1
|
Bischof L, Ussmann J, Platzbecker U, Jentzsch M, Franke GN. Allogeneic stem cell transplantation for MDS-clinical issues, choosing preparative regimens and outcome. Leuk Lymphoma 2025:1-14. [PMID: 40072896 DOI: 10.1080/10428194.2025.2476652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Despite the vast heterogeneity of myelodysplastic neoplasm (MDS), treatment options are limited and an allogeneic hematopoietic stem cell transplantation (allo-HSCT) remains the only curative approach. While, subsequently, allo-HSCT is the treatment of choice in fit high-risk MDS patients younger than 70 years, it should only be considered in young and fit low-risk MDS patients who suffer from severe and life threatening cytopenias, and fail all available conservative treatment options. With the increasing use of mismatched or haploidentical donors, the majority of MDS patients will have a suitable donor available, however, matched donors should still be preferred if rapidly available. Strategies to prevent relapse after allo-HSCT are scarce, and include the use of donor lymphocytes or the experimental use of hypomethylating agents in patients with impeding relapse detected by MRD or chimerism evaluation. Here, we summarize current treatment options and factors to consider in the context of allo-HSCT in MDS.
Collapse
Affiliation(s)
- Lara Bischof
- Department for Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Jule Ussmann
- Department for Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Uwe Platzbecker
- Department for Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Madlen Jentzsch
- Department for Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| | - Georg-Nikolaus Franke
- Department for Hematology, Cellular Therapy, Hemostaseology and Infectious Diseases, University of Leipzig Medical Center, Leipzig, Germany
| |
Collapse
|
2
|
Marumo A, Nagata Y, Fujioka M, Kurosawa S, Najima Y, Sakaida E, Doki N, Fukushima K, Ota S, Shono K, Ito A, Uchida N, Nishida T, Sawa M, Tsunemine H, Matsuoka KI, Makoto O, Kanda Y, Fukuda T, Atsuta Y, Itonaga H. Outcome of donor lymphocyte infusion after allogeneic hematopoietic stem cell transplantation in relapsed myelodysplastic syndrome. Cytotherapy 2025; 27:222-228. [PMID: 39503682 DOI: 10.1016/j.jcyt.2024.09.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/08/2024]
Abstract
BACKGROUND AIMS Allogeneic hematopoietic stem cell transplantation (HSCT) improves outcomes for myelodysplastic syndrome (MDS) patients, but relapse rates remain high, and postrelapse treatment options are limited. Therefore, this study aimed to identify the factors contributing to the response to donor lymphocyte infusion (DLI) in relapsed MDS patients post-HSCT. METHODS This study included 107 patients with relapsed and DLI-treated MDS who underwent their first HSCT between 2002 and 2022 and were registered in the Transplant Registry Unified Program. Univariate and multivariate survival analyses were conducted using log-rank tests and Cox proportional hazards models. Overall survival (OS) and response rates to DLI were also analyzed. RESULTS The 1-year OS was 30.0% and univariate analysis identified poor prognostic factors: age ≥58 years (P = 0.003), complex karyotype (P = 0.026), hematologic relapse (P = 0.026) and early relapse (P = 0.004). Azacitidine plus DLI also improved prognosis (P < 0.001). Multivariate analysis confirmed age ≥58 years, hematologic relapse, and early relapse as poor prognostic factors. The adjusted OS for patients aged ≥58 years who relapsed <110 days post-transplant showed that the 1-year OS in patients with cytogenetic/molecular relapse was 43.6%, compared to 9.4% for those with hematologic relapse. Acute graft-versus-host disease (GVHD) occurred in 62.3% of patients, and chronic GVHD in 30.8%, with manageable outcomes. CONCLUSIONS DLI may improve OS in younger patients, those with cytogenetic/molecular relapse, and those with late relapse. Despite the risk of GVHD, its impact on prognosis is minimal. Given the limited treatment options, DLI should be considered for relapsed MDS patients post-HSCT.
Collapse
Affiliation(s)
- Atsushi Marumo
- Department of Hematology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan.
| | - Yasunobu Nagata
- Department of Hematology, Graduate School of Medicine, Nippon Medical School, Tokyo, Japan
| | - Machiko Fujioka
- Department of Hematology, Sasebo City General Hospital, Nagasaki, Japan
| | - Shuhei Kurosawa
- Department of Hematology, Yokohama Municipal Citizen's Hospital, Kanagawa, Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Emiko Sakaida
- Department of Hematology, Chiba University Hospital, Chiba, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Kentaro Fukushima
- Department of Hematology and Oncology, Osaka University Hospital, Tokyo, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Hokkaido, Japan
| | - Katsuhiro Shono
- Department of Hematology, Chiba Aoba Municipal Hospital, Chiba, Japan
| | - Ayumu Ito
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Naoyuki Uchida
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations TORANOMON HOSPITAL, Tokyo, Japan
| | - Tetsuya Nishida
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Tokyo, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Aichi, Japan
| | | | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Onizuka Makoto
- Department of Hematology/Oncology, Tokai University School of Medicine, Kanagawa, Japan
| | - Yoshinobu Kanda
- Division of Hematology, Jichi Medical University, Tochigi, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagoya, Japan; Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Aichi, Japan
| | - Hidehiro Itonaga
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
3
|
Liu FT, Wan CL, Huang YH, Cao HY, Lyu XY, Wang ZH, Huang SM, Tan KW, Ge SS, Zhang Y, Xue SL. Chidamide combined with azacitidine as a novel double epigenetic preemptive treatment for a myelodysplastic syndrome patient showing molecular relapse after allogeneic hematopoietic stem cell transplantation. Ann Hematol 2024; 103:6003-6006. [PMID: 39668198 DOI: 10.1007/s00277-024-06137-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024]
Abstract
Allogeneic hematopoietic stem cell transplantation (allo-HSCT) is theoretically the only curative option for high-risk myelodysplastic syndrome (HR MDS) patients. However, the management of patients with relapsed disease post allo-HSCT remains a challenge with few standard treatments. Chidamide, a new selective histone deacetylase, has shown synergistic anti-leukemia effect combined with azacitidine in acute myeloid leukemia patients. Herein, we reported a 50-year-old HR MDS patient who experienced molecular relapse post allo-HSCT and was successfully salvaged by preemptive treatment of chidamide combined with azacitidine (CHI-AZA). This patient maintained a deep remission lasting over 2 years by regular maintenance treatment of CHI-AZA, without sever treatment-related adverse events or increased risk of graft versus host disease. This case report demonstrated that double epigenetic regimen of CHI-AZA was effective and tolerable. Formally evaluating this regimen in HR MDS patients post allo-HSCT may be meaningful.
Collapse
Affiliation(s)
- Fang-Tong Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Chao-Ling Wan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yuan-Hong Huang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Han-Yu Cao
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Xiao-Yu Lyu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Zi-Hao Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Si-Man Huang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Kai-Wen Tan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Shuai-Shuai Ge
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| | - Yang Zhang
- Canglang Hospital of Suzhou, Suzhou, China.
| | - Sheng-Li Xue
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Shizi Street 188, Suzhou, 215006, P. R. China.
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China.
| |
Collapse
|
4
|
Getz TM, Bewersdorf JP, Kewan T, Stempel JM, Bidikian A, Shallis RM, Stahl M, Zeidan AM. Beyond HMAs: Novel Targets and Therapeutic Approaches. Semin Hematol 2024; 61:358-369. [PMID: 39389839 DOI: 10.1053/j.seminhematol.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024]
Abstract
Myelodysplastic syndromes/neoplasms (MDS) constitute a heterogeneous group of clonal hematopoietic disorders with extremely variable clinical features and outcomes. Management of MDS is largely based on risk stratification of patients into either lower-risk or higher-risk categories using the International Prognostic Scoring System-Revised and, more recently, on the Molecular International Prognostic Scoring System. Lower-risk MDS is often managed with the goal of ameliorating cytopenias and improving quality of life, while higher-risk MDS is treated with therapies aimed at extending survival and delaying progression to acute myeloid leukemia (AML). Therapeutic strategies in lower-risk MDS patients may consist of erythropoiesis stimulating agents, luspatercept, and lenalidomide for selected patients. Furthermore, imetelstat has recently been added to the FDA-approved therapeutic armamentarium for lower-risk MDS. In higher-risk MDS, monotherapy with hypomethylating agents continues to be the standard of care. While several novel hypomethylating agent combinations have and are being studied in large randomized phase 3 clinical trials, including the combination of azacitidine and venetoclax, no combination to date have improved overall survival to azacitidine monotherapy. Moreover, biomarker-directed therapies as well as immonotherapeutic approaches are currently being evaluated in early phase trials. Despite recent advancements, the lack of therapeutic agents, particularly after the failure of first line therapy in higher risk MDS, continues to be a major hurdle in the management of MDS. In this review, we discuss the current treatment landscape of MDS and provide an overview of novel agents currently in clinical development that have the potential to alter our current treatment paradigms.
Collapse
Affiliation(s)
- Ted M Getz
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut.
| | - Jan P Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut; Department of Medicine, Memorial Sloan Kettering Cancer Center, Leukemia Service, New York, New York
| | - Tariq Kewan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Jessica M Stempel
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Aram Bidikian
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Rory M Shallis
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| |
Collapse
|
5
|
Schulz E, Pavletic SZ, Mina A. Allogeneic hematopoietic cell transplantation in elderly patients with myelodysplastic syndromes: Considerations and challenges. Semin Hematol 2024; 61:420-430. [PMID: 39523201 PMCID: PMC11646184 DOI: 10.1053/j.seminhematol.2024.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Myelodysplastic syndromes/neoplasms (MDS) and related diseases are highly heterogeneous myeloid stem cell cancers that predominantly affect the elderly. The only curative treatment is allogeneic hematopoietic cell transplantation (HCT). Given the prevalence of age-related comorbidities, HCT in patients aged 65 years or older requires a highly personalized approach. This review summarizes disease risk stratification, treatment modalities, and outcomes for patients with MDS and related disorders, and discusses specific considerations and challenges for elderly patients undergoing HCT, including geriatric assessment, timing, conditioning treatment, donor and graft selection, and graft-versus-host disease prophylaxis.
Collapse
Affiliation(s)
- Eduard Schulz
- NIH Myeloid Malignancies Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; Immune Deficiency - Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Steven Z Pavletic
- NIH Myeloid Malignancies Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; Immune Deficiency - Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Alain Mina
- NIH Myeloid Malignancies Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD; Immune Deficiency - Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| |
Collapse
|
6
|
Liu Y, Calzone K, McReynolds LJ. Genetic predisposition to myelodysplastic syndrome: Genetic counseling and transplant implications. Semin Hematol 2024; 61:370-378. [PMID: 39443230 DOI: 10.1053/j.seminhematol.2024.09.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/14/2024] [Accepted: 09/17/2024] [Indexed: 10/25/2024]
Abstract
The development of myelodysplastic syndromes (MDS) is influenced by various genetic predispositions. Several important genes contribute to disease susceptibility. This paper explores common genetic predisposition genes in MDS, including DDX41, CEBPA, and SAMD9/SAMD9L, which are linked to hereditary conditions presenting diagnostic and clinical challenges. It delves into hereditary conditions that affect platelet production and count, such as RUNX1, ETV6, and ANKRD26, detailing their clinical features and how they contribute to an increased risk of MDS. The discussion extends to additional genetic syndromes like GATA2 deficiency, telomere biology disorders, Fanconi anemia, and Li-Fraumeni syndrome, along with new findings on genes like ERG that offer new insights into disease etiology. The importance of genetic counseling in MDS is underscored, outlining its goals, methods for evaluating family history, risk assessment, and the ethical considerations involved. Furthermore, the role of hematopoietic cell transplantation in managing MDS, particularly in patients with germline syndromes, is reviewed, emphasizing the need for optimal donor selection and personalized treatment approaches. This comprehensive overview illustrates the critical role of genetic factors in MDS and highlights the need for continued research and tailored clinical practices to improve patient outcomes.
Collapse
Affiliation(s)
- Yi Liu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Kathleen Calzone
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lisa J McReynolds
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Rockville, MD
| |
Collapse
|
7
|
Chalandon Y, Eikema DJ, Moiseev I, Ciceri F, Koster L, Vydra J, Passweg J, Rovira M, Ozcelik T, Gedde-Dahl T, Kröger N, Potter V, Yakoub-Agha I, Rambaldi A, Itälä-Remes M, Tanase A, Onida F, Gurnari C, Scheid C, Drozd-Sokolowska J, Raj K, McLornan DP, Robin M. Unrelated donor transplantation with posttransplant cyclophosphamide vs ATG for myelodysplastic neoplasms. Blood Adv 2024; 8:4792-4802. [PMID: 39008719 PMCID: PMC11414667 DOI: 10.1182/bloodadvances.2024013468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 06/26/2024] [Accepted: 06/27/2024] [Indexed: 07/17/2024] Open
Abstract
ABSTRACT It has been reported in prospective randomized trials that antithymocyte globulin (ATG)-based graft-versus-host disease (GVHD) prophylaxis has benefits in the setting of allogeneic hematopoietic stem cell transplantation (allo-HSCT) with unrelated donors (UDs). However, the optimal GVHD prophylaxis strategy has been challenged recently by the increasing use of posttransplant cyclophosphamide (PTCY). We report from the European Society for Blood and Marrow Transplantation registry the outcomes of 960 patients with myelodysplastic neoplasms who underwent allo-HSCT from UD with PTCY or ATG as GVHD prophylaxis. The primary outcomes were overall survival (OS) and progression-free survival (PFS). The disease characteristics were similar in both groups. Day 28 neutrophil engraftment was significantly better with ATG (93% vs 85%). Over a median follow-up of 4.4 years, the 5-year OS was 58% with PTCY, and 49% in the ATG group. The 5-year PFS was higher for PTCY at 53% vs 44% for ATG. Grade 2 to 4 acute GVHD incidence was lower when PTCY was used (23%), whereas there was no difference in the incidence of chronic GVHD at 5 years. Multivariable analyses confirmed better OS and PFS with PTCY with a hazard ratio (HR) for ATG of 1.32 (1-1.74) and a better PFS for PTCY with a HR for ATG of 1.33. This study suggests that GVHD prophylaxis using PTCY instead of ATG in this setting remains a valid option. Further prospective randomized studies would be essential to confirm these results.
Collapse
Affiliation(s)
- Yves Chalandon
- Hôpitaux Universitaires de Genève and Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | | | - Ivan Moiseev
- RM Gorbacheva Research Institute, Pavlov University, St. Petersburg, Russia
| | | | | | - Jan Vydra
- Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | | | - Montserrat Rovira
- Bone Marrow Transplantation Unit, Haematology Department, Institute of Haematology and Oncology, IDIBAPS, Hospital Clinic, University of Barcelona, Barcelona, Spain
| | - Tulay Ozcelik
- Demiroglu Bilim University Istanbul Florence Nightingale Hospital, Istanbul, Turkey
| | | | | | | | | | - Alessandro Rambaldi
- Department of Oncology and Hematology, University of Milan and Azienda Socio Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | | | | | - Francesco Onida
- Hematology and BMT Unit, ASST Fatebenefratelli-Sacco, University of Milan, Milan, Italy
| | - Carmelo Gurnari
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | | | - Kavita Raj
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Donal P. McLornan
- University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Marie Robin
- Hôpital Saint-Louis, APHP, Université de Paris Cité, Paris, France
| |
Collapse
|
8
|
Jung D, Long-Boyle JR, Pang WW, Gobburu JVS. Pharmacokinetics of Briquilimab as a Conditioning Agent for Hematopoietic Stem Cell Transplantation in Patients With Severe Combined Immunodeficiency, Myelodysplastic Syndrome, or Acute Myeloid Leukemia. Transplant Cell Ther 2024; 30:923.e1-923.e9. [PMID: 38972509 DOI: 10.1016/j.jtct.2024.07.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/09/2024]
Abstract
For successful engraftment of donor hematopoietic stem cells (HSC), conditioning with chemotherapy and/or radiation prior to hematopoietic cell transplantation (HCT) has been required to open marrow niche space and minimize the risk of immune rejection. Briquilimab, a humanized IgG1 monoclonal antibody that blocks the interaction between the c-Kit receptor and stem cell factor on various C-Kit expressing tissues including HSC, is a potential nonmyeloablative conditioning agent in clinical development for patients with severe combined immunodeficiency (SCID), myelodysplastic syndromes (MDS), and acute myeloid leukemia (AML). This study aimed to characterize pharmacokinetics (PK) and develop a population PK model of briquilimab after single intravenous infusions of 4 different doses in patients with SCID, MDS, or AML receiving HCT. The PK data was collected from 2 different studies: JAS-BMT-CP-001 and JSP-CP-003. JAS-BMT-CP-001 is a phase 1/2 open-label study of briquilimab as a conditioning agent prior to allogenic HCT in SCID patients. The participants received single intravenous infusions of 0.1, 0.3, 0.6, or 1.0 mg/kg. JSP-CP-003 was a phase 1a/b open-label study of briquilimab in combination with a standard conditioning regimen of low dose total body irradiation and fludarabine in MDS or AML subjects undergoing HCT. The participants received a single intravenous dose of 0.6 mg/kg briquilimab. In both studies, briquilimab PK samples were obtained at pre-treatment, 5 minutes post-end of infusion, 4- and 24-hours post-start of infusion, any time between 2 days and 30 days postinfusion, and on the day of HCT prior to donor cell infusion.The population PK model was developed using the PK data from these 2 clinical studies, and the effect of participants' baseline characteristics on the briquilimab PK was evaluated. PK simulations were performed using the developed PK model to calculate the time to reach target concentrations for HCT. A total of 49 participants (21 SCID adult and pediatric participants with a median age of 12 yr and 28 MDS/AML adult participants with a median age of 70 yr) were included in the PK analysis. A 2-compartment model with combined linear and non-linear elimination best described the PK of briquilimab. Body weight was determined as the sole covariate of the PK parameters among the explored covariates. For a typical subject with a body weight 70 kg, the estimated parameters for clearance, maximum metabolic rate of Michaelis Menten elimination, Michaelis Menten constant, central volume, peripheral volume, and intercompartmental clearance were 17.6 mL/h, 51,434.8 ng/h, 71.5 ng/mL, 3444.0 mL, 1613.3 mL, and 21.2 mL/h, respectively. The median time to reach target concentrations of 500, 1000, and 2000 ng/mL after a single dose of 0.6 mg/kg was calculated as 12.3, 10.4, and 7.7 days, respectively. The PK of intravenous briquilimab was characterized in subjects with SCID, MDS, or AML receiving HCT, and a population PK model was developed to estimate briquilimab clearance to use as a guide to the timing of donor cell infusion post-briquilimab. Body weight was identified as a significant covariate on elimination and volume of distribution of briquilimab.
Collapse
Affiliation(s)
- Dawoon Jung
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland
| | - Janel R Long-Boyle
- Division of Allergy, Immunology, and Bone Marrow Transplantation, Department of Pediatrics, University of California San Francisco, San Francisco, California
| | - Wendy W Pang
- Jasper Therapeutics Inc., Redwood City, California
| | - Jogarao V S Gobburu
- Center for Translational Medicine, University of Maryland School of Pharmacy, Baltimore, Maryland.
| |
Collapse
|
9
|
Notarantonio AB, Robin M, D'Aveni M. Current challenges in conditioning regimens for MDS transplantation. Blood Rev 2024; 67:101223. [PMID: 39089962 DOI: 10.1016/j.blre.2024.101223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 07/07/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
Myelodysplastic syndrome (MDS) is a very heterogeneous clonal disorder. Patients with "higher-risk" MDS, defined by specific recurrent genetic abnormalities, have a poor prognosis because of a high risk of progression to secondary acute myeloid leukemia with low chemosensitivity. Allogeneic hematopoietic stem cell transplantation remains the only treatment that offers durable disease control because the donor immune system allows graft-versus-MDS effects. In terms of preparation steps before transplantation, targeting the malignant clone by increasing the conditioning regimen intensity is still a matter of intense debate. MDS is mainly diagnosed in older patients, and high toxicity related to common myeloablative conditioning regimens has been reported. Efforts to include new drugs in the conditioning regimen to achieve the best malignant clone control without increasing toxicity have been made over the past 20 years. We summarized these retrospective and prospective studies and evaluated the limitations of the available evidence to delineate the ideal conditioning regimen.
Collapse
Affiliation(s)
- A B Notarantonio
- Hematology Department, University Hospital of Nancy, France; CNRS 7365, IMoPA, University of Lorraine, F-54000, France
| | - M Robin
- Hematology Department, Saint-Louis Hospital, APHP, Paris, France
| | - M D'Aveni
- Hematology Department, University Hospital of Nancy, France; CNRS 7365, IMoPA, University of Lorraine, F-54000, France.
| |
Collapse
|
10
|
Sabile JMG, Swords R, Tyner JW. Evaluating targeted therapies in older patients with TP53-mutated AML. Leuk Lymphoma 2024; 65:1201-1218. [PMID: 38646877 DOI: 10.1080/10428194.2024.2344057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 04/23/2024]
Abstract
Mutation of thetumor suppressor gene, TP53 (tumor protein 53), occurs in up to 15% of all patients with acute myeloid leukemia (AML) and is enriched within specific clinical subsets, most notably in older adults, and including secondary AML cases arising from preceding myeloproliferative neoplasm (MPN), myelodysplastic syndrome (MDS), patients exposed to prior DNA-damaging, cytotoxic therapies. In all cases, these tumors have remained difficult to effectively treat with conventional therapeutic regimens. Newer approaches fortreatmentofTP53-mutated AML have shifted to interventions that maymodulateTP53 function, target downstream molecular vulnerabilities, target non-p53 dependent molecular pathways, and/or elicit immunogenic responses. This review will describe the basic biology of TP53, the clinical and biological patterns of TP53 within myeloid neoplasms with a focus on elderly AML patients and will summarize newer therapeutic strategies and current clinical trials.
Collapse
Affiliation(s)
- Jean M G Sabile
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Ronan Swords
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Division of Hematology & Medical Oncology, Department of Medicine, Oregon Health & Science University, Portland, OR, USA
| | - Jeffrey W Tyner
- Knight Cancer Institute, Oregon Health & Science University, Portland, OR, USA
- Department of Cell, Developmental & Cancer Biology, Oregon Health & Science University, Portland, OR, USA
| |
Collapse
|
11
|
Gener-Ricos G, Rodriguez-Sevilla JJ, Urrutia S, Bataller A, Bazinet A, Garcia-Manero G. Advances in the management of higher-risk myelodysplastic syndromes: future prospects. Leuk Lymphoma 2024; 65:1233-1244. [PMID: 38712556 DOI: 10.1080/10428194.2024.2344061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/08/2024]
Abstract
Higher-risk myelodysplastic syndromes (HR-MDS) are defined using a number of prognostic scoring systems that include the degree of cytopenias, percentage of blasts, cytogenetic alterations, and more recently genomic data. HR-MDS encompasses characteristics such as progressive cytopenias, increased bone marrow blasts, unfavorable cytogenetics, and an adverse mutational profile. Survival is generally poor, and patients require therapy to improve outcomes. Hypomethylating agents (HMAs), such as azacitidine, decitabine, and more recently, oral decitabine/cedazuridine, are the only approved therapies for HR-MDS. These are often continued until loss of response, progression, or unacceptable toxicity. Combinations including an HMA plus other drugs have been investigated but have not demonstrated better outcomes compared to single-agent HMA. Moreover, in a disease of high genomic complexity such as HR-MDS, therapy targeting specific genomic abnormalities is of interest. This review will examine the biological underpinnings of HR-MDS, its therapeutic landscape in the frontline and relapsed settings, as well as the impact of hematopoietic stem cell transplantation, the only known curative intervention for this disease.
Collapse
Affiliation(s)
- Georgina Gener-Ricos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Samuel Urrutia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
12
|
Cutler C. Revisiting Timing and Decision Modeling for Allogeneic Hematopoietic Stem-Cell Transplantation in Myelodysplastic Syndromes. J Clin Oncol 2024; 42:2843-2848. [PMID: 38941577 DOI: 10.1200/jco.24.00649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 04/22/2024] [Indexed: 06/30/2024] Open
Abstract
The Oncology Grand Rounds series is designed to place original reports published in the Journal into clinical context. A case presentation is followed by a description of diagnostic and management challenges, a review of the relevant literature, and a summary of the authors' suggested management approaches. The goal of this series is to help readers better understand how to apply the results of key studies, including those published in Journal of Clinical Oncology, to patients seen in their own clinical practice.
Collapse
Affiliation(s)
- Corey Cutler
- Division of Transplantation and Cellular Therapy, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| |
Collapse
|
13
|
Itonaga H, Miyazaki Y, Fujioka M, Aoki J, Doki N, Nishida T, Fukuda T, Uchida N, Ueda Y, Uehara Y, Katayama Y, Ota S, Kawakita T, Kato J, Matsuoka KI, Eto T, Onizuka M, Ichinohe T, Atsuta Y, Ishiyama K. Prognostic impact of the conditioning intensity on outcomes after allogeneic transplantation for MDS with low blasts: a nationwide retrospective study by the adult MDS working group of the Japan Society for Transplantation and Cellular Therapy. Bone Marrow Transplant 2024; 59:1107-1117. [PMID: 38714756 DOI: 10.1038/s41409-024-02297-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 04/23/2024] [Accepted: 04/24/2024] [Indexed: 05/10/2024]
Abstract
Poor prognostic factors, such as transfusion dependency and chromosomal risk, need to be considered in the indication of allogeneic hematopoietic cell transplantation (allo-HCT) for patients harboring myelodysplastic syndromes with less than 5% marrow blasts (MDS-Lo). We analyzed the post-transplant outcomes of 1229 MDS-Lo patients who received myeloablative (MAC)(n = 651), reduced-intensity (RIC)(n = 397), and non-myeloablative conditioning (NMAC) regimens (n = 181). The multivariate analysis revealed that the RIC group had better chronic graft-versus-host disease (GVHD)- and relapse-free survival (CRFS) (P = 0.021), and GVHD- and relapse-free survival (GRFS) than the MAC group (P = 0.001), while no significant differences were observed between the NMAC and MAC groups. In the subgroup analysis, the MAC group has better overall survival (P = 0.008) than the RIC group among patients with an HCT-comorbidity index (HCT-CI) score of 0, while the RIC group had better overall survival (P = 0.029) than the MAC group among those with an HCT-CI score ≥3. According to the type of conditioning regimen, total body irradiation 12 Gy-based MAC regimen showed better OS and CRFS than the other MAC regimen, and comparable outcomes to the RIC regimen. In conclusion, the RIC and NMAC regimens are promising options for MDS-Lo patients in addition to the MAC regimen.
Collapse
Affiliation(s)
- Hidehiro Itonaga
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan.
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Machiko Fujioka
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
- Department of Hematology, Sasebo City General Hospital, Sasebo, Japan
| | - Jun Aoki
- Department of Hematology, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Tetsuya Nishida
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Naoyuki Uchida
- Department of Hematology, Federation of National Public Service Personnel Mutual Aid Associations Toranomon Hospital, Tokyo, Japan
| | - Yasunori Ueda
- Department of Hematology/Oncology and Transfusion and Hemapheresis Center, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yasufumi Uehara
- Department of Hematology, Kitakyushu City Hospital Organization, Kitakyushu Municipal Medical Center, Kitakyushu, Japan
| | - Yuta Katayama
- Department of Hematology, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Toshiro Kawakita
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Jun Kato
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Ken-Ichi Matsuoka
- Department of Hematology and Oncology, Okayama University Hospital, Okayama, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Makoto Onizuka
- Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Tatsuo Ichinohe
- Department of Hematology and Oncology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
- Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Ken Ishiyama
- Department of Hematology, Center Hospital of the National Center for Global Health and Medicine, Tokyo, Japan
| |
Collapse
|
14
|
Sinanidis I, Hochman MJ, Tsai HL, Randall MP, Bonilla B, Varadhan R, Ambinder AJ, Jones RJ, DeZern AE, Karantanos T. Favorable outcomes in MDS and oligoblastic AML-MR after reduced-intensity conditioning allogeneic bone marrow transplantation with post-transplantation cyclophosphamide. Bone Marrow Transplant 2024; 59:1178-1180. [PMID: 38714755 DOI: 10.1038/s41409-024-02299-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Revised: 04/22/2024] [Accepted: 04/25/2024] [Indexed: 05/10/2024]
Affiliation(s)
- Ilias Sinanidis
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Michael J Hochman
- Division of Hematology, Department of Hematology and Medical Oncology, Emory University School of Medicine, Atlanta, GA, USA
| | - Hua-Ling Tsai
- Division of Quantitative Sciences, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Michael P Randall
- Division of Hematology and Oncology, Department of Medicine, University of California, San Francisco, CA, USA
| | - Brandon Bonilla
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Ravi Varadhan
- Division of Quantitative Sciences, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Alexander J Ambinder
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Richard J Jones
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA
| | - Amy E DeZern
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| | - Theodoros Karantanos
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD, USA.
| |
Collapse
|
15
|
Onida F, Gagelmann N, Chalandon Y, Kobbe G, Robin M, Symeonidis A, de Witte T, Itzykson R, Jentzsch M, Platzbecker U, Santini V, Sanz G, Scheid C, Solary E, Valent P, Greco R, Sanchez-Ortega I, Yakoub-Agha I, Pleyer L. Management of adult patients with CMML undergoing allo-HCT: recommendations from the EBMT PH&G Committee. Blood 2024; 143:2227-2244. [PMID: 38493484 DOI: 10.1182/blood.2023023476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 02/07/2024] [Accepted: 02/23/2024] [Indexed: 03/19/2024] Open
Abstract
ABSTRACT Chronic myelomonocytic leukemia (CMML) is a heterogeneous disease presenting with either myeloproliferative or myelodysplastic features. Allogeneic hematopoietic cell transplantation (allo-HCT) remains the only potentially curative option, but the inherent toxicity of this procedure makes the decision to proceed to allo-HCT challenging, particularly because patients with CMML are mostly older and comorbid. Therefore, the decision between a nonintensive treatment approach and allo-HCT represents a delicate balance, especially because prospective randomized studies are lacking and retrospective data in the literature are conflicting. International consensus on the selection of patients and the ideal timing of allo-HCT, specifically in CMML, could not be reached in international recommendations published 6 years ago. Since then, new, CMML-specific data have been published. The European Society for Blood and Marrow Transplantation (EBMT) Practice Harmonization and Guidelines (PH&G) Committee assembled a panel of experts in the field to provide the first best practice recommendations on the role of allo-HCT specifically in CMML. Recommendations were based on the results of an international survey, a comprehensive review of the literature, and expert opinions on the subject, after structured discussion and circulation of recommendations. Algorithms for patient selection, timing of allo-HCT during the course of the disease, pretransplant strategies, allo-HCT modality, as well as posttransplant management for patients with CMML were outlined. The keynote message is, that once a patient has been identified as a transplant candidate, upfront transplantation without prior disease-modifying treatment is preferred to maximize chances of reaching allo-HCT whenever possible, irrespective of bone marrow blast counts.
Collapse
Affiliation(s)
- Francesco Onida
- Department of Oncology and Hemato-Oncology, Hematology and Bone Marrow Transplantation Unit, Azienda Socio Sanitaria Territoriale Fatebenefratelli-Sacco, University of Milan, Milan, Italy
- European Society for Blood and Marrow Transplantation Practice Harmonization and Guidelines Committee, Barcelona, Spain
| | - Nico Gagelmann
- University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Yves Chalandon
- Division of Hematology, University Hospital of Geneva, Geneva, Switzerland
- Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Heinrich Heine University, Medical Faculty, Düsseldorf, Germany
| | - Marie Robin
- Service d'Hématologie Greffe, Hôpital Saint-Louis, L'Assistance Publique-Hôpitaux de Paris, Université de Paris Cité, Paris, France
| | - Argiris Symeonidis
- Department of Hematology, Olympion General Hospital and Rehabilitation Center, Patras, Greece
| | - Theo de Witte
- Department of Tumor Immunology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Raphael Itzykson
- Université Paris Cité, Génomes, Biologie Cellulaire et Thérapeutique U944, INSERM, Centre National de la Recherche Scientifique, Paris, France
- Département Hématologie et Immunologie, Hôpital Saint-Louis, L'Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Madlen Jentzsch
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Valeria Santini
- Myelodysplastic Syndromes Unit, Hematology, Dipartimento di Medicina Sperimentale e Clinica, Azienda Ospedaliero-Universitaria Careggi, University of Florence, Florence, Italy
| | - Guillermo Sanz
- University and Polytechnic Hospital La Fe and Health Research Institute La Fe, Valencia, Spain
- Centro de Investigacion Biomedica en Red Cancer, Instituto de Salud Carlos III, Madrid, Spain
| | - Christof Scheid
- Department of Internal Medicine I, University Hospital Cologne, Cologne, Germany
| | - Eric Solary
- Department of Hematology, INSERM Unité Mixte de Recherche 1287, Gustave Roussy Cancer Center, Villejuif, France
- Université Paris Saclay, Faculty of Medicine, Le Kremlin-Bicetre, France
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute for Hematology and Oncology, Medical University of Vienna, Vienna, Austria
| | - Raffaela Greco
- European Society for Blood and Marrow Transplantation Practice Harmonization and Guidelines Committee, Barcelona, Spain
- Unit of Hematology and Bone Marrow Transplantation, Istituto di Ricovero e Cura a Carattere Scientifico San Raffaele Hospital, Vita-Salute San Raffaele University, Milan, Italy
| | - Isabel Sanchez-Ortega
- European Society for Blood and Marrow Transplantation Practice Harmonization and Guidelines Committee, Barcelona, Spain
| | - Ibrahim Yakoub-Agha
- European Society for Blood and Marrow Transplantation Practice Harmonization and Guidelines Committee, Barcelona, Spain
- Centre Hospitalier Universitaire de Lille, University of Lille, INSERM U1286, Infinite, Lille, France
| | - Lisa Pleyer
- Austrian Group of Medical Tumor Therapy Study Group, Vienna, Austria
- Salzburg Cancer Research Institute, Center for Clinical Cancer and Immunology Trials, Salzburg, Austria
- 3rd Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
- Cancer Cluster Salzburg, Salzburg, Austria
| |
Collapse
|
16
|
Niscola P, Gianfelici V, Giovannini M, Piccioni D, Mazzone C, de Fabritiis P. Latest Insights and Therapeutic Advances in Myelodysplastic Neoplasms. Cancers (Basel) 2024; 16:1563. [PMID: 38672645 PMCID: PMC11048617 DOI: 10.3390/cancers16081563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Revised: 04/10/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic syndromes/neoplasms (MDSs) encompass a range of hematopoietic malignancies, commonly affecting elderly individuals. Molecular alterations in the hematopoietic stem cell compartment drive disease pathogenesis. Recent advancements in genomic profiling have provided valuable insights into the biological underpinnings of MDSs and have expanded therapeutic options, particularly for specific molecularly defined subgroups. This review highlights the diagnostic principles, classification updates, prognostic stratification systems, and novel treatments, which could inform future clinical trials and enhance the management of adult MDS patients, particularly for specific molecularly defined subgroups.
Collapse
Affiliation(s)
- Pasquale Niscola
- Division of Haematology, Sant’ Eugenio Hospital, 00144 Rome, Italy; (V.G.); (M.G.); (D.P.); (C.M.); (P.d.F.)
| | | | | | | | | | | |
Collapse
|
17
|
Mina A, Greenberg PL, Deeg HJ. How I reduce and treat posttransplant relapse of MDS. Blood 2024; 143:1344-1354. [PMID: 38306658 PMCID: PMC11443576 DOI: 10.1182/blood.2023023005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 02/04/2024] Open
Abstract
ABSTRACT Allogeneic hematopoietic stem cell transplantation (HSCT) is the only potentially curative option for patients with high-risk myelodysplastic syndromes (MDS). Advances in conditioning regimens and supportive measures have reduced treatment-related mortality and increased the role of transplantation, leading to more patients undergoing HSCT. However, posttransplant relapse of MDS remains a leading cause of morbidity and mortality for this procedure, necessitating expert management and ongoing results analysis. In this article, we review treatment options and our institutional approaches to managing MDS relapse after HSCT, using illustrative clinical cases that exemplify different clinical manifestations and management of relapse. We address areas of controversy relating to conditioning regimen intensity, chemotherapeutic bridging, and donor selection. In addition, we discuss future directions for advancing the field, including (1) the need for prospective clinical trials separating MDS from acute myeloid leukemia and focusing on posttransplant relapse, as well as (2) the validation of measurable residual disease methodologies to guide timely interventions.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Peter L. Greenberg
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
18
|
Karel D, Valburg C, Woddor N, Nava VE, Aggarwal A. Myelodysplastic Neoplasms (MDS): The Current and Future Treatment Landscape. Curr Oncol 2024; 31:1971-1993. [PMID: 38668051 PMCID: PMC11049094 DOI: 10.3390/curroncol31040148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic neoplasms (MDS) are a heterogenous clonal disorder of hemopoietic stem cells characterized by cytomorphologic dysplasia, ineffective hematopoiesis, peripheral cytopenias and risk of progression to acute myeloid leukemia (AML). Our understanding of this disease has continued to evolve over the last century. More recently, prognostication and treatment have been determined by cytogenetic and molecular data. Specific genetic abnormalities, such as deletion of the long arm of chromosome 5 (del(5q)), TP53 inactivation and SF3B1 mutation, are increasingly associated with disease phenotype and outcome, as reflected in the recently updated fifth edition of the World Health Organization Classification of Hematolymphoid Tumors (WHO5) and the International Consensus Classification 2022 (ICC 2022) classification systems. Treatment of lower-risk MDS is primarily symptom directed to ameliorate cytopenias. Higher-risk disease warrants disease-directed therapy at diagnosis; however, the only possible cure is an allogenic bone marrow transplant. Novel treatments aimed at rational molecular and cellular pathway targets have yielded a number of candidate drugs over recent years; however few new approvals have been granted. With ongoing research, we hope to increasingly offer our MDS patients tailored therapeutic approaches, ultimately decreasing morbidity and mortality.
Collapse
Affiliation(s)
- Daniel Karel
- Department of Hematology/Medical Oncology, The George Washington University, Washington, DC 20037, USA; (C.V.); (A.A.)
| | - Claire Valburg
- Department of Hematology/Medical Oncology, The George Washington University, Washington, DC 20037, USA; (C.V.); (A.A.)
| | - Navitha Woddor
- Department of Pathology, The George Washington University, Washington, DC 20037, USA; (N.W.); (V.E.N.)
| | - Victor E. Nava
- Department of Pathology, The George Washington University, Washington, DC 20037, USA; (N.W.); (V.E.N.)
- Department of Pathology, Washington DC VA Medical Center, Washington, DC 20422, USA
| | - Anita Aggarwal
- Department of Hematology/Medical Oncology, The George Washington University, Washington, DC 20037, USA; (C.V.); (A.A.)
- Department of Hematology/Medical Oncology, Washington DC VA Medical Center, Washington, DC 20422, USA
| |
Collapse
|
19
|
Shimomura Y, Kitamura T, Konuma T, Nakaya Y, Doki N, Sawa M, Nakamae H, Eto T, Nishida T, Ohigashi H, Ota S, Onizuka M, Hiramoto N, Kawakita T, Kanda J, Ichinohe T, Atsuta Y, Itonaga H. Hematopoietic stem cell transplantation from haploidentical offspring donors using post-transplant cyclophosphamide versus human leukocyte antigen-matched siblings in older patients with myelodysplastic syndrome. Am J Hematol 2024; 99:E42-E45. [PMID: 37966972 DOI: 10.1002/ajh.27160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 10/31/2023] [Accepted: 11/02/2023] [Indexed: 11/17/2023]
Affiliation(s)
- Yoshimitsu Shimomura
- Department of Hematology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Kobe, Japan
- Department of Environmental Medicine and Population Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Tetsuhisa Kitamura
- Department of Environmental Medicine and Population Science, Graduate School of Medicine, Osaka University, Suita, Japan
| | - Takaaki Konuma
- Department of Hematology/Oncology, The Institute of Medical Science, The University of Tokyo, Tokyo, Japan
| | - Yosuke Nakaya
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Anjo-shi, Japan
| | - Hirohisa Nakamae
- Department of Hematology, Osaka Metropolitan University Graduate School of Medicine, Osaka, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Tetsuya Nishida
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya-shi, Japan
| | - Hiroyuki Ohigashi
- Department of Hematology, Hokkaido University Hospital, Sapporo, Japan
| | - Shuichi Ota
- Department of Hematology, Sapporo Hokuyu Hospital, Sapporo, Japan
| | - Makoto Onizuka
- Department of Hematology/Oncology, Tokai University School of Medicine, Isehara, Japan
| | - Nobuhiro Hiramoto
- Department of Hematology, Kobe City Hospital Organization Kobe City Medical Center General Hospital, Kobe, Japan
| | - Toshiro Kawakita
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Junya Kanda
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tatsuo Ichinohe
- Department of Hematology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Nagakute, Japan
- Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Nagakute, Japan
| | - Hidehiro Itonaga
- Transfusion and Cell Therapy Unit, Nagasaki University Hospital, Nagasaki, Japan
| |
Collapse
|
20
|
Hellström-Lindberg ES, Kröger N. Clinical decision-making and treatment of myelodysplastic syndromes. Blood 2023; 142:2268-2281. [PMID: 37874917 DOI: 10.1182/blood.2023020079] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/22/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
ABSTRACT The myelodysplastic syndromes (MDSs) constitute a profoundly heterogeneous myeloid malignancy with a common origin in the hemopoietic stem cell compartment. Consequently, patient management and treatment are as heterogeneous. Decision-making includes identifying risk, symptoms, and options for an individual and conducting a risk-benefit analysis. The only potential cure is allogeneic stem cell transplantation, and albeit the fraction of patients with MDS who undergo transplant increase over time because of better management and increased donor availability, a majority are not eligible for this intervention. Current challenges encompass to decrease the relapse risk, the main cause of hematopoietic stem cell transplantation failure. Hypomethylating agents (HMAs) constitute firstline treatment for higher-risk MDSs. Combinations with other drugs as firstline treatment has, to date, not proven more efficacious than monotherapy, although combinations approved for acute myeloid leukemia, including venetoclax, are under evaluation and often used as rescue treatment. The treatment goal for lower-risk MDS is to improve cytopenia, mainly anemia, quality of life, and, possibly, overall survival. Erythropoiesis-stimulating agents (ESAs) constitute firstline treatment for anemia and have better and more durable responses if initiated before the onset of a permanent transfusion need. Treatment in case of ESA failure or ineligibility should be tailored to the main disease mechanism: immunosuppression for hypoplastic MDS without high-risk genetics, lenalidomide for low-risk del(5q) MDS, and luspatercept for MDS with ring sideroblasts. Approved therapeutic options are still scarcer for MDS than for most other hematologic malignancies. Better tools to match disease biology with treatment, that is, applied precision medicines are needed to improve patient outcome.
Collapse
Affiliation(s)
- Eva S Hellström-Lindberg
- Department of Medicine, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
21
|
Qi SS, Chen Z, Du Y, Sun M, Wang Z, Long F, Luo L, Xiong H. Prophylactic donor lymphocyte infusion after haploidentical hematopoietic cell transplantation and post-transplant cyclophosphamide for treatment of high-risk myeloid neoplasms in children: A retrospective study. Pediatr Blood Cancer 2023; 70:e30659. [PMID: 37669234 DOI: 10.1002/pbc.30659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 08/07/2023] [Accepted: 08/24/2023] [Indexed: 09/07/2023]
Abstract
BACKGROUND Post-transplant cyclophosphamide (PTCy) has been recommended for prevention of graft-versus-host disease (GvHD) following haploidentical hematopoietic cell transplantation (haplo-HCT) for treatment of malignant blood diseases, but disease relapse remains a problem. Although donor lymphocyte infusion (DLI) is reported to be effective for treating post-transplantation relapse, the efficacy and safety of prophylactic-DLI (pro-DLI) post haplo-HCT, and PTCy in pediatric patients with hematological malignancies is unknown. METHODS We retrospectively analyzed the outcomes of 54 pediatric patients with high-risk myeloid neoplasms who received a PTCy regimen for GvHD prophylaxis and pro-DLI after haploidentical peripheral blood stem cell transplantation. The high-risk myeloid neoplasms in this cohort included acute myeloid leukemia (n = 46) and myelodysplastic syndromes (n = 8). RESULTS Median follow-up was for 19.7 (range: 3.4-46.6) months. The cumulative incidences of grade II-IV and III-IV acute GvHD were 37.0% (95% CI: 22.7%-48.7%) and 16.7% (95% CI: 6.1%-26.0%), respectively. There were no graft-failure events, and the 2-year rate of moderate/severe chronic GvHD was 8.1% (95% CI: 0%-16.7%). The 2-year non-relapse mortality, relapse, disease-free survival, GvHD-free relapse-free survival, and overall survival rates were 5.1% (95% CI: 0%-11.7%), 16.6% (95% CI: 5.3%-26.6%), 78.9% (95% CI: 68.0%-91.6%), 62.2% (95% CI: 49.4%-78.3%), and 87.3% (95% CI: 78.3%-97.4%), respectively. CONCLUSIONS Prophylactic donor lymphocyte infusion in the setting of haploidentical hematopoietic cell transplantation with post-transplant cyclophosphamide appears to be effective and safe in pediatric patients with high-risk myeloid neoplasms.
Collapse
Affiliation(s)
- Shan-Shan Qi
- Laboratory of Pediatric Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhi Chen
- Department of Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Du
- Department of Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ming Sun
- Laboratory of Pediatric Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhuo Wang
- Department of Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Fei Long
- Laboratory of Pediatric Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Linlin Luo
- Department of Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hao Xiong
- Laboratory of Pediatric Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Hematology, Wuhan Children's Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
22
|
Kröger N. Chronological Age Alone Is not Decisive. DEUTSCHES ARZTEBLATT INTERNATIONAL 2023; 120:616. [PMID: 37811849 PMCID: PMC10568736 DOI: 10.3238/arztebl.m2023.0134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
|
23
|
Garcia-Manero G. Myelodysplastic syndromes: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol 2023; 98:1307-1325. [PMID: 37288607 DOI: 10.1002/ajh.26984] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
DISEASE OVERVIEW The myelodysplastic syndromes (MDS) are a very heterogeneous group of myeloid disorders characterized by peripheral blood cytopenias and increased risk of transformation to acute myelogenous leukemia (AML). MDS occurs more frequently in older males and in individuals with prior exposure to cytotoxic therapy. DIAGNOSIS Diagnosis of MDS is based on morphological evidence of dysplasia upon visual examination of a bone marrow aspirate and biopsy. Information obtained from additional studies such as karyotype, flow cytometry, and molecular genetics is usually complementary and may help refine diagnosis. A new WHO classification of MDS was proposed in 2022. Under this classification, MDS is now termed myelodysplastic neoplasms. RISK-STRATIFICATION Prognosis of patients with MDS can be calculated using a number of scoring systems. All these scoring systems include analysis of peripheral cytopenias, percentage of blasts in the bone marrow, and cytogenetic characteristics. The most commonly accepted system is the Revised International Prognostic Scoring System (IPSS-R). Recently, genomic data has been incorporated resulting in the new IPSS-M classification. RISK-ADAPTED THERAPY Therapy is selected based on risk, transfusion needs, percent of bone marrow blasts, cytogenetic and mutational profiles, comorbidities, potential for allogeneic stem cell transplantation (alloSCT), and prior exposure to hypomethylating agents (HMA). Goals of therapy are different in lower risk patients than in higher risk and in those with HMA failure. In lower risk, the goal is to decrease transfusion needs and transformation to higher risk disease or AML, as well as to improve survival. In higher risk, the goal is to prolong survival. In 2020, two agents were approved in the US for patients with MDS: luspatercept and oral decitabine/cedazuridine. In addition, currently other available therapies include growth factors, lenalidomide, HMAs, intensive chemotherapy, and alloSCT. A number of phase 3 combinations studies have been completed or are ongoing at the time of this report. At the present time there are no approved interventions for patients with progressive or refractory disease particularly after HMA based therapy. In 2021, several reports indicated improved outcomes with alloSCT in MDS as well as early results from clinical trials using targeted intervention.
Collapse
Affiliation(s)
- Guillermo Garcia-Manero
- Section of MDS, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| |
Collapse
|
24
|
Juckett M, Dandoy C, DeFilipp Z, Kindwall-Keller TL, Spellman SR, Ustun C, Waldman BM, Weisdorf DJ, Wood WA, Horowitz MM, Burns LJ, Khera N. How do we improve the translation of new evidence into the practice of hematopoietic cell transplantation and cellular therapy? Blood Rev 2023; 60:101079. [PMID: 37087394 PMCID: PMC10330269 DOI: 10.1016/j.blre.2023.101079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/05/2023] [Accepted: 04/06/2023] [Indexed: 04/24/2023]
Abstract
The field of hematopoietic cell transplantation and cell therapy (HCT/CT) is advancing rapidly to bring an ever-expanding collection of potentially curative therapies to patients with malignant and non-malignant diseases. The impact of these therapies depends on our ability to implement them as new evidence becomes available to advance the quality of care. There is often a long delay between evidence development and adoption of therapies based on that evidence into clinical practice. In this review, we describe the potential factors based on an implementation framework that could act as facilitators or barriers to adoption of therapies in the context of HCT/CT. We highlight two examples, the first to showcase the efforts to improve the efficiency of adoption of new findings and accelerate improvement in care of HCT/CT patients and the second to discuss the challenges in real world implementation of chimeric antigen receptor T cell therapy. We conclude by reviewing strategies to improve translation of evidence and ways to measure their success.
Collapse
Affiliation(s)
- Mark Juckett
- University of Minnesota, Minneapolis, MN, United States of America
| | - Christopher Dandoy
- University of Cincinnati College of Medicine, Cincinnati, OH, United States of America
| | | | | | - Stephen R Spellman
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Milwaukee, WI, United States of America
| | - Celalettin Ustun
- Rush University Medical Center, Chicago, IL, United States of America
| | - Bryce M Waldman
- Center for International Blood and Marrow Transplant, Milwaukee, WI, United States of America
| | | | - William A Wood
- University of North Carolina, Chapel Hill, NC, United States of America
| | - Mary M Horowitz
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Milwaukee, WI, United States of America; Medical College of Wisconsin, Milwaukee, WI, United States of America
| | - Linda J Burns
- CIBMTR® (Center for International Blood and Marrow Transplant Research), Milwaukee, WI, United States of America
| | - Nandita Khera
- College of Medicine, Mayo Clinic, Phoenix, AZ, United States of America.
| |
Collapse
|
25
|
Itonaga H, Miyazaki Y, Aoki K, Shingai N, Ozawa Y, Fukuda T, Kataoka K, Kawakita T, Ueda Y, Ara T, Tanaka M, Katayama Y, Sawa M, Eto T, Kanda J, Atsuta Y, Ishiyama K. Allogeneic transplantation of bone marrow versus peripheral blood stem cells from HLA-identical relatives in patients with myelodysplastic syndromes and oligoblastic acute myeloid leukemia: a propensity score analysis of a nationwide database. Ann Hematol 2023; 102:1215-1227. [PMID: 36918415 DOI: 10.1007/s00277-023-05167-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Accepted: 03/06/2023] [Indexed: 03/15/2023]
Abstract
Bone marrow (BM) and granulocyte colony-stimulating factor-mobilized peripheral blood stem cells (PBSC) are used as grafts from HLA-identical-related donors for adults with myelodysplastic syndrome (MDS). To assess the impact of graft sources on post-transplant outcomes in MDS patients, we conducted a retrospective analysis of a nationwide database. A total of 247 and 280 patients underwent transplantation with BM and PBSC, respectively. The inverse probability of treatment weighting (IPTW) methods revealed that overall survival (OS) was comparable between BM and PBSC (P = .129), but PBSC transplantation was associated with worse graft-versus-host disease (GVHD)-free/relapse-free survival (GRFS) (hazard rate [HR], 1.24; 95% confidence intervals [CIs], 1.00-1.53; P = 0.049) and chronic GVHD-free and relapse-free survival (CRFS) (HR, 1.29; 95% CIs, 1.13-1.73; P = 0.002) than BM transplantation. In the propensity score matched cohort (BM, n = 216; PBSC, n = 216), no significant differences were observed in OS and relapse; 3-year OS rates were 64.7% and 60.0% (P = 0.107), while 3-year relapse rates were 27.1% and 23.5% (P = 0.255) in BM and PBSC, respectively. Three-year GRFS rates (36.6% vs. 29.2%; P = 0.006), CRFS rate (37.7% vs. 32.5%; P = 0.003), and non-relapse mortality rates (13.9% vs. 21.1%; P = 0.020) were better in BM than in PBSC. The present study showed that BM transplantation provides a comparable survival benefit with PBSC transplantation and did not identify an enhanced graft-versus-MDS effect to reduce the incidence of relapse in PBSC transplantation.
Collapse
Affiliation(s)
- Hidehiro Itonaga
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan.
| | - Yasushi Miyazaki
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan.,Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Kazunari Aoki
- Laboratory of Stem Cell Genetics, Institute for Life and Medical Sciences, Kyoto University, Kyoto, Japan
| | - Naoki Shingai
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Yukiyasu Ozawa
- Department of Hematology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Aichi, Japan
| | - Takahiro Fukuda
- Department of Hematopoietic Stem Cell Transplantation, National Cancer Center Hospital, Tokyo, Japan
| | - Keisuke Kataoka
- Division of Hematology, Department of Medicine, Keio University School of Medicine, Tokyo, Japan.,Division of Molecular Oncology, National Cancer Center Research Institute, Tokyo, Japan
| | - Toshiro Kawakita
- Department of Hematology, National Hospital Organization Kumamoto Medical Center, Kumamoto, Japan
| | - Yasunori Ueda
- Department of Hematology/Oncology and Transfusion and Hemapheresis Center, Kurashiki Central Hospital, Okayama, Japan
| | - Takahide Ara
- Department of Hematology, Hokkaido University Hospital, Sapporo, Hokkaido, Japan
| | - Masatsugu Tanaka
- Department of Hematology, Kanagawa Cancer Center, Kanagawa, Japan
| | - Yuta Katayama
- Department of Hematology, Hiroshima Red Cross Hospital & Atomic-bomb Survivors Hospital, Hiroshima, Japan
| | - Masashi Sawa
- Department of Hematology and Oncology, Anjo Kosei Hospital, Aichi, Japan
| | - Tetsuya Eto
- Department of Hematology, Hamanomachi Hospital, Fukuoka, Japan
| | - Junya Kanda
- Department of Hematology and Oncology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yoshiko Atsuta
- Japanese Data Center for Hematopoietic Cell Transplantation, Aichi, Japan.,Department of Registry Science for Transplant and Cellular Therapy, Aichi Medical University School of Medicine, Aichi, Japan
| | - Ken Ishiyama
- Department of Hematology, Kanazawa University Hospital, Kanazawa, Japan
| |
Collapse
|