1
|
Huang N, Song Y, Wu L, He Q, Zhang Z, Guo J, Xu F, Chang C, Li X. Validation and improvement of the molecular international prognostic scoring system in Chinese patients with myelodysplastic syndromes. Ann Hematol 2024:10.1007/s00277-024-06162-4. [PMID: 39738836 DOI: 10.1007/s00277-024-06162-4] [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: 10/07/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
Various prognostic models have been proposed to improve the accuracy of prognostic assessment for Myelodysplastic syndromes (MDS). Recently, the Molecular International Prognostic Scoring System (IPSS-M) has been developed. Here, we validated the accuracy of IPSS-M in Chinese MDS patients, and proposed a prognostic model more suitable for Chinese patients. We analyzed the clinical, molecular and cytogenetic data of 798 primary MDS patients, and compared the accuracy of IPSS-R and IPSS-M in predicting overall survival (OS) and acute myeloid leukemia (AML) transformation. Using Cox proportional hazards model, we screened out 14 genes that had significant impacts on OS and AML progression. In our study, 44.86% of individuals were reclassified from IPSS-R to IPSS-M, of whom 64.80% were upstaged and 35.2% were downstaged. IPSS-M showed better performance than IPSS-R in predicting AML transformation (C-index: 0.84 vs. 0.81), but it was similar to IPSS-R in OS (C-index: 0.77 vs. 0.76). By combining age, mutational data and IPSS-R, we developed a new prognostic model more suitable for the Chinese patients (c-index was 0.81 for OS and 0.89 for AML transformation, respectively).
Collapse
Affiliation(s)
- Nanfang Huang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yang Song
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Lingyun Wu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi He
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juan Guo
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Xu
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chunkang Chang
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao Li
- Department of Hematology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
2
|
Wang J, Li S, Jiang H, Chang YJ, Zhao X, Jia J, Zhu X, Gong L, Liu X, Yu W, Huang X. Sintilimab plus decitabine for higher-risk treatment-naïve myelodysplastic syndromes: efficacy, safety, and biomarker analysis of a phase II, single-arm trial. J Immunother Cancer 2024; 12:e010355. [PMID: 39577869 PMCID: PMC11590843 DOI: 10.1136/jitc-2024-010355] [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: 08/14/2024] [Accepted: 10/29/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Immunotherapy combined with azacitidine was feasible in higher-risk myelodysplastic syndromes (MDSs) with limited sample size of treatment-naïve patients, while the optimization of treatment strategies, including the optimal immune checkpoint inhibitor and hypomethylating agent and possible benefiting population, remained undefined. This study first evaluates the efficacy and safety of sintilimab, a PD-1 blockade, plus decitabine in treatment-naïve higher-risk MDS patients and investigates biomarkers for predicting treatment response. METHODS In this phase II, single-arm trial (ChiCTR2100044393), treatment-naïve higher-risk MDS patients with an International Prognostic Scoring System-Revised score >3.5 received sintilimab (200 mg, days 1 and 22) and decitabine (20 mg/m2, day 1-5) over 6-week cycles. The primary endpoint was the overall response rate (ORR), including complete remission (CR), partial remission (PR) or marrow CR. RESULTS A total of 54 eligible patients were enrolled and treated, with 25 (46.3%) having very high-risk MDS. Among 53 evaluable patients, the ORR was 77.4% (n=41), including 26.4% CR (n=14). The overall clinical improvement rate (CR, PR, marrow CR or hematological improvement) reached 81.1%. With a median follow-up of 20.0 months, the median event-free survival was 23 months with 12 progressing to acute myeloid leukemia. Median overall survival was not reached. Treatment was generally well tolerated, with hematologic toxicities being the most common adverse events. Biomarker analysis highlighted a negative correlation between T cell exhaustion markers, particularly TIM-3 and PD-1, with ORR. CONCLUSIONS The combination of sintilimab and decitabine shows promise efficacy for higher-risk MDS, with a favorable safety profile. The potential predictive value of T cell exhaustion biomarkers might help screen the possible benefiting population. TRIAL REGISTRATION NUMBER ChiCTR210044393.
Collapse
Affiliation(s)
- Jing Wang
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Siqi Li
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Hao Jiang
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Ying-Jun Chang
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Xiaosu Zhao
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Jinsong Jia
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Xiaolu Zhu
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Lizhong Gong
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Xiaohong Liu
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Wenjing Yu
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
| | - Xiaojun Huang
- Peking University Institute of Hematology. National Clinical Research Center for Hematologic Disease, Beijing Key Laboratory of Hematopoietic Stem Cell Transplantation, Peking University People’s Hospital, Beijing, China
- Research Unit of Key Technique for Diagnosis and Treatments of Hematologic Malignancies, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
3
|
Kannan S, Vedia RA, Molldrem JJ. The immunobiology of myelodysplastic neoplasms: a mini-review. Front Immunol 2024; 15:1419807. [PMID: 39355256 PMCID: PMC11443505 DOI: 10.3389/fimmu.2024.1419807] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 08/27/2024] [Indexed: 10/03/2024] Open
Abstract
This mini review summarizes the immunobiology of myelodysplastic syndromes, specifically focusing on the interactions between immune cells, cytokines, and dysplastic cells within the tumor microenvironment in the bone marrow. We elucidate in detail how immune dysregulation and evasion influence the initiation and progression of myelodysplastic syndromes, as well as resistance to therapy and progression to AML. In addition, we highlight a range of therapeutic strategies, including the most recent breakthroughs and experimental therapies for treating MDS. Finally, we address the existing knowledge gaps in the understanding of the immunobiology of MDS and propose future research directions, promising advancements toward enhancing clinical outcomes and survival for patients with MDS.
Collapse
Affiliation(s)
- Shruthi Kannan
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Rolando A Vedia
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Jeffrey J Molldrem
- Department of Hematopoietic Biology and Malignancy, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Evolution of Cancer, Leukemia, and Immunity Post Stem cEll transplant (ECLIPSE), Therapeutics Discovery Division, The University of Texas MD Anderson Cancer Center, Houston, TX, United States
- Department of Stem Cell Transplantation and Cellular Therapy, Division of Cancer Medicine, UT MD Anderson Cancer Center, Houston, TX, United States
| |
Collapse
|
4
|
Adamska M, Kowal-Wiśniewska E, Czerwińska-Rybak J, Kiwerska K, Barańska M, Gronowska W, Loba J, Brzeźniakiewicz-Janus K, Wasilewska E, Łanocha A, Jarmuż-Szymczak M, Gil L. Defining the mutational profile of lower-risk myelodysplastic neoplasm patients with respect to disease progression using next-generation sequencing and pyrosequencing. Contemp Oncol (Pozn) 2024; 27:269-279. [PMID: 38405213 PMCID: PMC10883195 DOI: 10.5114/wo.2023.135365] [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: 11/29/2023] [Accepted: 01/18/2024] [Indexed: 02/27/2024] Open
Abstract
Introduction Lower-risk myelodysplastic neoplasms (LR-MDS) comprise the majority of MDS. Despite favourable prognoses, some patients remain at risk of rapid progression. We aimed to define the mutational profile of LR-MDS using next-generation sequencing (NGS), Sanger Sequencing (SSeq), and pyrosequencing. Material and methods Samples from 5 primary LR-MDS (67 exons of SF3B1, U2AF1, SRSF2, ZRSR2, TET2, ASXL1, DNMT3A, TP53, and RUNX1 genes) were subjected to NGS. Next, a genomic study was performed to test for the presence of identified DNA sequence variants on a larger group of LR-MDS patients (25 bone marrow [BM], 3 saliva [SAL], and one peripheral blood [PB] sample/s). Both SSeq (all selected DNA sequence variants) and pyrosequencing (9 selected DNA sequence variants) were performed. Results Next-generation sequencing results identified 13 DNA sequence variants in 7 genes, comprising 8 mutations in 6 genes (ASXL1, DNMT3A, RUNX1, SF3B1, TET2, ZRSR2) in LR-MDS. The presence of 8 DNA variants was detected in the expanded LR-MDS group using SSeq and pyrosequencing. Mutation acquisition was observed during LR-MDS progression. Four LR-MDS and one acute myeloid leukaemia myelodysplasia-related patient exhibited the presence of at least one mutation. ASXL1 and SF3B1 alterations were most commonly observed (2 patients). Five DNA sequence variants detected in BM (patients: 9, 13) were also present in SAL. Conclusions We suggest using NGS to determine the LR-MDS mutational profile at diagnosis and suspicion of disease progression. Moreover, PB and SAL molecular testing represent useful tools for monitoring LR-MDS at higher risk of progression. However, the results need to be confirmed in a larger group.
Collapse
Affiliation(s)
- Monika Adamska
- Department of Haematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
- Doctoral School, Poznań University of Medical Sciences, Poznań, Poland
| | - Ewelina Kowal-Wiśniewska
- Department of Haematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
- Institute of Human Genetics, Polish Academy of Sciences, Poznań, Poland
| | - Joanna Czerwińska-Rybak
- Department of Haematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
| | | | - Marta Barańska
- Doctoral School, Poznań University of Medical Sciences, Poznań, Poland
| | - Weronika Gronowska
- Student Scientific Society, Poznań University of Medical Sciences, Poznań, Poland
| | - Jagoda Loba
- Student Scientific Society, Poznań University of Medical Sciences, Poznań, Poland
| | - Katarzyna Brzeźniakiewicz-Janus
- Department of Haematology, Oncology, and Radiotherapy, University of Zielona Góra, Multi-specialist Hospital Gorzów Wielkopolski, Poland
| | - Ewa Wasilewska
- Department of Haematology, Medical University of Białystok, Białystok, Poland
| | - Aleksandra Łanocha
- Department of Haematology with Bone Marrow Transplantation Unit, University Hospital No. 1 of Pomeranian Medical University, Szczecin, Poland
| | - Małgorzata Jarmuż-Szymczak
- Department of Haematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
- Institute of Human Genetics, Polish Academy of Sciences, Poznań, Poland
| | - Lidia Gil
- Department of Haematology and Bone Marrow Transplantation, Poznań University of Medical Sciences, Poznań, Poland
| |
Collapse
|
5
|
Ning Y, Zhang Y, Kallen MA, Emadi A, Baer MR. Cytogenetics and molecular genetics of myelodysplastic neoplasms. Best Pract Res Clin Haematol 2023; 36:101512. [PMID: 38092472 DOI: 10.1016/j.beha.2023.101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 07/15/2023] [Accepted: 08/01/2023] [Indexed: 12/18/2023]
Abstract
According to the 2022 World Health Organization (WHO) Classification (5th edition), the term myelodysplastic neoplasms (abbreviated MDS) has been introduced to replace myelodysplastic syndromes. MDS are a group of clonal hematopoietic stem cell diseases characterized by cytopenia(s), dysplasia in one or more of lineages, ineffective hematopoiesis, and an increased risk of progression to bone marrow failure or to acute myeloid leukemia (AML). Current NCCN guidelines and recent review articles have provided in depth discussion on the clinical diagnosis and management of MDS. This review will focus on discussion of the WHO and International Consensus Classification (ICC) updates on the role of cytogenetics and molecular genetics in the diagnosis and risk stratification of MDS.
Collapse
Affiliation(s)
- Yi Ning
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Yanming Zhang
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Michael A Kallen
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD, USA.
| | - Ashkan Emadi
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.
| | - Maria R Baer
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA; University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA.
| |
Collapse
|
6
|
Block TJ, Shore‐Lorenti C, Zebaze R, Kerr PG, Kalff A, Perkins AC, Ebeling PR, Milat F. A Novel RUNX1 Genetic Variant Identified in a Young Male with Severe Osteoporosis. JBMR Plus 2023; 7:e10791. [PMID: 37701147 PMCID: PMC10494497 DOI: 10.1002/jbm4.10791] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 06/01/2023] [Accepted: 06/10/2023] [Indexed: 09/14/2023] Open
Abstract
This case describes a young man with an unusual cause of severe osteoporosis and markedly deranged bone microarchitecture resulting in multiple fractures. A potentially pathogenic germline variant in the runt-related transcription factor 1 (RUNX1) gene was discovered by a focused 51-gene myeloid malignancy panel during investigation for his unexplained normochromic normocytic anemia. Further bone-specific genetic testing and a pedigree analysis were declined by the patient. Recent experimental evidence demonstrates that RUNX1 plays a key role in the regulation of osteogenesis and bone homeostasis during skeletal development, mediated by the bone morphogenic protein and Wnt signaling pathways. Therefore, rarer causes of osteoporosis, including those affecting bone formation, should be considered in young patients with multiple unexpected minimal trauma fractures. © 2023 The Authors. JBMR Plus published by Wiley Periodicals LLC on behalf of American Society for Bone and Mineral Research.
Collapse
Affiliation(s)
- Tomasz J. Block
- Department of EndocrinologyMonash HealthMelbourneVictoriaAustralia
- Department of Diabetes, Central Clinical SchoolMonash UniversityMelbourneVictoriaAustralia
| | - Cat Shore‐Lorenti
- Centre for Endocrinology and MetabolismHudson Institute of Medical ResearchClaytonVictoriaAustralia
| | - Roger Zebaze
- Department of Medicine, School of Clinical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Peter G. Kerr
- Department of NephrologyMonash HealthMelbourneVictoriaAustralia
| | - Anna Kalff
- Department of HaematologyAlfred HealthMelbourneVictoriaAustralia
| | | | - Peter R. Ebeling
- Department of EndocrinologyMonash HealthMelbourneVictoriaAustralia
- Department of Medicine, School of Clinical SciencesMonash UniversityMelbourneVictoriaAustralia
| | - Frances Milat
- Department of EndocrinologyMonash HealthMelbourneVictoriaAustralia
- Centre for Endocrinology and MetabolismHudson Institute of Medical ResearchClaytonVictoriaAustralia
- Department of Medicine, School of Clinical SciencesMonash UniversityMelbourneVictoriaAustralia
| |
Collapse
|
7
|
Krishnan V. The RUNX Family of Proteins, DNA Repair, and Cancer. Cells 2023; 12:cells12081106. [PMID: 37190015 DOI: 10.3390/cells12081106] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 04/04/2023] [Accepted: 04/05/2023] [Indexed: 05/17/2023] Open
Abstract
The RUNX family of transcription factors, including RUNX1, RUNX2, and RUNX3, are key regulators of development and can function as either tumor suppressors or oncogenes in cancer. Emerging evidence suggests that the dysregulation of RUNX genes can promote genomic instability in both leukemia and solid cancers by impairing DNA repair mechanisms. RUNX proteins control the cellular response to DNA damage by regulating the p53, Fanconi anemia, and oxidative stress repair pathways through transcriptional or non-transcriptional mechanisms. This review highlights the importance of RUNX-dependent DNA repair regulation in human cancers.
Collapse
Affiliation(s)
- Vaidehi Krishnan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore 117599, Singapore
| |
Collapse
|
8
|
Huber S, Haferlach T, Müller H, Meggendorfer M, Hutter S, Hoermann G, Baer C, Kern W, Haferlach C. MDS subclassification-do we still have to count blasts? Leukemia 2023; 37:942-945. [PMID: 36813994 PMCID: PMC10079547 DOI: 10.1038/s41375-023-01855-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 02/10/2023] [Accepted: 02/14/2023] [Indexed: 02/24/2023]
Affiliation(s)
- Sandra Huber
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Torsten Haferlach
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Heiko Müller
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Manja Meggendorfer
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Stephan Hutter
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Gregor Hoermann
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Constance Baer
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Wolfgang Kern
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany
| | - Claudia Haferlach
- MLL Munich Leukemia Laboratory, Max-Lebsche-Platz 31, 81377, Munich, Germany.
| |
Collapse
|
9
|
Zavras PD, Sinanidis I, Tsakiroglou P, Karantanos T. Understanding the Continuum between High-Risk Myelodysplastic Syndrome and Acute Myeloid Leukemia. Int J Mol Sci 2023; 24:5018. [PMID: 36902450 PMCID: PMC10002503 DOI: 10.3390/ijms24055018] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/01/2023] [Accepted: 03/03/2023] [Indexed: 03/08/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a clonal hematopoietic neoplasm characterized by bone marrow dysplasia, failure of hematopoiesis and variable risk of progression to acute myeloid leukemia (AML). Recent large-scale studies have demonstrated that distinct molecular abnormalities detected at earlier stages of MDS alter disease biology and predict progression to AML. Consistently, various studies analyzing these diseases at the single-cell level have identified specific patterns of progression strongly associated with genomic alterations. These pre-clinical results have solidified the conclusion that high-risk MDS and AML arising from MDS or AML with MDS-related changes (AML-MRC) represent a continuum of the same disease. AML-MRC is distinguished from de novo AML by the presence of certain chromosomal abnormalities, such as deletion of 5q, 7/7q, 20q and complex karyotype and somatic mutations, which are also present in MDS and carry crucial prognostic implications. Recent changes in the classification and prognostication of MDS and AML by the International Consensus Classification (ICC) and the World Health Organization (WHO) reflect these advances. Finally, a better understanding of the biology of high-risk MDS and the mechanisms of disease progression have led to the introduction of novel therapeutic approaches, such as the addition of venetoclax to hypomethylating agents and, more recently, triplet therapies and agents targeting specific mutations, including FLT3 and IDH1/2. In this review, we analyze the pre-clinical data supporting that high-risk MDS and AML-MRC share the same genetic abnormalities and represent a continuum, describe the recent changes in the classification of these neoplasms and summarize the advances in the management of patients with these neoplasms.
Collapse
Affiliation(s)
| | | | | | - Theodoros Karantanos
- Division of Hematologic Malignancies and Bone Marrow Transplantation, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21231, USA
| |
Collapse
|