1
|
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
|
2
|
Gutmann DH, Boehm JS, Karlsson EK, Padron E, Seshadri M, Wallis D, Snyder JC. Precision preclinical modeling to advance cancer treatment. J Natl Cancer Inst 2025; 117:586-594. [PMID: 39383197 PMCID: PMC11972679 DOI: 10.1093/jnci/djae249] [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: 06/25/2024] [Revised: 09/17/2024] [Accepted: 10/02/2024] [Indexed: 10/11/2024] Open
Abstract
A new era of cancer management is underway in which treatments are being developed for the entire continuum of the disease process. The availability of genetically engineered and naturally occurring preclinical models serves as instructive platforms for evaluating therapeutic mechanisms. However, a major clinical challenge is that the entire malignancy process occurs across multiple scales including genetic mutations, malignant changes in cell behavior, dysregulated tumor microenvironments, and systemic adaptations in the host. A multidisciplinary group of investigators coalesced at the National Cancer Institute Oncology Models Forum with the overall goal to provide updates on the use of precision preclinical models of cancer. The benefits and limitations of preclinical models were discussed to identify strategies for maximizing opportunities in modeling that could inform future cancer prevention and treatment approaches. Our shared perspective is that the continuum of single cell, multicell, organoid, and in situ models are remarkable resources for the clinical challenges ahead. We provide a roadmap for parsing already available models and include preliminary recommendations for the application of next-generation preclinical modeling in cancer intervention.
Collapse
Affiliation(s)
- David H Gutmann
- Department of Neurology, Washington University, St Louis, MO 63110, United States
| | - Jesse S Boehm
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
- Koch Institute, Massachusetts Institute of Technology, Cambridge, MA 02139, United States
| | - Elinor K Karlsson
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, United States
- Genomics and Computational Biology, University of Massachusetts Chan Medical School, Worcester, MA 01655, United States
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, United States
| | - Mukund Seshadri
- Department of Oral Oncology, Roswell Park Comprehensive Cancer Center, Buffalo, NY 14263, United States
| | - Deeann Wallis
- Department of Genetics, University of Alabama at Birmingham, Birmingham, AL 35294, United States
| | - Joshua C Snyder
- Department of Surgery, Duke University, Durham, NC 27710, United States
| |
Collapse
|
3
|
Thomas X. Examining the safety and efficacy of imetelstat in low-risk myelodysplastic syndrome. Expert Opin Pharmacother 2025; 26:525-533. [PMID: 39989126 DOI: 10.1080/14656566.2025.2471518] [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: 01/13/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION The aim of treatment in very low-, low- and intermediate-1-risk myelodysplastic syndrome (MDS) is mainly to relieve symptoms due to cytopenias. Only a few therapeutic drugs are currently available, but novel drugs are under clinical investigations. In this setting, imetelstat, a telomerase inhibitor, is a promising new agent. AREAS COVERED This review summarizes promising emerging strategies using imetelstat for the treatment of lower-risk MDS. EXPERT OPINION Favorable results were demonstrated in the IMerge phase 3 clinical trial using imetelstat in transfusion-dependent patients with lower-risk MDS relapsed or refractory to erythropoiesis-stimulating agents (ESAs). This study led to imetelstat approval by the United States Food and Drug Administration (FDA) in June 2024.
Collapse
Affiliation(s)
- Xavier Thomas
- Hospices Civils de Lyon, Department of Clinical Hematology, Centre Hospitalier Lyon-Sud, Pierre Bénite, France
| |
Collapse
|
4
|
Poloni A, Raaschou-Jensen K, Mohedo FH, Paolini S, Oliva EN, Buccisano F, Vasconcelos A, Kim I, Makwana A, Bernasconi D, Rosettani B, Prebet T, Santini V. Lenalidomide in Transfusion-Dependent IPSS Low- or Intermediate-1-Risk Myelodysplastic Syndromes and Isolated Del(5q): Results of a European Postauthorization Safety Surveillance Study. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025; 25:e131-e142. [PMID: 39516085 DOI: 10.1016/j.clml.2024.10.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 10/02/2024] [Accepted: 10/02/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND This noninterventional postauthorization safety study assessed the safety and effectiveness of lenalidomide in patients with transfusion-dependent, International Prognostic Scoring System (IPSS) Low- or Intermediate (Int)-1-risk myelodysplastic syndromes (MDS) associated with isolated deletion of 5q (del[5q]) who were treated in routine care. PATIENTS AND METHODS Eligible adult patients in the lenalidomide cohort had transfusion-dependent, IPSS Low- or Int-1-risk MDS and isolated del(5q) and had received ≥ 1 dose of lenalidomide between 2014 and 2022. The primary endpoint was the 24-month cumulative incidence of acute myeloid leukemia (AML) progression. Overall survival (OS) was estimated by Kaplan-Meier analysis and safety data were collected. RESULTS In total, 296 patients received ≥ 1 dose of lenalidomide (lenalidomide cohort, safety population) and 277 had received ≥ 1 complete cycle of lenalidomide (primary population). In the safety population, 44.3% of patients completed 3-year follow-up and 55.1% discontinued, with 33.1% discontinuing due to death. In the primary population, 24-month cumulative incidence of AML progression was 12.7% (95% confidence interval, 8.9%-17.1%) and estimated OS probability was 78.3% at 24 months and 63.9% at 36 months. Grade 3/4 treatment-emergent adverse events were experienced by 67.2% of the safety population, and these led to discontinuation in 35.5% of patients. There were no new safety signals. CONCLUSION These real-world data support the established benefit-risk profile of lenalidomide in transfusion-dependent IPSS Low- or Int-1-risk MDS with isolated del(5q).
Collapse
Affiliation(s)
- Antonella Poloni
- Hematology Clinic, Azienda Ospedaliera Universitaria delle Marche, Università Politecnica delle Marche, Ancona, Italy.
| | | | - Francisca Hernandez Mohedo
- Instituto de Investigación Biosanitaria de Granada (ibs.GRANADA); Hematology Department, Virgen de las Nieves University Hospital, Granada, Spain
| | - Stefania Paolini
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli," Bologna, Italy
| | | | - Francesco Buccisano
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | | | - Iris Kim
- Bristol Myers Squibb, Princeton, NJ
| | | | | | | | | | - Valeria Santini
- MDS Unit, AOU Careggi, MD, PhD, University of Florence, Florence, Italy
| |
Collapse
|
5
|
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
|
6
|
Getz TM, Bewersdorf JP, Kewan T, Stempel JM, Bidikian A, Shallis RM, Stahl M, Zeidan AM. Beyond HMAs: Novel Targets and Therapeutic Approaches. Semin Hematol 2024; 61:358-369. [PMID: 39389839 DOI: 10.1053/j.seminhematol.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024]
Abstract
Myelodysplastic syndromes/neoplasms (MDS) constitute a heterogeneous group of clonal hematopoietic disorders with extremely variable clinical features and outcomes. Management of MDS is largely based on risk stratification of patients into either lower-risk or higher-risk categories using the International Prognostic Scoring System-Revised and, more recently, on the Molecular International Prognostic Scoring System. Lower-risk MDS is often managed with the goal of ameliorating cytopenias and improving quality of life, while higher-risk MDS is treated with therapies aimed at extending survival and delaying progression to acute myeloid leukemia (AML). Therapeutic strategies in lower-risk MDS patients may consist of erythropoiesis stimulating agents, luspatercept, and lenalidomide for selected patients. Furthermore, imetelstat has recently been added to the FDA-approved therapeutic armamentarium for lower-risk MDS. In higher-risk MDS, monotherapy with hypomethylating agents continues to be the standard of care. While several novel hypomethylating agent combinations have and are being studied in large randomized phase 3 clinical trials, including the combination of azacitidine and venetoclax, no combination to date have improved overall survival to azacitidine monotherapy. Moreover, biomarker-directed therapies as well as immonotherapeutic approaches are currently being evaluated in early phase trials. Despite recent advancements, the lack of therapeutic agents, particularly after the failure of first line therapy in higher risk MDS, continues to be a major hurdle in the management of MDS. In this review, we discuss the current treatment landscape of MDS and provide an overview of novel agents currently in clinical development that have the potential to alter our current treatment paradigms.
Collapse
Affiliation(s)
- Ted M Getz
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut.
| | - Jan P Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut; Department of Medicine, Memorial Sloan Kettering Cancer Center, Leukemia Service, New York, New York
| | - Tariq Kewan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Jessica M Stempel
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Aram Bidikian
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Rory M Shallis
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| |
Collapse
|
7
|
McGraw KL, Aplan PD, Pavletic SZ. A Call to Arms: Overcoming challenges in Myelodysplastic Syndromes therapy advances. Semin Hematol 2024; 61:345-347. [PMID: 39674608 DOI: 10.1053/j.seminhematol.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 11/27/2024] [Indexed: 12/16/2024]
Affiliation(s)
- Kathy L McGraw
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD; Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD; Myeloid Malignancies Program, National Institute of Health, Bethesda, MD.
| | - Peter D Aplan
- Myeloid Malignancies Program, National Institute of Health, Bethesda, MD; Genetics Branch, National Cancer Institute, National Institutes of Health, Bethesda, MD.
| | - Steven Z Pavletic
- Immune Deficiency Cellular Therapy Program, National Cancer Institute, National Institutes of Health, Bethesda, MD; Myeloid Malignancies Program, National Institute of Health, Bethesda, MD.
| |
Collapse
|
8
|
Kunimoto H, Sakuma T, Ohashi T, Shirafuta M, Teranaka H, Nakajima H. Successful treatment of low-risk myelodysplastic syndrome-related anemia in patients with chronic kidney disease with daprodustat: A report of two cases. EJHAEM 2024; 5:1335-1339. [PMID: 39691241 PMCID: PMC11647689 DOI: 10.1002/jha2.1057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/07/2024] [Accepted: 11/08/2024] [Indexed: 12/19/2024]
Abstract
Anemia is a major clinical manifestation seen in myelodysplastic syndromes (MDS). Treatment options for anemia in low-risk MDS are limited. Especially, oral medication which is uniformly effective for anemia in low-risk MDS is required. Hypoxia-inducible factor (HIF) prolyl hydroxylase (HIF-PH) inhibitors, such as daprodustat, are oral tablets effective for renal anemia. Pharmacological restoration of HIF activity by HIF-PH inhibitors may be beneficial for MDS-related anemia as well. Yet, their efficacy and safety against low-risk MDS are unclear. Here, we report two cases of low-risk MDS complicated with chronic kidney disease whose anemia responded to daprodustat treatment.
Collapse
Affiliation(s)
- Hiroyoshi Kunimoto
- Department of Stem Cell and Immune RegulationYokohama City University Graduate School of MedicineYokohamaJapan
| | - Takayuki Sakuma
- Department of Stem Cell and Immune RegulationYokohama City University Graduate School of MedicineYokohamaJapan
| | - Takuma Ohashi
- Department of Stem Cell and Immune RegulationYokohama City University Graduate School of MedicineYokohamaJapan
| | - Mayoko Shirafuta
- Department of Stem Cell and Immune RegulationYokohama City University Graduate School of MedicineYokohamaJapan
| | - Hiroshi Teranaka
- Department of Stem Cell and Immune RegulationYokohama City University Graduate School of MedicineYokohamaJapan
| | - Hideaki Nakajima
- Department of Stem Cell and Immune RegulationYokohama City University Graduate School of MedicineYokohamaJapan
| |
Collapse
|
9
|
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
|
10
|
Ball B, Xiao W, Borthakur G, Nguyen LXT, Valerio M, Venkatachalam A, Marcucci G, Stein A, Thai DL, Cook D, Chan K, Persaud S, Levine R, Abdel-Wahab O, Ben-Neriah Y, Stein E. Phase I First-in-Human Dose Escalation Study of the oral Casein Kinase 1α and Cyclin Dependent Kinase 7/9 inhibitor BTX-A51 in advanced MDS and AML. RESEARCH SQUARE 2024:rs.3.rs-4954060. [PMID: 39483885 PMCID: PMC11527261 DOI: 10.21203/rs.3.rs-4954060/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
BTX-A51, a first-in-class oral small molecule inhibitor of casein kinase 1α (CK1α) and cyclin dependent kinase (CDK) 7 and 9, induces apoptosis of leukemic cells by activating p53 and inhibiting expression of Mcl1. Here, we report on the results of the phase 1 clinical trial of BTX-A51 in patients with relapsed or refractory AML and MDS. Thirty-one patients were enrolled into 8 dose-escalation cohorts at BTX-A51 doses ranging from 1mg to 42mg dosed three days/week for 21 or 28 days out a 28-day cycle. The recommended phase 2 dose was 21mg dosed three days/week for 4 weeks of a 28-day cycle. BTX-A51 increased expression of p53 and reduced expression of MCL1 and RNA polymerase II phosphorylation on pre- and post-treatment immunocytochemistry studies. Overall, 3 patients (10%) experienced complete remission with incomplete count recovery (CRi). All 3 responding patients had RUNX1 mutations and the CR/CRi rate for RUNX1-mutated patients receiving BTX-A51 at efficacious doses (11mg or higher) was 30%. Ex-vivo studies confirmed higher efficacy of BTX-A51 on RUNX1-mutated myeloblasts and demonstrate synergy with azacitidine and venetoclax. Although the overall efficacy was modest, this study lays the groundwork for future studies with improved patient selection and combination approaches.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | - Yinon Ben-Neriah
- The Lautenberg Center for Immunology and Cancer Research, Institute of Medical Research Israel-Canada
| | | |
Collapse
|
11
|
Crisà E, Mora E, Germing U, Bally C, Diez Campelo M, Myllymäki M, Jädersten M, Komrokji R, Platzbecker U, Haase D, Hofmann WK, Al Ali NH, Barraco D, Bargay JJ, Bernal T, López Cadenas F, Calvisi A, Capodanno I, Cerrano M, Ciancia R, Crugnola M, Kündgen A, Finelli C, Fozza C, Frairia C, Freja E, Ganster C, Kubasch AS, Jimenez MJ, Latagliata R, Hernandez Mohedo F, Molero A, Vara Pampliega M, Perez CA, Pietrantuono G, Poloni A, Pomares H, Recasens V, Rüfer A, Signori A, Hellstrom-Lindberg E, Fenaux P, Sanz G, Santini V. Transfusion independence after lenalidomide discontinuation in patients with del(5q) myelodysplastic neoplasm: a HARMONY Alliance study. Leukemia 2024; 38:2259-2265. [PMID: 39103678 DOI: 10.1038/s41375-024-02360-1] [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: 04/12/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 08/07/2024]
Abstract
Lenalidomide (LEN) can induce red blood cell-transfusion independence (RBC-TI) in 60-70% of del(5q) myelodysplastic neoplasm (MDS) patients. Current recommendation is to continue LEN in responding patients until failure or progression, with likelihood of toxicity and a high cost for healthcare systems. This HARMONY Alliance study investigated the outcome of MDS del(5q) patients who discontinued LEN while RBC-transfusion independent. We enrolled 118 patients with IPSS-R low-intermediate risk. Seventy patients (59%) discontinued LEN for intolerance, 38 (32%) per their physician decision, nine (8%) per their own decision and one (1%) for unknown reasons. After a median follow-up of 49 months from discontinuation, 50/118 patients lost RBC-TI and 22/30 who underwent cytogenetic re-evaluation lost complete cytogenetic response. The median RBC-TI duration was 56 months. In multivariate analysis, RBC-TI duration after LEN discontinuation correlated with low transfusion burden before LEN therapy, treatment ≥ 12 LEN cycles, younger age and higher Hb level at LEN withdrawal. Forty-eight patients were re-treated with LEN for loss of response and 28 achieved again RBC-TI. These data show that stopping LEN therapy in MDS del(5q) patients who reached RBC-TI allows prolonged maintenance of TI in a large subset of patients.
Collapse
Affiliation(s)
- Elena Crisà
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Italy.
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy.
| | - Elvira Mora
- Hospital Universitario y Politécnico La Fe, Instituto de Investigación Sanitaria La Fe, Valencia, Spain
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Universitätsklinik Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Cecile Bally
- Hématologie clinique, hôpital Necker. Assistance publique hôpitaux de Paris (APHP), Paris, France
| | - Maria Diez Campelo
- Hematology, Institute for Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
| | - Mikko Myllymäki
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Martin Jädersten
- Department of Hematology M64 Karolinska University Hospital, Stockholm, Sweden
| | - Rami Komrokji
- Malignant Hematology Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy,Hemostaseology and Infectiology, University Hospital Leipzig, Leipzig, Germany
| | - Detlef Haase
- Clinics of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, University Hospital Mannheim, Heidelberg University, Heidelberg, Germany
| | - Najla H Al Ali
- Malignant Hematology Department, Moffitt Cancer Center, Tampa, FL, USA
| | - Daniela Barraco
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- Dipartimento di Ematologia - ASST Sette Laghi, Ospedale di Circolo e Fondazione Macchi, Varese, Italy
| | | | - Teresa Bernal
- Hematology Department. Hospital Central de Asturias, Oviedo, Spain
| | - Felix López Cadenas
- Hematology Department. Hospital Clinico Universitario de Salamanca, Salamanca, Spain
| | - Anna Calvisi
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- UOC Ematologia CTMO, Osp San Francesco Nuoro, Nuoro, Italy
| | - Isabella Capodanno
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- SOC Ematologia Azienda USL-IRCSS di Reggio Emilia, Reggio Emilia, Italy
| | - Marco Cerrano
- Hematology-University Hospital Città della Salute e della Scienza, Turin, Italy
- Hematology- Hospital Città della Salute e della Scienza, Turin, Italy
| | - Rosanna Ciancia
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- IRCCS Centro di Riferimento Oncologico (CRO) di Aviano (Pordenone), Aviano, Italy
| | - Monica Crugnola
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- Hematology Unit and BMT center University Hospital of Parma, Parma, Italy
| | - Andrea Kündgen
- Department of Hematology, Oncology and Clinical Immunology, Universitätsklinik Düsseldorf, Heinrich-Heine-University Düsseldorf, Düsseldorf, Germany
| | - Carlo Finelli
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Institute of Hematology "Seràgnoli", Bologna, Italy
| | - Claudio Fozza
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- Department of Clinical and Experimental Medicine, University of Sassari, Sassari, Italy
| | - Chiara Frairia
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- Hematology- Hospital Città della Salute e della Scienza, Turin, Italy
| | - Ebeling Freja
- Department of Medicine, Helsinki University Central Hospital, Helsinki, Finland
| | - Christina Ganster
- Clinics of Hematology and Medical Oncology, University Medical Center Göttingen, Göttingen, Germany
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy,Hemostaseology and Infectiology, University Hospital Leipzig, Leipzig, Germany
| | | | - Roberto Latagliata
- Unità Operativa Complessa (UOC) Ematologia, Ospedale Belcolle, Viterbo and Division of Cellular Biotechnology and Hematology, Sapienza University, Rome, Italy
| | | | - Antonieta Molero
- Hematology Department. Hospital Universitario Vall D'Hebron, Barcelona, Spain
| | | | | | - Giuseppe Pietrantuono
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- IRCCS CROB, Referral Cancer Center of Basilicata, Rionero In Vulture, Italy
| | - Antonella Poloni
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy
- Hematology, Università Politecnica Marche - AOU Marche, Ancona, Italy
| | - Helena Pomares
- Hematology Department. ICO Duran i Reynals, L'Hospitalet de LLobregat, LLobregat, Spain
| | - Valle Recasens
- Valle Recasens. Hematology Department. Hospital Miguel Servent, Zaragoza, Spain
| | - Axel Rüfer
- Division of Hematology, Luzerner Kantonsspital-in association with University Luzern, Luzern, Switzerland
| | - Alessio Signori
- Department of Health Sciences - Section of Biostatistics University of Genoa, Genova, Italy
| | - Eva Hellstrom-Lindberg
- Hematology Research Unit Helsinki, University of Helsinki and Helsinki University Hospital Comprehensive Cancer Center, Helsinki, Finland
| | - Pierre Fenaux
- Service d'Hématologie Séniors, Hôpital Saint-Louis, Assistance Publique-Hôpitaux de Paris, Paris, France
| | - Guillermo Sanz
- Hematology, Institute for Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
- CIBERONC, ISCIII, Madrid, Spain
| | - Valeria Santini
- Fondazione Sindromi Mielodisplastiche FISiM-ETS, Bologna, Italy.
- MDS Unit, Hematology, DMSC, AOU Careggi, University of Florence, Florence, Italy.
| |
Collapse
|
12
|
Patsialos I, Kontandreopoulou CN, Vlachopoulou D, Stafylidis C, Syriopoulou S, Kalala F, Anastasopoulou A, Mantzourani M, Diamantopoulos P. A myelodysplastic neoplasm with del(5q) treated with luspatercept uncovers unexplored mechanisms of action for the drug. Br J Haematol 2024; 205:1641-1644. [PMID: 39155048 DOI: 10.1111/bjh.19708] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 08/07/2024] [Indexed: 08/20/2024]
Affiliation(s)
- Iraklis Patsialos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christina-Nefeli Kontandreopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Dimitra Vlachopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Christos Stafylidis
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Stavroula Syriopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Fani Kalala
- Cancer Immunology Unit, Department of Immunology and Histocompatibility, School of Medicine, University of Thessaly, University Hospital of Larissa, Larissa, Greece
| | - Amalia Anastasopoulou
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Marina Mantzourani
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | - Panagiotis Diamantopoulos
- Hematology Unit, First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
13
|
Buckstein R. SintraREViewed: practice changing, or validation required? Lancet Haematol 2024; 11:e632-e634. [PMID: 39033768 DOI: 10.1016/s2352-3026(24)00185-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/06/2024] [Accepted: 06/07/2024] [Indexed: 07/23/2024]
Affiliation(s)
- Rena Buckstein
- Odette Cancer Center, Sunnybrook Health Sciences, Toronto, ON M4N3M5, Canada.
| |
Collapse
|
14
|
Díez-Campelo M, López-Cadenas F, Xicoy B, Lumbreras E, González T, Del Rey González M, Sánchez-García J, Coll Jordà R, Slama B, Hernández-Rivas JÁ, Thepot S, Bernal T, Guerci-Bresler A, Bargay J, Amigo ML, Preudhomme C, Fenwarth L, Platzbecker U, Götze KS, Arar A, Toribio S, Del Cañizo C, Hernández-Rivas JM, Fenaux P. Low dose lenalidomide versus placebo in non-transfusion dependent patients with low risk, del(5q) myelodysplastic syndromes (SintraREV): a randomised, double-blind, phase 3 trial. Lancet Haematol 2024; 11:e659-e670. [PMID: 39033767 DOI: 10.1016/s2352-3026(24)00142-x] [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: 03/16/2024] [Revised: 05/02/2024] [Accepted: 05/05/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Lenalidomide is the standard of care for patients who are transfusion dependent with chromosome 5q deletion (del[5q]) myelodysplastic syndromes. In the SintraREV trial, we aimed to investigate whether an early intervention of low lenalidomide doses for 2 years could delay transfusion dependency in patients with anaemia who were not transfusion dependent. METHODS This randomised, double-blind, phase 3 trial, was conducted at 22 sites (University Hospitals) in Spain, France, and Germany. Eligible patients were aged 18 years or older diagnosed with low-risk or intermediate-1-risk del(5q) myelodysplastic syndromes with non-transfusion-dependent anaemia (according to the IPSS), were erythropoietin-stimulating agents naive, and had an ECOG performance status of 2 or less. Patients were randomly assigned (2:1) by means of a telephone system to receive lenalidomide 5 mg daily in 28-day cycles versus placebo for 2 years. The primary endpoint was time to transfusion dependency based on blinded independent central review. Analysis were by intent-to-treat (ITT) and evaluable population. Safety analyses included all participants who received at least one dose of treatment. This trial is registered with ClinicalTrials.gov (NCT01243476) and EudraCT (2009-013619-36) and is complete. FINDINGS Between Feb 15, 2010, and Feb 21, 2018, 61 patients were randomly assigned to receive lenalidomide (n=40; two did not receive treatment) or placebo (n=21). The median age was 72·2 (IQR 65·4-81·9) years, 50 (82%) patients were female, and 11 (18%) were male. The median follow-up time was 60·6 (IQR 32·1-73·9) months. Regarding primary endpoint, median time to transfusion dependency was not reached (95% CI not applicable) in the lenalidomide group versus 11·6 months (95% CI 0·00-30·11) in the placebo group (p=0·0027). Lenalidomide significantly reduced the risk of transfusion dependency by 69·8% (hazard ratio 0·302, 95% CI 0·132-0·692; p=0·0046). The most frequent treatment-related adverse event was neutropenia, occurring in 24 (63%) of 38 patients in the lenalidomide group (grade 3 and 4 in 17 [45%] patients and one [3%], respectively) and in four (19%) of 21 patients in the placebo group (grade 3 in one [5%] patient). Thrombocytopenia was detected in seven (18%) of 38 patients receiving lenalidomide (grade 3 in two [5%] patients). Regarding the non-haematological toxicity, skin disorders (rash nine [23%] of 38 patients) were the most frequently described toxicities among patients receiving lenalidomide, being grade 3 in one (3%) of 38 patients. 19 serious adverse events were reported in 13 patients, 18 in the lenalidomide group and one in the placebo group, five of which were potentially related to the study drug. No treatment-related deaths were identified. INTERPRETATION An early approach with low doses of lenalidomide across two years delays the time to transfusion dependency and improves the rate and quality of the responses, with a manageable safety profile in patients who are non-transfusion dependent with del(5q) low-risk myelodysplastic syndromes. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- María Díez-Campelo
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain.
| | - Félix López-Cadenas
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Blanca Xicoy
- Department of Clinical Hematology, Institut Català d'Oncologia, Hospital Germans Trias i Pujol, Barcelona, Spain; Josep Carreras Leukemia Research Institute, Myeloid Neoplasm Group Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Eva Lumbreras
- IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Teresa González
- IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | | | - Joaquín Sánchez-García
- Department of Hematology, Hospital Universitario Reina Sofía, Córdoba, Spain; Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Córdoba, Spain; Universidad de Córdoba, Córdoba, Spain
| | - Rosa Coll Jordà
- Department of Hematology, Hospital Josep Trueta, Institut Català d'Oncologia, Girona, Spain
| | - Bohrane Slama
- Department of Clinical Hematology, CH Avignon, Avignon, France
| | - Jose-Ángel Hernández-Rivas
- Department of Hematology, Hospital Universitario Infanta Leonor, Universidad Complutense de Madrid, Madrid, Spain
| | - Sylvain Thepot
- Department of Clinical Hematology, Angers University Hospital, Angers, France
| | - Teresa Bernal
- Department of Hematology, Hospital Universitario Central de Asturias, Oviedo, Spain
| | | | - Joan Bargay
- Department of Hematology, Hospital Universitario Son Llatzer, Palma de Mallorca, Spain; Institut d'Investigacio Illes Balears (IdISBa), Palma de Mallorca, Spain
| | - María Luz Amigo
- Department of Hematology, Hospital Universitario Jose María Morales Meseguer, Murcia, Spain
| | - Claude Preudhomme
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), UMR9020-U1277, University of Lille, Lille, France; Laboratory of Hematology, CHU Lille, Lille, France
| | - Laurene Fenwarth
- Cancer Heterogeneity Plasticity and Resistance to Therapies (CANTHER), UMR9020-U1277, University of Lille, Lille, France; Laboratory of Hematology, CHU Lille, Lille, France
| | - Uwe Platzbecker
- Department for Hematology, Cell Therapy, Hemostaseology, and Infectious Medicine, University Hospital Leipzig, Leipzig, Germany
| | - Katharina S Götze
- Department of Medicine III, Technical University of Munich, Munich, Germany
| | - Ali Arar
- Department of Hematology, CHR Orleans, Orléans, France
| | - Sofía Toribio
- IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Consuelo Del Cañizo
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain
| | - Jesús María Hernández-Rivas
- Department of Hematology, Hospital Universitario de Salamanca, Salamanca, Spain; IBSAL, Instituto de investigación Biomédica de Salamanca, Salamanca, Spain; Department of Medicine, University of Salamanca, Salamanca, Spain
| | - Pierre Fenaux
- Département (DMU) d'hématologie et immunologie, Service d'hématologie Seniors, Hôpital St Louis, Université de Paris, APHP Nord Paris, France
| |
Collapse
|
15
|
Madanat YF, DeZern AE. A stimulating advance in erythropoiesis for patients with myelodysplastic syndromes. Lancet Haematol 2024; 11:e630-e631. [PMID: 39038478 DOI: 10.1016/s2352-3026(24)00211-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Accepted: 06/26/2024] [Indexed: 07/24/2024]
Affiliation(s)
- Yazan F Madanat
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, MD 21287, USA.
| |
Collapse
|
16
|
Brodermann MH, Henderson EK, Sellar RS. The emerging role of targeted protein degradation to treat and study cancer. J Pathol 2024; 263:403-417. [PMID: 38886898 DOI: 10.1002/path.6301] [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: 01/18/2024] [Revised: 04/18/2024] [Accepted: 04/30/2024] [Indexed: 06/20/2024]
Abstract
The evolution of cancer treatment has provided increasingly targeted strategies both in the upfront and relapsed disease settings. Small-molecule inhibitors and immunotherapy have risen to prominence with chimeric antigen receptor T-cells, checkpoint inhibitors, kinase inhibitors, and monoclonal antibody therapies being deployed across a range of solid organ and haematological malignancies. However, novel approaches are required to target transcription factors and oncogenic fusion proteins that are central to cancer biology and have generally eluded successful drug development. Thalidomide analogues causing protein degradation have been a cornerstone of treatment in multiple myeloma, but a lack of in-depth mechanistic understanding initially limited progress in the field. When the protein cereblon (CRBN) was found to mediate thalidomide analogues' action and CRBN's neo-targets were identified, existing and novel drug development accelerated, with applications outside multiple myeloma, including non-Hodgkin's lymphoma, myelodysplastic syndrome, and acute leukaemias. Critically, transcription factors were the first canonical targets described. In addition to broadening the application of protein-degrading drugs, resistance mechanisms are being overcome and targeted protein degradation is widening the scope of druggable proteins against which existing approaches have been ineffective. Examples of targeted protein degraders include molecular glues and proteolysis targeting chimeras (PROTACs): heterobifunctional molecules that bind to proteins of interest and cause proximity-induced ubiquitination and proteasomal degradation via a linked E3 ligase. Twenty years since their inception, PROTACs have begun progressing through clinical trials, with early success in targeting the oestrogen receptor and androgen receptor in breast and prostate cancer respectively. This review explores important developments in targeted protein degradation to both treat and study cancer. It also considers the potential advantages and challenges in the translational aspects of developing new treatments. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Elizabeth K Henderson
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| | - Rob S Sellar
- Department of Haematology, UCL Cancer Institute, University College London, London, UK
| |
Collapse
|
17
|
Song T, Zhao S, Luo S, Chen C, Liu X, Wu X, Sun Z, Cao J, Wang Z, Wang Y, Yu B, Zhang Z, Du X, Li X, Han Z, Chen H, Chen F, Wang L, Wang H, Sun K, Han Y, Xie L, Ji Y. SLC44A2 regulates vascular smooth muscle cell phenotypic switching and aortic aneurysm. J Clin Invest 2024; 134:e173690. [PMID: 38916960 PMCID: PMC11324303 DOI: 10.1172/jci173690] [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: 07/07/2023] [Accepted: 06/13/2024] [Indexed: 06/27/2024] Open
Abstract
Aortic aneurysm is a life-threatening disease with limited interventions that is closely related to vascular smooth muscle cell (VSMC) phenotypic switching. SLC44A2, a member of the solute carrier series 44 (SLC44) family, remains undercharacterized in the context of cardiovascular diseases. Venn diagram analysis based on microarray and single-cell RNA sequencing identified SLC44A2 as a major regulator of VSMC phenotypic switching in aortic aneurysm. Screening for Slc44a2 among aortic cell lineages demonstrated its predominant location in VSMCs. Elevated levels of SLC44A2 were evident in the aorta of both patients with abdominal aortic aneurysm and angiotensin II-infused (Ang II-infused) Apoe-/- mice. In vitro, SLC44A2 silencing promoted VSMCs toward a synthetic phenotype, while SLC44A2 overexpression attenuated VSMC phenotypic switching. VSMC-specific SLC44A2-knockout mice were more susceptible to aortic aneurysm under Ang II infusion, while SLC44A2 overexpression showed protective effects. Mechanistically, SLC44A2's interaction with NRP1 and ITGB3 activates TGF-β/SMAD signaling, thereby promoting contractile gene expression. Elevated SLC44A2 in aortic aneurysm is associated with upregulated runt-related transcription factor 1 (RUNX1). Furthermore, low-dose lenalidomide (LEN; 20 mg/kg/day) suppressed aortic aneurysm progression by enhancing SLC44A2 expression. These findings reveal that the SLC44A2-NRP1-ITGB3 complex is a major regulator of VSMC phenotypic switching and provide a potential therapeutic approach (LEN) for aortic aneurysm treatment.
Collapse
MESH Headings
- Animals
- Humans
- Male
- Mice
- Angiotensin II/pharmacology
- Aortic Aneurysm/genetics
- Aortic Aneurysm/metabolism
- Aortic Aneurysm/pathology
- Aortic Aneurysm, Abdominal/metabolism
- Aortic Aneurysm, Abdominal/pathology
- Aortic Aneurysm, Abdominal/genetics
- Membrane Transport Proteins/genetics
- Membrane Transport Proteins/metabolism
- Mice, Knockout
- Mice, Knockout, ApoE
- Muscle, Smooth, Vascular/metabolism
- Muscle, Smooth, Vascular/pathology
- Myocytes, Smooth Muscle/metabolism
- Myocytes, Smooth Muscle/pathology
- Phenotype
- Signal Transduction
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/metabolism
Collapse
Affiliation(s)
- Tianyu Song
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shuang Zhao
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Shanshan Luo
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Chuansheng Chen
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xingeng Liu
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Xiaoqi Wu
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Zhongxu Sun
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Jiawei Cao
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Ziyu Wang
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yineng Wang
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Bo Yu
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), and
| | - Zhiren Zhang
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), and
- Department of Cardiology, Central Laboratory, The First Affiliated Hospital of Harbin Medical University, NHC Key Laboratory of Cell Transplantation, Harbin Medical University, China
| | - Xiaolong Du
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Xiaoqiang Li
- Department of Vascular Surgery, The Affiliated Nanjing Drum Tower Hospital, Nanjing University Medical School, Nanjing, China
| | - Zhijian Han
- Department of Urology, First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongshan Chen
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Feng Chen
- Department of Forensic Medicine, and
| | - Liansheng Wang
- Department of Cardiology, the First Affiliated Hospital of Nanjing Medical University, Nanjing Medical University, Nanjing, China
| | - Hong Wang
- Center for Metabolic Disease Research, Department of Microbiology and Immunology, Temple University Lewis Katz School of Medicine, Philadelphia, Pennsylvania, USA
| | - Kangyun Sun
- Department of Cardiology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, China
| | - Yi Han
- Critical Care Department, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Liping Xie
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yong Ji
- Gusu School, Nanjing Medical University, Suzhou, China
- Key Laboratory of Cardiovascular and Cerebrovascular Medicine, Key Laboratory of Targeted Intervention of Cardiovascular Disease, Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing, Jiangsu, China
- State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), and
| |
Collapse
|
18
|
Hernandez-Lopez P, Vijaykumar T, Anand P, Auclair D, Frede J, Knoechel B, Lohr JG. Dual role of signaling pathways in myeloma requires cell type-specific targeting of ligand-receptor interactions. Blood Adv 2024; 8:3173-3185. [PMID: 38603572 PMCID: PMC11225681 DOI: 10.1182/bloodadvances.2023011463] [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: 08/17/2023] [Revised: 01/18/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
ABSTRACT Although most patients with multiple myeloma respond to treatment initially, therapy resistance develops almost invariably, and only a subset of patients show durable responses to immunomodulatory therapies. Although the immune microenvironment has been extensively studied in patients with myeloma, its composition is currently not used as prognostic markers in clinical routine. We hypothesized that the outcome of immune signaling pathway engagement can be highly variable, depending on which 2 cellular populations participate in this interaction. This would have important prognostic and therapeutic implications, suggesting that it is crucial for immune pathways to be targeted in a specific cellular context. To test this hypothesis, we investigated a cohort of 25 patients with newly diagnosed multiple myeloma. We examined the complex regulatory networks within the immune compartment and their impact on disease progression. Analysis of immune cell composition and expression profiles revealed significant differences in the B-cell compartment associated with treatment response. Transcriptional states in patients with short time to progression demonstrated an enrichment of pathways promoting B-cell differentiation and inflammatory responses, which may indicate immune dysfunction. Importantly, the analysis of molecular interactions within the immune microenvironment highlights the dual role of signaling pathways, which can either be associated with good or poor prognosis depending on the cell types involved. Our findings therefore argue that therapeutic strategies targeting ligand-receptor interactions should take into consideration the composition of the microenvironment and the specific cell types involved in molecular interactions.
Collapse
Affiliation(s)
- Pablo Hernandez-Lopez
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Tushara Vijaykumar
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
| | - Praveen Anand
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | | | - Julia Frede
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| | - Birgit Knoechel
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston, MA
| | - Jens G. Lohr
- Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, Dana-Farber Cancer Institute, Boston, MA
- Harvard Medical School, Boston, MA
- Broad Institute of MIT and Harvard, Cambridge, MA
| |
Collapse
|
19
|
Serrano G, Berastegui N, Díaz-Mazkiaran A, García-Olloqui P, Rodriguez-Res C, Huerga-Dominguez S, Ainciburu M, Vilas-Zornoza A, Martin-Uriz PS, Aguirre-Ruiz P, Ullate-Agote A, Ariceta B, Lamo-Espinosa JM, Acha P, Calvete O, Jimenez T, Molero A, Montoro MJ, Díez-Campelo M, Valcarcel D, Solé F, Alfonso-Pierola A, Ochoa I, Prósper F, Ezponda T, Hernaez M. Single-cell transcriptional profile of CD34+ hematopoietic progenitor cells from del(5q) myelodysplastic syndromes and impact of lenalidomide. Nat Commun 2024; 15:5272. [PMID: 38902243 PMCID: PMC11189937 DOI: 10.1038/s41467-024-49529-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 06/06/2024] [Indexed: 06/22/2024] Open
Abstract
While myelodysplastic syndromes with del(5q) (del(5q) MDS) comprises a well-defined hematological subgroup, the molecular basis underlying its origin remains unknown. Using single cell RNA-seq (scRNA-seq) on CD34+ progenitors from del(5q) MDS patients, we have identified cells harboring the deletion, characterizing the transcriptional impact of this genetic insult on disease pathogenesis and treatment response. Interestingly, both del(5q) and non-del(5q) cells present similar transcriptional lesions, indicating that all cells, and not only those harboring the deletion, may contribute to aberrant hematopoietic differentiation. However, gene regulatory network (GRN) analyses reveal a group of regulons showing aberrant activity that could trigger altered hematopoiesis exclusively in del(5q) cells, pointing to a more prominent role of these cells in disease phenotype. In del(5q) MDS patients achieving hematological response upon lenalidomide treatment, the drug reverts several transcriptional alterations in both del(5q) and non-del(5q) cells, but other lesions remain, which may be responsible for potential future relapses. Moreover, lack of hematological response is associated with the inability of lenalidomide to reverse transcriptional alterations. Collectively, this study reveals transcriptional alterations that could contribute to the pathogenesis and treatment response of del(5q) MDS.
Collapse
Affiliation(s)
- Guillermo Serrano
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology (KAUST), Thuwal, Saudi Arabia
| | - Nerea Berastegui
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Aintzane Díaz-Mazkiaran
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Paula García-Olloqui
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Carmen Rodriguez-Res
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
| | - Sofia Huerga-Dominguez
- Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
| | - Marina Ainciburu
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Amaia Vilas-Zornoza
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | - Patxi San Martin-Uriz
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
| | - Paula Aguirre-Ruiz
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
| | - Asier Ullate-Agote
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
| | - Beñat Ariceta
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
| | | | - Pamela Acha
- MDS Research Group, Josep Carreras Leukaemia Research Institut, Universitat Autònoma de Barcelona, Barcelona, Spain
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Oriol Calvete
- MDS Research Group, Josep Carreras Leukaemia Research Institut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Tamara Jimenez
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - Antonieta Molero
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Maria Julia Montoro
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Maria Díez-Campelo
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Department of Hematology, Hospital Universitario de Salamanca-IBSAL, Salamanca, Spain
| | - David Valcarcel
- Service of Hematology, Hospital Universitari Vall d'Hebron, Barcelona; Vall d'Hebron Instituto de Oncología (VHIO), Barcelona, Spain
| | - Francisco Solé
- MDS Research Group, Josep Carreras Leukaemia Research Institut, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Ana Alfonso-Pierola
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain
- Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain
| | - Idoia Ochoa
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), University of Navarra, Pamplona, Spain
- Department of Electrical and Electronics engineering, School of Engineering (Tecnun), University of Navarra, Donostia, Spain
| | - Felipe Prósper
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
- Hematology and Cell Therapy Service, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain.
| | - Teresa Ezponda
- Hematology-Oncology Program, CIMA, Cancer Center Clínica Universidad de Navarra (CCUN), IdiSNA, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
| | - Mikel Hernaez
- Computational Biology Program CIMA-Universidad de Navarra, Cancer Center Clínica Universidad de Navarra (CCUN), IdISNA, Pamplona, Spain.
- Centro de Investigación Biomédica en Red de Cáncer, CIBERONC, Madrid, Spain.
- Instituto de Ciencia de los Datos e Inteligencia Artificial (DATAI), University of Navarra, Pamplona, Spain.
| |
Collapse
|
20
|
Battaglia MR, Cannova J, Madero-Marroquin R, Patel AA. Treatment of Anemia in Lower-Risk Myelodysplastic Syndrome. Curr Treat Options Oncol 2024; 25:752-768. [PMID: 38814537 DOI: 10.1007/s11864-024-01217-0] [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] [Accepted: 05/13/2024] [Indexed: 05/31/2024]
Abstract
OPINION STATEMENT A majority of patients with lower-risk myelodysplastic syndrome (MDS) will present with or develop anemia. Anemia in MDS is associated with decreased quality of life and may correlate with decreased progression-free survival and overall survival. In this state of the art review we summarize current risk stratification approaches to identify lower-risk MDS (LR-MDS), the natural history of the disease, and meaningful clinical endpoints. The treatment landscape of LR-MDS with anemia is also rapidly evolving; we review the role of supportive care, erythropoietin stimulating agents, lenalidomide, luspatercept, hypomethylating agents (HMAs), and immunosuppressive therapy (IST) in the management of LR-MDS with anemia. In patients with deletion 5q (del5q) syndrome lenalidomide has both efficacy and durability of response. For patients without del5q who need treatment, the management approach is impacted by serum erythropoietin (EPO) level, SF3B1 mutation status, and ring sideroblast status. Given the data from the Phase III COMMANDS trial, we utilize luspatercept in those with SF3B1 mutation or ring sideroblasts that have an EPO level < 500 U/L; in patients without an SF3B1 mutation or ring sideroblasts there is equipoise between luspatercept and use of an erythropoietin stimulating agent (ESA). For patients who have an EPO level ≥ 500 U/L or have been previously treated there is not a clear standard of care. For those without previous luspatercept exposure it can be considered particularly if there is an SF3B1 mutation or the presence of ring sideroblasts. Other options include HMAs or IST; the Phase III IMERGE trial supports the efficacy of the telomerase inhibitor imetelstat in this setting and this may become a standard option in the future as well.
Collapse
Affiliation(s)
| | - Joseph Cannova
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Avenue MC 2115, Chicago, IL, 60637, USA
| | - Rafael Madero-Marroquin
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Avenue MC 2115, Chicago, IL, 60637, USA
| | - Anand A Patel
- Section of Hematology-Oncology, Department of Medicine, University of Chicago, 5841 S Maryland Avenue MC 2115, Chicago, IL, 60637, USA.
| |
Collapse
|
21
|
Kewan T, Stahl M, Bewersdorf JP, Zeidan AM. Treatment of Myelodysplastic Syndromes for Older Patients: Current State of Science, Challenges, and Opportunities. Curr Hematol Malig Rep 2024; 19:138-150. [PMID: 38632155 DOI: 10.1007/s11899-024-00733-y] [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] [Accepted: 03/20/2024] [Indexed: 04/19/2024]
Abstract
PURPOSE OF REVIEW Myelodysplastic syndromes/neoplasms (MDS) represent a diverse group of pathologically distinct diseases with varying prognoses and risks of leukemia progression. This review aims to discuss current treatment options for elderly patients with MDS, focusing on patients ineligible for intensive chemotherapy or allogenic hematopoietic stem cell transplantation (HSCT). The challenges associated with treatment in this population and emerging therapeutic prospects are also explored. RECENT FINDINGS Recent advancements in molecular diagnostics have enhanced risk stratification by incorporating genetic mutations, notably through the molecular International Prognostic Scoring System (IPSS-M). Lower-risk MDS (LR-MDS) treatment ranges from observation to supportive measures and erythropoiesis-stimulating agents (ESAs), with emerging therapies like luspatercept showing promise. High-risk MDS (HR-MDS) is treated with hypomethylating agents (HMAs) or allogenic HSCT, but outcomes remain poor. Elderly MDS patients, often diagnosed after 70, pose challenges in treatment decision-making. The IPSS-M aids risk stratification, guiding therapeutic choices. For LR-MDS, supportive care, ESAs, and novel agents like luspatercept are considered. Treatment of HR-MDS involves HMAs or allogenic HSCT. Emerging treatments, including oral HMAs and novel agents targeting FLT3, and IDH 1/2 mutations, show promise. Future research should refine treatment strategies for this elderly population focusing on quality-of-life improvement.
Collapse
Affiliation(s)
- Tariq Kewan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, and Yale Comprehensive Cancer Center, Yale University, New Haven, CT, USA
| | - Maximillian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, and Yale Comprehensive Cancer Center, Yale University, New Haven, CT, USA.
| |
Collapse
|
22
|
Bruzzese A, Martino EA, Mendicino F, Lucia E, Olivito V, Capodanno I, Neri A, Morabito F, Vigna E, Gentile M. Myelodysplastic syndromes del(5q): Pathogenesis and its therapeutic implications. Eur J Haematol 2024; 112:860-869. [PMID: 38294126 DOI: 10.1111/ejh.14181] [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: 12/14/2023] [Revised: 01/20/2024] [Accepted: 01/22/2024] [Indexed: 02/01/2024]
Abstract
Myelodysplastic syndromes (MDS) encompass a heterogeneous set of acquired bone marrow neoplastic disorders characterized by ineffective hematopoiesis within one or more bone marrow lineages. Nearly half of MDS patients carry cytogenetic alterations, with del(5q) being the most prevalent. Since its first description, del(5q) was consistently correlated with a typical clinical phenotype marked by anemia, thrombocytosis, and a low risk of evolving into acute leukemia. Presently, the World Health Organization (WHO) classification of myeloid neoplasms recognizes a specific subtype of MDS known as "myelodysplastic neoplasm with low blast and isolated del(5q)" identified by the sole presence of 5q deletion or in combination with one other abnormality excluding -7/del(7q). Several studies have sought to unravel the biological processes triggered by del(5q) in the development of MDS, revealing the involvement of various genes localized in specific regions of chromosome 5 referred to as common deleted regions (CDR). This intricate biological landscape makes the MDS cells with del(5q) exceptionally sensitive to lenalidomide. Several studies have confirmed the efficacy of lenalidomide in this context. Regrettably, the response to lenalidomide is not conclusive, prompting ongoing research into biological mechanisms that drive patients toward leukemia and strategies to circumvent lenalidomide resistance and disease progression.
Collapse
Affiliation(s)
- Antonella Bruzzese
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | | | - Francesco Mendicino
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Eugenio Lucia
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Virginia Olivito
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | | | - Antonino Neri
- Scientific Direction Azienda USL-IRCCS of Reggio Emilia, Reggio Emilia, Italy
| | - Fortunato Morabito
- Biotechnology Research Unit, Aprigliano, A.O./ASP of Cosenza, Cosenza, Italy
| | - Ernesto Vigna
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
| | - Massimo Gentile
- Department of Onco-hematology, Hematology Unit, A.O. of Cosenza, Cosenza, Italy
- Department of Pharmacy, Health and Nutritional Science, University of Calabria, Rende, Italy
| |
Collapse
|
23
|
Karel D, Valburg C, Woddor N, Nava VE, Aggarwal A. Myelodysplastic Neoplasms (MDS): The Current and Future Treatment Landscape. Curr Oncol 2024; 31:1971-1993. [PMID: 38668051 PMCID: PMC11049094 DOI: 10.3390/curroncol31040148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 03/16/2024] [Accepted: 03/30/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic neoplasms (MDS) are a heterogenous clonal disorder of hemopoietic stem cells characterized by cytomorphologic dysplasia, ineffective hematopoiesis, peripheral cytopenias and risk of progression to acute myeloid leukemia (AML). Our understanding of this disease has continued to evolve over the last century. More recently, prognostication and treatment have been determined by cytogenetic and molecular data. Specific genetic abnormalities, such as deletion of the long arm of chromosome 5 (del(5q)), TP53 inactivation and SF3B1 mutation, are increasingly associated with disease phenotype and outcome, as reflected in the recently updated fifth edition of the World Health Organization Classification of Hematolymphoid Tumors (WHO5) and the International Consensus Classification 2022 (ICC 2022) classification systems. Treatment of lower-risk MDS is primarily symptom directed to ameliorate cytopenias. Higher-risk disease warrants disease-directed therapy at diagnosis; however, the only possible cure is an allogenic bone marrow transplant. Novel treatments aimed at rational molecular and cellular pathway targets have yielded a number of candidate drugs over recent years; however few new approvals have been granted. With ongoing research, we hope to increasingly offer our MDS patients tailored therapeutic approaches, ultimately decreasing morbidity and mortality.
Collapse
Affiliation(s)
- Daniel Karel
- Department of Hematology/Medical Oncology, The George Washington University, Washington, DC 20037, USA; (C.V.); (A.A.)
| | - Claire Valburg
- Department of Hematology/Medical Oncology, The George Washington University, Washington, DC 20037, USA; (C.V.); (A.A.)
| | - Navitha Woddor
- Department of Pathology, The George Washington University, Washington, DC 20037, USA; (N.W.); (V.E.N.)
| | - Victor E. Nava
- Department of Pathology, The George Washington University, Washington, DC 20037, USA; (N.W.); (V.E.N.)
- Department of Pathology, Washington DC VA Medical Center, Washington, DC 20422, USA
| | - Anita Aggarwal
- Department of Hematology/Medical Oncology, The George Washington University, Washington, DC 20037, USA; (C.V.); (A.A.)
- Department of Hematology/Medical Oncology, Washington DC VA Medical Center, Washington, DC 20422, USA
| |
Collapse
|
24
|
Pereira MP, Herrity E, Kim DDH. TP53-mutated acute myeloid leukemia and myelodysplastic syndrome: biology, treatment challenges, and upcoming approaches. Ann Hematol 2024; 103:1049-1067. [PMID: 37770618 DOI: 10.1007/s00277-023-05462-5] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Accepted: 09/17/2023] [Indexed: 09/30/2023]
Abstract
Improved understanding of TP53 biology and the clinicopathological features of TP53-mutated myeloid neoplasms has led to the recognition of TP53-mutated acute myeloid leukemia/myelodysplastic syndrome (TP53m AML/MDS) as a unique entity, characterized by dismal outcomes following conventional therapies. Several clinical trials have investigated combinations of emerging therapies for these patients with the poorest molecular prognosis among myeloid neoplasms. Although some emerging therapies have shown improvement in overall response rates, this has not translated into better overall survival, hence the notion that p53 remains an elusive target. New therapeutic strategies, including novel targeted therapies, immune checkpoint inhibitors, and monoclonal antibodies, represent a shift away from cytotoxic and hypomethylating-based therapies, towards approaches combining non-immune and novel immune therapeutic strategies. The triple combination of azacitidine and venetoclax with either magrolimab or eprenetapopt have demonstrated safety in early trials, with phase III trials currently underway, and promising interim clinical results. This review compiles background on TP53 biology, available and emerging therapies along with their mechanisms of action for the TP53m disease entity, current treatment challenges, and recently published data and status of ongoing clinical trials for TP53m AML/MDS.
Collapse
Affiliation(s)
- Mariana Pinto Pereira
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Elizabeth Herrity
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada
| | - Dennis D H Kim
- Hans Messner Allogeneic Blood and Marrow Transplantation Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, M5G2M9, Toronto, ON, Canada.
- Leukemia Program, Department of Medical Oncology and Hematology, Princess Margaret Cancer Centre, Toronto, ON, Canada.
- Department of Hematology, Faculty of Medicine, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
25
|
Streuer A, Jann JC, Boch T, Mossner M, Riabov V, Schmitt N, Altrock E, Xu Q, Demmerle M, Nowak V, Oblaender J, Palme I, Weimer N, Rapp F, Metzgeroth G, Hecht A, Höger T, Merz C, Hofmann WK, Nolte F, Nowak D. Treatment with the apoptosis inhibitor Asunercept reduces clone sizes in patients with lower risk Myelodysplastic Neoplasms. Ann Hematol 2024; 103:1221-1233. [PMID: 38413410 PMCID: PMC10940491 DOI: 10.1007/s00277-024-05664-5] [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: 12/19/2023] [Accepted: 02/14/2024] [Indexed: 02/29/2024]
Abstract
In low-risk Myelodysplastic Neoplasms (MDS), increased activity of apoptosis-promoting factors such as tumor necrosis factor (TNFα) and pro-apoptotic Fas ligand (CD95L) have been described as possible pathomechanisms leading to impaired erythropoiesis. Asunercept (APG101) is a novel therapeutic fusion protein blocking CD95, which has previously shown partial efficacy in reducing transfusion requirement in a clinical phase I trial for low-risk MDS patients (NCT01736436; 2012-11-26). In the current study we aimed to evaluate the effect of Asunercept therapy on the clonal bone marrow composition to identify potential biomarkers to predict response. Bone marrow samples of n = 12 low-risk MDS patients from the above referenced clinical trial were analyzed by serial deep whole exome sequencing in a total of n = 58 time points. We could distinguish a mean of 3.5 molecularly defined subclones per patient (range 2-6). We observed a molecular response defined as reductions of dominant clone sizes by a variant allele frequency (VAF) decrease of at least 10% (mean 20%, range: 10.5-39.2%) in dependency of Asunercept treatment in 9 of 12 (75%) patients. Most of this decline in clonal populations was observed after completion of 12 weeks treatment. Particularly early and pronounced reductions of clone sizes were found in subclones driven by mutations in genes involved in regulation of methylation (n = 1 DNMT3A, n = 1 IDH2, n = 1 TET2). Our results suggest that APG101 could be efficacious in reducing clone sizes of mutated hematopoietic cells in the bone marrow of Myelodysplastic Neoplasms, which warrants further investigation.
Collapse
Affiliation(s)
- Alexander Streuer
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany.
| | - Johann-Christoph Jann
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Tobias Boch
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Maximilian Mossner
- Centre for Genomics and Computational Biology, Barts Cancer Institute, London, UK
| | - Vladimir Riabov
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Nanni Schmitt
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Eva Altrock
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Qingyu Xu
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Marie Demmerle
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Verena Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Julia Oblaender
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Iris Palme
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Nadine Weimer
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Felicitas Rapp
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Georgia Metzgeroth
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Anna Hecht
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | | | | | - Wolf-Karsten Hofmann
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Florian Nolte
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim of the Heidelberg University, Pettenkoferstr. 22, 68169, Mannheim, Germany
| |
Collapse
|
26
|
van de Loosdrecht AA, Cremers EMP, Alhan C, Duetz C, In 't Hout FEM, Visser-Wisselaar HA, Chitu DA, Verbrugge A, Cunha SM, Ossenkoppele GJ, Janssen JJWM, Klein SK, Vellenga E, Huls GA, Muus P, Langemeijer SMC, de Greef GE, Te Boekhorst PAW, Raaijmakers MHG, van Marwijk Kooy M, Legdeur MC, Wegman JJ, Deenik W, de Weerdt O, van Maanen-Lamme TM, Jobse P, van Kampen RJW, Beeker A, Wijermans PW, Biemond BJ, Tanis BC, van Esser JWJ, Schaar CG, Noordzij-Nooteboom HS, Jacobs EMG, de Graaf AO, Jongen-Lavrencic M, Stevens-Kroef MJPL, Westers TM, Jansen JH. Determinants of lenalidomide response with or without erythropoiesis-stimulating agents in myelodysplastic syndromes: the HOVON89 trial. Leukemia 2024; 38:840-850. [PMID: 38297135 PMCID: PMC10997501 DOI: 10.1038/s41375-024-02161-6] [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: 06/30/2023] [Revised: 01/11/2024] [Accepted: 01/17/2024] [Indexed: 02/02/2024]
Abstract
A randomized phase-II study was performed in low/int-1 risk MDS (IPSS) to study efficacy and safety of lenalidomide without (arm A) or with (arm B) ESA/G-CSF. In arm B, patients without erythroid response (HI-E) after 4 cycles received ESA; G-CSF was added if no HI-E was obtained by cycle 9. HI-E served as primary endpoint. Flow cytometry and next-generation sequencing were performed to identify predictors of response. The final evaluation comprised 184 patients; 84% non-del(5q), 16% isolated del(5q); median follow-up: 70.7 months. In arm A and B, 39 and 41% of patients achieved HI-E; median time-to-HI-E: 3.2 months for both arms, median duration of-HI-E: 9.8 months. HI-E was significantly lower in non-del(5q) vs. del(5q): 32% vs. 80%. The same accounted for transfusion independency-at-week 24 (16% vs. 67%), but similar in both arms. Apart from presence of del(5q), high percentages of bone marrow lymphocytes and progenitor B-cells, a low number of mutations, absence of ring sideroblasts, and SF3B1 mutations predicted HI-E. In conclusion, lenalidomide induced HI-E in patients with non-del(5q) and del(5q) MDS without additional effect of ESA/G-CSF. The identified predictors of response may guide application of lenalidomide in lower-risk MDS in the era of precision medicine. (EudraCT 2008-002195-10).
Collapse
Affiliation(s)
- A A van de Loosdrecht
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands.
| | - E M P Cremers
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - C Alhan
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - C Duetz
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - F E M In 't Hout
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | | | - D A Chitu
- HOVON Foundation, Rotterdam, The Netherlands
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - A Verbrugge
- HOVON Foundation, Rotterdam, The Netherlands
| | - S M Cunha
- HOVON Foundation, Rotterdam, The Netherlands
| | - G J Ossenkoppele
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - J J W M Janssen
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - S K Klein
- Department of Hematology, Meander Medisch Centrum, Amersfoort, The Netherlands
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - E Vellenga
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - G A Huls
- Department of Hematology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - P Muus
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Haematology, St. James University Hospital, Leeds, UK
| | - S M C Langemeijer
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - G E de Greef
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - P A W Te Boekhorst
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - M H G Raaijmakers
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | | | - M C Legdeur
- Department of Hematology, Medisch Spectrum Twente, Enschede, The Netherlands
| | - J J Wegman
- Department of Hematology, Deventer Ziekenhuis, Deventer, The Netherlands
- Department of Hematology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - W Deenik
- Department of Internal Medicine, Tergooi Ziekenhuis, Hilversum, The Netherlands
- Department of Internal Medicine, Rijnstate, Arnhem, the Netherlands
| | - O de Weerdt
- Department of Internal Medicine, St. Antonius Ziekenhuis, Nieuwegein, The Netherlands
| | | | - P Jobse
- Department of Internal Medicine, Admiraal de Ruyter Ziekenhuis, Goes, The Netherlands
| | - R J W van Kampen
- Department of Internal Medicine, Zuyderland Ziekenhuis, Geleen, The Netherlands
| | - A Beeker
- Department of Hematology, Spaarne Gasthuis, Hoofddorp, The Netherlands
| | - P W Wijermans
- Department of Hematology, Haaglanden Ziekenhuis, Den Haag, The Netherlands
| | - B J Biemond
- Department of Hematology, Amsterdam UMC, location AMC, Amsterdam, The Netherlands
| | - B C Tanis
- Department of Internal Medicine, Groene Hart Ziekenhuis, Gouda, The Netherlands
- Department of General Practice Erasmus MC, Rotterdam, The Netherlands
| | - J W J van Esser
- Department of Internal Medicine, Amphia Ziekenhuis, Breda, The Netherlands
| | - C G Schaar
- Department of Internal Medicine, Gelre Ziekenhuis, Apeldoorn, The Netherlands
| | - H S Noordzij-Nooteboom
- Department of Internal Medicine, Van Weel Bethesda Ziekenhuis, Dirksland, The Netherlands
| | - E M G Jacobs
- Department of Internal Medicine, Elkerliek Ziekenhuis, Helmond, The Netherlands
| | - A O de Graaf
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - M Jongen-Lavrencic
- Department of Hematology, Erasmus MC Cancer Institute, Rotterdam, The Netherlands
| | - M J P L Stevens-Kroef
- Department of human genetics, Radboud University Medical Center, Nijmegen, The Netherlands
| | - T M Westers
- Department of Hematology, Amsterdam UMC, location VUmc, Cancer Center Amsterdam, Amsterdam, The Netherlands
| | - J H Jansen
- Department of Laboratory Medicine - Laboratory of Hematology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|
27
|
Chandhok NS, Sekeres MA. Combining lenalidomide with erythropoiesis stimulating agents: a party of one. Leukemia 2024; 38:473-474. [PMID: 38360864 DOI: 10.1038/s41375-024-02176-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/17/2024]
Affiliation(s)
- Namrata S Chandhok
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Mikkael A Sekeres
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA.
| |
Collapse
|
28
|
Brown A, Batra S. Rare Hematologic Malignancies and Pre-Leukemic Entities in Children and Adolescents Young Adults. Cancers (Basel) 2024; 16:997. [PMID: 38473358 DOI: 10.3390/cancers16050997] [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: 02/05/2024] [Revised: 02/27/2024] [Accepted: 02/27/2024] [Indexed: 03/14/2024] Open
Abstract
There are a variety of rare hematologic malignancies and germline predispositions syndromes that occur in children and adolescent young adults (AYAs). These entities are important to recognize, as an accurate diagnosis is essential for risk assessment, prognostication, and treatment. This descriptive review summarizes rare hematologic malignancies, myelodysplastic neoplasms, and germline predispositions syndromes that occur in children and AYAs. We discuss the unique biology, characteristic genomic aberrations, rare presentations, diagnostic challenges, novel treatments, and outcomes associated with these rare entities.
Collapse
Affiliation(s)
- Amber Brown
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Riley Hospital for Children, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA
| | - Sandeep Batra
- Division of Pediatric Hematology, Oncology and Stem Cell Transplant, Department of Pediatrics, Riley Hospital for Children, 705 Riley Hospital Drive, Indianapolis, IN 46202, USA
| |
Collapse
|
29
|
Hatakeyama K, Kikushige Y, Ishihara D, Yamamoto S, Kawano G, Tochigi T, Miyamoto T, Sakoda T, Christoforou A, Kunisaki Y, Fukata M, Kato K, Ito T, Handa H, Akashi K. Thrombospondin-1 is an endogenous substrate of cereblon responsible for immunomodulatory drug-induced thromboembolism. Blood Adv 2024; 8:785-796. [PMID: 38163319 PMCID: PMC10847748 DOI: 10.1182/bloodadvances.2023010080] [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: 02/27/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/03/2024] Open
Abstract
ABSTRACT Immunomodulatory drugs (IMiDs) are key drugs for treating multiple myeloma and myelodysplastic syndrome with chromosome 5q deletion. IMiDs exert their pleiotropic effects through the interaction between cell-specific substrates and cereblon, a substrate receptor of the E3 ubiquitin ligase complex. Thus, identification of cell-specific substrates is important for understanding the effects of IMiDs. IMiDs increase the risk of thromboembolism, which sometimes results in fatal clinical outcomes. In this study, we sought to clarify the molecular mechanisms underlying IMiDs-induced thrombosis. We investigated cereblon substrates in human megakaryocytes using liquid chromatography-mass spectrometry and found that thrombospondin-1 (THBS-1), which is an inhibitor of a disintegrin-like and metalloproteinase with thrombospondin type 1 motifs 13, functions as an endogenous substrate in human megakaryocytes. IMiDs inhibited the proteasomal degradation of THBS-1 by impairing the recruitment of cereblon to THBS-1, leading to aberrant accumulation of THBS-1. We observed a significant increase in THBS-1 in peripheral blood mononuclear cells as well as larger von Willebrand factor multimers in the plasma of patients with myeloma, who were treated with IMiDs. These results collectively suggest that THBS-1 represents an endogenous substrate of cereblon. This pairing is disrupted by IMiDs, and the aberrant accumulation of THBS-1 plays an important role in the pathogenesis of IMiDs-induced thromboembolism.
Collapse
Affiliation(s)
- Kiwamu Hatakeyama
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Yoshikane Kikushige
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Daisuke Ishihara
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Shunsuke Yamamoto
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Gentaro Kawano
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Taro Tochigi
- Miyazaki Prefectural Miyazaki Hospital, Miyazaki, Japan
| | - Toshihiro Miyamoto
- Haematology/Respiratory Medicine, Faculty of Medicine, Institute of Medical Pharmaceutical and Health Sciences, Kanazawa University Hospital, Ishikawa, Japan
| | - Teppei Sakoda
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | | | - Yuya Kunisaki
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
- Center for Cellular and Molecular Medicine, Kyushu University Hospital, Fukuoka, Japan
| | - Mitsuhiro Fukata
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Koji Kato
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| | - Takumi Ito
- Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Hiroshi Handa
- Center for Future Medical Research Institute of Medical Science, Tokyo Medical University, Tokyo, Japan
| | - Koichi Akashi
- Department of Medicine and Biosystemic Sciences, Kyushu University Graduate School of Medicine, Fukuoka, Japan
| |
Collapse
|
30
|
Merz AMA, Platzbecker U. Beyond the horizon: emerging therapeutic approaches in myelodysplastic neoplasms. Exp Hematol 2024; 130:104130. [PMID: 38036096 DOI: 10.1016/j.exphem.2023.11.004] [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: 07/30/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023]
Abstract
Management of myelodysplastic neoplasms (MDS) requires a personalized approach, with a focus on improving quality of life and extending lifespan. The International Prognostic Scoring System-Revised and the molecular International Prognostic Scoring System are key tools for risk stratification and management of MDS. They provide a framework for predicting survival and the risk of transformation to acute myeloid leukemia. However, a major challenge in MDS management remains the limited therapeutic options available, especially after the failure of first-line therapies. In lower-risk MDS, the failure of erythropoietin-stimulating agents often leaves few alternatives, although in higher-risk MDS, the prognosis after hypomethylating agent failure is dismal. This highlights the urgent need for novel, more personalized therapeutic approaches. In this review, we discuss emerging novel therapeutic approaches in the treatment of MDS. Several new therapeutic targets are currently being evaluated, offering hope for improved management of MDS in the future.
Collapse
Affiliation(s)
- Almuth Maria Anni Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, University of Leipzig Faculty of Medicine Leipzig, Germany.
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, University of Leipzig Faculty of Medicine Leipzig, Germany.
| |
Collapse
|
31
|
Merz AMA, Sébert M, Sonntag J, Kubasch AS, Platzbecker U, Adès L. Phase to phase: Navigating drug combinations with hypomethylating agents in higher-risk MDS trials for optimal outcomes. Cancer Treat Rev 2024; 123:102673. [PMID: 38176221 DOI: 10.1016/j.ctrv.2023.102673] [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: 11/03/2023] [Revised: 12/14/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024]
Abstract
Recent developments in high-risk Myelodysplastic Neoplasms (HR MDS) treatment are confronted with challenges in study design due to evolving drug combinations with Hypomethylating Agents (HMAs). The shift from the International Prognostic Scoring System (IPSS) to its molecular revision (IPSS-M) has notably influenced research and clinical practice. Introducing concepts like the MDS/AML overlap complicate classifications and including chronic myelomonocytic leukemia (CMML) in MDS studies introduces another layer of complexity. The International Consortium for MDS emphasizes aligning HR MDS criteria with the 2022 ELN criteria for AML. Differences in advancements between AML and MDS treatments and hematological toxicity in HR MDS underline the importance of detailed trial designs. Effective therapeutic strategies require accurate reporting of adverse events, highlighting the need for clarity in criteria like the Common Terminology Criteria for Adverse Events (CTCAE). We provide an overview on negative clinical trials in HR MDS, analyze possible reasons and explore possibilities to optimize future clinical trials in this challenging patient population.
Collapse
Affiliation(s)
- Almuth Maria Anni Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany
| | - Marie Sébert
- Service Hématologie Séniors, Hôpital Saint-Louis (AP-HP), Paris Cité University and INSERM U944, Paris, France
| | - Jan Sonntag
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany.
| | - Lionel Adès
- Service Hématologie Séniors, Hôpital Saint-Louis (AP-HP), Paris Cité University and INSERM U944, Paris, France.
| |
Collapse
|
32
|
Panizo Inogés M, Alfonso-Pierola A. [Myelodysplastic neoplasms]. Med Clin (Barc) 2024; 162:77-82. [PMID: 37604730 DOI: 10.1016/j.medcli.2023.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/23/2023]
Affiliation(s)
- María Panizo Inogés
- Departamento de Hematología y Hemoterapia, Clínica Universidad de Navarra, Pamplona, Navarra, España
| | - Ana Alfonso-Pierola
- Departamento de Hematología y Hemoterapia, Clínica Universidad de Navarra, Pamplona, Navarra, España.
| |
Collapse
|
33
|
Platzbecker U, Santini V, Fenaux P, Sekeres MA, Savona MR, Madanat YF, Díez-Campelo M, Valcárcel D, Illmer T, Jonášová A, Bělohlávková P, Sherman LJ, Berry T, Dougherty S, Shah S, Xia Q, Sun L, Wan Y, Huang F, Ikin A, Navada S, Feller F, Komrokji RS, Zeidan AM. Imetelstat in patients with lower-risk myelodysplastic syndromes who have relapsed or are refractory to erythropoiesis-stimulating agents (IMerge): a multinational, randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 2024; 403:249-260. [PMID: 38048786 DOI: 10.1016/s0140-6736(23)01724-5] [Citation(s) in RCA: 47] [Impact Index Per Article: 47.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 08/11/2023] [Accepted: 08/14/2023] [Indexed: 12/06/2023]
Abstract
BACKGROUND Unmet medical needs remain in patients with red blood cell transfusion-dependent (RBC-TD) lower-risk myelodysplastic syndromes (LR-MDS) who are not responding to or are ineligible for erythropoiesis-stimulating agents (ESAs). Imetelstat, a competitive telomerase inhibitor, showed promising results in a phase 2 trial. We aimed to compare the RBC transfusion independence (RBC-TI) rate with imetelstat versus placebo in patients with RBC-TD LR-MDS. METHODS In phase 3 of IMerge, a double-blind, placebo-controlled trial conducted in 118 sites including university hospitals, cancer centres, and outpatient clinics in 17 countries, patients (aged ≥18 years) with ESA-relapsed, ESA-refractory, or ESA-ineligible LR-MDS (low or intermediate-1 risk disease as per International Prognostic Scoring System [IPSS] criteria) were randomly assigned via a computer-generated schedule (2:1) to receive imetelstat 7·5 mg/kg or placebo, administered as a 2-h intravenous infusion, every 4 weeks until disease progression, unacceptable toxic effects, or withdrawal of consent. Randomisation was stratified by previous RBC transfusion burden and IPSS risk group. Patients, investigators, and those analysing the data were masked to group assignment. The primary endpoint was 8-week RBC-TI, defined as the proportion of patients without RBC transfusions for at least 8 consecutive weeks starting on the day of randomisation until subsequent anti-cancer therapy, if any. Primary efficacy analyses were performed in the intention-to-treat population, and safety analyses were conducted in patients who received at least one dose of trial medication or placebo. This trial is registered with ClinicalTrials.gov (NCT02598661; substudy active and recruiting). FINDINGS Between Sept 11, 2019, and Oct 13, 2021, 178 patients were enrolled and randomly assigned (118 to imetelstat and 60 to placebo). 111 (62%) were male and 67 (38%) were female. 91 (77%) of 118 patients had discontinued treatment by data cutoff in the imetelstat group versus 45 (75%) in the placebo group; a further one patient in the placebo group did not receive treatment. Median follow-up was 19·5 months (IQR 12·0-23·4) in the imetelstat group and 17·5 months (12·1-22·7) in the placebo group. In the imetelstat group, 47 (40% [95% CI 30·9-49·3]) patients had an RBC-TI of at least 8 weeks versus nine (15% [7·1-26·6]) in the placebo group (rate difference 25% [9·9 to 36·9]; p=0·0008). Overall, 107 (91%) of 118 patients receiving imetelstat and 28 (47%) of 59 patients receiving placebo had grade 3-4 treatment-emergent adverse events. The most common treatment-emergent grade 3-4 adverse events in patients taking imetelstat were neutropenia (80 [68%] patients who received imetelstat vs two [3%] who received placebo) and thrombocytopenia (73 [62%] vs five [8%]). No treatment-related deaths were reported. INTERPRETATION Imetelstat offers a novel mechanism of action with durable transfusion independence (approximately 1 year) and disease-modifying activity for heavily transfused patients with LR-MDS who are not responding to or are ineligible for ESAs. FUNDING Janssen Research & Development before April 18, 2019, and Geron Corporation thereafter.
Collapse
Affiliation(s)
- Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Infectious Diseases, and Hemostaseology, University Hospital Leipzig, Leipzig, Germany.
| | - Valeria Santini
- MDS Unit, AOU Careggi, University of Florence, Florence, Italy
| | - Pierre Fenaux
- Hôpital Saint-Louis, Université de Paris 7, Paris, France
| | - Mikkael A Sekeres
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Michael R Savona
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yazan F Madanat
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Maria Díez-Campelo
- Hematology Department, University Hospital of Salamanca, IBSAL, Salamanca, Spain
| | - David Valcárcel
- Department of Hematology, Hospital Universitari Vall d'Hebron, Barcelona, Spain
| | | | - Anna Jonášová
- 1st Medical Department-Hematology, Charles University General Hospital, Prague, Czech Republic
| | - Petra Bělohlávková
- 4th Department of Internal Medicine-Haematology, Charles University Hospital, Hradec Kralove, Czech Republic
| | | | | | | | | | - Qi Xia
- Geron Corporation, Parsippany, NJ, USA
| | - Libo Sun
- Geron Corporation, Parsippany, NJ, USA
| | - Ying Wan
- Geron Corporation, Parsippany, NJ, USA
| | - Fei Huang
- Geron Corporation, Parsippany, NJ, USA
| | | | | | | | | | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine and Yale Comprehensive Cancer Center, Yale University, New Haven, CT, USA
| |
Collapse
|
34
|
Nishiguchi G, Mascibroda LG, Young SM, Caine EA, Abdelhamed S, Kooijman JJ, Miller DJ, Das S, McGowan K, Mayasundari A, Shi Z, Barajas JM, Hiltenbrand R, Aggarwal A, Chang Y, Mishra V, Narina S, Thomas M, Loughran AJ, Kalathur R, Yu K, Zhou S, Wang X, High AA, Peng J, Pruett-Miller SM, Daniels DL, Urh M, Shelat AA, Mullighan CG, Riching KM, Zaman GJR, Fischer M, Klco JM, Rankovic Z. Selective CK1α degraders exert antiproliferative activity against a broad range of human cancer cell lines. Nat Commun 2024; 15:482. [PMID: 38228616 PMCID: PMC10791743 DOI: 10.1038/s41467-024-44698-1] [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: 05/01/2023] [Accepted: 12/21/2023] [Indexed: 01/18/2024] Open
Abstract
Molecular-glue degraders are small molecules that induce a specific interaction between an E3 ligase and a target protein, resulting in the target proteolysis. The discovery of molecular glue degraders currently relies mostly on screening approaches. Here, we describe screening of a library of cereblon (CRBN) ligands against a panel of patient-derived cancer cell lines, leading to the discovery of SJ7095, a potent degrader of CK1α, IKZF1 and IKZF3 proteins. Through a structure-informed exploration of structure activity relationship (SAR) around this small molecule we develop SJ3149, a selective and potent degrader of CK1α protein in vitro and in vivo. The structure of SJ3149 co-crystalized in complex with CK1α + CRBN + DDB1 provides a rationale for the improved degradation properties of this compound. In a panel of 115 cancer cell lines SJ3149 displays a broad antiproliferative activity profile, which shows statistically significant correlation with MDM2 inhibitor Nutlin-3a. These findings suggest potential utility of selective CK1α degraders for treatment of hematological cancers and solid tumors.
Collapse
Affiliation(s)
- Gisele Nishiguchi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Lauren G Mascibroda
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sarah M Young
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Elizabeth A Caine
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Sherif Abdelhamed
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | | | - Darcie J Miller
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Sourav Das
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kevin McGowan
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anand Mayasundari
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Zhe Shi
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Juan M Barajas
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ryan Hiltenbrand
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anup Aggarwal
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Yunchao Chang
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Vibhor Mishra
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Shilpa Narina
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Melvin Thomas
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Allister J Loughran
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Ravi Kalathur
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kaiwen Yu
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Suiping Zhou
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Xusheng Wang
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Anthony A High
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Junmin Peng
- Department of Structural Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Center for Proteomics and Metabolomics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Shondra M Pruett-Miller
- Center for Advanced Genome Engineering, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
- Department of Cell and Molecular Biology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, Memphis, TN, 38105, USA
| | - Danette L Daniels
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Marjeta Urh
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Anang A Shelat
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Charles G Mullighan
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA
| | - Kristin M Riching
- Promega Corporation, 5430 East Cheryl Drive, Madison, WI, 53711, USA
| | - Guido J R Zaman
- Oncolines B.V., Kloosterstraat 9, 5349 AB, Oss, The Netherlands
| | - Marcus Fischer
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| | - Jeffery M Klco
- Department of Pathology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| | - Zoran Rankovic
- Department of Chemical Biology and Therapeutics, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN, 38105, USA.
| |
Collapse
|
35
|
Yoon H, Rutter JC, Li YD, Ebert BL. Induced protein degradation for therapeutics: past, present, and future. J Clin Invest 2024; 134:e175265. [PMID: 38165043 PMCID: PMC10760958 DOI: 10.1172/jci175265] [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] [Indexed: 01/03/2024] Open
Abstract
The concept of induced protein degradation by small molecules has emerged as a promising therapeutic strategy that is particularly effective in targeting proteins previously considered "undruggable." Thalidomide analogs, employed in the treatment of multiple myeloma, stand as prime examples. These compounds serve as molecular glues, redirecting the CRBN E3 ubiquitin ligase to degrade myeloma-dependency factors, IKZF1 and IKZF3. The clinical success of thalidomide analogs demonstrates the therapeutic potential of induced protein degradation. Beyond molecular glue degraders, several additional modalities to trigger protein degradation have been developed and are currently under clinical evaluation. These include heterobifunctional degraders, polymerization-induced degradation, ligand-dependent degradation of nuclear hormone receptors, disruption of protein interactions, and various other strategies. In this Review, we will provide a concise overview of various degradation modalities, their clinical applications, and potential future directions in the field of protein degradation.
Collapse
Affiliation(s)
- Hojong Yoon
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Justine C. Rutter
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Yen-Der Li
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
| | - Benjamin L. Ebert
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
- Cancer Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts, USA
- Howard Hughes Medical Institute, Boston, Massachusetts, USA
| |
Collapse
|
36
|
Hellström-Lindberg ES, Kröger N. Clinical decision-making and treatment of myelodysplastic syndromes. Blood 2023; 142:2268-2281. [PMID: 37874917 DOI: 10.1182/blood.2023020079] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/22/2023] [Accepted: 10/12/2023] [Indexed: 10/26/2023] Open
Abstract
ABSTRACT The myelodysplastic syndromes (MDSs) constitute a profoundly heterogeneous myeloid malignancy with a common origin in the hemopoietic stem cell compartment. Consequently, patient management and treatment are as heterogeneous. Decision-making includes identifying risk, symptoms, and options for an individual and conducting a risk-benefit analysis. The only potential cure is allogeneic stem cell transplantation, and albeit the fraction of patients with MDS who undergo transplant increase over time because of better management and increased donor availability, a majority are not eligible for this intervention. Current challenges encompass to decrease the relapse risk, the main cause of hematopoietic stem cell transplantation failure. Hypomethylating agents (HMAs) constitute firstline treatment for higher-risk MDSs. Combinations with other drugs as firstline treatment has, to date, not proven more efficacious than monotherapy, although combinations approved for acute myeloid leukemia, including venetoclax, are under evaluation and often used as rescue treatment. The treatment goal for lower-risk MDS is to improve cytopenia, mainly anemia, quality of life, and, possibly, overall survival. Erythropoiesis-stimulating agents (ESAs) constitute firstline treatment for anemia and have better and more durable responses if initiated before the onset of a permanent transfusion need. Treatment in case of ESA failure or ineligibility should be tailored to the main disease mechanism: immunosuppression for hypoplastic MDS without high-risk genetics, lenalidomide for low-risk del(5q) MDS, and luspatercept for MDS with ring sideroblasts. Approved therapeutic options are still scarcer for MDS than for most other hematologic malignancies. Better tools to match disease biology with treatment, that is, applied precision medicines are needed to improve patient outcome.
Collapse
Affiliation(s)
- Eva S Hellström-Lindberg
- Department of Medicine, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Nicolaus Kröger
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
37
|
Madanat YF, Zeidan AM. Treatment Considerations of Myelodysplastic Syndromes/Neoplasms for Pathologists. Clin Lab Med 2023; 43:685-698. [PMID: 37865511 DOI: 10.1016/j.cll.2023.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2023]
Abstract
The diagnosis of myelodysplastic syndromes/neoplasms (MDS) has evolved over the years with the incorporation of genetic abnormalities to establish a diagnosis, their impact on risk stratification, prognostication, and therapeutic options. Hematopathologists are the cornerstone to establish an accurate diagnosis and ensure patients receive the best available treatment option. Hematopathologists and clinicians must work closely together to establish the best disease subclassification, by combining pathologic findings with the clinical presentation. This will ensure patients receive the best therapeutic approach by better understanding the disease entity. In this review, we discuss how we approach a bone marrow biopsy report in the management of MDS.
Collapse
Affiliation(s)
- Yazan F Madanat
- Eugene P. Frenkel M.D. Scholar in Clinical Medicine, Division of Hematology and Medical Oncology, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA. https://twitter.com/MadanatYazan
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Smilow Cancer Center, Yale University, New Haven, CT, USA.
| |
Collapse
|
38
|
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
|
39
|
Bosi A, Barcellini W, Passamonti F, Fattizzo B. Androgen use in bone marrow failures and myeloid neoplasms: Mechanisms of action and a systematic review of clinical data. Blood Rev 2023; 62:101132. [PMID: 37709654 DOI: 10.1016/j.blre.2023.101132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2023] [Revised: 08/22/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
Despite recent advancements, treatment of cytopenia due to bone marrow failures (BMF) and myeloid neoplasms remains challenging. Androgens promote renewal and maturation of blood cells and may be beneficial in these forms. Here we report a systematic review of androgens use as single agent in hematologic conditions. Forty-six studies, mainly retrospective with various androgen types and doses, were included: 12 on acquired aplastic anemia (AA), 11 on inherited BMF, 17 on myelodysplastic syndromes (MDS), and 7 on myelofibrosis. Responses ranged from 50 to 70% in inherited BMF, 40-50% in acquired AA and MDS, while very limited evidence emerged for myelofibrosis. In acquired AA, response was associated with presence of non-severe disease; in MDS androgens were more effective on thrombocytopenia or mild to moderate anemia, whilst limited benefit was observed for transfusion dependent anemia. Toxicity profile mainly consisted of virilization and liver enzyme elevation, whilst the risk of leukemic evolution remains controversial.
Collapse
Affiliation(s)
- Alessandro Bosi
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Wilma Barcellini
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Francesco Passamonti
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy
| | - Bruno Fattizzo
- Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy; Department of Oncology and Hemato-Oncology, University of Milan, Milan, Italy.
| |
Collapse
|
40
|
Petzer V, Wolf D. [Recent findings in myelodysplastic syndrome]. Dtsch Med Wochenschr 2023; 148:1431-1436. [PMID: 37918427 DOI: 10.1055/a-1968-3106] [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: 11/04/2023]
Abstract
Myelodysplastic syndromes (MDS) represent a heterogeneous group of myeloid disorders characterized by peripheral blood cytopenias and increased risk of transformation to acute myeloid leukemia (AML).Recent developments include the classification and the estimation of prognosis. In 2022 the former 2016 WHO classification was replaced by the ICC and WHO 2022 classification. Both classifications have included precursor lesions (CHIP and ICUS), both distinguish between three molecularly cytogenetically defined subgroups - del(5q), TP53, SF3B1 - and morphologically defined subgroups with differences in blast threshold (WHO: 20%; ICC: 10%) for the differentiation from AML. However, although prognostic factors influenced the classification-subgroups, it is important to distinguish the prognosis, which is crucial for optimal therapeutic decision making. Since 2022, the IPSS-M has been available for this purpose, which represents an expansion of the well-established IPSS-R. It could improve prognosis estimation by adding molecular data, recently this could have been confirmed in real world cohorts. The IPSS-M also represents an important extension with regard to prognosis estimation for patients with therapy-related MDS.In 2020 Luspatercept has been approved for transfusion-dependent lower risk MDS patients harboring ring sideroblasts ± an SF3B1 mutation after failure of an erythropoiesis stimulating agent. The COMMANDS trial has just reported an interim analysis, where the superiority of luspatercept in the 1st line compared to erythropoietin could be demonstrated. In addition, data from the phase III trial with Imeltelstat give reason to hope that we will be able to offer a new second-line therapy to LR-MDS patients. For higher risk MDS patients azacitidine therapy remains the standard of care, results of phase III trials of combination therapies must be awaited.
Collapse
|
41
|
Badar T, Madanat YF, Zeidan AM. Updates on risk stratification and management of lower-risk myelodysplastic syndromes/neoplasms. Future Oncol 2023; 19:1877-1889. [PMID: 37750305 DOI: 10.2217/fon-2023-0454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2023] Open
Abstract
The majority of lower-risk myelodysplastic syndromes/neoplasms patients present with anemia. Historically, these patients were treated with erythropoiesis-stimulating agents (ESA), with modest responses. A subset of these patients with del(5q) may do better with lenalidomide. Recently, in randomized trials, luspatercept has shown better responses compared with ESAs in treatment-naive patients and imetelstat in patients refractory to ESAs. Other evaluated novel compounds (fostamatinib, H3B-880, roxadustat, pyruvate kinase receptor activator) have not yet shown meaningful efficacy. More needs to be done to improve outcomes; in pursuance of this, participation in clinical trials evaluating novel therapies should be encouraged. While lower-risk myelodysplastic syndromes/neoplasms tend to have an indolent course, a subset of them has a dismal prognosis. Improving prognostication and serial monitoring will help in identifying high-risk patients for appropriate management.
Collapse
Affiliation(s)
- Talha Badar
- Division of Hematology & Oncology, Department of Medicine, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Yazan F Madanat
- Division of Hematology & Medical Oncology, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX 75390, USA
- Leukemia Program, Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine & Yale Cancer Center, New Haven, CT 06510, USA
| |
Collapse
|
42
|
Yamanaka S, Furihata H, Yanagihara Y, Taya A, Nagasaka T, Usui M, Nagaoka K, Shoya Y, Nishino K, Yoshida S, Kosako H, Tanokura M, Miyakawa T, Imai Y, Shibata N, Sawasaki T. Lenalidomide derivatives and proteolysis-targeting chimeras for controlling neosubstrate degradation. Nat Commun 2023; 14:4683. [PMID: 37596276 PMCID: PMC10439208 DOI: 10.1038/s41467-023-40385-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 07/21/2023] [Indexed: 08/20/2023] Open
Abstract
Lenalidomide, an immunomodulatory drug (IMiD), is commonly used as a first-line therapy in many haematological cancers, such as multiple myeloma (MM) and 5q myelodysplastic syndromes (5q MDS), and it functions as a molecular glue for the protein degradation of neosubstrates by CRL4CRBN. Proteolysis-targeting chimeras (PROTACs) using IMiDs with a target protein binder also induce the degradation of target proteins. The targeted protein degradation (TPD) of neosubstrates is crucial for IMiD therapy. However, current IMiDs and IMiD-based PROTACs also break down neosubstrates involved in embryonic development and disease progression. Here, we show that 6-position modifications of lenalidomide are essential for controlling neosubstrate selectivity; 6-fluoro lenalidomide induced the selective degradation of IKZF1, IKZF3, and CK1α, which are involved in anti-haematological cancer activity, and showed stronger anti-proliferative effects on MM and 5q MDS cell lines than lenalidomide. PROTACs using these lenalidomide derivatives for BET proteins induce the selective degradation of BET proteins with the same neosubstrate selectivity. PROTACs also exert anti-proliferative effects in all examined cell lines. Thus, 6-position-modified lenalidomide is a key molecule for selective TPD using thalidomide derivatives and PROTACs.
Collapse
Affiliation(s)
- Satoshi Yamanaka
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan
- Division of Proteo-Interactome, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan
| | - Hirotake Furihata
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Yuta Yanagihara
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, 791-0295, Japan
| | - Akihito Taya
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Takato Nagasaka
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Mai Usui
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Koya Nagaoka
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan
| | - Yuki Shoya
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan
| | - Kohei Nishino
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, 770-8503, Japan
| | - Shuhei Yoshida
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, 791-0295, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Tokushima University, Tokushima, 770-8503, Japan
| | - Masaru Tanokura
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Takuya Miyakawa
- Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, 113-8657, Japan
- Graduate School of Biostudies, Kyoto University, Kyoto, 606-8502, Japan
| | - Yuuki Imai
- Division of Integrative Pathophysiology, Proteo-Science Center, Ehime University, Toon, 791-0295, Japan
| | - Norio Shibata
- Department of Life Science and Applied Chemistry, Nagoya Institute of Technology, Nagoya, 466-8555, Japan
| | - Tatsuya Sawasaki
- Division of Cell-Free Sciences, Proteo-Science Center, Ehime University, Matsuyama, 790-8577, Japan.
| |
Collapse
|
43
|
Garcia-Manero G. Myelodysplastic syndromes: 2023 update on diagnosis, risk-stratification, and management. Am J Hematol 2023; 98:1307-1325. [PMID: 37288607 PMCID: PMC12002404 DOI: 10.1002/ajh.26984] [Citation(s) in RCA: 53] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 05/21/2023] [Accepted: 05/23/2023] [Indexed: 06/09/2023]
Abstract
DISEASE OVERVIEW The myelodysplastic syndromes (MDS) are a very heterogeneous group of myeloid disorders characterized by peripheral blood cytopenias and increased risk of transformation to acute myelogenous leukemia (AML). MDS occurs more frequently in older males and in individuals with prior exposure to cytotoxic therapy. DIAGNOSIS Diagnosis of MDS is based on morphological evidence of dysplasia upon visual examination of a bone marrow aspirate and biopsy. Information obtained from additional studies such as karyotype, flow cytometry, and molecular genetics is usually complementary and may help refine diagnosis. A new WHO classification of MDS was proposed in 2022. Under this classification, MDS is now termed myelodysplastic neoplasms. RISK-STRATIFICATION Prognosis of patients with MDS can be calculated using a number of scoring systems. All these scoring systems include analysis of peripheral cytopenias, percentage of blasts in the bone marrow, and cytogenetic characteristics. The most commonly accepted system is the Revised International Prognostic Scoring System (IPSS-R). Recently, genomic data has been incorporated resulting in the new IPSS-M classification. RISK-ADAPTED THERAPY Therapy is selected based on risk, transfusion needs, percent of bone marrow blasts, cytogenetic and mutational profiles, comorbidities, potential for allogeneic stem cell transplantation (alloSCT), and prior exposure to hypomethylating agents (HMA). Goals of therapy are different in lower risk patients than in higher risk and in those with HMA failure. In lower risk, the goal is to decrease transfusion needs and transformation to higher risk disease or AML, as well as to improve survival. In higher risk, the goal is to prolong survival. In 2020, two agents were approved in the US for patients with MDS: luspatercept and oral decitabine/cedazuridine. In addition, currently other available therapies include growth factors, lenalidomide, HMAs, intensive chemotherapy, and alloSCT. A number of phase 3 combinations studies have been completed or are ongoing at the time of this report. At the present time there are no approved interventions for patients with progressive or refractory disease particularly after HMA based therapy. In 2021, several reports indicated improved outcomes with alloSCT in MDS as well as early results from clinical trials using targeted intervention.
Collapse
Affiliation(s)
- Guillermo Garcia-Manero
- Section of MDS, Department of Leukemia, University of Texas MD Anderson Cancer Center, Houston, Texas, United States
| |
Collapse
|
44
|
Gerke MB, Christodoulou I, Karantanos T. Definitions, Biology, and Current Therapeutic Landscape of Myelodysplastic/Myeloproliferative Neoplasms. Cancers (Basel) 2023; 15:3815. [PMID: 37568631 PMCID: PMC10417399 DOI: 10.3390/cancers15153815] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Revised: 07/24/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
Myelodysplastic/myeloproliferative neoplasms (MDS/MPN) are hematological disorders characterized by both proliferative and dysplastic features. According to the 2022 International Consensus Classification (ICC), MDS/MPN consists of clonal monocytosis of undetermined significance (CMUS), chronic myelomonocytic leukemia (CMML), atypical chronic myeloid leukemia (aCML), MDS/MPN with SF3B1 mutation (MDS/MPN-T-SF3B1), MDS/MPN with ring sideroblasts and thrombocytosis not otherwise specified (MDS/MPN-RS-T-NOS), and MDS/MPN-NOS. These disorders exhibit a diverse range of genetic alterations involving various transcription factors (e.g., RUNX1), signaling molecules (e.g., NRAS, JAK2), splicing factors (e.g., SF3B, SRSF2), and epigenetic regulators (e.g., TET2, ASXL1, DNMT3A), as well as specific cytogenetic abnormalities (e.g., 8 trisomies, 7 deletions/monosomies). Clinical studies exploring therapeutic options for higher-risk MDS/MPN overlap syndromes mostly involve hypomethylating agents, but other treatments such as lenalidomide and targeted agents such as JAK inhibitors and inhibitors targeting PARP, histone deacetylases, and the Ras pathway are under investigation. While these treatment modalities can provide partial disease control, allogeneic bone marrow transplantation (allo-BMT) is the only potentially curative option for patients. Important prognostic factors correlating with outcomes after allo-BMT include comorbidities, splenomegaly, karyotype alterations, and the bone marrow blasts percentage at the time of transplantation. Future research is imperative to optimizing therapeutic strategies and enhancing patient outcomes in MDS/MPN neoplasms. In this review, we summarize MDS/MPN diagnostic criteria, biology, and current and future treatment options, including bone marrow transplantation.
Collapse
Affiliation(s)
- Margo B. Gerke
- School of Medicine, Emory University, Atlanta, GA 30322, USA;
| | - Ilias Christodoulou
- Department of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | | |
Collapse
|
45
|
Frumm SM, Shimony S, Stone RM, DeAngelo DJ, Bewersdorf JP, Zeidan AM, Stahl M. Why do we not have more drugs approved for MDS? A critical viewpoint on novel drug development in MDS. Blood Rev 2023; 60:101056. [PMID: 36805300 DOI: 10.1016/j.blre.2023.101056] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2022] [Revised: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 02/16/2023]
Abstract
Approval of new agents to treat higher risk (HR) myelodysplastic syndrome (MDS) has stalled since the approval of DNA methyltransferase inhibitors (DNMTi). In addition, the options for patients with lower risk (LR) MDS who have high transfusion needs and do not harbor ring sideroblasts or 5q- syndrome are limited. Here, we review the current treatment landscape in MDS and identify areas of unmet need, such as treatment after failure of erythropoiesis-stimulating agents or DNMTis, TP53-mutated disease, and MDS with potentially targetable mutations. We discuss how our understanding of MDS pathogenesis can inform therapy development, including treating HR-MDS similarly to AML and pursuing therapies to address splicing factor mutations and dysregulated inflammation. We then bring a critical lens to current methodology of MDS studies and propose solutions to improve the efficiency and yield of these clinical trials, including using the most meaningful response metrics and expanding enrollment.
Collapse
Affiliation(s)
- Stacey M Frumm
- Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Shai Shimony
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA; Rabin Medical Center and Faculty of Medicine, Tel Aviv University, Israel
| | - Richard M Stone
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Daniel J DeAngelo
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jan Phillipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, and Yale Cancer Center, Yale University, New Haven, CT, USA
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| |
Collapse
|
46
|
Venugopal S, Shallis RM, Zeidan AM. Oral therapy for myelodysplastic syndromes/neoplasms and acute myeloid leukemia: a revolution in progress. Expert Rev Anticancer Ther 2023; 23:903-911. [PMID: 37470508 DOI: 10.1080/14737140.2023.2238897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 07/17/2023] [Indexed: 07/21/2023]
Abstract
INTRODUCTION Patients with myeloid neoplasms such as myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukemia (AML) are generally older, and many are not eligible for curative intent intensive therapies and/or allogeneic hematopoietic stem cell transplantation. While lower intensity, hypomethylating agent (HMA)-based therapies such as azacitidine+venetoclax have improved patient outcomes significantly, responses are not durable, and most patients die from disease-related complications. The approvals of oral HMAs such as cedazuridine-decitabine (C-DEC) and oral azacitidine (CC-486) have kindled the hope that myeloid malignancies may soon be treated with total oral therapy. AREAS COVERED We review all-oral therapies including the approvals of C-DEC and CC-486 in MDS and AML, respectively, in addition to emerging all-oral therapies, both monotherapy and combination, in higher-risk (HR) MDS and AML. EXPERT OPINION Oral HMAs have the potential to be a convenient and efficacy-equivalent treatment option for patients with HR-MDS or AML and improve their quality of life by reducing clinic visits for medication administration. Total-oral therapy combinations, largely including an oral HMA 'backbone,' are in the early phases of clinical development, and it is our hope that well-designed trials employing these agents may soon allow the identification of optimal regimens that deliver effective disease-directed therapy with good tolerability.
Collapse
Affiliation(s)
- Sangeetha Venugopal
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Rory M Shallis
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
47
|
Lucero J, Al-Harbi S, Yee KWL. Management of Patients with Lower-Risk Myelodysplastic Neoplasms (MDS). Curr Oncol 2023; 30:6177-6196. [PMID: 37504319 PMCID: PMC10377892 DOI: 10.3390/curroncol30070459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/15/2023] [Accepted: 06/25/2023] [Indexed: 07/29/2023] Open
Abstract
Myelodysplastic neoplasms (MDS) are a heterogenous group of clonal hematologic disorders characterized by morphologic dysplasia, ineffective hematopoiesis, and cytopenia. In the past year, the classification of MDS has been updated in the 5th edition of the World Health Organization (WHO) Classification of Haematolymphoid Tumours and the International Consensus Classification (ICC) of Myeloid Neoplasms and Acute Leukemia with incorporation of morphologic, clinical, and genomic data. Furthermore, the more comprehensive International Prognostic Scoring System-Molecular (IPSS-M) allows for improved risk stratification and prognostication. These three developments allow for more tailored therapeutic decision-making in view of the expanding treatment options in MDS. For patients with lower risk MDS, treatment is aimed at improving cytopenias, usually anemia. The recent approval of luspatercept and decitabine/cedazuridine have added on to the current armamentarium of erythropoietic stimulating agents and lenalidomide (for MDS with isolated deletion 5q). Several newer agents are being evaluated in phase 3 clinical trials for this group of patients, such as imetelstat and oral azacitidine. This review provides a summary of the classification systems, the prognostic scores and clinical management of patients with lower risk MDS.
Collapse
Affiliation(s)
- Josephine Lucero
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, 700 University Avenue, 6th Floor, Toronto, ON M5G 1Z5, Canada; (J.L.); (S.A.-H.)
| | - Salman Al-Harbi
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, 700 University Avenue, 6th Floor, Toronto, ON M5G 1Z5, Canada; (J.L.); (S.A.-H.)
| | - Karen W. L. Yee
- Division of Medical Oncology and Hematology, Princess Margaret Cancer Centre, University Health Network, 700 University Avenue, 6th Floor, Toronto, ON M5G 1Z5, Canada; (J.L.); (S.A.-H.)
- Division of Hematology, University of Toronto, Toronto, ON M5S 3H2, Canada
| |
Collapse
|
48
|
Stempel JM, Xie Z, Bewersdorf JP, Stahl M, Zeidan AM. Evolution of Therapeutic Benefit Measurement Criteria in Myelodysplastic Syndromes/Neoplasms. Cancer J 2023; 29:203-211. [PMID: 37195777 DOI: 10.1097/ppo.0000000000000666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDS) are heterogeneous, clonal myeloid neoplasms characterized by ineffective hematopoiesis, progressive cytopenias, and an increased risk of progression to acute myeloid leukemia. The diversity in disease severity, morphology, and genetic landscape challenges not only novel drug development but also therapeutic response assessment. The MDS International Working Group (IWG) response criteria were first published in the year 2000 focusing on measures of blast burden reduction and hematologic recovery. Despite revision of the IWG criteria in 2006, correlation between IWG-defined responses and patient-focused outcomes, including long-term benefits, remains limited and has potentially contributed to failures of several phase III clinical trials. Several IWG 2006 criteria also lacked clear definitions leading to problems in practical applications and interobserver and intraobserver consistency of response reporting. Although the 2018 revision addressed lower-risk MDS, the most recent update in 2023 redefined responses for higher-risk MDS and has set out to provide clear definitions to enhance consistency while focusing on clinically meaningful outcomes and patient-centered responses. In this review, we analyze the evolution of the MDS response criteria, limitations, and areas of improvement.
Collapse
Affiliation(s)
- Jessica M Stempel
- From the Department of Internal Medicine, Hematology Section, Yale School of Medicine, New Haven, CT
| | - Zhuoer Xie
- Department of Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Harvard University, Boston, MA
| | - Amer M Zeidan
- From the Department of Internal Medicine, Hematology Section, Yale School of Medicine, New Haven, CT
| |
Collapse
|
49
|
Barone P, Patel S. Myelodysplastic syndrome: Approach to diagnosis in the era of personalized medicine. Semin Diagn Pathol 2023; 40:172-181. [PMID: 37121781 DOI: 10.1053/j.semdp.2023.04.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2023] [Revised: 04/11/2023] [Accepted: 04/12/2023] [Indexed: 05/02/2023]
Affiliation(s)
- Paul Barone
- NewYork-Presbyterian Hospital, Weill Cornell Campus, United States of America.
| | - Sanjay Patel
- Weill Cornell Medicine, United States of America
| |
Collapse
|
50
|
Wang C, Sallman DA. Current Therapeutic Landscape in Lower Risk Myelodysplastic Syndromes. Curr Treat Options Oncol 2023; 24:387-408. [PMID: 36966266 DOI: 10.1007/s11864-023-01062-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/19/2023] [Indexed: 03/27/2023]
Abstract
OPINION STATEMENT Lower risk myelodysplastic syndromes are typically characterized by an indolent disease course with a relatively low risk of transformation into acute myeloid leukemia. These patients are classically identified using the revised International Prognostic Scoring System and most likely its molecular version in the near future which may change the paradigm of treatment. The overall goals of care are symptomatic control to reduce transfusion requirements and improve quality of life. Symptomatic anemia is the most common indication to initiate disease-specific therapies after the optimization of supportive measures. Currently, erythropoiesis-stimulating agents remain the standard upfront therapy for anemia, and patients with del(5q) cytogenetic changes can benefit from lenalidomide monotherapy. Other therapeutic options after failure of upfront treatment include luspatercept, hypomethylating agents, and immunosuppressive therapies after taking into account of individualized disease features. Allogeneic hematopoietic stem cell transplant is the only potentially curative option and is usually reserved for medically fit patients with severe symptomatic cytopenias who failed all standard options and/or the disease is progressing toward higher risk categories. Fortunately, novel investigational therapies are rapidly emerging by targeting different biological processes contributing to MDS pathogenesis, and eligible patients should be managed in clinical trials if available.
Collapse
Affiliation(s)
- Chen Wang
- Department of Internal Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL, 33612, USA.
| |
Collapse
|