days) period was 995 and 150, respectively. Forty-three patients received HLA-mismatched rPBSCT/rBMT with PTCy as GVHD prophylaxis, 214 received HLA-mismatched rPBSCT/rBMT with prophylaxis other than PTCy (non-PTCy) and 888 received cord blood transplantation (CBT) as a 2nd allo-HSCT. Disease subtypes were acute myeloid/lymphoid leukemia in 640 patients, MDS/CML/MPN/AA in 42, ATL in 56, NHL/HL in 92 and other disease in 42. Among those receiving PTCy, the total dose of cyclophosphamide was <80mg/kg in 7 patients, 80mg/kg in 17 and 100mg/kg in 17. One-year OS in those receiving their 2nd allo-HSCT in the early or late period were 38.1%/41.1% in PTCy rPBSCT/rBMT, 44.6% /55.0% in non-PTCy rPBSCT/rBMT and 33.7%/41.6% in CBT, respectively. Neutrophil engraftment at day28 was achieved in 78.7%/71.0%, 79.3% /78.6% and 52.6%/56.3% in each group, respectively. In the multivariate analysis, PTCy rPBSCT/rBMT showed comparable OS with those of other donor sources both in early (vs non-PTCy: HR 0.83, p=0.480; vs CBT: HR 0.87, p=0.577) and late (vs non-PTCy-rPBSCT/BMT: HR 1.07, p=0.909; vs CBT: HR 0.79, p=0.677) period, with higher frequency of neutrophil engraftment compared to CBT especially in early period (HR 1.61, p=0.022). Other transplant outcomes as relapse (vs non-PTCy, PTCy: HR 0.75, p=0.543; CBT: HR 1.28, p=0.141) and non-relapse mortality (vs non-PTCy, PTCy: HR 0.99, p=0.969; CBT: HR 0.88, p=0.248) was also comparable between the three donor sources.

Conclusions: The outcomes of 2nd allo-HSCT for graft failure remains poor regardless of the timing of transplantation. Related PBSCT/BMT using PTCy could be a tolerable and effective transplant strategy in them, comparable or even better to CBT especially in terms of engraftment. Considering the clinically difficult characteristics of patients receiving the 2nd allo-HSCT, toxicity and other risks of PTCy prophylaxis and its association with patient factors should be further examined in a larger cohort with long follow-up.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P697 UNRELATED DONOR SELECTION FOR STEM CELL TRANSPLANT USING POST-TRANSPLANT CYCLOPHOSPHAMIDE FOR ACUTE MYELOID LEUKEMIA: A STUDY FROM THE ACUTE LEUKEMIA WORKING PARTY OF THE EBMT

Jaime Sanz 1, Myriam Labopin2, Goda Choi3, Alexander Kulagin4, Jacopo Peccatori5, Jan Vydra6, Péter Reményi7, Jurjen Versluis8, Montserrat Rovira9, Didier Blaise10, Hélène Labussière-Wallet11, Juan Montoro12, Simona Sica13, Ellen Meijer14, Maija Itäla-Remes14, Nicolaas Schaap15, Claude Eric Bulabois16, Simona Piemontese5, Mohamad Mohty17, Fabio Ciceri5

1Hospital Universitari i politècnic La Fe, Valencia, Spain, 2EBMT Paris Office, Hospital Saint Antoine, Paris, France, 3University Medical Center Groningen (UMCG), Groningen, Netherlands, 4RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russian Federation, 5Hematology and Bone Marrow transplant Unit, San Raffaele Scientific Institute IRCCS, Milano, Italy, 6Institute of Hematology and Blood Transfusion, Prague, Czech Republic, 7Dél-pesti Centrumkórház, Budapest, Hungary, 8Erasmus MC Cancer Institute, Rotterdam, Netherlands, 9BMT Unit, Institute of Haematology and Oncology, IDIBAPS, Hospital Clinic, University of Barcelona. Josep Carreras Leukaemia Research Foundation, Barcelona, Spain, 10Programme de Transplantation & Therapie Cellulaire, Marseille, France, 11Centre Hospitalier Lyon Sud, Lyon, France, 12Hospital Universitari i Politècnic La Fe, Valencia, Spain, 13Universita Cattolica del Sacro Cuore, Rome, Italy, 14VU University Medical Center, Amsterdam, Netherlands, 15Radboud University Medical Center, Nijmegen, Netherlands, 16CHU Grenoble Alpes - Université Grenoble Alpes, Grenoble, France, 17Hôpital Saint-Antoine, Sorbonne University, INSERM UMRs 938, Paris, France

Background: Post-transplant cyclophosphamide (PTCy) is increasingly used in matched unrelated donor (MUD) or mismatched unrelated donor (MMUD) hematopoietic stem cell transplant (HSCT). However, there is a paucity of information on how to select the most appropriate donor in this setting.

Methods: We retrospectively analyzed the characteristics of MUDs and 9/10 MMUDs that may affect transplant outcomes in patients with acute myeloid leukemia (AML) in first or second complete remission (CR1 or CR2) who received a first peripheral blood allogeneic HSCT with PTCy between January 2010 and December 2022, registered in the EBMT database. The primary endpoint was leukemia-free survival (LFS).

Results: Overall, 1011 patients were included with a median age of 54 years (range, 18-77); 873 (86%) were in CR1, 612 (61%) had intermediate-risk cytogenetics, and 548 (54%) received myeloablative conditioning. Regarding donors, 621 (61%) were MUDs and 390 (39%) were MMUDs. The median age was 29 years (range, 18-64), 304 (30%) were females of which 150 (15% of whole group) were donors to male recipients. Of donors, 522 (52%) had negative cytomegalovirus (CMV-neg) serostatus of which 189 (19%) were used for CMV-neg recipients. The cumulative incidence of acute GVHD grade II-IV and III-IV 180-day was 24% (95% CI 22-27), 7% (95% CI 6-9), while the 2-year cumulative incidence of chronic and chronic extensive GVHD was 31% (95% CI 28-35) and 12% (95% CI 10-15), respectively. After a median follow-up of 26 months, the cumulative incidence of relapse and non-relapse mortality and the probabilities of LFS, overall survival (OS) and GVHD-free, relapse-free survival (GRFS) were 25% (95% CI 22-27), 12% (95% CI 10-14), 64% (95% CI 60-67), 70% (95% CI 67-73) and 52% (95% CI 49-56), respectively. On multivariable analysis, donor age older than 30 years had a negative impact on LFS (hazard ratio [HR] 1.4; 95% CI 1.12-1.74), while CMV-neg donor for CMV-neg recipient was associated with improved LFS (HR 0.74; 95% CI 0.55-0.99). Donor age older than 30 years also showed a detrimental effect on relapse (HR 1.38; 95% CI 1.06-1.8), OS (HR 1.45; 95% CI 1.14-1.85) and GRFS (HR 1.29; 95% CI 1.07-1.56). The use of MMUD and female donors for male recipients did not significantly impact any transplant outcomes.

Conclusions: For patients undergoing HSCT from an unrelated donor with PTCy for AML in CR, younger donor age significantly improves survival and should be the primary donor-related characteristic guiding donor selection. In this context, donor age might be prioritized over HLA match considerations. In addition, CMV neg donors are preferable for CMV neg recipients.

Disclosure: The authors declare no competing interests.

10: Stem Cell Donor

P698 PATIENT HLA GENOTYPE AND REGISTRY DIVERSITY STRONGLY INFLUENCE DONOR SEARCH DURATION AND SUCCESS

Stefanie V. Dorner 1, Henning Baldauf2, Carina Rave2, Christine Urban3, Thilo Mengling3, Johannes Schetelig2, Alexander H. Schmidt3,1, Julia Pingel1

1DKMS Registry gGmbH, Tübingen, Germany, 2DKMS Group gGmbH, Dresden, Germany, 3DKMS Group gGmbH, Tübingen, Germany

Background: The availability of unrelated stem cell donors is of increasing importance for HSCT. In this study, success and duration of stem cell donor search have been investigated from the perspective of a donor registry.

Methods: The DKMS Registry analysed 12,444 search requests received between July 2021 and June 2022. The analysis considered unrelated donor searches requested from transplant centres with sufficient data availability (Germany and USA were n/a) and sample size. These were located in Europe (n=8,480), Canada (n=1,309), India (n=1,085), Australia (n=980), South Africa (n=248), Singapore (n=235) and South America (n=107). As cancellation reasons are not systematically available, this study focused on three questions: (1) In how many initial donor searches at least one DKMS donor has been selected for a workup request (=successful case in this study)? (2) How long did it take from first search request to workup request within these successful cases? (3) Which variables did influence the search?.

Univariable and multivariable logistic models and Cox models have been applied to analyse impact factors on search success and duration, respectively.

Results: Patients with a malignant haematological disease are almost twice as likely to request a workup compared to patients with non-malignant or other diseases. A DKMS donor was requested for workup in 26-30% of searches for malignant diseases, but only 15% for non-malignant or other diseases. Patients with AML/MDS had the lowest median search duration until start of workup of 56 days. Patients with ALL, other malignant haematological diseases and patients with non-malignant or other diseases had a median search duration of 62, 71 and 61 days, respectively.

European patients had a workup rate of 31%. Correspondingly, Canada, Australia and India showed 19%, 17% and 8.5%, respectively. The median search duration of successful Indian searches was only 47 days, whereas successful searches of patients from Europe, Canada, Australia took 57, 67 and 87 days, respectively.

Patients with three or more 10/10-donors had a workup request in 38% of cases after a median search duration of 58 days. Respectively, patients with only one or more 9/10-donors were successful in only 15% of cases after a median of 66 days (p<0.001).

In a second analysis, the period between pre-workup and workup request has been investigated. In 36% of the patient cases where one or two donors had pre-workup requests like Confirmatory Typing, also a workup was requested. In cases where 3-5 (≥6) donors had a pre-workup request, the workup rate was 53% (58%).

Median search duration in cases with urgent pre-workup requests was 6 days shorter than in non-urgent cases (47 vs. 53 days; p=0.051).

Conclusions: Our study confirms that the unrelated donor search success depends on the patient’s HLA-frequency and the size and diversity of the genetically relevant DKMS donor pool (e.g. DKMS-BMST India ~50,000 donors), whereas this pool is “increased” for malignant diagnoses by higher acceptance of mismatched donors. The duration of a search varies by the patient’s country and is positively influenced by an “urgent”-flag at pre-workup stage.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P699 FEASIBILITY AND SAFETY OF GRANULOCYTE COLONY-STIMULATING FACTOR (G-CSF) PERIPHERAL BLOOD STEM CELL MOBILIZATION AND COLLECTION BY APHERESIS IN RELATED DONORS AGED 65 AND OVER

Kahina Amokrane 1, Kamelia Alexandrova1, Cristina Castilla-Llorente1, Sylvain Pilorge1, Tereza Coman1, Jean-Henri Bourhis1, Valerie Lapierre1

1Gustave Roussy, Villejuif, France

Background: Allogeneic hematopoietic stem cells (HSC) transplantation (AlloHSCT) is the only curative treatment for several hematological malignancies. The advances in supportive care, better donor selection and the development of reduced intensity conditioning allows the increasingly use of this treatment for elderly patients suffering from hematological malignancies. The increase in the age of recipients correlates with that of related donors, and raises the question of the feasibility and safety of HSC collection from elderly donors.

Methods: From January 2014 to November 2023, we performed 749 alloHSCT at the Gustave Roussy Hematology Dpt, of which 346 (46%) were related donors. In this population, we retrospectively evaluated HSC mobilisation in 35 (10%) donors aged 65 years or more. All these patients followed the same mobilisation protocol based on the administration of a dose of 10µg/kg/day of G-CSF divided into two daily doses. Peripheral blood (PB) HSC collections were performed by apheresis using continuous mononuclear cell collection procedure, with Spectra Optia (Terumo BCT) with an extracorporeal circuit of 250 ml. PB-HSC collection was performed on day 4 (after 7 doses) and/or day 5 (after 9 doses), depending on the PB CD34+ cell count and on CD34+ cell graft quantity expected (usually more than 4x106/Kg of body weight of the recipient or 6x106 in case of cryopreservation). None of the donors were treated with MOZOBIL (Plerixafor®). All donors were previously checked by clinical examination and standard biological check-up for donor validation, and had, in addition, a systematic evaluation including a blood lipid profile, cardiac ultrasound, arterial ultrasound of the supra-aortic vessels and a low-dose non-injection chest CT scan. All donors had a Karnowski index of 100%.

Results: The median age of the donors was 67 years [65 to 73]. For 34 (97%) of the 35 donors, the HSC collection provided a graft, of which 20 donors (57%) required only one PB-HSC collection (15 (42%) on day 4 and 5 (15%) on day 5). See table 1. Mobilization failed for one donor in whom the graft was obtained by bone marrow collection. No apheresis was interrupted for safety reason. The median number of total body blood mass treated was 2.7 [2.0-3.8]. The only adverse events reported were low back pains and headaches, each of which responded to paracetamol administration. No adverse event related to apheresis was reported. There was no graft failures reported in recipients.

Full size table

.

Table1: Graft characteristics according to the number of days of G-CSF administration and the day of harvesting.

Conclusions: This study unequivocally establishes the feasibility and safety of apheresis collection of G-CSF-mobilized PB-HSC from related donors aged 65 and above, subject to meticulous donor selection based on our concise set of pre-donation assessments. As the population ages, these findings provide crucial insights into expanding the donor pool for alloHSCT, ensuring safer and more accessible treatment options for hematologic malignancies.

Disclosure: no conflict of interest.

10: Stem Cell Donor

P700 HAPLOIDENTICAL VERSUS MISMATCHED UNRELATED ALLOGENEIC STEM CELL TRANSPLANTATION FOR MYELOID MALIGNANCIES: AN ANALYSIS ON 1413 PATIENTS FROM THE GERMAN REGISTRY

Andrea Gantner 1, Daniel Fürst2,3, Aysenur Arslan2,3, Svenja Labuhn4, Mark Ringhoffer5, Johannes Schetelig6,7, Thomas Schroeder8, Gesine Bug9, Elisa Amann2,3, Christine Neuchel2,3, Sandra Schmeller4, Jan Beyersmann4, Hubert Schrezenmeier2,3, Joannis Mytilineos10, Nicolaus Kröger11, Elisa Sala1

1University Hospital Ulm, Ulm, Germany, 2Institute of Clinical Transfusion Medicine and Immunogenetics Ulm, German Red Cross Blood Transfusion Service, Baden Wuerttemberg – Hessen, Ulm, and University Hospital Ulm, Ulm, Germany, 3Institute of Transfusion Medicine, University of Ulm, Ulm, Germany, 4Institute of Statistics, University of Ulm, Ulm, Germany, 5Medizinische Klinik III, Städtisches Klinikum Karlsruhe, Karlsruhe, Germany, 6Medizinische Klinik und Poliklinik I, Universitätsklinikum Carl Gustav Carus, Dresden, Germany, 7DKMS Group, Clinical Trials Unit Dresden, Dresden, Germany, 8West German Cancer Center Essen, University Hospital Essen, Essen, Germany, 9Goethe University Frankfurt, Frankfurt, Germany, 10ZKRD - Zentrales Knochenmarkspender-Register für Deutschland, German National Bone Marrow Donor Registry, Ulm, Germany, 11University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: Allogenic stem cell transplantation (allo-SCT) is the only curative option for high-risk myeloid diseases. Donor selection represents a fundamental issue, significantly impacting on outcome. In absence of an HLA-matched donor, allo-SCT from 9/10-matched unrelated donors (9/10-MUD) or haploidentical donors (haplo) can be performed. The incidence of graft-versus-host-disease (GVHD) correlates with the degree of HLA-disparity under standard GVHD-prophylaxis with calcineurin inhibitors, methotrexate/mycophenolate-mofetil plus anti-thymocyte globulin (ATG). Post-transplantation cyclophosphamide (PT-Cy) has revolutionized GVHD-prophylaxis, reducing non-relapse mortality (NRM) especially in the setting of mismatched donors. The aim of this study was to retrospectively compare outcomes of patients with myeloid malignancies undergoing 9/10-MUD-allo-SCT using ATG versus haplo-allo-SCT with PT-Cy.

Methods: We conducted a retrospective analysis evaluating patients who underwent first allo-SCT for myeloid malignancies from a 9/10-MUD or haplo-donor between 2009-2020 in Germany. Outcomes for overall survival (OS), disease-free survival (DFS), GVHD-relapse-free survival (GRFS), NRM, relapse incidence as well as acute (aGVHD) and chronic GvHD (cGVHD) incidences were analysed. Multivariable Cox proportional hazard models and competing risks regression models were fitted to analyse the impact of donor type on outcome.

Results: We identified 1413 adults receiving allo-SCT, 279 (19,7%) from a haplo-donor plus PT-Cy and 1134 (80,3%) from 9/10-MUD with ATG. Patients’ characteristics are listed in Table1. The 1-year cumulative incidence of aGVHD was 16,8% in the 9/10-MUD-cohort and 16,0% in the haplo-cohort (p=0,702). The 5-years (5y) cumulative incidence of cGVHD was significantly higher for haplo-allo-SCT (haplo: 42,4% versus 9/10-MUD: 33,1%, p=0,006). In univariate analysis, 5y-GRFS was comparable (9/10-MUD: 22,5% versus haplo: 24,7%, p=0,774) as well as 5y-NRM (haplo: 29,4% versus 9/10-MUD: 36,8%; p=0,054). The 5y-DFS was significantly better in the haplo-group (9/10-MUD: 32,9% versus haplo: 38,4%, p=0.009), with no differences in 5y-relapse incidence (9/10-MUD: 30,3%; haplo: 32,2%, p=0,378). Overall, this resulted in significantly better 5y-OS for haplo-allo-SCT (9/10-MUD: 37,8% versus haplo: 49,0%, p=0,009). Considering the multivariable analysis, the use of a 9/10-MUD compared to haplo-donor revealed no significant effect on OS (HR 1,21, CI 0,97-1,5, p=0,085), as well as on DFS (HR 1,21, CI 0,99-1,48, p=0,065) and GRFS (HR 0,97, CI 0,81-1,16, p=0,72).

Variable

Values

PT-Cy-Haplo

9/10-MUD

Total (n)

P-value

Patient Age

Years median (range)

58 (18-79)

58 (18-77)

1413

0,995

Disease Type

AML n (%)

233 (83.5)

866 (76.4)

1099

0,013

MDS n (%)

46 (16.5)

268 (23.6)

314

Disease Stage

Early n (%)

142 (50.9)

535 (47.2)

677

0,128

Intermediate n (%)

67 (24)

304 (26.8)

371

Advanced n (%)

68 (24.4)

294 (25.9)

362

Missing n (%)

2 (0.7)

1 (0.1)

3

Year of Tx

2000 - 2005 n (%)

0 (0)

17 (1.5)

17

<0.001

2006 - 2010 n (%)

1 (0.4)

346 (30.5

347

2011 - 2015 n (%)

35 (12.5)

449 (39.6)

484

2016 - 2020 n (%)

243 (87.1)

322 (28.4)

565

Stem cell source

BM n (%)

71 (25.4)

55 (4.9)

126

<0.001

PBSC n (%)

205 (73.5)

1078 (95.1)

1283

Missing n (%)

3 (1.1)

1 (0.1)

4

Conditioning intensity

MAC n (%)

157 (56.3)

580 (51.1)

737

0,165

RIC n (%)

122 (43.7)

548 (48.3)

670

Missing n (%)

0 (0)

6 (0.5)

6

  1. Abbreviations: AML= acute myeloid leukemia, BM=Bone Marrow, D=Donor, MAC=Myeloablative conditioning, MDS= myelodysplastic syndrome, MUD=Matched unrelated donor, PBSC=Peripheral blood stem cells, PT-CY=Post transplant cyclophosphamide, RIC=Reduced intensity conditioning, Tx=Transplantation

Conclusions: Our data showed comparable outcome for patients with high-risk myeloid malignancies after haplo-allo-SCT plus PT-Cy versus 9/10-MUD plus ATG, with a trend for better OS and DFS in the haplo-group (multivariable analysis). The significantly better OS and DFS in the haplo-allo-SCT plus PT-Cy-cohort also emerging in univariate analysis could have different interpretations: (1) potentially higher graft-versus-leukemia effect for haplo-donors delivered by higher degree of mismatch (also concordant with a significantly higher incidence of cGVHD in this cohort); (2) year of transplantation, with the majority of haplo-allo-SCT being performed after 2016, while most of 9/10-MUD-allo-SCT before. Better DFS with comparable relapse incidence may partly be attributable to recent improvements in treatment options for myeloid malignancies. In conclusion, our findings support the hypothesis that PT-Cy could potentially abrogate the role of HLA-differences in allo-SCT. In order to better evaluate the impact of PT-Cy on outcome, prospective clinical trials comparing PT-Cy with ATG in mismatched setting are needed.

Disclosure: Johannes Schetelig: Advisory Boards participation: Abbvie, AstraZeneca, BeiGene, BMS, Janssen, and MSD; Lecture Fees from Astellas, AstraZeneca, BeiGene, BMS, Novartis, Eurocept, Medac and Janssen.

Gesine Bug: GB has received honoraria from Novartis, Jazz, BMS and Gilead and travel grants from Jazz, Gilead and Neovii.

Elisa Sala: honoraria for consultancy or travel support from Gilead, Novartis, BMS, Jazz, Neovii, Therakos/Mallinckrodt, MSD, Priothera.

10: Stem Cell Donor

P701 THE POST-PANDEMIC HAEMATOPOIETIC STEM CELL (HSC) GRAFT CRYOPRESERVATION TRENDS AND THEIR EFFECTS ON UNRELATED DONOR REGISTRIES: THE UK EXPERIENCE

Ruta Barkauskiene1, Sara Lozano Cerrada1, Angharad Pryce1, Joanne Badger1, Paul Johnson1, Ann O’Leary1, Robert Danby 1, Chloe Anthias1

1Anthony Nolan, London, United Kingdom

Background: To address the challenges and disruptions posed by the COVID-19 pandemic on the haematopoietic stem cell (HSC) provision, international bodies, such as European Society for Blood and Marrow Transplantation (EBMT) and World Marrow Donor Association (WMDA) recommended HSC graft cryopreservation.

We conducted a retrospective review of transplant centre practices to determine how cryopreservation practice has changed since the end of the pandemic. With concerns of primary/ secondary graft failure and emerging evidence of reduced progression free survival (PFS) in recipient of cryopreserved grafts, we also examined whether cryopreservation affected subsequent donation rates of HSC and donor lymphocytes.

Methods: Data were collected from Anthony Nolan (AN) and aligned UK registries database for 3 separate time periods to allow comparison between practices before, during and after the pandemic. These time periods were defined as follows:

  • Pre-pandemic: 11/03/2016 to 05/05/2019.

  • During the COVID-19 pandemic: 11/03/2020 to 05/05/2023 (as defined by World Health Organisation (WHO)).

  • Post-pandemic: 06/05/2023 to 01/12/2023.

.

Bone marrow (BM) harvest and umbilical cord (UC) donations were excluded from analysis concentrating on PBSC and DLC.

Results: AN and aligned UK registries continued PBSC provision during the pandemic at similar levels to pre-pandemic rates: 3577 vs 3614 pre and during the pandemic, respectively (Table 1).

Cryopreserved grafts comprised 5.7% of all PBSC provisions pre-pandemic and in keeping with international recommendations rose to 68.8% during the pandemic. Post-pandemic graft cryopreservation remains at higher than pre-pandemic levels at 18.2% (Table 1).

TCs have provided the following reasons for on-going PBSC graft cryopreservation: logistical challenges - 18%; delay due to patient condition (relapse, other causes of deterioration) - 32%; high or rising COVID-19 rates in the recipient’s or donor’s country – 10%; concerns with donor suitability to proceed – 3%; and other – 37%. Within the 34 UK TCs the rates of cryopreservation of PBSC grafts vary highly between 0 to 100% of all unrelated donor transplants performed (Graph 1).

Unused product rates have significantly increased during the pandemic and remained high during the post-pandemic period (Table 1). Majority of unused products were not infused because of patient related reasons (Table 1).

There was no increase in subsequent PBSC or DLI requests during and post-pandemic when compared to pre-pandemic levels (Table 1).

Pre-pandemic

(11th March 2016 – 5th May 2019)

COVID-19 pandemic

(11th March 2020- 5thMay 2023)

Post-pandemic

(6th May – 1st December 2023)

Total PBSC provisions

3577

3614

734

Total DLC provisions

434

477

101

% DLC of total DLC + PBSC

10.8%

11.7%

12.1%

Cryopreserved PBSCs (% of total PBSC)

206 (5.7%)

2487 (68.8%)

134 (18.2%)

Unused products – PBSC (% of total PBSC and % of cryo PBSC)

4 (< 0.1% and 1.9%)

100 (2.7% and 4%)

16 (2.1% and 11.9%)

Unused products – DLC (% of total DLC)

4 (0.9%)

21 (4.4%)

8 (7.9%)

Number of unused products due to patient reasons (%)

-

79 (65.3%)

20 (83.3%)

Number of subsequent PBSC donations (% of total PBSC donations)

46 (1.3%)

45 (1.2%)

6 (0.8%)

Number of subsequent DLC donations (% of total DLC donations)

229 (52.7%)

196 (41.1%)

31 (30.7%)

Table 1. Comparison of PBSC and DLC provision, cryopreservation percentage, rates of unused products and subsequent donation requests during the pre-pandemic, COVID-19 pandemic and post-pandemic periods.

Conclusions: AN and the aligned UK registries demonstrated resilience and adaptability which allowed to maintain continuous unrelated PBSC graft provision during the COVID-19 pandemic.

Post-pandemic graft cryopreservation rates remain higher than pre-pandemic rates but there is a significant variability between individual UK TC practices with some TCs reverting back to exclusive use of fresh PBSC grafts. Logistical difficulties and patient deterioration are among the most frequent reasons for graft cryopreservation as stated by the UK TCs.

Despite concerns of primary/ secondary graft failure and potentially reduced PFS with cryopreserved grafts, there has not been an increase in subsequent PBSC or DLC requests during pandemic and post-pandemic period.

Number of unused PBSC and DLC product collections was higher during pandemic period and remains so post-pandemic. Improved communication between TCs, collection centres and registries is essential to address this issue.

Clinical Trial Registry: N/A.

Disclosure: None of the authors have any interests of conflict to declare.

10: Stem Cell Donor

P702 ALLOGENIC STEM CELL TRANSPLANTATION AND PERI TRANSPLANT STRATEGIES IN PATIENTS WITH MISMATCH DONORS – A SINGLE CENTER EXPERIENCE IN 151 PATIENTS

Paul Jäger1, Benno Biermann 1, Felicitas Schulz1, Kathrin Nachtkamp1, Patrick Tressin1, Ben-Niklas Baermann1, Annika Kasprzak1, Felix Matkey1, Ulrich Germing1, Sascha Dietrich1, Guido Kobbe1

1University Hospital Düsseldorf, Düsseldorf, Germany

Background: The feasibility of allogenic hematopoietic stem cell transplantation (HCT) with HLA-mismatched related (haplo) and unrelated (9/10 MMUD) donors has shifted because of the adoption of posttransplantation cyclophosphamide (PTCy). The most common transplant-specific complication of HLA-mismatched unrelated donor (MMUD) HCT are GvHD and relapse. The optimal strategy to prevent GvHD without increasing the risk of relapse in this setting is unclear. Whereas in the haplo setting the administration of PTCy is common practice, in vivo T-cell depletion with peritransplant antithymocyte globulin (ATG) is often performed in patients undergoing 9/10 MMUD HCT. If both haplo donors and 9/10 MMUD are available for a patient, individual patient-, disease- and therapy-specific factors must be considered when deciding which donor to use. Here we report our single center experience of peri transplant strategies in patients with mismatch donors.

Methods: A total of 151 patients (AML n=58, MDS n=29, ALL n=17, NHL n=14, CMML n=11, MPN n=7, excluded n=15) were analyzed who received transplants from mismatched donors from 2012 to 2023 (haplo PTCy n=38, 9/10 MMUD ATG n=77, 9/10 MMUD PTCy n=21). Fifteen patients with 2nd/3rd HCT were excluded from further analysis. The EBMT- and Armand-Scores were distributed evenly in the individual groups.

Results: After a median follow-up of 3.1 years from HCT patients with haplo donor (all PTCy) and patients with 9/10 MMUD who received PTCy had superior 2-y-OS of 73.05% and 70.89%, compared to patients with 9/10 MMUD who received ATG with a 2-y-OS of 53.95% (p=0.1532). The overall non relapse mortality was 18% in patients with haplo donors, 10% in patients with 9/10 MMUD with PTCy and 25% in patients with 9/10 MUD who received ATG.

Patients with haplo donor who were transplanted in CR (<5% BM-blasts) had a 2-y-OS and a 2-y-PFS of 83.33% and 62.22%, respectively. Interestingly, patients with 9/10 MMUD who were transplanted in CR had similar 2-y-OS and 2-y-PFS of 66.96% and 55.27%, respectively. Patients with haplo donors who had active disease at transplant had 2-y-OS and 2-y-PFS of 64.62% and 52.14%, respectively. In contrast, patients who had received a transplant from 9/10 MMUD with active disease had a markedly lower 2-y-OS and 2-y-PFS of 47.35% and 23.29%, respectively (p=0.0414). The organ toxicities were similar in the individual groups, but WBC engraftment (>1000µl) was prolonged by a median of 3 days in patients who got PTCy and the median duration of hospitalization was three days longer in the haplo group. In transplants from 9/10 MMUD, the administration of PTCy did not lead to a reduction in overall GvHD activity, but to a reduction of chronic and severe acute GvHD.

Conclusions: Our data suggests that transplants from haplo donors may be preferable to 9/10 MMUD, especially in patients not in CR at the time of transplantation. If only 9/10 MMUD are available in patients not in CR, poor outcome is likely and alternative strategies should be considered. The use of PTCy in patients transplanted with 9/10 MMUD may be a good alternative to ATG and results in less chronic and severe acute GvHD.

Disclosure: The authors have no conflicts of interest.

10: Stem Cell Donor

P703 SURVIVAL PROGNOSTIC FACTORS AFTER A SECOND ALLOGENEIC HEMATOPOIETIC TRANSPLANTATION

Rodrigo Cantera Estefanía1, Raquel García Ruiz1, Miriam Sánchez Escamilla1, Leddy Patricia Vega Suárez1, María Terán1, Dianis Escorcio Fariab2, Sara Fernández Luis1, Juan José Domínguez García1, Tatiana Fernández Barge 1, Ana Gea Peña1, Irene Francés Alexandre1, Ana Tobalina García1, María Oviedo Madrid1, Irene Gorostidi Álvarez1, Julia Bannatyne Undabeitia1, Juan Manuel Cerezo Martín1, Mercedes Colorado Araujo1, Mónica López Duarte1, Guillermo Martín Sánchez1, Lucrecia Yáñez San Segundo1, Noemí Fernández Escalada1, Andrés Insunza Gaminde1, Monserrat Briz del Blanco1, Jose Iñigo Romón Alonso1, Gala Aglaia Méndez Navarro1, Enrique Ocio San Miguel1, Arancha Bermúdez Rodríguez1

1Hospital Universitario Marqués de Valdecilla, Santander, Spain, 2Hospital de Cabueñes, Gijón, Spain

Background: A second allogeneic stem cell transplant (2nd-alloSCT) may be a curative option for patients with relapse/refractory hematological disorders. However, it is associated with a high morbi-mortality. The primary objective of this study is to evaluate outcomes (overall survival [OS], progression-free survival [PFS], non-relapse mortality [NRM] and relapses) after a 2nd-alloSCT. Secondary objective is to analyze prognostic factors that impact on survival.

Methods: We retrospectively evaluated 40 patients who underwent a 2nd-alloSCT in our institution between January 2008 and May 2023.

Results: Median age at transplant was 44 years (range 8-59). The primary indication for transplant was relapsed disease after 1st-alloSTC in 28 (70%) patients, followed by graft failure (GF) in 11 (27.5%) patients. Disease status at transplant was complete remission in 21/28 patients. Patient´s characteristics are shown in table 1.

With a median follow-up of 8.6 years (range 0.01-15.2), 1-year and 5-year OS and PFS rates were 59.8%, 36.2%, and 59.7%, and 29.5%, respectively. Eight patients (20%) relapsed with a median of 1.1 years (range 0.16-2.66) post-2nd-alloSCT. The Cumulative incidence (CI) of NRM at day +100 and 1-year was 32.5% and 40.4%, respectively. Fourteen patients (35%) died due to toxicity (4 infections, 4 graft versus host disease (GVHD), 2 neurological toxicity, 2 secondary lymphoproliferative disorders, 1 pulmonary toxicity, 1 secondary neoplasm). Four patients (10%) died due to GF (2, secondary GF and 2, primary GF). Reason of 2nd-alloSCT in these patients was 2 GF and 2 relapses.

Six patients (15%) died due to relapse (relapse was the indication for the 2nd-alloSCT in all of them), in 5 of relapsed patients time from 1st to 2nd-alloSCT was >12 months and in 1 patient was <6 months, 5 of them were in remission and only 1 patient had active disease at time of 2nd-alloSCT.

In the univariate analysis, donor change, graft source, conditioning intensity, GVHD prophylaxis, T-cell depletion, acute GVHD (aGVHD) and reason of 2nd-alloSCT (relapse vs GF) did not impact on OS and PFS. Time between first and 2nd-alloSCT less than 12 months impacted negatively in OS (0.14 years vs 3.27, p<0.01).

In multivariate analysis, unrelated mismatched donor negatively affected OS (HR 6.00, p=0.04) and both, OS and PFS, were negatively affected by pretransplant refractoriness (OS HR 5.91, p=0.06; PFS HR 6.23, p=0.05) and HCT-CI ≥3 (OS HR 5.01, p=0.03; PFS HR 4.48, p=0.03). On the other hand, post-transplant preventive interventions with lymphocyte infusion or maintenance improved OS (HR 0.12, p=0.07) and chronic GVHD (cGVHD) improved PFS (HR 0.19, p=0.04).

Patients (n=40)

Median Age: 44,5 years (8 – 59)

DRI

Indication of 2º-alloSCT

T-depletion

Sex

Low risk

1 (2.5%)

Relapse

28 (70%)

ATG

20 (50%)

Men

23 (57.5%)

Intermediate risk

4 (10%)

Primary graft failure

4 (10%)

Cy post

7 (17.5%)

Women

17 (42.5%)

High risk

18 (45%)

Secondary graft failure

7 (17.5%)

Time between ALLO-SCT

Diagnosis

Very High risk

6 (15%)

Secondary MDS

1 (2.5%)

<6 months

7 (17.5%)

AML/MDS

20 (50%)

Source

Disease Status

6-12 months

3 (7.5%)

ALL

10 (25%)

Peripheral Blood

32 (80%)

2ª CR

12 (30%)

> 12 months

30 (75%)

Lymphoma

4 (10%)

Bone Marrow

8 (20%)

≥ 3ª CR

9 (22.5%)

Donor

Chronic MPS

2 (5%)

Donor Change

PR

2 (5%)

Matched sibling

10 (25%)

MM

3 (7.5%)

Yes

16 (40%)

RR

6 (15%)

Missmatched sibling

1 (2.5%)

Aplastic anemia

1 (2,5%)

No

24 (60%)

Graft failure

11 (27.5%)

Matched unrelated

13 (32.5%)

Karnofsky

Prophylaxis GVHD

HCT-CI

Missmatched unrelated

9 (22.5%)

<80%

10 (25%)

Tacrolimus+MMF/MTX

16 (40%)

< 3

16 (40%)

Haploidentical

7 (17.5%)

>80

26 (65%)

Ciclosporine+MMF/MTX

24 (60%)

≥ 3

20 (50%)

Table 1. Patient’s characteristics. ALLOSCT: Allogeneic Transplant. AML/MDS: Acute Myeloid Leukemia/Myelodysplastic Syndrome. ATG: Antithymocyte Globulin. CMV: Cytomegallovirus. CR: Complete Remission. Cy post: Post-Transplant Cyclophosphamide. GF: Graft Failure. GVHD: Graft vs. Host Disease. MM: multiple myeloma. MMF: Mycophenolate. MPS: Myeloproliferative Syndrome. MTX: Methotrexate. PR: Partial Response. RR: Relapse/Refractoriness.

Conclusions: Second-alloSCT can salvage 36% of patients at 5 years, but high NRM rates are expected (36% and 40% after 100 days and 1 year). Disease control prior transplant, HCT-CI<3, cGVHD and time between both alloSCT >12 months significantly improved OS in our study.

Disclosure: The authors declare no conflicts of interest.

10: Stem Cell Donor

P704 QUALITY OF ENGRAFTMENT AND INCIDENCE OF ACUTE GVHD GRADE II – IV POST PERIPHERAL HAEMOPOIETIC CELLS CRYOPRESERVATION IN PATIENTS UNDERGOING ALLOGENEIC TRANSPLANTATION FROM HLA IDENTICAL DONORS

Anna Maria Raiola 1, Massimiliano Gambella1, Barabino Luca2, Stefania Bregante1, Carmen Di Grazia1, Alida Dominietto1, Anna Ghiso1, Livia Giannoni1, Alberto Serio1, Silvia Luchetti1, Riccardo Varaldo1, Monica Passannante1, Antonella Laudisi1, Alessandra Scalas2, Paola Contini3, Federico Ivaldi3, Raffaele De Palma3, Alessandra Bo1, Emanuele Angelucci1

1IRCCS Ospedale Policlinico San Martino, Genoa, Italy, 2University of Cagliari, Cagliari, Italy, 3University of Genoa, Genoa e IRCCS Ospedale Policlinico San Martino, Genoa, Italy

Background: Cryopreservation of allogeneic peripheral blood stem cells (PBSC) was commonly employed during the SARS-CoV-2 pandemic. The purpose of this study was to evaluate hematopoietic reconstitution and acute GvHD incidence and severity in patients transplanted from an HLA-identical related (MRD) or unrelated donor (MUD) donor with cryopreserved PBSC.

Methods: We performed a comparative analysis of 46 patients who underwent an allo-HSCT with cryopreserved (cryo group) and 39 patients with fresh (fresh group) apheresis at our transplant unit. The analysis was retrospective for 61 patients transplanted from January 1, 2016 to May 31, 2022 and prospective for 24 patients transplanted from June 1, 2022 to May 30, 2023. The protocol was approved by the Local Ethics Committee (346/2022).

All patients had an HLA-matched sibling or unrelated donor. GVHD prophylaxis consisted of thymoglobulin, cyclosporine and metotrexate.

The primary clinical endpoints were time to engraftment (absolute neutrophil count, platelet count), incidence of graft failure (GF) (lack of full donor chimerism), poor graft function (PGF) (severe cytopenia with full donor chimerism) and acute GvHD. Secondary endpoints were overall survival (OS), event-free survival (EFS), non-relapse mortality (NRM) and relapse incidence (RI). To further characterize immune correlates of clinical outcome, we performed Mass Flow cytometry which allows a detailed characterization of immune cells.

Results: Patient characteristics were comparable between the fresh and cryopreserved group, except for a higher rate of transplants from MUD in the cryo group (p=0.008). Quality parameters of cryopreserved cells (i.e., total nucleated and CD34+ cell enumeration, post-thawing viability) achieved the established requirements for release. One patient who received a fresh graft was not available for engraftment evaluation due to early death.

Median time to neutrophil recovery was longer in the cryo group (18 days) (95%CI: 17-18.9) vs. the fresh group (16 days) (95%CI: 15.3-16.6), (p = 0.007), as for platelet recovery: 20 days (95%CI: 17.8-22.1) vs. 14 days (13.1-14.8) (p= <0.001). For the cryo group GF occurred in 6/46 (13%) patients, while for the fresh group all patients achieved full donor chimerism 1 month after transplant (p = 0.03). Among evaluable patients, 100-days incidence of PGF was 12/40 (30%) in the cryo and 1/38 (2,6%) in the fresh group (p = <0.001).

The cumulative incidence (CI) of grade II - IV acute GvHD was significantly higher in the cryo group, 36.8% (95%CI: 24.9-54.5) vs. 10.2% (95%CI: 4.1-25.9) in the fresh group (p = 0.002). Similar results were observed when the analysis was limited to the MUD subgroup (37% vs. 10%) (p = 0.05). The CI of grade III – IV was 13.2% (95%CI: 6.2-27.8) in the cryo group vs. 2.5% (95%CI: 0.3-17.7) in the fresh group (p=0.04). No differences in terms of OS (p = 0.65), EFS (p = 0.72) RI (p = 0.18) and NRM (p = 0.19) were observed. Of note, by Mass Flow, we found that expansion or loss of given cell subsets correlated with clinical outcomes.

Conclusions: In our experience PBSC cryopreservation was detrimental for hematological reconstitution and acute GvHD incidence. Further studies are ongoing to confirm and explain our data.

Clinical Trial Registry: Local Ethics Committee (346/2022).

Disclosure: no conflict of interest.

10: Stem Cell Donor

P705 WHAT WORKS AND WHAT DOES NOT? THE IMPACT OF DONOR CHARACTERISTICS IN T REPLETE HAPLOIDENTICAL STEM CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE IN CHILDREN

Anuraag Reddy Nalla 1, Revathi Raj1, Ramya Uppuluri1, Venkateswaran Vellaichamy1, Suresh Duraisamy1, Kavitha Ganesan1, Anupama Nair1, Vijayshree Muthukumar1, Indira Jayakumar1, Ravindra Prasad1, Usha Surianarayanan1

1Apollo Cancer Hospitals, Chennai, India

Background: The advent of post-transplant cyclophosphamide (PTCY) has considerably increased the number patients who can benefit from a life saving haematopoietic stem cell transplantation (HSCT). Our study aimed to analyze the impact of donor characteristics on the graft versus host disease and relapse free survival (GRFS) to help make recommendations on optimal donor choice in children undergoing T replete haploidentical HSCT in low-middle-income countries (LMIC).

Methods: We performed a retrospective analysis of patients who had been transplanted at our centre between 2019 to 2023 with a minimum follow up period of 6 months. We included all children had a T replete haploidentical HSCT with PTCY below age 18 years. We targeted a minimum CD34 cell dose of over 7 x 10^/kg and a CD3 dose of over 1.5 x 108/ kg in all our patients. Data collected included the patient diagnosis, age at HSCT, sex and conditioning regimen. The donor data collected included the age, sex, blood group incompatibility, the presence or absence of B leader mismatch and its impact on graft versus host disease, relapse and overall survival. The data was analysed using SPSS software and the study was approved by the Institutional Ethics Committee.

Results: A total of 163 children underwent haploidentical HSCT with PTCY at our centre with male: female ratio of 1:1. The diagnosis was an underlying malignancy in 62 children and a non-malignant disorder in 101 children. The conditioning regimen was myeloablative in 75% (123/163) children and all patients had a peripheral blood stem cell source. The donor was the father in 76% (126/163) children and in 70.6% (115/163) the donor was over 35 years of age. We documented blood group incompatibility in 41% (67/163) children and B leader mismatch in 22% (36/163) children. The graft failure rate was 10.9% (17/163). The incidence of acute graft versus host disease (a GVHD) was low at 7.4% with a higher limited chronic graft versus host disease at 43.6% (p value <0.001). The incidence of relapse in the children with malignancy was 33.8% (21/62).

Sex and blood group mismatch did not impact on overall survival (OS) (p value 0.056). The outcomes with donor age over 35 years were good and 70.5% (115/163) were transplanted using donor over 35 years. The median OS was 45.6 months as compared 36.7 months in donor age less than 35 years of age (p value 0.007). We documented B leader mismatch in 38.1% children and the median OS was 42.1 months in B leader matched donors versus 50 months in B leader mismatched donors (p value 0.008). The overall survival in the cohort was 77% as depicted in the Kaplan Meier curve.

Conclusions: T replete haploidentical HSCT with PTCY offers access to a curative option in children with lethal blood disorders. Our data clearly shows that all children can be offered HSCT with over 75% survival in LMIC with optimal cell dose with albeit with high chronic GVHD. The donor age, sex mismatch, blood group mismatch and B leader mismatch did not impact on the survival.

Clinical Trial Registry: NA.

Disclosure: NOTHING TO DECLARE.

10: Stem Cell Donor

P706 OUTCOMES OF OLDER ADULTS UNDERGOING ALLOGENEIC HEMATOPOIETIC CELL TRANSPLANTATION WITH POST-TRANSPLANT CYCLOPHOSPHAMIDE BASED PROPHYLAXIS

Victoria Murillo Cortés 1, Paola Charry2, María Suárez-Lledó3,4, María Teresa Solano4, Anna Serrahima4, Joan Cid2,4, Miquel Lozano2,4, Laura Rosiñol3,4, Jordi Esteve3,4, Álvaro Urbano-Ispizua3,4, Enric Carreras5, Francesc Fernandez-Avilés3,4, Carmen Martínez3,4, Montserrat Rovira3,4, María Queralt Salas3,4

1Universidad de Zaragoza. Servicio de Hematología y Hemoterapia, Hospital Clínico Universitario Lozano Blesa, Zaragoza, Spain, 2Apheresis and Cellular Therapy Unit, Clinical Institute of Hematology and Oncology (ICMHO) Hospital Clínic de Barcelona, Barcelona, Spain, 3Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Barcelona, Spain, 4Clinical Institute of Hematology and Oncology (ICMHO) Hospital Clínic de Barcelona, Barcelona, Spain, 5Fundació i Institut de Recerca Josep Carreras Contra la Leucèmia, Barcelona, Spain

Background: The landscape of allo-HCT for patients aged 65 and older has evolved significantly over the years. Secondary to its efficacy, using post-transplant cyclophosphamide (PTCY) for GVHD prevention is becoming prevalent across allo-HCT from all donor types. However, the wider and progressive implementation of PTCY has highlighted potential risks such as delayed engraftment, increased infective episodes, and specific organ toxicity.

Considering its wide use and the potential consequences, the safety of using PTCY in older adults has not yet been extensively investigated. This study evaluates the results obtained from using PTCY in adults aged 65 or older from a single institution, and compares the tolerability of PTCY in allo-HCT in the group of patients aged 65 to 69 and aged 70 or older.

Methods: We retrospectively analyzed data from 57 consecutive adults with hematological malignancies aged 65 or older, who underwent their first reduced intensity allo-HCT with PTCY-based prophylaxis between January 2011 and January 2023. For GVHD prevention, patients receiving grafts from 10/10 HLA-matched and 9/10 mismatched donors received PTCY and tacrolimus. Patients undergoing haplo-HCT received PTCY, tacrolimus and mycophenolate mofetil. Key outcomes such as overall survival (OS), relapse-free survival (RFS), non-relapse mortality (NRM), cumulative incidence of relapse (CIR) and GVHD-Free/RFS (GRFS), were analyzed.

Results: Main information of the study cohort grouped according to age ranges is detailed in Table 1. The median age of the cohort was 68 years and 11 patients (18%) aged 70 years or more. Thirty seven (60.7%) patients were males, 14 (23%) had a KPS<90%, and 42 (68.9%) an HCT-CI>3. Acute myeloid leukemia was the most prevalent underlying diagnosis. Transplant baseline information demonstrated uniformity across age categories.

As described in Table 1, 54 (94.8%) patients engrafted, in a median duration of 19 days for neutrophil and 21 days for platelet engraftments. Grade III-IV mucositis (34%), neutropenic colitis (40.4%), hospitalization duration (median 30 days), and readmission rates (56.1%) showed no significant age-related variations. Cumulative incidences of infectious complications, including bacterial bloodstream infections (43.9% in the first 30 days) and CMV reactivation and disease (42.1% and 5.3% at 6 months) showed no differences across age groups. Seven (12.3%) adults required outpatient rehabilitation after HCT discharge.

From Table 1, the cumulative incidences of grade II-IV and III-IV aGVHD at day +180, and extensive cGVHD at 2-years of the 57 patients included were, respectively, 26.3%, 10.5%, and 4.8%. Two-year OS, RFS, NRM, CIR and GRFS were, respectively, 45.5%, 39%, 27,1%, 33.9% and 37%. The differences in the proportions of these transplant outcomes across age groups were not statistically significant.

Table 1 Main Results

All Patients

N=57 (100%)

Patients <70 years

N=46 (100%)

Patients >= 70 years

N= 11 (100%)

P value

Age at alloHCT median, years (range)

68 (65-75)

67 (65-69)

70 (69-75)

<0.001

Sex

Male

37 (60.7)

28 (60.9)

9 (81.8)

0.191

Karnofsky Performance Status

<90%

14 (23)

13 (28.9)

1 (9.1)

0.258

HCT-CI

>=3

42 (68.9)

13 (28.3)

9 (81.9)

0.709

Disease Risk Index

High / Very High

25 (43.9)

21 (45.7)

4 (36.4)

0.739

Donor selection

MSD

3 (4.9)

3 (6.5)

0.701

10/10 HLA MUD

26 (42.6)

20 (43.5)

6 (54.5)

9/10 HLA MMUD

15 (24.6)

13 (28.3)

2 (18.2)

Haploidentical

13 (21.3)

10 (21.7)

3 (27.3)

Median follow-up: median months (range)

14.9 (5.6-33.2)

15.4 (5.1-42)

9.8 (7.2-24)

0.491

Engraftment information

Median days neutrophil engraftment (IQR)

19 (17-24)

20 (17-24)

19 (15-23)

0.767

Median days platelet engraftment (IQR)

21 (14-19)

21 (13-28)

28 (20-34)

0.143

Primary Graft Failure

Yes

3 (5.2)

2 (4.4)

1 (9.1)

0.481

Cumulative incidence of ICU admission [% (95% CI)]

Day +180

21.1 (11.6-32.5)

19.6 (9.6-32.2)

27.3 (5.8-55.2)

0.368

Median duration of transplant hospitalization

Days (IQR)

30 (26-39)

30 (23-37)

29 (23-39)

0.441

Readmission

Yes, n (%)

32 (56.1)

27 (58.7)

5 (45.5)

0.427

Days from HCT discharge to readmission (IQR)

27 (12-155)

25 (12-87)

157 (13-173)

0.516

Cumulative incidence infectious complications [% (95% CI)]

Day +30 Bacterial bloodstream infection

43.9 (30.7-56.3)

45.7 (30.7-59.4)

36.4 (10.1-54.0)

0.656

Day +180 CMV reactivation

42.1 (29.1-54.6)

45.7 (30.7-59.4)

27.3 (5.8-55.2)

0.231

Day +180 CMV disease

5.3 (1.4-13.3)

4.3 (0.8-13.2)

9.1 (0.4-34.8)

0.549

Day +180 VHH6 reactivation / disease

14.0 (6.5-24.4)

10.9 (3.9-21.8)

27.3 (5.7-55.4)

0.171

Day +180 Respiratory viral infection

12.3 (5.3-22.3)

10.9 (3.9-21.8)

18.2 (2.5-45.6)

0.153

Cumulative incidence of GVHD [% (95% CI)]

Day +180 Grade II-IV aGVHD

26.3 (15.6-38.3)

28.3 (16.1-41.8)

18.2 (2.5-45.6)

0.478

Day +180 Grade III-IV aGVHD

10.5 (4.2-20.1)

13.0 (5.2-24.5)

0.211

2-year any grade cGVHD

19.0 (8.7-32.5)

21.3 (9.0-36.9)

10.0 (0.4-37.9)

0.535

2-year Moderate/ Severe cGVHD

4.8 (0.8-14.4)

6.0 (1.0-17.8)

0.467

Median months to immunosuppression

discontinuation (IQR)

6.07(3.07-7.93)

6.43 (3.1-7.9)

5.83 (5.5-6.03)

0.533

Dead

22 (36.1)

16 (34.8)

6 (54.5)

0.887

Causes of death

Relapse

10 (45.5)

7 (43.8)

3 (50)

Infection

7 (31.8)

4 (25)

3 (50)

GVHD

3 (13.6)

3 (18.8)

Graft failure

0 (0)

Other causes

2 (4.1)

2 (12.5)

Overall Survival [% (95% CI)]

2-year

45.5 (31.5-58.6)

47.3 (31.7-61.4)

34.1 (6.5-65.5)

0.776

Relapse-Free Survival [% (95% CI)]

2-year

39 (25.7-52.1)

38.9 (24.3-53.2)

39 (20.6-67.3)

0.771

Non-Relapse Mortality [% (95% CI)]

2-year

27.1 (16-39.4)

24.5 (13-38)

39 (10.3-67.7)

0.437

CIR [% (95% CI)]

2-year

33.9 (21.2-47.1)

36.6 (22.2-51.1)

22.1 (2.2-54.8)

0.318

GRFS [% (95% CI)]

2-year

37.0 (24.4-49.7)

35.7 (21.9-49.7)

42.4 (13.7-69.1)

0.527

Conclusions: The safety of using PTCY-based prophylaxis was similar in the subgroup of patients aged between 65 and 69 years and in the group of patients aged 70 or older. These results provide preliminary evidence that PTCY prophylaxis can be effective also in older patients, challenging previous perceptions. Further analyses on specific risk factors and a larger cohort of patients are needed to refine strategies for this growing demographic group in the allo-HCT setting.

Disclosure: None of the authors declare any conflicts of interest.

10: Stem Cell Donor

P707 COMPARING THE CLINICAL OUTCOMES OF ALLOGENEIC STEM CELL TRANSPLANTATION FROM MATCHED-SIBLING DONORS AND ALTERNATIVE DONORS IN PATIENTS WITH SEVERE THALASSEMIA : SYSTEMATIC REVIEW AND META-ANALYSIS

Sirichai Srichairatanakool 1, Wuttipat Kiratipaisarl1, Sarunsorn Krintratun1, Adisak Tantiworawit1, Chatree Chai-Adisaksopha1

1Chiang Mai University, Chiang Mai, Thailand

Background: The only approved curative modality in thalassemia is allogeneic stem cell transplantation; however, the probability of finding a matched sibling donor (MSD) within the family is less than 30%. Within the past few years, the use of alternative donors of hematopoietic stem cell transplantation in thalassemic patients have been increasingly investigated; however, there are lack of homogeneity of outcomes among different donor types.

Methods: We performed a systemic review and meta-analysis to assess the transplant outcomes, including overall survival (OS), thalassemia free survival (TFS), and transplant-related adverse outcomes such as (graft-versus-host disease (GVHD), graft failure (GF), and treatment related mortality (TRM) between matched-sibling donor compared with alternative donors in patients with severe thalassemia. We searched MEDLINE, Cochrane Central Register of Controlled trials, and EMBASE published in English language would be included. Four authors would review and perform methods following the guidance of PRISMA guideline independently. Our primary objectives were a pairwise, pooled estimate of hazard ratio (HR) with 95% confidence interval for all-cause mortality and thalassemia recurrence comparing between the standard donor source, MSD, to an alternative source, non-MSD.

Results: From 1st January 1964 to 12th September 2023, 9 studies meeting our inclusion criteria were considered in the systematic review. 6 studies in the meta-analysis for OS. The primary outcome of our meta-analysis showed significant reduction in all-cause mortality in MSD group compared with alternative donors (hazard ratio 0.50, 95% CI, 0.31-0.83; p= 0.01, I2=41.11%). In addition, MSD group was associated with a statistically significant reduction in transplant-related adverse outcomes including acute GVHD (RR 0.59, p<0.01), chronic GVHD (RR 0.55, p=0.02), and TRM (RR 0.45, p=0.03).

Conclusions: MSDs remains the preferred donor for transplantation in thalassemic patients with more favourable outcomes compared with alternative donor, associated with less all-cause mortality, acute GVHD, chronic GVHD, and TRM.

Clinical Trial Registry: -.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P708 IMPACT OF DONOR BLOOD GROUP AND CMV MATCH ON ALLOGENEIC STEM CELL TRANSPLANTATION: A SINGLE CENTER LONG TERM FOLLOW UP STUDY

Mohamed Debes1, James Clarke1, Yeong Lim1, Shahid Iqbal1, Leah Credidio1, Hannah Williams2, Jennifer Gibson1, Sajid Pervaiz1, Angela Milner1, Thomas Seddon1, Arpad Toth1, Muhammad Saif 1

1Clatterbridge Cancer Centre, Liverpool, United Kingdom, 2Liverpool Clinical Laboratories, Liverpool, United Kingdom

Background: Limited availability of HLA matched donors in allogeneic stem cell transplant (SCT) often leads to selection of donors who are blood group or cytomegalovirus (CMV) sero-status incompatible with the recipient. The extent to which this effects both short term and long term outcomes remains unclear. We present our single center long term follow up data in allogeneic stem cell transplantation focusing in particular on the impact of donor and recipient ABO/Rh status and CMV sero-status incompatibility.

Methods: We undertook retrospective analysis of patients who underwent allogeneic SCT between 2000 and 2017 (n=446, males=267 females=179). Data were extracted from electronic patient record. Primary end-point was overall survival (OS) at 5 years. We used the GraphPad (Prism) software. Kaplan-Meir’s test was used for survival analysis, log-rank analysis for survival comparison, Fischer’s exact test for comparison of categorical variables and two-sided t-test as well as one way ANOVA for comparing continuous variables in study groups. For multiple co-variates, we used Cox proportional hazard regression analysis and multiple logistic regression where appropriate.

Results: OS at five years in CMV matched group was 44.35% (median survival of 1027 days) vs 34.3% in CMV mismatched group, HR=0.71, CI(0.555 to 0.924) p = 0.007. Our subgroup data analysis show that CMV status had no impact on survival in related donor transplants. In this cohort, median survival in CMV matched transplants was 1340d compared to 1571 days in CMV mismatched transplants (p=0.84). However in unrelated donor subgroup, CMV-matched patients had superior median survival of 593 days compared to 248 days in CMV mismatched group, HR=0.61, CI(0.44 – 0.85), P=0.0021.

Our data show no significant difference in overall survival at five year between ABO/Rh compatible and incompatible transplants (40.95% vs 39.28% p=0.39). However, non-relapse mortality was significantly higher in ABO/Rh incompatible (both major and minor) transplants compared to ABO/Rh compatible transplants (62.32% vs 48.42% p=0.043). Subgroup analysis showed that Rh incompatibility (12.2% of cohort) alone conferred a lower median survival at 1 year compared to ABO/Rh identical group (320.5d vs 365.5d p=0.008). Of note, we also found there was a higher post relapse survival (PRS) at 1 year in ABO/Rh compatible transplants compared to incompatible transplants (44%, median survival vs 28%, p=0.0031). However, at 5 years post relapse, this PRS advantage in ABO/Rh compatible group was no longer observed (14% vs 25% p=0.21).

Conclusions: Our results show that CMV matched transplants had superior overall survival compared to CMV mismatched, though this advantage was only significant in unrelated donor transplants. ABO/Rh incompatibility did not influence overall survival following allo SCT but resulted in a higher NRM. ABO/Rh matching also appeared to confer benefit on OS post relapse though this effect was only observed in the first year following relapse and was abrogated at 5 years. Interestingly, RhD incompatibility was independently associated with a lower overall survival at one year compared to ABO/Rh compatible transplant.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P709 DIFFERENCES IN OVERALL SURVIVAL AFTER ALLOGENEIC HEMATOPOIETIC STEM CELL TRANSPLANT WITH IDENTICAL AND HAPLOIDENTICAL DONORS IN MEXICO

Marco Alejandro Jiménez-Ochoa 1, Elsa Avila-Arreguin2, Severiano Baltazar-Arellano3, María Margarita Contreras-Serratos1, Martha Leticia González Bautista1, Alba Morales-Hernandez2, Uendy Pérez-Lozano4, María Guadalupe Rodríguez González2, Guillermo Sotomayor-Duque3, Elizabeth Urbina-Escalante1

1Instituto Mexicano del Seguro Social, Centro Médico Nacional Siglo XXI, Mexico City, Mexico, 2Instituto Mexicano del Seguro Social, Centro Médico Nacional la Raza, Mexico City, Mexico, 3Instituto Mexicano del Seguro Social, Centro Médico Nacional del Noreste, Monterrey, Mexico, 4Instituto Mexicano del Seguro Social, Centro Médico Nacional Puebla, Puebla, Mexico

Background: Allogeneic hematopoietic stem cell transplantation (HSCT) is a therapy that offers potential cure for hematological malignancies. A limitation is that only 25% of patients will have an identical donor. The use of haploidentical donors allows 95% of patients to have a donor. The experience in Mexico with haploidentical HSCT is limited.

Usually in our country, the most accessible donor option if an identical donor is not available is the haploidentical one. The Instituto Mexicano del Seguro Social has the program in Mexico with the highest national productivity in the number of transplants. The objective is to describe the overall survival of patients undergoing allogeneic transplantation with identical and haploidentical donors in four public health hospitals.

Methods: An observational, retrospective, analytical study was carried out in four Transplant Units. Patients transplanted with malignant hematological pathologies in the allogeneic variety from January 2018 to December 2022 were included. Patients with umbilical cord transplants were excluded. Epidemiological and survival data were taken from the clinical record and recorded in a common database. Statistical analysis was performed with the SPSS 25 program. The group with an allogeneic donor was compared with the haploidentical donor, using Chi2 or Mann-Whitney U depending on the type of variable. For the survival analysis, Kaplan-Meier curves were performed and compared with Log Rank.

Results: 302 allogeneic transplants were performed, 176 (58%) identical and 126 (42%) haploidentical. The median age was 32 and 29 years respectively. The most frequent diagnosis in both groups was acute lymphoblastic leukemia, followed by acute myeloblastic leukemia. 80% of the conditionings were myeloablative. The most frequent scheme in the identical group was BUCY2 (79%) while in the haploidentical group BUCYFLU (61%). Differences were found in the CD34 dose of the identical (6.7 million) and haploidentical (8.2 million) groups, as well as in the median myeloid graft 13 vs 16 days respectively. The median overall survival was not reached in the identical group and was 919 days in the haploidentical group. The median follow-up was 337 days. The most common cause of death in the identical group was relapse (14%), in the haploidentical group infection (17%). There were no differences in the causes of death. Acute GVHD occurred in 48% of the identical group and 43% of the haploidentical group, without differences. Chronic GVHD occurred in 37% of the identical group and 28% of the haploidentical group, being lower in the latter group.

Table 1. Clinical features of hematopoietic stem cell transplanted patients

Variable

Identical (176/58%)

Haploidentical (126/42%)

Significance

Age (years)

32 (16-65)

29 (16-64)

p > 0.05

Male

95 (55%)

78 (62%)

p > 0.05

Diagnosis

ALL

95 (54%)

69 (55%)

p > 0.05

AML

31 (18%)

16 (13%)

p > 0.05

CML

17 (9%)

15 (11%)

p > 0.05

MDS

13 (8%)

9 (8%)

p > 0.05

HL

9 (5%)

17 (13%)

0.01

MM

4 (2%)

NA

Myelofibrosis

3 (2%)

NA

Other

4 (2%)

NA

CD34 Dose

6.7 (1.7-15)

8.2 (1.8-18)

0.01

Day of engraftment

13 (9-32)

16 (13-23)

0.002

Conclusions: Haploidentical donor transplants accounted for 42% of all procedures; overall survival in the identical group was higher than in the haploidentical group. The haploidentical donor is a safe alternative option, when an identical unrelated donor is not available, with an overall survival of 50% of cases and a lower prevalence of chronic GVHD.

Disclosure: The authors declares that they have no relevant material conflict of interest that relate to the research described in this abstract.

10: Stem Cell Donor

P710 ANTI-T LYMPHOCYTE GLOBULIN (ATLG) TO PREVENT GRAFT FAILURE AFTER HAPLOIDENTICAL STEM CELL TRANSPLANTATION WITH DONOR SPECIFIC ANTIBODIES (DSA)

Christian Niederwieser 1, Radwan Massoud1, Evgeny Klyuchnikov1, Gagelmann Nico1, Johanna Richter1, Kristin Rathje1, Tatiana Urbanowicz1, Ameya Kunte1, Janik Engelmann1, Christina Ihne1, Iryna Lastovytska1, Cecilia Lindhauer1, Franziska Marquard1, Mirjam Reichard1, Alla Ryzhkova1, Rusudan Sabauri1, Mathias Schaeferskuepper1, Niloufar Seyedi1, Georgios Kalogeropoulos1, Silke Heidenreich1, Ina Rudolph1, Normann Steiner1, Julia Kienast1, Maroly Bohorquezmanjarres1, Dietlinde Janson1, Christine Wolschke1, Francis Ayuk1, Nicolaus Kröger1

1University Medical Center Hamburg-Eppendorf, Hamburg, Germany

Background: The presence of donor-specific antibodies (DSA) in patients undergoing haploidentical (haplo) hematopoietic cell transplantation (HCT) is a high risk factor for graft failure and/or delayed engraftment. Especially in heavily transfused patients, the presence of DSA is frequent. In this ongoing study, we analyzed the use of anti-T-lymphocyte globulin (ATLG) as part of preconditioning to avoid rejection in patients with detectable donor specific antibodies (DSA) who received HCT from haploidentical donors.

Methods: So far, four patients (median age: 59.9 years), who underwent related haplo-HCT with DSA, median DSA mean fluorescence intensity (MFI) was 7964.61 (range; 1530-16182.99), received ATLG [median total dose 12.5 mg/kg body weight (BW)], given on day -7 (median) as part of the conditioning to reduce the risk of rejection. Conditioning regimen consisted of thiotepa/busulfan (n=1) or fludarabine/thiotepa/busulfan (n=3) and graft-versus-host disease (GvHD) prophylaxis consisted of post-transplant cyclophosphamide (50mg/kg BW on day +3 and +4) and tacrolimus and MMF.

To detect DSA, serum of the patients was tested against donor mononuclear or lineage specific cells (crossmatch), against a panel of mononuclear cells (PRA, panel reactive antibodies) and/or against a solid surface of HLA antigens (Luminex test; Austin, Texas, USA).

Results: As for inclusion, all four patients had DSA tested by serum against solid surface HLA-antigens (Luminex). However, only three patients had a positive PRA test except one patient with HLA-Cw DSA (3rd patient). DSA were found against (ordered by HLA numbering), HLA -A3 (4th patient), -A24 (1st patient), -B13 (2nd patient), -Cw12 (3rd patient),- DR4 (4th patient), -DQB1 0302 (4th patient), but negative for the second HCT (1st patient). All but one (1st patient) showed leucocyte engraftment on day +21 (median) and platelet engraftment in three transplants was reached on day +24 (median). One patient (1st patient) with DSA against HLA-A24 (MFI: 6424.93; 25% of all four patients) rejected the haploidentical graft. The patient was successfully transplanted (day +36) with a one antigen, mismatched unrelated donor, but without DSA and is alive 2.7 years after HCT. Another one [3rd patient; not the one who rejected; DSA in Cw12 (MFI: 1530)], died of infection of viral origin on day +33 after transplantation. The OS, NRM and RI were 75%, 25% and 0%, respectively. Median follow-up of those alive was +415 days.

Conclusions: The preliminary results using ATLG preconditioning resulted in a rejection rate of 25%, which may compare or group favourably to rejection rates, published recently on matched/one-antigen mismatched HCT with conventional GvHD prophylaxis and DSA, of 36.4% [1].

[1] Lima, A. C. M., J. Getz, G. B. do Amaral, G. Loth, V. A. M. Funke, S. K. Nabhan, R. R. Petterle, R. de Marco, M. Gerbase-DeLima, N. F. Pereira, C. Bonfim and R. Pasquini (2023). “Donor-Specific HLA Antibodies Are Associated with Graft Failure and Delayed Hematologic Recovery after Unrelated Donor Hematopoietic Cell Transplantation.” Transplant Cell Ther 29(8): 493.e491-493.e410.

Disclosure: There are no conflicts of interests.

10: Stem Cell Donor

P711 HAPLOIDENTICAL TRANSPLANT WITH POST- CY WITHOUT T DEPLETION. PEDIATRIC EXPERIENCE IN HIGH-RISK LEUKEMIAS FROM GATMO [ARGENTINIAN GROUP OF BONE MARROW TRANSPLANT]

Maria Laura Rizzi 1, Natalia Gonzalez1, Evelin Colombo1, Sol Jarchum1, Emilia Mas2, Victoria Suen2, Sandra Formisano3, Vera Milovik4, Rquel Staciuk5, Pablo Longo6, Silvina Palmer3, German Stemelin3, Silvina Wilberger4, Valeria Santidrian5

1Sanatorio Allende, Cordoba, Argentina, 2Hospital Privado, Cordoba, Argentina, 3Hospital Britanico, Capital federal, Argentina, 4Hospital Aleman, Buenos Aires, Argentina, 5Hospital Garraham, CABA, Argentina, 6Hospital Universitario Austral, Pilar, Argentina

Background: The availability of an ideal histoidentical donor is only 25%, unrelated identical donors are an option, but in situations where the urgency to perform a transplant is imminent, haploidentical transplants emerge as a fast, safe source.

Objectives: To evaluate overall and relapse-free survival.

Methods: A total of 120 patients were reviewed from January 2015 to December 2022 in six centers in Argentina, with an median age of 8.8 years. The study included 50% male and 50% female patients, all with high-risk malignancies: 86 cases of ALL, 26 cases of AML, and 7 other cases underwent Haploidentical Transplant (haplo - HTC) using post-cyclophosphamide 50mg/kg on day+3 + 4 as in vivo T depletion. The source of graft was peripheral blood, bone marrow, cryopreserved peripheral blood, and BM + PB. Disease status was 1CR, 2CR, MRD + >3CR, refractory PR and failure to an unrelated transplant. Donor-recipient sex was 50% match and non-match: being 33 years the average donor age. The statistical analysis InfoStat version 2020 included:Pie graph for population distribution, Chi square independence test, Kaplan Meyer for survival, Multivariate descriptive correspondence analysis.

Results: With a median follow-up of 13.6 months,78 patients (66%)received peripheral blood, and only 25 patients 21%received bone marrowIn 97% of cases had a 100% chimera on day 30. The median white blood cell (WBC) recovery time was 16.7 days, and for platelets, it was 20.5 days. Graft failure occurred in 17.5% of cases. In addition, 35% of patients experienced relapse, and the occurrence of relapse was directly related to disease status, with a p-value of 0.013. Acute Graft vs host disease, GI-II,(a GVHD) was 40%, G III-IV of 8%, and 51% of cases were without GVHD. Chronic GVHD was 21% GII-III. CMV reactivation occurred in 35% of patients. Toxicity was represented by febrile neutropenia, severe infections (including aspergillosis and mucormycosis), hemorrhagic cystitis and others including (VOD, renal failure, septic shock, seizures).50% of patients were alive without relapse at 7.3 months. The progression-free survival time (PFS) was 7.3 months. Overall survival was 50% at a median follow-up of 40 months.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (93)

Conclusions: While this study is retrospective, the results reveals promising findings in a high-risk population, with toxicity comparable to histoidentical and unrelated transplants; and the higher relapse incidence associated with the status of the disease to the transplantation is comparable to other reports. Although in the multivariate analysis, we could not confirm the higher incidence of GVHD associated with the use of peripheral blood as a source, there was a marked tendency towards higher GVHD incidence with the peripheral blood source. HAPLO-PCY is a valid option for pediatric patients without an ideal donor who require a prompt transplant. Prospective follow-up of the data may position haploidentical PCY as an alternative to unrelated and related transplant.

Clinical Trial Registry: not applicable.

Disclosure: I am not disclosures.

10: Stem Cell Donor

P712 DONOR SELECTION CRITERIA IN ALLOGENEIC STEM CELL TRANSPLANTATION FOR ACUTE MYELOID LEUKAEMIA: RETROSPECTIVE SINGLE-CENTER EVALUATION

Ursula La Rocca1,2, Saveria Capria 1, Claudia Lucci1, Roberto Ricci1, Adele Delli Paoli1, Martina Salvatori1, Maurizio Martelli1, Anna Paola Iori1, Walter Barberi1

1Sapienza University of Rome, Rome, Italy, 2Italian National Institute of Health, Rome, Italy

Background: Donor selection criteria are essential for allogeneic hematopoietic stem cell transplant (HSCT) outcomes in patients with acute myeloid leukaemia (AML). HLA identical siblings, available in 25% of cases, still represent the best option. In all the other cases, it is necessary to select alternatives, such as haploidentical family donors, matched (MUD) or mismatched unrelated donors (MMUD). Besides HLA compatibility, CMV donor matching for patient/recipient serostatus, DSAs, stem cell source, donor age and gender, female parity, weight, and ABO match should be considered in the decision-making process whenever more than one donor is available. Even though variants to be preferred are clear, a consensus regarding their hierarchical order to choose the best donor in not well established.

Methods: We conducted a retrospective, single-centre study aimed at evaluating the outcome (OS, DFS) of AML patients’ candidates for allogeneic HSCT from MUD, MMUD or haploidentical donor considering criteria for donor selection (HLA match, stem cell source, ABO match, donor’s age and gender). All consecutive AML patients, stratified according to European Leukemia Network (ELN) 2017, who underwent HSCT from haploidentical, MMUD or MUD from January 2017 to December 2020, at the Hematology Center of Sapienza, University of Rome, were analized. Kaplan-Meier survival analysis of OS and DFS was performed. The log-rank test was used to compare the survival rate between the different categorical variables, and the Mann-Whitney test for continuous-level variables.

Results: Twenty-nine patients (15F/14M) with a median age of 55.6 years (24.1 - 69.2) and a median follow-up of 47 months (range 31-64) were enrolled. The 2017 ELN risk distribution was 17.2% favourable, 48.3% intermediate and 34.5% adverse. Overall, 58,6% underwent HSCT in I complete remission (CR), 20,7% in IRC with positive MRD, 6,9% in IICR, and 13,8% were first-line treatment refractory. The 5-year OS and DFS were 54.4% and 51.7%, respectively. Among the variables analysed, we observed a better trend in terms of 5-years DFS for patients who received a haploidentical donor (72.7%) respect to MUD (41.2%), from a male donor (58.8%) respect to female donors (41.7%), in case of bone marrow (83.3%) respect to peripheral blood stem cell source (43.5%). Overall, no variable had an impact on survival except for ABO match on DFS (p=0.031).

Conclusions: We confirm the role of allogeneic HSC transplantation in AML patients, with OS and DFS exceeding 50%. Donor selection is essential for the success of HSCT, but in our experience only ABO match was significant on DFS. Prospective evaluations with larger samples of patients would be helpful to establish a donor selection algorithm to guide transplant specialists in this critical choice for each disease scenario.

Disclosure: We have no conflicts of interest to disclose.

10: Stem Cell Donor

P713 IRANIAN DONORS SAVE LIVES: COLLABORATING WITH COMMUNITY ADVOCATES TO ADDRESS RACIAL DISPARITY IN ACCESS TO UNRELATED DONORS FOR ALLOGENEIC TRANSPLANTATION

Farnaz Farahbakhsh 1,2,3, Warren Fingrut1,4,5, Bonnie Lu1

1Stem Cell Club, Toronto, Canada, 2Northumbria University, Newcastle Upon Tyne, United Kingdom, 3St. George’s University, True Blue, Grenada, 4Harvard T.H. Chan School of Public Health, Boston, United States, 5University of Texas MD Anderson Cancer Center, Houston, United States

Background: While engaging with underserved communities is critical to address disparities impacting these communities, little work has explored how to collaborate with community advocates to accomplish this goal. Here, we describe a campaign developed with Iranian-Canadian advocates to engage Iranian peoples, an underserved population & largely untapped donor pool, to stem cell donation, & we examine barriers & facilitators to community engagement.

Methods: We collaborated with Iranian-Canadian advocates to develop Iranian Donors Save Lives, a campaign to address racial disparity in donor pools impacting Iranian peoples. English & Farsi multimedia resources were developed to engage Iranian peoples to donation. Multimedia were reviewed for accuracy by a transplant physician & for appeal by Iranian advocates, then published to stemcellclub.ca/Iranian & deployed by advocates to recruit Iranian donors. Focus groups were conducted with Iranian-Canadian young adults, with qualitative & quantitative analyses evaluating the multimedia’s impact on participants’ knowledge & attitudes to donation, & exploring barriers & facilitators to community engagement.

Results: Multimedia developed featured 33 Iranian advocates & included infographics; video testimonials & TikToks starring Iranian advocates & social media influencers; stories & appeals from Iranian patients; & statements from Iranian physicians.

In surveys of 39 Iranian-Canadians (all 17-35 yrs; 49% male), after viewing the multimedia, donation knowledge improved (72% vs 91%, p = .001); ambivalence decreased (47% vs 30%, p = .002); & willingness to register as donors increased (54% vs 77%, p = .032). Qualitative analysis of feedback from focus groups with these participants identified barriers (lack of knowledge of racial disparity; stigma around health; mistrust of healthcare) & facilitators (stories from community members personally impacted; cultural references; use of languages spoken; tailored communication) to community engagement & provided rich examples of how the multimedia overcame barriers & incorporated facilitators to engagement.

In 2023, advocates spearheaded a Nowruz recruitment campaign including 18 drives at Iranian community events across Canada. Ahead of these drives, multimedia resources were shared by Iranian partner groups & amplified by media outlets (ie CBC News, CTV News). Overall, 510 potential donors were recruited.

Conclusions: Through collaboration with Iranian advocates, we developed high quality multimedia which overcame barriers & incorporated facilitators to community engagement, & we spearheaded a national campaign to engage Iranian peoples to donation, recruiting hundreds of Iranian donors. Our work is a model for healthcare organization partnership with community advocates from underserved populations to address health disparities.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P714 INVESTIGATE THE POTENTIAL ASSOCIATION OF HLA ALLELES WITH HEMATOPOIETIC STEM CELL TRANSPLANTATION OUTCOMES

Fatma Al Lawati 1,2, Murtadha Alkhbouri3, Salma Al Harrasi2, Aliya Al Ansari1

1Sultan Qaboos University, Muscat, Oman, 2Royal Hospital/MOH, Muscat, Oman, 3Sultan Qaboos University Hospital, Muscat, Oman

Background: Human leukocyte antigens (HLA) are major determinants of successful hematopoietic stem cell transplantation (HSCT) via their matching status between donors and recipients. Recently, numerous reports revealed that specific HLA alleles could affect transplantation outcomes. This retrospective study aims to investigate the impact of HLA alleles on HSCT outcomes.

Methods: Data were collected for Omani patients registered at Sultan Qaboos University Hospital (SQUH) system, who underwent allogenic HSCT from fully HLA-matched related donors in the period from 2012 to 2022 (n=154). Baseline parameters included date of HSCT, donor and recipient age, gender, blood group and CMV status, stem cell source and dose, conditional regimen, and graft versus host disease (GVHD) prophylaxis.

HSCT outcomes including acute GVHD (aGVHD) and/or chronic GVHD (cGVHD), chimerism status (partial or complete) at 6 to 12 months after transplantation, neutrophil and platelet engraftment, and patient overall survival. All HLA-typing records for patients and donors were at low-resolution level (2 digits) by molecular SSOP method for five HLA loci: HLA-A, B, C, DRB1 and DQB1.

For Statistical analysis, HLA alleles in Omanis with frequency > 5% based on a previous report were considered. There were 9 alleles for HLA-A, 4 for B, 7 for C, 6 for DRB1 and 4 for DQB1. HSCT outcomes were compared among allele positive and negative groups using R (version 4.3.1). GVHD and chimerism were analyzed by regression analysis. Platelet and neutrophil engraftment time were assessed by Mann-Whitney test. Patient overall survival was evaluated by Kaplan-Meier model and Log-rank testing. At 95% confidence interval, p-value threshold was corrected using Bonferroni correction.

HLA-A*

Allele

01

02

11

24

26

30

32

33

68

%

13.6

43

14

14.2

14.9

11

19.4

11

14.9

HLA-B*

Allele

08

35

51

58

%

10.3

26.6

37.7

12.3

HLA-C*

Allele

03

04

06

07

12

15

16

%

14.9

31.1

16.2

27.9

22.7

21.4

18.8

HLA-DRB1*

Allele

03

04

07

11

15

16

%

29.2

13.6

14.9

17.5

14.2

63

HLA-DQB1

Allele

02

03

05

06

%

42.2

26.6

71.4

22.7

Results: The median age for patients was 21 years. The cumulative rate of aGVHD and cGVHD were 16% and 15%, respectively. Thirty-one patients (20%) showed mixed chimerism at least once during the 6 to 12 months after HSCT and three showed complete rejection (2%). The median neutrophil and platelet engraftment times were 14 days and 15 days, respectively.

None of the considered HLA alleles was associated with increase or decrease the risk of cGVHD and/or aGVHD. Also, none of them showed a significant effect on chimerism status. Survival curve analyses indicated a significant protective effect of HLA-DQB1*05 (P=0.010). Patients carrying this allele had a better estimated 5-year overall survival (90%) than did DQB1*05 negative patients (70%). Two other HLA alleles showed a weak impact on neutrophil engraftment time (insignificant after P-value correction). HLA-DRB1*03 was positively associated (P=0.011), while HLA-DQB1*06 had a weak negative effect (P=0.02).

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (94)

Figure: Kaplan-Meier probability of overall survival for DQB1*05- positive and DQB1*05-negative patients.

Conclusions: This study reveals that HLA-DQB1*05 improves patient over-all survival after identical allogenic HSCT. Further studies on larger scale are needed to validate our findings.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P715 SUCCESSFUL OUT-PATIENT SELF-ADMINISTRATION OF MOBILISATION AGENTS FOR HAEMATOPOIETIC STEM CELL TRANSPLANT

Yona Skosana 1, Emma Smith1, Bridged Mofokeng1, Thabo Gcayiya1, Tanya Glatt1,2,3

1South African National Blood Service, Johannesburg, South Africa, 2University of Johannesburg, Johannesburg, South Africa, 3University of Witwatersrand, Johannesburg, South Africa

Background: The Specialised Therapeutic Services (STS) department of the South African National Blood Services (SANBS) is a JACIE-audited Collection Facility (CF). The CF medical director is responsible for prescribing mobilisation agents and managing the mobilisation process for autologous and allogeneic donors. The routine method of mobilisation includes hospital admission of the donor/patient and subcutaneous administration of granulocyte-colony stimulating factor (G-CSF) by a nurse. Inpatient care can be associated with additional cost to the donor and/or recipient, undue discomfort of being away from home and from family as well as increased risk of prolonged admission and hospital-acquired infections. Recently, donors/ patients have been trained to self-administer the G-CSF regimen as outpatients during this mobilisation period. This study will review the efficacy and safety of self-administered mobilisation by donors/patients at our centre.

Methods: All the peripheral blood stem cell (PBSC) collections performed in donors and patients who self-administered G-CSF as outpatients from June 2022 to November 2023 at the Cellular and Immunotherapy Centre (CITC) were included. Donors who were admitted for mobilisation and/or opted to have nursing assistance at home with the administration of G-CSF were excluded. Donors receiving chemotherapy plus G-CSF mobilisation and those receiving Plerixafor were also excluded.

The following data were assessed: type of haematopoietic stem cell transplant (HSCT) (autologous/ allogeneic), gender, age, pre-CD34 count, number of days of collection, CD34 yield, self-reporting of any adverse events (AEs)A donor was defined as being adequately mobilised if they had a pre-CD34 of >20 cells/µl. A target yield is defined as >2 x106/kg.

Donors were allowed to return with the used injections for proper waste disposal by the collection facility. All adverse events were discussed during the consenting process. Analgesia was prescribed as needed.

Results: During this 17 month period, 44 donors and patients had PBSCs of which eight (18.2%) fulfilled inclusion criteria. One (12.5%) was a patient, two (25%) were registry donors and 5 (62.5%) were sibling donors. All donors and patients were adequately mobilised and achieved the target yield (Table 1).

Table 1: Procedure-related results for outpatient self-administered mobilisation agents

Donor

Gender

Age

Recipient

Pre CD34 (cells/µL)

Total CD34 Yield (cells/kg)

Venous Access

Number of days collection

Reported AEs (Y/N)

1

Female

51

Autologous

65

7.4

Central

1

N

2

Male

20

Matched related

70

28.3

Peripheral

1

N

3

Female

74

Matched related

29

4.11

Central

1

N

4

Female

31

Matched related

33

3.6

Central

1

Y (bone pain)

5

Female

28

Matched related

97

7.6

Central

1

N

6

Male

64

Matched related

210

8.5

Peripheral

1

N

7

Male

34

Matched unrelated

194

22.2

Peripheral

1

Y (bone pain)

8

Female

31

Matched unrelated

54; 28

5.1

Peripheral

2

Y (bone pain and headache)

Conclusions: The results above show that outpatient self-administered G-CSF is an effective and safe option in stem cell mobilisation, especially in healthy donors. A directed risk assessment must be performed and adequate training must be given before commencing with the self-administration of G-CSF to ensure donor/patient safety. Donors must also have accessibility to emergency healthcare facilities if required.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P716 ANALYSIS OF BONE MARROW TRANSPLANTATION OUTCOMES BETWEEN JAPANESE AND NON-JAPANESE DONORS AND RECIPIENTS

Ryu Yanagisawa 1, Michiho Shindo2, Akihito Shinohara2, Yachiyo Kuwatsuka3, Koichi Nakase4, Fumihiko Kimura5, Naoki Shingai6, Tetsuya Nishida7, Takahiro Fukuda8, Masatoshi Sakurai9, Mineo Kurokawa10, Takashi Koike11, Shuichi Ota12, Satoru Takada13, Makoto Onizuka11, Tatsuo Ichinohe14, Yoshiko Atsuta15,16, Junya Kanda17, Hideki Nakasone18,19, Tomomi Toubai20

1Shinshu University Hospital, Matsumoto, Japan, 2Tokyo Women’s Medical University, Tokyo, Japan, 3Nagoya University Hospital, Nagoya, Japan, 4Ehime Prefectural Central Hospital, Ehime, Japan, 5National Defense Medical College, Tokorozawa, Japan, 6Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan, 7Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan, 8National Cancer Center Hospital, Tokyo, Japan, 9Keio University School of Medicine, Tokyo, Japan, 10The University of Tokyo Hospital, Tokyo, Japan, 11Tokai University School of Medicine, Isehara, Japan, 12Sapporo Hokuyu Hospital, Sapporo, Japan, 13Saiseikai Maebashi Hospital, Maebashi, Japan, 14Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan, 15Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan, 16Aichi Medical University School of Medicine, Nagakute, Japan, 17Graduate School of Medicine, Kyoto University, Kyoto, Japan, 18Jichi Medical University Saitama Medical Center, Saitama, Japan, 19Jichi Medical University, Shimotsuke, Japan, 20Yamagata University Hospital, Yamagata, Japan

Background: Genetic diversity might be lower in the Japanese population than in other ethnic populations. Therefore, ethnicity might have an impact on outcomes in patients who receive allogeneic hematopoietic cell transplantation from donors of other ethnicities. However, evidence explaining the impact of different ethnicities is limited.

Methods: The Japanese National Database was used to examine the outcomes of first allogeneic bone marrow transplantation (BMT) between Japanese and non-Japanese individuals from 1996 to 2021. Propensity score matching was performed using sex, age, underlying disease, human leukocyte antigen mismatch, conditioning regimen intensity, and year of BMT implementation. Japanese-to-Japanese BMT cases were selected as controls. Overall survival (OS), nonrelapse mortality (NRM), grade II or higher acute graft-versus-host disease (GVHD), chronic GVHD, and neutrophil and platelet engraftment were compared.

Results: The final analysis included 48 and 75 non-Japanese-to-Japanese and Japanese-to-non-Japanese BMT cases, respectively. OS, NRM, acute and chronic GVHD, and neutrophil engraftment were not significantly different between the two groups. However, platelet engraftment was delayed in the Japanese-to-non-Japanese BMT group but not in the non-Japanese-to-Japanese BMT group.

Conclusions: These results suggest that BMT performed between the Japanese and non-Japanese had minimal impact on outcomes. These results may be useful for donor registries and for securing potential donors in the increasingly internationalizing future.

Disclosure: MS owns stock in Celaid Therapeutics.

10: Stem Cell Donor

P717 PSYCHOLOGICAL ASSESSMENT OF HEMATOPOIETIC STEM CELL DONORS UNDERGOING PERIPHERAL BLOOD STEM CELL MOBILISATION

Alka Khadwal 1, Ritwik Lahiri1, Sandeep Grover1, Rekha Hans1, Nabhajit Mallik1, Charanpreet Singh1, Arihant Jain1, Deepesh Lad1, Gaurav Prakash1, Aditya Jandial1, Pankaj Malhotra1

1Post Graduate Institute of Medical Education & Research, Chandigarh, India

Background: The data on related donor safety issues has been limited and, to the best of our knowledge, has not been reported from India, where societal, cultural, educational and awareness level is drastically different to that in developed countries from where most of the data about donor safety issues come. Hence, in this year-long single-centre study, we assessed the psychological status of Indian PBSC donors prospectively.

Methods: All consenting donors >18 years, allogeneic as well as autologous, irrespective of sex, underwent psychological evaluation using Brief Resilient Coping Scale (BRCS), Optimism scale, DUKE UNC Functional Social Support scale Patient Health Questionnaire-9 (PHQ), Generalized Anxiety Disorder-7 (GAD-7), EuroQol 5D-5L scale and Insomnia severity index at baseline and post-donation within a week post donation and after one month.

Results: Thirty donors -median age 38 years, allogeneic =17, 56%, autologous = 13,44%, M: F- 1.5:1, constituted the study cohort. On the Brief Resilience Coping Scale assessment, 36%, 33.3% and 30.0% of donors possessed high, medium and low resilient coping mechanisms, respectively, irrespective of the type of donors (p=0.989). On the optimism scale, 66.7% were optimistic, 23.3% were highly optimistic, and only 10% were pessimistic, with no difference amongst autologous or allogeneic donors (p=0.474). The DUKE UNC Functional Social Support scale assessment revealed that all the donors felt they were receiving ample support from their families and friends. At baseline evaluation on Physical Health Questionnaire-9, minimal depression was found in 63.3%, while one(3.3%) donor had a moderately severe depressive state, and 16.7% each had mild and moderate depression. Significant improvement in depressive symptoms was seen at one week (p=0.00); however, no further significant change was seen (p=0.05)at one month. Autologous donors experienced an exacerbation of depression in both one week (p=0.000) and one month(p=0.001). A majority (76.7%) of donors were minimally anxious at baseline, while 6.7% each had mild and moderate amounts of anxiety, while 10% of donors had severe anxiety. There was an increase in anxiety scores of the donors at one week (p=0.00), and without any increase by one month (p=0.07) and it mainly was contributed by autologous donors at one week (p=0.001) and one month (p=0.000). On the EuroQol 5D-5L scale at baseline, the majority had no problems in mobility (73.3%), self-care (86.7%), usual activities (66.6%), pain (56.7%) and anxiety (70%) with no statistically significant changes noted in the various domains at various study points. Autologous donors did not differ at baseline (p=0.255), but they had significantly more problems in all domains at one week(p=0.000) and one month (p=0.000) post-HSC donation in comparison to allogeneic donors.

Clinically significant insomnia was observed in 16.7% of donors at baseline with no significant change at one week (p=0.02) and one month (p=0.34). Autologous donors had significant insomnia severity at one week (p=0.001) and one month (p=0.001).

Conclusions:

All donors, irrespective of being an allogeneic or autologous donor, face anxiety, stress, insomnia and health-related concerns at baseline as well as post-stem cell donation, even at one month post-procedure, although to a lesser extent in allogeneic donors.

Disclosure: Nothing to declare.

10: Stem Cell Donor

P718 IMPACT AND EFFICACY OF DONOR TYPE IN ALLOGENEIC STEM CELL TRANSPLANTATION IN PATIENTS WITH ACUTE LEUKEMIA, SINGLE CENTER EXPERIENCE

Lazar Chadievski 1, Irina Panovska Stavridis1, Sanja Trajkova1, Nevenka Ridova1, Milche Cvetanoski1, Simona Stojanovska1, Bozidar Kocoski1, Lidija Chevreska1, Svetlana Krstevska Balkanov1, Aleksandra Pivkova Veljanovska1

1University Clinic for Hematology, Skopje, North Macedonia, The Republic of

Background: Performing allogeneic stem cell transplantation in patients with acute leukemia is most of the times an imperative in improving survival rates. But, not always a sibling donor is available, so an alternative must be made with unrelated and haploidentical donors. But is it safe and justified.

Methods: In our analysis we have included 85 patients with acute leukemia treated on the University Clinic for Hematology in Skopje from 2010 to 2022, where an allogeneic HSCT was done. All patients were between 14 and 70 years af age, initially treated with chemotherapy in an attempt to achieve complete remission (CR) before transplant, but even patients that were not in CR and were MRD positive were included. All patients had to have a good performance status, and Karnnofsky score ≥70%.

Results: 49 (57.6%) patients were male and 36 (42.4 %) female. Of all patients, 69 (81.1%) were diagnosed with AML, while 16 (18.8%) with ALL. The median age of the patients was 39.5 years. 80 % of all patients were transplanted in CR, while 20% were not in CR, or were MRD positive before the allogeneic HSCT. The median time from diagnosis till performing the transplantation was 7.66 months. According to the type of donor, in the majority of patients 46 (54.1%) we used sibling donors, in 29 (34.1%) matched unrelated donors (MUD) and in 10 (11.8%) haploidentical donors. In patients with sibling transplantation the rate of some degree of GvHD was 30.4%, while in patinets with MUD was 27.6%. The OS of all patients for 24 months was 55.6%. The OS of patients with sibling allogeneic HSCT was 56.7% for 24 months, in MUD allogeneic HSCT 61.6%, which did not show any statistical significance, but that was not the case in patients with haplodientical HSCT where the OS was 33.3% in 24 months. The disease free survival (DFS) in patients with sibling, MUD and haploidentical HSCT in 36 months was 42%, 63.2% and 30% respectively, and showed a statistical significance in favor of the unrelated donors. The overall transplant related mortality (TRM) was 13.3%. If we analyze the TRM according to the type of donor, in allogeneic HSCT for sibling donor the TRM was 7.2% and in unrelated/haploidentical HSCT was 15% and it was not statistically significant (Log-rank (Mantel-Cox) test, P value = 0,1422, Chi square = 2,154). The incidence of relapse was 28.5% in 24 months for sibling and unrelated donors HSCT.

Conclusions: Allogeneic HSCT is well established therapeutic option and an imperative in treating leukemia patients. In lack of sibling donors, MUD allogeneic HSCT is an adequate alternative providing effective therapeutic approach, making the haploidentical HSCT the least preferable option.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P719 THE EFFECT OF GRAFT MANIPULATION AND CRYOPRESERVATION ON DIFFERENT GRAFT COMPONENT AND ITS POTENTIAL CLINICAL APPLICATIONS

Mohammed Kawari 1,2, Emily Fu1, Shiyi Chen1, Mileidys Alvarez1, Jessica McLeod1, Muhammad Badawi1, Arpita Parikh1, Rashied Kawshermolla1, Racheljihye Kim1, Bramdeo Motiram1, Lynn Jean1, Miyada Himmat1, Lydia Morrison1, Keanu Herzog1, Madhavi Gerbitz3, Megan Nelles1, Eunyoung Cho1, Ahmed Najemeldin1, Igor Novitzky-Basso1,2, Armin Gerbitz1,2,3

1University of Toronto, Toronto, Canada, 2Princess Margaret Cancer Center, Toronto, Canada, 3Philip Orsino Cell Processing Laboratory, Toronto, Canada

Background: Allogeneic hematopoietic stem cell transplant (AlloSCT) is used to treat various medical conditions. While the active substance of a stem cell graft is considered the CD34+CD38- hematopoietic stem cell, successful AlloSCT relies heavily on the presence of functional T cells to facilitate engraftment and ensure graft versus leukemia effects. While fresh grafts are commonly used, the COVID-19 pandemic led to the widespread adoption of cryopreservation for risk mitigation. Concerns were raised about the impact of prolonged travel, graft cell density, and cryopreservation on cell viability and the overall success of AlloSCT.

Methods: At the onset of pandemic restrictions in late March 2020, in Canada, we conducted a prospective analysis of the cellular components of all stem cell grafts on arrival at the institution and after cryopreservation at the Princess Margaret Cancer Center in Toronto. We continued this study after restrictions were lifted until the end of 2022 in the sense of a natural experiment. In addition, we collected data on ex vivo time (time from the end of apheresis to infusion or cryopreservation) and cell concentration in the apheresis bag. Viability analysis was conducted across all cell populations, including hematopoietic progenitors (CD34), monocytes (CD14), T-cells (CD3, CD4, and CD8), B-cells (CD19), and NK-cells (CD56). In total, we analyzed 425 grafts from matched related, haploidentical, and matched or mismatched unrelated donors. During this period 305 were infused fresh, and 120 were infused after cryopreservation.

Full size table

Characteristic

Fresh

Thawed

P value

Ex-Vivo Time in Hours

26.7 (2.2-64.5)

33 (1.6-87.6)

0.43

Donor Age in Years

34 (13-72)

28 (18-65)

<0.001

Donor

Haploidentical

69 (22.6%)

18 (15%)

Matched Related

85 (27.8%)

9 (7.5%)

Matched Unrelated

151 (49.5%)

93 (77.5%)

Results: The median ex-vivo time for fresh grafts was 26.7h (n=305, 2.2-64.5), and cryopreserved grafts were 33h (n=120, 1.6-87.6, p=0.43, Table 1). Upon arrival at our centre, the viability of all cell components was minimally affected by cell concentration in the bag and ex vivo time. However, there was a significant drop in viable cells with cryopreservation, with variable effects on cell type. CD34 was the most tolerant with an overall viability of 87%, while CD3 cells were the most sensitive, with 68.2% viability. Among different donor types, the viability of unrelated donor cells was the lowest which could be explained by the longer ex-vivo time.

Ex-vivo time and cell concentration did not exert a statistically significant impact on CD34 viability, p=0.24 and p= 0.21, respectively. A significant correlation was identified between cell concentration and ex-vivo time, and viabilities of CD3, CD4, and CD8. Higher cell concentration and extended ex-vivo time were ultimately associated with lower T-cell viability, especially post-thaw.

Conclusions: In summary, our findings indicate that the components of peripheral blood grafts are influenced by travel time, cell concentration, and cryopreservation, with variable impact on different cell types. CD34 cells exhibit greater resilience, while T cells are profoundly affected. Moreover, we highlight that extended ex-vivo time and high cell concentration disproportionately affect T-cell viability when grafts undergo cryopreservation. The significant adverse effects on T cell viability due to prolonged ex-vivo time and cryopreservation might explain the higher incidence of poor graft function and delayed engraftment previously reported. Further work includes correlation with clinical outcomes, and post-transplant immune reconstitution of affected cell types.

Disclosure: Nothing to declare for all authors.

2: Stem Cell Mobilization, Collection and Engineering

P720 A SIMPLE PREDICTIVE ALGORITHM FOR ESTIMATION OF THE BLOOD VOLUME TO BE PROCESSED TO YIELD TARGET CELL NUMBERS FOR MANUFACTURING OF TISA-CEL® AND AXI-CEL®

Stephanie Winter 1, Andreas Tanzmann1, Manuela Branka1, Patricija Rajsp1, Philipp Wohlfarth1, Philipp Staber1, Ulrich Jaeger1, Antonia Mueller1, Nina Worel1

1Medical University Vienna, Vienna, Austria

Background: Chimeric antigen-receptor (CAR) T cells are approved for the treatment of adults with relapsed/refractory non-Hodgkin’s lymphoma (NHL), multiple myeloma (MM), and children and adults with acute lymphoblastic leukemia (ALL). In addition, several clinical CAR-T cell trials are ongoing with in parts so far not approved products. CAR-T manufacturers demand starting materials with different target cell populations (e.g. mononuclear or CD3+ cells) and cell numbers. To facilitate the respective target cell collection, we established a simple formula to estimate the blood volume (BV) to be processed to yield defined counts of mononuclear cells (MNCs) or CD3+ cells, respectively.

Methods: Leukapheresis procedures of 99 patients (64% male) from January 2016 to July 2023 were retrospectively analyzed for their MNC (50-100x108 for axi-cel) and CD3+ cell (1-4x109 for tisa-cel) collection efficiency (CE2). All collections have been performed with the Spectra Optia cMNC platform software version 11 and an ACD-A ratio of 1:12.

Based on these retrospective data, a tailored value for CE2 was chosen to avoid undercollection and further used in a prospective approach for the estimation of the BV to be processed to collect the required amount of the respective target cell population.

Results: Patients in the retrospective dataset had a median age of 62 years (range, 21-83) and where diagnosed with ALL (n=2), chronic lymphocytic leukemia (n=4), DLBCL (n=80), follicular lymphoma (FL; n=10), high-grade lymphoma (n=2), and CNS lymphoma (n=1). Patients had a median of 1.5x109/L MNC (range, 0.5-21,2x109/L) and 651x106/L CD3+ (range, 75-3777x106/L) cells before apheresis. The median CE2 was 56% for MNC and 66% for CD3+ cells, respectively. To predict optimal cell yields two simple algorithms were generated. The CE2 was tailored to 50% for MNCs and 60% for CD3+ cells to avoid insufficient collection, while allowing only <30% of patients to have a lower CE2.

Testing these formulas retrospectively in the 99 patients verified its accuracy by comparing the actually processed BV to the estimated BV with a median difference of 11.9% in MNC and 10.2% in CD3+ guided collections. In additional 10 patients (29% male, median age 63 years, range 38-74) with DLBCL (n=7), FL (n=1), mantle cell lymphoma (n=1), CNS lymphoma (n=1) the algorithm was applied prospectively. The prospective cohort had a median of 1.55x109/L MNC (range, 0.11-1.98x109/L) and 477x106/L CD3+ (range, 61-1133x106/L) cells before apheresis and collection procedures achieved similar results with <30% undercollection.

Conclusions: Our collection results by using these two formulas are very promising although deviations due to patient and procedure related factors can negatively impact the CE2. Nevertheless, our algorithm facilitates the daily routine of the operators by generating clear guidance on the calculable duration of the leukapheresis. When CE2 changes over time, the algorithm can easily be adjusted by exchanging the corrected CE2 value in the formulas.

Disclosure: Stepahnie Winter received speakers fees from Kite GILEAD. Nina Worel received speakers fee and honoraria from Novartis and Kite GILEAD.

2: Stem Cell Mobilization, Collection and Engineering

P721 REAL-WORLD MOBILIZATION AND HARVEST OUTCOMES IN NEWLY DIAGNOSED MULTIPLE MYELOMA PATIENTS RECEIVING D-VTD: UK EXPERIENCE

Jose Aspa Cilleruelo 1, Christianne Bourlon2, Elisa Roldan Galvan1, Reuben Benjamin2, Kirsty Cuthill2, Arief Gunawan2, Majid Kazmi3, Pramila Krishnamurthy2, Victoria Potter2, Robin Sanderson2, Michelle Kenyon2, Mili Shah2, Matthew Streetly3, Fabio Serpenti2, Benjamin Robinson1, Andrew Chantry1, Stella J. Bowcock2, Lalita Banerjee4, John Robert Jones5, Sabia Rashid6, Ana Isabel Duran Maestre6, Satyajit Sahu6, Sudarshan Gurung7, Lianwea Chia8, Haili Cui2, Alison Pratt2, Maheen Ahsan2, Stella Bouziana2, John A. Snowden1, Katharine Elizabeth Bailey2

1Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, United Kingdom, 2King’s College Hospital NHS Foundation Trust, London, United Kingdom, 3Guy´s and St Thomas´NHS Foundation Trust, London, United Kingdom, 4Maidstone and Tunbridge Wells NHS Trust, Kent, United Kingdom, 5East Sussex Healthcare NHS Trust, Kent, United Kingdom, 6Lewisham and Greenwich NHS Trust, London, United Kingdom, 7Medway NHS Trust, London, United Kingdom, 8Dartford and Gravesham NHS Trust, London, United Kingdom

Background: In the United Kingdom, The National Institute for Health and Care Excellence (NICE) approved the use of the D-VTd regimen as a first-line treatment for transplant-eligible newly diagnosed multiple myeloma patients (NDMM) in November 2022, establishing it as the standard of care for transplant candidates. The adverse impact of incorporating daratumumab on the mobilization and harvest of haematopoietic stem cell (HSC) outcomes was initially outlined in a subanalysis of the CASSIOPEIA trial, revealing increased utilization of plerixafor (PLX) and reduced HSC yield. However, there is limited availability of real-world data derived from clinical experience after its approval.

Our aim was to provide a service evaluation describing real-world experiences with mobilization, harvest, and autologous stem cell transplantation (ASCT) outcomes in NDMM patients receiving D-VTd.

Methods: All consecutive NDMM patients who completed HSC harvest and ASCT in two transplant centres in the UK were included. Patients received D-VTd as induction and achieved at least a partial remission (PR) before proceeding to ASCT. Mobilization was conducted with either cyclophosphamide 1.5 to 2.0 g/m² (on D + 1) plus G-CSF 5µg/kg (on D + 5 to D + 10) or G-CSF alone, and harvesting occurred from D + 11 or D + 5 respectively, completing 1 to 3 apheresis procedures as needed. The use of PLX was indicated in cases where the CD34+ cell count prior to apheresis was between 5 and 15/µL or in the case of a second mobilization attempt. A successful harvest was defined as ≥2.0x10^6 CD34+ cells/kg.

A descriptive analysis was conducted using Stata 17. Variables are expressed as percentages, median, and interquartile range. Age and procedures required are expressed as the maximum and minimum.

Results: A total of 156 patients were included. The median age was 63 years (range 34-75). 32% of the patients had anaemia at diagnosis, 21% had thrombocytopenia, and additionally, a total of 12 patients (8%) received radiotherapy. MM response pre-apheresis was ≥ very good partial response (VGPR) in 81% (n= 126). Forty-six patients (26%) received additional VTd (n=44) or D-VTd (n=2) cycles before completing mobilization, as a “bridging” strategy. Regarding mobilization and harvest outcomes, the median time from diagnosis to harvest was 28 weeks (24-33). Fifteen patients (9.6%) required a second mobilization attempt. The median total dose of CD34+ cells was 4.3x10^6/kg (2.9-5.8), and the median number of apheresis procedures required was 2 (1-3). The total number of patients who needed PLX was 46 (30%), of whom 67% were preemptive.

The median time from mobilization to transplant was 7 weeks (6-10). All patients achieved neutrophil engraftment in a median of 13 days (13-15) and 20 days (17-25) for platelets. One patient died after engraftment, on day +49, due to pneumonia.

Conclusions: To our knowledge, this is the largest real-world experience reported. Our results confirm that the addition of daratumumab is related to a high rate of PLX use and low HSC yield.

Clinical Trial Registry: N/A.

Disclosure: John Snowden: Advisory board for Jazz, Vertex and Medac, none directly relevant to the abstract.

Elisa Roldan Galvan: Travel epenses from Janssen.

2: Stem Cell Mobilization, Collection and Engineering

P722 FACTORS AFFECTING CD34 + CRUDE COLLECTION EFFICIENCY IN MOBILIZED DONORS UNDERGOING LEUKOCYTAPHERESIS

Jesús Fernandez 1, Monica Linares1, David Gomez-Vives1, Sara Lewandowski2, Hugo Fabre3, Ana Garcia-Buendia1, Marina Rierola1, Veronica Pons1, Julia Ayats1, Juan Porras1, Carmen Montanero1, Teresa Contijoch1, Isabel Amaya1, Esperanza Ribas1, Marc Vilches1, Marta Guerrero1, Alba Acedo1, Sofia Alonso1, Nerea Castillo-Flores1, Sergi Querol1, Rafael Parra1

1Banc de Sang i Teixits, Barcelona, Spain, 2Terumo bct, USA, United States, 3Terumo bct, France, France

Background: Traditional collection efficiency (CE) metrics for leukocytapheresis, such as pre- and post-apheresis cell pool calculations (CE1), or solely pre-procedure cell pool calculations (CE2), fail to consider the dynamic kinetics and variability in CD34+ cell recruitment during the procedure. A more precise measure, termed “crude” CE (cruCE), recently coined by Castillo-Aleman evaluates the harvested cell pool against the total number of cells trafficking outside the bone marrow, inclusive of the pre-procedure CD34+ pool and cells newly recruited during the procedure. This preliminary report highlights factors influencing CD34+ cruCE in mobilized donors undergoing leukocytapheresis for hematopoietic stem cell transplantation (HSCT).

Methods: We retrospectively reviewed leukocytapheresis performed at our institution with the Spectra Optia apheresis device (Terumo BCT) on autologous and allogeneic donors. We considered patient- and disease-specific factors, pre and post peripheral blood (PB) counts, product cell counts, dynamic kinetics and parameters related to apheresis and device alarms to assess their impact on CD34+ cruCE. Secondary, we compared classical CE2 and CE1 versus cruCE.

Results: The analysis included 151 autologous and 59 allogeneic aphereses. The median (range) of CD34 + CE2, CE1 and cruCE was 49 (7-104), 64 (5-125) and 75% (8-99), respectively. The median age of patients was 53 years (range: 0-71). Central catheters were utilized in 55 procedures (26%), and Plerixafor was administered in 35 cases (17%). Table 1 summarizes the analyzed parameters:

Parameter

Median

Range

WBC/µL (pre PB counts)

34000

3000-109000

% MNC (pre PB counts)

18

7 -58

CD34 + /µL (pre PB counts)

39

2 - 584

Hematocrit (pre PB counts)

35

22- 49

Platelets/µL (pre PB counts)

166000

14000 - 432000

CD34 + /kg x10^6 (apheresis product)

4.9

0.2 - 27.0

WBC/µL (apheresis product)

152000

30000 - 385000

% MNC (apheresis product)

80

23 - 100

Hematocrit (apheresis product)

1.5

0.4 - 3.0

Platelets/µL (apheresis product)

809000

49000 - 3361000

Times TBV processed

3.6

1.1 - 7.0

% CD34+ loses after procedure

43

-313 - 95

Total CD34+ recruited x10^8

2.1

-3.3 – 17.6

Recruitment factor (RF)

2.4

0.2 - 6.2

Inlet Pump Flow (mL/min)

50

12 - 91

Collection Pump Flow (mL/min)

1

0.5 - 3.0

Weighted collection preference (CP)

49

35 - 64

Inlet-Pressure Low Alarms

6

0 - 188

% Time Reestablishing Interface

6

0 - 43

In univariate analysis, cruCE significantly correlated with the number of TBV processed (p=<0.001; Pearson=0.694), prior PB counts (WBC/µL: p=0.005; Pearson=0.191; CD34 + /µL: p<0.001; Pearson=-0.454; platelets/µL: p=0.007; Pearson=0.186, % MNC: p=0.001; Pearson=-0.225, hematocrit: p=<0.001; Pearson=0.266), and kinetic measures of CD34+ cells (% CD34 loses: p<0.001; Pearson=0.706, RF: p<0.001; Pearson=0.413). Device settings and alarms showed a negative trend with cruCE (Weighted CP: p=0.084; Pearson=-0.12, inlet pressure low alarms: p=0.079; Pearson=-0.122), although not statistically significant. No differences were observed in relation to mobilization techniques, devices used, or operators.

Classical CE significantly correlated with cruCE (CE2: p<0.001; Pearson=0.368, CE1: p<0.001; Pearson=0.684).

Conclusions: Processing a large TBV associated low CD34+ presence in PB appears to enhance CE, likely due to the increased procedure time and the proportional ease of collecting fewer cells in PB. Elevated PB hematocrit and platelet levels may facilitate the interface, thereby improving cruCE. Other factors like intra-apheresis CD34+ recruitment also augments cruCE, whereas device settings and inlet pressure low alarms may negatively influence it. Multivariate analysis is underway to corroborate these findings.

Lower CE2 and CE1 than cruCE is observed in our series because they assume an unreal higher constant CD34+ pool during whole procedure, in contrast to cruCE that consider CD34+ dynamic kinetics. Therefore, traditional CE2 and CE1 correlate weakly and moderately to cruCE, respectively.

Overall, a high median CD34+ cruCE is evident in our reported leukocytapheresis procedures indicating a very good capacity of CD34+ cell harvesting using this technology.

Disclosure: Lewandowski S: Data Services Manager, Veda Solutions, Global Services, Terumo Blood and Cell Technologies.

Fabre H: Head - EMEA Medical Affairs Terumo Blood and Cell Technologies.

2: Stem Cell Mobilization, Collection and Engineering

P723 EFFICACY OF PROPHYLACTIC ANTIBIOTICS FOR THE PREVENTION OF NEUTROPENIC FEVER IN PATIENTS WITH MULTIPLE MYELOMA RECEIVING HIGH-DOSE CYCLOPHOSPHAMIDE FOR STEM CELL MOBILIZATION

Liqiong Hou1, Juan Li 1

1The First Affiliated Hospital of SunYat-sen University, Guangzhou, China

Background: High-dose cyclophosphamide (HD-Cy) (3 g/m2) plus granulocyte colony-stimulating factor (G-CSF) is a very effective regimen for peripheral blood stem cell (PBSC) mobilization. Unfortunately, it is associated with an increased risk of neutropenic fever (NF). We analyzed the effect of NF on PBSC apheresis results and the efficacy of prophylactic antibiotics for the prevention of NF associated with HD-Cy plus G-CSF for PBSC mobilization in patients with newly diagnosed multiple myeloma (MM).

Methods: First, patients were divided into NF (+)and NF (-) groups according to whether they suffered from NF during mobilization. Second, we divided patients into an antibiotic prophylaxis group and a nonantibiotic prophylaxis group according to whether antibiotic prophylaxis was used during the mobilization period.

Results: Our study showed that NF(+) patients (n=44) had lower CD34+ cell dose collection (median 2.60 versus 5.34 x106/kg, P <0.001) and slower neutrophil engraftment and platelet engraftment (median 11 versus 10 days, P=0.002 and median 13 versus 11 days, P=0.043, respectively) than NF(-) patients (n=234). Of note, the nonantibiotic prophylaxis group patients (n=30) had a 26.7% incidence of NF. In the patients receiving antibiotic prophylaxis (n=227), the incidence was reduced to 9.3% (P =0.01). The antibiotic prophylaxis patients had higher CD34+ cell collection (median 5.41 versus 2.27x106/kg, P< 0.001) and lower hospitalization cost of mobilization (median3,108.02 versus 3702.39; p=0.012).

Conclusions: Thus, our results demonstrate that NF is associated with lower CD34+ cell collection and that antibiotic prophylaxis can reduce the incidence of NF, improve stem cell mobilization and collection outcomes, which reduces the hospitalization cost of mobilization.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P724 EFFICACY OF PLERIXAFOR IN CD34+ STEM CELL MOBILIZATION IN HEALTHY PEDIATRIC STEM CELL DONORS

Rishab Bharadwaj1, Vimal Kumar 1, Deenadayalan Munirathnam1

1Dr Rela Institute and Medical Centre, Chennai, India

Background: Plerixafor’s efficacy in enhancing stem cell collection for patients with hematological malignancies undergoing autologous transplants is well documented, but its potential in healthy pediatric stem cell donors for allogeneic transplants remains largely unexplored. We describe our experience with the usage of single dose of Plerixafor in addition to Granulocyte Colony Stimulating Factor (G-CSF) in healthy pediatric stem cell donors for patients undergoing hematopoietic stem cell transplants.

Methods: A retrospective review of stem cell collection outcomes from donors who received plerixafor as an adjunct to the standard mobilization regimen, was performed. The mobilisation protocol included: (1) G-CSF (10 mcg/kg body weight/day) administered subcutaneously once a day for 5 days, (2) A single dose of subcutaneous plerixafor (0.24 mg/kg BW) on day 4, around 10 hours before apheresis. Peripheral blood CD34+ counts were assessed on the morning of day 5 before apheresis. Apheresis was carried out via a Spectra Optia system, either through peripheral veins where feasible, but more likely through a central venous catheter placed the previous day. Data on donor demographics, CD34+ cell counts in the peripheral blood (before apheresis) and stem cell product (post apheresis), apheresis characteristics and adverse events was collected and reviewed.

Results: The median age of the donors was 10 years (range 2-18 years). Six donors (40%) had an unfavorable Donor/Recipient body weight (D/R) ratio, with a median D/R ratio of 1 (range 0.6-2.1). Minimal side effects such as myalgia and headache were reported in a few patients, which resolved with analgesics. No significant adverse effects necessitation disruption of the mobilization protocol was noticed. The median peripheral blood and stem cell product CD34+ was 216.3 cells/µL (range 70-673 cells/µL) and 1889 cells/µL (range 733-4486 cells/µL) respectively. None of the donors required a second apheresis, with median harvest time being 219 minutes (range 107-326 minutes), and median blood volume processed 3.1 (range 1.2-3.8). No complications were reported during apheresis. The median CD34+ cells x10^6/kg body weight collected was 10.2 (range 5.5-17.3), which surpassed the target for all patients. 11 of the patients (stem cell recipients) who have crossed 100 days post-transplant show no signs of chronic GvHD.

Conclusions: Plerixafor can be used in conjunction with standard G-CSF mobilization protocols for optimizing apheresis outcomes, by boosting the peripheral blood stem cell count on the day of collection. It is safe and well tolerated in the pediatric population. It is postulated to alter the stem cell graft composition favorably, reducing the risk of developing chronic graft-versus-host disease (GvHD).

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P725 PEGFILGRASTIM FOR STEM CELL MOBILIZATION FOLLOWED BY FRESH STEM CELL INFUSION IN MULTIPLE MYELOMA

Hegta Tainá Rodrigues FIgueiroa 1, Pedro Paulo Faust1, Phillip Scheinberg1, Juliana Matos Pessoa1, Fauze Lutfe Ayoub1, André Dias Américo1, Dimitra Delijaicov1, Breno Moreno Gusmão1, André Larrubia1, Eurides Leite da Rosa1, Ana Cynira Franco Marret1, Arlette Edna Lazar1, Germano Glauber de Medeiros Lima1, Isabella Silva Pimentel Pitol1, Fabio Rodrigues Kerbauy1,2

1Beneficência Portuguesa de São Paulo, São Paulo, Brazil, 2Universidade Federal de São Paulo, São Paulo, Brazil

Background: The mobilization of CD34+ cells for autologous hematopoietic stem cell transplantation (aHSCT) can be effectively achieved with a single dose of pegfilgrastim. Amidst the COVID-19 pandemic, this approach was adopted at Beneficencia Portuguesa of São Paulo. The primary goal was to minimize patient contact with healthcare facilities, thereby reducing the risk of SARS-CoV-2 infection. Additionally, we investigated the possibility of eliminating the use of G-CSF post-aHSCT, considering pegfilgrastim’s extended half-life and the implementation of fresh cell infusion.

Methods: We carried out a retrospective cohort study encompassing all initial aHSCT for Multiple Myeloma(MM) conducted at our center since September 2016. This analysis compared patients who received pegfilgrastim 6 mg(PEG) with those who received filgrastim at a dosage of 0.3mg/Kg/day for 5 days (FILG), followed by plerixafor administered ‘just-in-time’ approach. Our pegfilgrastim mobilization protocol, initiated in January 2020, avoided G-CSF pos autograft.

Data regarding mobilization outcomes were gathered, including the initial stem cell count, the total yield of stem cells (expressed as CD34*10^6/Kg), the necessity for plerixafor, the number of apheresis sessions, and the frequency of visits to healthcare facilities. Additionally, key clinical outcomes were recorded, such as the need for transfusions of red blood cells (RBC) and platelets (PA), the duration until patient discharge, and the time to neutrophil engraftment.

Results:

333 mobilizations for HSCT were carried out from September 2016 until December 2022, detailed data on stem cell mobilization were available for 312. Of those 156 received initial filgrastim and 150 received pegfilgrastim, 6 patients received different mobilization regimens (lipegfilgrastim and chemo-mobilization) and were excluded from the present analysis. Mobilization failure was a rare event (1/156vs1/150). First peripheral CD34/mm³ count was higher for FILG (44vs25, p < 0.001), consequently fewer patients required plerixafor (23.2%vs57.3%, p < 0.001) with this approach. Use of PEG was associated with a reduction in number of visits to the apheresis center (6vs8 visits, p < 0.001) and also a reduction in the number of apheresis sessions, 91% of patients required only one session in contrast to 81% of patients receiving filgrastim (p = 0.02). The yield of stem cells was higher for FILG (6*106/Kg vs 5*106/Kg, p = 0.02). Hospital discharge was somewhat faster for patients receiving PEG with more patients being discharged home by day 20 (85%[78- 59] vs 67%[IC95 59-74%] p =0.087), neutrophilic engraftment was similar for both mobilization. Patients receiving PEG needed fewer RBC (0.61vs1.38, p = <0.001) as well as fewer PA (2.12 vs 3.61 p = 0.001). Since the beginning of the pandemic, i.e. March 2020 in Brazil, there were 153 transplants at own center, and 1 patient acquired a covid-19 during mobilization.

General Characteristics

Variable

Filgrastim (n = 155)

Pegfilgrastim (n = 150)

P value

Male, n (%)

78 (48.4)

75(50)

0.8

Age, years median (IQR)

64 (58-68)

63 (55-68)

0.3

Conditioning regimen, n (%)

0.6

Melphalan 200 mg/m²

133 (85.8)

125 (83.3)

Melphalan 140 mg/m²

21(13.5)

24 (16)

Melphalan 100 mg/m²

1 (0.6)

BuMel

1 (0.7%)

ISS, n (%)

0.5

I

48 (40.3)

43 (33.6)

II

31 (26.1)

36 (28.1)

III

40 (33.6)

49 (38.3)

Missing

36

22

Disease Status, n (%)

<0.001

Progressive Disease

1 (0.7)

4 (2.7)

Partial Response

31 (20.5)

29 (19.3)

Very Good Partial Response

90 (59.6)

109 (72.7)

Complete Response

29 (19.2)

8 (5.3)

Missing

4

Outcomes

First CD34 level, cells/mm³, mean, (IC95%)

26 (14- 44)

13 (8 – 25)

< 0.001

Yield CD34 *106/Kg, mean, (IC95%)

6 (4 – 12)

5 (4- 7)

0.002

Neutrophil engraftment by day 20, (IC95%)

92% (86 - 95)

99% (94- 100)

0.2

Hospital discharge by day 20, % (IC95%)

67% (59-74)

85% (78-89)

0.087

Mean RBPC per patient, median, (IQR)

1.38 (0.98- 1.9)

0.61 (0.38 – 0.84)

< 0.001

Mean platelet apheresis per patient, median, (IQR)

3.61 (2.9 – 4.3)

2.12 (1.8-2.5)

< 0.001

Use of plerixafor %, (IC95%)

36 (23%)

86 (57,3%)

< 0.001

Conclusions: Peg-filgrastim provided adequate stem cell yield however required a higher usage of plerixafor rescue compared to FILG and this might have an impact on the number of apheresis session. Health resource utilization reduced modestly with PEG during mobilization. Transplant short term outcomes were comparable without the requirement of GCSF to hasten neutrophil engraftment, and with a reduction in the transfusion requirements needs in the PEG group.

Disclosure: H egta Figueiroa, Fabio Kerbauy., Juliana Pessoa, Isabella Pittol, Ana Marret, André Larrubia., Arlette Lazar, Eurides Rosa, P.edro Faust, Dimitra Delijaicov and Germano Lima. have no conflicts of interest to declare Breno Gusmão has conglicts, working with AMGEN, BMS, JANSSEN, GSK, Pfizer, takeda. Phillip Sheinberg. has been a Scientific Presentations: Novartis, Roche, Alexion, Janssen, AstraZeneca, Grants/Research Support: Alnylam, Pfizer, consultant/Advisory: Roche, Alexion, Pfizer, BioCryst, Novartis, Astellas, speaker: Novartis, Pfizer, Alexion, he declare no equity, stock options, patents, or royalties from any companies.

2: Stem Cell Mobilization, Collection and Engineering

P726 A MID-APHERESIS CD34 COUNT UTILITY IN EARLY STOPPAGE OF APHERESIS IN PATIENTS/DONORS IN A PBSCT PROCEDURE – ANALYSIS OF EXPERIENCE IN TWO SOUTH INDIAN CANCER CENTRES

Veni Prasanna Gedala 1, Rakesh Reddy Boya1, Pradeep Ventrapati1, Anuradha Dasari1, Chandrasekhar Bendi2, Praveena Voonna2

1Apollo Cancer Centres, Visakhapatnam, India, 2Mahatma Gandhi Cancer Institute, Visakhapatnam, India

Background: The aim of the study is to see if doing a mid-apheresis CD34 count would help in early termination of the apheresis procedure without compromising on the final CD34+ stem cell yield. For patients who had a suboptimal collection in mid-CD34, we sought to see if extending procedure beyond Standard Volume Leukapheresis (SVL) is successful in getting adequate stem cell yield in a single session of apheresis.

Methods: Data of patients who underwent apheresis procedures between January 2020 and November 2023 in stem cell units of two tertiary-care cancer centres of south India was analysed retrospectively. Mobilization regimen was G-CSF based, with plerixafor used as pre-emptive strategy. The apheresis machine used was Fresenius Kabi Com. Tec in one unit and Spectra Optia in another unit. The criterion for standard volume leukapheresis (SVL) is processing of 2-3 times of total blood volume (TBV) for donors in an allogeneic transplant setting and 3-4 times of TBV for a patient in the case of autologous PBSCT. Mid-apheresis CD34 count is assessed after 1- 1.5×TBV was processed in allogeneic PBSCT and 2×TBV in autologous PBSCT. Adequate CD34+ stem cell collection was defined as a count of 3×106 CD34+ cells/kg body weight of patients for autologous PBSCT; for matched sibling donor Allo-PBSCT its 4-6 ×106 CD34+ cells/kg body weight and for Haplo-identical allo-PBSCT its 8-10 ×106 CD34+ cells/kg body weight of patient. The strategy to do a mid CD34 count is deemed to be successful if the yield obtained is able to denote an expected adequate collection at double the volume it was checked at AND if the resultant stoppage is earlier than doing the total planned SVL. For patients, who had less than expected mid-cd34, procedure is extended beyond SVL to see if adequate stem cell yield can be collected for a maximum of 6 × TBV.

Results: A total of 124 apheresis procedures were done in the period, 73 of them for auto-PBSCT and 51 for allo-PBSCT. Mid-CD34 count enabled early stoppage in 54 of auto-PBSCT procedures (74%) and in 48 of allo-PBSCT (94%) procedures. For the 19 patients of auto-PBSCT which couldn’t stopped early, extending procedure up to and beyond SVL resulted in adequate yield in 15 patients in the first session itself; 2 patients had successful harvest on the 2nd day and the remaining 2 were truly poor mobilizers. For allo PBSCT setting, extending procedure beyond SVL could salvage the remaining three. The median time of procedure for auto and allo-PBSCT in patients who had successful early stoppage and those who needed extended procedure was shown in table 1.

Table 1: Time taken for procedure and stem cell yield in relation to the procedure time.

Median time of the procedure (min)

Range (in min)

Stem cell yield mean, range (x106CD34+cells/kg body wt)

AUTOLOGOUS PBSCT (n=73)

Successful early stoppage (n=54)

225

118-480

7.15 (4.3-22.6)

Extended procedure (n=19)

295

185-675

4.2 (0.57-5.7)

ALLOGENIC PBSCT (n=51)

Successful early stoppage (n=48)

215

70-470

9.62 (5.1-30.6)

Extended procedure (n=3)

340

210-489

6.85 (5.9-7.9)

Conclusions: Mid-CD34 count has successfully enabled early termination of apheresis procedure without compromising on the CD34+ stem cell yield in the final product, thereby minimising the side effects and discomfort to the patient/donor.

Disclosure: NOTHING TO DECLARE.

2: Stem Cell Mobilization, Collection and Engineering

P727 PLERIXAFOR AS A MOBILIZING AGENT FOR SALVAGE HIGH-DOSE CHEMOTHERAPY IN RELAPSED/REFRACTORY TESTICULAR CANCER: AN EXPERIENCE OF 22 CASES

Sara Bleve1, Maria Concetta Cursano1, Cecilia Menna1, Caterina Gianni1, Giuseppe Schepisi1, Valentina Gallà2, Chiara Casadei1, Serena Mangianti3, Stefano Baravelli4, Accursio Fabio Augello4, Francesco Lanza5, Simona Secondino6, Paolo Pedrazzoli6, Giovanni Rosti1, Ugo De Giorgi 1

1IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy, 2Biostatistics and Clinical Trials Unit, IRCCS Istituto Romagnolo per lo Studio dei Tumori (IRST) “Dino Amadori”, Meldola, Italy, 3Cell Processing Laboratory- PS, Cesena, Italy, 4Morgagni-Pierantoni Hospital, Forlì, Italy, 5Hematology Unit, Ravenna Hospital, Ravenna, Italy, 6Fondazione Istituto di Ricerca e Cura a Carattere Scientifico (IRCCS) Policlinico S. Matteo, Pavia, Italy

Background: Up to 10% of advanced germ cell tumor (GCT) patients (pts) may necessitate high-dose chemotherapy (HDC) supported by stem cells, with cure rates ranging between 20–70%.

Plerixafor plays a significant role in enhancing the success of harvesting CD34+ cells, even in cases where previous attempts at mobilization have failed even if its use is authorized only in poor mobilizer adult/pediatric pts with a diagnosis of lymphoma or multiple myeloma and in pediatric solid tumors. Small retrospective studies and a few case series describe its role in adults with solid tumors. We present a monocentric clinical experience on the use of plerixafor in the mobilization of relapsed/refractory GTC pts.

Methods: We retrospectively analyzed clinical data from pts with relapsed/refractory GCTs candidate to HDC who provided informed consent to undergoing mobilization and collection of peripheral blood progenitor cells (PBSCs) with the use of plerixafor through an off-label program. The monocentric acquisition of data goes from September 2009 to August 2023. Plerixafor was administered to all pts at the dose of 240 ug per Kg body weight to contribute to collecting 6 x 106 CD34+ cells needed to perform a subsequent program of 3 cycles of HDC.

Results: A total of 22 pts were included in our analysis. All pts were affected by relapsed/refractory poor prognosis GCT, with a median of 2.5 prior lines of treatment. All pts failed prior mobilization with approved G-CSF (filgrastim 10 ug/Kg) in combination with chemotherapy. In 15 pts of 22 (68%), leukapheresis was not attempted because of the insufficient number of CD34+ cells in peripheral blood and in 7 pts (32%), an adequate number of CD34+ cells was not achieved for the following HDC program. The most common mobilization chemotherapy regimens administered were paclitaxel-ifosfamide and cisplatin, etoposide, and ifosfamide, selected according to the prior clinical history. Successful PBSC collection was performed in all pts treated with plerixafor and pts obtained a median of 5.4 106 CD34 cells per kg body weight in a median of 2.3 leukapheresis. A subsequent HDC with autologous stem cell support was received from 15 of 22 pts (68%). The median time to leukocyte engraftment was 12.5 days and the median time to platelet engraftment was 14.8 days, consistent with other cases treated with HDC.

Conclusions: This retrospective study represents a single-center experience and the largest data collection on the use of plerixafor as a molecule able to increase PBSC mobilization in solid tumors. These findings, consistent with the reports of previous small series in GCTs, prove the efficacy of plerixafor in these pts who failed previous mobilization therapies and support its use in clinical practice.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P728 ASSESSING AND REFINING PREDICTIVE MODELS OF POTENTIAL POOR MOBILIZERS

Nelson Chan1, Kai Wan 2, Sinju Thomas1, Sandra Loaiza1, Noora Buti1, Sophie Zemenides1, Eitan Mirvis1, Lucy Cook1,2, Edward Bataillard1, Edward Kanfer1, Maria Atta2, Aristeidis Chaidos2,1, Jane Apperley2,1, Edurado Olavarria2,1, Renuka Palanicawandar1

1Imperial College NHS Healthcare Trust, London, United Kingdom, 2Imperial College London, London, United Kingdom

Background: In autologous hematopoietic stem cell transplantation (Auto SCT) for multiple myeloma and lymphoma, a notable 10-25% of patients exhibit poor mobilization (PM) of peripheral blood stem cells (PBSC), requiring additional apheresis or alternative mobilization strategies. Accurate predictive models for potential poor mobilizers (PPM) are critical to optimize mobilization strategies. This study evaluates existing predictive models for their effectiveness in real-life scenarios and aims to develop a refined predictive model based on our in-house data at Hammersmith Hospital, London.

Methods: We analysed PBSC mobilization data from an internal audit undertaken in 2022, of all myeloma patients (n=55) and lymphoma patients (n=21). The predictive models assessed included the Pamplona protocol, the current EBMT Handbook Risk Factors for PPM, and GITMO’s simplified PPM score. PM was defined according to EBMT criteria. The efficacy of these models was evaluated through chi-square tests, sensitivity, specificity, and multiple logistic regression analysis. Bootstrapping was applied to account for small sample size. Additionally, an in-house predictive model was developed and assessed using receiver operating curve (ROC) and area under curve (AUC) analysis.

Results: Among 54 myeloma and 20 lymphoma patients with complete data sets, 24 patients (39.2%) were classified as PM (Table 1). The established predictive models showed limited sensitivity: Pamplona Protocol (sensitivity 6.9%, specificity 93.3%, p-value 1.0), EBMT Handbook risk factors (sensitivity 13.8%, specificity 88.9%, p-value 1.0), GITMO score (sensitivity 13.8%, specificity 71.1%, p-value 0.221). In contrast, our in-house prediction model showed over 80% sensitivity and specificity (sensitivity 82.8%, specificity 82.2%, p-value 0.00017*). In-house analysis identified key predictors of PPM, including age over 60, lack of complete response/very good partial response (CR/VGPR) and exposure to daratumumab. It also suggested that incorporating the International Myeloma Working Group (IMWG) response criteria could enhance predictive accuracy in cases without pre-mobilization bone marrow assessment.

All (n=74)

Proven Poor Mobilizers (n=29)

Non-Poor Mobilizers (n=45)

Relative risk

p-value

Age

Median

59.0

63.0

57.0

Age>60 (n)

17 (23.0%)

10 (34.5%)

7 (15.6%)

61.4

0.004*

Treatment response before mobilization

Non-CR/VGPR (n)

15 (20.2%)

10 (34.5%)

5 (11.1%)

18.8

0.006*

Treatment

Lenalidomide (n)

10 (13.5%)

4 (13.8%)

6 (13.3%)

0.842

0.898

Daratumumab (n)

28 (37.8)

16 (55.2%)

12 (26.7%)

5.707

0.035*

Lines of chemotherapy received

One (n)

56 (75.7%)

24 (82.8%)

32 (71.1%)

0.109

0.058

Two (n)

16 (21.6%)

5 (17.2%)

11 (24.4%)

Three (n)

2 (2.7%)

0 (0%)

2 (4.4%)

Pre-mobilization blood counts (Median)

Haemoglobin (g/L)

119

115

125

0.958

0.062

White cell count (x 10e9/L)

6.15

6

6.7

1.070

0.423

Platelet (x10e9/L)

222

213

234

0.997

0.381

Table 1. Clinical features and multivariate analysis of proven poor mobilizers and non-poor mobilizers. CR, Complete Response; VGPR, Very Good Partial Response. * p-value < 0.05.

Conclusions: This study underscores the limited sensitivity of current predictive models for PM in our patient cohort, emphasizing the need for model refinement. The significant impact of daratumumab, a relatively recent treatment, on PM prediction is a notable finding, as it is not yet incorporated into existing models. Our analysis suggests that an age threshold of 60 is a more effective predictor for PM. The in-house model, integrating IMWG criteria and specific patient characteristics, shows promise for more accurately predicting PPM. However, it requires further validation, particularly given the small sample size and limited exposure to diverse chemotherapy regimens. This study paves the way for more precise and individualized prediction of PM, potentially leading to more effective and tailored mobilization approaches.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P729LEFT OUT IN THE COLD: STEM CELL GRAFTS ARE STILL FUNCTIONAL AFTER MORE THAN 20 YEARS IN CRYOSTORAGE

Rebecca Axelsson Robertson 1,2, Lyda Osorio Fernandez1, Michael Uhlin2

1Karolinska University Hospital, Stockholm, Sweden, 2Karolinska Institutet, Stockholm, Sweden

Background: In autologous transplantation hematopoietic stem and progenitor cells (HSPC) are harvested when patients are in remission, cryopreserved and later reinfused after consecutive treatment strategies. Data is lacking on how extended long-term cryopreservation affects the quality of HSPC. Cytokine production is a parameter that characterize functional potent cells especially in response to viral antigens since infections are a severe risk post HSCT.

Methods: This study assesses the functional state of 21 HSPC grafts in a unique library of grafts cryopreserved for up to 34 years. The samples were divided into 3 groups based on their time in cryopreservation and analyzed with non-parametrical Kruska-Wallis’ test. Functional analysis of the ability of the grafts to produce cytokines (GM-CSF, IFN-γ, IL-10, IL-12p70, IL-13, IL-1b, IL-2, IL-4, IL-5, IL-6, IL-7, IL-8 and TNF-α) in response to 48h of incubation with mitogens and antigens (SEB, CMV, EBV and adenovirus) was evaluated by Luminex.

Results: After up to 34 years in cryopreservation all grafts retained some ability to produce cytokines. In response to SEB the Th1 cytokines IFN-γ, 5230 pg/mL, IL-2, 3113 pg/mL, and TNF-α, 2826 pg/mL were most strongly produced. A significant decrease in the production of TNF-α (p = 0.018), IL-2 (p = 0.018), IL-1b (p = 0.032), IL-4 (p = 0.050), IL-5 (p = 0.002), IL-6 (p = 0.026), IL-10 (0.033), IL-12p70 (p = 0.013) and IL-13 (p = 0.037) between the grafts cryopreserved for <10 years and the older samples could be seen. In response to viral antigens, cytokine production could also be detected for all grafts. The cytokine production was highest in response to CMV antigens with the cytokines TNF-α, 3485 pg/mL, IL-6, 2037 pg/mL and IL-1b, 814 pg/mL most strongly produced. In response to CMV, a significant difference between the grafts cryopreserved for <10 years and older samples could be detected for IFN-γ (p = 0.035), TNF-α (p = 0.042), IL-2 (p = 0.032), IL-8 (p = 0.040) and IL-13 (p = 0.037). In response to EBV a significant difference for IL-2 (p = 0.021) between the youngest and oldest grafts could be detected. It is also worth noting that the production of IL-8 was higher in the samples cryostored for longer time than in the younger samples throughout the assay.

Conclusions: The sustained ability to produce cytokines in all the grafts after cryostorage is a functional marker of retained quality. However, the significant decrease in production of almost all Th1 and Th2 cytokines by cryostorage time, indicates a loss of function or could also be a sign of the decreased viability after thawing and incubation of the grafts previously detected. However, since all grafts showed viable HSPC cells with at least some functional capacity it is, based on this study, hard to identify a time-limit for cryostorage. It may still be wise to use caution if considering using grafts cryostored for more than 20 years or at least as previously suggested harvest higher cell numbers and do extensive testing prior to transplantation.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P730 SAFETY AND EFFICACY OF PLERIXAFOR IN POOR MOBILIZERS: AN INTERIM ANALYSIS AND MULTICENTER PROSPECTIVE STUDY

Weiping Liu1, Yao Liu2, Hui Liu3, Mingxing Zhong4, Zengjun Li5, Caixia Li6, Yuerong Shuang7, Jun Luo8, Jian Zhou9, Jie Ji 10, Sujun Gao11, Wenzheng Yu12, Xiaopei Wang1, Yuqin Song1, Jun Zhu1

1Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China, 2Chongqing University Cancer Hospital, Chongqing, China, 3Beijing Hospital, Beijing, China, 4Ganzhou People’s Hospital, Ganzhou, China, 5Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China, 6National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China, 7Jiangxi Cancer Hospital, Nanchang, China, 8The First Affiliated Hospital of Guangxi Medical University, Nanning, China, 9Affiliated Cancer Hospital of Zhengzhou University and Henan Cancer Hospital, Zhengzhou, China, 10West China Hospital, Sichuan University, Chengdu, China, 11First Affiliated Hospital of Jilin University, Changchun, China, 12Binzhou Medical University Hospital, Binzhou, China

Background: Autologous stem cell transplantation is an important treatment option for lymphoma. Plerixafor, a novel CXCR4 antagonist, can increase the number of circulating haematopoietic stem cells (HSCs) and improve harvest efficacy. Post-hoc analysis of a phase III clinical trial revealed that lymphoma patients with peripheral blood CD34+ cells <10/μl at the time of collection benefited from plerixafor treatment. This study aimed to assess the safety and efficacy of plerixafor in poor mobilizers.

Methods: In this multicenter, prospective, open-label, single-arm clinical study, lymphoma patients with poor mobilization were included. All patients met the following inclusion criteria: 18-70 years old, male or female, ECOG 0 or 1, and eligible for autologous haematopoietic stem-cell transplantation. Poor mobilization was defined as follows: (1) Poor steady-state mobilization: Peripheral blood CD34+ count <10/μL at day 4 when G-CSF (10μg/kg/day subcutaneously) was initiated three weeks or more after the last chemotherapy, (2) Poor chemotherapy mobilization: Peripheral blood CD34+ cells <10/μL when chemotherapy combined with G-CSF mobilization strategy was used, (3) Poor collection: CD34+ cell collection yield <1.0×106/kg at day 1, or <1.5×106/kg at days 1 + 2. All patients received G-CSF 10μg/kg/day and plerixafor injection at 0.24 mg/kg/day for a maximum of 4 consecutive days. The primary endpoint was the proportion of patients who collected at least 2×106 CD34+ cells/kg within 4 apheresis sessions. This trial was registered with clinicaltrials.gov (NCT05510544).

Results: As of October 31, 2023, 70 patients were enrolled. The median age was 49 years (range: 18-67 years); male/female ratio was 1.3:1. A total of 37 (52.86%) patients had poor steady-state mobilization, 25 (35.71%) had poor chemotherapy mobilization, and 8 (11.43%) had poor collection mobilization. A total of 56 patients (80%) reached the primary endpoint. The mean time to reach the target was 2.2 days. The proportion reaching the primary endpoint was 73% in patients with poor steady-state mobilization, 84% in patients with poor chemotherapy mobilization, and 100% in patients with poor collection mobilization. Regarding safety, we observed adverse effects in 42.3% of patients. Most of these patients were classified as grade 1.

Conclusions: Plerixafor plus G-CSF regimen was a tolerable and effective mobilization strategy for poor mobilizers.

Clinical Trial Registry: Clinicaltrials.gov (NCT05510544).

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P731 BONE MARROW HARVESTING IN ALLOGENEIC DONORS: HOW TO PREDICT SUCCESSFUL AND SATISFYING BM PRODUCT

Tigran Torosian 1, Alicja Wozniak1, Grzegorz Hensler1, Katarzyna Szymanska1, Iwona Tomanek1

1Fundacja DKMS, Warsaw, Poland

Background: Bone marrow (BM) collections account for approx. 10% of all donations according to our data. BM is most commonly collected for pediatric patients (66.1%, with 27.9% for children under 5 years old), but collections for adult patients are still in use in certain situations. It is difficult to estimate the expected total count of nuclear cells (TNC), which may be crucial for successful engraftment. The knowledge based on evidence on how to predict TNC count may impact proper donor and source selection.

Methods: We analyzed self-reported collection data from January 2018 to August 2023 (881 BM procedures) and selected those from 5 CCs with more than 40 BM collections performed. A total of 784 results were analyzed. To assess the efficacy of collections, the overall volume and cellularity of products (TNC) were correlated with donors’ gender and weight. Based on product volume and TNC count – the cell concentration was calculated (NC/nl). The results of the concentration of cells were then analyzed, taking as a variable the CC where the collection was performed.

Results: 70% of donors were male. The average weight of men was 83.8 kg, of women 67.0 kg. The mean product volume was 1048 ml (SD 355 ml). The mean TNC was 198.8x108/product (SD 79.0 x 108). The average product concentration was 19.1x108 NC/nl (SD 7.3 x 108 NC/nl).

The assessment of correlation between donor weight and TNC was performed. There was a moderate positive relationship between donor weight and TNC (r= 0.335; p < 0.001). The effect of gender indicated very weak male advantage (r = 0.175, p < 0.001).

We determined also if there was a difference in product NC concentration between CCs. The sample size of groups varied from 477 to 44. There was a difference, revealing, that CCs with the highest number of BM collections had higher results of NC concentrations (p < 0.001).

Conclusions:

  • NC count/kg and concentration per nl of BM are predictable. Thus, donors with higher body weight and acceptable BMI are a better choice for obtaining a satisfactory BM product.

  • Collection Center’s experience is essential for a product with high TNC concentration.

  • For the CC: to calculate the reasonable volume of BM product, knowing the expected NC concentration and requested TNC: BM-Vol (BM volume ml) = requested TNC (x 108) / NC concentration (which is 11-26/nl, with average 20x108/nl product).

  • For the TC: to calculate if the donor is feasible for BM donation: TNC (expected TNC) = donor weight (DW) x average NC/per kg (NC/kg is 1.5-3.5 x 108/kg donor weight, with average 2.5 x 108/kg).

.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P732 COMPARISON OF MOBILIZATION REGIMENS USED IN LYMPHOMA AND FACTORS THAT INFLUENCE PERIPHERAL BLOOD STEM CELL MOBILIZATION

Laxma Reddy 1,2, Anant Gokarn1,2, Akanksha Chichra1,2, Sachin Punatar1,2, Sumeet Mirgh1,2, Nishant Jindal1,2, Lingaraj Nayak1,2, Navin Khattry1,2, Sumathi Hiregoudar1,2, Minal Poojary1,2, Suryatapa Saha1,2, Shashank Ojha1,2, Bhausaheb Bagal1,2

1Tata Memorial Hospital, Mumbai, India, 2Homi Bhaba National Institute, Mumbai, India

Background: Steady-state mobilization using granulocyte colony stimulating factor (GCSF) is the most common method of mobilization. However, GCSF alone fails to mobilize stem cells in 20-30% of lymphoma patients, though the use of plerixafor with GCSF has improved mobilization rates. Use of chemotherapy to mobilize stem cells can reduce failure rates in relapsed/refractory lymphomas. Chemo-mobilization using the same salvage protocol used for remission-induction in relapsed/refractory lymphomas offers advantage of achieving harvest goals without impairing dose intensity of chemotherapy. Primary objective of this study was to compare mobilization success rates (i.e., collection of at least 2x106/Kg of CD34+ progenitor-cells during one mobilization attempt) using different regimens and secondary objective was to analyze factors that influenced mobilization failures.

Methods: This is a single-center retrospective analysis of lymphoma patients who were attempted peripheral blood stem cell (PBSC) mobilization at Tata Memorial Centre, ACTREC, Mumbai from December 2007 to June 2022. Salvage chemotherapy that was being used for treatment of relapse was used as a chemo-mobilization regimen along with GCSF. For patients who had already completed all salvage-chemotherapy cycles, GCSF + /-plerixafor was used. Data of all lymphoma patients who were attempted apheresis was obtained from medical records. Clinical parameters (age, sex, diagnosis, lines of treatment, disease status at time of harvest, regimen used for harvest) and stem cell collection parameters (peripheral blood CD34 cell count on day1 of harvest, number of days of harvest, CD34 cells collected on day1 harvest and total CD34 cells collected) were analyzed and compared. If a patient underwent mobilization attempt twice then each attempt was considered an independent event to assess these factors.

Results: Total of 287 PBSC mobilization attempts were made in 272 patients. Median age was 29 years(5-78 years). Of 287 mobilization attempts, 174 were in Hodgkin lymphoma(HL) patients and 113 attempts in non-Hodgkin lymphoma(NHL). Of these, 243 harvests were successful (84.6%) and 42 (14.6%) were failure (i.e. < 2 × 106 cells/kg collected or apheresis was not attempted due to low peripheral-blood CD34 levels). Most of the patients had between 1 to 3 apheresis sessions in one mobilisation attempt to achieve maximal stem cell yield. For purpose of comparison, regimens were divided into 5 groups (Table.1). Chemo-mobilization with gemcitabine-dexamethasone-platinum(GDP) and cytarabine-based regimen had 95% and 91% successful harvest rates respectively. Ifosfamide-based regimen had highest harvest failures (30%). Proportion of patients with successful chemo-mobilization with GDP and cytarabine-based regimen were significantly higher than ifosfamide-based and GCSF mobilization(p=0,000017). Patients who had received radiation(RT) to mediastinum, spine, or pelvis followed by ifosfamide based mobilization had a failure rate of 48%. Odds-ratio (15.6) of failure of harvest was significantly higher for patients who received RT and ifosfamide-based regimen compared to GDP regimen without RT(p=0.000). No other clinical parameter had any impact on harvest success.

Full size table

.

Conclusions: GDP and cytarabine-based regimen have better mobilization success compared to ifosfamide-based regimen or GCSF (with or without plerixafor) in lymphoma patients. Ifosfamide-based mobilization regimens are best avoided in patients who receive radiation to mediastinum, spine, or pelvis due to high failure rates.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P733 THIRTEEN-YEAR EXPERIENCE WITH PLERIXAFOR IN CHILDREN FROM A SINGLE CENTRE: TOLERABILITY, GRAFT QUALITY AND ENGRAFTMENT

Lucy Metayer1, Charlotte Rigaud1, Valerie Lapierre1, Sylvie Psyche1, Angelique Benoit-Tricoire1, Claudia Pasqualini1, Christelle Dufour1, Kamélia Alexandrova 1

1Gustave Roussy, Villejuif, France

Background: Plerixafor is widely accepted to be safe and effective for adults undergoing peripheral blood stem cell (PBSC) mobilisation, either in steady state or post chemotherapy, but there is less published data concerning children. This single-centre retrospective study aimed to evaluate the safety, efficacy and the quality of engraftment post Plerixafor in a large cohort of children treated for solid malignant tumours and lymphoma, and having apheresis prior to treatment intensification with high-dose chemotherapy (HDC) with autologous stem-cell transplant.

Methods: A single centre retrospective analysis of children ≤18 years old treated for solid tumours receiving at least one dose of Plerixafor for PBSC apheresis at Gustave Roussy between 2010-2023. The patient list was created by combing records from the paediatric stem-cell transplant register, pharmacy dispensation records, and haematology PBSC harvest files. PBSC collections were performed using continuous mononuclear cell collection procedure, with Spectra Optia (Terumo BCT) with an extracorporeal circuit of 250 ml; a blood prime was required for children weighing ≤12kg. Engraftment was defined as neutrophil count >0.5G/L for 48 hours or >1.5G/L once, and for platelets as >50G/L without transfusion in the preceding 48 hours. Data was collected from medical files.

Results: In total, 64 children with a median age of 7 years (range 1–18) were identified. Apheresis was performed in 10 patients under ≤2 years, and 7 weighing ≤12kg.

The most frequent cancer diagnoses were neuroblastoma (23/73, 36%) and Ewing sarcoma (17/64, 27%). The majority (38/64, 59%) received single intensification; 25/64 (39%) received 2 and 1/64 received 3 high dose chemotherapy (HDC) courses.

The indication for Plerixafor was likely failure (low CD34+ prior to apheresis or poor yield on Day 1 of harvest) in 43/64 (67%) patients, or previous failed apheresis attempt in 21/64 (33%). The median total blood volume processed was 3.2 (range 2.5–3.8); median number of apheresis days with plerixafor was 2 (1–4).

An insufficient harvest (<3cx106 CD34 + /kg/autograft) despite plerixafor occurred for 7/64 (11%) patients aged 6–16 years, 5/7 with Ewing sarcoma. The therapeutic plan was modified for 2/7 with single instead of tandem intensification; for 2/7 a decision was made to continue with conventional chemotherapy instead of high dose chemotherapy, and the remainder had a bone marrow harvest to supplement the graft.

In total, 56/64 (88%) patients proceeded to CHD with autograft. The stem cell harvest quality was good; 27/126 (21%) PBSC harvests had >50%PNN affecting 13 patients. Clinical neutrophil and platelet engraftment occurred at a median 12 (range 8–38) and 19 (9–68) days respectfully.

Plerixafor was well tolerated with only 4 patients having significant side effects, all diarrhoea (1 grade 4, 2 grade 3, 1 grade 1). These patients were aged 3–18 years old and had diagnoses of neuroblastoma, Ewing sarcoma and Hodgkin lymphoma.

Conclusions: These results contribute to smaller previously published cohorts demonstrating that not only is plerixafor a well-tolerated and effective treatment, enabling stem cell harvests, even in young and heavily pre-treated patients but clinical engraftment is also acceptable.

Disclosure: No conflicts of interest.

2: Stem Cell Mobilization, Collection and Engineering

P734 AN ALTERNATIVE USE OF A COLLOIDAL SUBSTITUTE (SUCCINYLATED GELATINE) FOR QUALITY CONTROLS IN PERIPHERAL BLOOD STEM TRANSPLANTATION

Claudia Del Fante 1, Eugenio Barone1, Simona De Vitis1, Rosalia Cacciatore1, Daniela Troletti1, Cesare Perotti1

1Cell Manipulation Laboratory, Immunohaematology and Transfusion Service, Fondazione IRCCS Policlinico San Matteo, Pavia, Italy

Background: Quality evaluation of cryopreserved stem cells before transplantation is a cornerstone for transplant success. Notwithstanding the improvement of quality and safety standards, there is still a variable practice among laboratories due to the variability of results obtained from the analysis of thawed samples, related to dimethylsulfoxide (DMSO) cryoprotectant interference, able to lower cell viability. The recently discontinued production of Dextran 40, previously validated in our laboratory as a diluting solution for quality controls on the thawed peripheral blood stem cell (PBSC) samples, forced us to find an alternative solution. We selected succinylated gelatine 4% w/v (Gelofusine, B Braun) after its validation as washing solution during automated DMSO removal from cryopreserved PBSC. Herein we show the results of an in-process validation study using Gelofusine to dilute thawed PBSC samples before cytofluorimetric analysis.

Methods: The study was performed on 6 PBSC, cryopreserved for autologous transplantation. Before the patient’s conditioning regimen, quality controls were performed by flow cytometry (FACS Lyric, BD) using the HISAGE single platform, as per routine. The same sample preparation method, previously validated for Dextran 40, was applied to Gelofusine. Each cryopreserved PBSC satellite vial was thawed in a water bath at 37°C. Then, a 250 microliter sample was immediately diluted with Gelofusine (1:2), incubated for 5 minutes in the dark, and further diluted with PBS buffer; finally, cell count and viability of CD34+ cells, mononuclear cells (MNCs), and total nucleated cells (TNC) were evaluated. At PBSC infusion, additional 6 samples (each from the first bag infused to the patient) were drawn and analyzed using the same method described above. Finally, cell recovery versus the pre cryopreservation samples of CD34+ cells, MNCs and TNC cells on both satellite vial and bag samples at the time of infusion was compared with historical data (Dextran 40 as diluent solution).

Results: Our preliminary results show that Gelofusine can be employed as a diluting solution to reduce DMSO damage after cell thawing. Viable MNCs and CD34+ cells did not significantly differ from fresh leukapheresis products. On the contrary, TNC were significantly lower at infusion compared to the fresh product. Finally, MNCs and CD34+ cell recovery at infusion showed a trend in favor of Gelofusine when compared to historical results obtained with Dextran 40.

Conclusions: Our preliminary results show that Gelofusine is a valid diluting solution for flow cytometric quality controls performed both on the PBSC satellite vials and at infusion. Our data need to be confirmed in a larger study.

Clinical Trial Registry: N.A.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P735 ANTI-CD38 TREATMENT AS A PREDICTIVE FACTOR FOR STEM CELL MOBILIZATION FAILURE AND DELAYED GRAFT RECOVERY IN MULTIPLE MYELOMA: A TERTIARY HOSPITAL EXPERIENCE

Paula Zoco Gallardo 1, Ariadna García Ascacibar1, Juan Mateos Mazón1, Laura Posada Alcón1, Javier Arzuaga Méndez1, Julia Arrieta Aguilar1, Amaia Uresandi Iruin1, Amaia Balerdi Malcorra1, Maria Elena Amutio Díez1, María Casado Sánchez1, Sara Hormaza De Jauregui1, Gorka Pinedo Martín1, Laura Aranguren Del Castillo1, Ana Lobo Olmedo1, Maria Elena Amutio Díez1, Juan Carlos García Ruiz1

1Hospital Universitario Cruces, Barakaldo, Spain

Background: Multiple myeloma (MM) is the leading indication for autologous stem cell transplant (ASCT) worldwide. Administration of high-dose Melphalan followed by infusion of CD34+ cells has been shown to increase depth of response, progression-free survival and overall survival, and is currently the standard of care for elegible patients. This procedure is conditioned by the collection of sufficient stem cells. Therefore, we collected and analyzed variables in our MM patient cohort to identify potential predictive factors for failure of stem cells mobilization. We also investigated how the graft recovery was after ASCT.

Methods: Two hundred and five patients with MM who underwent stem cell mobilization procedure at the University Hospital of Cruces between 2018 and 2023 were retrospectively analyzed. Inclusion criteria were age >18 years, ECOG≤2 and adequate organic function. The target CD34+ cell count was ≥2 x106/kg per planned ASCT. All patients were mobilized with G-CSF at 10μg/kg/12h +/- plerixafor until the day we started leukapheresis, which was when an amount of ≥50 x109/L leukocytes in peripheral blood (PB) or ≥10 CD34 + /μL was reached. We defined mobilization failure as achieving a CD34+ peak <10/μL in PB after 6 doses of GCSF, a yield <2x106CD34 + /kg after 3 consecutive apheresis sessions and/or a decrease in the progressive yield of the procedure, which was the moment we consider adding plerixafor to the mobilization strategy.

Full size table

Results: Sixteen point one percent of our sample (33) were poor mobilizers. Age and previous anti-CD38 treatment were the only factors shown to predict poor mobilization (p-value 0.024 and 0.009, respectively). 41.1% (7) of those treated with anti-CD38 were classified as poor mobilizers, and no differences were found when this variable was adjusted for other complementary therapies received. Anti-CD38 treatment is the only factor we found to be associated with Plerixafor use (p-value 0.006). in addition, we observed slower platelet recovery in poor mobilizers: they needed 1.5 days more than non poor mobilizers (p-value 0.029).

Conclusions: - The results obtained in our study are consistent with the published literature, showing that previous anti-CD38 therapy and age are predisposing factors for poor mobilization of hematopoietic progenitors. This should be taken into account to adjust the mobilization approach and to evaluate the “preemptive” use of plerixafor.

- In our sample, we did not found differences in patient mobilization adjusted for the number of previous cycles of chemotherapy or treatment with Lenalidomide, Thalidomide and/or radiotherapy.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P736 INCREASED DOSE OF G-CSF OVERCOMES AGE AS A RISK FACTOR FOR POOR MOBILIZATION

Yuliya Shestovska 1, Shalina Joshi1, Dzhirgala Mandzhieva1, Regina Mullaney1, Ey Luu1, Rashmi Khanal1, Asya Varshavsky Yanovsky1, Michael Styler1, Henry Fung1, Peter Abdelmessieh1

1Fox Chase Cancer Center - Temple University Hospital Bone Marrow Transplant and Cellular Therapy Program, Phildelphia, United States

Background: Autologous transplantation is a standard treatment modality for many hematological malignancies. The procedure, however, relies on the effective mobilization and collection of CD34+ cells. The most prevalent mobilization regimen combines granulocyte colony-stimulating factor (G-CSF) with Plerixafor and, in some cases, a chemotherapy agent. However, patient characteristics such as advanced age, a history of extensive prior treatments, bone marrow involvement by disease, and reduced bone marrow cellularity have been associated with poor mobilization outcomes.

Methods: We retrospectively reviewed the mobilization outcomes of autologous transplant donors at our center, focusing on two distinct mobilization regimens. For low-risk patients, we used G-CSF at 10 mcg/kg on days 1-5 (total dose 50 mcg/kg) and Plerixafor on day 4, prior to the first apheresis on day 5. High-risk patients received a more intensive regimen: G-CSF at 20 mcg/kg on days 1-4, G-CSF at 10 mcg/kg on day 5 (total dose 90 mcg/kg), and Plerixafor on day 4, with apheresis commencing on day 5. Baseline characteristics of our patients are summarized in Table 1. To evaluate the efficacy of these mobilization strategies, we primarily focused on the CD34+ cell count in peripheral blood collected in the morning prior to the first apheresis. This approach was chosen to measure mobilization efficacy independently of the apheresis procedure, thereby eliminating it as a potential confounder.

Results: Our analysis revealed distinct outcomes based on age groups and mobilization regimens. In patients under 65 years of age, there was no significant difference in mobilization efficacy between the standard and intensified mobilization protocols. However, in patients aged 65 years and older, the intensified mobilization regimen (G-CSF at a total dose of 90mcg/kg + Plerixafor) demonstrated significantly higher efficacy in mobilizing CD34+ cells, as indicated by the pre-apheresis peripheral blood CD34 counts. Consequently, this older age group demonstrated favorable mobilization efficacy outcomes, suggesting an age-specific benefit from the intensified mobilization approach. Patients<65yo who received intensified mobilization had an extremely high risk of poor mobilization due to their primary disease and bone marrow function but still were able to collect an adequate number of cells for safe autologous transplant. Detailed results and statistical analyses are available in Table 1. No severe adverse reactions were observed during mobilization.

Table 1: Baseline Characteristics and Results

G-CSF 90mcg/kg total dose+ Plerixafor

G-CSF 50 mcg/kg total dose+ Plerixafor

Baseline characteristics

n

347

163

Sex M : F (%)

58:42

57:43

Primary diagnosis (%)

PCD

70%

72%

NHL

21%

20%

HL

5%

8%

Solid Tumor

2.5%

0%

Other

1.5%

0%

Age (Median (range))

65 (19-78)

58 (19-72)

Age 65+y.o (%)

52%

17%

Baseline characteristics: patients 65yo+

n

182

28

Sex M : F (%)

54:46

64:36

Primary diagnosis (%)

PCD

75%

75%

NHL

22%

21%

HL

2%

4%

Solid Tumor

0%

0%

Other

1%

0%

G-CSF 90mcg/kg total dose+ Plerixafor

G-CSF 50 mcg/kg total dose+ Plerixafor

p-value

Mobilization efficacy in 65y.o+ patients

Pre-collection CD34 + /µl on D1 apheresis

Average (Range)

100.5 (0-429.23)

68.3 (13.4-152.43)

<.05

CD34 yield (x10^6/ kg) on D1 apheresis

Average (Range)

8.9 (0.1-40.6)

7.0(2-18.1)

Not significant

Mobilization efficacy in <65y.o patients

Pre-collection CD34 + /µl on D1 apheresis

Average (Range)

93.2(0.9-406.6)

110.0 (4.5-533.1)

Not significant

CD34 yield (x10^6/ kg) on D1 apheresis

Average (Range)

7.9(0.2-27.3)

11.3 (0.6-61)

<.01

Conclusions: This study demonstrates that an intensified mobilization regimen involving higher doses of G-CSF and Plerixafor is safe and effective for the older population of patients undergoing autologous transplantation. Notably, this approach appears to mitigate the impact of age as a risk factor for mobilization failure. These findings suggest that adjusting mobilization strategies based on patient age can significantly improve the efficacy of CD34+ cell collection, particularly in older patients.

Clinical Trial Registry: NA.

Disclosure: Asya Varshavsky Yanovsky: Pfizer, Janssen, BMS- advisory.

Henry Fung: Astra Zeneca, Incyte, Kite, Janssen - advisory, honoraria.

Peter Abdelmessieh: AbbVie, Sobi - advisory.

2: Stem Cell Mobilization, Collection and Engineering

P737 THE IMPACT OF TRANSPORTATION TIME ON STEM HEMATOPOIETIC CELL VIABILITY AFTER CRYOPRESERVATION

Iwona Mitrus 1, Marcin Wilkiewicz1, Agata Kawulok1, Wojciech Fidyk1, Małgorzata Sobczyk-Kruszelnicka1, Sebastian Giebel1

1Maria Sklodowska-Curie National Research Institute of Oncology, Gliwice, Poland

Background: The quality of hematopoietic stem cells is important for allogeneic transplantation outcome. In the case of unrelated donors, obtained cells must be transported from collection to the transplant center, which sometimes are located in different countries or on different continents. According to EBMT and WMDA guidelines, the product should be transported between 2-8ºC and delivered within 72 hours after collection, to ensure cell viability. Nevertheless, the quality of graft may decrease during transportation.

During COVID-19 pandemic, allogeneic HSCs (related and unrelated) in our hospital were being frozen and infused thawed; this practice is still used in our center, to increase patient safety. However, the cryopreservation - as well as the transportation time - has negative impact on the viability of HSCs.

The main aim of the study was to evaluate the impact of transport time on stem cell viability after thawing the cryopreserved product.

Methods: This retrospective study included 111 unrelated, cryopreserved grafts, that were used for transplantation between January 2022 and October 2023. Grafts were divided into two groups based on the time of transport (1 day vs 2 days). All products were transported in 4ºC. Overall, 57 of the grafts came from Poland, 39 from Germany, from other 10 European centers, and 5 from the USA.

Obtained cells were cryopreserved according to the local procedures. In short, they were diluted to a concentration of <200 × 106/mL with concurrent plasma or 5% human albumin serum and DMSO to a final concentration of 5%. Next, they were aliquoted into cryobags, frozen in controlled-rate freezers, and stored in liquid nitrogen.

The viability of the cells was measured using trypan blue dye staining after pick-up and a second time immediately after thawing.

Results: The concentration of cells in apheresis products and the time between cryopreservation and transplantation were similar for both groups. However, the median viability of fresh graft was higher in the case of cells transported for 1 day (a median viability of 99%) compared to products transported for 2 days (a median viability of 98%). This difference was statistically significant (p <0.001). Similar results were obtained for thawed products (respectively: median viability 85.5% range: 50.0-99.0% for the first group, and median 81.0%, range: 51-98% for the second group, p=0.006).

Conclusions: The difficulty in collecting and transporting donor cells, especially during the COVID-19 pandemic, caused a change in procedures for transplant centers, including an increased use of cryopreserved allogeneic HPCs. Cryopreservation ensures that the recipient has the graft before the preparative regimen begins. This modification of routine has been analyzed by many centers, however, the results are controversial.

In our results, presented during 48th Annual Meeting of EBMT in 2022, we reported, that cryopreservation of unrelated donor products has no negative impact on clinical outcomes after transplantation. However, the decrease in hematopoietic cell number must be taken into account, especially when long transport is planned.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P738 LENOGRASTIM AND FILGRASTIM: REAL-LIFE COMPARISON IN MOBILIZATION OF ALLOGENEIC HEMATOPOIETIC STEM CELL DONORS

Rosamaria Nitti 1, Filippo Magri1, Alessia Orsini1, Michela Giulia Tassara1, Raffaella Milani1, Salvatore Gattillo2, Milena Coppola1, Luca Santoleri1, Fabio Ciceri3

1Immunohematology and Transfusion Medicine Unit, IRCCS San Raffaele Hospital, Milan, Italy, 2Medical Laboratory, Bolognini Hospital, Seriate, Italy, 3Unit of Hematology and Bone Marrow Transplantation, IRCCS San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy

Background: Lenograstim and filgrastim are granulocyte colony-stimulating factors (G-CSF) used in mobilization of stem cells in healthy hematopoietic stem cell (HSC) donors, however, few real-life comparison data are available.

Methods: We retrospectively analyzed consecutive allogeneic HSC donors at their first donation treated in IRCCS San Raffaele Hospital between 2012 and 2023. According to label, mobilization was performed with lenograstim, substituted by filgrastim whenever unavailable due to drug shortage. Both G-CSFs were prescribed at a dose of 10 μg/kg/day for 5 days, adjusting the last two doses according to day 4 counts. We evaluated white blood cell (WBC), platelet, CD34+ cells absolute counts and CD34+ percentage over WBC count, at day 4 and 5 of G-CSF administration. The detection of CD34+ cells was based on a four-parameter flow cytometry analysis (CD45FITC/CD34PE staining, side and forward angle light scatter). The absolute count was determined with a single-platform method, using Flow Count fluorospheres, with lyse no-wash processing. The ISHAGE sequential Boolean gating strategy was applied to identify CD34+cells. We compared continuous variables with Mann-Whitney test and frequencies with chi-squared test.

Results: We analyzed 588 donors, 526 treated with lenograstim and 62 with filgrastim. There were no differences between the groups for gender (females: lenograstim 202/526, filgrastim 24/62, p 0.96), age (median: lenograstim 37 years, filgrastim 38 years, p 0.17), weight (median: lenograstim 72kg, filgrastim 75kg, p 0.4) or basal ferritin (median: lenograstim 105ng/ml, filgrastim 114ng/ml, p 0.50). At day 4, there were no differences in WBC count (median: lenograstim 40200/μl, filgrastim 42350/μl, p 0.11) or platelet count (median: lenograstim 217000/μl, filgrastim 213000/μl, p 0.93). However, day 4 CD34+ cells were significantly higher in the filgrastim-treated group, both as absolute count (median: lenograstim 37/μl, filgrastim 50/μl, p 0.0069) and as percentage over WBC count (median: lenograstim 0.09%, filgrastim 0.11%, p 0.03).

Donors were classified as poor mobilizer at day 4 if CD34+ were <20/μl and plerixafor was administered on-demand on day 5 to 10 donors, in an equal fraction in the two groups (lenograstim 9/526, filgrastim 1/62, p 0.94); these donors were excluded from day 5 counts analysis as only post-plerixafor counts were available.

At day 5, there were no differences in WBC count (median: lenograstim 45200/μl, filgrastim 44800/μl, p 0.88) or platelet count (median: lenograstim 206000/μl, filgrastim 214000/μl, p 0.6). At day 5, CD34+ cells were significantly higher in filgrastim-treated donors as percentage over WBC counts (median: lenograstim 0.17%, filgrastim 0.18%, p 0.027), while their absolute counts were no longer significantly higher (median: lenograstim 74/μl, filgrastim 80/μl, p 0.057).

Then analysis on AEs on both groups showed no difference on reported events.

Conclusions: Lenograstim and filgrastim have comparable effects on WBC and platelet counts at day 4 and 5 of administration. Day 4 CD34+ cells counts are higher during filgrastim treatment, but this difference decreases at day 5, suggesting a possible slower effect of lenograstim, or a successful modulation of the last G-CSF doses according to day 4 counts. Filgrastim should still be considered a valid alternative option to lenograstim as mobilizing agent in normal donors.

Disclosure: None related to this work.

2: Stem Cell Mobilization, Collection and Engineering

P739 NIVOLUMAB-IFOSFAMIDE-CARBOPLATIN-ETOPOSIDE (NICE) AS MOBILIZING REGIMEN FOR RELAPSED/REFRACTORY HODGKIN LYMPHOMA (R/R HL)

Maria K. Angelopoulou1, Eleni Lalou1, Athanasios Liaskas 1, Aikaterini Benekou1, Maria-Aikaterini Lefaki1, Ioannis Vasilopoulos1, Angeliki Georgopoulou1, Chrysovalantou Chatzidimitriou1, Alexandros Machairas1, Alexia Piperidou1, Kalliopi Zerzi1, Konstantinos Keramaris1, Marina P. Siakantaris1, Panayiotis Panayiotidis1, Theodoros P. Vassilakopoulos1

1National and Kapodistrian University of Athens/ School of Medicine, Athens, Greece

Background: Salvage chemotherapy followed by autologous hematopoietic stem cell transplantation (ASCT) remains the treatment of choice for transplant-eligible r/r HL. Ifosfamide-based (IGEV, ICE) or platinum-based (ESHAP, DHAP) regimens have been traditionally used as salvage chemotherapy, based both on their stem cell mobilizing potential and favorable disease control prior to ASCT. Over the last few years, novel agents such as brentuximab vedotin and PD-1 inhibitors have been incorporated into salvage regimens.

Methods: We have implemented NICE as salvage treatment for r/r HL patients since April 2023, following approval by the Greek Health authorities based on excellent reported outcomes in moderately-sized phase 2 trials. We report the mobilizing potential and kinetics of the first 10 consecutive patients mobilized with NICE. NICE consisted of: Nivolumab 240 mg day 1, etoposide 100 mg/m2 days 1-3, carboplatin AUC 5 (5 × [25 + CrCl] (750 mg maximum) day 2, ifosfamide 5000 mg/m2 day 2 with MESNA.

Results: Six patients were females and four males. All except one had received ABVD as 1st line treatment. Six patients had primary refractory disease, three had early and one late relapse. The distribution of clinical stage at relapse was as follows: IIA (three patients), IIB (one), IIIA (three), IVA (two) and IVB (one). Their median age at mobilization was 25.5 years (21-50). According to NICE protocol, 9/10 received one cycle of Nivolumab two weeks prior to 1st NICE cycle. One patient received five Nivolumab cycles before commencing NICE due to practical issues. Six patients were mobilized after the 2nd cycle of NICE, two after the 1st and two after the third. At the expected day of maximum mobilization (day 12), the median circulating CD34+ counts were 17/μL (1.08-92.1); only 3/10 patients achieved CD34+ counts >20/μL (institutional threshold) and were subjected to apheresis. One patient (#2) was considered as complete mobilization failure. Plerixafor was necessary for six patients (67%) in order to proceed with apheresis on the following day. Median day of peak CD34+ counts was +13. Median CD34+ collected cells was 6.85x106/kg (0-9.9x106). Two patients were collected for two days and two patients (#2, #3) subsequently underwent a second mobilization course with cyclophosphamide and achieved high circulating CD34+ counts (42/μL and 80/μL) and successful stem cell collection (7.2 and 5.2X106/kg). Table 1 depicts mobilization kinetics. All eight assessed patients had a complete metabolic response after NICE and proceeded to ASCT; in two patients response evaluation is pending. Historical controls treated in our Institution with salvage ifosfamide-containing regimen IGEV had median circulating CD34+ counts 198.6/μL and the number of total CD34+ collected cells was 11.6x106/kg without the use of plerixafor (Angelopoulou, et al, EHA 2010).

Table 1.

Mobilization kinetics after NICE chemotherapy

Patient #

Day 12 circulating CD34+ counts (/μL)

Plerixafor

Peak day

Peak circulating CD34+ counts (/μL)

Collection day

CD34+ cells collected (X106/kg)

1

21.6

Yes

13

57.35

13

6.59

2

1.08

No

13

3.52

NA

3

2.92

Yes

14

19.41

13, 14

2.76

4

17.50

Yes

13

135.39

13

9.90

5

6.65

Yes

14

24.31

13,14

4.49

6

16.56

Yes

13

91.12

13

7.10

7

50.79

No

12

50.79

12

7.40

8

13.25

Yes

13

54.95

13

7.49

9

92.10

No

12

92.10

12

8.02

10

34.19

No

12

41.39

13

6.28

Conclusions: Our preliminary data confirm the excellent disease control achieved by NICE salvage therapy for r/r HL patients prior to ASCT, but also raise a concern that it might be associated with reduced mobilizing capacity and the need of plerixafor in order to achieve an acceptable stem cell collection. A potential negative impact of PD-1 inhibitors on stem cell mobilization needs to be further studied.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P740 IMPACT OF DARATUMUMAB, BORTEZOMIB, THALIDOMIDE DEXAMETHASONE INDUCTION THERAPY ON STEM CELL MOBILIZATION: A REAL WORLD EXPERIENCE

Giuseppe Sapienza 1, Marika Porrazzo1, Fabrizio Accardi1, Roberto Bono1, Stefania Tringali1, Cristina Rotolo1, Cirino Botta2, Caterina Patti1, Alessandra Santoro1, Laura Di Noto1, Lucia Sbriglio1, Luca Castagna1

1Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy, 2Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone”, Palermo, Italy

Background: The clinical benefit in adding daratumumab to the VTD-induction-therapy in the newly-diagnosed-multiple-Myeloma (NDMM) was widely demonstrated in the phase-III-CASSIOPEIA-study. The emerging concern related to the addition of daratumumab to a triplet backbone is its negative impact on stem cell mobilization. Data from the largest phase-3 trials CASSIOPEIA-(DaraVTD-vs-VTD) and phase-2-MASTER (DaraKrd) and GRIFFIN-(DaraRVD-vs-RVD) showed a lower stem cell yield and higher plerixafor use, independently from the type of mobilization therapy used (cyclophosphamide + GCSF or only G-CSF).

The aim of this multicentric, retrospective study was to describe our experience on stem cell mobilization with DARA-VTD in a real life setting.

Methods: Patients with NDMM treated with DARA-VTD were included in this analysis. Stem cell mobilization performed with G-CSF 10 mg/day with leukapheresis planned at day +5. The minimum target of CD34+cells was 2.5 x 106 cells/kg for 1 ASCT and 5.0 x 106 cells/kg when 2 ASCT were planned. Plerixafor was used on demand when the number of circulating CD34+ cells was less than 20 mcl/L at day +5 or when the number of CD34 harvested was less than 2.5 x 106 cells after the first leukapheresis. Categorical data are presented as count with percentage, and continuous data are presented as median with range. Statistical comparisons were done using the chi square or Fisher exact test for categorical variables and the Mann-Whitney-U-test for continuous variables. NCSS-2019-version was used for statistical analysis, and a P value <.05 was considered to indicate statistical significance.

Results: From 2022, 40 patients were included. Median age was 61 (41-71). Most of the patients were mobilized after the third (52%) and fourth course (42%). Before mobilization, 31 (77%) patients were in VGPR and 9 (23%) in CR. The median number of CD34+ cells harvested was 4 (0-11) x 10e6/Kg. 10% pts. failed the first mobilization attempt. 50% and 45% of patients collected stem cells with 1 and 2 apheresis respectively. 22% patients required plerixafor use. Stem cell mobilization was performed after a median of 21 days after last daratumumab administration. There was no difference in total CD 34 yield between patients mobilized after more and less of 21 days after last daratumumab infusion (p=0,57). Median CD 34 /Kg total yield was 4.7 and 3.7 x 10e6/Kg for patients harvested after 3 and 4 cycles of induction respectively (p=0.07). After high dose Melphalan, the median day to achieve ANC more than 500 and PLTS>20.000 was 12 (6-24) and 13,5 (7-26) respectively. With a median follow up of 175 days, 1 year OS was 94%. 6 patients had planned two ASCT before mobilization and only 4 patient underwent second ASCT.

Conclusions: Our retrospective analysis showed a low rate of mobilization failures after dara vtd induction. We furthermore found no difference on CD 34 total yeald after 3 or 4 cicles of dara VTD and when mobilization was performed after more or less of 21 days ater last daratumumab administration.

Prospective trials are needed to confirm feasibility of chemo free G CSF stem cell mobilization after Daratumomab VTD induction.

Disclosure: No disclosures to declare.

2: Stem Cell Mobilization, Collection and Engineering

P741 INSTITUTIONAL THRESHOLD OF PREAPHERESIS PERIPHERAL BLOOD CD34+ CELL COUNT: A SINGLE-CENTER EXPERIENCE

Yandy Marx Castillo Aleman 1,2, Carlos Agustin Villegas Valverde1, Antonio Alfonso Bencomo Hernandez1, Yendry Ventura Carmenate1,2, Fatema Mohammed Al Kaabi1,2, Shinnette Lumame2, Charisma Castelo2, Nameer Abdul Al-Saadawi1,2, Inas El-Najjar2, Mohamed Ibrahim Abu Haleeqa1,2, Jayakumar David Dennison2, Mansi Sachdev2, Fatma Abdou1, Shadi Shamat1, Anil Kumar Sarode1, Dina El Mouzain1,2, Hiba Mohamad Abdelrahman2, Rene Antonio Rivero Jimenez1

1Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates, 2Yas Clinic Khalifa City, Abu Dhabi, United Arab Emirates

Background: Preapheresis peripheral blood CD34+ (PB-CD34+) cell count is widely recognized as a predictor of the number of harvested hematopoietic progenitor cells (HPCs). However, there is currently no consensus on the minimum threshold of PB-CD34+ cells required for apheresis, and institutional practices vary from 5 to 20 cells/μL. To optimize apheresis resources and mitigate potential adverse events in cases of predicted low yields, we reviewed our institutional threshold (15 cells/μL) as a starting point for HPC apheresis.

Methods: Apheresis procedures conducted between January 1, 2023, and November 30, 2023, were included in this analysis. The retrospective data collection was exempted from ethics review by the Institutional Review Board. The data collected included the patient’s diagnosis, type of transplant (autologous or allogeneic), PB-CD34+ counts immediately before and after apheresis, CD34+ cell count in the apheresis bag, dose of CD34+ cells per kilogram of the recipient’s body weight achieved, apheresis system, total blood volume processed, and product volume to calculate the fold enrichment (FE) and the collection efficiency (CE).

Results: A total of 64 aphereses were conducted using the Spectra Optia® apheresis system (Terumo BCT, Japan) in autologous (n = 52) or allogeneic (n = 12) settings. Table 1 summarizes the comparison of procedures in terms of preapheresis PB-CD34+ cell count, cell count in the apheresis bag, dose achieved per apheresis, FE, and CE.

Table 1. Comparison of autologous and allogeneic procedures in terms of CD34+ cell counts and collection efficiency

Parameters

Autologous

Allogeneic

Mann-Whitney test

Total

Median

(2.5 - 97.5 percentile)

Median

(2.5 - 97.5 percentile)

Pre-apheresis peripheral blood CD34+ cell count (cells/μL)

34 (6 - 434)

52 (28 - 123)

*0.0392

38 (7 - 291)

Product CD34+ cell count (cells/μL)

780 (96 - 4987)

988 (536 - 5010)

0.1821

880 (113 - 5236)

CD34+ cell dose (x 106 cells/Kg of recipient’s body weight)

3.30 (0.20 - 23.24)

4.45 (2.54 - 14.68)

*0.0419

3.68 (0.24 - 19.49)

CD34+ fold enrichment (FE)

23.08 (7.00 - 92.66)

18.54 (11.15 - 43.62)

0.6929

21.98 (7.91 - 85.73)

CD34+ collection efficiency 1 (CE1)

65.71 (8.59 - 125.40)

76.23 (50.54 - 98.42)

*0.0312

68.79 (8.64 - 119.50)

As shown the correlation analysis demonstrated a strong and significant correlation between the preapheresis PB-CD34+ cell count and the product CD34+ cell count. The linear regression model suggests that 37% of the variability in the concentration of CD34+ cells in the product can be attributed to the preapheresis cell count.

Similarly, when analyzing the dose achieved per procedure, a direct and proportional association was observed. This linear model suggests that a significant proportion of the dose depends on and can be predetermined by the preapheresis PB-CD34+ cell count. Furthermore, there were no statistically significant differences in the overall study regarding the type of transplant, indicating that analyzing all the data together was appropriate and sufficient for examining these associations.

Lastly, based on a graphic distribution of the data from 2023, it was observed that any preapheresis CD34+ value equal to or greater than 17 cells/μL corresponded to a cell dose greater than 2.0 × 106 cells/kg. However, to avoid biases in the series, the AUC was calculated, and a cutoff value of 20 cells/μL was obtained by applying the maximum value of Youden’s index with greater specificity.

Conclusions: A preapheresis PB-CD34+ cell count of 20 cells/μL may be considered the revised optimal threshold to initiate apheresis in our center. However, the previous value of 15 cells/μL can still be used on a case-by-case basis.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P742 APPLICATION OF GRANULOCYTE COLONY STIMULATING FACTOR DURING CYTAPHERESIS LEADS TO BETTER STEM CELL YIELDS

Ivan Tonev 1, Stanislav Simeonov1, Milcho Mincheff1

1Specialized Hospital For Active Treatment Of Haematological Diseases, Sofia, Bulgaria

Background: Patients with Hodgkin’s disease or Non-Hodgkin lymphomas who had underwent big number of chemotherapies and most of the patients with multiple myeloma are poor mobilizers. There is a controversy if application of granulocyte colony stimulating factor (G-CSF) during harvesting of peripheral blood hematopoietic stem cells (PBHSC) may result in better yields.

Methods: In an attempt to reach better yields, we applied additional dose of G-CSF at the beginning of cytapheresis and an hour later. To analyze if this approach leads to better harvests of PBSCs, we compared two groups of myeloma patients – with or without application of G-CSF during the procedure. We compared the initial number of CD34+ cells per microliter at the beginning and their number per kilogram BW in the apheresis product. We also calculated the ratio of the percentage of these cells in the peripheral blood and in the apheresis product.

Results: In both groups the initial number of CD34+ cells per microliter is comparable (33.885 in the group without G-CSF and 31.705 in the group with G-CSF application). The number of harvested CD34+ cells is higher in the group with application of growth factor 3.489 (1.93 to 5.22) versus 2.542 (0.83 to 4.42). The percentage ratio of CD 34+ cells in peripheral blood and apheresis product was 8.6 (2.22 to 4.49) vs. 6 (1.13 to 3.18) in the group receiving G-CSF during apheresis.

Conclusions: Receiving necessary dose of stem cells for transplantation can be a real challenge, especially in bad mobilizers. Application of granulocyte colony stimulating factor during cytapheresis is a feasible approach to improve stem cell harvests.

Disclosure: nothing to disclose.

2: Stem Cell Mobilization, Collection and Engineering

P743 ADAPTATION OF THE VOLUMETRIC METHOD ON THE FACSLYRIC CYTOMETER FOR THE ENUMERATION OF CD34 + STEM CELLS

Mohamed Brahimi 1, Amel Mihoubi2, Kamilia Amani2, Nawel Bounoua2, Aoudia Fatima2, Benaissa Mohamed Amine2, Amel Bendimerad2, Nabil Yafour1

1Établissement Hospitalier et Universitaire 1(er) Novembre 1954, Service d’Hématologie et de Thérapie Cellulaire, BP 4166 Ibn Rochd, Université d’Oran 1, Ahmed Ben Bella, Faculté de Médecine, Oran, Algeria, 2Établissement Hospitalier et Universitaire 1(er) Novembre 1954, Service d’Hématologie et de Thérapie Cellulaire, Oran, Algeria

Background: The enumeration of hematopoietic stem cells (HSCs) CD34+ by flow cytometry (FCM) is necessary to assess quality of the leukapheresis harvests. CD34+ HSCs can be counted by various methods. Currently, standardized FCM procedures by ISHAGE double and single platforms are most commonly used. Alternatively, single-platform procedures based on volumetric principles can be performed, thus avoiding the use of beads or counting white blood cell by a hematology analyzer. In our laboratory, we adapted the volumetric method on the FACS Lyric and compared the results obtained by this technique with those obtained by the double platform method.

Methods: This study is a prospective analysis conducted from 27/11/22 to 20/11/23.

160 samples (2ml) from apheresis products for autologous patients and allogeneic donors were analyzed on a BD FACS Lyric cytometer following the ISHAGE double-platform method.

The FACS Lyric cytometer, set to “Low” flow rate, can analyze an acquisition volume of 10 μl of sample per minute. The number of CD34+ cells acquired in 10 minutes (100 μl) is divided by 100 and then multiplied by the dilution factor to obtain the number of CD34+ cells/μl. The values obtained by this technique are compared to those obtained from the ISHAGE double-platform method.

Results: The report of evaluation on all 160 samples with the two Cytometric methods gave the following least squares equation by comparing the Volumetric (y) to the double plateform method (x): y=0.875x (r2= 0.97). The paired t-test showed no significant difference between the two methods (p>0.05).

The Intra-class Correlation Coefficient (ICC) was equal to 0.958. The plot of the differences between the volumetric and double platform values against their means according to Band and Altman design showed that the difference mean was 166.31 with a standard deviation (SD=258.7). We noticed that 91.25% of the differences were within the agreement limits (mean±2SD).

Conclusions: In our study, both the double-platform and volumetric methods yielded similar results for CD34+ cell enumeration. The results obtained demonstrate that the volumetric technique based on flow stability in “LOW” mode is feasible on the FACS Lyric cytometer. This method represents a simple and cost-effective approach, reducing the error due to hematology analyzers.

Disclosure: the authors declare no conflict of interest.

2: Stem Cell Mobilization, Collection and Engineering

P744 COMPARISON OF CE CHEMOTHERAPY PLUS G-CSF VS. G-CSF ALONE FOR AUTOLOGOUS HEMATOPOIETIC STEM CELL MOBILIZATION IN PATIENTS WITH MULTIPLE MYELOMA: A SINGLE CENTER RETROSPECTIVE ANALYSIS

Martin Hildebrandt1, Veronika Dill 1, Philipp Blüm2, Anja Lindemann1, Alexander Biederstädt3, Marion Högner1, Katharina Götze1, Florian Basssermann1

1Technical University of Munich, Munich, Germany, 2Medizinische Fakultät Mannheim der Universität Heidelberg, DRK-Blutspendedienst Baden-Württemberg | Hessen, Mannheim, Germany, 3The University of Texas MD Anderson Cancer Center, Houston, United States

Background: Despite recent advances in the treatment of multiple myeloma, high dose chemotherapy followed by autologous stem cell transplantation (ASCT) remains an essential keystone in the therapy of eligible patients. As for the stem cell mobilization procedure, different regimens have been established, usually consisting of a cycle of chemotherapy followed by application of G-CSF, with febrile neutropenia being a common complication. During the COVID-19 Pandemic, our institution decided to primarily use G-CSF only mobilization in order to minimize the patients’ risk of infection and to reduce the burden on the health system, in accordance with national guidelines.

Methods: This is a single-center, retrospective analysis of 106 consecutive patients with multiple myeloma undergoing hematopoietic stem cell mobilization and collection at our institution from January 2019 until October 2021. Based on protocols issued by the German Multiple Myeloma Group, a chemotherapy regimen with Cyclophosphamide and Etoposide (CE) plus G-CSF was used as the standard mobilization regimen for patients with MM. Alternatively, mobilisation of hematopoietic stem cells was performed by application of G-CSF only, in accordance with the recommendations of the European Myeloma Network (EMN) (Terpos et al., Leukemia, 2020). The efficacy and safety of G-CSF only mobilization was retrospectively analyzed and compared to a historical cohort undergoing chemotherapy-based mobilization with CE.

Results: CE offered a higher mobilization efficacy and greater CD34+ yield than G-CSF alone, but was also associated with a relevant proportion of patients developing infectious complications, febrile neutropenia, and a higher need for blood transfusions. Mobilization with G-CSF alone, although associated with a higher need for plerixafor administration and a higher number of apheresis sessions per patient, allowed the collection of the target dose of hematopoietic stem cells in the majority of our patients. With regard to efficacy of the products, i.e. engraftment after ASCT, there was no difference between patients mobilized with chemotherapy plus G-CSF vs. G-CSF alone.

Conclusions: In preparation for high-dose chemotherapy in patients with multiple myeloma, stem cell mobilization using G-CSF alone provides a valid alternative to chemotherapy-based mobilization, especially in selected patients with comorbidities and an increased risk of serious infections, in the winter season and also in future pandemics.

Clinical Trial Registry: The investigation was approved by the Local Ethics Committee (ethical approval # 2023-412-S-NP).

Disclosure: The authors declare no competing interests.

2: Stem Cell Mobilization, Collection and Engineering

P745 BLOOD TRANSFUSION NEEDS DURING PERIPHERAL BLOOD STEM CELL COLLECTION: A SINGLE-CENTER RETROSPECTIVE STUDY

Liliana Fonseca1, Catarina Almeida2, Pedro Leite-Silva3, Ana Salselas 3, Susana Roncon3

1Centro Hospitalar Tondela-Viseu, Viseu, Portugal, 2Centro Hospitalar do Baixo Vouga, Aveiro, Portugal, 3Portuguese Oncology Institute of Porto (IPO Porto), Porto, Portugal

Background: Peripheral blood stem cell (PBSC) collecting is a key step for an autologous hematopoietic cell transplantation (HCT). We aim to know the impact of apheresis on patient need for red blood cell and platelet transfusion.

Methods: Clinical details from patients proposed for PBSC collection were retrospectively reviewed from 2020 to 2022. A total of 254 patients (25 were children) and 429 apheresis (49 in pediatrics) were included. The demographic characteristics of the patient, number of apheresis procedures, granulocyte colony-stimulating factor (G-CSF) doses, laboratory values, red blood cell/platelet transfusions, and the apheresis method used (continuous in adults vs by cycles in pediatrics) were recorded. The transfusion threshold was hemoglobin ≤7 g/L and platelet ≤50x109/L. The patients were divided into 2 groups according to the number of apheresis: 1 and ≥2. Statistical analysis was performed using the Wilcoxon rank-sum test.

Results: Fifty percent of the patients were male, and the mean age was 47.9 years. The mean time between the last chemotherapy treatment and collection was 59.2 days. In most patients (57,5%), 1-day collection was enough (mean 1.46). Only 8 cases (1.9%) required red blood cell transfusion and 112 (26.1%) platelets. Before the procedure, the mean hemoglobin was 11.6 g/dL, hematocrit 34.6%, and platelets 160x109/L. In the group of ≥2 apheresis, an association was found between the number of procedures and the need for platelet transfusion (p<0.001). Also in this group, the value of hemoglobin and hematocrit has an impact on the need for red blood cell transfusion (p<0.001). Moreover, in both groups, a statistical significance was found between platelet count before the procedure and platelet transfusion needs. The G-CSF dose doesn’t seem to significantly impact the patient’s platelet count, but there appears to be a tendency to lower platelet counts after the administration (platelets mean was 210x109/L and after 160x109/L). We also observed that a high flow during the procedure resulted in an augment of both platelet (p<0.001) and red blood cell transfusions (p=0.026); therefore, the apheresis technique doesn’t influence the need for transfusions.

Conclusions: We conclude that the red blood cell transfusion can be impacted by the value of hemoglobin and hematocrit prior to the procedure when the patient needs more than 1 day of collection and by the flow during the procedure. The need for platelet transfusion is associated with the platelet count before the apheresis, the number of procedures, and the flow during the procedure. Despite literature reporting an effect of G-CSF on platelet count, resulting in thrombocytopenia, we only found a tendency to reduce platelet count in our population. The PBSC collection by apheresis is an effective and safe procedure; no severe adverse events transfusion-related were reported. We consider that preparing and optimizing clinical and analytical patient status could lead to a successful collection and later to a timely HCT.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P746 ENHANCING PRECISION IN PERIPHERAL BLOOD HAEMATOPOIETIC PROGENITOR CELL APHERESIS: A NOVEL APPROACH TO INTEGRATE COLLECTION EFFICIENCY VARIANCE FOR PRECISE CD34 + YIELD PREDICTION

Nelson Chan1, Kai Wan 1, Sinju Thomas1, Sandra Loaiza1, Shab Uddin1, Edward Aina1, Emma Bray1, Kaivalya Kulkarni1, Romeo Valeza1, Ruth Emhazion1, Jane Apperley2,1, Edurado Olavarria2,1, Renuka Palanicawandar1

1Imperial College NHS Healthcare Trust, London, United Kingdom, 2Imperial College London, London, United Kingdom

Background: Traditionally, the CE2 formula has been utilised to predict yields in apheresis and to guide the volume of blood processed. A recent study indicated substantial variability in Collection Efficiency (CE), with ranges from 2.0 to 8.8 (median 5.3), calling into question the reliability of the CE2 formula. This investigation reviews the distribution and variability of CE in non-plerixafor mobilised apheresis and explores contributory factors to this variability. Additionally, the study proposes a refined approach to predict CD34+ apheresis yield by incorporating CE variance.

Methods: A retrospective review of peripheral blood stem cell (PBSC) mobilisation data from 2022 was conducted at the Hammersmith Hospital’s Haematology apheresis unit. The dataset comprised of 160 non-plerixafor mobilised collections, with CE data available for 85 cases. An Anderson-Darling test assessed the normality of CE, and one-way ANOVA was employed to explore differences in CE across various subgroups including diagnosis, mobilisation regimen, operator, apheresis protocol and apheresis device.

Results: CE demonstrated a normal distribution (p=0.865), with a mean of 4.96, and a 95% confidence interval (CI) extending from 2.28 to 7.64. Analysis revealed no significant differences in CE across the subgroups investigated (Table 1). This finding suggests an intrinsic variability in CE that is not readily explained by the subgroups analysed.

Subgroups (Mean CE)

p-value

Diagnosis

Multiple myeloma (4.92), Multiple Sclerosis (5.00), Lymphoma (4.23), Germ Cell Tumor (5.66) and Healthy Donors (5.04)

0.2084

Apheresis protocol

MNC (4.88) and CMNC (4.98)

0.8110

Mobilization regime

G-CSF alone (4.92), G-CSF + Etoposide (4.72), G-CSF + Cyclophosphamide (5.96)

0.4060

Operator

Operators A - E

0.6630

Apheresis device

Devices A - F

0.2234

Table 1. Subgroup CE analysis. CE, Collection Efficiency.

Conclusions: This study described substantial real-life variability in Collection Efficiency (CE), with no discernible factors contributing to this variability. The conventional application of the CE2 formula provides only a 50% one-tailed probability of achieving the target yield. In our proposed model, we generate the probability distribution of the CD34+ yield by incorporating both the mean and variance of CE into the CE2 formula. For instance, consider an autologous peripheral blood stem cell collection for a donor weighing 60 kg, with a total blood volume (TBV) of 4339 mL, and a pre-apheresis peripheral blood CD34+ count of 25 cells/µL. The predicted CD34+ yield with processing 3 TBV is 2.69 x 10^6/kg (95% CI: 1.20-4.18 x 10^6/kg), and the probability of achieving the target dose of 2 x 10^6/kg is 69.3%. Further prospective validation of this prediction model is underway to evaluate its role in apheresis planning.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P747 STEM CELL MOBILIZATION IN MULTIPLE MYELOMA: DESCRIPTIVE ANALYSIS AND PREDICTIVE FACTORS FOR STEM CELL MOBILIZATION FAILURE

Laura Pardo Gambarte 1, Isabel Iturrate Basarán1, Begoña Pérez de Camino Gaisse1, Soledad Sánchez Fernández1, Javier Cornago Navascués1, Laura Solán Blanco1, Jose Luis López Lorenzo1, Elham Askari1, Amalia Domingo González1, Elena Prieto Pareja1, Alberto Velasco Valdazo2, Rafael Martos Martínez3, Daniel Naya Errea4, Pilar Llamas Sillero1

1Fundación Jiménez Díaz University Hospital, Madrid, Spain, 2Rey Juan Carlos University Hospital, Madrid, Spain, 3General de Villalba University Hospital, Madrid, Spain, 4Infanta Elena University Hospital, Madrid, Spain

Background: For patients with newly diagnosed multiple myeloma (MM) undergoing high-dose chemotherapy followed by autologous stem cell transplantation (ASCT) is considered the standard of care, being the apheresis of stem cell essential for first-line ASCT.

A minimum of 2x106 CD34+ cells/kg is needed to carry out the transplant. However, 5-40% of patients fail to collect this minimum threshold, with mobilization failures described in up to 17% of cases.

Lenalidomide and new schemes with Daratumumab have shown to have an adverse impact in the stem cell transplant yield.

The principal objectives of our study were to describe the characteristics of our population and to identify risks factors associated with a mobilization failure in order to optimize the use of Plerixafor and plan an appropriate mobilization strategy for each patient.

Methods: A multicenter, observational and retrospective study was performed, including 124 patients with MM who performed stem cell mobilization from January 2019 to October 2023.

Patients received Zarzio® (filgastrim) 10-15 µg/kg/day sc for 5 days, using Plerixafor in case of CD34 + < 10/ µg on day +4. All apheresis was performed with Spectra Optia® (Terumo).

To compare different groups, univariate logistic regression and multivariate models were performed. Statistical analysis was performed with R software (R Development Core Team). Values of p<0.05 were considered statistically significant.

Results: The mean number of cycles prior to mobilization was 4, with a mean of 11 days from the end of the cycle to the start of mobilization. Seventy-one patients (57%) were treated with VTd, 19 (15%) with VRd, and 24 (19%) received Daratumumab-based schemes. A total of 56 patients (45%) received Lenalidomide. Twenty-two patients (18%) switched from first line, 13 of them (54%) to VRd. Of the total number of patients, 59 (48%) required Plerixafor, 7 of them (12%) needed 2 doses. Eight patients (7%) had mobilization failure.

Two groups were compared: patients with optimal mobilization (CD34 + /microliter > 25 on day +5) without the need of Plerixafor, versus the rest (mobilization failure, suboptimal mobilization or optimal mobilization after Plerixafor).

In the univariate analysis, higher body weight and platelet count were found to be associated with better mobilization rates. However, shorter time from the last cycle received to mobilization, as well as higher medullary infiltration, monoclonal component and light chain ratio were associated with a higher probability of poor mobilization. In terms of treatment, previous use of Lenalidomide and Daratumumab was found to be significantly associated with risk of insufficient mobilization.

In multivariate analysis, prior Lenalidomide use, lower body weight and lower platelet count were associated with poor mobilization.

Conclusions: Previous use of Lenalidomide and Daratumumab hinders the mobilization of hematopoietic progenitors. A higher number of cycles received and a shorter rest time between treatment and mobilization is associated with worse mobilization. Higher myeloma tumor burden and lower body weight are also factors for insufficient mobilization. Plerixafor rescues most patients and its administration is safe, allowing stem cell collection and subsequent autologous transplantation.

Disclosure: The authors declare no conflict of interest.

2: Stem Cell Mobilization, Collection and Engineering

P748 MOBILIZATION AND HEMATOPOIETIC STEM CELL TRANSPLANTATION IN PATIENTS WITH MULTIPLE MYELOMA WITH DARATUMUMAB-BASED INDUCTION TREATMENS

Isabel Izquierdo 1, Ana Gomez2, Ana Godoy1, Juan Gimeno1, Beatriz Martinez3, Vicente Carrasco4

1Hospital Miguel Servet, Zaragoza, Spain, 2Hospital Ernest Lluch, Calatayud, Spain, 3Hospital Clinico Lozano Blesa, Zaragoza, Spain, 4Hospital Royo Villanova, Zaragoza, Spain

Background: Standard first-line treatment for patients with newly diagnosed Multiple Myeloma (MM) transplantation candidates includes combinations with Bortezomib and Dexamethasone, Thalidomide, Lenalidomide, Cyclophosphamide or Doxorubicin. In September 2022, Daratumumab was approved in combination with bortezomib, thalidomide and dexamethasone (VTd) for these patients.

Data collected from the MASTER and GRIFFIN trials show that the introduction of Daratumumab in the 1st line doesn´t affect the feasibility and safety when performing autologous transplantation, although a significant reduction in stem cell mobilization and stem cell yield, even with the use of plerixafor-containing mobilization regimens has been described.

Methods: An observational, retrospective and multicenter study was carried out in our region. We included patients with a diagnosis of MM who underwent autologous stem cell mobilization and collection during first-line treatment from January 2022 to November 2023. A descriptive analysis was carried out in which mean, median, standard deviation (SD) and range [Range] were calculated in quantitative variables; and absolute and relative frequencies in qualitative variables. The analysis was carried out based on the sociodemographic, clinical, analytical and therapeutic variables previously collected.

Results: 53 patients were included in the analysis. The median age was 61,6 (range: 40-73), 30% were male.

50,9% received daratumumab during induction. Of those, the median number of cycles administered was 3 (range: 3-15). 49% received daratumumab in combination with bortezomib, lenalidomide and dexamethasone.

All patients underwent mobilization using G-CSF at a dose of 1mg/kg/12h for 5 days. In case of failure, rescue was performed with administration of Plerixafor 20 mg or 0.24 mg/kg body weight for patients weighing ≤ 83 kg and 0.24 mg/kg body weight for patients weighing > 83 kg, followed by a new apheresis. A second mobilization was performed with G-CSF only in 1 patient without Plerixafor administration.

In the group receiving Daratumumab, 8 patients (29,6%) presented mobilization failure, requiring rescue with plerixafor.

Patients with daratumumab exposure had a mean mobilization of 3,18 x106 CD34+ cells compared to 4,41 for those without. Two patients failed to collect 2.0 x 106 cells/kg, the institutional minimum for transplantation.

Of all patients, 39 autologous transplantation were performed (73.58%), 22 patients (41.51%) had not previously received Daratumumab. The group that received Daratumumab presented a median number of hemoperipheral recovery evaluated through the neutrophilic white cell (0.5x109/L) and platelet (20x109/L) graft of 11.0 days [8.0-17.0] and 12.5 days [10.0-17.0] respectively.

With Daratumumab

(n=27)

Without Daratumumab (n=26)

Total

patients

(n=53)

Stem cells CD34 x106, Median [Range]

3,18 [1,75-7,18]

4,41 [2,8-11,42]

3,69 [1,75-11,42]

Mobilization with G-CSF (dose 1mg/kg/12h x 4-5 days), n (%)

27 (100,0%)

26 (100,0%)

53 (100,0%)

Mobilization failure with G-CSF, n (%)

8 (29,63%)

9 (34,62%)

17 (32,08%)

Use of Plerixafor (mobilization rescue), n (%)

7 (25,93%)*

9 (34,62%)

16 (30,19%)

Cycle number apheresis

Median [Range]

3 [3,0-15,0]

4 [3,0-12,0]

4 [3,0-15,0]

Days to neutrophil engraftment (0,5x109/L), Median [Range]

11 [8,0-17,0]

11 [7,0-17,0]

11 [8,0-17,0]

Days until platelet engraftment

(20x109/L) Median [Range]

12,5 [10,0-17,0]

12 [7,0-16,0]

12 [7,0-17,0]

Conclusions: In our experience, inclusion of Daratumumab in the induction regimen for transplant-eligible patients with MM resulted in a reduction in stem cell mobilization and stem cell yield, although our number of cases is still low to drawing conclusions.

Treatment with quadruplets with Daratumumab in first line does not preclude successful mobilization or subsequent HSCT, although an increased use of Plerixafor and novel strategies such as early mobilization may be required, given the speed and depth of responses that are achieved with this type of regimen.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P749 STEM CELL MOBILIZATION IN MULTIPLE MYELOMA PATIENTS: A SINGLE CENTER STUDY IN TUNISIA

Imen Bellalah1, Karima Kacem 1, Malek Sayadi1, Dorra Jabeur1, Raoudha Mansouri1, Raihane Banlakhal1

1Aziza Othmana Hospital, Tunis, Tunisia

Background: The management of multiple myeloma (MM) in young patients (≤ 65 years) involves induction chemotherapy, followed by intensification through single or double autologous stem cell transplantation (ASCT). Inadequate mobilization and peripheral stem cell (HPC) harvest can pose significant challenges to the successful execution of ASCT. The objective of this study was to assess the strategy employed for HPC mobilization and collection in MM patients eligible for transplantation at Aziza Othmana Hospital in Tunisia.

Methods: This retrospective study involves patients aged less than 70, diagnosed between January 2020 and December 2022 with symptomatic MM (based on CRAB/SLIM criteria), eligible for ASCT irrespective of the type of induction chemotherapy (CTD/VTD/VCD). The induction therapy regimen comprises a four-cycle protocol, followed by mobilization. Patients who did not undergo stem cell mobilization after induction chemotherapy were excluded.

The mobilization protocol used Etoposide (375mg/m2: D1-D2) + G-CSF at 10γ/kg/day from D3 subcutaneously until peripheral stem cell harvest. CD34+ cells were measured in peripheral blood from day 10. A CD34+ cutoff level of 20 μL was necessary to initiate apheresis which be continued untel having a harvest ≥4 × 10^6/kg for double autograft.

Cobe Spectra or Optia were used for apheresis. Stem cell mobilization failure was defined by a persistent circulating CD34+ cell count < 20/µl or the collection of CD34+ cells < 4 × 10^6 CD34+ cells/kg.

Results: A total of 118 patients were included. The baseline clinical and demographic characteristics of patients are listed in Table 1. The mobilization procedure was discontinued in 3 patients due to febrile neutropenia, resulting in death from septic shock in one patient.

Fifty-six percent of patients were collected via a central venous line. The median number of circulating CD34 cells on the day of apheresis was 302/μl (range, 54-1169). The median number of apheresis sessions was 2 (range, 1-3) at D12 (range, 10-16).

Adequate (≥4 × 10^6/kg) CD34+ cells were collected at the first apheresis in 86 patients, while 24 patients required 2 apheresis sessions. A median of de 12.98 x 106 /Kg CD34 cells were collected on the first mobilization trial (range, 3.11-38).

The median number of CD34+ cells ×10^6/kg collected on days 1, 2, and 3 of apheresis was respectively 10.79 (1.02-38), 4.44 (0.6-10.07), and 1.45 (0.33-3.07). Failure was noted in 2 among 115 (1.73%) patients, and a second mobilization trial was successful in one patient with Etoposide+G-CSF. The other patient showed progression.

In this study, 24.34% of patients had thrombocytopenia grade 3, and 7% with anemia grade 3.

N

118

Male/female

69/49

Median age at diagnosis

56 (39-68)

(range), years

ISS at diagnosis : 98 patients

Stage I (%)

30 (30.61%)

Stage II (%)

33 (33.67%)

Stage III (%)

35 (35.71%)

R-ISS: 89 patients

Stage I (%)

14 (15.73%)

Stage II (%)

36 (40.44%)

Stage III (%)

39 (43.82%)

Type of the MM

Ig G kappa

41 (34.75%)

Ig G lambda

22 (18.64%)

Ig A kappa

15 (13%)

Ig A lambda

12 (9.3%)

Kappa light chain

20 (16.94%)

Lambda light chain

8 (6.77%)

Induction chemotherapy

VTD

112 (94.91%)

VCD

4 (3.38%)

CTD

2 (1.7%)

Radiotherapy

18 (15.25%)

Disease status before stem

cell mobilization 111

CR

26 (23.42%)

VGPR

47 (42.34%)

PR

34 (28,8%)

SD

3 (2.7%)

Active disease

1 (0.9%)

Conclusions: Due to the low rate of mobilization failure (1.7%), the investigation into predictive factors for failure could not be conducted. This mobilization strategy has proven effective in obtaining a sufficient number of CD34+ cells.

Disclosure: Nothing to declare.

2: Stem Cell Mobilization, Collection and Engineering

P750 CHALLENGES OF HEMATOPOIETIC STEM CELL COLLECTION IN BIG PEDIATRIC TRANSPLANT CENTER: SINGLE CENTER EXPERIENCE

Nara Stepanyan 1, Kirill Kirgizov1, Elena Machneva2, Ramil Fatkhullin1, Timur Aliev1, Irina Kostareva1, Nune Matinyan1, Vladimir Zhogov1, Marina Rubanskaya1, Olga Romantsova1, Natalia Batmanova1, Timur Valiev1, Tatiyana Gorbunova1, Vladimir Polyakov1, Svatlana Varfolomeeva1

1Research Institute of Pediatric Oncology and Hematology, N.N. Blokhin National Medical Research Center for Oncology, Russian Ministry of Health, Moscow, Russian Federation, 2Russian Children’s Clinical Hospital of the N.I. Pirogov Russian National Research Medical University, Ministry of Health of Russia, Moscow, Russian Federation

Background: HSCT for children requires advanced HSC harvesting and processing unit. Organization of such unit is the challenge. We aimed to show our experience in organization of such unit for big pediatric transplant center.

Methods: During the period of January 2021 – November 2023 we have collected 333 HSC both as PBSC (n=258) and BM (n=75). Before the start of operations in 2021 we established our HSC harvest protocol. Autologous HSC (n=223) collected for pts. with solid non-hematological tumors and lymphomas. Forty-seven collections were made for tandem HSCT. Median body weight of the patients (underwent autoHSCT) was 28.3 (7.0-95.0) kg, height – 124 (55-188) cm, and age – 92.0 (3.0-215.0) months. Median body weight of healthy donors was 58.3 (13.0-102.0) kg, median height – 148.1 (74-182) cm and the median age was 432 (12.0-636.0) months. All PBSC (n=258) collected with the help of Spectra Optia separator using MNC 6,9,12 software (mononuclear cell boron). Our protocol includes both G-CSF and CXCR4 antagonists (plerixafor) stimulation.

Results: Most of the pts. (94%) and all donors successfully donated HSC. Only 10 pts. and 3 donors were poor mobilizers and received CXCR4 agonists. Median content of CD 34+ cells in the apheresis product from pts. was 230 (0.7-1291) cells/μl, the median apheresis duration was 259 (160-435) minutes. The median content of CD 34+ cells in the apheresis product from donors was 267.6 (54-832) cells/μl, the median duration apheresis - 185 (75-406) minutes. BM harvest: CD34+ cells in the bone marrow concentrate being 107.8 (31-322.7) cells/μl, the median duration of the operation was 75 minutes (55-125) minutes, excluding the time for anesthesia. No severe complications were found during the procedures both for pts. and donors. Main challenge was to establish team and protocol.

Conclusions: Our experience showed possibility of organization of effective harvest and processing team in our institution. Our major challenges was establishing team and protocol. Application of modern technologies allowed to improve the results of our harvest and processing unit.

Disclosure: No conflict of Interest statement.

2: Stem Cell Mobilization, Collection and Engineering

P751 IMMUNOPHENOTYPING OF PERIPHERAL BLOOD MONONUCLEAR CELLS COLLECTED BY APHERESIS

Carlos Agustin Villegas-Valverde 1, Antonio Bencomo-Hernandez1, Yandy Max Castillo-Aleman1, Fatrma Abdou1, Shadi Shamat1, Anil Kumar1, Yendry Ventura-Carmenate2, Fatema Al Kaabi3, Inas El-Najjar3, Shinnette Lumame3, Charisma Castelo3, Nameer Abdul Al Saadawi3, Dina El Mouzain3, Hiba Abdelrahman3, Jayakumar Dennison3, Mansi Sachdev3, Rene Antonio Rivero-Jimenez1

1Abu Dhabi Stem Cells Center, Abu Dhabi, United Arab Emirates, 2Abu Dhabi Stem Cells Center/Yas Clinic Khalifa City, Abu Dhabi, United Arab Emirates, 3Yas Clinic Khalifa City, Abu Dhabi, United Arab Emirates

Background: Many hematological and non-hematological disorders, circulating mononuclear cells (MNCs) also influence the hematopoietic reconstitution. (1, 2) In allogeneic transplants, among the chimerism of various cell populations, different donor-derived cell subsets are also indicators for a successful engraftment or graft-versus-host disease (GvHD). (3, 4, 5) However, publications reporting the immunophenotyping of peripheral blood MNCs collected by apheresis are still scarce.

Methods: The initial apheresis procedures conducted between July 1, 2020, and July 1, 2022, in the launching period of the Abu Dhabi Bone Marrow Transplant (AD-BMT) Program comprised this analysis, being a retrospective data collection exempted from ethics review as confirmed by the Institutional Review Board. The sample was made up of 14 patients with diagnosis of malignant hemopathies. The data collected included the immunophenotype of cell subset count in peripheral blood immediately before the apheresis and apheresis product. Immune profiles were done with flow cytometry using two polychromatic tubes (DURAClone IM Phenotyping Basic Tube and DURAClone IM T cell subsets Tube from Beckman Coulter). A Navios EX Flow Cytometer (Beckman Coulter, USA) was used, and data acquisition was processed with Kaluza C Software v. 1.2 (Beckman Coulter, USA).

Results: A total of 14 apheresis were conducted using the Amicus® apheresis system (Fresenius Kabi, Germany) for autologous HSC transplantations (HSCTs). All patients received a standard mobilization protocol with filgrastim (G-CSF) or G-CSF plus plerixafor. Table 1 compares the median cell count of selected subsets in preapheresis peripheral blood and apheresis product. The apheresis procedure does not compromise the relative proportions of the different subsets of MNCs harvested. Moreover, apheresis increased MNCs’ relative and absolute concentrations as expected, mainly due to the poor collection of most granulocytes, which is influenced by their specific gravity and lower collection efficiency.

As shown in Table 1, there were no significant changes between the relative concentrations (%) of monocytic and lymphocytic cell subpopulations between peripheral blood and the apheresis product. However, T cytotoxic (Tc) naïve lymphocytes increased significantly in the apheresis product, and terminally differentiated effector memory (TEMRA) cells decreased significantly compared to the preapheresis peripheral blood count.

Table 1. Comparison between the median cell count of selected subsets in preapheresis peripheral blood and apheresis product. (n=14)

Subsets

Median % (5th - 95th percentile)

Wilcoxon test

Preaphersis peripheral blood

Apheresis product

Monocytes (M)

9.5 (5.1-13.6)

29.3 (16.5-37.1)

**0.0078

Classical M

70.7 (46.9-80.3)

66.7 (27.0-77.6)

0.2500

Non-classical M

29.2 (19.8-53.2)

33.3 (22.5-68.7)

0.2500

Total Lymphocytes

8.3 (3.2-17.2)

24.9 (10.6-59.0)

**0.0078

B lymphocytes

3.0 (2.1-10.9)

3.4 (1.5-9.2)

0.8438

T lymphocytes

83.2 (66.0-89.6)

86.2 (41.8-90.9)

0.7539

T helper (Th) cells

42.1 (34.9-60.5)

42.0 (32.1-59.8)

>0.9999

Th Naïve

22.0 (14.6-42.7)

20.7 (12.6-50.6)

>0.9999

Th Central Memory

55.8 (36.5-78.1)

56.2 (36.5-79.0)

>0.9999

Th Effector Memory

19.2 (7.0-34.9)

17.5 (8.3-33.6)

0.6406

Th TEMRA

0.1 (0.0-2.1)

0.1 (0.0-2.6)

0.2500

T cytotoxic (Tc) cells

25.0 (22.3-41.6)

28.3 (19.2-45.1)

0.6523

Tc Naïve

16.7 (7.7-39.2)

27.2 (12.0-66.7)

*0.0391

Tc Central Memory

16.8 (6.7-49.6)

14.4 (5.2-41.6)

0.6406

Tc Effector Memory

46.5 (11.1-62.0)

32.1 (12.4-59.9)

0.1484

Tc TEMRA

17.6 (5.7-39.9)

13.3 (5.2-35.3)

*0.0156

Natural Killer Cells

5.0 (3.0-23.9)

7.3 (1.9-21.9)

0.8203

Natural Killer T Cells

4.4 (0.3-9.4)

3.4 (0.5-7.1)

0.1484

Conclusions: Although HSCTs have been performed as a standard of care for many disorders, little is known about the effects of mobilization and apheresis collection on the graft MNC composition, which may impact the transplant outcomes. Therefore, further studies are required to verify its effects on engraftment kinetics and its clinical implications.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

9: Stem Cell Source

P752 REAPPRAISING CORD BLOOD AS A PRIORITY CELL SOURCE IN ALTERNATE DONOR TRANSPLANT AND IN DIFFICULT-TO-CURE PAEDIATRIC HEMATOLOGIC MALIGNANCY

Abdul Wajid Moothedath 1, Denise Bonney1, Madeleine Powys1, Ramya Nataraj1, Omima Mustafa1, Srividhya Senthil1, Robert Wynn1

1Manchester University NHS Foundation Trust, Manchester, United Kingdom

Background: Cord blood (CB) has declined as a cell source, it’s decline paralleled by a rise in haploidentical transplant. We continue to prioritize CB as a donor cell source. In metabolic disease, the wild-type donor with high chimerism delivers augmented enzyme to the recipient tissues and is rapidly available and early age at transplant affects disease outcomes. HLA mismatch is better tolerated than with adult donors with low rates of chronic GVHD. In malignant disease, CB allows T-cell replete and mismatched transplant, thereby optimizing graft versus leukaemia and reducing relapse, especially for highest risk disease.

Methods: Ten year retrospective, observational, single-centre study of paediatric patients who received an allogeneic transplant using CB (T-deplete for non-malignant and T-replete for high risk malignant) diseases. Data was collected using case and electronic records for transplant characteristics, engraftment, graft failure, GVHD and survival.

Results: A total of 103 patients (51 non-malignant and 52 malignant) received CB transplant. Median age at transplant was 7 years (IQR 2; 11) for malignant and 1.2 years (IQR 0.6; 2) for non-malignant cohorts. Among non-malignant diseases, 59% had a matched cord whereas 83% of malignant cohort had mismatched cord. Baseline characteristics are summarised in Table 1.

Neutrophil engraftment occured after a median duration of 17 days in both cohorts. In the non-malignant cohort, graft failure was seen in 7 patients (2 had primary and 5 had secondary). Cell dose was adequate in all these patients (median TNC-11.7 * 107/kg and CD34- 4.18 * 105 /kg). Median interval to second transplant was 101 days (IQR 63;129). Neutrophil engraftment was observed after a median of 14 days (IQR 11;25) post second transplant. One patient died due to co-morbidities associated with late diagnosis of Hurler’s. Among the malignant TRCB cohort, 4 patients had primary graft failure, of which 3 had persistent disease and 1 patient engrafted after a second cord transplant.

For non-malignant cohort, after a median follow up of 36.5 (IQR 13; 75 months), the overall survival(OS) was 80% and the event free survival(EFS) was 70%. No difference in OS was observed on the basis of HLA match (HLA-match 81% vs HLA-mismatched 78%; P=0.92). For malignant cohort, after a median follow up of 11.5 (IQR 4.25:48.5) months, OS was 59% and EFS was 53%. The OS and EFS was 87% and 83% in patients who were MRD negative pre-transplant and 44% and 39% in MRD positive patients, respectively.

Among non-malignant cohort, grade 3-4 acute GVHD was seen in 11/51 (21%), whereas it was seen in 16/52 (31%) among malignant cohort. The incidence of cGVHD was 10% (5/51) and 7.7% (4/52) among non-malignant and malignant patients respectively, who received a CB transplant. In majority, it was limited to skin only.

Table 1

Malignant

Number (n)

TRCB = 52

Non-malignant

TDCB =51

Age (Years)

Median (IQR)

7 (2-11)

1.2 (0.6-2)

Diagnosis (%)

• AML

38 (73)

• MPS-1

41 (80)

• MDS

5 (9.6)

• PID

2 (4)

• T-AML

6 (11.5)

• Fanconi anemia

1 (2)

• MPAL

1 (1.9)

• Leukodystrophies

2 (4)

• Myeloid sarcoma

1 (1.9)

• Other storage

5 (10)

• Downs AML

1 (1.9)

HLA match

Matched cord (8/8)

9 (17.3)

30 (59)

Mismatched cord

43 (82.7)

21 (41)

Flow MRD at HSCT

Not applicable

Positive

35 (67.3)

Negative

17 (32.7)

Morphology

Positive

11 (21.1)

Not applicable

Negative

41 (78.9)

Previous HSCT

17 (32.7)

0 (0)

Serotherapy

ATG

49 (96)

Alemtuzumab

2 (4)

Days to neutrophil engraftment

Median (IQR)

17 (15-21)

17 (14-21)

Graft failure

4 (7.7)

7 (13)

Management of graft failure

Second HSCT

2

7

Conclusions: Umbilical CB transplant with serotherapy offers an excellent curative option for non-malignant disease without a matched related donor. Incidence of graft failure is low and majority can be salvaged by an early second transplant. In malignant diseases, TRCB offers a potential for cure in relapsed-refractory AML/MDS with low rates of chronic GVHD.

Disclosure: Nothing to declare.

9: Stem Cell Source

P753 CORTICOSTEROIDS IMPAIR HEMATOPOIETIC MICROENVIRONMENT VIA INDUCING SENESCENCE OF THE MESENCHYMAL STROMAL NICHE

Yuqing Wang 1, Cong Wang1, Zheng Wang1, Xi Zhang1

1Army Medical University, Xinqiao Hospital, Chongqing, China

Background: Mesenchymal stem cells play a crucial role in maintaining the immune homeostasis of the post-transplant bone marrow microenvironment, which is essential for the successful engraftment of allogeneic hematopoietic stem cells. Graft-versus-host disease (GVHD) induced by allogeneic hematopoietic stem cell transplantation is a key factor contributing to transplant failure. Corticosteroids with or without calcineurin inhibitors comprise the first-line treatment option for graft-versus-host disease (GVHD). But there is still up 65% of patients experience steroid-resistant or steroid-refractory GVHD. The pathogenesis of steroid-resistant or steroid-refractory GVHD is complicated, and the impacts of steroid on bone marrow microenvironment are lacking.

Methods: To simulate the changes of bone marrow microenvironment during steroid treatment, mesenchymal stromal cells (MSCs), the pivotal component of bone microenvironment, were isolated from healthy donor and subjected to dexamethasone (Dex) treatment. Bulk RNA sequencing (RNA-seq) was employed to identify the significant molecular changes, followed by in vitro cell-based validation.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (95)

Results: Bulk RNA-seq analysis revealed downregulation of genes associated with cell cycle and mitosis following Dex treatment. The percentage of SA-β-gal positive cells increased under Dex treatment (Figure 1A). Additionally, Dex-treated BM-MSCs upregulated P16, indicating the potential induction of senescence in bone marrow derived MSCs (BM-MSCs). Besides, in BM-MSCs with glucocorticoid receptor (GR) knockdown, the percentage of Dex-induced senescent cells significantly decreased (Figure 1B). Thus, Dex-induced senescence in BM-MSCs appears to be GR-dependent. Moreover, Dex-treatment attenuated the osteogenic ability of BM-MSCs, while concurrently enhancing their adipogenic potential.

Figure 1. (A) The proportion of SA-β-Gal+ cells with or without Dex treatment. (B) The proportion of SA-β-Gal+ cells with si-NC or si-NR3C1 with or without Dex treatment.

Conclusions: Steroid treatment triggers senescence of BM-MSCs, resulting in an elevated secretion of inflammatory factors. This may underlie the diminished capacity of MSCs to support hematopoiesis observed in patients with steroid-resistant or steroid-refractory GVHD patients. And the compromised osteogenic ability of BM-MSCs following steroid treatment may lead to osteoporosis. Elucidating the pathogenesis of steroid on bone marrow environment may provide promising target for refractory/relapsed GVHD treatment.

Disclosure: Nothing to declare.

9: Stem Cell Source

P754 THE CORD BLOOD POST THAW CLINIC – HELPING YOU WORK OUT “WHAT GOOD LOOKS LIKE”

Roger Horton 1, Ben Cash2, Kennedy Davies2, Irina Evseeva3, Phoebe Groves3, Sophie Macleod3, Alex Ross2, Claire Simpson1, Daniel Gibson1

1Anthony Nolan Cell Therapy Centre, Nottingham, United Kingdom, 2NHS Blood and Transplant, Bristol, United Kingdom, 3Anthony Nolan, London, United Kingdom

Background: As part of the UK Cord Initiative, Anthony Nolan (AN) and NHS Blood and Transplant (NHSBT) runs the post thaw clinic to help answer queries around cord blood unit (CBU) post thaw data interpretation, which can be both complex and confusing. This service is designed to complement/ follow on from the BSHI selection advisory panel (BSHI SAP).

Methods: Queries were submitted to a dedicated inbox along with the relevant CBU reports and any additional information. The teams from AN and NHSBT cord blood banks, a panel consisting of 6 experts, then interpreted the data replying with a consensus opinion within 24 hours to give the requesting Transplant Centre (TC) guidance.

Results: Our 1st query was submitted on 27/04/2022. We have since had 73 queries for 56 patient cases from 11 different TCs. 94% of queries were responded to within our 24-hour limit. The majority of our queries (more than 50%) were about international CBUs.

The topics we were asked about varied, with 44% of requests asking simply for an overview and opinion from the panel. The remaining queries covered a variety of parameters including TNC viability, CFU values, help with calculations, CD34+ viability, nRBC numbers, TNC recovery, CD34+ recovery, RBC volume, age of the CBU and cell dose.

Example 1: An international CBU was reviewed as the TC were looking to submit workup forms, but CFU came back as no growth. This was a tricky one to judge with the lack of CFU growth, but the CBU report looked fine otherwise. Opinion – a risk assessment was required on the lack of CFUs vs urgency. This CBU was from a reputable bank but still a leap of faith for clinical team. Outcome – An alternative international CBU was requested for shipment and the patient was infused.

Example 2: Review of international CBU - the data was poor and not suitable for the patient if the cell dose calculations were correct (numbers were then confirmed by CBB). Opinion – the panel agreed they would not release this CBU if it were theirs. Outcome - 2 x URD WUs were requested and cancelled, then an alternative CBU pair was requested, and the patient was infused.

In 45% of cases TC’s were able to proceed with their cord of interest, for 22% an alternative was recommended and utilised, 5% utilised an alternative graft, 3% on hold, 13% did not proceed to workup and 13% are still ongoing (TC in the early stages of graft selection).

Conclusions: This service has proved valuable to TCs; 72% of cases proceeded straight after advice was given. In 27% of cases alternative CBUs or grafts were sourced after identifying quality issues thereby maximising patient outcomes. An additional bonus to TCs of the process was that late cancellations due to product related issues, reduced from 7% to 2.6% thereby reducing TC workload and facilitating faster patient treatment as well.

Clinical Trial Registry: Not applicable.

Disclosure: Nothing to declare.

9: Stem Cell Source

P755 USE ON NON-CRYOPRESERVED HEMATOPOIETIC STEM CELLS FOR AUTOLOGOUS HSCT IN ADULTS - MULTICENTER STUDY, FROM THE IDWP AND TCWP EBMT – PRELIMINARY RESULTS

Emilian Snarski 1, Gloria Tridello2, Lotus Wendel2, Zinaida Peric3, William Boreland4, Dina Averbuch5, Rafael de la Camara6

1University of Zielona Góra, Zielona Góra, Poland, 2EBMT, Leiden Study Unit, Leiden, Belgium, 3University Hospital Centre Rijeka and School of Medicine, Rijeka, Croatia, 4EBMT, Paris Study Unit, Paris, France, 5Pediatric Infectious Diseases, Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel, 6Hospital de la Princesa, Madrid, Spain

Background: Autologous HSCT is mostly performed with cryopreserved hematopoietic stem cell products. There are reports of use of non-cryopreserved grafts in AHSCT for lymphoma, multiple myeloma and autoimmune diseases – which show reduction of infectious complications and very good engraftment times. The data on cryopreservation of the autograft are not collected by EBMT so there is no information on how many AHSCT are performed with non-cryopreserved grafts in EBMT centers. Moreover, there is no large multicenter study that addresses impact of this practice on the risk of typical infectious complications and long term outcomes of AHSCT. We aimed at analyzing use of non-cryopreserved grafts for AHSCT in adults in EBMT centers.

Methods: In collaboration between EBMT Infectious Diseases Working Party and Transplant Complications Working Party, we designed a questionnaire that was distributed to EBMT centers. The first phase of the study aimed to determine the number of centers performing non-cryo and cryo AHSCT. Second phase is planned to analyse the long term outcomes of non-cryo versus cryo AHSCT.

Results: 207 centers from 45 countries responded to the questionnaire. 28 centers that use non-cryopreserved cells for AHSCT in some patients and 50 centers that only use cryopreserved grafts agreed to take part in the survey. In the years 2010 to 2021, these centers performed at least 1220 AHSCTs with non-cryopreserved grafts and 3429 AHSCTs with cryopreserved grafts. The numbers of non-cryo increased from 39 in 2011 to 175 in 2021. The centers performing non-cryo grafts were located in: South America (9), Southern Europe (9), Western Asia (4), Southern Asia (2), Western Europe (2), Eastern Europe (1), Northern Africa (1) and Southern Africa (1). The diseases most commonly transplanted with non-cryopreserved grafts included: multiple myeloma (11 out of 11 centers), amyloidosis (7 out of 10 centers), lymphomas (3 out of 11 centers), autoimmune diseases (2 out of 9 centers). The use of growth factors, antibiotic prophylaxis, antifungal prophylaxis and antiviral prophylaxis varied greatly between the centers.

Conclusions: There are at least 28 centers performing AHSCTs with non-cryopreserved grafts. The number of recorded cases allows future analysis of long term outcomes and comparison with similar patients recorded in the EBMT database. The use of non-cryografts for AHSCT has not been yet analysed in EBMT centers and early data from our study show feasibility of such analysis.

Disclosure: Nothing to declare.

The 50th Annual Meeting of the European Society for Blood and Marrow Transplantation: Physicians – Poster Session (P001-P755) (2025)

References

Top Articles
Latest Posts
Recommended Articles
Article information

Author: Kareem Mueller DO

Last Updated:

Views: 5542

Rating: 4.6 / 5 (46 voted)

Reviews: 85% of readers found this page helpful

Author information

Name: Kareem Mueller DO

Birthday: 1997-01-04

Address: Apt. 156 12935 Runolfsdottir Mission, Greenfort, MN 74384-6749

Phone: +16704982844747

Job: Corporate Administration Planner

Hobby: Mountain biking, Jewelry making, Stone skipping, Lacemaking, Knife making, Scrapbooking, Letterboxing

Introduction: My name is Kareem Mueller DO, I am a vivacious, super, thoughtful, excited, handsome, beautiful, combative person who loves writing and wants to share my knowledge and understanding with you.