1
|
Appelbaum JS, Percival ME, Scott BL. A cure for the kiss of death? Blood Adv 2025; 9:2855-2856. [PMID: 40493363 DOI: 10.1182/bloodadvances.2025015850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/12/2025] Open
Affiliation(s)
- Jacob S Appelbaum
- Division of Hematology/Oncology, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Mary-Elizabeth Percival
- Division of Hematology/Oncology, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| | - Bart L Scott
- Division of Hematology/Oncology, University of Washington, Seattle, WA
- Clinical Research Division, Fred Hutchinson Cancer Center, Seattle, WA
| |
Collapse
|
2
|
Lontos K, Saliba RM, Kanagal-Shamanna R, Özcan G, Ramdial J, Chen G, Kadia T, Short NJ, Daver NG, Kantarjian H, Marin D, Kebriaei P, Popat U, Champlin R, Shpall E, Oran B. TP53-mutant variant allele frequency and cytogenetics determine prognostic groups in MDS/AML for transplantation. Blood Adv 2025; 9:2845-2854. [PMID: 39853269 DOI: 10.1182/bloodadvances.2024014499] [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: 08/12/2024] [Revised: 12/04/2024] [Accepted: 12/26/2024] [Indexed: 01/26/2025] Open
Abstract
ABSTRACT Results after hematopoietic stem cell transplantation (HSCT) for TP53-mutated myeloid malignancies are disappointing. Several HSCT centers decline to perform HSCT for patients with TP53 mutation because of poor outcomes. In this study, we analyzed 240 patients with TP53-mutated myelodysplastic syndrome (MDS) or acute myeloid leukemia (AML) who underwent HSCT. We aimed to identify the patients who benefit most from HSCT. The primary outcome was progression-free survival (PFS). Of the cohort, 52% had AML and the median age was 62 years. AML and MDS outcomes were similar. We identified several favorable prognostic factors for PFS, including absence of complex cytogenetics/5q deletion/7q deletion, a lower variant allele frequency (VAF), a monohit status, and use of a matched-related donor. Using classification and regression tree analysis, we identified VAF and cytogenetics as the 2 most important prognostic factors. Patients with TP53mut VAF ≥ 50% had a 2-year PFS of 3%, and patients with TP53mut VAF < 50% and complex/5q/7q cytogenetic abnormalities had 2-year PFS of 22%. Patients with TP53mut VAF < 50% and without complex/5q/7q cytogenetics had 2-year PFS of 60%. These data inform clinical practice and help patients decide whether to pursue HSCT.
Collapse
Affiliation(s)
- Konstantinos Lontos
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rima M Saliba
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gonca Özcan
- Department of Hematology Oncology, National Institutes of Health, Bethesda, MD
| | - Jeremy Ramdial
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - George Chen
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Nicholas James Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Partow Kebriaei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Uday Popat
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Richard Champlin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elizabeth Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Betül Oran
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
3
|
Shah MV, Arber DA, Hiwase DK. TP53 -Mutated Myeloid Neoplasms: 2024 Update on Diagnosis, Risk-Stratification, and Management. Am J Hematol 2025; 100 Suppl 4:88-115. [PMID: 40066944 PMCID: PMC12067166 DOI: 10.1002/ajh.27655] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 02/25/2025] [Accepted: 02/27/2025] [Indexed: 05/13/2025]
Abstract
Alterations in the tumor suppressor gene TP53 are common in human cancers and are associated with an aggressive nature. Approximately 8%-12% of myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) harbor TP53 mutations (TP53 mut) and present immense challenges due to inherent chemoresistance and poor outcomes. As TP53 mut are more common in older individuals and those with secondary/therapy-related myeloid neoplasms (MN), their incidence is expected to increase with an aging population and rising proportion of cancer survivors. Treatments used for other MN-intensive chemotherapy, hypomethylating agents, and the BCL-2 inhibitor venetoclax-do not improve the survival of TP53 mut MN patients meaningfully. Additionally, further development of many promising agents has been discontinued, highlighting the challenges. Widespread acknowledgment of these problems led to the recognition of TP53 mut MN as a distinct entity in the 5th edition of the World Health Organization and International Consensus Classifications. However, critical discrepancies between the two classifications may lead to under- or overestimation of the prognostic risk. Here, we review recent advances in the biology, diagnosis, and treatment of TP53 mut MN. The development of TP53 mut MN is positioned at the intersection of age, hereditary predisposition, and anti-cancer therapies. Precursor TP53 mut clones can be detected years prior to the eventual leukemic transformation-raising the possibility of early intervention. We discuss the two classification systems and the bearing of the discrepancies between the two on timely and effective management. We provide novel evidence in the areas of discrepancies. Finally, we review the current therapeutic landscape and the obvious limitations of the currently used therapies.
Collapse
Affiliation(s)
| | - Daniel A. Arber
- Department of PathologyUniversity of ChicagoChicagoIllinoisUSA
| | - Devendra K. Hiwase
- Department of Haematology, Royal Adelaide HospitalCentral Adelaide Local Health NetworkAdelaideSouth AustraliaAustralia
- Precision Medicine ThemeSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideSouth AustraliaAustralia
- Adelaide Medical SchoolUniversity of AdelaideAdelaideSouth AustraliaAustralia
| |
Collapse
|
4
|
Masuda Y, Sadato D, Toya T, Hosoda Y, Hirama C, Shimizu H, Najima Y, Harada H, Harada Y, Doki N. Transplantation outcomes of TP53-mutant AML and MDS: a single transplantation center experience of 63 patients. Int J Hematol 2025; 121:820-832. [PMID: 40011351 DOI: 10.1007/s12185-025-03951-z] [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: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/13/2025] [Indexed: 02/28/2025]
Abstract
Allogeneic hematopoietic stem cell transplantation is recommended for TP53-mutant acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS) despite a high relapse rate and poor survival. To understand TP53 alterations on a molecular level and define stratified prognostic outcomes following transplantation, we performed targeted next-generation sequencing on 63 patients who underwent transplantation for TP53-mutant AML/MDS and profiled their molecular spectrum. Sixty-eight TP53 mutations were detected, with a median variant allele frequency of 46.8%. Copy number alterations at the TP53 locus were present in 19 patients (30%). Complex karyotype was detected in 48 patients (76%) and was significantly associated with larger TP53 clone size, bi-allelic status, and the absence of concurrent mutations, reflecting the high TP53 mutational burden. Specifically, 51 patients (81%) with the dominant TP53 clone greatly overlapped with those with the complex karyotype. Multivariable overall survival (OS) analysis identified AML (hazard ratio [HR], 2.51; P = 0.03) and TP53 clonal dominance (HR, 5.30; P = 0.002) as prognostic factors. One-year OS was worse in AML with the dominant TP53 clone than in others (13% vs 61%; P < 0.001). Our results underscore the utility of mutational profile-guided risk stratification in patients with TP53-mutant AML/MDS, and could aid in transplantation-related decision-making.
Collapse
Affiliation(s)
- Yasutaka Masuda
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Daichi Sadato
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Takashi Toya
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan.
| | - Yuzuru Hosoda
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Chizuko Hirama
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Hiroaki Shimizu
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Yuho Najima
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| | - Hironori Harada
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
- Laboratory of Oncology, School of Life Sciences, Tokyo University of Pharmacy and Life Sciences, Tokyo, Japan
| | - Yuka Harada
- Clinical Research and Trials Center, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, Tokyo, Japan
| | - Noriko Doki
- Hematology Division, Tokyo Metropolitan Cancer and Infectious Diseases Center, Komagome Hospital, 3-18-22 Honkomagome, Bunkyo-Ku, Tokyo, 113-8677, Japan
| |
Collapse
|
5
|
Gagelmann N, Kröger N. Allogeneic Stem Cell Transplant for Myelofibrosis and Myelodysplastic Syndromes: A Contemporary Review. Am J Hematol 2025; 100 Suppl 4:16-29. [PMID: 40079242 PMCID: PMC12067163 DOI: 10.1002/ajh.27660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2024] [Revised: 12/31/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025]
Abstract
Allogeneic hematopoietic stem-cell transplantation (HCT) remains the only potentially curative therapy for patients with myelodysplastic neoplasms (MDS) and myelofibrosis (MF) and is the standard care for eligible patients with higher-risk disease. Despite significant advancements, both diseases pose unique challenges due to their clinical and molecular heterogeneity, necessitating personalized approaches to patient selection, timing, and transplant management. For MDS, genomic profiling has revolutionized prognostic frameworks such as IPSS-M, enabling tailored therapeutic decisions. In MF, driver mutations (e.g., JAK2, CALR, MPL) and additional high-risk molecular markers provide critical insights into disease biology and transplant outcomes. Optimal timing of HCT is critical, and recent models might help personalize treatment approaches. Molecular measurable residual disease monitoring has demonstrated prognostic value in both diseases, guiding preemptive strategies to mitigate relapse risk. Harnessing molecular technologies, clinical expertise, patient-centered decision-making, and innovative pharmaceutical strategies offers an exciting opportunity to shape a transformative and curative treatment framework. Here, we provide a contemporary review on HCT for MDS and MF, highlighting up-to-date insights into disease biology, standard of care, and recommendations, as well as open avenues.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell TransplantationUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| | - Nicolaus Kröger
- Department of Stem Cell TransplantationUniversity Medical Center Hamburg‐EppendorfHamburgGermany
| |
Collapse
|
6
|
McMahon C, Raddi MG, Mohan S, Santini V. New Approvals in Low- and Intermediate-Risk Myelodysplastic Syndromes. Am Soc Clin Oncol Educ Book 2025; 45:e473654. [PMID: 40334184 DOI: 10.1200/edbk-25-473654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Recent advances in defining low- and intermediate-risk myelodysplastic syndromes (MDSs) have emphasized the critical role of molecular characterization using next-generation sequencing (NGS). Molecular profiling significantly enhances diagnostic precision, classification, and risk stratification, thereby informing therapeutic decisions, including the timing of hematopoietic stem-cell transplantation (HSCT). The Molecular International Prognostic Scoring System integrates clinical and molecular data, reclassifying and upstaging a substantial number of patients compared with previous prognostic systems, possibly allowing for more tailored therapeutic interventions. The novel therapeutic agents luspatercept and imetelstat have been particularly impactful. Luspatercept, which is effective in lower-risk (LR)-MDS, especially but not only in SF3B1-mutated cases, promotes late-stage erythroid maturation and transfusion independence (TI). Imetelstat, a telomerase inhibitor, induces TI while demonstrating disease-modifying effects as it significantly reduces mutation allele frequencies in patients who respond. These agents exemplify personalized medicine, emphasizing treatment selection and timing based on individual molecular and clinical features. Current research focuses on optimizing therapeutic strategies and exploring combination treatments to improve clinical outcomes.
Collapse
Affiliation(s)
| | - Marco G Raddi
- MDS Unit, Hematology, DMSC, University of Florence, AOUC, Florence, Italy
| | - Sanjay Mohan
- Vanderbilt University School of Medicine, Nashville, TN
| | - Valeria Santini
- MDS Unit, Hematology, DMSC, University of Florence, AOUC, Florence, Italy
| |
Collapse
|
7
|
Richman LP, Waller BF, Lovitch SB, Jambhekar A. Detection of TP53 mutations by immunohistochemistry in acute myeloid leukemia varies with interpreter expertise and mutation status. Am J Clin Pathol 2025:aqaf047. [PMID: 40414698 DOI: 10.1093/ajcp/aqaf047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 04/14/2025] [Indexed: 05/27/2025] Open
Abstract
OBJECTIVE TP53 mutations, including missense and inactivating (frameshift, splice site, and nonsense) mutations, occur in approximately 10% of myeloid neoplasms and confer adverse outcomes. Classification of myeloid neoplasms by World Health Organization and International Consensus Classification standards recognizes the importance of early detection of TP53 mutations. p53 immunohistochemistry (IHC) is a widely accessible method used to detect mutations; however, previous studies have demonstrated variable accuracy, especially for inactivating TP53 mutations. Recently, sequencing using targeted panels has seen increased use. Although highly accurate, sequencing is resource intensive and not universally available. METHODS Using 134 bone marrow samples from patients with acute myeloid leukemia evaluated for TP53 mutation by sequencing, we assessed the concordance of p53 IHC with sequencing as well as the interrater-reliability for IHC intensity and percent positivity. RESULTS Consistent with previous studies, we found that p53 IHC was strongly specific and modestly sensitive for missense mutations and that overall performance improved with dedicated hematopathology training. We also found that IHC performed poorly for inactivating mutations and was even variable between cases harboring identical amino acid changes. Low predicted transcriptional activity of p53 missense proteins correlated with a mutant pattern of IHC staining. The status of the second allele and variant allele frequency also affected the accuracy of p53 IHC as a surrogate for TP53 allele status. CONCLUSION Cases of acute myeloid leukemia with TP53 mutations predicted to have low transcriptional activity showed reduced overall survival. Our results demonstrate limited practical utility of p53 IHC for accurate evaluation of TP53 mutation status because of multifactorial confounders.
Collapse
Affiliation(s)
- Lee P Richman
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Brianna F Waller
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Scott B Lovitch
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, United States
| | - Ashwini Jambhekar
- Department of Systems Biology, Harvard Medical School, Boston, MA, United States
- Ludwig Center at Harvard, Boston, MA, United States
| |
Collapse
|
8
|
Reed HD, Do H, Weller E, Harris MH, Reilly CR, Tsai HK, Farrar JE, Lau BW, Pommert L, Gamper C, Atkinson I, Coyne K, Joos M, Loveless S, Steltz S, Cheng L, Malsch M, Myers K, Shimamura A. Lymphoid malignancies in patients with Shwachman-Diamond syndrome. Blood 2025; 145:2528-2532. [PMID: 40090004 DOI: 10.1182/blood.2024026507] [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: 08/19/2024] [Revised: 02/05/2025] [Accepted: 02/18/2025] [Indexed: 03/18/2025] Open
Abstract
ABSTRACT This study identified an increased risk of lymphoid malignancy in Shwachman-Diamond syndrome (SDS) with an observed risk 38-fold higher than expected based on population data. Increased toxicity was observed with standard therapies in patients with SDS.
Collapse
Affiliation(s)
- Helen D Reed
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Hyunwoo Do
- Biostatistics and Research Design Center, Boston Children's Hospital, Boston, MA
| | - Edie Weller
- Biostatistics and Research Design Center, Boston Children's Hospital, Boston, MA
- Division of Hematology and Oncology, Boston Children's Hospital, Boston, MA
| | - Marian H Harris
- Department of Pathology, Boston Children's Hospital, Boston, MA
| | | | - Harrison K Tsai
- Department of Pathology, Boston Children's Hospital, Boston, MA
| | - Jason E Farrar
- Arkansas Children's Research Institute, Little Rock, AR
- Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR
| | - Bonnie W Lau
- Department of Pediatrics, Dartmouth Health, Geisel School of Medicine at Dartmouth College, Lebanon, NH
| | - Lauren Pommert
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Oncology, Cancer and Blood Diseases Institute, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | | | - Ian Atkinson
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Katherine Coyne
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Margret Joos
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| | - Sara Loveless
- Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Sarah Steltz
- Clinical Research Operations Center, Boston Children's Hospital, Boston, MA
| | - Leah Cheng
- Clinical Research Operations Center, Boston Children's Hospital, Boston, MA
| | | | - Kasiani Myers
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH
- Division of Bone Marrow Transplantation and Immune Deficiency, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Akiko Shimamura
- Dana-Farber and Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, Boston, MA
| |
Collapse
|
9
|
Gooptu M, Murdock HM, Soiffer RJ. How I treat AML relapse after allogeneic HSCT. Blood 2025; 145:2128-2137. [PMID: 39719042 DOI: 10.1182/blood.2024025705] [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: 10/10/2024] [Revised: 12/03/2024] [Accepted: 12/16/2024] [Indexed: 12/26/2024] Open
Abstract
ABSTRACT Allogeneic hematopoietic stem cell transplantation (HSCT) is one of the principal curative approaches in the treatment of acute myeloid leukemia (AML); however, relapse after transplantation remains a catastrophic event with poor prognosis. The incidence of relapse has remained unchanged over the last 3 decades despite an evolving understanding of the immunobiology of the graft-versus-leukemia effect and the immune escape mechanisms that lead to post-HSCT relapse. The approach to posttransplant relapse is highly individualized and is dictated both by disease biology and genomics as well as the patient's clinical status at the time of relapse and the interval between relapse and transplantation. With the help of 3 illustrative cases, we discuss our approach to early, late, and incipient relapse. Current therapeutic strategies incorporate immunosuppression taper when feasible, a variety of targeted and nontargeted chemotherapeutic agents, and consolidative cellular therapies including donor lymphocyte infusions or a second allogeneic transplant. We then summarize evolving frontiers in the treatment and prognostication of relapse, including the critical role of measurable residual disease. Finally, we emphasize enrollment on clinical trials and thoughtful discussions regarding goals of care and supporting frail patients as universal principles that should be incorporated in approaches to treatment of AML relapse after transplantation.
Collapse
Affiliation(s)
- Mahasweta Gooptu
- Department of Hematology/Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - H Moses Murdock
- Department of Hematology/Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Robert J Soiffer
- Department of Hematology/Oncology, Dana-Farber Cancer Institute, Boston, MA
| |
Collapse
|
10
|
Shah MV, Hung K, Baranwal A, Wechalekar G, Al-Kali A, Toop CR, Greipp P, Kutyna MM, Matin A, Ladon D, Saliba A, Chen D, Begna K, Brown A, Rud D, Litzow MR, Hogan WJ, Bardy P, Badar T, Kumar S, Yeung DT, Patnaik MM, Foran JM, He R, Gangat N, Hefazi M, Scott HS, Arana Yi CY, Alkhateeb H, Mangaonkar AA, Thomas D, Hahn CN, Orazi A, Arber DA, Kok CH, Tefferi A, Hiwase D. Validation of the 5th edition of the World Health Organization and International Consensus Classification guidelines for TP53-mutated myeloid neoplasm in an independent international cohort. Blood Cancer J 2025; 15:88. [PMID: 40335478 PMCID: PMC12059121 DOI: 10.1038/s41408-025-01290-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 04/09/2025] [Accepted: 04/16/2025] [Indexed: 05/09/2025] Open
Abstract
The World Health Organization (WHO-5) and International Consensus Classification (ICC) acknowledge the poor prognosis of TP53-mutated (TP53mut) myeloid neoplasm (MN). However, there are substantial differences between the two classifications that may lead to under- or overestimation of the prognostic risk. We retrospectively applied WHO-5 and ICC to 603 MN cases harboring TP53mut (variant allele frequency, VAF ≥ 2%). WHO-5 and ICC would not classify 64% and 20% of these cases as TP53mut MN, respectively. Moreover, of those classified, 67.5% would be classified discrepantly. Primary drivers of discrepancies included: (i) prognostic importance of TP53mut acute myeloid leukemia (AML), (ii) interaction of the blast percentage and allelic status, (iii) 17p.13.1 deletion detected by cytogenetics, (iv) complex karyotype (CK) as multi-hit equivalent, and (v) TP53mut VAF threshold, we analyzed survival outcomes of each of these groups with an aim to provide clarity. TP53mut AML was associated with significantly poor survival compared to TP53-wild type TP53wt AML, myelodysplasia-related (AML, MR 4.7 vs. 18.3 months; P < 0.0001), supporting its inclusion within TP53mut MN as a distinct subentity. Secondly, the survival of TP53mut with blast 10-19% was poor regardless of the allelic status. Thirdly, for cases with a single TP53mut with VAF < 50%, 17p13.1 del or CK serve as practical surrogates of biallelic inactivation, obviating the need for an additional copy number analysis. Finally, TP53mut AML, MDS multi-hit/multi-hit equivalent with VAF < 10% had significantly poorer survival compared to TP53mut MDS VAF < 10% without CK and 17p del, and were comparable to those with VAF ≥ 10% (14.1 vs. 48.8 vs.7.8 months, P < 0.0001). Collectively, these findings address key areas of contention and provide valuable insights that will guide future revisions of the WHO and ICC classifications.
Collapse
Affiliation(s)
| | - Kevin Hung
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | | | - Gauri Wechalekar
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Aref Al-Kali
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Carla R Toop
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | - Patricia Greipp
- Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | - Monika M Kutyna
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Aasiya Matin
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Dariusz Ladon
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
| | | | - Dong Chen
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | - Kebede Begna
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Anna Brown
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Danielle Rud
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | - Mark R Litzow
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | - Peter Bardy
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Talha Badar
- Department of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Sharad Kumar
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - David T Yeung
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
| | | | - James M Foran
- Department of Hematology/Oncology, Mayo Clinic, Jacksonville, FL, USA
| | - Rong He
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| | | | | | - Hamish S Scott
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | | | | | | | - Daniel Thomas
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
| | - Christopher N Hahn
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia
| | - Attilio Orazi
- Texas Tech University Health Sciences Center, El Paso, TX, USA
| | | | - Chung Hoow Kok
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA, Australia
- Division of Hematopathology, Mayo Clinic, Rochester, MN, USA
| | | | - Devendra Hiwase
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia.
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA, Australia.
- Adelaide Medical School, University of Adelaide, Adelaide, SA, Australia.
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, Australia.
| |
Collapse
|
11
|
Mina A, Komrokji R. How I treat higher-risk MDS. Blood 2025; 145:2002-2011. [PMID: 39808802 DOI: 10.1182/blood.2024025271] [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: 08/30/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDS) are a widely heterogenous group of myeloid malignancies characterized by morphological dysplasia, a defective hematopoiesis, and recurrent genetic abnormalities. The original International Prognostic Scoring System (IPSS) and revised IPSS have been used to risk-stratify patients with MDS to guide treatment strategies. In higher-risk MDS, the therapeutic approach is geared toward delaying leukemic transformation and prolonging survival. For more than a decade, the hypomethylating agents azacitidine and decitabine have been the standard of care and, when feasible, an allogeneic hematopoietic stem cell transplantation should be considered. However, the IPSS scoring systems solely rely on clinical, morphological, and cytogenetic features and do not account for somatic mutations present in >80% of cases. These genetic abnormalities have been shown to play a crucial role in prognostication, prompting the development of molecular IPSS, and the integration of genomic features into MDS classification systems in recent years. In this review, we delineate our approach to higher-risk MDS in the context of updated classifications and the latest prognostication tools. We use illustrative clinical cases to support our discussion and share insights from recent clinical trials, highlighting lessons learned.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
12
|
Qiu H, Zhang C, Ma X, Li Y. Molecular insights and treatment innovations: Advancing outcomes in acute myeloid leukemia with myelodysplasia‑related changes (Review). Oncol Rep 2025; 53:54. [PMID: 40116086 DOI: 10.3892/or.2025.8887] [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: 11/05/2024] [Accepted: 02/10/2025] [Indexed: 03/23/2025] Open
Abstract
Acute myeloid leukemia, myelodysplasia‑related (AML‑MR), a challenging and aggressive subtype of AML, is characterized by unique genetic abnormalities and molecular features, which contribute to its poor prognosis compared with other AML subtypes. The present review summarizes the current understanding of AML‑MR pathogenesis, highlighting notable advancements in genetic and cytogenetic insights. Critical mutations, such as those in the tumor antigen p53 and additional sex combs like 1 genes, and their role in disease progression and resistance to treatment, are explored. The review further investigates how clonal evolution and cellular microenvironment alterations drive AML‑MR transformation and impact patient outcomes. Despite the poor outlook typically associated with AML‑MR, developments in treatment approaches offer hope. The present review considers the efficacy of novel therapeutic agents, including CPX‑351, hypomethylating agents and targeted molecular therapies. Additionally, innovations in immunotherapy and allogeneic hematopoietic stem cell transplantation are discussed as promising avenues to improve patient survival rates. The challenges of treating AML‑MR, particularly in elderly and pretreated patients, underline the necessity for individualized treatment strategies that consider both the biological complexity of the disease and the overall health profile of the patient. The present review focuses on the mechanisms of AML‑MR transformation, highlighting factors that may offer a crucial theoretical foundation and pave the way for future applications in precision medicine. Future research directions include exploring novel targeted therapies and combination regimens to mitigate the transformation risks and enhance the quality of life of patients with AML‑MR.
Collapse
Affiliation(s)
- Hong Qiu
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Chaowei Zhang
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| | - Xiaochen Ma
- Department of Public Health, Shandong Second Medical University, Weifang, Shandong 261053, P.R. China
| | - Ying Li
- Department of Hematology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
13
|
Gurnari C, Robin M, Adès L, Aljurf M, Almeida A, Duarte FB, Bernard E, Cutler C, Della Porta MG, De Witte T, DeZern A, Drozd-Sokolowska J, Duncavage E, Fenaux P, Gagelmann N, Garcia-Manero G, Haferlach C, Haferlach T, Hasserjian R, Hellström-Lindberg E, Jacoby M, Kulasekararaj A, Lindsley RC, Maciejewski JP, Makishima H, Malcovati L, Mittelman M, Myhre AE, Ogawa S, Onida F, Papaemmanuil E, Passweg J, Platzbecker U, Pleyer L, Raj K, Santini V, Sureda A, Tobiasson M, Voso MT, Yakoub-Agha I, Zeidan A, Walter M, Kröger N, McLornan DP, Cazzola M. Clinical-genomic profiling of MDS to inform allo-HCT: recommendations from an international panel on behalf of the EBMT. Blood 2025; 145:1987-2001. [PMID: 39970324 DOI: 10.1182/blood.2024025131] [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: 10/21/2024] [Revised: 01/09/2025] [Accepted: 01/20/2025] [Indexed: 02/21/2025] Open
Abstract
ABSTRACT For patients with myelodysplastic neoplasm/syndrome (MDS), allogeneic hematopoietic cell transplantation (allo-HCT) represents the only potentially curative treatment, capable of eradicating disease-related mutant hematopoietic cells and establishing normal donor hematopoiesis. Biologic-assignment clinical trials have indicated that in eligible patients, allo-HCT is associated with superior clinical outcomes compared with nontransplant therapy. However, this therapeutic option is only available to a subset of patients, and the outcome is influenced by multiple factors inherent to the patient, the MDS subtype, and the allo-HCT procedure itself. In 2017, the European Society for Blood and Marrow Transplantation (EBMT) published recommendations for allo-HCT in MDS to guide practical decision making. In the contemporary era, genomic profiling has become routine clinical practice in many centers, and the most recent classification systems include MDS entities that are defined by genetic abnormalities. In particular, the molecular International Prognostic Scoring System offers more precise prognostication across all clinical end points and currently represents the standard tool for estimating patient survival in the absence of disease-modifying treatment. Evidence from multiple sources increasingly indicates that allo-HCT should be considered at the time of diagnosis in all eligible patients with MDS. Therefore, genomic profiling for somatic mutations and testing for germ line predisposition variants are integral to determining a patient's eligibility for transplantation. Although all patients with higher-risk MDS are potential candidates for immediate transplantation, a subset of those with lower-risk MDS may also derive benefit from this procedure at an earlier disease stage. Comprehensive recommendations on behalf of an expert international panel for clinical practice and future clinical studies of relevance are presented.
Collapse
Affiliation(s)
- Carmelo Gurnari
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
- Translational Hematology and Oncology Research Department, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Marie Robin
- Department of Hematology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Lionel Adès
- Department of Hematology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Mahmoud Aljurf
- Cancer Center for Excellence, King Faisal Cancer Center, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia
| | - Antonio Almeida
- Department of Hematology, Hospital da Luz Lisboa, Lisbon, Portugal, and Faculdade de Medicina, Universidade Católica Portuguesa, Rio de Mouro, Portugal
| | - Fernando Barroso Duarte
- Department of Hematology, Hospital Universitario Walter Cantídio, Universidade Federal do Ceara, Fortaleza, Brazil
| | - Elsa Bernard
- Department of Computational Oncology, UMR 981, Gustave Roussy, Villejuif, France
| | - Corey Cutler
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Matteo Giovanni Della Porta
- Comprehensive Cancer Center, IRCCS Humanitas Clinical and Research Center and Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Theo De Witte
- Department of Tumor Immunology, Radboud Institute of Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Amy DeZern
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins University School of Medicine, Baltimore, MD
| | - Joanna Drozd-Sokolowska
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Eric Duncavage
- Department of Pathology and Immunology, Washington University School of Medicine, St Louis, MO
| | - Pierre Fenaux
- Department of Hematology, Hôpital Saint Louis, Assistance Publique-Hôpitaux de Paris, Université Paris Cité, Paris, France
| | - Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | | | | | | | | | - Eva Hellström-Lindberg
- Department of Hematology, Karolinska University Hospital, and Centre of Hematology and Regenerative Medicine, Institution of Medicine, Karolinska Institute, Huddinge, Sweden
| | - Meagan Jacoby
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO
| | | | | | - Jaroslaw P Maciejewski
- Translational Hematology and Oncology Research Department, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH
| | - Hideki Makishima
- Department of Hematology and Medical Oncology, Shinshu University School of Medicine, Matsumoto, Japan
| | - Luca Malcovati
- Fondazione IRCCS Policlinico San Matteo and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Moshe Mittelman
- Department of Hematology, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Anders E Myhre
- Department of Haematology, Oslo University Hospital, Oslo, Norway
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Francesco Onida
- Department of Hematology-Bone Marrow Transplant, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, University of Milan, Milan, Italy
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Jakob Passweg
- Divisions of Hematology and Internal Medicine, Department of Medicine, University Hospital Basel, Basel, Switzerland
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
- University Hospital Dresden, Dresden, Germany
| | - Lisa Pleyer
- Third Medical Department with Hematology, Medical Oncology, Hemostaseology, Rheumatology and Infectiology, Oncologic Center, Paracelsus Medical University, Salzburg, Austria
| | - Kavita Raj
- Department of Haematology and Stem Cell Transplantation, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Valeria Santini
- MDS Unit, Hematology, Department of Experimental and Clinical Medicine, Azienda Ospedaliero Universitaria Careggi Medical School, University of Florence, Florence, Italy
| | - Anna Sureda
- Institut Català d'Oncologia, Hospital Duran i Reynals, IDIBELL, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Magnus Tobiasson
- Department of Hematology, Karolinska University Hospital, and Centre of Hematology and Regenerative Medicine, Institution of Medicine, Karolinska Institute, Huddinge, Sweden
| | - Maria Teresa Voso
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Ibrahim Yakoub-Agha
- Centre Hospitalier Universitaire de Lille, University of Lille, INSERM U1286, INFINITE 59000, Lille, France
| | - Amer Zeidan
- Department of Hematology and Oncology, Yale University, New Haven, CT
| | - Matthew Walter
- Division of Oncology, Department of Medicine, Washington University School of Medicine, St Louis, MO
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Donal P McLornan
- Department of Haematology and Stem Cell Transplantation, University College London Hospitals NHS Foundation Trust, London, United Kingdom
| | - Mario Cazzola
- Fondazione IRCCS Policlinico San Matteo and Department of Molecular Medicine, University of Pavia, Pavia, Italy
| |
Collapse
|
14
|
Shimony S, Stahl M, Stone RM. Acute Myeloid Leukemia: 2025 Update on Diagnosis, Risk-Stratification, and Management. Am J Hematol 2025; 100:860-891. [PMID: 39936576 PMCID: PMC11966364 DOI: 10.1002/ajh.27625] [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: 11/22/2024] [Revised: 01/23/2025] [Accepted: 01/25/2025] [Indexed: 02/13/2025]
Abstract
DISEASE OVERVIEW Acute myeloid leukemia (AML) is a bone marrow stem cell cancer that is often fatal despite available treatments. Diagnosis, risk assessment, monitoring, and therapeutic management of AML have changed dramatically in the last decade due to increased pathophysiologic understanding, improved assessment technology, and the addition of at least 12 approved therapies. DIAGNOSIS The diagnosis is based on the presence of immature leukemia cells in the blood, and/or bone marrow or less often in extra-medullary tissues. New biological insights have been integrated into recent classification systems. RISK ASSESSMENT The European Leukemia Network has published risk classification algorithms for both intensively and non-intensively treated patients based on cytogenetic and on molecular findings. Prognostic factors may differ based on the therapeutic approach. MONITORING Our increasing ability to quantify lower levels of measurable residual disease (MRD) potentially allows better response assessment, as well as dynamic monitoring of disease status. The incorporation of MRD findings into therapeutic decision-making is rapidly evolving. RISK ADAPTED THERAPY The availability of 12 newly approved agents has been welcomed; however, optimal strategies incorporating newer agents into therapeutic algorithms are debated. The overarching approach integrates patient and caregiver goals of care, comorbidities, and disease characteristics.
Collapse
Affiliation(s)
- Shai Shimony
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Maximilian Stahl
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| | - Richard M. Stone
- Department of Medical OncologyDana‐Farber Cancer InstituteBostonMassachusettsUSA
| |
Collapse
|
15
|
Oyarbide U, Bezzerri V, Staton M, Boni C, Shah A, Cipolli M, Calo E, Corey SJ. Reduced EIF6 dosage attenuates TP53 activation in models of Shwachman-Diamond syndrome. J Clin Invest 2025; 135:e187778. [PMID: 39964763 PMCID: PMC11996912 DOI: 10.1172/jci187778] [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: 10/17/2024] [Accepted: 02/13/2025] [Indexed: 02/20/2025] Open
Abstract
Shwachman-Diamond syndrome (SDS) is characterized by neutropenia, exocrine pancreatic insufficiency, and bony abnormalities with an increased risk of myeloid neoplasia. Almost all cases of SDS result from biallelic mutations in Shwachman-Bodian-Diamond syndrome (SBDS). SBDS interacts with elongation factor-like 1 (EFL1) to displace eukaryotic initiation factor 6 (EIF6) from the 60S ribosomal subunit. Released EIF6 permits the assembly of ribosomal large and small subunits in the cytoplasm. Decreased EIF6 levels due to haploinsufficiency or missense mutations, which lead to decreased protein expression, may provide a somatic genetic rescue and antileukemic effects. We observed accumulation of EIF6 protein in sbds-KO zebrafish models, confirmed this accumulation in patient-derived tissues, and correlated these with changes in ribosomal proteins and tumor protein p53 (TP53) pathways. The mechanism of action for this adaptive response is unknown. To address this, we generated eif6-KO zebrafish, which do not survive more than 10 days after fertilization. We also created 2 mutants with low Eif6 expression, i.e., 5%-25% of WT levels, that could survive until adulthood. We bred them with sbds-null strains and analyzed their phenotype and biochemical properties. Low Eif6 levels reduced Tp53 pathway activation but did not rescue neutropenia in Sbds-deficient zebrafish. Further studies elucidating the interplay between SBDS, EIF6, and TP53 and cellular stress responses offer promising insights into SDS pathogenesis, somatic genetic rescue, and therapeutic strategies.
Collapse
Affiliation(s)
- Usua Oyarbide
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Valentino Bezzerri
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
- Department of Life Sciences, Health and Health Professions, Link Campus University, Rome, Italy
| | - Morgan Staton
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| | - Christian Boni
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Arish Shah
- Department of Biology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Marco Cipolli
- Cystic Fibrosis Center, Azienda Ospedaliera Universitaria Integrata, Verona, Italy
| | - Eliezer Calo
- Department of Biology and David H. Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Seth J. Corey
- Departments of Pediatrics and Cancer Biology, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Inoue Y, Cioccio J, Mineishi S, Minagawa K. Evolution of Allogeneic Stem Cell Transplantation: Main Focus on AML. Cells 2025; 14:572. [PMID: 40277898 PMCID: PMC12026247 DOI: 10.3390/cells14080572] [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: 03/13/2025] [Revised: 04/04/2025] [Accepted: 04/09/2025] [Indexed: 04/26/2025] Open
Abstract
In recent years, treatments in the field of hematologic malignancies have undergone significant evolution; allogeneic hematopoietic stem cell transplantation (allo-HSCT) has shifted from an "ultimate" therapy to becoming a component of a comprehensive therapeutic strategy for acute myeloid leukemia (AML). Advances in risk stratification (including molecular profiling and measurable residual disease assessment), conditioning regimens, and graft-versus-host disease (GVHD) prophylaxis-such as post-transplant cyclophosphamide-have improved outcomes and expanded donor selection and transplant eligibility. We should not only focus on the transplantation procedure but also consider various therapeutic components, including chemotherapy, targeted therapy (possibly including chimeric antigen receptor T-cell therapy), and post-transplant maintenance therapy, which need to be orchestrated within the broader context of leukemia treatment. In this review, we summarized key developments in allo-HSCT for AML and aim to "decipher" each component of transplantation.
Collapse
Affiliation(s)
- Yoshitaka Inoue
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, PA 17033, USA; (J.C.); (S.M.); (K.M.)
- Cancer Institute, Penn State College of Medicine, Hershey, PA 17033, USA
| | - Joseph Cioccio
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, PA 17033, USA; (J.C.); (S.M.); (K.M.)
| | - Shin Mineishi
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, PA 17033, USA; (J.C.); (S.M.); (K.M.)
| | - Kentaro Minagawa
- Blood and Marrow Transplant Program, Division of Hematology and Oncology, Penn State Cancer Institute, Hershey, PA 17033, USA; (J.C.); (S.M.); (K.M.)
| |
Collapse
|
17
|
Zhou C, Xu L, Zhang X, Chang Y, Mo X, Sun Y, Huang X, Wang Y. Impact of TP53 alteration on allogeneic hematopoietic cell transplantation outcomes for myelodysplastic syndromes. Bone Marrow Transplant 2025; 60:467-473. [PMID: 39838079 DOI: 10.1038/s41409-025-02511-7] [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: 08/09/2024] [Revised: 12/24/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025]
Abstract
The poor outcome of TP53 alteration has been reported in myelodysplastic syndrome (MDS) patients. However, the role of allogeneic hematopoietic stem cell transplantation (allo-HSCT) in TP53 alteration patients remains debated. Previous studies showed that TP53 mutations had no effect on the prognosis of patients with acute leukemia after haploidentical HSCT (haplo-HSCT). The effect of haplo-HSCT on MDS patients with TP53 alterations remains to be further elucidated. We aimed to reveal the role of TP53 alterations in the prognosis of MDS patients undergoing allo-HSCT, especially haplo-HSCT. 261 MDS patients with known TP53 status were enrolled, including thirty-seven patients with TP53 mutation/deletion (TP53mut/del). TP53mut/del patients showed a worse rate of 2-year cumulative incidence of relapse (CIR) and 2-year disease-free survival (DFS) than TP53 wild type (TP53wt) patients (46.2% vs 17.0%, P < 0.001; 41.8% vs 68.9, P < 0.001) after allo-HSCT, even for those with haplo-HSCT (CIR: P < 0.001; DFS: P = 0.002). However, the prognostic effect of TP53 alteration on overall survival (OS) was not observed in patients with haplo-HSCT (66.7% vs 75.2%, P = 0.108). Positivity of post-transplantation measurable residual disease (post-MRD) and time from diagnosis to transplantation were independent risk factors for MDS patients. TP53 alterations do not affect OS in patients undergoing haplo-HSCT requires further validation.
Collapse
Affiliation(s)
- Cuiyan Zhou
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Lanping Xu
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiaohui Zhang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yingjun Chang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiaodong Mo
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Yuqian Sun
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
| | - Xiaojun Huang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China
- Peking-Tsinghua Center for Life Sciences, 100871, Beijing, China
| | - Yu Wang
- Peking University People's Hospital, Peking University Institute of Hematology, National Clinical Research Center for Hematologic Disease, Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Beijing, China; Collaborative Innovation Center of Hematology, Peking University, Beijing, China.
| |
Collapse
|
18
|
Ma C, Huang X, Chen Y, Shi F, Li X, Tian S, Ma W. Outcomes of allogeneic hematopoietic stem cell transplantation in Shwachman-Diamond syndrome: a systematic review and meta-analysis. Cytotherapy 2025; 27:465-474. [PMID: 39745406 DOI: 10.1016/j.jcyt.2024.12.004] [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: 10/04/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 03/26/2025]
Abstract
We conducted a systematic review and meta-analysis to evaluate the outcomes of Allogeneic hematopoietic stem cell transplantation (Allo-HSCT) in the treatment of Shwachman-Diamond syndrome (SDS). A literature search was performed on PubMed, Embase, and Web of Science. After screening 397 articles, 10 studies were included. Data was extracted in accordance with the PRISMA guidelines and analyzed using the R 'meta' package. The pooled median 3 (1-5)-year overall survival (OS) after Allo-HSCT were 63.7% (95% CI 56.9-70.2%), 80.3% (95% CI 68.% 5-92.1%), 41.1% (95% CI 21.7-60.4%), 48.9% (95% CI 29.0-68.9%), and 8.7% (95% CI 0.0-60.8%) in SDS patients, SDS patients with bone marrow failure (BMF), SDS patients with myeloid neoplasms (MN), SDS patients with myelodysplastic syndrome (MDS), and SDS patients with acute myeloid leukemia (AML), respectively. Allo-HSCT is an efficacious approach for treating SDS patients with severe hematologic complications. However, poor outcomes were revealed in SDS patients with MN with a pooled 3 (1-5)-year relapse rate (RR) after Allo-HSCT of 25.8% (95% CI 12.5-39.0%), and a pooled 3-year non-relapse mortality (NRM) was 52.6% (95% CI 34.2-70.9%). These findings were consistent with the clinical findings that transplant-related complications are the main cause of the poor transplantation prognosis of SDS patients with MN. Efficacious bone marrow conditioning regimens, graft-versus-host disease (GVHD) prevention, and bridging treatment regimens are potential means to improve the transplantation prognosis of SDS patients.
Collapse
Affiliation(s)
- Cuiping Ma
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaohua Huang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Yuhan Chen
- Shenzhen Luohu District Hospital of Traditional Chinese Medicine, Guangdong, China
| | - Fengqin Shi
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Li
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Shaodan Tian
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| | - Wei Ma
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
19
|
Kugler E, Dasdemir E, Bataller A, Wang B, DiNardo CD, Daver N, Yilmaz M, Short NJ, Borthakur G, Kadia TM, Sasaki K, Hammond D, Bazinet A, Irajizad E, Thakral B, Pierce S, Reville P, Ravandi F, Abbas HA. Mutation dynamics from diagnosis to relapse in acute myeloid leukemia with chromosomal 7 deletions. Leuk Lymphoma 2025:1-13. [PMID: 40164144 DOI: 10.1080/10428194.2025.2477723] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 02/20/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025]
Abstract
Monosomy 7 and 7q deletions (-7/del(7q)) are the most common adverse cytogenetic event in acute myeloid leukemia (AML), linked to high relapse rates. We analyzed 115 AML patients with -7/del(7q) who achieved remission after induction therapy to characterize the mutational landscape from diagnosis to relapse. Median overall survival (OS) was 10.4 months, with improved survival in patients without TP53 mutation (13.04 vs. 8.6 months) or complex karyotype (12.4 vs. 8.6 months). TP53 mutations were most frequent (67% of cases at diagnosis) and persisted in 97% of patients at relapse. At time of relapse, patients with TP53 mutations had fewer co-occurring mutations in ASXL1, RUNX1, NRAS, PTPN11 and SRSF2 compared to TP53 wild-type patients. Patients with mutated TP53 and co-mutation in NF1, BCORL1, GATA2, or RUNX1 had shorter relapse-free survival (2 vs. 5 months) and OS (7.2 vs. 10.4 months) than those with TP53 mutation alone. Allogeneic transplant improved OS significantly, regardless of TP53 status.
Collapse
Affiliation(s)
- Eitan Kugler
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enes Dasdemir
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Biology and Biochemistry, University of Houston, Houston, TX, USA
| | - Alex Bataller
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bofei Wang
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney D DiNardo
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Musa Yilmaz
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gautam Borthakur
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan M Kadia
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danielle Hammond
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Bazinet
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ehsan Irajizad
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Beenu Thakral
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sherry Pierce
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Patrick Reville
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hussein A Abbas
- Department of Leukemia, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Genomic Medicine, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
20
|
Libby DM, Libby LJ, Ma X, Chua J, Blow T, Razavi P, Saxena A. Association of Oncogene Driver Mutations with Recurrence and Survival in Stage I Nonsmall Cell Lung Cancer. Clin Lung Cancer 2025; 26:116-123. [PMID: 39674767 DOI: 10.1016/j.cllc.2024.10.016] [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: 07/11/2024] [Revised: 10/30/2024] [Accepted: 10/31/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Stage I nonsmall cell lung cancer (NSCLC) is primarily treated with surgical resection and has a favorable prognosis with an expected recurrence rate of 30%. New methods to risk stratify patients with stage I NSCLC are needed to help select those that might benefit from more active surveillance or adjuvant therapy. METHODS We analyzed clinical data from 1330 patients (1469 tumors) with NSCLC and correlated it with next-generation sequencing (NGS). To reduce the potential confounding variables of stage and treatment, this analysis only included patients with stage I NSCLC in whom surgical resection was the primary treatment. RESULTS In 570 patients (600 tumors), 75 (12.5%) developed recurrence. Recurrence occurred in 37.5% of patients with KRAS G12V mutation versus 11.1% of patients without this mutation (P < .001). A lower chance of recurrence was associated with "any EGFR" mutation (6.74% vs. 14.9%, P = .006). A history of coronary artery disease (CAD) increased the chance of recurrence: OR 2.7 (1.57-4.89, P < .001). Shorter survival was predicted by KRAS G12V (P = .009) and "other TP53" mutation (P = .025). KRAS G12V, KRAS G13D, MET E168D, PTEN, and "other TP53" were oncogene mutations associated with reduced survival in stage I NSCLC. CAD, type 2 diabetes (DM2), and "other cancer" were medical comorbidities associated with reduced survival in stage I NSCLC. CONCLUSIONS Oncogene mutations such as KRAS G12V and EGFR may have implications for cancer surveillance strategies and inform future treatment trials of stage I NSCLC.
Collapse
Affiliation(s)
- Daniel M Libby
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Medical College of Cornell University, New York, NY.
| | - Laura J Libby
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Weill Medical College of Cornell University, New York, NY
| | - Xiaoyue Ma
- Division of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, NY
| | - Jason Chua
- Division of Biostatistics and Epidemiology, Weill Medical College of Cornell University, New York, NY
| | - Tahj Blow
- Weill Medical College of Cornell University, New York, NY
| | - Peyman Razavi
- Weill Medical College of Cornell University, New York, NY
| | - Ashish Saxena
- Division of Hematology and Medical Oncology, Department of Medicine, Weill Medical College of Cornell University, New York, NY
| |
Collapse
|
21
|
Jiang B, Yang T, Zhao Y, Luo Y, Ouyang G, Yu J, Ye Y, Lan J, Lu Y, Lai X, Ye B, Chen Y, Liu L, Xu Y, Shi P, Xiao H, Hu H, Guo Q, Fu H, Wang X, Sun J, Zheng W, He J, Zhao Y, Wu W, Cai Z, Wei G, Huang H, Shi J. Better pre-transplant treatment options for TP53-mutated MDS: cytoreductive or non-cytoreductive therapy? Bone Marrow Transplant 2025; 60:326-334. [PMID: 39627346 DOI: 10.1038/s41409-024-02486-x] [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: 08/07/2024] [Revised: 10/12/2024] [Accepted: 11/25/2024] [Indexed: 03/12/2025]
Abstract
Patients with TP53-mutated myelodysplastic neoplasms (MDS) have unfavorable prognoses; the benefit of cytoreductive treatment before hematopoietic stem cell transplantation (HSCT) is debated. We retrospectively analyzed 284 MDS patients undergoing allogeneic HSCT; among which 49 had TP53 mutation, with 38 receiving cytoreduction and 11 treated exclusively with best supportive care (BSC) before transplantation. Regardless of TP53 allelic state, patients with mutated-TP53 had a lower overall survival rate and higher relapse rate than those with wild-type TP53 (P < 0.001, P = 0.002, respectively). Among the TP53-mutated cohort, the 2-year overall survival rate in the cytoreduction group was comparable to that in the BSC group (34.6% vs. 45.5%, P = 0.53), and no other prognostic benefit was observed as well (all P < 0.05). Moreover, no prognostic difference was found among the chemotherapy subgroup, hypomethylating agent subgroup, and BSC subgroup (all P > 0.05). Patients in the pre-HSCT measurable residual disease (MRD) negative subgroup, pre-HSCT MRD-positive subgroup, and BSC subgroup exhibited similar prognoses (all P > 0.05). Multivariate analyses showed that pre-HSCT cytoreduction was not associated with post-transplant survival (all P > 0.05). In conclusion, TP53-mutated MDS patients have poor post-HSCT outcomes; compared to BSC, pre-HSCT cytoreduction doesn't improve prognosis, even in those with MRD negative before transplantation.
Collapse
Affiliation(s)
- Bingqian Jiang
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Tingting Yang
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yanmin Zhao
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yi Luo
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Guifang Ouyang
- Department of Hematology, Ningbo First Hospital, Ningbo, China
| | - Jian Yu
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yishan Ye
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Jianping Lan
- Department of Hematology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Ying Lu
- Department of Hematology, Yinzhou People's Hospital, Ningbo, China
| | - Xiaoyu Lai
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Baodong Ye
- Department of Hematology, The First Hospital Affiliated to Zhejiang Chinese Medical University, Hangzhou, China
| | - Yi Chen
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lizhen Liu
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yang Xu
- Department of Hematology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Pengfei Shi
- Department of Hematology, The Affiliated Hangzhou First People's Hospital, Westlake University School of Medicine, Hangzhou, China
| | - Haowen Xiao
- Department of Hematology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Huixian Hu
- Department of Hematology, Jinhua Central Hospital, Jinhua, China
| | - Qunyi Guo
- Department of Hematology, Taizhou Hospital of Zhejiang, Wenzhou Medical University, Taizhou, China
| | - Huarui Fu
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Xinyu Wang
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Jie Sun
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Weiyan Zheng
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Jingsong He
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Yi Zhao
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Wenjun Wu
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Zhen Cai
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - Guoqing Wei
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China
- Institute of Hematology, Zhejiang University, Hangzhou, China
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China
| | - He Huang
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University, Hangzhou, China.
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China.
| | - Jimin Shi
- Bone Marrow Transplantation Center of The First Affiliated Hospital & Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, China.
- Institute of Hematology, Zhejiang University, Hangzhou, China.
- Zhejiang Province Engineering Research Center for Stem Cell and Immunity Therapy, Hangzhou, China.
| |
Collapse
|
22
|
Hart SA, Lee LA, Seegmiller AC, Mason EF. Diagnosis of TP53-mutated myeloid disease by the ICC and WHO fifth edition classifications. Blood Adv 2025; 9:445-454. [PMID: 39536285 PMCID: PMC11808619 DOI: 10.1182/bloodadvances.2024014140] [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: 07/05/2024] [Revised: 10/17/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
ABSTRACT The International Consensus (ICC) and World Health Organization fifth edition (WHO5) classifications introduced new, but differing, categories of myeloid disease defined by TP53 mutations. We reviewed a cohort of 188 cases of TP53-mutated myeloid disease to determine how diagnoses and outcomes differ between the 2 classifications. Overall, 120 (64%) cases were classified differently by the ICC and WHO5, including 24 of 80 (30%) cases with <20% blasts. These cases were discrepantly categorized primarily because of inclusion of complex karyotype (CK) as a surrogate for biallelic TP53 inactivation only in the ICC. However, there were no significant differences in clinicopathologic characteristics or overall survival between cases categorized as TP53-mutated disease by both classifications and those with a single TP53 mutation and CK, suggesting that CK reliably identifies TP53-mutated cases with biallelic TP53 inactivation. Most cases of acute myeloid leukemia (AML; 96/102 [94%]) were discrepantly diagnosed between the ICC and WHO5 because of the introduction of AML with mutated TP53 as a distinct category only in the ICC. Nearly all of these were instead diagnosed as AML, myelodysplasia related (AML-MR) by WHO5. However, when compared with a separate cohort of patients with AML-MR without TP53 mutations, patients with TP53-mutated AML showed a distinct genetic profile and significantly worse overall survival, supporting the inclusion of AML with mutated TP53 as a distinct disease category. Overall, our results show that a significant percentage of TP53-mutated myeloid disease is classified differently by the ICC and WHO5 and highlight areas to address in future classification systems.
Collapse
Affiliation(s)
- Stephanie A. Hart
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Laura A. Lee
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Adam C. Seegmiller
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| | - Emily F. Mason
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
23
|
Thalambedu N, Mohan Lal B, Harbaugh B, Alapat DV, Gaddam M, Gentille Sanchez CG, Kumaran M, Varma A. Unveiling Myelodysplastic Syndromes: Exploring Pathogenic Mechanisms and Therapeutic Advances. Cancers (Basel) 2025; 17:508. [PMID: 39941875 PMCID: PMC11816122 DOI: 10.3390/cancers17030508] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/14/2025] [Accepted: 01/26/2025] [Indexed: 02/16/2025] Open
Abstract
Myelodysplastic syndromes (MDSs), either primary or secondary, are a heterogeneous group of clonal hematological neoplasms characterized by bone marrow dyshematopoiesis, peripheral blood cytopenia, and the potential risk of acute myeloid leukemia (AML) transformation. The clinical heterogeneity in MDS is a reflection of the underlying multitude of genetic defects playing a role in the pathogenesis. Recent advances in the clinicopathological, immunophenotypic, and molecular landscape in understanding the pathophysiology of MDS lead to evolving and refined classification systems with newer entities. Evolving MDS therapies will target the disease's core mechanisms, allowing for personalized treatment based on individual patient's genes and leading to better outcomes. This review provides an overview of MDS pathogenesis to enhance comprehension of its various subgroups. Additionally, we examine the updated classification systems of the World Health Organization (WHO) and the International Consensus Classification (ICC) pertaining to MDS, along with relevant therapeutic approaches.
Collapse
Affiliation(s)
- Nishanth Thalambedu
- Department of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.T.); (B.M.L.); (M.K.)
| | - Bhavesh Mohan Lal
- Department of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.T.); (B.M.L.); (M.K.)
| | - Brent Harbaugh
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (B.H.); (D.V.A.)
| | - Daisy V. Alapat
- Department of Pathology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (B.H.); (D.V.A.)
| | - Mamatha Gaddam
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Slot # 508, Little Rock, AR 72205, USA; (M.G.); (C.G.G.S.)
| | - Cesar Giancarlo Gentille Sanchez
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Slot # 508, Little Rock, AR 72205, USA; (M.G.); (C.G.G.S.)
| | - Muthu Kumaran
- Department of Hematology and Oncology, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA; (N.T.); (B.M.L.); (M.K.)
| | - Ankur Varma
- Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, 4301 W. Markham Street, Slot # 508, Little Rock, AR 72205, USA; (M.G.); (C.G.G.S.)
| |
Collapse
|
24
|
Kröger N. Treatment of high-risk myelodysplastic syndromes. Haematologica 2025; 110:339-349. [PMID: 39633555 PMCID: PMC11788630 DOI: 10.3324/haematol.2023.284946] [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: 10/11/2024] [Accepted: 11/19/2024] [Indexed: 12/07/2024] Open
Abstract
Myelodysplastic syndrome (MDS) is considered to be a heterogeneous myeloid malignancy with a common origin in the hematopoietic stem cell compartment and is generally divided into lower- and higher-risk forms. While the treatment goals for lower-risk MDS are to decrease transfusion requirements and transformation into acute leukemia, the major aims for higher-risk MDS are to prolong survival and ultimately cure the patient. Although novel agents such as luspatercept and imetelstat have recently been approved as new treatment options for lower-risk MDS, hypomethylating agents currently remain the only approved non-transplant option for higher-risk MDS and are the standard of care for patients not eligible for allogeneic hematopoietic stem cell transplantation (HSCT). Combinations with other drugs as first-line treatment have to date not proven more efficacious than monotherapy in higher-risk MDS, and outcome after the failure of treatment with hypomethylating agents is poor. The only potential cure and standard of care for eligible patients is HSCT and even though the number of transplanted - especially older - MDS patients has increased over time due to better management and greater donor availability, the majority of MDS patients will not be eligible for this curative approach. Current challenges include decreasing the relapse risk, the main cause of HSCT failure. This review summarizes current knowledge on the options of transplant and non-transplant treatment approaches for these patients and demonstrate the unmet clinical need for more effective therapies.
Collapse
Affiliation(s)
- Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf.
| |
Collapse
|
25
|
Guerrettaz RD, Spellman SR, Page KM. Role of the CIBMTR biorepository and registry in precision transplantation research. Bone Marrow Transplant 2025; 60:161-164. [PMID: 39516706 PMCID: PMC11810599 DOI: 10.1038/s41409-024-02446-5] [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: 07/10/2024] [Revised: 10/06/2024] [Accepted: 10/11/2024] [Indexed: 11/16/2024]
Abstract
Observational studies using registry-based data have become indispensable in our management of patients receiving hematopoietic cell transplantation (HCT) or cellular therapies (CT) for hematologic malignancies. These registries support studies that describe the natural history of disease, evaluate clinical effectiveness, or measure safety and quality of patient care. To complement registries, biobanks exist as an essential partner to provide insight on genetic, molecular, and cellular processes. In this review, we discuss and provide examples of several biobank and registry collaborations that have advanced our knowledge in the field of HCT and CT, using the Center for International Blood and Marrow Transplantation Research (CIBMTR) as a highlighted example. The CIBMTR has collected data on over 650,000 patients and has over 60,000 paired donor recipient banked samples available for research. Biospecimens and clinical data from the CIBMTR have led to advancements in our knowledge and care of conditions such as acute myeloid leukemia and myelodysplastic syndrome (MDS), as well as our understanding of the roles of HLA typing and social determinants of health. Despite the utility in advancing research, registries and biobanks do not exist without challenges which are briefly reviewed. Nevertheless, the contributions of these resources in supporting HCT/CT research are noteworthy.
Collapse
Affiliation(s)
- Ryan D Guerrettaz
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, USA.
| | - Stephen R Spellman
- Center for International Blood and Marrow Transplantation Research (CIBMTR), NMDP, Minneapolis, MN, USA
| | - Kristin M Page
- Department of Pediatrics, Medical College of Wisconsin (MCW), Milwaukee, WI, USA
- Center for International Blood and Marrow Transplantation Research (CIBMTR), Medical College of Wisconsin (MCW), Milwaukee, WI, USA
| |
Collapse
|
26
|
Mediavilla C, D'Aveni M, Huynh A, Joris M, Masouridi-Levrat S, Nguyen S, Turlure P, Magro L, Guillaume T. [Place of hematopoietic stem cell transplantation for very high risk acute myeloblastic leukemia and myelodysplastic syndromes (SFGM-TC)]. Bull Cancer 2025; 112:S54-S67. [PMID: 39242254 DOI: 10.1016/j.bulcan.2024.06.016] [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: 04/18/2024] [Revised: 06/14/2024] [Accepted: 06/20/2024] [Indexed: 09/09/2024]
Abstract
Allogeneic hematopoietic stem cell transplantation (alloHSCT) is a potentially curative treatment for acute myeloid leukemia (AML) and myelodysplastic syndromes (MDS). However, these transplants are complicated by a high rate of relapse in very high cytogenetic risk or refractory diseases. The benefit of this therapeutic strategy for these serious malignant hemopathies could therefore be reassessed. As part of the 14th workshop for the harmonization of allograft practices organized by the francophone society of bone marrow transplantation and cellular therapy (SFGM-TC) (SFGM-TC) in Lille in September 2023, the role of allograft for very high risk or refractory AML and MDS was challenged after analysis of published studies.
Collapse
Affiliation(s)
| | - Maud D'Aveni
- Service d'hématologie, CHU de Nancy, Nancy, France
| | - Anne Huynh
- Service d'hématologie, IUCT, CHU de Toulouse, Toulouse, France
| | | | | | - Stéphanie Nguyen
- Service d'hématologie, CHU La Pitié-Salpétrière, Sorbonne Université, Paris, France
| | - Pascal Turlure
- Service d'hématologie, CHU Dupuytren Limoges, 2, avenue Martin Luther King, 87000 Limoges, France
| | - Leonardo Magro
- Service des maladies du sang, CHU de Lille, 2, rue Michel Polonowski, 59000 Lille, France
| | - Thierry Guillaume
- Service d'hématologie clinique, CHU de Nantes, Hôtel Dieu, 1, place Ricordeau, 44000 Nantes, France.
| |
Collapse
|
27
|
Della Porta MG, Bewersdorf JP, Wang YH, Hasserjian RP. Future directions in myelodysplastic syndromes/neoplasms and acute myeloid leukaemia classification: from blast counts to biology. Histopathology 2025; 86:158-170. [PMID: 39450427 DOI: 10.1111/his.15353] [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: 08/14/2024] [Revised: 10/01/2024] [Accepted: 10/03/2024] [Indexed: 10/26/2024]
Abstract
Myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukaemia (AML) are neoplastic haematopoietic cell proliferations that are diagnosed and classified based on a combination of morphological, clinical and genetic features. Specifically, the percentage of myeloblasts in the blood and bone marrow is a key feature that has historically separated MDS from AML and, together with several other morphological parameters, defines distinct disease entities within MDS. Both MDS and AML have recurrent genetic abnormalities that are increasingly influencing their definitions and subclassification. For example, in 2022, two new MDS entities were recognised based on the presence of SF3B1 mutation or bi-allelic TP53 abnormalities. Genomic information is more objective and reproducible than morphological analyses, which are subject to interobserver variability and arbitrary numeric cut-offs. Nevertheless, the integration of genomic data with traditional morphological features in myeloid neoplasm classification has proved challenging by virtue of its sheer complexity; gene expression and methylation profiling also can provide information regarding disease pathogenesis, adding to the complexity. New machine-learning technologies have the potential to effectively integrate multiple diagnostic modalities and improve on historical classification systems. Going forward, the application of machine learning and advanced statistical methods to large patient cohorts can refine future classifications by advancing unbiased and robust previously unrecognised disease subgroups. Future classifications will probably incorporate these newer technologies and higher-level analyses that emphasise genomic disease entities over traditional morphologically defined entities, thus promoting more accurate diagnosis and patient risk stratification.
Collapse
Affiliation(s)
- Matteo G Della Porta
- Comprehensive Cancer Center, IRCCS Humanitas Clinical and Research Center and Humanitas University, Milan, Italy
| | - Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Cancer Center, New Haven, CT, USA
| | - Yu-Hung Wang
- Epigenetics of Haematopoiesis Laboratory, Division of Cancer Sciences, The University of Manchester, Manchester, UK
- Division of Hematology, National Taiwan University Hospital, Taipei, Taiwan
| | - Robert P Hasserjian
- Department of Pathology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
28
|
Theron A, Alonso-Saladrigues A, Dapena JL, López-Duarte M, Diaz de Heredia C, Verdú-Amorós J, Sarrate E, Esperanza-Cebollada E, Cuatrecasas E, Andreu S, Conde N, Sanchez-Sierra N, Isola I, Camós M, Torrebadell M, Rives S, Català A. Secondary haematological dysplasia after CAR-T-cell therapy for acute lymphoblastic leukaemia in children. Br J Haematol 2025; 206:186-194. [PMID: 39463072 DOI: 10.1111/bjh.19862] [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: 07/04/2024] [Accepted: 10/15/2024] [Indexed: 10/29/2024]
Abstract
The use of CAR-T is becoming more widespread in the treatment of haematological malignancies. In adults, secondary myelodysplastic syndromes (MDS) after CAR-T have been described. However, there are currently no data on the risk of MDS following CAR-T in children treated for acute lymphoblastic leukaemia (ALL). We studied all children treated with CAR-T cells at Hospital Sant Joan de Déu in Barcelona and those with persistent cytopenias were evaluated at the cytological, cytogenetic, and molecular levels to look for MDS. A total of 106 patients received CAR-T for ALL. Among 40 patients without early relapse or subsequent therapy after CAR-T, four fulfilled the WHO criteria for myelodysplasia. These four patients had received a haematopoietic stem cell transplantation (HSCT) prior to CAR-T and presented cytopenias with severe dysplastic changes in bone marrow after CAR-T. One patient had clonal MDS with high-risk cytogenetics arising from the host cells requiring a HSCT. Three patients had non-progressive dysplasia arising from the donor cells. Two are alive in complete remission with stable cytopenias and one succumbed to ALL relapse. This is the first description of post-CAR-T MDS and haematological dysplasia in children and highlights the need to monitor children with persistent post-CAR-T cytopenias.
Collapse
Affiliation(s)
- Alexandre Theron
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Department of Pediatric Oncology and Hematology, Hôpital Arnaud de Villeneuve, Univ Montpellier, CHU Montpellier, Montpellier, France
| | - Anna Alonso-Saladrigues
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Faculty of Medicine PhD Student, University of Barcelona, Barcelona, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Jose-Luis Dapena
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Mónica López-Duarte
- Pediatric Hematology Unit, Hematology Department, Hospital de Valdecilla, Santander, Spain
| | - Cristina Diaz de Heredia
- Pediatric Hematology and Oncology Department, Hospital Universitari Vall d'Hebron, and Vall d'Hebron Institut of Research (VHIR), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Jaime Verdú-Amorós
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Department of Pediatric Hematology and Oncology, Hospital Clínico Universitario, Biomedical Research Institute (INCLIVA), Valencia, Spain
| | - Edurne Sarrate
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Elena Esperanza-Cebollada
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
| | - Esther Cuatrecasas
- Cytogenetics Laboratory, Genetics Medicine Section, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Sandra Andreu
- Cytogenetics Laboratory, Genetics Medicine Section, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Nuria Conde
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
| | - Nazaret Sanchez-Sierra
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
| | - Ignacio Isola
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Mireia Camós
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Montse Torrebadell
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
- Hematology Laboratory, Hospital Sant Joan de Déu de Barcelona, Esplugues de Llobregat, Spain
| | - Susana Rives
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Albert Català
- Leukemia and Lymphoma Department, CAR-T-Cell Unit, Pediatric Cancer Center Barcelona (PCCB), Hospital Sant Joan de Déu de Barcelona, University of Barcelona, Esplugues de Llobregat, Spain
- Developmental Tumors Biology Group, Leukemia, and Other Pediatric Hemopathies, Pediatric Cancer Center Barcelona (PCCB), Institut de Recerca Hospital Sant Joan de Déu, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
29
|
Xiao Y, Yang K, Huang Q, Wei C, Wei M, Geng Z, Wu H, Zhou T, Yin X, Zhou Y. Selinexor in combination with venetoclax and decitabine in patients with refractory myelodysplastic syndrome previously exposed to hypomethylating agents: three case reports. Front Oncol 2024; 14:1477697. [PMID: 39749030 PMCID: PMC11693730 DOI: 10.3389/fonc.2024.1477697] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 12/02/2024] [Indexed: 01/04/2025] Open
Abstract
The management of patients with myelodysplastic syndrome (MDS) refractory to hypomethylating agents (HMAs) remains a challenge with few reliably effective treatments. Preclinical studies have shown that the inhibition of the nuclear export protein XPO1 causes nuclear accumulation of p53 and disruption of NF-κB signaling; both of which are relevant targets for MDS. Selinexor is an XPO1 inhibitor with demonstrated efficacy in MDS patients. Herein, we report three patients with MDS refractory to HMAs, however, when selinexor and venetoclax were added to the treatment regimen, the patients achieved a complete response and a significant reduction in spleen size. All patients successfully underwent hematopoietic stem cell transplantation. These cases demonstrate that the combination therapy can achieve CR and significant reductions in spleen size, offering a promising therapeutic option for patients with limited treatment choices. Combination therapy would also offer a potential way for patients to bridge to transplantation. Formal evaluations of this regimen in patients with MDS refractory to HMAs may be meaningful.
Collapse
Affiliation(s)
- Yunshuo Xiao
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Kun Yang
- Department of Hematology, Zigong First People’s Hospital, Zigong, China
- Department of Hematology, West China Hospital, Chengdu, China
| | - Qiuying Huang
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Changqing Wei
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Manlv Wei
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Zhili Geng
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Hui Wu
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Tianhong Zhou
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Xialoin Yin
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| | - Yali Zhou
- Department of Hematology, The 923rd Hospital of the Joint Logistics Support Force of the People’s Liberation Army, Nanning, China
| |
Collapse
|
30
|
Han WM, Sazzad HMS, Bloch M, Baker DA, Roth N, Bowden-Reid E, Smith DE, Hoy JF, Woolley I, Finlayson R, Templeton DJ, Matthews GV, Costello J, Dawson MA, Dawson SJ, Polizzotto MN, Petoumenos K, Yeh P, Dharan NJ. Age-related clonal hematopoiesis and HIV infection are associated with geriatric outcomes: The ARCHIVE study. Cell Rep Med 2024; 5:101835. [PMID: 39626674 PMCID: PMC11722090 DOI: 10.1016/j.xcrm.2024.101835] [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: 06/30/2024] [Revised: 09/12/2024] [Accepted: 11/04/2024] [Indexed: 12/20/2024]
Abstract
While HIV infection and clonal hematopoiesis (CH) have been linked with inflammatory dysregulation and an increased risk of aging-related comorbidities, their relationship with clinical geriatric syndromes has not been well defined. In the Age-related Clonal Haematopoiesis in an HIV Evaluation Cohort (ARCHIVE) study (NCT04641013), we measure associations between HIV and CH and geriatric syndromes. Of 345 participants (176 with HIV and 169 without HIV), 23% had at least one mutation associated with CH: 27% with HIV and 18% without HIV (p = 0.048). In adjusted analyses, HIV infection is independently associated with increased phenotypic age acceleration (coefficient 1.73, 95% confidence interval [CI] 0.3, 3.16) and CH is independently associated with being frail (vs. pre-frail/robust; odds ratio 2.38, 95% CI 1.01, 5.67) and with having reduced quality of life (coefficient -2.18, 95% CI -3.92, -0.44). Our findings suggest that HIV is associated with increased biological age and that CH may be used as a biomarker for adverse geriatric outcomes.
Collapse
Affiliation(s)
- Win Min Han
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia.
| | - Hossain M S Sazzad
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia
| | - Mark Bloch
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia; Holdsworth House Medical Practice, Sydney, NSW 2010, Australia
| | | | - Norman Roth
- Prahran Market Clinic, Melbourne, VIC 3181, Australia
| | - Ellen Bowden-Reid
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia
| | - Don E Smith
- Albion Centre, South Eastern Sydney Local Health District, Sydney, NSW 2010, Australia; School of Population Health, University of New South Wales Sydney, Sydney, NSW 2033, Australia
| | - Jennifer F Hoy
- Department of Infectious Diseases, Alfred Hospital and Monash University, Melbourne, VIC 3004, Australia
| | - Ian Woolley
- Monash Infectious Diseases, Monash Health, Clayton, VIC 3168, Australia; Centre for Inflammatory Diseases, Monash University, Clayton, VIC 3168, Australia
| | | | - David J Templeton
- Department of Sexual Health Medicine and Sexual Assault Medical Service, Sydney Local Health District, Sydney, NSW 2050, Australia; Discipline of Medicine, Central Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2050, Australia
| | - Gail V Matthews
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia; St Vincent's Hospital, Darlinghurst, NSW 2010, Australia
| | | | - Mark A Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia; Centre for Cancer Research, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Sarah-Jane Dawson
- Peter MacCallum Cancer Centre, Melbourne, VIC 3052, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, VIC 3052, Australia; Centre for Cancer Research, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Mark N Polizzotto
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia; Australian National University, Canberra, ACT 2601, Australia
| | - Kathy Petoumenos
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia
| | - Paul Yeh
- Department of Medicine, School of Clinical Sciences at Monash Health, Monash University, Clayton, VIC 3168, Australia; Monash Haematology, Monash Health, Clayton, VIC 3168, Australia
| | - Nila J Dharan
- Kirby Institute, University of New South Wales Sydney, Sydney, NSW 2033, Australia.
| |
Collapse
|
31
|
Fernández HF, Mishra A. Transplant options and outcomes for TP53 myeloid disease. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2024; 2024:335-339. [PMID: 39644031 DOI: 10.1182/hematology.2024000558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/09/2024]
Abstract
TP53-mutated myeloid disease is a constellation of abnormalities seen in both de novo and therapy-related acute myeloid leukemia and myelodysplastic syndrome. Historically, this group of disorders has had a poor prognosis. Newer treatment combinations allow patients to be treated with less toxicity. If response to induction therapy is achieved, fit and willing patients should be considered for allogeneic hematopoietic cell transplantation (HCT). The addition of allogeneic HCT to the treatment approach has modestly improved outcomes compared to chemotherapy alone, more so for those patients with disease control. Tailoring the conditioning regimen and maintenance therapy may improve outcomes in TP53 myeloid patients. In addition to chemotherapy, disease-modulating and immunological treatments continue to be studied to further improve outcomes.
Collapse
Affiliation(s)
- Hugo F Fernández
- Department of Malignant Hematology and Cellular Therapy, H. Lee Moffitt Cancer Center, Pembroke Pines, FL
| | - Asmita Mishra
- Department of Malignant Hematology and Cellular Therapy, H. Lee Moffitt Cancer Center, Pembroke Pines, FL
| |
Collapse
|
32
|
Yoshida K. Clonal hematopoiesis in cancer predisposition syndromes. Int J Hematol 2024:10.1007/s12185-024-03878-x. [PMID: 39643764 DOI: 10.1007/s12185-024-03878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 11/02/2024] [Accepted: 11/10/2024] [Indexed: 12/09/2024]
Abstract
After recent advances in sequencing technologies led to the discovery of novel genes associated with predisposition to hematological malignancies, studies have now shown that myeloid neoplasms associated with germline variants are more common than previously estimated. Based on these findings, myeloid neoplasms with germline predisposition have emerged as a unique category in the recent World Health Organization classification of Haematolymphoid Tumors. Clonal hematopoiesis is common in healthy individuals, particularly in older people. In patients with germline predisposition to hematological malignancies, clonal hematopoiesis is frequently observed at younger ages and is often associated with unique disease-specific driver mutations, some of which are hypothesized to compensate for the inherited defect. This review summarizes recent findings on clonal hematopoiesis in cancer predisposition syndromes.
Collapse
Affiliation(s)
- Kenichi Yoshida
- Division of Cancer Evolution, National Cancer Center Research Institute, Tokyo, Japan.
| |
Collapse
|
33
|
Aakash F, Gisriel SD, Zeidan AM, Bennett JM, Bejar R, Bewersdorf JP, Borate UM, Boultwood J, Brunner AM, Buckstein R, Carraway HE, Churpek JE, Daver NG, DeZern AE, Efficace F, Fenaux P, Figueroa ME, Garcia-Manero G, Gore SD, Greenberg PL, Griffiths EA, Halene S, Hourigan CS, Kim TK, Kim N, Komrokji RS, Kutchroo VK, List AF, Little RF, Majeti R, Nazha A, Nimer SD, Odenike O, Padron E, Patnaik MM, Platzbecker U, Della Porta MG, Roboz GJ, Sallman DA, Santini V, Sanz G, Savona MR, Sekeres MA, Stahl M, Starczynowski DT, Steensma DP, Taylor J, Abdel-Wahab O, Wei AH, Xie Z, Xu ML, Hasserjian RP, Loghavi S. Contemporary Approach to the Diagnosis and Classification of Myelodysplastic Neoplasms/Syndromes-Recommendations From the International Consortium for Myelodysplastic Neoplasms/Syndromes (MDS [icMDS]). Mod Pathol 2024; 37:100615. [PMID: 39322118 DOI: 10.1016/j.modpat.2024.100615] [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: 04/21/2024] [Revised: 08/23/2024] [Accepted: 09/01/2024] [Indexed: 09/27/2024]
Abstract
Myelodysplastic neoplasms/syndromes (MDS) are a heterogeneous group of biologically distinct entities characterized by variable degrees of ineffective hematopoiesis. Recently, 2 classification systems (the 5th edition of the World Health Organization Classification of Haematolymphoid tTumours and the International Consensus Classification) further subcharacterized MDS into morphologically and genetically defined groups. Accurate diagnosis and subclassification of MDS require a multistep systemic approach. The International Consortium for MDS (icMDS) summarizes a contemporary, practical, and multimodal approach to MDS diagnosis and classification.
Collapse
Affiliation(s)
- Fnu Aakash
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas; Department of Pathology, University of Texas Medical Branch, Galveston, Texas
| | - Savanah D Gisriel
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut; Department of Pathology and Laboratory Medicine, University of Wisconsin-Madison, Madison, Wisconsin
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, Yale Cancer Center, New Haven, Connecticut
| | - John M Bennett
- James P. Wilmot Cancer Center, Division of Hematopathology, University of Rochester Medical Center, Rochester, New York
| | - Rafael Bejar
- Division of Hematology and Oncology, Moores Cancer Center, UC San Diego, La Jolla, California
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Uma M Borate
- Division of Hematology, The Ohio State University Comprehensive Cancer Center, Columbus, Ohio
| | - Jacqueline Boultwood
- Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Andrew M Brunner
- Division of Hematology, Massachusetts General Hospital Brigham, Boston, Massachusetts
| | - Rena Buckstein
- Division of Medical Oncology/Hematology, Department of Medicine, Sunnybrook Health Sciences Centre, Toronto, Ontario, Canada
| | - Hetty E Carraway
- Leukemia Program, Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Jane E Churpek
- Division of Haematology, Oncology, and Palliative Care, Department of Medicine, Carbone Cancer Center, University of Wisconsin-Madison, Madison, Wisconsin
| | - Naval G Daver
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, Maryland
| | - Fabio Efficace
- Health Outcomes Research Unit, Italian Group for Adult Hematologic Diseases (GIMEMA), Rome, Italy
| | - Pierre Fenaux
- Service d'hématologie, Hôpital Saint-Louis (Assistance Publique Hôpitaux de Paris), Université de Paris-Cité, Paris, France
| | - Maria E Figueroa
- Biochemistry & Molecular Biology, Sylvester Comprehensive Cancer Center. University of Miami Miller School of Medicine, Miami, Florida
| | | | - Steven D Gore
- Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, Maryland
| | - Peter L Greenberg
- Division of Hematology, Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | | | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine, Yale Cancer Center, New Haven, Connecticut
| | - Christopher S Hourigan
- Fralin Biomedical Research Institute, Virginia Tech FBRI Cancer Research Center, Washington, District of Columbia
| | - Tae Kon Kim
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | | | - Rami S Komrokji
- Malignant Hematology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | | | - Alan F List
- Chief Scientific Officer, Stelexis Therapeutics, New York, New York
| | - Richard F Little
- Cancer Therapy Evaluation Program, National Cancer Institute, Rockville, Maryland
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, California
| | - Aziz Nazha
- Department of Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, Pennsylvania
| | - Stephen D Nimer
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Olatoyosi Odenike
- Leukemia Program, Section of Hematology/Oncology, University of Chicago Medicine and University of Chicago Comprehensive Cancer Center, Chicago, Illinois
| | - Eric Padron
- Malignant Hematology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, Minnesota
| | - Uwe Platzbecker
- Department of Hematology and Cellular Therapy, University Hospital Leipzig, Leipzig, Germany
| | - Matteo G Della Porta
- IRCCS Humanitas Research Hospital, Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Gail J Roboz
- Weill Cornell Medical College and New York Presbyterian Hospital, New York, New York
| | - David A Sallman
- Malignant Hematology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Valeria Santini
- Myelodysplastic Syndromes Unit, Department of Experimental and Clinical Medicine, Hematology, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Guillermo Sanz
- Health Research Institute La Fe, Valencia, Spain; Hospital Universitario y Politécnico La Fe, Valencia, Spain; CIBERONC, IS Carlos III, Madrid, Spain
| | - Michael R Savona
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, Tennessee
| | - Mikkael A Sekeres
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Maximilian Stahl
- Department of Medical Oncology, Division of Leukemia, Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital, Cincinnati, Ohio
| | | | - Justin Taylor
- Division of Hematology, Department of Medicine, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, Florida
| | - Omar Abdel-Wahab
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Andrew H Wei
- Department of Haematology, Royal Melbourne Hospital, Walter and Eliza Hall Institute of Medical Research, University of Melbourne, Victoria, Australia
| | - Zhuoer Xie
- Malignant Hematology Program, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Mina L Xu
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut
| | - Robert P Hasserjian
- Department of Pathology, Massachusetts General Hospital Brigham, Boston, Massachusetts
| | - Sanam Loghavi
- Department of Hematopathology, University of Texas MD Anderson Cancer Center, Houston, Texas.
| |
Collapse
|
34
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [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: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
35
|
Tashakori M, Yohe S, Linden MA, McKenna RW. The prognostic effect of blast count in TP53 mutant myeloid neoplasms -the Minnesota experience. Leuk Lymphoma 2024; 65:2151-2162. [PMID: 39257174 DOI: 10.1080/10428194.2024.2400213] [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: 07/04/2024] [Revised: 08/23/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024]
Abstract
In 2022, the World Health Organization (WHO) and International Consensus Classification (ICC) recognized TP53 as an entity-defining alteration in myeloid neoplasms, yet with differing criteria that could lead to discrepant diagnoses and affect clinical trial eligibility. We studied 67 patients with TP53 mutant myeloid neoplasms, reclassifying them using both criteria. While most cases fulfill the criteria for TP53 mutant defined entities, most discrepancies were found in cases with ≥20% blasts. Patients were stratified into three groups based on blast count (<10%, 10-19%, and ≥20%) which revealed comparable clinicopathologic features, genetic characteristics, and outcomes. Notably, patients with ≥10% blasts had shorter overall survival compared to those with <10% blasts (8.1 vs. 12.4 months; p = 0.03). This study is among the few to examine TP53 mutant myeloid neoplasms as a single entity and suggests that the 10% blast count threshold could serve as a gateway to a more harmonized classification for these patients.
Collapse
Affiliation(s)
- Mehrnoosh Tashakori
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Sophia Yohe
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Michael A Linden
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| | - Robert W McKenna
- Department of Laboratory Medicine and Pathology, University of Minnesota, Minneapolis, MN, USA
| |
Collapse
|
36
|
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
|
37
|
Patel SA. Managing the unmanageable: evidence-driven approaches to real-world patient prototypes of TP53-mutant myelodysplastic neoplasms and acute myeloid leukemia. Leukemia 2024; 38:2544-2551. [PMID: 39349613 DOI: 10.1038/s41375-024-02417-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 09/13/2024] [Accepted: 09/17/2024] [Indexed: 11/27/2024]
Abstract
Patients with TP53 aberrations comprise the highest risk subset of all myeloid malignancies. The managerial conundrum of TP53-mutant myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML) stems from refractoriness to or relapse after conventional chemotherapy, as well as the limited translational success of investigational therapies targeting TP53-mutant cells. Thus far, no targeted therapies have been commercially approved for this mutational subset. As a result, management plans for patients with TP53-mutant MDS and AML are often driven by clinical judgment and/or physician preference rather than consensus guidelines backed by a rigorous evidence basis. This clinical case-based, evidence-driven review highlights the most salient data that guides the management of commonly encountered patient prototypes. This review discusses the therapeutic menu of first-line options that derive from multi-institutional experiences as well as from disease-centric consortia and discusses how these first-line options can be optimally tailored to heterogeneous groups of patients. The debate regarding whether allogeneic stem cell transplant should be offered to these patients is summarized. Finally, this review explores the recent unfortunate news of pauses in clinical trials for the leading investigational agents - eprenetapopt, magrolimab, sabatolimab, and idasanutlin - and offers solutions toward re-invigorating the pipeline of precision therapeutics in 2025.
Collapse
Affiliation(s)
- Shyam A Patel
- Department of Medicine - Division of Hematology/Oncology; Center for Clinical and Translational Science; Morningside Graduate School of Biomedical Sciences; UMass Chan Medical School, 55 Lake Avenue North, Worcester, MA, 01655, USA.
| |
Collapse
|
38
|
Shahzad M, Amin MK, Daver NG, Shah MV, Hiwase D, Arber DA, Kharfan-Dabaja MA, Badar T. What have we learned about TP53-mutated acute myeloid leukemia? Blood Cancer J 2024; 14:202. [PMID: 39562552 PMCID: PMC11576745 DOI: 10.1038/s41408-024-01186-5] [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: 08/07/2024] [Revised: 10/31/2024] [Accepted: 11/04/2024] [Indexed: 11/21/2024] Open
Abstract
TP53 is a tumor suppressor gene frequently mutated in human cancers and is generally associated with poor outcomes. TP53 mutations are found in approximately 5% to 10% of patients with de novo acute myeloid leukemia (AML), more frequently observed in elderly patients and those with therapy-related AML. Despite recent advances in molecular profiling and the emergence of targeted therapies, TP53-mutated AML remains a challenge to treat. Current treatment strategies, including conventional chemotherapy, hypomethylating agents, and venetoclax-based therapies, have shown limited efficacy in TP53-mutated AML, with low response rates and poor overall survival. Allogeneic hematopoietic stem cell transplantation is a potentially curative option; however, its efficacy in TP53-mutated AML depends on comorbid conditions and disease status at transplantation. Novel therapeutic modalities, including immune-based therapies, did show promise in early-phase studies but did not translate into effective therapies in randomized controlled trials. This review provides a comprehensive overview of TP53 mutations in AML, outcomes based on allelic burden, clinical implications, and therapeutic challenges.
Collapse
Affiliation(s)
- Moazzam Shahzad
- Division of Hematology and Oncology, Moffitt Cancer Center, Tampa, FL, USA
| | - Muhammad Kashif Amin
- Division of Hematologic Malignancies & Cellular Therapeutics, The University of Kansas Medical Center, Kansas City, KS, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Devendra Hiwase
- Department of Hematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA, Australia
| | - Daniel A Arber
- Department of Pathology, University of Chicago, Chicago, IL, USA
| | | | - Talha Badar
- Division of Hematology and Medical Oncology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
39
|
Ahmadi SE, Rahimian E, Rahimi S, Zarandi B, Bahraini M, Soleymani M, Safdari SM, Shabannezhad A, Jaafari N, Safa M. From regulation to deregulation of p53 in hematologic malignancies: implications for diagnosis, prognosis and therapy. Biomark Res 2024; 12:137. [PMID: 39538363 PMCID: PMC11565275 DOI: 10.1186/s40364-024-00676-9] [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: 07/16/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024] Open
Abstract
The p53 protein, encoded by the TP53 gene, serves as a critical tumor suppressor, playing a vital role in maintaining genomic stability and regulating cellular responses to stress. Dysregulation of p53 is frequently observed in hematological malignancies, significantly impacting disease progression and patient outcomes. This review aims to examine the regulatory mechanisms of p53, the implications of TP53 mutations in various hematological cancers, and emerging therapeutic strategies targeting p53. We conducted a comprehensive literature review to synthesize recent findings related to p53's multifaceted role in hematologic cancers, focusing on its regulatory pathways and therapeutic potential. TP53 mutations in hematological malignancies often lead to treatment resistance and poor prognosis. Current therapeutic strategies, including p53 reactivation and gene therapy, show promise in improving treatment outcomes. Understanding the intricacies of p53 regulation and the consequences of its mutations is essential for developing effective diagnostic and therapeutic strategies in hematological malignancies, ultimately enhancing patient care and survival.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Rahimian
- Department of Medical Translational Oncology, National Center for Tumor Diseases (NCT) Dresden, Dresden, Germany
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mehran Bahraini
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Maral Soleymani
- Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Seyed Mehrab Safdari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Ashkan Shabannezhad
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Jaafari
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Pharmacology and Chemical Biology, University of Pittsburgh, Pittsburgh, PA, 15261, USA
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
40
|
Kubota Y, Kimura S. Current Understanding of the Role of Autophagy in the Treatment of Myeloid Leukemia. Int J Mol Sci 2024; 25:12219. [PMID: 39596291 PMCID: PMC11594995 DOI: 10.3390/ijms252212219] [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: 09/29/2024] [Revised: 11/12/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024] Open
Abstract
The most important issues in acute myeloid leukemia are preventing relapse and treating relapse. Although the remission rate has improved to approximately 80%, the 5-year survival rate is only around 30%. The main reasons for this are the high relapse rate and the limited treatment options. In chronic myeloid leukemia patients, when a deep molecular response is achieved for a certain period of time through tyrosine kinase inhibitor treatment, about half of them will reach treatment-free remission, but relapse is still a problem. Therefore, potential therapeutic targets for myeloid leukemias are eagerly awaited. Autophagy suppresses the development of cancer by maintaining cellular homeostasis; however, it also promotes cancer progression by helping cancer cells survive under various metabolic stresses. In addition, autophagy is promoted or suppressed in cancer cells by various genetic mutations. Therefore, the development of therapies that target autophagy is also being actively researched in the field of leukemia. In this review, studies of the role of autophagy in hematopoiesis, leukemogenesis, and myeloid leukemias are presented, and the impact of autophagy regulation on leukemia treatment and the clinical trials of autophagy-related drugs to date is discussed.
Collapse
MESH Headings
- Humans
- Autophagy
- Animals
- Leukemia, Myeloid/pathology
- Leukemia, Myeloid/genetics
- Leukemia, Myeloid/therapy
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/drug therapy
- Antineoplastic Agents/therapeutic use
- Antineoplastic Agents/pharmacology
- Hematopoiesis
Collapse
Affiliation(s)
- Yasushi Kubota
- Department of Clinical Laboratory Medicine, Saga-Ken Medical Centre Koseikan, Saga 840-8571, Japan
| | - Shinya Kimura
- Division of Hematology, Respiratory Medicine and Oncology, Department of Internal Medicine, Faculty of Medicine, Saga University, Saga 849-8501, Japan;
| |
Collapse
|
41
|
Zhang L, Abro B, Campbell A, Ding Y. TP53 mutations in myeloid neoplasms: implications for accurate laboratory detection, diagnosis, and treatment. Lab Med 2024; 55:686-699. [PMID: 39001691 PMCID: PMC11532620 DOI: 10.1093/labmed/lmae048] [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] [Indexed: 11/05/2024] Open
Abstract
Genetic alterations that affect the function of p53 tumor suppressor have been extensively investigated in myeloid neoplasms, revealing their significant impact on disease progression, treatment response, and patient outcomes. The identification and characterization of TP53 mutations play pivotal roles in subclassifying myeloid neoplasms and guiding treatment decisions. Starting with the presentation of a typical case, this review highlights the complicated nature of genetic alterations involving TP53 and provides a comprehensive analysis of TP53 mutations and other alterations in myeloid neoplasms. Currently available methods used in clinical laboratories to identify TP53 mutations are discussed, focusing on the importance of establishing a robust testing protocol within clinical laboratories to ensure the delivery of accurate and reliable results. The treatment implications of TP53 mutations in myeloid neoplasms and clinical trial options are reviewed. Ultimately, we hope that this review provides valuable insights into the patterns of TP53 alterations in myeloid neoplasms and offers guidance to establish practical laboratory testing protocols to support the best practices of precision oncology.
Collapse
Affiliation(s)
- Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, US
| | - Brooj Abro
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, US
| | - Andrew Campbell
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA, US
| | - Yi Ding
- Department of Laboratory Medicine, Geisinger Medical Center, Danville, PA, US
| |
Collapse
|
42
|
Singhal D, Kutyna MM, Hahn CN, Shah MV, Hiwase DK. Therapy-Related Myeloid Neoplasms: Complex Interactions among Cytotoxic Therapies, Genetic Factors, and Aberrant Microenvironment. Blood Cancer Discov 2024; 5:400-416. [PMID: 39422544 PMCID: PMC11528189 DOI: 10.1158/2643-3230.bcd-24-0103] [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: 04/26/2024] [Revised: 07/21/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024] Open
Abstract
Therapy-related myeloid neoplasm (t-MN), characterized by its association with prior exposure to cytotoxic therapy, remains poorly understood and is a major impediment to long-term survival even in the era of novel targeted therapies due to its aggressive nature and treatment resistance. Previously, cytotoxic therapy-induced genomic changes in hematopoietic stem cells were considered sine qua non in pathogenesis; however, recent research demonstrates a complex interaction between acquired and hereditary genetic predispositions, along with a profoundly senescent bone marrow (BM) microenvironment. We review emerging data on t-MN risk factors and explore the intricate interplay among clonal hematopoiesis, genetic predisposition, and the abnormal BM microenvironment. Significance: t-MN represents a poorly understood blood cancer with extremely poor survival and no effective therapies. We provide a comprehensive review of recent preclinical research highlighting complex interaction among emerging therapies, hereditary and acquired genetic factors, and BM microenvironment. Understanding the risk factors associated with t-MN is crucial for clinicians, molecular pathologists, and cancer biologists to anticipate and potentially reduce its incidence in the future. Moreover, better understanding of the molecular pathogenesis of t-MN may enable preemptive screening and even intervention in high-risk patients.
Collapse
Affiliation(s)
- Deepak Singhal
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | - Monika M. Kutyna
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
| | - Christopher N. Hahn
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
- Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
| | | | - Devendra K. Hiwase
- Department of Haematology, Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, Australia
- Adelaide Medical School, University of Adelaide, Adelaide, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, Australia
| |
Collapse
|
43
|
Zak T, Sukhanova M, Gao J, Fu L, Chen YH, Chen QC, Behdad A, Tariq H. Therapy-related myeloid neoplasms with single-hit TP53 mutations share the clinical, molecular, and survival characteristics of their multi-hit counterparts. Leuk Lymphoma 2024; 65:1691-1697. [PMID: 38884125 DOI: 10.1080/10428194.2024.2367699] [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: 04/03/2024] [Revised: 05/10/2024] [Accepted: 06/09/2024] [Indexed: 06/18/2024]
Abstract
Recent updates in the classification of myeloid neoplasms (MNs) recognize the poor prognostic impact of TP53 mutations, with particular emphasis on the TP53 allele status. Studies on the effect of TP53 allele status exclusively in therapy-related MNs (t-MNs) are lacking. We compared the clinicopathologic and survival characteristics of t-MNs with single-hit (SH) and multi-hit (MH) TP53 mutations. A total of 71 TP53-mutated t-MNs were included, including 56 (78.9%) MH and 15 (21.1%) SH. Both groups showed comparable genetic profiles with an excess of high-risk karyotypes and a paucity of other co-mutated genes. TP53 was the sole detectable mutation in 73.3% of SH and 75.0% of MH cases. The overall survival (OS) of SH TP53-mutated t-MNs was not significantly different from MH cases (median survival: 233 vs.273 days, p = 0.70). Our findings suggest that t-MNs with SH TP53 mutations share the poor prognostic and biologic profile of their MH counterparts.
Collapse
Affiliation(s)
- Taylor Zak
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Madina Sukhanova
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Juehua Gao
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lucy Fu
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Yi-Hua Chen
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Qing Ching Chen
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Amir Behdad
- Department of Pathology and Laboratory Medicine, Cleveland Clinic Florida, Weston, FL, USA
| | - Hamza Tariq
- Department of Pathology, Northwestern Memorial Hospital, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| |
Collapse
|
44
|
Bidikian A, Bewersdorf JP, Shallis RM, Getz TM, Stempel JM, Kewan T, Stahl M, Zeidan AM. Targeted therapies for myelodysplastic syndromes/neoplasms (MDS): current landscape and future directions. Expert Rev Anticancer Ther 2024; 24:1131-1146. [PMID: 39367718 DOI: 10.1080/14737140.2024.2414071] [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: 08/28/2024] [Revised: 10/01/2024] [Accepted: 10/04/2024] [Indexed: 10/06/2024]
Abstract
INTRODUCTION Myelodysplastic syndromes/neoplasms (MDS) are a heterogeneous group of hematologic malignancies that are stratified into high-risk (HR-MDS) and low-risk (LR-MDS) categories. Until recently, LR-MDS has been typically managed by supportive measures and erythropoiesis-stimulating agents (ESAs); whereas management of HR-MDS typically included hypomethylating agents and allogeneic hematopoietic stem cell transplant. However, the limited rates and durations of response observed with these interventions prompted the search for targeted therapies to improve the outcomes among patients with MDS. AREAS COVERED Here, we review the current landscape of targeted therapies in MDS. These include pyruvate kinase and hypoxia-inducible factor (HIF) activators; TGF-beta, telomerase, BCL2 and isocitrate dehydrogenase (IDH) inhibitors; as well as novel approaches targeting inflammation, pyroptosis, immune evasion, and RNA splicing machinery. EXPERT OPINION This review highlights the progress and challenges in MDS treatment. Despite some promising results, many therapies remain in early development or have faced setbacks, emphasizing the need for a more comprehensive understanding of the disease's pathobiology. Continued research into targeted therapies, homogenous clinical trial designs, as well as increased incorporation of molecular prognostic tools and artificial intelligence into trial design are essential for developing effective treatments for MDS and improving patient outcomes.
Collapse
Affiliation(s)
- Aram Bidikian
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Jan P Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Rory M Shallis
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Ted M Getz
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Jessica M Stempel
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Tariq Kewan
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale School of Medicine and Yale New Haven Hospital, New Haven, CT, USA
| |
Collapse
|
45
|
Wang H, Li X, Qi J, Liu H, Chu T, Xu X, Qiu H, Fu C, Tang X, Ruan C, Wu D, Han Y. Prognostic mutations identified by whole-exome sequencing and validation of the Molecular International Prognostic Scoring System in myelodysplastic syndromes after allogeneic haematopoietic stem cell transplantation. Br J Haematol 2024; 205:1899-1909. [PMID: 39138006 DOI: 10.1111/bjh.19707] [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: 04/28/2024] [Accepted: 08/05/2024] [Indexed: 08/15/2024]
Abstract
In this study, we used the whole-exome sequencing (WES) approach to obtain genomic profiles from 92 marrow samples of myelodysplastic syndrome (MDS) patients before haematopoietic stem cell transplantation. We identified 129 mutations in 45 driver genes. Fifty-five patients (59.8%) carried at least 1 driver mutation. The splicing factor U2AF1 was the most frequently mutated in the cohort (21 cases, 23%), followed by BCOR (9 cases, 10%), ASXL1 (8 cases, 9%), TET2 (6 cases, 7%), NPM1 (5 cases, 5%), RUNX1 (5 cases, 5%), and SETBP1 (5 cases, 5%). WES also identified 49 possible oncogenic variants in six genes (PIEZO1, LOXHD1, MYH13, DNAH5, DPH1, and USH2A) that were associated with overall survival (OS) or relapse-free survival (RFS) in MDS after transplantation. Multivariate analysis showed mutations in DNAH5 and USH2A to be independent risk factors for OS. Mutations in DNAH5 and LOXHD1 were risk factors for worse RFS. The Molecular International Prognostic Scoring System retained its independent prognostic significance for RFS after multivariate analysis.
Collapse
Affiliation(s)
- Hong Wang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Xueqian Li
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
| | - Jiaqian Qi
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Hong Liu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Tiantian Chu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Xiaoyan Xu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Huiying Qiu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Chengcheng Fu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Xiaowen Tang
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Changgeng Ruan
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Depei Wu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| | - Yue Han
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
- Key Laboratory of Thrombosis and Hemostasis of Ministry of Health, Suzhou, China
- State Key Laboratory of Radiation Medicine and Protection, Soochow University, Suzhou, China
| |
Collapse
|
46
|
Puzo CJ, Hager KM, Rinder HM, Weinberg OK, Siddon AJ. Overall survival in TP53-mutated AML and MDS. Ann Hematol 2024:10.1007/s00277-024-06054-7. [PMID: 39443370 DOI: 10.1007/s00277-024-06054-7] [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: 08/08/2024] [Accepted: 10/18/2024] [Indexed: 10/25/2024]
Abstract
TP53 mutations in patients with AML and MDS frequently portend a poor prognosis, related to both p53 allele status and blast count. In 2022, the ICC and WHO released updated guidelines for classifying p53-mutated AML/MDS. The characteristics of p53 mutations, their associated co-mutations, and their effects on overall survival (OS) are not known in the context of these new guidelines. A retrospective chart review was undertaken for all patients with AML or MDS and at least one TP53 mutation detected on next generation sequencing (NGS) at Yale New Haven Hospital from 2015 to 2023. All patients (N = 210) met criteria for one of the 5 diagnostic classes based on WHO and ICC guidelines. Kaplan-Meier curves with associated log-rank testing and Cox proportional hazards model quantified the effects of clinical and molecular data on survival. Multi-hit pathogenic mutations were related to poorer OS in MDS but not AML using either the WHO (p = .02) or the ICC (p = .01) diagnostic criteria. The most significant predictors of OS in the sample overall were platelet count < 50 K (HR: 2.01, 95% CI [1.47, 2.75], p < .001) and TP53 VAF ≤ 40% (HR: 0.68, 95% CI[0.50, 0.91], p = .01). Blast count ranges, complex karyotype, and p53 mutation type or location, showed no association with OS. In our cohort defined by the 2022 ICC and WHO criteria, VAF and thrombocytopenia, rather than blast count or p53 mutation features, significantly predicted OS. These results speak to each criteria's ability to identify cases of similarly aggressive disease biology and prognosis.
Collapse
Affiliation(s)
| | - Karl M Hager
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Henry M Rinder
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA
| | - Olga K Weinberg
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alexa J Siddon
- Department of Laboratory Medicine, Yale School of Medicine, 333 Cedar Street, New Haven, CT, 06520, USA.
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
47
|
Saba L, Nasr L, Manrique-Succar J, Diacovo J, Chaulagain C. How we manage multiple myeloma with clonal hematopoiesis of indeterminate potential (CHIP): a case report. ANNALS OF TRANSLATIONAL MEDICINE 2024; 12:96. [PMID: 39507461 PMCID: PMC11534749 DOI: 10.21037/atm-23-1945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 06/28/2024] [Indexed: 11/08/2024]
Abstract
Background Clonal hematopoiesis of indeterminate potential (CHIP) is characterized by genetic alterations associated with hematologic neoplasms, posing clinical challenges in managing concurrent hematological malignancies. CHIP may complicate the treatment landscape due to its potential to influence disease progression and treatment response. We report a 73-year-old male with multiple myeloma (MM) harboring a CHIP PPM1D mutation, elucidating the complexities and therapeutic considerations in such cases. Case Description After four cycles of cyclophosphamide, bortezomib, and dexamethasone therapy, he achieved a partial response, followed by complete hematologic response (CR) post eight cycles of lenalidomide, dexamethasone, and bortezomib therapy. Despite this, upfront autologous hematopoietic stem cell transplantation (HSCT) was initially deemed unsuitable due to positive PPM1D CHIP status. HSCT proceeded after aggressive relapse with clonal evolution but yielded short-lived response. Following failure of >4 lines of therapy, he received chimeric antigen receptor T (CAR-T) cell therapy (ciltacabtagene autoleucel) for salvage. This approach successfully induced remission, which was maintained for 6 months. Conclusions This case report highlights MM management complexities in CHIP presence, suggesting potential utility of HSCT and CAR-T cell therapy. Prospective studies are necessary to evaluate the safety and efficacy of these therapies in myeloma patients with concurrent CHIP, aiming to optimize treatment strategies and improve outcomes in this challenging clinical context.
Collapse
Affiliation(s)
- Ludovic Saba
- Department of Hematology and Medical Oncology, Cleveland Clinic Florida, Weston, FL, USA
| | - Lewis Nasr
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Julia Diacovo
- Pathology and Laboratory Medicine Institute, Cleveland Clinic Florida, Weston, FL, USA
| | - Chakra Chaulagain
- Department of Hematology and Medical Oncology, Cleveland Clinic Florida, Weston, FL, USA
| |
Collapse
|
48
|
Montoro MJ, Palomo L, Haferlach C, Acha P, Chan O, Navarro V, Kubota Y, Schulz FI, Meggendorfer M, Briski R, Al Ali N, Xicoy B, López-Cadenas F, Bosch F, González T, Eder LN, Jerez A, Wang YH, Campagna A, Santini V, Bernal Del Castillo T, Such E, Tien HF, Diaz Varela N, Platzbecker U, Haase D, Díez-Campelo M, Della Porta M, Garcia-Manero G, Wiseman DH, Germing U, Maciejewski JP, Komrokji RS, Sole F, Haferlach T, Valcárcel D. Influence of TP53 gene mutations and their allelic status in myelodysplastic syndromes with isolated 5q deletion. Blood 2024; 144:1722-1731. [PMID: 39074355 DOI: 10.1182/blood.2024023840] [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: 01/05/2024] [Revised: 06/10/2024] [Accepted: 06/30/2024] [Indexed: 07/31/2024] Open
Abstract
ABSTRACT Mutations in the TP53 gene, particularly multihit alterations, have been associated with unfavorable clinical features and prognosis in patients diagnosed with myelodysplastic syndrome (MDS). Despite this, the role of TP53 gene aberrations in MDS with isolated deletion of chromosome 5 [MDS-del(5q)] remains unclear. This study aimed to assess the impact of TP53 gene mutations and their allelic state in patients with MDS-del(5q). To that end, a comprehensive analysis of TP53 abnormalities, examining both TP53 mutations and allelic imbalances, in 682 patients diagnosed with MDS-del(5q) was conducted. Twenty-four percent of TP53-mutated patients exhibited multihit alterations, whereas the remaining patients displayed monoallelic mutations. TP53-multihit alterations were predictive of an increased risk of leukemic transformation. The impact of monoallelic alterations was dependent on the variant allele frequency (VAF); patients with TP53-monoallelic mutations and VAF <20% exhibited behavior similar to TP53 wild type, and those with TP53-monoallelic mutations and VAF ≥20% presented outcomes equivalent to TP53-multihit patients. This study underscores the importance of considering TP53 allelic state and VAF in the risk stratification and treatment decision-making process for patients with MDS-del(5q).
Collapse
Affiliation(s)
- Maria Julia Montoro
- Department of Hematology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Laura Palomo
- Department of Hematology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | | | - Pamela Acha
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Badalona, Spain
| | - Onyee Chan
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Víctor Navarro
- Oncology Data Science Group, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Yasuo Kubota
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Felicitas Isabel Schulz
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | | | - Robert Briski
- Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, TX
| | - Najla Al Ali
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Blanca Xicoy
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Badalona, Spain
| | | | - Francesc Bosch
- Department of Hematology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| | - Teresa González
- Department of Hematology, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Lea Naomi Eder
- Clinics of Hematology and Medical Oncology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - Andrés Jerez
- Department of Hematology, Hospital Morales Meseguer, Murcia, Spain
| | - Yu-Hung Wang
- Division of Cancer Sciences, Epigenetics of Haematopoiesis Laboratory, The University of Manchester, Manchester, United Kingdom
| | - Alessia Campagna
- Department of Biomedical Sciences, Humanitas Clinical and Research Center-IRCCS and Humanitas University, Milan, Italy
| | - Valeria Santini
- Hematology, MDS Unit, University of Florence, Azienda Ospedaliero Universitaria Careggi, Florence, Italy
| | - Teresa Bernal Del Castillo
- Department of Hematology, Servicio de Hematología, Hospital Universitario Central de Asturias Instituto de Investigación del Principado de Asturias, Oviedo, Spain
| | - Esperanza Such
- Department of Hematology, Servicio de Hematología, Hospital La Fe, Valencia, Spain
| | - Hwei-Fang Tien
- Division of Hematology, Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Nicolás Diaz Varela
- Department of Hematology, Servicio de Hematología, Hospital Universitario Central de Asturias Instituto de Investigación del Principado de Asturias, Oviedo, Spain
| | - Uwe Platzbecker
- Department of Hematology, University Hospital of Leipzig, Dresden, Germany
| | - Detlef Haase
- Clinics of Hematology and Medical Oncology, University Medical Center Göttingen, Georg August University, Göttingen, Germany
| | - María Díez-Campelo
- Department of Hematology, Hospital Universitario de Salamanca, Instituto de Investigación Biomédica de Salamanca, Salamanca, Spain
| | - Matteo Della Porta
- Department of Biomedical Sciences, Humanitas Clinical and Research Center-IRCCS and Humanitas University, Milan, Italy
| | | | - Daniel H Wiseman
- Division of Cancer Sciences, Epigenetics of Haematopoiesis Laboratory, The University of Manchester, Manchester, United Kingdom
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, University Hospital of Düsseldorf, Düsseldorf, Germany
| | - Jaroslaw P Maciejewski
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Rami S Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Francesc Sole
- MDS Group, Institut de Recerca Contra la Leucèmia Josep Carreras, Badalona, Spain
| | | | - David Valcárcel
- Department of Hematology, Vall d'Hebron Barcelona Hospital Campus, Vall d'Hebron Institute of Oncology, Barcelona, Spain
| |
Collapse
|
49
|
Bernard E, Hasserjian RP, Greenberg PL, Arango Ossa JE, Creignou M, Tuechler H, Gutierrez-Abril J, Domenico D, Medina-Martinez JS, Levine M, Liosis K, Farnoud N, Sirenko M, Jädersten M, Germing U, Sanz G, van de Loosdrecht AA, Nannya Y, Kosmider O, Follo MY, Thol F, Zamora L, Pinheiro RF, Pellagatti A, Elias HK, Haase D, Ganster C, Ades L, Tobiasson M, Palomo L, Della Porta MG, Fenaux P, Belickova M, Savona MR, Klimek VM, Santos FPS, Boultwood J, Kotsianidis I, Santini V, Solé F, Platzbecker U, Heuser M, Valent P, Finelli C, Voso MT, Shih LY, Fontenay M, Jansen JH, Cervera J, Gattermann N, Ebert BL, Bejar R, Malcovati L, Ogawa S, Cazzola M, Hellström-Lindberg E, Papaemmanuil E. Molecular taxonomy of myelodysplastic syndromes and its clinical implications. Blood 2024; 144:1617-1632. [PMID: 38958467 PMCID: PMC11487646 DOI: 10.1182/blood.2023023727] [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: 01/03/2024] [Revised: 05/13/2024] [Accepted: 06/05/2024] [Indexed: 07/04/2024] Open
Abstract
ABSTRACT Myelodysplastic syndromes (MDS) are clonal hematologic disorders characterized by morphologic abnormalities of myeloid cells and peripheral cytopenias. Although genetic abnormalities underlie the pathogenesis of these disorders and their heterogeneity, current classifications of MDS rely predominantly on morphology. We performed genomic profiling of 3233 patients with MDS or related disorders to delineate molecular subtypes and define their clinical implications. Gene mutations, copy-number alterations, and copy-neutral loss of heterozygosity were derived from targeted sequencing of a 152-gene panel, with abnormalities identified in 91%, 43%, and 11% of patients, respectively. We characterized 16 molecular groups, encompassing 86% of patients, using information from 21 genes, 6 cytogenetic events, and loss of heterozygosity at the TP53 and TET2 loci. Two residual groups defined by negative findings (molecularly not otherwise specified, absence of recurrent drivers) comprised 14% of patients. The groups varied in size from 0.5% to 14% of patients and were associated with distinct clinical phenotypes and outcomes. The median bone marrow (BM) blast percentage across groups ranged from 1.5% to 10%, and the median overall survival ranged from 0.9 to 8.2 years. We validated 5 well-characterized entities, added further evidence to support 3 previously reported subsets, and described 8 novel groups. The prognostic influence of BM blasts depended on the genetic subtypes. Within genetic subgroups, therapy-related MDS and myelodysplastic/myeloproliferative neoplasms had comparable clinical and outcome profiles to primary MDS. In conclusion, genetically-derived subgroups of MDS are clinically relevant and might inform future classification schemas and translational therapeutic research.
Collapse
Affiliation(s)
- Elsa Bernard
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
- Department of Computational Oncology, UMR 981, Gustave Roussy, Villejuif, France
| | | | - Peter L. Greenberg
- Division of Hematology, Stanford University Cancer Institute, Stanford, CA
| | - Juan E. Arango Ossa
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria Creignou
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | | | - Jesus Gutierrez-Abril
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Dylan Domenico
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Juan S. Medina-Martinez
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Max Levine
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Konstantinos Liosis
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Noushin Farnoud
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maria Sirenko
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Martin Jädersten
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Ulrich Germing
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - Guillermo Sanz
- Department of Hematology, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red Cáncer, Instituto de Salud Carlos III, Madrid, Spain
| | - Arjan A. van de Loosdrecht
- Department of Hematology, Amsterdam University Medical Center, Vrije University Medical Center, Amsterdam, The Netherlands
| | - Yasuhito Nannya
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
| | - Olivier Kosmider
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin and Université de Paris, Université Paris Descartes, Paris, France
| | - Matilde Y. Follo
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Bologna, Italy
| | - Felicitas Thol
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Lurdes Zamora
- Hematology Department, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Barcelona, Spain
- Myelodysplastic Syndromes Group, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Ronald F. Pinheiro
- Drug Research and Development Center, Federal University of Ceara, Ceara, Brazil
| | - Andrea Pellagatti
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, United KIngdom
| | - Harold K. Elias
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Detlef Haase
- Clinics of Hematology and Medical Oncology, University Medical Center, Göttingen, Germany
| | - Christina Ganster
- Clinics of Hematology and Medical Oncology, University Medical Center, Göttingen, Germany
| | - Lionel Ades
- Department of Hematology, Hôpital St Louis, and Paris University, Paris, France
| | - Magnus Tobiasson
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Laura Palomo
- Hematology Department, Catalan Institute of Oncology, Hospital Germans Trias i Pujol, Barcelona, Spain
- Myelodysplastic Syndromes Group, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Matteo Giovanni Della Porta
- Department of Biomedical Sciences, Humanitas Clinical and Research Center and Humanitas University, Milan, Italy
| | - Pierre Fenaux
- Department of Hematology, Hôpital St Louis, and Paris University, Paris, France
| | - Monika Belickova
- Department of Genomics, Institute of Hematology and Blood Transfusion, Prague, Czech Republic
| | - Michael R. Savona
- Department of Medicine, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN
| | - Virginia M. Klimek
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Fabio P. S. Santos
- Oncology-Hematology Center, Hospital Israelita Albert Einstein, São Paulo, Brazil
| | - Jacqueline Boultwood
- Radcliffe Department of Medicine, Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, United KIngdom
| | - Ioannis Kotsianidis
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, Alexandroupolis, Greece
| | - Valeria Santini
- Myelodysplastic Syndromes Unit, Department of Experimental and Clinical Medicine, Hematology, Azienda Ospedaliero Universitaria Careggi, University of Florence, Florence, Italy
| | - Francesc Solé
- Myelodysplastic Syndromes Group, Josep Carreras Leukemia Research Institute, Barcelona, Spain
| | - Uwe Platzbecker
- Medical Clinic and Policlinic 1, Hematology and Cellular Therapy, University of Leipzig, Leipzig, Germany
| | - Michael Heuser
- Department of Hematology, Hemostasis, Oncology, and Stem Cell Transplantation, Hannover Medical School, Hannover, Germany
| | - Peter Valent
- Division of Hematology and Hemostaseology, Department of Internal Medicine I, Medical University of Vienna, Vienna, Austria
- Department of Internal Medicine I, Ludwig Boltzmann Institute for Hematology and Hemostaseology, Medical University of Vienna, Vienna, Austria
| | - Carlo Finelli
- Institute of Hematology Seràgnoli, Istituti di Ricovero e Cura a Carattere Scientifico Azienda Ospedaliero-Universitaria di Bologna, Bologna, Italy
| | - Maria Teresa Voso
- Myelodysplastic Syndromes Cooperative Group Gruppo Laziale Mielo-displasie (GROM-L), Department of Biomedicine and Prevention, Tor Vergata University, Rome, Italy
| | - Lee-Yung Shih
- Division of Hematology, Chang Gung Memorial Hospital at Linkou, Chang Gung University, Taoyuan City, Taiwan
| | - Michaela Fontenay
- Department of Hematology, Assistance Publique-Hôpitaux de Paris, Hôpital Cochin and Université de Paris, Université Paris Descartes, Paris, France
| | - Joop H. Jansen
- Laboratory of Hematology, Department of Laboratory Medicine, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - José Cervera
- Department of Hematology and Genetics Unit, University Hospital La Fe, Valencia, Spain
| | - Norbert Gattermann
- Department of Hematology, Oncology, and Clinical Immunology, Heinrich Heine University, Düsseldorf, Germany
| | - Benjamin L. Ebert
- Department of Medical Oncology and Howard Hughes Medical Institute, Dana-Farber Cancer Center, Boston, MA
| | - Rafael Bejar
- University of California San Diego Moores Cancer Center, La Jolla, CA
| | - Luca Malcovati
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Seishi Ogawa
- Department of Pathology and Tumor Biology, Kyoto University, Kyoto, Japan
- Institute for the Advanced Study of Human Biology, Kyoto University, Kyoto, Japan
| | - Mario Cazzola
- Department of Hematology Oncology, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Policlinico San Matteo, Pavia, Italy
- Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Eva Hellström-Lindberg
- Department of Medicine Huddinge, Center for Hematology and Regenerative Medicine, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Elli Papaemmanuil
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY
| |
Collapse
|
50
|
Canichella M, Molica M, Mazzone C, de Fabritiis P. Maintenance Therapy Post-Hematopoietic Stem Cell Transplantation in Acute Myeloid Leukemia. Curr Oncol 2024; 31:6050-6060. [PMID: 39451755 PMCID: PMC11506619 DOI: 10.3390/curroncol31100451] [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: 09/11/2024] [Revised: 10/05/2024] [Accepted: 10/08/2024] [Indexed: 10/26/2024] Open
Abstract
High-risk acute myeloid leukemia has been associated with a poor outcome. Hematopoietic stem cell transplantation (HSCT) represents the only curative option for eligible patients. Relapse after HSCT is a dramatic event with poor chances of survival. With the aim of reducing the rate of post-HSCT relapse, maintenance treatment has been investigated in this setting. Results from clinical trials suggest an advantage in the use of a maintenance strategy; however, standardized guidelines are not yet available due to the lack of prospective clinical trials. In this review, we have reported the most important strategies adopted as post-HSCT maintenance, highlighting their efficacy, but the current research also opens questions.
Collapse
Affiliation(s)
| | - Matteo Molica
- Department of Hematology-Oncology, Azienda Ospedaliera Pugliese-Ciaccio, 88100 Catanzaro, Italy;
| | - Carla Mazzone
- Hematology, St. Eugenio Hospital, ASL Roma2, 00144 Rome, Italy
| | - Paolo de Fabritiis
- Hematology, St. Eugenio Hospital, ASL Roma2, 00144 Rome, Italy
- Department of Biomedicina e Prevenzione, Tor Vergata University, 00133 Rome, Italy
| |
Collapse
|