1
|
Borate U, Pugh K, Waller A, Welkie RL, Huang Y, Bewersdorf JP, Stahl M, DeZern AE, Platzbecker U, Sekeres MA, Wei AH, Buckstein RJ, Roboz GJ, Savona MR, Loghavi S, Hasserjian RP, Fenaux P, Sallman DA, Hourigan CS, Della Porta MG, Nimer S, Little RF, Santini V, Efficace F, Taylor J, Garcia-Manero G, Odenike O, Kim TK, Halene S, Komrokji RS, Griffiths EA, Greenberg PL, Xu ML, Xie Z, Bejar R, Sanz GF, Patnaik MM, Figueroa M, Carraway HE, Abdel-Wahab O, Starczynowski D, Padron E, Boultwood J, Gore S, Daver NG, Churpek JE, Majeti R, Bennett JM, List AF, Brunner AM, Zeidan AM. Reducing clinical trial eligibility barriers for patients with MDS: an icMDS position statement. Blood 2025; 145:1369-1381. [PMID: 40146152 PMCID: PMC11969261 DOI: 10.1182/blood.2023023717] [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: 05/20/2024] [Accepted: 11/14/2024] [Indexed: 03/28/2025] Open
Abstract
ABSTRACT Excessively restrictive inclusion and exclusion criteria in clinical trials are one of many barriers to clinical trial enrollment for patients with myelodysplastic syndromes/neoplasms (MDSs). Many organizations are developing efforts to increase clinical trial eligibility; yet, several recent publications focused on patients with MDS suggest that many patients with this disease may be excluded from clinical trials unnecessarily. Clinical trial eligibility should reflect the phase of the study and risks of the agent being studied. Phase 3 trials should be less restrictive than early-phase trials to represent the real-world population as closely as possible. We hypothesize that many clinical trials, particularly phase 3 trials, have unnecessarily restrictive eligibility criteria. This study aims to evaluate the most common eligibility criteria according to phase of trial and to determine whether criteria correspond with drug safety signals. We identified MDS clinical trials registered on ClinicalTrials.gov from 1 January 2000 to 1 September 2023 and analyzed the eligibility criteria of 191 therapeutic MDS trials. We found that categorical inclusion and exclusion criteria are remarkably similar in representation across trial phases. Additionally, only 13% of trials are concordant with drug safety signals, suggesting that the eligibility criteria are often arbitrary. On behalf of the icMDS (International Consortium for Myelodysplastic Syndromes), an association of international MDS experts, we provide a position statement on restrictive eligibility criteria for MDS clinical trials that should be avoided with the aim of removing barriers to clinical trial enrollment.
Collapse
Affiliation(s)
- Uma Borate
- Division of Hematology, The Ohio State University Comprehensive Cancer Center/James Cancer Hospital, The Ohio State University, Columbus, OH
| | - Kelly Pugh
- Division of Hematology, The Ohio State University Comprehensive Cancer Center/James Cancer Hospital, The Ohio State University, Columbus, OH
| | - Allyson Waller
- Division of Hematology, The Ohio State University Comprehensive Cancer Center/James Cancer Hospital, The Ohio State University, Columbus, OH
| | - Rina Li Welkie
- Division of Hematology, The Ohio State University Comprehensive Cancer Center/James Cancer Hospital, The Ohio State University, Columbus, OH
| | - Ying Huang
- Division of Hematology, The Ohio State University Comprehensive Cancer Center/James Cancer Hospital, The Ohio State University, Columbus, OH
| | - Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA
| | - Amy E. DeZern
- Division of Hematology Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD
| | - Uwe Platzbecker
- Section of Hematology, Leipzig University Hospital, Leipzig, Germany
| | - Mikkael A. Sekeres
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Andrew H. Wei
- Department of Haematology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital, Walter and Eliza Hall Institute of Medical Research, The University of Melbourne, VIC, Australia
| | - Rena J. Buckstein
- Department of Medical Oncology/Hematology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Gail J. Roboz
- Division of Hematology and Medical Oncology, Weill Cornell Medical College and New York Presbyterian Hospital, New York, NY
| | - Michael R. Savona
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Pierre Fenaux
- Hematology Department, Hôpital Saint Louis, Assistance Publique Hôpitaux de Paris and Paris Cité University, Paris, France
| | - David A. Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Christopher S. Hourigan
- Fralin Biomedical Research Institute, Virginia Tech Fralin Biomedical Research Institute Cancer Research Center, Washington, DC
| | - Matteo Giovanni Della Porta
- Department of Biomedical Sciences, IRCCS Humanitas Clinical and Research Center and Humanitas University, Milan, Italy
| | - Stephen Nimer
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Richard F. Little
- National Cancer Institute, Cancer Therapy Evaluation Program, Rockville, MD
| | - Valeria Santini
- Myelodysplastic Syndromes Unit, Hematology, Azienda Ospedaliera Universitaria Careggi, University of Florence, Italy
| | - Fabio Efficace
- Italian Group for Adult Hematologic Diseases, Health Outcomes Research Unit, Rome, Italy
| | - Justin Taylor
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | | | - Olatoyosi Odenike
- Section of Hematology/Oncology, Leukemia Program, University of Chicago Medicine and University of Chicago Comprehensive Cancer Center, Chicago, IL
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT
| | - Rami S. Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | | | - Peter L. Greenberg
- Division of Hematology, Department of Medicine, Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - Mina L. Xu
- Departments of Pathology and Laboratory Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Rafael Bejar
- Division of Hematology and Oncology, Moores Cancer Center, La Jolla, CA
| | - Guillermo F. Sanz
- Hematology Department, Hospital Universitario y Politécnico La Fe, Valencia, Spain
- Health Research Institute La Fe, Valencia, Spain
- Centro de Investigación Biomédica en Red de Cáncer, Instituto de Salud Carlos III, Madrid, Spain, Valencia, Spain
| | - Mrinal M. Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN
| | - Maria Figueroa
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | - Hetty E. Carraway
- Division of Hematologic Oncology and Blood Disorders, Leukemia Program, Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH
| | - Omar Abdel-Wahab
- Department of Medicine, Molecular Pharmacology Program, Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Daniel Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH
| | - Eric Padron
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Jacqueline Boultwood
- Blood Cancer UK Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Steven Gore
- National Cancer Institute, Cancer Therapy Evaluation Program, Rockville, MD
| | - Naval G. Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jane E. Churpek
- Department of Hematology, Oncology, and Palliative Care, Carbone Cancer Center, University of Wisconsin-Madison, Madison, WI
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Cancer Institute, Stanford University School of Medicine, Stanford, CA
| | - John M. Bennett
- Department of Pathology and Laboratory Medical Center, University of Rochester Medical Center, Rochester, NY
| | | | - Andrew M. Brunner
- Division of Hematology and Oncology, Leukemia Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT
| |
Collapse
|
2
|
Teymouri F, Dasanu CA. Selecting optimal therapy for higher-risk myelodysplastic syndromes: present and future projections. Expert Opin Pharmacother 2025; 26:345-347. [PMID: 39956939 DOI: 10.1080/14656566.2025.2468467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/10/2025] [Accepted: 02/13/2025] [Indexed: 02/18/2025]
Affiliation(s)
- Farzad Teymouri
- Department of Medicine, Eisenhower Health, Rancho Mirage, CA, USA
| | - Constantin A Dasanu
- Lucy Curci Cancer Center, Eisenhower Health, Rancho Mirage, CA, USA
- Department of Medical Oncology and Hematology, UC San Diego Health System, San Diego, CA, USA
| |
Collapse
|
3
|
Nikolopoulos T, Bochalis E, Chatzilygeroudi T, Chondrou V, Dereki I, Athanasopoulou K, Zafeiropoulos J, Bourikas K, Patrinos GP, Symeonidis A, Sgourou A. Integrating advanced analytical methods to assess epigenetic marks affecting response to hypomethylating agents in higher risk myelodysplastic syndrome. Mol Med 2025; 31:59. [PMID: 39953389 PMCID: PMC11829487 DOI: 10.1186/s10020-025-01123-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 02/08/2025] [Indexed: 02/17/2025] Open
Abstract
BACKGROUND Patients with higher-risk (HR) myelodysplastic syndrome (MDS), ineligible for allogeneic hematopoietic stem cell transplantation (alloHSCT), require prompt therapeutic interventions, such as treatment with hypomethylating agents (HMAs) to restore normal DNA methylation patterns, mainly of oncosuppressor genes, and consequently to delay disease progression and increase overall survival (OS). However, response assessment to HMA treatment relies on conventional methods with limited capacity to uncover a wide spectrum of underlying molecular events. METHODS We implemented liquid chromatography-tandem mass spectrometry (LC-MS/MS) to assess 5' methyl-2' deoxycytidine (5mdC), 5' hydroxy-methyl-2'-deoxycytidine (5hmdC) levels and global adenosine/thymidine ([dA]/[T]) ratio in bone marrow aspirates from twenty-one HR MDS patients, pre- and post-HMA treatment. Additionally, targeted methylation analysis was performed by interpretation of NGS-methylation (MeD-seq) data obtained from the same patient cohort. RESULTS LC/MS-MS analysis revealed a significant hypomethylation status in responders (Rs), already established at baseline and a trend for further DNA methylation reduction post-HMA treatment. Non-responders (NRs) reached statistical significance for DNA hypomethylation only post-HMA treatment. The 5hmdC epigenetic mark was approximately detected at 37.5-40% among NRs and Rs, implying the impairment of the natural active demethylation pathway, mediated by the ten-eleven (TET) 5mdC dioxygenases. R and NR subgroups displayed a [dA]/[T] ratio < 1 (0.727 - 0.633), supporting high frequences of 5mdC transition to thymidine. Response to treatment, according to whole genome MeD-seq data analysis, was associated with specific, scattered hypomethylated DMRs, rather than presenting a global effect across genome. MeD-seq analysis identified divergent epigenetic effects along chromosomes 7, 9, 12, 16, 18, 21, 22, X and Y. Within statistically significant selected chromosomal bins, genes encoding for proteins and non-coding RNAs with reversed methylation profiles between Rs and NRs, were highlighted. CONCLUSIONS Implementation of powerful analytical tools to identify the dynamic DNA methylation changes in HR MDS patients undergoing HMA therapy demonstrated that LC-MS/MS exerts high efficiency as a broad-based but rapid and cost-effective methodology (compared to MeD-seq) to decode different perspectives of the epigenetic background of HR MDS patients and possess discriminative efficacy of the response phenotype to HMA treatment.
Collapse
Affiliation(s)
- Theodoros Nikolopoulos
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Eleftherios Bochalis
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Theodora Chatzilygeroudi
- School of Health Sciences, Faculty of Medicine, Hematology Division, University of Patras, Patras, Greece
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Baltimore, MD, USA
| | - Vasiliki Chondrou
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Irene Dereki
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Katerina Athanasopoulou
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - John Zafeiropoulos
- Chemistry Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - Kyriakos Bourikas
- Chemistry Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece
| | - George P Patrinos
- Laboratory of Pharmacogenomics and Inaffiliationidualized Therapy, Department of Pharmacy, School of Health Sciences, University of Patras, University Campus, Rion, Patras, Greece
- College of Medicine and Health Sciences, Department of Genetics and Genomics, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Zayed Center for Health Sciences, United Arab Emirates University, Al Ain, Abu Dhabi, UAE
- Clinical Bioinformatics Unit, Department of Pathology, Faculty of Medicine and Health Sciences, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Argiris Symeonidis
- School of Health Sciences, Faculty of Medicine, Hematology Division, University of Patras, Patras, Greece.
| | - Argyro Sgourou
- Biology Laboratory, School of Science and Technology, Hellenic Open University, Patras, Greece.
| |
Collapse
|
4
|
Getz TM, Bewersdorf JP, Kewan T, Stempel JM, Bidikian A, Shallis RM, Stahl M, Zeidan AM. Beyond HMAs: Novel Targets and Therapeutic Approaches. Semin Hematol 2024; 61:358-369. [PMID: 39389839 DOI: 10.1053/j.seminhematol.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024]
Abstract
Myelodysplastic syndromes/neoplasms (MDS) constitute a heterogeneous group of clonal hematopoietic disorders with extremely variable clinical features and outcomes. Management of MDS is largely based on risk stratification of patients into either lower-risk or higher-risk categories using the International Prognostic Scoring System-Revised and, more recently, on the Molecular International Prognostic Scoring System. Lower-risk MDS is often managed with the goal of ameliorating cytopenias and improving quality of life, while higher-risk MDS is treated with therapies aimed at extending survival and delaying progression to acute myeloid leukemia (AML). Therapeutic strategies in lower-risk MDS patients may consist of erythropoiesis stimulating agents, luspatercept, and lenalidomide for selected patients. Furthermore, imetelstat has recently been added to the FDA-approved therapeutic armamentarium for lower-risk MDS. In higher-risk MDS, monotherapy with hypomethylating agents continues to be the standard of care. While several novel hypomethylating agent combinations have and are being studied in large randomized phase 3 clinical trials, including the combination of azacitidine and venetoclax, no combination to date have improved overall survival to azacitidine monotherapy. Moreover, biomarker-directed therapies as well as immonotherapeutic approaches are currently being evaluated in early phase trials. Despite recent advancements, the lack of therapeutic agents, particularly after the failure of first line therapy in higher risk MDS, continues to be a major hurdle in the management of MDS. In this review, we discuss the current treatment landscape of MDS and provide an overview of novel agents currently in clinical development that have the potential to alter our current treatment paradigms.
Collapse
Affiliation(s)
- Ted M Getz
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut.
| | - Jan P Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut; Department of Medicine, Memorial Sloan Kettering Cancer Center, Leukemia Service, New York, New York
| | - Tariq Kewan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Jessica M Stempel
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Aram Bidikian
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Rory M Shallis
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| |
Collapse
|
5
|
Chien KS, Rodriguez-Sevilla JJ, Alvarado Y, Montalban-Bravo G, Hammond DE, Swaminathan M, Bazinet A, Kimberley J, Bodden K, Schneider H, Dong XQ, Pierce SA, Huang X, Jabbour EJ, Kantarjian HM, Garcia-Manero G. A phase I study of the myeloid cell leukemia 1 (MCL1) inhibitor tapotoclax (AMG 176) in patients with myelodysplastic syndromes after hypomethylating agent failure. Leuk Res 2024; 147:107602. [PMID: 39461095 DOI: 10.1016/j.leukres.2024.107602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 10/11/2024] [Accepted: 10/21/2024] [Indexed: 10/29/2024]
Affiliation(s)
- Kelly S Chien
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA.
| | | | - Yesid Alvarado
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | | | - Danielle E Hammond
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Mahesh Swaminathan
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Alexandre Bazinet
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Jacqueline Kimberley
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Kristy Bodden
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Heather Schneider
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Xiao Qin Dong
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Sherry A Pierce
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Xuelin Huang
- The University of Texas MD Anderson Cancer Center, Department of Biostatistics, Houston, TX, USA
| | - Elias J Jabbour
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | - Hagop M Kantarjian
- The University of Texas MD Anderson Cancer Center, Department of Leukemia, Houston, TX, USA
| | | |
Collapse
|
6
|
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
|
7
|
Zhang L, Deeb G, Deeb KK, Vale C, Peker Barclift D, Papadantonakis N. Measurable (Minimal) Residual Disease in Myelodysplastic Neoplasms (MDS): Current State and Perspectives. Cancers (Basel) 2024; 16:1503. [PMID: 38672585 PMCID: PMC11048433 DOI: 10.3390/cancers16081503] [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: 02/17/2024] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/28/2024] Open
Abstract
Myelodysplastic Neoplasms (MDS) have been traditionally studied through the assessment of blood counts, cytogenetics, and morphology. In recent years, the introduction of molecular assays has improved our ability to diagnose MDS. The role of Measurable (minimal) Residual Disease (MRD) in MDS is evolving, and molecular and flow cytometry techniques have been used in several studies. In this review, we will highlight the evolving concept of MRD in MDS, outline the various techniques utilized, and provide an overview of the studies reporting MRD and the correlation with outcomes.
Collapse
Affiliation(s)
- Linsheng Zhang
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - George Deeb
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Kristin K. Deeb
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Colin Vale
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| | - Deniz Peker Barclift
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Nikolaos Papadantonakis
- Department of Hematology and Medical Oncology, Winship Cancer Institute of Emory University, Atlanta, GA 30322, USA
| |
Collapse
|
8
|
Bewersdorf JP, Shallis RM, Sharon E, Park S, Ramaswamy R, Roe CE, Irish JM, Caldwell A, Wei W, Yacoub A, Madanat YF, Zeidner JF, Altman JK, Odenike O, Yerrabothala S, Kovacsovics T, Podoltsev NA, Halene S, Little RF, Piekarz R, Gore SD, Kim TK, Zeidan AM. A multicenter phase Ib trial of the histone deacetylase inhibitor entinostat in combination with pembrolizumab in patients with myelodysplastic syndromes/neoplasms or acute myeloid leukemia refractory to hypomethylating agents. Ann Hematol 2024; 103:105-116. [PMID: 38036712 PMCID: PMC11838822 DOI: 10.1007/s00277-023-05552-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023]
Abstract
Patients with myelodysplastic syndromes/neoplasms (MDS) or acute myeloid leukemia (AML) with hypomethylating agent failure have a poor prognosis. Myeloid-derived suppressor cells (MDSCs) can contribute to MDS progression and mediate resistance to anti-PD1 therapy. As histone deacetylase inhibitors (HDACi) decrease MDSCs in preclinical models, we conducted an investigator-initiated, NCI-Cancer Therapy Evaluation Program-sponsored, multicenter, dose escalation, and expansion phase Ib trial (NCT02936752) of the HDACi entinostat and the anti-PD1 antibody pembrolizumab. Twenty-eight patients (25 MDS and 3 AML) were enrolled. During dose escalation (n=13 patients), there was one dose-limiting toxicity (DLT) on dose level (DL) 1 (G5 pneumonia/bronchoalveolar hemorrhage) and two DLTs at DL 2 (G3 pharyngeal mucositis and G3 anorexia). Per the 3 + 3 dose escalation design, DL 1 (entinostat 8 mg PO days 1 and 15 + pembrolizumab 200 mg IV day 1 every 21 days) was expanded and another 15 patients were enrolled. Hematologic adverse events (AEs) were common. The most common non-hematologic ≥G3 AEs were infection (32%), hypoxia/respiratory failure (11%), and dyspnea (11%). There were no protocol-defined responses among the 28 patients enrolled. Two patients achieved a marrow complete remission (mCR). Using a systems immunology approach with mass cytometry and machine learning analysis, mCR patients had increased classical monocytes and macrophages but there was no significant change of MDSCs. In conclusion, combining entinostat with pembrolizumab in patients with advanced MDS and AML was associated with limited clinical efficacy and substantial toxicity. Absence of an effect on MDSCs could be a potential explanation for the limited efficacy of this combination. ClinicalTrial.gov Identifier: NCT02936752.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Elad Sharon
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Silvia Park
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rahul Ramaswamy
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Caroline E Roe
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN, USA
| | - Jonathan M Irish
- Department of Cell and Developmental Biology, Vanderbilt University, Nashville, TN, USA
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN, USA
| | - Anne Caldwell
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Wei Wei
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Abdulraheem Yacoub
- The Division of Hematologic Malignancies and Cellular Therapeutics (HMCT), The University of Kansas Cancer Center, Westwood, KS, USA
| | - Yazan F Madanat
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
| | - Joshua F Zeidner
- Lineberger Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Jessica K Altman
- Robert H. Lurie Comprehensive Cancer Center of Northwestern University, Chicago, IL, USA
| | | | | | | | - Nikolai A Podoltsev
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Richard F Little
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Richard Piekarz
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Steven D Gore
- Cancer Therapy Evaluation Program, National Cancer Institute, Bethesda, MD, USA
| | - Tae Kon Kim
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Division of Hematology/Oncology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN, USA.
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale Cancer Center, Yale School of Medicine, Yale University, New Haven, CT, USA.
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, Yale University, 333 Cedar Street, PO Box 208028, New Haven, CT, 06520-8028, USA.
| |
Collapse
|
9
|
Brunner AM, Platzbecker U, DeZern AE, Zeidan AM. Are We Ready For "Triplet" Therapy in Higher-Risk MDS? Clin Hematol Int 2023; 5:88301. [PMID: 37933301 PMCID: PMC10625655 DOI: 10.46989/001c.88301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2023] [Accepted: 08/26/2023] [Indexed: 11/08/2023] Open
Abstract
Higher-risk Myelodysplastic Syndromes/Neoplasms (MDS) represent an ongoing therapeutic challenge, with few effective therapies, many of which may have limited use in this older patient population often with considerations around comorbidities. Outside of transplant, azacitidine and decitabine remain the only disease-modifying therapies, and are palliative in nature. Recent interest has grown in extending combination chemotherapies used to treat acute myeloid leukemia (AML) to patients with MDS, including novel combination chemotherapy "doublets" and "triplets." In this review, we discuss considerations around combination chemotherapy in MDS, specifically as relates to study design, appropriate endpoints, supportive considerations, and how to integrate these into the current treatment paradigm. New therapies in MDS are desperately needed but also require considerations particular to this unique patient population.
Collapse
Affiliation(s)
- Andrew M Brunner
- Leukemia Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | | | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Centre at John Hopkins, Baltimore, MD, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| |
Collapse
|
10
|
Sharplin K, Proudman W, Chhetri R, Tran ENH, Choong J, Kutyna M, Selby P, Sapio A, Friel O, Khanna S, Singhal D, Damin M, Ross D, Yeung D, Thomas D, Kok CH, Hiwase D. A Personalized Risk Model for Azacitidine Outcome in Myelodysplastic Syndrome and Other Myeloid Neoplasms Identified by Machine Learning Model Utilizing Real-World Data. Cancers (Basel) 2023; 15:4019. [PMID: 37627047 PMCID: PMC10452100 DOI: 10.3390/cancers15164019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2023] [Revised: 08/04/2023] [Accepted: 08/06/2023] [Indexed: 08/27/2023] Open
Abstract
Azacitidine is an approved therapy for higher-risk myelodysplastic syndrome (MDS). However, only 30-40% patients respond to azacitidine, and the responses may take up to six cycles to become evident. Delayed responses and the myelosuppressive effects of azacitidine make it challenging to predict which patients will benefit. This is further compounded by a lack of uniform prognostic tools to identify patients at risk of early treatment failure. Hence, we performed a retrospective analysis of 273 consecutive azacytidine-treated patients. The median overall survival was 16.25 months with only 9% alive at 5 years. By using pre-treatment variables incorporated into a random forest machine learning model, we successfully identified those patients unlikely to benefit from azacytidine upfront (7.99 vs. 22.8 months, p < 0.0001). This model also identified those who required significantly more hospitalizations and transfusion support. Notably, it accurately predicted survival outcomes, outperforming the existing prognostic scoring system. By integrating somatic mutations, we further refined the model and identified three distinct risk groups with significant differences in survival (5.6 vs. 10.5 vs. 43.5 months, p < 0.0001). These real-world findings emphasize the urgent need for personalized prediction tools tailored to hypomethylating agents, reducing unnecessary complications and resource utilization in MDS treatment.
Collapse
Affiliation(s)
- Kirsty Sharplin
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - William Proudman
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - Rakchha Chhetri
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Elizabeth Ngoc Hoa Tran
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Jamie Choong
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - Monika Kutyna
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Philip Selby
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Aidan Sapio
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - Oisin Friel
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Beaumont Hospital, D09 V2N0 Dublin, Ireland
| | - Shreyas Khanna
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Deepak Singhal
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
| | - Michelle Damin
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
| | - David Ross
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
- Genetic and Molecular Pathology, SA Pathology, Adelaide, SA 5000, Australia
| | - David Yeung
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Daniel Thomas
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Chung H. Kok
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| | - Devendra Hiwase
- Royal Adelaide Hospital, Central Adelaide Local Health Network, Adelaide, SA 5000, Australia
- Precision Medicine Theme, South Australian Health and Medical Research Institute (SAHMRI), Adelaide, SA 5000, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA 5000, Australia
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA 5000, Australia
| |
Collapse
|
11
|
Karantanos T, Teodorescu P, Arvanitis M, Perkins B, Jain T, DeZern AE, Dalton WB, Christodoulou I, Paun BC, Varadhan R, Esteb C, Rajkhowa T, Bonifant C, Gondek LP, Levis MJ, Yegnasubramanian S, Ghiaur G, Jones RJ. CCRL2 affects the sensitivity of myelodysplastic syndrome and secondary acute myeloid leukemia cells to azacitidine. Haematologica 2023; 108:1886-1899. [PMID: 36519323 PMCID: PMC10316237 DOI: 10.3324/haematol.2022.281444] [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: 05/24/2022] [Accepted: 12/02/2022] [Indexed: 12/23/2022] Open
Abstract
Better understanding of the biology of resistance to DNA methyltransferase (DNMT) inhibitors is required to identify therapies that can improve their efficacy for patients with high-risk myelodysplastic syndrome (MDS). CCRL2 is an atypical chemokine receptor that is upregulated in CD34+ cells from MDS patients and induces proliferation of MDS and secondary acute myeloid leukemia (sAML) cells. In this study, we evaluated any role that CCRL2 may have in the regulation of pathways associated with poor response or resistance to DNMT inhibitors. We found that CCRL2 knockdown in TF-1 cells downregulated DNA methylation and PRC2 activity pathways and increased DNMT suppression by azacitidine in MDS/sAML cell lines (MDS92, MDS-L and TF-1). Consistently, CCRL2 deletion increased the sensitivity of these cells to azacitidine in vitro and the efficacy of azacitidine in an MDS-L xenograft model. Furthermore, CCRL2 overexpression in MDS-L and TF-1 cells decreased their sensitivity to azacitidine. Finally, CCRL2 levels were higher in CD34+ cells from MDS and MDS/myeloproliferative neoplasm patients with poor response to DNMT inhibitors. In conclusion, we demonstrated that CCRL2 modulates epigenetic regulatory pathways, particularly DNMT levels, and affects the sensitivity of MDS/sAML cells to azacitidine. These results support CCRL2 targeting as having therapeutic potential in MDS/sAML.
Collapse
Affiliation(s)
- Theodoros Karantanos
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore.
| | - Patric Teodorescu
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Marios Arvanitis
- Division of Cardiology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore
| | - Brandy Perkins
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Tania Jain
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Amy E DeZern
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - W Brian Dalton
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Ilias Christodoulou
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Bogdan C Paun
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Ravi Varadhan
- Division of Biostatistics and Bioinformatics, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Christopher Esteb
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Trivikram Rajkhowa
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Challice Bonifant
- Department of Pediatrics, Johns Hopkins University School of Medicine, Baltimore, MD; Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Lukasz P Gondek
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Mark J Levis
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Srinivasan Yegnasubramanian
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Gabriel Ghiaur
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| | - Richard J Jones
- Division of Hematological Malignancies, Department of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore
| |
Collapse
|
12
|
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
|
13
|
Awada H, Gurnari C, Xie Z, Bewersdorf JP, Zeidan AM. What's Next after Hypomethylating Agents Failure in Myeloid Neoplasms? A Rational Approach. Cancers (Basel) 2023; 15:2248. [PMID: 37190176 PMCID: PMC10137017 DOI: 10.3390/cancers15082248] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2023] [Revised: 04/07/2023] [Accepted: 04/07/2023] [Indexed: 05/17/2023] Open
Abstract
Hypomethylating agents (HMA) such as azacitidine and decitabine are a mainstay in the current management of patients with myelodysplastic syndromes/neoplasms (MDS) and acute myeloid leukemia (AML) as either single agents or in multidrug combinations. Resistance to HMA is not uncommon, and it can result due to several tumor cellular adaptations. Several clinical and genomic factors have been identified as predictors of HMA resistance. However, the management of MDS/AML patients after the failure of HMA remains challenging in the absence of standardized guidelines. Indeed, this is an area of active research with several potential therapeutic agents currently under development, some of which have demonstrated therapeutic potential in early clinical trials, especially in cases with particular mutational characteristics. Here, we review the latest findings and give a rational approach for such a challenging scenario.
Collapse
Affiliation(s)
- Hussein Awada
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Carmelo Gurnari
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH 44195, USA
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, 00133 Rome, Italy
| | - Zhuoer Xie
- Department of Hematology, H. Lee Moffitt Cancer Center, Tampa, FL 33612, USA
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Comprehensive Cancer Center, New York, NY 10065, USA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University and Yale Cancer Center, New Haven, CT 06511, USA
| |
Collapse
|
14
|
Gurnari C, Xie Z, Zeidan AM. How I Manage Transplant Ineligible Patients with Myelodysplastic Neoplasms. Clin Hematol Int 2023; 5:8-20. [PMID: 36574201 PMCID: PMC10063738 DOI: 10.1007/s44228-022-00024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 11/14/2022] [Indexed: 12/28/2022] Open
Abstract
Myelodysplastic neoplasms, formerly known as myelodysplastic syndromes (MDS), represent a group of clonal disorders characterized by a high degree of clinical and molecular heterogeneity, and an invariable tendency to progress to acute myeloid leukemia. MDS typically present in the elderly with cytopenias of different degrees and bone marrow dysplasia, the hallmarks of the disease. Allogeneic hematopoietic stem cell transplant is the sole curative approach to date. Nonetheless, given the disease's demographics, only a minority of patients can benefit from this procedure. Currently used prognostic schemes such as the Revised International Prognostic Scoring System (R-IPSS), and most recently the molecular IPSS (IPSS-M), guide clinical management by dividing MDS into two big categories: lower- and higher-risk cases, based on a cut-off score of 3.5. The main clinical problem of the lower-risk group is represented by the management of cytopenias, whereas the prevention of secondary leukemia progression is the goal for the latter. Herein, we discuss the non-transplant treatment of MDS, focusing on current practice and available therapeutic options, while also presenting new investigational agents potentially entering the MDS therapeutic arsenal in the near future.
Collapse
Affiliation(s)
- Carmelo Gurnari
- Translational Hematology and Oncology Research Department, Taussig Cancer Center, Cleveland Clinic, Cleveland, OH, USA
- Department of Biomedicine and Prevention, University of Rome Tor Vergata, Rome, Italy
| | - Zhuoer Xie
- Department of Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Medicine, Yale School of Medicine, and Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
15
|
Wang C, Sallman DA. Therapeutic approaches for the management of higher risk myelodysplastic syndromes. Leuk Lymphoma 2023; 64:511-524. [PMID: 36433645 DOI: 10.1080/10428194.2022.2140287] [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: 11/28/2022]
Abstract
The heterogeneous nature of myelodysplastic syndromes (MDS) demands a risk-adapted therapeutic approach, and higher risk MDS, characterized by an increased risk of transformation into acute myeloid leukemia and inferior survival, is typically defined based on an integrated assessment of cytopenias, bone marrow blast percentage, and cytogenetic findings using the revised International Prognostic Scoring System. Incorporating mutational data could further refine the risk assessment and identify those with higher-than-expected disease risk. The principal therapeutic goal in this disease subset is to modify the natural history and prolong survival. Allogeneic stem cell transplant, the only potentially curative treatment, should be offered to eligible patients. Hypomethylating agents are the only approved treatment with unsatisfactory response rates and duration, and patients who failed prior hypomethylating agents unfortunately have dismal outcomes with urgent need of novel therapeutic agents. In this review, we provide the therapeutic landscape in higher risk MDS based on the current evidence and discuss the investigational treatment options under development.
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, Tampa, FL, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
16
|
Abstract
PURPOSE OF REVIEW Therapies that target the immune system are increasingly used across oncology, including in hematologic malignancies such as myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). While allogeneic transplant has been a key therapy in these cancers, new approaches that target the immune system are being explored including immune checkpoint therapies, antibody-drug conjugates, and cellular therapies. RECENT FINDINGS This review outlines updates in the preclinical rationale for immune directed therapies in MDS and AML, as well as recent clinical trials exploring these therapies. This manuscript summarizes the development of therapies targeting T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) and CD47, which are being evaluated in late phase studies in MDS and AML. It also reviews the landscape of other immune based therapies including antibody-drug conjugates, chimeric antigen receptor-T cells, bispecific antibodies, and tumor vaccines. SUMMARY The treatment landscape in MDS and AML is rapidly changing; with a goal of improving the quality and duration of responses, a number of immune based therapies are under investigation. This review outlines recent advances with these therapies as well as some of the challenges that remain to incorporate them into leukemia care.
Collapse
Affiliation(s)
- Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
17
|
Bewersdorf JP, Patel KK, Goshua G, Shallis RM, Podoltsev NA, Stahl M, Stein EM, Huntington SF, Zeidan AM. Cost-effectiveness of azacitidine and ivosidenib in newly diagnosed older, intensive chemotherapy-ineligible patients with IDH1-mutant acute myeloid leukemia. Leuk Lymphoma 2023; 64:454-461. [PMID: 36493798 PMCID: PMC9957935 DOI: 10.1080/10428194.2022.2140288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2022] [Revised: 10/10/2022] [Accepted: 10/16/2022] [Indexed: 12/14/2022]
Abstract
Ivosidenib + azacitidine (IVO/AZA) is approved in the United States for newly diagnosed, older or intensive chemotherapy-ineligible patients with IDH1-mutated acute myeloid leukemia. We created a partitioned survival analysis model to evaluate the health economic implications of this approval. Model outputs were used to calculate the incremental cost-effectiveness ratio (ICER) of IVO/AZA versus AZA. One-way and probabilistic sensitivity analyses were conducted. In the base case scenario, IVO/AZA and AZA resulted in life-time costs of $403,062 and $161,887, respectively. With an incremental gain of 0.95 QALYs, the ICER of IVO/AZA was $252,782/QALY. In sensitivity analyses, only a reduction in the price of IVO by 59.3% lowered the ICER to below $150,000/QALY and 99.95% of model calculations yielded ICERs of >$150,000/QALY. In a model in which all patients received IVO monotherapy after progression on AZA monotherapy, the ICER was $155,453/QALY and various model inputs that would make IVO/AZA cost-effective were identified.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Kishan K. Patel
- Department of Medicine, Brigham and Women’s Hospital, Boston, MA
| | - George Goshua
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Department of Health Policy and Management, Harvard T.H. Chan School of Public Health, Boston, MA
| | - Rory M. Shallis
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - Nikolai A. Podoltsev
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - Maximilian Stahl
- Department of Medical Oncology, Adult Leukemia Program, Dana-Farber Cancer Institute, Boston, MA
| | - Eytan M. Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Scott F. Huntington
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | - Amer M. Zeidan
- Hematology Section, Department of Internal Medicine, Yale School of Medicine, New Haven, CT
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| |
Collapse
|
18
|
Aubrey BJ, Brunner AM. SOHO State of the Art and Next Questions: Treatment of Higher-Risk Myelodysplastic Syndromes. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:869-877. [PMID: 36030175 DOI: 10.1016/j.clml.2022.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 01/26/2023]
Abstract
Higher-risk myelodysplastic syndromes (MDS) carry a dismal prognosis with rapid disease progression, disease-related complications that impact quality of life, high risk of transformation to acute myeloid leukemia (AML), and poor long-term survival. Higher-risk disease is determined by a number of factors including the depth and type of cytopenias, percentage of myeloblasts occupying the bone marrow, cytogenetic abnormalities, and increasingly also by the presence of higher-risk molecular alterations. In addition to disease characteristics, a patient's performance status and degree of co-morbidity strongly influence treatment decisions and clinical outcomes. A critical first step in the management of patients with higher-risk MDS is evaluating eligibility for allogeneic hematopoietic stem cell transplant (HCT), which currently remains the only curative therapy, and is available to an ever-increasing number of patients. Outside of stem cell transplant, treatment with hypomethylating agent chemotherapy, azacitidine or decitabine, remains the cornerstone of therapy with improvements in overall survival and reduced transformation to AML; however, these approaches are palliative in nature and outcomes remain very poor overall. With a deepening understanding of disease pathophysiology has come a burgeoning array of novel targeted therapies that are currently in pre-clinical and early phase clinical trials offering hope for new treatment options for this malignancy.
Collapse
Affiliation(s)
- Brandon J Aubrey
- Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - Andrew M Brunner
- Harvard Medical School, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
19
|
Brunner AM, Gavralidis A, Ali NA, Hunter A, Komrokji R, Zeidan A, Sallman DA. Evaluating complete remission with partial hematologic recovery (CRh) as a response criterion in myelodysplastic syndromes (MDS). Blood Cancer J 2022; 12:153. [PMID: 36379923 PMCID: PMC9666661 DOI: 10.1038/s41408-022-00748-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/26/2022] [Accepted: 10/28/2022] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndromes (MDS) treated with DNMTI therapy have responses according to the 2006 IWG response criteria. CR responses have had the strongest association with OS. Recently, CR with partial hematologic recovery (CRh; i.e. blasts <5%, ANC > 500, platelets > 50) has been evaluated in AML, but its relevance is unknown in MDS. We identified adult patients with MDS treated with DNMTIs. We assessed best overall response to therapy according to IWG 2006 criteria, and subsequently identified patients meeting CRh criteria from the subgroup with SD or mCR. We evaluated duration of therapy and overall survival according to response. We identified 311 patients with MDS who received treatment between 2007 and 2018. The median age at the time of therapy was 69 years (range 23-91). Median follow up was 60 months. According to IWG 2006, responses included CR (n = 43, 14%), PR (n = 2, 1%), mCR (n = 57, 18%), SD (n = 149, 48%) and PD (n = 60, 19%). 79 patients (25%) achieved HI. A total of 62 patients (20%) met CRh criteria leading to reclassification of mCR (now n = 26, 8%) or SD (now n = 118, 38%). Patients achieving CR had similar time on therapy (median 8.1mo) compared to CRh (median 6mo, HR 1.4, 95% CI 0.9-2.0), and longer than other responses (p < 0.001). OS varied according to response; median OS was similar between CR (23.3mo) and CRh (25mo, HR 1.28 [0.79-2.08]), which was longer than those with mCR (17.2mo, HR 1.71 [0.96-3.05]), SD (16.3mo, HR 1.61 [1.04-2.48]), and PD (8.7mo, HR 3.04 [1.91-4.83]) (p < 0.001). OS associations with CR/CRh were confirmed in multivariable analysis accounting for allogeneic transplant. MDS patients who achieve a CRh response had similar survival and duration on therapy as patients who achieve CR response and superior to other IWG responses. These data support further evaluation of CRh into future response criteria and clinical trials.
Collapse
Affiliation(s)
| | | | | | | | | | - Amer Zeidan
- Yale University School of Medicine, New Haven, CT, USA
| | | |
Collapse
|
20
|
Kota V, Ogbonnaya A, Farrelly E, Schroader BK, Raju A, Kristo F, Dalal M. Clinical impact of transformation to acute myeloid leukemia in patients with higher-risk myelodysplastic syndromes. Future Oncol 2022; 18:4017-4029. [PMID: 36622291 DOI: 10.2217/fon-2022-0334] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Aim: Forty percent of patients with higher-risk myelodysplastic syndromes (HR-MDS) transform to acute myeloid leukemia (AML). Materials & methods: This retrospective study assessed the impact of HR-MDS transformation to AML on OS in a 6-month landmark analysis and the results were validated using a time-varying analysis. Results: The rate of AML transformation was 26.9% at 1 year. Patients who transformed to AML had a higher risk of death than patients who did not in the 6-month landmark analysis (HR: 1.82; p: 0.0072) and time-varying analysis at 1 year (HR: 2.85; p < 0.0001). Patients treated with azacitidine and decitabine in first-line therapy had similar results. Conclusion: HR-MDS transformation to AML is associated with inferior OS in patients with HR-MDS initiating first-line therapy.
Collapse
Affiliation(s)
- Vamsi Kota
- Georgia Cancer Center at Augusta University, 1411 Laney Walker Blvd, Augusta, GA 30912, USA
| | | | - Eileen Farrelly
- Xcenda LLC, 5025 Plano Pkwy Ste 500, Carrollton, TX 75010-5039, USA
| | | | - Aditya Raju
- Xcenda LLC, 5025 Plano Pkwy Ste 500, Carrollton, TX 75010-5039, USA
| | - Fjoralba Kristo
- Takeda Pharmaceuticals America, 95 Hayden Ave, Lexington, MA 02421, USA
| | - Mehul Dalal
- Takeda Pharmaceuticals America, 95 Hayden Ave, Lexington, MA 02421, USA
| |
Collapse
|
21
|
Zeidan AM, Joshi N, Kale H, Wang WJ, Corman S, Salimi T, Epstein RS. Impact of Hypomethylating Agent Use on Hospital and Emergency Room Visits, and Predictors of Early Discontinuation in Patients With Higher-Risk Myelodysplastic Syndromes. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:670-679. [PMID: 35614009 DOI: 10.1016/j.clml.2022.04.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 04/05/2022] [Accepted: 04/13/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Previous analyses using the SEER-Medicare database have reported substantial underutilization of hypomethylating agents (HMAs) among patients with higher-risk myelodysplastic syndromes (MDS), and an association between poor HMA persistence and high economic burden. We aimed to compare rates of hospitalizations and emergency room (ER) visits among patients with higher-risk MDS according to use or non-use of HMA therapy, and to explore factors associated with early discontinuation of HMA therapy. PATIENTS AND METHODS We used the 2010-2016 SEER-Medicare database to identify patients aged ≥66 years with a new diagnosis of refractory anemia with excess blasts (RAEB; a surrogate for higher-risk MDS) between 2011 and 2015. New hospitalizations and ER visits during the 12 months following MDS diagnosis were determined. Treatment discontinuation was defined as stopping HMA therapy before 4 cycles. RESULTS Overall, 664 (55.8%) patients were HMA users and 526 (44.2%) non-users. Non-users had more hospitalizations (mean 0.47 vs. 0.30, P < .001) and ER visits (mean 0.69 vs. 0.41, P = .005) per month than HMA users. Among HMA users, 193 (29.1%) discontinued HMA therapy before 4 cycles, and 91 (47.2%) of these after 1 cycle. Older age and poor performance status were associated with higher risk of HMA discontinuation. CONCLUSION An increased rate of hospitalizations and ER visits occurred in HMA non-users vs. HMA users. Approximately one-third of patients discontinued HMA therapy early. Predictors of discontinuation included older age and poor performance status. Novel approaches are needed to improve utilization and persistence with HMA therapy and associated outcomes, particularly among these higher-risk groups.
Collapse
Affiliation(s)
- Amer M Zeidan
- Section of Hematology, Department of Medicine, Yale School of Medicine, and Yale Cancer Center, Smilow Cancer Hospital, New Haven, CT.
| | | | | | | | | | | | | |
Collapse
|
22
|
Mohty R, Al Hamed R, Bazarbachi A, Brissot E, Nagler A, Zeidan A, Mohty M. Treatment of myelodysplastic syndromes in the era of precision medicine and immunomodulatory drugs: a focus on higher-risk disease. J Hematol Oncol 2022; 15:124. [PMID: 36045390 PMCID: PMC9429775 DOI: 10.1186/s13045-022-01346-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Accepted: 08/22/2022] [Indexed: 11/22/2022] Open
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous clonal disease of myeloid neoplasms characterized by ineffective hematopoiesis, variable degree of cytopenias, and an increased risk of progression to acute myeloid leukemia (AML). Molecular and genetic characterization of MDS has led to a better understanding of the disease pathophysiology and is leading to the development of novel therapies. Targeted and immune therapies have shown promising results in different hematologic malignancies. However, their potential use in MDS is yet to be fully defined. Here, we review the most recent advances in therapeutic approaches in MDS, focusing on higher-risk disease. Allogeneic hematopoietic cell transplantation is beyond the scope of this article.
Collapse
Affiliation(s)
- Razan Mohty
- Division of Hematology-Oncology and Blood and Marrow Transplantation Program, Mayo Clinic, Jacksonville, FL, USA
| | - Rama Al Hamed
- Department of Internal Medicine, Jacobi Medical Center/Albert Einstein College of Medicine, Bronx, NY, USA
| | - Ali Bazarbachi
- Bone Marrow Transplantation Program, Department of Internal Medicine, American University of Beirut Medical Center, Beirut, Lebanon
| | - Eolia Brissot
- Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, AP-HP, Sorbonne University, and INSERM, Saint-Antoine Research Centre, 75012, Paris, France
| | - Arnon Nagler
- Hematology and Bone Marrow Transplant Unit, Chaim Sheba Medical Center, Tel Hashomer, Israel
| | - Amer Zeidan
- Division of Hematology/Oncology, Department of Internal Medicine, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Mohamad Mohty
- Department of Clinical Hematology and Cellular Therapy, Saint-Antoine Hospital, AP-HP, Sorbonne University, and INSERM, Saint-Antoine Research Centre, 75012, Paris, France.
| |
Collapse
|
23
|
Geng S, Xu R, Huang X, Li M, Deng C, Lai P, Wang Y, Wu P, Chen X, Weng J, Du X. Dynamics of PD-1 expression are associated with treatment efficacy and prognosis in patients with intermediate/high-risk myelodysplastic syndromes under hypomethylating treatment. Front Immunol 2022; 13:950134. [PMID: 36003379 PMCID: PMC9393298 DOI: 10.3389/fimmu.2022.950134] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2022] [Accepted: 07/14/2022] [Indexed: 11/13/2022] Open
Abstract
Hypomethylating agents (HMAs) are widely used in patients with higher-risk MDS not eligible for stem cell transplantation. However, the general response rate by HMAs is lesser than 50% in MDS patients, while the relapse rate is high. Emerging evidence indicates that demethylating effects committed by HMAs may facilitate the up-regulation of a range of immune checkpoints or cancer suppressor genes in patients with MDS, among which the programmed death protein 1 (PD-1) and its ligands are demonstrated to be prominent and may contribute to treatment failure and early relapse. Although results from preliminary studies with a limited number of enrolled patients indicate that combined administration of PD-1 inhibitor may yield extra therapeutic benefit in some MDS patients, identifications of this subgroup of patients and optimal timing for the anti-PD-1 intervention remain significant challenges. Dynamics of immune checkpoints and associated predictive values during HMA-treatment cycles remained poorly investigated. In this present study, expression levels of immune checkpoints PD-1 and its ligands PD-L1 and PD-L2 were retrospectively analyzed by quantitative PCR (Q-PCR) in a total of 135 myelodysplastic syndromes (MDS) cohort with higher-risk stratification. The prognostic value of dynamics of these immune checkpoints during HMA cycles was validated in two independent prospective cohorts in our center (NCT01599325 and NCT01751867). Our data revealed that PD-1 expression was significantly higher than that in younger MDS patients (age ≤ 60) and MDS with lower IPSS risk stratification (intermediate risk-1). A significantly up-regulated expression of PD-1 was seen during the first four HMA treatment cycles in MDS patients, while similar observation was not seen concerning the expression of PD-L1 or PD-L2. By utilizing binary logistic regression and receiver operating characteristic (ROC) models, we further identified that higher or equal to 75.9 PD-1 expressions after 2 cycles of HMA treatment is an independent negative prognostic factor in predicting acute myeloid leukemia (AML) transformation and survival. Collectively, our data provide rationales for monitoring the expression of PD-1 during HMA treatment cycles, a higher than 75.9 PD-1 expression may identify patients who will potentially benefit from the combined therapy of HMA and PD-1 inhibitors.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Xin Du
- *Correspondence: Xin Du, ; ; Jianyu Weng,
| |
Collapse
|
24
|
Brunner AM, Fell G, Steensma DP. Historical expectations with DNA methyltransferase inhibitor monotherapy in MDS: when is combination therapy truly "promising"? Blood Adv 2022; 6:2854-2866. [PMID: 35143613 PMCID: PMC9092413 DOI: 10.1182/bloodadvances.2021006357] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Accepted: 02/09/2022] [Indexed: 11/20/2022] Open
Abstract
DNA methyltransferase inhibitors (DNMTIs) for patients with higher risk myelodysplastic syndromes (HR-MDS) have low complete remission rates and are not curative. Early DNMTI combination clinical trials in HR-MDS are often termed "promising," but many randomized trials subsequently failed to show benefit. Clearer understanding of when a combination is likely to improve upon DNMTI monotherapy would inform randomized studies. We reviewed MDS azacitidine or decitabine monotherapy studies. We collected baseline demographics including International Prognostic Scoring System (IPSS) risk, DNMTI, disease characteristics; and response variables including survival and marrow and hematologic responses. Aggregate estimates across studies were calculated using meta-analyses techniques. Using a binomial design, we estimated the necessary operating characteristics to design a phase 2 study showing improved efficacy of a combination over monotherapy. Among 1908 patients, the overall response rate (ORR) was 24% (n = 464; 95% confidence interval [CI], 0.22-0.26): 267 complete response (CR, 14%), 68 partial response (4%), and 129 marrow complete remission (7%). Among 1604 patients for whom a hematologic response was reported, 476 (30%; 95% CI, 0.27-0.32) reported hematologic improvement (HI). More patients treated with azacitidine achieved HI (38%; 95% CI, 0.35-0.41) compared with decitabine (15%; 95% CI, 0.13-0.19), whereas the marrow ORR rate was higher with decitabine (29%; 95% CI, 0.26-0.33) compared with azacitidine (21%; 95% CI, 0.19-0.23). CR rates were similar between DNMTIs: 13% with azacitidine and 16% with decitabine. Variables that influence MDS response include the specific DNMTI backbone and the distribution of IPSS risk of patients enrolled on a trial. Considering these factors can help identify which early combination approaches are worth assessing in larger randomized trials.
Collapse
Affiliation(s)
- Andrew M Brunner
- Department of Hematology and Oncology, Massachusetts General Hospital, Harvard Medical School, Boston, MA
| | - Geoffrey Fell
- Department of Biostatistics and Computational Biology, Dana-Farber Cancer Institute, Boston, MA; and
| | | |
Collapse
|
25
|
Goksu SY, Ozer M, Goksu BB, Wang R, Khatib J, Patel PA, Vusirikala M, Cole S, Seyhanli A, Collins RH, Chung S, Zeidan AM, Madanat YF. The impact of race and ethnicity on outcomes of patients with myelodysplastic syndromes: a population-based analysis. Leuk Lymphoma 2022; 63:1651-1659. [PMID: 35133215 DOI: 10.1080/10428194.2022.2032034] [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: 10/19/2022]
Abstract
Race and ethnic backgrounds affect the disease characteristics and clinical outcomes in many cancers, including acute myeloid leukemia; however, the association of race/ethnicity on myelodysplastic syndrome (MDS) is still controversial. Therefore, we aimed to study the impact of race/ethnicity on the disease characteristics and survival outcomes in patients with MDS. Adult patients with MDS diagnosed in 2004-2016 were selected using the SEER database. Race/ethnicity was categorized as non-Hispanic White (NHW), non-Hispanic Black (NHB), and Hispanic. Hispanic and NHB patients had significantly lower incidence rate ratio (IRR) in age group ≥01 years (p < .001) compared to NHW; however, in the age group <50 years, NHB patients had significantly higher IRR with an increased incidence rate of 49%. NHB patients had better overall survival than Hispanic and NHW patients (p < .001), even after adjusting for confounding variables. MDS have significant differences in age at diagnosis, disease risk, and survival outcomes based on racial/ethnic backgrounds.
Collapse
Affiliation(s)
- Suleyman Yasin Goksu
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Muhammet Ozer
- Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA.,Department of Internal Medicine, Capital Health Regional Medical Center, Trenton, NJ, USA
| | - Busra B Goksu
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, USA
| | - Rong Wang
- Department of Population and Data Sciences, Yale University, New Haven, CT, USA
| | - Jude Khatib
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Prapti A Patel
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Madhuri Vusirikala
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Suzanne Cole
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Ahmet Seyhanli
- Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Robert H Collins
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Stephen Chung
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale Cancer Center and Smilow Cancer Hospital, Yale University School of Medicine, New Haven, CT, USA
| | - Yazan F Madanat
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.,Division of Hematology and Oncology, Harold C. Simmons Comprehensive Cancer Center, Dallas, TX, USA
| |
Collapse
|
26
|
Griffiths EA. Oral hypomethylating agents: beyond convenience in MDS. HEMATOLOGY. AMERICAN SOCIETY OF HEMATOLOGY. EDUCATION PROGRAM 2021; 2021:439-447. [PMID: 34889435 PMCID: PMC8791154 DOI: 10.1182/hematology.2021000278] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Oral hypomethylating agents (HMAs) represent a substantial potential boon for patients with myelodysplastic syndrome (MDS) who have previously required between 5 and 7 visits per month to an infusion clinic to receive therapy. For patients who respond to treatment, ongoing monthly maintenance visits represent a considerable burden to quality of life, and for those who are early in therapy, these sequential visits may tax transportation and financial resources that would be optimally distributed over the treatment cycle to facilitate transfusion support. The availability of oral HMAs may support the optimal application of these agents by contributing to adherence and lessening the burden of therapy, potentially encouraging patients to stay on longer-term treatment. Distinct pharmacokinetic profiles for the recently approved oral HMAs (oral azacitidine and decitabine-cedazuridine) result in differential toxicity profiles and have prompted their clinical trial development in lower- and higher-risk MDS, respectively.
Collapse
Affiliation(s)
- Elizabeth A. Griffiths
- Correspondence Elizabeth A. Griffiths, Roswell Park Comprehensive Cancer Center, Elm and Carlton St, Buffalo, NY 14263; e-mail:
| |
Collapse
|
27
|
Aguirre LE, Komrokji R, Padron E. It is time to shift the treatment paradigm in myelodysplastic syndromes: A focus on novel developments and current investigational approaches exploring combinatorial therapy in high-risk MDS. Best Pract Res Clin Haematol 2021; 34:101325. [PMID: 34865697 DOI: 10.1016/j.beha.2021.101325] [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: 10/20/2022]
Abstract
Higher risk myelodysplastic syndromes are defined as a subset of disease with higher risk of AML transformation and poor overall survival. For decades, therapeutic options for high-risk MDS have been limited to allogeneic stem cell transplant (the only option for cure but limited to only a handful of patients) or hypomethylating agents, with the goal to alter the natural history of disease, delay progression and improve survival, while addressing cytopenias, transfusion requirements and improving quality of life. Recent developments in DNA sequencing and other technologies have shed significant light into the pathogenesis of MDS and led to rational and targeted drug development across a variety of therapeutic vulnerabilities, including disruption of protein ubiquitination through NAE inhibition, selective modulation of macrophage activity and immune checkpoint inhibition through blockade of TIM-3. This review highlights some of the most promising agents in recent drug development and their therapeutic efficacy in the management of high-risk MDS, and further explores the rationale behind potential combinatorial approaches using an HMA backbone to synergistically improve treatment outcomes.
Collapse
Affiliation(s)
- Luis E Aguirre
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Rami Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Eric Padron
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| |
Collapse
|
28
|
Inhibition of CBP synergizes with the RNA-dependent mechanisms of Azacitidine by limiting protein synthesis. Nat Commun 2021; 12:6060. [PMID: 34663789 PMCID: PMC8523560 DOI: 10.1038/s41467-021-26258-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 09/22/2021] [Indexed: 01/25/2023] Open
Abstract
The nucleotide analogue azacitidine (AZA) is currently the best treatment option for patients with high-risk myelodysplastic syndromes (MDS). However, only half of treated patients respond and of these almost all eventually relapse. New treatment options are urgently needed to improve the clinical management of these patients. Here, we perform a loss-of-function shRNA screen and identify the histone acetyl transferase and transcriptional co-activator, CREB binding protein (CBP), as a major regulator of AZA sensitivity. Compounds inhibiting the activity of CBP and the closely related p300 synergistically reduce viability of MDS-derived AML cell lines when combined with AZA. Importantly, this effect is specific for the RNA-dependent functions of AZA and not observed with the related compound decitabine that is only incorporated into DNA. The identification of immediate target genes leads us to the unexpected finding that the effect of CBP/p300 inhibition is mediated by globally down regulating protein synthesis.
Collapse
|
29
|
Li X, Xu F, Zhang Z, Guo J, He Q, Song LX, Wu D, Zhou LY, Su JY, Xiao C, Chang CK, Wu LY. Dynamics of epigenetic regulator gene BCOR mutation and response predictive value for hypomethylating agents in patients with myelodysplastic syndrome. Clin Epigenetics 2021; 13:169. [PMID: 34461985 PMCID: PMC8404357 DOI: 10.1186/s13148-021-01157-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Accepted: 08/23/2021] [Indexed: 12/02/2022] Open
Abstract
Background BCOR (BCL6 corepressor) is an epigenetic regulator gene involved in the specification of cell differentiation and body structure development. Recurrent somatic BCOR mutations have been identified in myelodysplastic syndrome (MDS). However, the clinical impact of BCOR mutations on MDS prognosis is controversial and the response of hypomethylating agents in MDS with BCOR mutations (BCORMUT) remains unknown. Results Among 676 MDS patients, 43 patients (6.4%) harbored BCOR mutations. A higher frequency of BCOR mutations (8.7%) was investigated in patients with normal chromosome, compared to 4.2% in patients with abnormal karyotype (p = 0.040). Compared to the BCORWT patients, the BCORMUT patients showed a higher ratio of refractory anemia with excess blasts subset (p = 0.008). The most common comutations with BCOR genes were ASXL1 (p = 0.002), DNMT3A (p = 0.114) and TET2 (p = 0.148). When the hierarchy of somatic mutations was analyzed, BCOR mutations were below the known initial mutations (ASXL1 or TET2) but were above U2AF1 mutations. Transformation-free survival was significantly shorter in BCORMUT patients than that in BCORWT patients (16 vs. 35 months; p = 0.035). RNA-sequencing was performed in bone marrow mononuclear cells from BCORMUT and BCORWT patients and revealed 2030 upregulated and 772 downregulated genes. Importantly, HOXA6, HOXB7, and HOXB9 were significantly over-expressed in BCORMUT patients, compared to BCORWT patients. Eight of 14 BCORMUT patients (57.1%) achieved complete remission (CR) with decitabine treatment, which was much higher than that in BCORWT patients (28.7%, p = 0.036). Paired sequencing results (before and after decitabine) showed three of 6 CR patients lost the mutated BCOR. The median survival of CR patients with a BCORMUT was 40 months, which was significantly longer than that in patients with BCORWT (20 months, p = 0.036). Notably, prolonged survival was observed in three BCORMUT CR patients even without any subsequent therapies. Conclusions BCOR mutations occur more frequently in CN MDS patients, predicting higher risk of leukemia transformation. BCORMUT patients showed a better response to decitabine and achieved longer post-CR survival. Supplementary Information The online version contains supplementary material available at 10.1186/s13148-021-01157-8.
Collapse
Affiliation(s)
- Xiao Li
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Feng Xu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Zheng Zhang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Juan Guo
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Qi He
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Lu-Xi Song
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Dong Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Li-Yu Zhou
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ji-Ying Su
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Chao Xiao
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Chun-Kang Chang
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China
| | - Ling-Yun Wu
- Department of Hematology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, 200233, China.
| |
Collapse
|
30
|
Shallis RM, Zeidan AM. Management of the Older Patient with Myelodysplastic Syndrome. Drugs Aging 2021; 38:751-767. [PMID: 34342860 DOI: 10.1007/s40266-021-00881-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/11/2021] [Indexed: 01/19/2023]
Abstract
No two diagnoses of myelodysplastic syndrome are genuinely alike, owing to differing and dynamic mutational topography and epigenetic aberrancy. Consequently, no two patients with myelodysplastic syndrome are identical and disease-specific and patient-specific factors are considered in formulating the optimal treatment, which includes few that are disease modifying. Age itself should not be an absolute contraindication to therapy, including intensive therapy such as allogeneic hematopoietic stem cell transplantation, which is the only curative therapy. However, age associates with an increased prevalence of frailty and comorbidities that must be considered and may preclude a path to cure. Palliative therapies are the mainstay for many patients with myelodysplastic syndrome, which is a disease of older adults with the majority of patients diagnosed at age ≥ 75 years. The older patient requires heightened attention to end organ function/reserve and drug-drug interactions as well as insurance, income, cost, and socioeconomic and psychosocial issues that influence management. Many prior studies have included relatively younger populations or have not specifically performed high-quality subgroup analyses of older patients. In this review, we discuss the available standard-of-care therapies for myelodysplastic syndrome as they specifically relate to the older population and assess the emerging therapeutics that may further the pursuit for personalized treatment and improve both the outcomes and quality of life of the older patient with myelodysplastic syndrome.
Collapse
Affiliation(s)
- Rory M Shallis
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, Yale University, 333 Cedar Street, PO Box 208028, New Haven, CT, 06520-8028, USA
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, Yale University, 333 Cedar Street, PO Box 208028, New Haven, CT, 06520-8028, USA.
| |
Collapse
|
31
|
Chien KS, Kim K, Nogueras-Gonzalez GM, Borthakur G, Naqvi K, Daver NG, Montalban-Bravo G, Cortes JE, DiNardo CD, Jabbour E, Alvarado Y, Andreeff M, Bose P, Jain N, Kadia TM, Huang X, Sheppard KB, Klingner-Winton C, Pierce SA, Dong XQ, Soltysiak KA, Kantarjian HM, Garcia-Manero G. Phase II study of azacitidine with pembrolizumab in patients with intermediate-1 or higher-risk myelodysplastic syndrome. Br J Haematol 2021; 195:378-387. [PMID: 34340254 DOI: 10.1111/bjh.17689] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 06/01/2021] [Accepted: 06/07/2021] [Indexed: 02/02/2023]
Abstract
Programmed cell death protein 1 (PD-1) and PD-ligand 1 (PD-L1) expression is upregulated in cluster of differentiation 34 (CD34)+ bone marrow cells from patients with myelodysplastic syndromes (MDS). Hypomethylating agent (HMA) treatment results in further increased expression of these immune checkpoints. We hypothesised that combining an anti-PD-1 antibody with HMAs may have efficacy in patients with MDS. To test this concept, we designed a phase II trial of the combination of azacitidine and pembrolizumab with two cohorts. In the 17 previously untreated patients, the overall response rate (ORR) was 76%, with a complete response (CR) rate of 18% and median overall survival (mOS) not reached after a median follow-up of 12·8 months. For the HMA-failure cohort (n = 20), the ORR was 25% and CR rate was 5%; with a median follow-up of 6·0 months, the mOS was 5·8 months. The most observed toxicities were pneumonia (32%), arthralgias (24%) and constipation (24%). Immune-related adverse events requiring corticosteroids were required in 43%. Overall, this phase II trial suggests that azacitidine and pembrolizumab is safe with manageable toxicities in patients with higher-risk MDS. This combined therapy may have anti-tumour activity in a subset of patients and merits further studies in the front-line setting.
Collapse
Affiliation(s)
- Kelly S Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kunhwa Kim
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kiran Naqvi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jorge E Cortes
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Courtney D DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yesid Alvarado
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Michael Andreeff
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Prithviraj Bose
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nitin Jain
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xuelin Huang
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kimberly B Sheppard
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Cheri Klingner-Winton
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sherry A Pierce
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xiao Qin Dong
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly A Soltysiak
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
32
|
Erythroid nuclear dysplasia is associated with inferior outcomes for patients with myelodysplastic syndrome undergoing allogeneic hematopoietic cell transplantation. Leuk Res 2021; 109:106625. [PMID: 34062365 DOI: 10.1016/j.leukres.2021.106625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 05/12/2021] [Accepted: 05/16/2021] [Indexed: 11/23/2022]
Abstract
Disease burden prior to hematopoietic cell transplantation (HCT) is difficult to assess in myelodysplastic syndrome (MDS), particularly in patients without excess blasts. We assessed whether morphologic dysplasia at the time of transplant can be a metric of disease burden that is associated with post-transplant outcomes in MDS patients. We identified 84 MDS patients undergoing allogeneic HCT at our institution between 2010 and 2017 who received a bone marrow evaluation immediately prior to HCT. Dysplasia was independently determined by two hematopathologists blinded to existing pathology reports. Erythroid nuclear dysplasia, but not megakaryocytic or myeloid, was associated with post-HCT outcomes. Presence compared to absence of erythroid nuclear dysplasia was associated with lower 2-year progression-free survival (PFS; 34 % vs 62 %, p = 0.0495) and 2-year overall survival (OS; 34 % vs 62 %, p = 0.042). In a multivariate analysis including age, IPSS-R at the time of transplant, pre-HCT therapy, and donor type as covariates, erythroid nuclear dysplasia remained associated with lower PFS (HR 2.6, p = 0.036) and OS (HR 2.7, p = 0.028). Dysplasia assessment prior to transplant may serve as an estimate of disease burden in MDS and identify high-risk patients who merit additional therapies pre- or post-transplant.
Collapse
|
33
|
Patel AA, Cahill K, Saygin C, Odenike O. Cedazuridine/decitabine: from preclinical to clinical development in myeloid malignancies. Blood Adv 2021; 5:2264-2271. [PMID: 33904891 PMCID: PMC8095139 DOI: 10.1182/bloodadvances.2020002929] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Accepted: 03/23/2021] [Indexed: 12/23/2022] Open
Abstract
Since the US Food and Drug Administration (FDA) approvals of parenteral decitabine and azacitidine, DNA methyltransferase inhibitors, otherwise referred to as DNA hypomethylating agents (HMAs), have been a mainstay in the treatment of higher-risk myelodysplastic syndromes. The development of oral HMAs has been an area of active interest; however, oral bioavailability has been quite poor due to rapid metabolism by cytidine deaminase (CDA). This led to the development of the novel CDA inhibitor cedazuridine, which was combined with an oral formulation of decitabine. Preclinical work demonstrated a pharmacokinetic and pharmacodynamic profile approximate to parenteral decitabine, leading to early-phase clinical trials of oral cedazuridine-decitabine (C-DEC) in myelodysplastic syndromes and chronic myelomonocytic leukemia (CMML). A combination of oral decitabine 35 mg with oral cedazuridine 100 mg was established as the recommended phase 2 dose. Phase 2 data confirmed bioequivalence of C-DEC when compared with parenteral decitabine, and a larger phase 3 trial has demonstrated similar results, leading to the FDA approval of C-DEC for use in intermediate/high-risk myelodysplastic syndrome (MDS) and CMML. This review will focus upon the current role of HMA therapy in MDS/CMML, preclinical and clinical development of C-DEC, and potential roles of oral HMA therapy in myeloid malignancies moving forward.
Collapse
Affiliation(s)
- Anand A Patel
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Kirk Cahill
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Caner Saygin
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, University of Chicago Medicine, Chicago, IL
| |
Collapse
|
34
|
Epigenetics in a Spectrum of Myeloid Diseases and Its Exploitation for Therapy. Cancers (Basel) 2021; 13:cancers13071746. [PMID: 33917538 PMCID: PMC8038780 DOI: 10.3390/cancers13071746] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Revised: 04/01/2021] [Accepted: 04/02/2021] [Indexed: 02/06/2023] Open
Abstract
Simple Summary The genome is stored in the limited space of the nucleus in a highly condensed form. The regulation of this packaging contributes to determining the accessibility of genes and is important for cell function. Genes affecting the genome’s packaging are frequently mutated in bone marrow cells that give rise to the different types of blood cells. Here, we first discuss the molecular functions of these genes and their role in blood generation under healthy conditions. Then, we describe how their mutations relate to a subset of diseases including blood cancers. Finally, we provide an overview of the current efforts of using and developing drugs targeting these and related genes. Abstract Mutations in genes encoding chromatin regulators are early events contributing to developing asymptomatic clonal hematopoiesis of indeterminate potential and its frequent progression to myeloid diseases with increasing severity. We focus on the subset of myeloid diseases encompassing myelodysplastic syndromes and their transformation to secondary acute myeloid leukemia. We introduce the major concepts of chromatin regulation that provide the basis of epigenetic regulation. In greater detail, we discuss those chromatin regulators that are frequently mutated in myelodysplastic syndromes. We discuss their role in the epigenetic regulation of normal hematopoiesis and the consequence of their mutation. Finally, we provide an update on the drugs interfering with chromatin regulation approved or in development for myelodysplastic syndromes and acute myeloid leukemia.
Collapse
|
35
|
Diamantopoulos PT, Viniou NA. Factors affecting response to 5-azacytidine and prognosis of myelodysplastic syndrome. Is long-term survival a realistic goal? Leuk Res 2021; 103:106543. [PMID: 33640709 DOI: 10.1016/j.leukres.2021.106543] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 02/15/2021] [Accepted: 02/18/2021] [Indexed: 12/14/2022]
Abstract
The introduction of hypomethylating agents (HMAs) 5-azacytidine and decitabine has altered the prognosis of patients with myelodysplastic syndrome (MDS). Over the past few years, the International Prognostic Scoring System (IPSS) and the revised IPSS (IPSS-R) have been used both to define the prognosis of patients with MDS and to select patients to be treated with HMAs. Nevertheless, the prognosis of individual patients with MDS can differ considerably from the one calculated with the use of the above-mentioned prognostic systems. Thus, some patients may achieve long-term survival irrespective of their initial prognostic score. Several factors besides those used to define the IPSS/IPSS-R are analyzed in this review article; these include age and gender, the baseline hematologic characteristics, the comorbidities, the cytogenetic and molecular profile of the patients, as well as their response to treatment with 5-azacytidine. Thus, insight into a more personalized way of managing patients with MDS is given and long-term survival is set as a more realistic goal of treatment with 5-azacytidine.
Collapse
Affiliation(s)
- Panagiotis T Diamantopoulos
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece.
| | - Nora-Athina Viniou
- First Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| |
Collapse
|
36
|
Bagla S, Regling KA, Wakeling EN, Gadgeel M, Buck S, Zaidi AU, Flore LA, Chicka M, Schiffer CA, Chitlur MB, Ravindranath Y. Distinctive phenotypes in two children with novel germline RUNX1 mutations - one with myeloid malignancy and increased fetal hemoglobin. Pediatr Hematol Oncol 2021; 38:65-79. [PMID: 32990483 DOI: 10.1080/08880018.2020.1814463] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
RUNX1 associated familial platelet disorder (FPD) is a rare autosomal dominant hematologic disorder characterized by thrombocytopenia and/or altered platelet function. There is an increased propensity to develop myeloid malignancy (MM) - acute myeloid leukemia, myeloproliferative neoplasms or myelodysplastic syndrome often in association with secondary somatic variants in other genes. To date, 23 FPD-MM pediatric cases have been reported worldwide. Here, we present two new kindreds with novel RUNX1 pathogenic variants in which children are probands. The first family is a daughter/mother diad, sharing a heterozygous frameshift variant in RUNX1 gene (c.501delT p.Ser167Argfs*9). The daughter, age 13 years, presented with features resembling juvenile myelomonocytic leukemia - severe anemia, thrombocytopenia, high white cell count with blast cells, monocytosis, increased nucleated red cells and had somatic mutations with high allele burden in CUX1, PHF6, and SH2B3 genes. She also had increased fetal hemoglobin and increased LIN28B expression. The mother, who had a long history of hypoplastic anemia, had different somatic mutations- a non-coding mutation in CUX1 but none in PHF6 or SH2B3. Her fetal hemoglobin and LIN28B expression were normal. In the second kindred, the proband, now 4 years old with thrombocytopenia alone, was investigated at 3 months of age for persistent neonatal thrombocytopenia with large platelets. Molecular testing identified a heterozygous intragenic deletion in RUNX1 encompassing exon 5. His father is known to have increased bruising for several years but is unavailable for testing. These two cases illustrate the significance of secondary mutations in the development and progression of RUNX1-FPD to MM.
Collapse
Affiliation(s)
- Shruti Bagla
- Department of Pediatrics-Hematology/Oncology, Wayne State University-School of Medicine, Detroit, Michigan, USA
| | - Katherine A Regling
- Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Erin N Wakeling
- DMC University Laboratories, Detroit Medical Center, Detroit, Michigan, USA
| | - Manisha Gadgeel
- Department of Pediatrics-Hematology/Oncology, Wayne State University-School of Medicine, Detroit, Michigan, USA
| | - Steven Buck
- Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Ahmar U Zaidi
- Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Leigh A Flore
- Department of Pediatrics-Hematology/Oncology, Wayne State University-School of Medicine, Detroit, Michigan, USA.,Division of Genetic, Genomic and Metabolic Disorders, Children's Hospital of Michigan, Detroit, Michigan, USA
| | | | - Charles A Schiffer
- Department of Pediatrics-Hematology/Oncology, Wayne State University-School of Medicine, Detroit, Michigan, USA.,Department of Oncology, Karmanos Cancer Institute, Detroit, Michigan
| | - Meera B Chitlur
- Department of Pediatrics-Hematology/Oncology, Wayne State University-School of Medicine, Detroit, Michigan, USA.,Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| | - Yaddanapudi Ravindranath
- Department of Pediatrics-Hematology/Oncology, Wayne State University-School of Medicine, Detroit, Michigan, USA.,Division of Hematology/Oncology, Children's Hospital of Michigan, Detroit, Michigan, USA
| |
Collapse
|
37
|
Lewis R, Bewersdorf JP, Zeidan AM. Clinical Management of Anemia in Patients with Myelodysplastic Syndromes: An Update on Emerging Therapeutic Options. Cancer Manag Res 2021; 13:645-657. [PMID: 33531837 PMCID: PMC7846829 DOI: 10.2147/cmar.s240600] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2020] [Accepted: 01/13/2021] [Indexed: 12/18/2022] Open
Abstract
For the majority of patients with lower-risk myelodysplastic syndrome (LR-MDS), one of the primary clinical goals is to alleviate the symptoms associated with the resultant cytopenias and to minimize the transfusion burden. While supportive red blood cell (RBC) transfusions and erythropoiesis-stimulating agents (ESAs) may lead to clinical improvement, frequent transfusions are often complicated by iron overload and decreased quality of life; furthermore, most patients either do not respond to ESAs or will eventually develop resistance. As such, there is a great need for further therapeutic options in the management of anemia related to MDS. Several additional therapeutics are now available in select patients with LR-MDS and symptomatic anemia including luspatercept, lenalidomide, and immunosuppressive therapy. Furthermore, several novel agents are currently in development to address this area of clinical need such as imetelstat and roxadustat. In this article, we review the currently available therapeutic options for symptomatic anemia in LR-MDS as well as review the therapeutic agents in development.
Collapse
Affiliation(s)
- Russell Lewis
- Department of Medicine, Section of Hematology, Yale University, New Haven, CT, USA
| | | | - Amer M Zeidan
- Department of Medicine, Section of Hematology, Yale University, New Haven, CT, USA
| |
Collapse
|
38
|
Stein EM, Bonifacio G, Latremouille-Viau D, Shi S, Guerin A, Wu EQ, Sadek I, Cao X. Treatment patterns and outcomes in patients with myelodysplastic syndromes treated with hypomethylating agents: a SEER-Medicare analysis. Leuk Lymphoma 2021; 62:1411-1421. [PMID: 33430673 DOI: 10.1080/10428194.2020.1869959] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
To describe real-world treatment patterns and outcomes among adult patients with myelodysplastic syndromes (MDS) treated with hypomethylating agents (HMA), patients were identified in the SEER-Medicare database (01/2006-12/2016); 3,046 patients with MDS treated with HMA were included. An algorithm was developed to categorize patients into MDS risk groups: the majority of patients were classified as Higher-risk (70.9%), 8.0% as Intermediate-risk, and 21.1% as Unknown-risk. Overall, 77.4% of patients initiated azacitidine and 22.6% decitabine; they received an average of 5.1 index-HMA cycles, of which 90.9% were complete with a median cycle duration of 28 days. Median survival was 11.6, 18.4, and 19.1 months for the Higher-risk, Intermediate-risk, and Unknown-risk groups, respectively. Median time-to-AML transformation was 19.3 months for the Higher-risk group and 50.4 months for the Intermediate-risk group (not reached for Unknown-risk). Data highlight the unmet medical needs of patients with MDS treated with HMA, particularly for the Higher-risk MDS group.
Collapse
Affiliation(s)
- Eytan M Stein
- Leukemia Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gaetano Bonifacio
- Health Economics and Outcomes Research, US Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | | | - Sherry Shi
- Analysis Group, Inc., Montreal, QC, Canada
| | | | - Eric Q Wu
- Analysis Group, Inc., Boston, MA, USA
| | - Islam Sadek
- Health Economics and Outcomes Research, US Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| | - Xiting Cao
- Health Economics and Outcomes Research, US Oncology, Novartis Pharmaceuticals Corporation, East Hanover, NJ, USA
| |
Collapse
|
39
|
Joshi N, Kale H, Corman S, Wert T, Hill K, Zeidan AM. Direct Medical Costs Associated With Treatment Nonpersistence in Patients With Higher-Risk Myelodysplastic Syndromes Receiving Hypomethylating Agents: A Large Retrospective Cohort Analysis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2021; 21:e248-e254. [PMID: 33422471 DOI: 10.1016/j.clml.2020.12.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 12/02/2020] [Accepted: 12/03/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Suboptimal use of hypomethylating agents (HMAs) among higher-risk myelodysplastic syndrome (HR-MDS) patients can translate into worse health outcomes and economic burden. We estimated the direct medical costs associated with HMA treatment nonpersistence among HR-MDS patients. PATIENTS AND METHODS Using the Surveillance, Epidemiology, and End Results-Medicare linked database, a retrospective cohort of patients diagnosed with refractory anemia with excess blasts (RAEB), a diagnosis that substantially overlaps with HR-MDS, between January 2011 and December 2015 was analyzed. Patients who had ≥ 1 year of continuous Medicare enrollment before diagnosis and who did not receive stem cell transplant or lenalidomide in the follow-up period were included. Patients receiving HMAs were stratified into HMA persistent (≥4 HMA cycles) and HMA nonpersistent (<4 cycles or a gap of ≥ 90 days between cycles) groups. Healthcare resource use and costs during the follow-up period were reported descriptively as total and per patient per month (PPPM). Weighted generalized linear models (GLM) were used to compare estimated healthcare resource use and costs between HMA groups. RESULTS Among the 664 patients with RAEB, 295 (44.4%) were HMA nonpersistent and 369 (55.6%) HMA persistent. On the basis of weighted GLM analysis, the HMA nonpersistent group incurred significantly (P < .05) higher total PPPM costs compared to the HMA persistent group ($18,039 vs. $13,893), particularly for hospitalization ($3,375 vs. $2,131), and emergency room ($5,517 vs. $2,867) costs. CONCLUSION There is a substantial economic burden associated with early discontinuation of guideline-recommended HMA therapy in RAEB patients. The study findings necessitate closer care management in this population in order to improve outcomes and reduce healthcare spending.
Collapse
Affiliation(s)
- Namita Joshi
- Real-world Evidence/Data Analytics Center of Excellence, Pharmerit International LP, Bethesda, MD
| | - Hrishikesh Kale
- Real-world Evidence/Data Analytics Center of Excellence, Pharmerit International LP, Bethesda, MD
| | - Shelby Corman
- Real-world Evidence/Data Analytics Center of Excellence, Pharmerit International LP, Bethesda, MD.
| | - Tim Wert
- Market Access, Taiho Oncology, Princeton, NJ
| | - Kala Hill
- Market Access, Taiho Oncology, Princeton, NJ
| | - Amer M Zeidan
- Yale Cancer Center, Smilow Cancer Hospital at Yale New Haven, New Haven, CT
| |
Collapse
|
40
|
Corman S, Joshi N, Wert T, Kale H, Hill K, Zeidan AM. Under-use of Hypomethylating Agents in Patients With Higher-risk Myelodysplastic Syndrome in the United States: A Large Population-based Analysis. CLINICAL LYMPHOMA MYELOMA & LEUKEMIA 2020; 21:e206-e211. [PMID: 33293239 DOI: 10.1016/j.clml.2020.10.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 10/20/2020] [Accepted: 10/22/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Recent data suggest significant underutilization of hypomethylating agents (HMAs) that are recommended treatments for patients with myelodysplastic syndromes (MDS) with refractory anemia with excess blasts (RAEB). The study objective was to assess the degree of HMA use and predictors of HMA underuse in this population. PATIENTS AND METHODS This was a retrospective study including patients diagnosed with the RAEB form of MDS between January 2011 and December 2015 using the Surveillance, Epidemiology, and End Results-Medicare linked database. Patients were excluded if they had < 1 year of continuous enrollment before diagnosis or received stem cell transplant or lenalidomide during the follow-up period. HMA non-peristence was defined as use of < 4 cycles (3-10 HMA days/28 days) of HMAs or a gap of ≥ 90 days between consecutive cycles. Patients were characterized as HMA never-users, HMA-persistent users, and HMA-non-persistent users. Descriptive statistics were used to summarize patient characteristics. Multivariable logistic regression was used to assess predictors of HMA underuse and persistence. RESULTS Of the 1190 patients, 526 (44%) were never-users, 295 (25%) were non-persistent users, and 369 (31%) were persistent users. Age at diagnosis (eg, 66-70 years vs. ≥ 80 years; odds ratio [OR], 2.36; 95% confidence interval [CI], 1.56-3.56), marital status (single vs. married; OR, 0.67; 95% CI, 0.51-0.89), National Cancer Institute comorbidity index (≥ 3 vs. 0-1; OR, 0.62; 95% CI, 0.46-0.83), and performance status (poor vs. good; OR, 0.67; 95% CI, 0.51-0.87) were significantly associated with HMA underuse. CONCLUSION Several demographic and clinical factors were associated with underuse of HMAs. There is need for a better understanding of suboptimal HMA use and its relationship with clinical response.
Collapse
Affiliation(s)
| | | | | | | | - Kala Hill
- Pharmerit International, LP, Bethesda, MD
| | - Amer M Zeidan
- Section of Hematology, Department of Medicine, Yale University, and Yale Cancer Center, New Haven, CT
| |
Collapse
|
41
|
Brierley CK, Zabor EC, Komrokji RS, DeZern AE, Roboz GJ, Brunner AM, Stone RM, Sekeres MA, Steensma DP. Low participation rates and disparities in participation in interventional clinical trials for myelodysplastic syndromes. Cancer 2020; 126:4735-4743. [PMID: 32767690 DOI: 10.1002/cncr.33105] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 05/12/2020] [Accepted: 05/14/2020] [Indexed: 11/11/2022]
Abstract
BACKGROUND The development of novel therapies for the myelodysplastic syndromes (MDS) is hampered by inadequate trial recruitment. Factors contributing to low trial accrual are incompletely understood. METHODS This study analyzed a pooled patient database from institutions of the US MDS Clinical Research Consortium to compare the characteristics of participants in interventional trials with those of patients who did not enroll in a trial. RESULTS Data were identified for 1919 patients with MDS, and 449 of these patients (23%) participated in an interventional clinical trial. The median age of all patients was 68 years, and 64% were male. Patients who participated in trials were significantly younger than nonparticipants (P = .014), and men were more likely to participate in a trial (71% of trial participants were male, whereas 61% of nonparticipants were; P < .001). Race and ethnicity were not associated with trial enrollment. Patients in more affluent ZIP codes had a higher participation rate (P < .001). Patients with intermediate- and high-risk disease according to the revised International Prognostic Scoring System were overrepresented (P = .004), and trial participants less frequently had treatment-related disease (P < .001). In multivariable analyses, participation in a clinical trial was associated with a reduced hazard of death (P = .004). Even at large referral centers, only a minority of patients with MDS enrolled in interventional trials. CONCLUSIONS Restrictive trial eligibility criteria that exclude patients with MDS on account of age, comorbidities, or a history of another cancer are limit enrollment of MDS patients to clinical trials. Gaining insight into the barriers to trial accrual may help investigators and study sponsors to design trials that will accrue more rapidly and augment treatment options for patients with MDS.
Collapse
Affiliation(s)
| | - Emily C Zabor
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - Rami S Komrokji
- H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida
| | - Amy E DeZern
- Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | - Gail J Roboz
- Weill Cornell Medical College, New York, New York
| | - Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Richard M Stone
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| | - Mikkael A Sekeres
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio.,Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, Ohio
| | - David P Steensma
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, Massachusetts
| |
Collapse
|
42
|
Zhao HG, Liu F, Qin TJ, Bai H, Hou M, Yu K, Hu Y, Liu L, Li Y, Yu L. [Efficacy and safety of generic azacitidine in Chinese patients with higher-risk myelodysplastic syndromes: a multicenter, prospective, single-arm study]. ZHONGHUA XUE YE XUE ZA ZHI = ZHONGHUA XUEYEXUE ZAZHI 2020; 41:811-817. [PMID: 33190437 PMCID: PMC7656073 DOI: 10.3760/cma.j.issn.0253-2727.2020.10.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Indexed: 11/29/2022]
Abstract
Objective: To evaluate the efficacy, safety, and pharmacokinetics of the generic azacitidine in Chinese patients with higher-risk myelodysplastic syndromes(MDS). Methods: Between October 2013 and 2016, 72 patients were eligible for enrollment at 9 sites from China received generic subcutaneous azacitidine 75 mg·m(-2)·d(-1) for 7 days per 28-day cycle, for ≥6 cycles. Pharmacokinetic blood samples were collected on day 1 of a single-dose. Results: For each patient at cycle 6 or at the time of study discontinuation, whichever came first, the overall response rate, which included complete remission (CR)and partial remission(PR), was 6.9%(5/72), the rate of patients who had the best effect with CR or PR during the treatment was 12.5%(9/72). Patients who were dependent on red-blood-cell transfusions and platelet transfusions at baseline became transfusion independent were 46.3%(19/41)and 41.2% (7/17), respectively. The median time of treatment was 6 cycles, and the median OS was 16.1 months (95%CI 10.9-20.6 months). For 36 patients(50%)received treatment at ≥6 cycles, and the median OS was 22.3 months(95%CI 16.1- not evaluative). Most common grade Ⅲ-Ⅳ hematologic treatment-emergent adverse events were neutropenia(55%), leukopenia(47%), and thrombocytopenia(61%). Pharmacokinetic profiles were similar for generic and original azacitidine in Chinese patients. Conclusion: Generic azacitidine treatment was favorable and safe and can be used as a standard treatment for patients with higher-risk MDS.
Collapse
Affiliation(s)
- H G Zhao
- Department of Hematology, The Affiliated Hospital of Qingdao University, Qingdao 266000, China
| | - F Liu
- Department of Hematology, Xiyuan Hospital, Chinese Academy of Medical Sciences, Beijing 100091, China
| | - T J Qin
- National Clinical Research Center for Blood Diseases, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - H Bai
- Department of Hematology, Chinese People's Liberation Army Joint Logistics Support Unit 940 Hospital, Lanzhou 730050, China
| | - M Hou
- Department of Hematology, Qilu Hospital of Shandong University, Jinan 250012, China
| | - K Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325000, China
| | - Y Hu
- Department of Hematology, Union Hospital Affiliated to Tongji Medical College of Huazhong University of Science and Technology, Wuhan 430022, China
| | - L Liu
- Department of Hematology, Tangdu Hospital, Air Force Medical University, Xi'an 710038, China
| | - Y Li
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China
| | - L Yu
- Department of Hematology, Chinese PLA General Hospital, Beijing 100853, China; Department of Hematology-Oncology, International Cancer Center, Shenzhen University General Hospital, Shenzhen University Health Science Center, Shenzhen 518060, China
| |
Collapse
|
43
|
Bewersdorf JP, Zeidan AM. Following in the footsteps of acute myeloid leukemia: are we witnessing the start of a therapeutic revolution for higher-risk myelodysplastic syndromes? Leuk Lymphoma 2020; 61:2295-2312. [PMID: 32421403 PMCID: PMC7670856 DOI: 10.1080/10428194.2020.1761968] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2020] [Revised: 04/16/2020] [Accepted: 04/19/2020] [Indexed: 12/21/2022]
Abstract
For most patients with higher-risk myelodysplastic syndromes (HR-MDS) the hypomethylating agents (HMA) azacitidine and decitabine remain the mainstay of therapy. However, the prognosis mostly remains poor and aside from allogeneic hematopoietic stem cell transplantation no curative treatment options exist. Unlike acute myeloid leukemia, which has seen a dramatic expansion of available therapies recently, no new agents have been approved for MDS in the United States since 2006. However, various novel HMAs, HMA in combination with venetoclax, immune checkpoint inhibitors, and targeted therapies for genetically defined patient subgroups such as APR-246 or IDH inhibitors, have shown promising results in early stages of clinical testing. Furthermore, the wider availability of genetic testing is going to allow for a more individualized treatment of MDS patients. Herein, we review the current treatment approach for HR-MDS and discuss recent therapeutic advances and the implications of genetic testing on management of HR-MDS.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| | - Amer M. Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT
| |
Collapse
|
44
|
Bewersdorf JP, Zeidan AM. Management of higher risk myelodysplastic syndromes after hypomethylating agents failure: are we about to exit the black hole? Expert Rev Hematol 2020; 13:1131-1142. [DOI: 10.1080/17474086.2020.1819233] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| | - Amer M. Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
45
|
Robin M, Fenaux P. Which lower risk myelodysplastic syndromes should be treated with allogeneic hematopoietic stem cell transplantation? Leukemia 2020; 34:2552-2560. [PMID: 32661295 DOI: 10.1038/s41375-020-0967-x] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 06/23/2020] [Accepted: 07/01/2020] [Indexed: 12/12/2022]
Abstract
Indications of allogeneic hematopoietic stem cell transplantation (HSCT) remain controversial in patients with lower risk myelodysplastic syndrome. We review prognostic factors in lower risk MDS, delineating patients with relatively poor risk who may potentially benefit from HSCT during the disease course. Results of HSCT in those patients, and main efforts to decrease non-relapse mortality (NRM) are detailed. Prospective studies are needed to determine more precisely which lower risk MDS patients may benefit from transplantation.
Collapse
Affiliation(s)
- Marie Robin
- Service d'hématologie-greffe, hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP) and Université de Paris, Paris, France.
| | - Pierre Fenaux
- Service d'hématologie-sénior, hôpital Saint-Louis, Assistance Publique Hôpitaux de Paris (APHP) and Université de Paris, Paris, France
| |
Collapse
|
46
|
Davidoff AJ, Hu X, Bewersdorf JP, Wang R, Podoltsev NA, Huntington SF, Gore SD, Ma X, Zeidan AM. Hypomethylating agent (HMA) therapy use and survival in older adults with Refractory Anemia with Excess Blasts (RAEB) in the United States (USA): a large propensity score-matched population-based study †. Leuk Lymphoma 2020; 61:1178-1187. [PMID: 31878809 PMCID: PMC7735409 DOI: 10.1080/10428194.2019.1703970] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2019] [Revised: 11/16/2019] [Accepted: 12/06/2019] [Indexed: 12/13/2022]
Abstract
Hypomethylating agents (HMA) showed overall survival (OS) benefits in patients with higher-risk myelodysplastic syndromes (HR-MDS) in clinical trials. We conducted a retrospective cohort study of Surveillance, Epidemiology, and End Results (SEER)-Medicare data of patients ≥66 years diagnosed with refractory anemia with excess blasts (RAEB), a proxy for HR-MDS, in 01/2001-04/2004 (pre-period) or 01/2006-12/2011 (post-period). Association between post-period diagnosis and OS was examined using propensity scores (PS)-matched samples. Among 1876 RAEB patients, median OS was 9 months and 30.8% received HMAs (3.6% in pre-period; 43.0% in post-period) with no association between post-period diagnosis and OS. In the top PS quartile, post-period diagnosis was associated with a 74% lower risk of death (Hazard ratio [HR] = 0.26, 95%-CI: 0.10-0.69, p = 0.007), while outcomes were worse in the lowest PS quartile (HR = 2.80, 95%-CI: 1.06-7.36, p = 0.037). HMA lead to a 3-month OS benefit for patients most likely to receive HMA but not for unselected RAEB cohort.
Collapse
Affiliation(s)
- Amy J. Davidoff
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Chronic Disease Epidemiology, School of Public Health, Yale University, New Haven, CT
| | - Xin Hu
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
| | | | - Rong Wang
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Health Policy and Management, School of Public Health, Yale University, New Haven, CT
| | - Nikolai A. Podoltsev
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| | - Scott F. Huntington
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| | - Steven D. Gore
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| | - Xiaomei Ma
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Health Policy and Management, School of Public Health, Yale University, New Haven, CT
| | - Amer M. Zeidan
- Cancer Outcomes, Public Policy, and Effectiveness Research (COPPER) Center, Yale University, New Haven, CT
- Department of Internal Medicine, School of Medicine, Yale University, New Haven, CT
| |
Collapse
|
47
|
Zeidan AM, Shallis RM, Wang R, Davidoff A, Ma X. Epidemiology of myelodysplastic syndromes: Why characterizing the beast is a prerequisite to taming it. Blood Rev 2019; 34:1-15. [DOI: 10.1016/j.blre.2018.09.001] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2018] [Revised: 09/01/2018] [Accepted: 09/17/2018] [Indexed: 02/08/2023]
|
48
|
How I treat MDS after hypomethylating agent failure. Blood 2018; 133:521-529. [PMID: 30545832 DOI: 10.1182/blood-2018-03-785915] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2018] [Accepted: 12/04/2018] [Indexed: 12/20/2022] Open
Abstract
Hypomethylating agents (HMA) azacitidine and decitabine are standard of care for myelodysplastic syndrome (MDS). Response to these agents occurs in ∼50% of treated patients, and duration of response, although variable, is transient. Prediction of response to HMAs is possible with clinical and molecular parameters, but alternative approved treatments are not available, and in the case of HMA failure, there are no standard therapeutic opportunities. It is important to develop a reasoned choice of therapy after HMA failure. This choice should be based on evaluation of type of resistance (primary vs secondary, progression of disease [acute leukemia or higher risk MDS] vs absence of hematological improvement) as well as on molecular and cytogenetic characteristics reassessed at the moment of HMA failure. Rescue strategies may include stem-cell transplantation, which remains the only curative option, and chemotherapy, both of which are feasible in only a minority of cases, and experimental agents. Patients experiencing HMA failure should be recruited to clinical experimental trials as often as possible. Several novel agents with different mechanisms of action are currently being tested in this setting. Drugs targeting molecular alterations (IDH2 mutations, spliceosome gene mutations) or altered signaling pathways (BCL2 inhibitors) seem to be the most promising.
Collapse
|
49
|
Steensma DP, Brunner AM, DeZern AE, Garcia-Manero G, Komrokji RS, Odenike OS, Roboz GJ, Savona MR, Stone RM, Sekeres MA. Low clinical trial accrual of patients with myelodysplastic syndromes: Causes and potential solutions. Cancer 2018; 124:4601-4609. [PMID: 30289970 DOI: 10.1002/cncr.31769] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2018] [Revised: 08/03/2018] [Accepted: 08/21/2018] [Indexed: 11/11/2022]
Abstract
Despite few effective therapies, only a small percentage of patients diagnosed with myelodysplastic syndromes (MDS) in the United States are enrolled in prospective, interventional clinical trials. MDS-specific barriers to trial accrual include a high frequency of elderly patients with comorbid conditions, atypical disease features and uncertainty regarding the diagnosis (because other nonclonal processes also can cause dysplasia and cytopenias), a history of another nonmyeloid neoplasm resulting in therapy-related MDS, rapid disease recurrence after allogeneic stem cell transplantation, and an arbitrary division between MDS and acute myeloid leukemia. In addition, barriers to accrual that are common to other oncology populations, such as difficulty traveling to clinical trial enrollment sites and narrow trial eligibility criteria, also prevent patients with MDS from enrolling in studies. Collectively these barriers must be assessed systematically, and creative solutions are needed to improve outcomes for this needy patient population.
Collapse
Affiliation(s)
- David P Steensma
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Andrew M Brunner
- Division of Hematology and Medical Oncology, Department of Medicine, Massachusetts General Hospital, Boston, Massachusetts
| | - Amy E DeZern
- Division of Hematological Malignancies, Sidney Kimmel Cancer Center, Johns Hopkins University, Baltimore, Maryland
| | | | - Rami S Komrokji
- Moffitt Cancer Center, Malignant Hematology Department, Tampa, Florida
| | - Olatoyosi S Odenike
- Division of Hematology and Medical Oncology, Department of Medicine, University of Chicago, Chicago, Illinois
| | - Gail J Roboz
- Division of Hematology & Oncology, Joan and Sanford I. Weill Department of Medicine, Weill Cornell Medicine, New York, New York
| | - Michael R Savona
- Division of Hematology & Oncology, Vanderbilt-Ingram Cancer Center, Nashville, Tennessee
| | - Richard M Stone
- Division of Hematological Malignancies, Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Mikkael A Sekeres
- Department of Hematology and Medical Oncology, Cleveland Clinic Taussig Cancer Institute, Cleveland, Ohio
| |
Collapse
|
50
|
Counseling patients with higher-risk MDS regarding survival with azacitidine therapy: are we using realistic estimates? Blood Cancer J 2018; 8:55. [PMID: 29891916 PMCID: PMC5995881 DOI: 10.1038/s41408-018-0081-8] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2018] [Revised: 03/21/2018] [Accepted: 04/03/2018] [Indexed: 11/30/2022] Open
|