1
|
Braish JS, Montalban-Bravo G, Ravandi F, Short N, Kadia T, Ohanian M, Chien K, Masarova L, Sasaki K, Yilmaz M, Logahvi S, Daver N, Borthakur G, Jabbour E, Schneider H, Romero LT, Kantarjian H, Garcia-Manero G. Safety and efficacy of the combination of azacitidine with venetoclax after hypomethylating agent failure in higher-risk myelodysplastic syndrome. Leuk Res 2025; 153:107692. [PMID: 40252309 DOI: 10.1016/j.leukres.2025.107692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 02/28/2025] [Accepted: 04/05/2025] [Indexed: 04/21/2025]
Abstract
OBJECTIVE Therapies for patients with higher-risk myelodysplastic syndromes (HR-MDS) who have failed hypomethylating agents (HMAs) are needed. This Phase I/II study evaluates the safety, tolerability, and efficacy of venetoclax, an orally bioavailable BCL-2 inhibitor, in combination with azacitidine in this population. METHODS We conducted a single-center, dose-escalation, Phase I/II trial (NCT04550442) involving 33 patients with HR-MDS or CMML (IPSS ≥ 1.5) who had progressed after prior HMA therapy. Patients received intravenous or subcutaneous azacitidine (SC) (75 mg/m² for 5 days) and venetoclax (100-400 mg for 7-14 days in a 28-day cycle). The primary endpoints were safety/tolerability (Phase 1) and overall response rate (ORR) (Phase 2). RESULTS Patients received a median of 3 cycles (range, 1-22). The maximum tolerated dose of venetoclax was 400 mg. Common grade 3-4 adverse events included neutropenia (19 %) and thrombocytopenia (10 %). The 4-week early mortality rate was 9 %. The ORR was 49 %, and the median overall survival (OS) was 7 months (95 % CI, 3.5-10.5). The median progression-free survival (PFS) was 6 months (95 % CI, 3.0-9.0). Four patients (12 %) underwent stem cell transplantation, with 3 of 4 alive at last follow-up (75 %). CONCLUSION Combining venetoclax with azacitidine is feasible for HR-MDS and CMML patients who failed prior HMA therapy. However, this combination did not significantly improve clinical outcomes in this patient population.
Collapse
Affiliation(s)
- Julie S Braish
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | | | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Nicholas Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Maro Ohanian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Kelly Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Lucia Masarova
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Sanam Logahvi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Naval Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Elias Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Heather Schneider
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Lizabeth T Romero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, TX, United States.
| |
Collapse
|
2
|
Mina A, Komrokji R. How I treat higher-risk MDS. Blood 2025; 145:2002-2011. [PMID: 39808802 DOI: 10.1182/blood.2024025271] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 12/05/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDS) are a widely heterogenous group of myeloid malignancies characterized by morphological dysplasia, a defective hematopoiesis, and recurrent genetic abnormalities. The original International Prognostic Scoring System (IPSS) and revised IPSS have been used to risk-stratify patients with MDS to guide treatment strategies. In higher-risk MDS, the therapeutic approach is geared toward delaying leukemic transformation and prolonging survival. For more than a decade, the hypomethylating agents azacitidine and decitabine have been the standard of care and, when feasible, an allogeneic hematopoietic stem cell transplantation should be considered. However, the IPSS scoring systems solely rely on clinical, morphological, and cytogenetic features and do not account for somatic mutations present in >80% of cases. These genetic abnormalities have been shown to play a crucial role in prognostication, prompting the development of molecular IPSS, and the integration of genomic features into MDS classification systems in recent years. In this review, we delineate our approach to higher-risk MDS in the context of updated classifications and the latest prognostication tools. We use illustrative clinical cases to support our discussion and share insights from recent clinical trials, highlighting lessons learned.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| |
Collapse
|
3
|
Bataller A, Sasaki K, Urrutia S, Montalban-Bravo G, Bazinet A, Chien K, Hammond D, Bouligny IM, Swaminathan M, Issa G, Short N, Daver N, DiNardo CD, Kadia T, Jabbour E, Ravandi F, Roboz GJ, Savona M, Griffiths EA, McCloskey J, Odenike O, Oganesian A, Keer HN, Azab M, Kantarjian H, Garcia-Manero G. Oral decitabine cedazuridine with and without venetoclax in higher-risk myelodysplastic syndromes or chronic myelomonocytic leukemia: a propensity score-matched study. Blood Cancer J 2025; 15:50. [PMID: 40164584 PMCID: PMC11958769 DOI: 10.1038/s41408-025-01245-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/14/2025] [Accepted: 02/27/2025] [Indexed: 04/02/2025] Open
Abstract
Hypomethylating agents (HMA) are indicated in the treatment of higher-risk myelodysplastic syndromes (MDS) and chronic myelomonocytic leukemia (CMML). The combination of hypomethylating agents with venetoclax (Ven) has demonstrated promising results in these diseases, although randomized clinical trials are needed for validation. In this retrospective study, we compared two matched cohorts of patients with MDS or CMML: one receiving oral decitabine-cedazuridine (DEC-C, n = 73) and one receiving DEC-C and Ven (DEC-C-Ven, n = 51), in three contemporary clinical trials. The aim is to determine the impact of the addition of Ven to HMA in MDS and CMML. Individuals were matched using a propensity score approach that was based on the IPSS-M score and age. All patients had excess blasts; 84% were diagnosed with MDS and 16% with CMML. Most patients had high- or very high-risk disease, according to the revised IPSS-R. The overall response rate was superior in the DEC-C-Ven cohort (90% vs 64%, P = 0.002). The median times to best response were 1.1 and 2.7 months for the DEC-C-Ven and DEC-C cohorts, respectively (P < 0.001). More patients underwent hematopoietic stem cell transplantation in the DEC-C-Ven cohort (47%) than in the DEC-C cohort (16%, P < 0.001). The 4- and 8-week mortality did not significantly differ between the DEC-C and DEC-C-Ven cohorts. Patients in the DEC-C-Ven cohort had a more profound neutropenia at days 15 and 21 of the first cycle. The median overall survival was 24 and 19 months for the DEC-C-Ven and DEC-C cohorts, respectively (P = 0.89), and the median event-free survival durations were 18 and 10 months (P = 0.026). In conclusion, the addition of Ven resulted in improved response rates and outcomes in specific subgroups; prospective clinical trials are needed to confirm these findings.
Collapse
Affiliation(s)
- Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Samuel Urrutia
- Division of Oncology, Washington University in St. Louis, St. Louis, MO, USA
| | | | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ian M Bouligny
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahesh Swaminathan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Naval Daver
- 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
| | - Tapan Kadia
- 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
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gail J Roboz
- Weill Cornell Medicine, New York-Presbyterian Hospital, New York, NY, USA
| | - Michael Savona
- Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | | | | | - Olatoyosi Odenike
- The University of Chicago Pritzker School of Medicine, Chicago, IL, USA
| | | | | | | | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
4
|
Garcia JS, Platzbecker U, Odenike O, Fleming S, Fong CY, Borate U, Jacoby MA, Nowak D, Baer MR, Peterlin P, Chyla B, Wang H, Ku G, Hoffman D, Potluri J, Garcia-Manero G. Efficacy and safety of venetoclax plus azacitidine for patients with treatment-naive high-risk myelodysplastic syndromes. Blood 2025; 145:1126-1135. [PMID: 39652823 PMCID: PMC11923426 DOI: 10.1182/blood.2024025464] [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: 05/21/2024] [Accepted: 11/01/2024] [Indexed: 03/14/2025] Open
Abstract
ABSTRACT Outcomes are poor in patients with higher-risk myelodysplastic syndromes (HR MDS) and frontline treatment options are limited. This phase 1b study investigated safety and efficacy of venetoclax, a selective B-cell lymphoma 2 inhibitor, at the recommended phase 2 dose (RP2D; 400 mg for 14 days per 28-day cycle), in combination with azacitidine (75 mg/m2 for 7 days per 28-day cycle) for treatment-naive HR MDS. Safety was the primary outcome, and complete remission (CR) rate was the primary efficacy outcome. Secondary outcomes included rates of modified overall response (mOR), hematologic improvement (HI), overall survival (OS), and time to next treatment (TTNT). As of May 2023, 107 patients received venetoclax and azacitidine combination at the RP2D. Best response of CR or marrow CR was observed in 29.9% and 50.5% (mOR, 80.4%), respectively. Median OS was 26.0 months, with 1- and 2-year survival estimates of 71.2% and 51.3%, respectively. Among 59 patients with baseline red blood cell and/or platelet transfusion-dependence, 24 (40.7%) achieved transfusion independence on study, including 11 (18.6%) in CR. Fifty-one (49.0%) of 104 evaluable patients achieved HI. Median TTNT excluding transplantation was 13.4 months. Adverse events reflected known safety profiles for venetoclax and azacitidine, including constipation (53.3%), nausea (49.5%), neutropenia (48.6%), thrombocytopenia (44.9%), febrile neutropenia (42.1%), and diarrhea (41.1%). Overall, venetoclax plus azacitidine at the RP2D was well tolerated and had favorable outcomes. A phase 3 study (NCT04401748) is ongoing to confirm survival benefit of this combination. This trial was registered at www.clinicaltrials.gov as #NCT02942290.
Collapse
Affiliation(s)
| | - Uwe Platzbecker
- Department for Hematology, Cell Therapy, Hemostaseology and Infectious Diseases, Universitätsklinikum Leipzig, Leipzig, Germany
| | - Olatoyosi Odenike
- Section of Hematology/Oncology, Department of Medicine, The University of Chicago Medicine and Comprehensive Cancer Center, Chicago, IL
| | - Shaun Fleming
- Department of Malignant Haematology and Stem Cell Transplantation, Alfred Health, Melbourne, Australia
| | - Chun Yew Fong
- Department of Clinical Haematology, Austin Health, Melbourne, Australia
| | - Uma Borate
- Division of Hematology, Department of Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH
| | - Meagan A. Jacoby
- Department of Internal Medicine, Washington University-School of Medicine, St. Louis, MO
| | - Daniel Nowak
- Department of Hematology and Oncology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Maria R. Baer
- Department of Medicine, University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD
| | - Pierre Peterlin
- Department of Hematology, Nantes University Hospital, Nantes, France
| | | | | | - Grace Ku
- Genentech Inc, South San Francisco, CA
| | | | | | | |
Collapse
|
5
|
Carraway HE, Brunner AM, Lai CE, Luskin MR, Park J, Perl AE, Stein EM, Wang ES, Zeidan AM, Zeidner JF, Komrokji R. Advancing the Management of CH, MDS, and AML From the First Bridging the Gaps in Leukemia, Lymphoma, and Multiple Myeloma Conference. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2025:S2152-2650(25)00082-5. [PMID: 40187939 DOI: 10.1016/j.clml.2025.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/28/2025] [Accepted: 03/03/2025] [Indexed: 04/07/2025]
Abstract
PURPOSE The management of myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) have evolved substantially in recent years with the development of targeted therapies and novel nontargeted approaches. However, many questions remain about how to best use current therapies, and there is a large unmet need for effective therapies, particularly for patients with higher-risk MDS, AML, and those with MDS/AML relapsed/refractory (R/R) to prior therapy. METHODS AND RESULTS A panel of experts was assembled to discuss current controversies and unanswered questions in the care of patients with MDS and AML. Workshop topics included: molecular testing and new classification systems, clonal hematopoiesis, treatment of MDS (lower-risk and higher-risk), frontline treatment of AML, treatment of special populations, treatment of R/R AML, and novel approaches. CONCLUSIONS We identified many areas of ongoing controversy in the diagnosis and management of MDS and AML related to classification and risk assessment, treatment selection, sequencing of therapies, and monitoring of responses. Many clinical trials are ongoing to further improve outcomes for patients with MDS and AML, and we noted potential areas of debate related to study design, selection of endpoints, and assessment of responses. The controversies and gaps in knowledge identified by this panel will inform a follow-up conference in 2025 that will employ a modified Delphi method with a goal of developing and publishing formal consensus recommendations that can provide actionable guidance to clinicians in practice.
Collapse
Affiliation(s)
- Hetty E Carraway
- Division of Hematologic Oncology and Blood Disorders, Leukemia Program, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH; Department of Medicine, Cleveland Clinic Lerner College of Medicine of Case Western Reserve University, Cleveland, OH.
| | - Andrew M Brunner
- Leukemia Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA
| | - Catherine E Lai
- Leukemia Clinical Research Unit, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA
| | | | - Jae Park
- Cellular Therapy Service, Deparment of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Alexander E Perl
- Leukemia Clinical Research Unit, Abramson Cancer Center of the University of Pennsylvania, Philadelphia, PA
| | - Eytan M Stein
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Eunice S Wang
- Leukemia Service, Department of Medicine, Roswell Park Comprehensive Cancer Center, Buffalo, NY
| | - Amer M Zeidan
- Department of Internal Medicine, Yale University, New Haven, CT
| | - Joshua F Zeidner
- Division of Hematology, Department of Medicine, University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | - Rami Komrokji
- Malignant Hematology Department, Moffitt Cancer Center, Tampa, FL
| |
Collapse
|
6
|
Mina A, McGraw KL, Cunningham L, Kim N, Jen EY, Calvo KR, Ehrlich LA, Aplan PD, Garcia-Manero G, Foran JM, Garcia JS, Zeidan AM, DeZern AE, Komrokji R, Sekeres MA, Scott B, Buckstein R, Tinsley-Vance S, Verma A, Wroblewski T, Pavletic S, Norsworthy K. Advancing drug development in myelodysplastic syndromes. Blood Adv 2025; 9:1095-1104. [PMID: 39786387 PMCID: PMC11914162 DOI: 10.1182/bloodadvances.2024014865] [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: 09/20/2024] [Revised: 12/05/2024] [Accepted: 12/05/2024] [Indexed: 01/12/2025] Open
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDSs) are heterogeneous stem cell malignancies characterized by poor prognosis and no curative therapies outside of allogeneic hematopoietic stem cell transplantation. Despite some recent approvals by the US Food and Drug Administration, (eg, luspatercept, ivosidenib, decitabine/cedazuridine, and imetelstat), there has been little progress in the development of truly transformative therapies for the treatment of patients with MDS. Challenges to advancing drug development in MDS are multifold but may be grouped into specific categories, including criteria for risk stratification and eligibility, response definitions, time-to-event end points, transfusion end points, functional assessments, and biomarker development. Strategies to address these challenges and optimize future clinical trial design for patients with MDS are presented here.
Collapse
Affiliation(s)
- Alain Mina
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
| | - Kathy L. McGraw
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
- Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | - Lea Cunningham
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
| | - Nina Kim
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Emily Y. Jen
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Katherine R. Calvo
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
- Department of Laboratory Medicine, National Institutes of Health Clinical Center, Bethesda, MD
| | - Lori A. Ehrlich
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Peter D. Aplan
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
| | | | | | - Jacqueline S. Garcia
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA
| | - Amer M. Zeidan
- Section of Hematology, Department of Internal Medicine, Yale School of Medicine and Yale Comprehensive Cancer Center, Yale University, New Haven, CT
| | - Amy E. DeZern
- Division of Hematologic Malignancies, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Mikkael A. Sekeres
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| | - Bart Scott
- Fred Hutchinson Cancer Research Center, Seattle, WA
| | - Rena Buckstein
- Odette Cancer Center, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Sara Tinsley-Vance
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center & Research Institute, Tampa, FL
| | - Amit Verma
- Department of Oncology, Albert Einstein College of Medicine, New York, NY
| | - Tanya Wroblewski
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| | - Steven Pavletic
- Immune Deficiency Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD
- Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD
| | - Kelly Norsworthy
- Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD
| |
Collapse
|
7
|
Yan L, Li J, Yang Y, Zhang X, Zhang C. Old drug, new use: Recent advances for G-CSF. Cytokine 2024; 184:156759. [PMID: 39293182 DOI: 10.1016/j.cyto.2024.156759] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 09/08/2024] [Accepted: 09/10/2024] [Indexed: 09/20/2024]
Abstract
Granulocyte colony-stimulating factor (G-CSF), also known as colony-stimulating factor 3 (CSF3), is a proinflammatory cytokine that primarily stimulates the survival, proliferation, differentiation and function of neutrophil granulocyte progenitor cells and mature neutrophils. Over the past years, G-CSF has mainly been used to cure patients with neutropenia and as a part of chemotherapy to induct the remission for refractory/relapse leukemia. Recent studies showed that C-CSF can been used as condition regimens and as a part of preventive methods after allogeneic transplantation to improve the survival of patients and also has immunoregulation, and has promote or inhibit the proliferation of solid tumors. Therefore, in this review, we firstly describe the structure for G-CSF. Then its functions and mechanism were reviewed including the neutrophil mobilization, differentiation, migration, and inhibiting apoptosis of neutrophils, and its immunoregulation. Finally, the clinical applications were further discussed.
Collapse
Affiliation(s)
- Lun Yan
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing 400037 China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing 400037 China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400037 China
| | - Jing Li
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing 400037 China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing 400037 China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400037 China
| | - Yang Yang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing 400037 China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing 400037 China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400037 China
| | - Xi Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing 400037 China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing 400037 China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400037 China.
| | - Cheng Zhang
- Medical Center of Hematology, Xinqiao Hospital of Army Medical University, Chongqing 400037 China; Chongqing Key Laboratory of Hematology and Microenvironment, Chongqing 400037 China; State Key Laboratory of Trauma and Chemical Poisoning, Army Medical University, Chongqing 400037 China.
| |
Collapse
|
8
|
Gener-Ricos G, Rodriguez-Sevilla JJ, Urrutia S, Bataller A, Bazinet A, Garcia-Manero G. Advances in the management of higher-risk myelodysplastic syndromes: future prospects. Leuk Lymphoma 2024; 65:1233-1244. [PMID: 38712556 DOI: 10.1080/10428194.2024.2344061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/09/2024] [Accepted: 04/12/2024] [Indexed: 05/08/2024]
Abstract
Higher-risk myelodysplastic syndromes (HR-MDS) are defined using a number of prognostic scoring systems that include the degree of cytopenias, percentage of blasts, cytogenetic alterations, and more recently genomic data. HR-MDS encompasses characteristics such as progressive cytopenias, increased bone marrow blasts, unfavorable cytogenetics, and an adverse mutational profile. Survival is generally poor, and patients require therapy to improve outcomes. Hypomethylating agents (HMAs), such as azacitidine, decitabine, and more recently, oral decitabine/cedazuridine, are the only approved therapies for HR-MDS. These are often continued until loss of response, progression, or unacceptable toxicity. Combinations including an HMA plus other drugs have been investigated but have not demonstrated better outcomes compared to single-agent HMA. Moreover, in a disease of high genomic complexity such as HR-MDS, therapy targeting specific genomic abnormalities is of interest. This review will examine the biological underpinnings of HR-MDS, its therapeutic landscape in the frontline and relapsed settings, as well as the impact of hematopoietic stem cell transplantation, the only known curative intervention for this disease.
Collapse
Affiliation(s)
- Georgina Gener-Ricos
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Samuel Urrutia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | |
Collapse
|
9
|
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
|
10
|
Bazinet A, Desikan SP, Li Z, Rodriguez-Sevilla JJ, Venugopal S, Urrutia S, Montalban-Bravo G, Sasaki K, Chien KS, Hammond D, Kanagal-Shamanna R, Ganan-Gomez I, Kadia TM, Borthakur G, DiNardo CD, Daver NG, Jabbour EJ, Ravandi F, Kantarjian H, Garcia-Manero G. Cytogenetic and Molecular Associations with Outcomes in Higher-Risk Myelodysplastic Syndromes Treated with Hypomethylating Agents plus Venetoclax. Clin Cancer Res 2024; 30:1319-1326. [PMID: 38300723 PMCID: PMC11815990 DOI: 10.1158/1078-0432.ccr-23-2860] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/30/2023] [Accepted: 01/29/2024] [Indexed: 02/03/2024]
Abstract
PURPOSE Hypomethylating agents (HMA) combined with venetoclax are an emerging therapeutic strategy for higher-risk myelodysplastic syndromes (HR-MDS). The cytogenetic and molecular factors associated with outcomes with this combination for HR-MDS are incompletely understood. EXPERIMENTAL DESIGN We pooled patient data from 3 prospective trials evaluating HMA-venetoclax in HR-MDS to study associations between cytogenetic and molecular factors and overall response rate (ORR), overall survival (OS), and event-free survival (EFS). The Kaplan-Meier method was used to estimate time-to-event endpoints. Univariate and multivariate analyses using logistic regression (for ORR) or the Cox proportional hazards model (for OS and EFS) were used to identify associations between clinical, cytogenetic, and molecular factors and outcomes. RESULTS A total of 80 patients (52 HMA-naïve, 28 HMA-failure) were included. ORR was 90% in HMA-naïve and 57% in HMA-failure. Median OS was 28.2 and 8.3 months in HMA-naïve and HMA-failure, respectively. Median EFS was 17.9 and 5.5 months in HMA-naïve and HMA-failure, respectively. In addition, 24/52 (46%) of the HMA-naïve and 3/28 (11%) of the HMA-failure patients proceeded to allogeneic stem cell transplantation (SCT). Factors associated with inferior outcomes were prior HMA failure, complex cytogenetics, trisomy 8, TP53 mutations, and RAS pathway mutations. Mutations in RNA splicing, DNA methylation, and ASXL1 appeared favorable. Blast percentage was not predictive of outcomes. CONCLUSIONS Knowledge of cytogenetic and molecular alterations may help identify which patients with HR-MDS benefit the most from venetoclax.
Collapse
Affiliation(s)
- Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sai Prasad Desikan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ziyi Li
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Sangeetha Venugopal
- Department of Leukemia, The University of Miami Sylvester Comprehensive Cancer Center, Miami, FL
| | - Samuel Urrutia
- Department of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kelly S. Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Irene Ganan-Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Tapan M. Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Courtney D. DiNardo
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Naval G. Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Elias J. Jabbour
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Farhad Ravandi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| | | |
Collapse
|
11
|
Bataller A, Montalban-Bravo G, Bazinet A, Alvarado Y, Chien K, Venugopal S, Ishizawa J, Hammond D, Swaminathan M, Sasaki K, Issa GC, Short NJ, Masarova L, Daver NG, Kadia TM, Colla S, Qiao W, Huang X, Kanagal-Shamanna R, Hendrickson S, Ravandi F, Jabbour E, Kantarjian H, Garcia-Manero G. Oral decitabine plus cedazuridine and venetoclax in patients with higher-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia: a single-centre, phase 1/2 study. Lancet Haematol 2024; 11:e186-e195. [PMID: 38316133 DOI: 10.1016/s2352-3026(23)00367-8] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 02/07/2024]
Abstract
BACKGROUND Hypomethylating agents are approved in higher-riskmyelodysplastic syndromes. The combination of a hypomethylating agent with venetoclax is standard of care in acute myeloid leukaemia. We investigated the safety and activity of the first totally oral combination of decitabine plus cedazuridine and venetoclax in patients with higher-risk-myelodysplastic syndromes and chronic myelomonocytic leukaemia. METHODS We did a single-centre, dose-escalation and dose-expansion, phase 1/2, clinical trial. Patients with treatment-naive higher-risk-myelodysplastic syndromes or chronic myelomonocytic leukaemia (risk level categorised as intermediate-2 or higher by the International Prognostic Scoring System) with excess blasts (>5%). Treatment consisted of oral decitabine 35 mg plus cedazuridine 100 mg on days 1-5 and venetoclax (variable doses of 100-400 mg, day 1 to 14, 28-day cycle). The primary outcomes were safety for the phase 1 part and the overall response for the phase 2 part of the study. The trial is ongoing and this analysis was not prespecified. This study is registered with ClinicalTrials.gov, NCT04655755, and is currently enrolling participants. FINDINGS Between Jan 21, 2021, and Jan 20, 2023, we enrolled 39 patients (nine in phase 1 and 30 in phase 2). The median age was 71 years (range 27-94), 28 (72%) patients were male, and 11 (28%) were female. The maximum tolerated dose was not reached, and the recommended phase 2 dose was established as oral decitabine 35 mg plus cedazuridine 100 mg for 5 days and venetoclax (400 mg) for 14 days. The most common grade 3-4 adverse events were thrombocytopenia (33 [85%] of 39), neutropenia (29 [74%]), and febrile neutropenia (eight [21%]). Four non-treatment-related deaths occurred on the study drugs due to sepsis (n=2), lung infection (n=1), and undetermined cause (n=1). The median follow-up time was 10·8 months (IQR 5·6-16·4). The overall response rate was 95% (95% CI 83-99; 37/39). 19 (49%) patients proceeded to hematopoietic stem-cell transplantation. INTERPRETATION This early analysis suggests that the combination of oral decitabine plus cedazuridine with venetoclax for higher-risk-myelodysplastic syndromes and chronic myelomonocytic leukaemia is safe in most patients, with encouraging activity. Longer follow-up will be needed to confirm these data. FUNDING MD Anderson Cancer Center, MDS/AML Moon Shot, Genentech/AbbVie, and Astex Pharmaceuticals.
Collapse
Affiliation(s)
- Alex Bataller
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yesid Alvarado
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Sangeetha Venugopal
- Leukemia Program, Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami Leonard M Miller School of Medicine, Miami, FL, USA
| | - Jo Ishizawa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Danielle Hammond
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mahesh Swaminathan
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koji Sasaki
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ghayas C Issa
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas J Short
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lucia Masarova
- 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
| | - Tapan M Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Qiao
- Department of Biostatistics, 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
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Stephany Hendrickson
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- 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
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
12
|
Kasprzak A, Andresen J, Nachtkamp K, Kündgen A, Schulz F, Strupp C, Kobbe G, MacKenzie C, Timm J, Dietrich S, Gattermann N, Germing U. Infectious Complications in Patients with Myelodysplastic Syndromes: A Report from the Düsseldorf MDS Registry. Cancers (Basel) 2024; 16:808. [PMID: 38398198 PMCID: PMC10887010 DOI: 10.3390/cancers16040808] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/15/2024] [Indexed: 02/25/2024] Open
Abstract
Despite notable advancements in infection prevention and treatment, individuals with hematologic malignancies still face the persistent threat of frequent and life-threatening complications. Those undergoing chemotherapy or other disease-modifying therapies are particularly vulnerable to developing infectious complications, increasing the risk of mortality. Myelodysplastic syndromes (MDS) predominantly affect the elderly, with the incidence rising with age and peaking at around 70 years. Patients with MDS commonly present with unexplained low blood-cell counts, primarily anemia, and often experience varying degrees of neutropenia as the disease progresses. In our subsequent retrospective study involving 1593 patients from the Düsseldorf MDS Registry, we aimed at outlining the incidence of infections in MDS patients and identifying factors contributing to heightened susceptibility to infectious complications in this population.
Collapse
Affiliation(s)
- Annika Kasprzak
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Julia Andresen
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Kathrin Nachtkamp
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Andrea Kündgen
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Felicitas Schulz
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Corinna Strupp
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Colin MacKenzie
- Institute of Medical Microbiology and Hospital Hygiene, University Hospital Duesseldorf, 40225 Duesseldorf, Germany
| | - Jörg Timm
- Institute of Virology, Medical Faculty, Heinrich-Heine-University, 40225 Düsseldorf, Germany
| | - Sascha Dietrich
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Norbert Gattermann
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| | - Ulrich Germing
- Department of Hematology, Oncology and Clinical Immunology, Heinrich-Heine University, 40225 Duesseldorf, Germany (A.K.); (G.K.); (N.G.); (U.G.)
| |
Collapse
|
13
|
Sevoyan A, Mekinian A, Chermat F, Adès L, Ivanyan A, Fenaux P, Hakobyan Y. MDS/CMML from resource-limited region: Characteristics and comparison to tertiary reference European center. Eur J Haematol 2024; 112:296-300. [PMID: 37822035 DOI: 10.1111/ejh.14115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/13/2023]
Abstract
INTRODUCTION Myelodysplastic Syndromes (MDS) and Chronic Myelomonocytic Leukemia (CMML) are clonal myeloid malignancies, characterized by bone marrow failure leading to cytopenias (and possible myeloproliferation for CMML) and a high propensity to evolve to Acute Myeloid Leukemia (AML). OBJECTIVE AND METHODS The aim of our retrospective study was to evaluate the clinical and hematological features; the prevalence of MDS subtypes, R-IPSS, and the outcome of 106 Armenian MDS/CMML patients diagnosed over the 2008-2020 period in a single Armenian Hematology center and compare them to French MDS patients included in the GFM registry. RESULTS Median age in the Armenian cohort was 64 years (range 19-84) and 55% were males. The main MDS subtypes were MDS-MLD (29.2%) and MDS-SLD (27.3%), the least frequent was del 5q (0.9%). By comparison, a higher prevalence of MDS-MLD, MDS-EB2, and MDS-RS was found in the French cohort. Armenian patients' cohort generally had poor access to standard MDS treatment and 42.3% of the patients were transfusion dependent. Overall survival, however, did not significantly differ between Armenian and French cohorts. CONCLUSION Our study stresses issues regarding epidemiology, access to diagnosis, difficulties of risk stratification, and access to treatment.
Collapse
Affiliation(s)
- Anna Sevoyan
- Department of Hematology, Hôpital of Hematology, Yerevan, Armenia
| | - Arsène Mekinian
- Service de Médecine Interne and Inflammation-Immunopathology-Biotherapy Department (DHU i2B), Hôpital Saint-Antoine, Paris, France
- French Armenian research center, Yerevan, Armenia
| | - Fatiha Chermat
- Department of Hematology, Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U944 Institut de Recherche Saint-Louis (IRSL) and Université de Paris, Paris, France
| | - Lionel Adès
- Department of Hematology, Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U944 Institut de Recherche Saint-Louis (IRSL) and Université de Paris, Paris, France
| | - A Ivanyan
- Department of Hematology, Hôpital of Hematology, Yerevan, Armenia
| | - Pierre Fenaux
- Department of Hematology, Hôpital Saint-Louis, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
- INSERM U944 Institut de Recherche Saint-Louis (IRSL) and Université de Paris, Paris, France
| | - Yervand Hakobyan
- Department of Hematology, Hôpital of Hematology, Yerevan, Armenia
| |
Collapse
|
14
|
Panizo Inogés M, Alfonso-Pierola A. [Myelodysplastic neoplasms]. Med Clin (Barc) 2024; 162:77-82. [PMID: 37604730 DOI: 10.1016/j.medcli.2023.07.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 08/23/2023]
Affiliation(s)
- María Panizo Inogés
- Departamento de Hematología y Hemoterapia, Clínica Universidad de Navarra, Pamplona, Navarra, España
| | - Ana Alfonso-Pierola
- Departamento de Hematología y Hemoterapia, Clínica Universidad de Navarra, Pamplona, Navarra, España.
| |
Collapse
|
15
|
Bouchla A, Papageorgiou SG, Symeonidis A, Sakellari I, Zikos P, Thomopoulos TP, Hatzimichael E, Galanopoulos A, Vyniou NA, Kotsianidis I, Pappa V. Evaluation of complete response to azacitidine according to the revised International Working Group 2023 response criteria for higher risk MDS. Does it make a difference in patients' outcome? Leukemia 2023; 37:2517-2519. [PMID: 37816955 PMCID: PMC10681887 DOI: 10.1038/s41375-023-02051-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/12/2023] [Accepted: 09/21/2023] [Indexed: 10/12/2023]
Affiliation(s)
- Anthi Bouchla
- Second Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Sotirios G Papageorgiou
- Second Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | - Argyris Symeonidis
- Department of Internal Medicine, University Hospital of Patras, Rio, Greece
| | - Ioanna Sakellari
- Department of Hematology and Stem cell Transplantation, Georgios Papanicolaou General Hospital, Thessaloniki, Greece
| | - Panagiotis Zikos
- Department of Hematology, Aghios Andreas General Hospital, Patras, Greece
| | - Thomas P Thomopoulos
- Second Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | | | - Ioannis Kotsianidis
- Department of Hematology, Democritus University of Thrace Medical School, Alexandroupolis, Greece
| | - Vasiliki Pappa
- Second Propaedeutic Department of Internal Medicine, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
16
|
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
|
17
|
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
|
18
|
Sallman DA, Al Malki MM, Asch AS, Wang ES, Jurcic JG, Bradley TJ, Flinn IW, Pollyea DA, Kambhampati S, Tanaka TN, Zeidner JF, Garcia-Manero G, Jeyakumar D, Komrokji R, Lancet J, Kantarjian HM, Gu L, Zhang Y, Tan A, Chao M, O'Hear C, Ramsingh G, Lal I, Vyas P, Daver NG. Magrolimab in Combination With Azacitidine in Patients With Higher-Risk Myelodysplastic Syndromes: Final Results of a Phase Ib Study. J Clin Oncol 2023; 41:2815-2826. [PMID: 36888930 PMCID: PMC10414740 DOI: 10.1200/jco.22.01794] [Citation(s) in RCA: 85] [Impact Index Per Article: 42.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 11/21/2022] [Accepted: 01/19/2023] [Indexed: 03/10/2023] Open
Abstract
PURPOSE Magrolimab is a monoclonal antibody that blocks cluster of differentiation 47, a don't-eat-me signal overexpressed on cancer cells. Cluster of differentiation 47 blockade by magrolimab promotes macrophage-mediated phagocytosis of tumor cells and is synergistic with azacitidine, which increases expression of eat-me signals. We report final phase Ib data in patients with untreated higher-risk myelodysplastic syndromes (MDS) treated with magrolimab and azacitidine (ClinicalTrials.gov identifier: NCT03248479). PATIENTS AND METHODS Patients with previously untreated Revised International Prognostic Scoring System intermediate-/high-/very high-risk MDS received magrolimab intravenously as a priming dose (1 mg/kg) followed by ramp-up to a 30 mg/kg once-weekly or once-every-2-week maintenance dose. Azacitidine 75 mg/m2 was administered intravenously/subcutaneously once daily on days 1-7 of each 28-day cycle. Primary end points were safety/tolerability and complete remission (CR) rate. RESULTS Ninety-five patients were treated. Revised International Prognostic Scoring System risk was intermediate/high/very high in 27%, 52%, and 21%, respectively. Fifty-nine (62%) had poor-risk cytogenetics and 25 (26%) had TP53 mutation. The most common treatment-emergent adverse effects included constipation (68%), thrombocytopenia (55%), and anemia (52%). Median hemoglobin change from baseline to first postdose assessment was -0.7 g/dL (range, -3.1 to +2.4). CR rate and overall response rate were 33% and 75%, respectively. Median time to response, duration of CR, duration of overall response, and progression-free survival were 1.9, 11.1, 9.8, and 11.6 months, respectively. Median overall survival (OS) was not reached with 17.1-month follow-up. In TP53-mutant patients, 40% achieved CR with median OS of 16.3 months. Thirty-four patients (36%) had allogeneic stem-cell transplant with 77% 2-year OS. CONCLUSION Magrolimab + azacitidine was well tolerated with promising efficacy in patients with untreated higher-risk MDS, including those with TP53 mutations. A phase III trial of magrolimab/placebo + azacitidine is ongoing (ClinicalTrials.gov identifier: NCT04313881 [ENHANCE]).
Collapse
Affiliation(s)
| | | | - Adam S. Asch
- Stephenson Cancer Center-University of Oklahoma Health Sciences Center, Oklahoma City, OK
| | | | | | - Terrence J. Bradley
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL
| | | | | | | | - Tiffany N. Tanaka
- University of California San Diego Moores Cancer Center, San Diego, CA
| | - Joshua F. Zeidner
- University of North Carolina, Lineberger Comprehensive Cancer Center, Chapel Hill, NC
| | | | | | | | | | | | - Lin Gu
- Gilead Sciences, Inc, Foster City, CA
| | | | | | - Mark Chao
- Gilead Sciences, Inc, Foster City, CA
| | | | | | - Indu Lal
- Gilead Sciences, Inc, Foster City, CA
| | - Paresh Vyas
- MRC Molecular Haematology Unit, Oxford BRC, Department of Hematology, Weatherall Institute of Molecular Medicine, University of Oxford and Oxford University Hospitals NHS Trust, Oxford, United Kingdom
| | - Naval G. Daver
- The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
19
|
Stempel JM, Xie Z, Bewersdorf JP, Stahl M, Zeidan AM. Evolution of Therapeutic Benefit Measurement Criteria in Myelodysplastic Syndromes/Neoplasms. Cancer J 2023; 29:203-211. [PMID: 37195777 DOI: 10.1097/ppo.0000000000000666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
ABSTRACT Myelodysplastic syndromes/neoplasms (MDS) are heterogeneous, clonal myeloid neoplasms characterized by ineffective hematopoiesis, progressive cytopenias, and an increased risk of progression to acute myeloid leukemia. The diversity in disease severity, morphology, and genetic landscape challenges not only novel drug development but also therapeutic response assessment. The MDS International Working Group (IWG) response criteria were first published in the year 2000 focusing on measures of blast burden reduction and hematologic recovery. Despite revision of the IWG criteria in 2006, correlation between IWG-defined responses and patient-focused outcomes, including long-term benefits, remains limited and has potentially contributed to failures of several phase III clinical trials. Several IWG 2006 criteria also lacked clear definitions leading to problems in practical applications and interobserver and intraobserver consistency of response reporting. Although the 2018 revision addressed lower-risk MDS, the most recent update in 2023 redefined responses for higher-risk MDS and has set out to provide clear definitions to enhance consistency while focusing on clinically meaningful outcomes and patient-centered responses. In this review, we analyze the evolution of the MDS response criteria, limitations, and areas of improvement.
Collapse
Affiliation(s)
- Jessica M Stempel
- From the Department of Internal Medicine, Hematology Section, Yale School of Medicine, New Haven, CT
| | - Zhuoer Xie
- Department of Hematology, H. Lee Moffitt Cancer Center, Tampa, FL
| | - Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Harvard University, Boston, MA
| | - Amer M Zeidan
- From the Department of Internal Medicine, Hematology Section, Yale School of Medicine, New Haven, CT
| |
Collapse
|
20
|
Abaza Y, Patel AA. Novel Therapies in Myelodysplastic Syndrome: Where Do Venetoclax and Isocitrate Dehydrogenase Inhibitors Fit in? Cancer J 2023; 29:188-194. [PMID: 37195775 DOI: 10.1097/ppo.0000000000000657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/18/2023]
Abstract
ABSTRACT Myelodysplastic syndromes (MDSs) are a heterogeneous group of clonal hematopoietic stem cell disorders with treatment approaches tailored to the presence of cytopenias, disease risk, and molecular mutation profile. In higher-risk MDSs, the standard of care are DNA methyltransferase inhibitors, otherwise referred to as hypomethylating agents (HMAs), with consideration for allogeneic hematopoietic stem cell transplantation in appropriate candidates. Given modest complete remission rates (15%-20%) with HMA monotherapy and median overall survival of approximately 18 months, there is much interest in the investigation of combination and targeted treatment approaches. Furthermore, there is no standard treatment approach in patients with progression of disease after HMA therapy. In this review, we aim to summarize the current evidence for the B-cell lymphoma-2 inhibitor, venetoclax, and a variety of isocitrate dehydrogenase inhibitors in the treatment of MDSs along with discussing their potential role in the treatment paradigm of this disease.
Collapse
Affiliation(s)
- Yasmin Abaza
- From the Department of Hematology and Oncology, Northwestern University, Robert Lurie Cancer Center
| | - Anand Ashwin Patel
- Section of Hematology/Oncology, Department of Medicine, University of Chicago, IL
| |
Collapse
|
21
|
Montoro MJ, Pomares H, Coll R, Bernal Del Castillo T, Tormo M, Jiménez A, Brunet S, Casaño J, Oiartzabal I, Díez-Campelo M, Ramos F, Romero R, Salido-Fiérrez E, Pedro C, Bargay J, Muñoz-Novas C, López R, Rafel M, Valcárcel D. Evaluation of the outcomes of newly diagnosed patients with high-risk myelodysplastic syndrome according to the initial therapeutical strategies chosen in usual clinical practice. Leuk Lymphoma 2023; 64:679-690. [PMID: 36577016 DOI: 10.1080/10428194.2022.2154604] [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: 12/29/2022]
Abstract
Myelodysplastic syndromes (MDS) are a heterogeneous group of diseases without a care standard and show variability in treatment outcomes. This Spanish, observational, prospective study ERASME (CEL-SMD-2012-01) assessed the evolution of newly diagnosed and treatment-naïve high-risk MDS patients (according to IPPS-R). 204 patients were included: median age 73.0 years, 54.4% males, 69.6% 0-1 ECOG, and 94.6% with comorbidities. Active treatment was the most common strategy (52.0%) vs. stem cell transplantation (25.5%) and supportive care/watchful-waiting (22.5%). Overall (median) event-free survival was 7.9 months (9.1, 8.3, and 5.3); progression-free survival: 10.1 months (12.9, 12.8, and 4.3); and overall survival: 13.8 months (15.4, 14.9; 8.4), respectively, with significant differences among groups. Adverse events (AEs) of ≥3 grade were reported in 72.6% of patients; serious AEs reported in 60.6%. 33.1% of patients died due to AEs. Three patients developed second primary malignant neoplasms (median: 8.2 months). Our study showed better outcomes in patients receiving active therapy early after diagnosis.
Collapse
Affiliation(s)
- Maria Julia Montoro
- Department of Hematology, Hospital Universitari Vall d´Hebron, Barcelona, Spain
| | - Helena Pomares
- ICO-Hospital Duran i Reynals, IDIBELL, L'Hospitalet de Llobregat, Spain
| | - Rosa Coll
- ICO-Hospital Universitari Doctor Josep Trueta, Girona, Spain
| | | | - Mar Tormo
- Hospital Clínico Universitario de Valencia e Instituto de Investigación INCLIVA, Valencia, Spain
| | - Ana Jiménez
- Hospital Universitario Ramón y Cajal, Madrid, Spain
| | - Salut Brunet
- Hospital de la Santa Creu i Sant Pau, Barcelona, Spain
| | - Javier Casaño
- Hospital Universitario Reina Sofía, IMIBIC, Córdoba, Spain
| | | | | | | | - Rafael Romero
- Complejo Hospitalario Universitario de Pontevedra, Pontevedra, Spain
| | | | | | - Joan Bargay
- Hospital Son Llàtzer, Palma de Mallorca, Spain
| | | | - Rocío López
- Celgene S.L.U., a Bristol-Myers Squibb Company, Madrid, Spain
| | | | - David Valcárcel
- Department of Hematology, Hospital Universitari Vall d´Hebron, Barcelona, Spain
| | | |
Collapse
|
22
|
Hasegawa K, Wei AH, Garcia-Manero G, Daver NG, Rajakumaraswamy N, Iqbal S, Chan RJ, Hu H, Tse P, Yan J, Zoratti MJ, Xie F, Sallman DA. Azacitidine Monotherapy in Patients With Treatment-Naïve Higher-risk Myelodysplastic Syndrome: A Systematic Literature Review and Meta-analysis. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2023; 23:127-137. [PMID: 36428152 DOI: 10.1016/j.clml.2022.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 10/26/2022] [Accepted: 11/03/2022] [Indexed: 11/09/2022]
Abstract
BACKGROUND The global incidence of myelodysplastic syndromes (MDS) has been estimated as 0.06 to 0.26/100,000. Since their introduction, hypomethylating agents have played a central role in the treatment of MDS, with heterogeneous real-world outcomes. MATERIALS AND METHODS We assessed and synthesized clinical outcomes of azacitidine (AZA) monotherapy in treatment-naïve patients with higher-risk MDS. A systematic literature review was conducted by searching MEDLINE, Embase, and CENTRAL to identify randomized clinical trials (RCTs) and observational studies, both prospective and retrospective, reporting complete remission (CR), partial remission (PR), overall survival (OS), duration of response (DOR), time-to-response (TTR), and myelosuppressive adverse events (AEs) for patients treated with AZA monotherapy. Noncomparative meta-analyses were used to summarize effects. RESULTS The search identified 3250 abstracts, of which 34 publications describing 16 studies (5 RCTs, 3 prospective, and 8 retrospective observational) were included. Across all studies, pooled CR was 16%; PR was 6%; Median OS was 16.4 months; median DOR was 10.1 months; median TTR was 4.6 months. Proportions of grade 3/4 anemia and thrombocytopenia AEs were 10% and 30%. CONCLUSIONS The effectiveness and efficacy of AZA monotherapy-as measured by CR and median OS-was limited. These findings highlight a significant unmet medical need for effective treatments for patients with higher-risk MDS.
Collapse
Affiliation(s)
| | - Andrew H Wei
- Peter MacCallum Cancer Centre, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Melbourne, VIC
| | | | - Naval G Daver
- University of Texas MD Anderson Cancer Center, Houston, TX
| | | | | | | | - Hao Hu
- Gilead Sciences, Inc., Foster City, CA
| | | | - Jiajun Yan
- McMaster University, Hamilton, ON, Canada
| | | | - Feng Xie
- McMaster University, Hamilton, ON, Canada
| | | |
Collapse
|
23
|
Bazinet A, Darbaniyan F, Jabbour E, Montalban-Bravo G, Ohanian M, Chien K, Kadia T, Takahashi K, Masarova L, Short N, Alvarado Y, Yilmaz M, Ravandi F, Andreeff M, Kanagal-Shamanna R, Ganan-Gomez I, Colla S, Qiao W, Huang X, McCue D, Mirabella B, Kantarjian H, Garcia-Manero G. Azacitidine plus venetoclax in patients with high-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia: phase 1 results of a single-centre, dose-escalation, dose-expansion, phase 1-2 study. Lancet Haematol 2022; 9:e756-e765. [PMID: 36063832 DOI: 10.1016/s2352-3026(22)00216-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Revised: 06/28/2022] [Accepted: 06/30/2022] [Indexed: 12/28/2022]
Abstract
BACKGROUND Therapies beyond hypomethylating agents such as azacitidine are needed in high-risk myelodysplastic syndromes. Venetoclax is an orally bioavailable small molecule BCL-2 inhibitor that is synergistic with hypomethylating agents. We therefore aimed to evaluate the safety, tolerability, and preliminary activity of azacitidine combined with venetoclax for treatment-naive and relapsed or refractory high-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia. METHODS We did a single centre, dose-escalation, dose-expansion, phase 1-2 trial at the University of Texas MD Anderson Cancer Center (Houston, TX, USA). This Article details the phase 1 results. We enrolled patients (≥18 years) with treatment-naive or relapsed or refractory high-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia and bone marrow blasts of more than 5%. No specific Eastern Cooperative Oncology Group status restriction was used. Patients were treated with intravenous or subcutaneous azacitidine (75 mg/m2) for 5 days and oral venetoclax (100-400 mg) for 7-14 days. The primary outcome was safety and tolerability as well as determination of the maximum tolerated dose and recommended phase 2 dose of the azacitidine and venetoclax combination using a 3 + 3 study design. All patients who received one dose of study drug were included in the analyses. This study is registered with ClinicalTrials.gov, number NCT04160052. The phase 2 dose-expansion part of the trial is ongoing. FINDINGS Between Nov 12, 2019, and Dec 17, 2021, a total of 23 patients were enrolled in the phase 1 portion of this study (17 [74%] hypomethylating agent naive and six [26%] post-hypomethylating agent failure). 18 (78%) patients were male and five (22%) were female; 21 (91%) were white and two (9%) were Asian. Median follow-up was 13·2 months (IQR 6·8-18·3). The maximum tolerated dose was not reached and the recommended phase 2 dose was established as azacitidine 75 mg/m2 for 5 days plus venetoclax 400 mg for 14 days. The most common grade 3-4 treatment-emergent adverse events were neutropenia (nine [39%] of 23), thrombocytopenia (nine [39%]), lung infection (seven [30%]), and febrile neutropenia (four [17%]). Three deaths due to sepsis, which were not deemed treatment-related, occurred on the study drugs. The overall response rate was 87% (95% CI 66-97; 20 of 23 patients). INTERPRETATION Azacitidine with venetoclax is safe and shows encouraging activity in patients with high-risk myelodysplastic syndromes or chronic myelomonocytic leukaemia. FUNDING MD Anderson Cancer Center.
Collapse
Affiliation(s)
- Alexandre Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Faezeh Darbaniyan
- Department of Biostatistics, 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
| | | | - Maro Ohanian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kelly Chien
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tapan Kadia
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Koichi Takahashi
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Lucia Masarova
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nicholas Short
- 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
| | - Musa Yilmaz
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad Ravandi
- 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
| | - Rashmi Kanagal-Shamanna
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Irene Ganan-Gomez
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Wei Qiao
- Department of Biostatistics, 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
| | - Deborah McCue
- Pharmacy Division, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bailey Mirabella
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hagop Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
24
|
Adès L, Girshova L, Doronin VA, Díez-Campelo M, Valcárcel D, Kambhampati S, Viniou NA, Woszczyk D, De Paz Arias R, Symeonidis A, Anagnostopoulos A, Munhoz EC, Platzbecker U, Santini V, Fram RJ, Yuan Y, Friedlander S, Faller DV, Sekeres MA. Pevonedistat plus azacitidine vs azacitidine alone in higher-risk MDS/chronic myelomonocytic leukemia or low-blast-percentage AML. Blood Adv 2022; 6:5132-5145. [PMID: 35728048 PMCID: PMC9631625 DOI: 10.1182/bloodadvances.2022007334] [Citation(s) in RCA: 76] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Accepted: 05/03/2022] [Indexed: 11/20/2022] Open
Abstract
PANTHER is a global, randomized phase 3 trial of pevonedistat+azacitidine (n = 227) vs azacitidine monotherapy (n = 227) in patients with newly diagnosed higher-risk myelodysplastic syndromes (MDS; n = 324), higher-risk chronic myelomonocytic leukemia (n = 27), or acute myeloid leukemia (AML) with 20% to 30% blasts (n = 103). The primary end point was event-free survival (EFS). In the intent-to-treat population, the median EFS was 17.7 months with pevonedistat+azacitidine vs 15.7 months with azacitidine (hazard ratio [HR], 0.968; 95% confidence interval [CI], 0.757-1.238; P = .557) and in the higher-risk MDS cohort, median EFS was 19.2 vs 15.6 months (HR, 0.887; 95% CI, 0.659-1.193; P = .431). Median overall survival (OS) in the higher-risk MDS cohort was 21.6 vs 17.5 months (HR, 0.785; P = .092), and in patients with AML with 20% to 30% blasts was 14.5 vs 14.7 months (HR, 1.107; P = .664). In a post hoc analysis, median OS in the higher-risk MDS cohort for patients receiving >3 cycles was 23.8 vs 20.6 months (P = .021) and for >6 cycles was 27.1 vs 22.5 months (P = .008). No new safety signals were identified, and the azacitidine dose intensity was maintained. Common hematologic grade ≥3 treatment emergent adverse events were anemia (33% vs 34%), neutropenia (31% vs 33%), and thrombocytopenia (30% vs 30%). These results underscore the importance of large, randomized controlled trials in these heterogeneous myeloid diseases and the value of continuing therapy for >3 cycles. The trial was registered on clinicaltrials.gov as #NCT03268954.
Collapse
Affiliation(s)
- Lionel Adès
- INSERM U944, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Saint Louis and University of Paris, Paris, France
| | - Larisa Girshova
- Federal Almazov North-West Medical Research Centre, Saint-Petersburg, Russia
| | | | - María Díez-Campelo
- Institute for Biomedical Research of Salamanca (IBSAL), University Hospital of Salamanca, Salamanca, Spain
| | - David Valcárcel
- Hematology Department, Vall Hebron Institute of Oncology (VHIO), Vall d'Hebron, University Hospital, Barcelona, Spain
| | | | - Nora-Athina Viniou
- Hematology Unit, First, Department of Internal Medicine, Laikon General Hospital, National and Kapodistrian University of Athens, Athens, Greece
| | | | | | - Argiris Symeonidis
- Hematology Division, Department of Internal Medicine, University Hospital Patras, Patras, Greece
| | | | | | - Uwe Platzbecker
- Department of Hematology and Cellular Therapy, Leipzig University Hospital, Leipzig, Germany
| | - Valeria Santini
- MDS (Myelodysplastic Syndrome) Unit, Hematology, Azienda Ospedaliero-Universitaria (AOU) Careggi, University of Florence, Florence, Italy
| | - Robert J. Fram
- Takeda Development Center Americas, Inc (TDCA), Lexington, MA; and
| | - Ying Yuan
- Takeda Development Center Americas, Inc (TDCA), Lexington, MA; and
| | | | | | - Mikkael A. Sekeres
- Division of Hematology, Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL
| |
Collapse
|
25
|
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
|
26
|
Adès L, Duployez N, Guerci-Bresler A, Laribi K, Peterlin P, Vey N, Thepot S, Wickenhauser S, Zerazhi H, Stamatoullas A, Wattel E, Recher C, Toma A, Dimicoli-Salazar S, Braun T, Beyne-Rauzy O, Marolleau JP, Cheze S, Park S, Cluzeau T, Nimubona S, Bordessoule D, Benramdane R, Quesnel B, Amé S, de Botton S, Chermat F, Preudhomme C, Chevret S, Fenaux P. A randomised phase II study of azacitidine (AZA) alone or with Lenalidomide (LEN), Valproic acid (VPA) or Idarubicin (IDA) in higher-Risk MDS or low blast AML: GFM's "pick a winner" trial, with the impact of somatic mutations. Br J Haematol 2022; 198:535-544. [PMID: 35438802 DOI: 10.1111/bjh.18193] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/25/2022] [Accepted: 03/28/2022] [Indexed: 11/30/2022]
Abstract
In order to improve the outcome observed with azacitidine (AZA) in higher-risk Myelodysplastic syndrome (MDS), its combination with other drugs in MDS must be evaluated. So far, no combination has not been shown to be more effective than AZA alone. AZA-PLUS was a phase II trial that, in a "pick a winner" approach, randomly assigned patients with higher-risk MDS, CMML and low blast count AML to: AZA; AZA plus lenalidomide; AZA plus Valproic Acid or AZA plus Idarubicin. 322 patients were included. After six cycles, 69 (21.4%) CR + PR were observed with no benefit from any combination. Median EFS and OS were 17.2 and 19.7 months in the whole cohort, respectively, with no difference across randomised arms. Infection and rates of hospitalisation during the first six cycles were higher in the AZA-LEN And AZA-IDA arm, related to increased myelosuppression. Factors associated with better response were IPSS, favourable or intermediate karyotype, haemoglobin, lower circulating blast count, fibrinogen level and lower LDH, while poorer survival was seen in therapy-related MDS and, in the case of TP53, PTPN11 or CSF3R mutation. The combinations used did not improve the outcome obtained with AZA alone. However, our "pick a winner" randomised strategy may remain useful with potentially more active drugs to be tested in combination with AZA.
Collapse
Affiliation(s)
- Lionel Adès
- Hématologie Sénior Hôpital Saint Louis, Assistance publique hôpitaux de paris, and Université de Paris Cité, Paris, France
| | - Nicolas Duployez
- Lille University Hospital, Lille and INSERM UMR-S 1277, Lille, France
| | | | | | - Pierre Peterlin
- Hematology Department, Nantes University Hospital, Nantes, France
| | | | - Sylvain Thepot
- Hematology Department, Angers University Hospital, Angers, France
| | | | - Hacene Zerazhi
- Hematology Department, Centre Hospitalier d'Avignon, Avignon, Fran, France
| | | | - Eric Wattel
- Hematology Department, Lyon University Hospital, Lyon, France
| | - Christian Recher
- Hematology Department, Toulouse University Hospital, Toulouse, France
| | - Andrea Toma
- Hematology Department, Creteil, University Hospital, Creteil, France
| | | | - Thorsten Braun
- Hematology Department, Hôpital Avicenne Assistance publique hôpitaux de paris, and Université Paris 13, Bobigny, France
| | - Odile Beyne-Rauzy
- Hematology Department, Toulouse University Hospital, Toulouse, France
| | | | - Stéphane Cheze
- Hematology Department, Caen, University Hospital, Caen, France
| | - Sophie Park
- Hematology Department, Grenoble, University Hospital, Grenoble, France
| | - Thomas Cluzeau
- Hematology Department, Nice, University Hospital, Nice, France
| | | | | | - Riad Benramdane
- Department of Hematology, Centre Hospitalier, Pontoise, France
| | - Bruno Quesnel
- Univ. Lille, CNRS, Inserm, CHU Lille, UMR9020, UMR-S 1277, Lille, France
| | - Shanti Amé
- Hematology Department, Strasbourg, University Hospital, Strasbourg, France
| | | | | | - Claude Preudhomme
- Lille University Hospital, Lille and INSERM UMR-S 1277, Lille, France
| | - Sylvie Chevret
- Biostatistics and Clinical Epidemiology Research Team (ECSTRRA), INSERM UMR-1153 (CRESS), Université de Paris, Paris, France
| | - Pierre Fenaux
- Hématologie Sénior Hôpital Saint Louis, Assistance publique hôpitaux de paris, and Université de Paris Cité, Paris, France
| |
Collapse
|
27
|
Bazinet A, Bravo GM. New Approaches to Myelodysplastic Syndrome Treatment. Curr Treat Options Oncol 2022; 23:668-687. [PMID: 35320468 DOI: 10.1007/s11864-022-00965-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2022] [Indexed: 12/19/2022]
Abstract
OPINION STATEMENT The treatment of myelodysplastic syndromes (MDS) begins with risk stratification using a validated tool such as the International Prognostic Scoring System (IPSS) or its revised version (IPSS-R). This divides patients into lower- and higher- risk categories. Although treatment objectives in lower-risk MDS (LR-MDS) have traditionally been directed at improving cytopenias (usually anemia) as well as quality of life, recent data supports a potential role for early intervention in delaying transfusion dependency. In addition, careful individualized risk stratification incorporating clinical, cytogenetic, and mutational data might help identify patients at higher-than-expected risk for progression. Given the need for supportive care with red blood cell (RBC) transfusions leading to iron overload, iron chelation should be considered for patients with heavy transfusion requirements at risk for end-organ complications. For patients with LR-MDS and isolated anemia, no high-risk features, and endogenous erythropoietin (EPO) levels below 500 U/L, erythropoiesis-stimulating agents (ESAs) can be attempted to improve anemia. Some LR-MDS patient subgroups may also benefit from specific therapies including luspatercept (MDS with ring sideroblasts), lenalidomide (MDS with deletion 5q), or immunosuppressive therapy (hypocellular MDS). LR-MDS patients failing the above options, or those with multiple cytopenias and/or higher-risk features, can be considered for oral low-dose hypomethylating agent (HMA) therapy. Alternatively, these patients may be enrolled on a clinical trial with promising agents targeting the transforming-growth factor beta (TGF-β) pathway, the hypoxia-inducible factor (HIF) pathway, telomerase activity, inflammatory signaling, or the splicing machinery. In higher-risk MDS (HR-MDS), therapy seeks to modify the natural history of the disease and prolong survival. Eligible patients should be considered for curative allogeneic hematopoietic stem cell transplantation (aHSCT). Despite promising novel combinations, the HMAs azacitidine (AZA) or decitabine (DAC) are still the standard of care for these patients, with intensive chemotherapy-based approaches being a potential option in a small subset of patients. Individuals who fail to respond or progress after HMA experience dismal outcomes and represent a major unmet clinical need. Such patients should be treated as part of a clinical trial if possible. Experimental agents to consider include venetoclax, myeloid cell leukemia 1 (MCL-1) inhibitors, eprenetapopt, CPX-351, immunotherapies (directed towards CD47, TIM3, or CD70), interleukin-1 receptor-associated kinase 4 (IRAK4) inhibitors, pevonedistat, seclidemstat, and eltanexor. In this review, we extensively discuss the current landscape of experimental therapies for both LR- and HR-MDS.
Collapse
Affiliation(s)
- Alexandre Bazinet
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Box 428, Houston, TX, 77030, USA
| | - Guillermo Montalban Bravo
- Department of Leukemia, University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd., Box 428, Houston, TX, 77030, USA.
| |
Collapse
|
28
|
Kim N, Pavletic S, Norsworthy KJ. Meaningful response criteria for myelodysplastic syndromes. Br J Haematol 2021; 196:1137-1148. [PMID: 34628648 DOI: 10.1111/bjh.17838] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2021] [Revised: 08/13/2021] [Accepted: 09/02/2021] [Indexed: 01/11/2023]
Abstract
Standardizing response criteria for myelodysplastic syndromes (MDS), a clinically and biologically heterogeneous group of disorders, has been historically challenging. The International Working Group (IWG) response criteria, first proposed in 2000 and modified in 2006 and 2018, represent the best effort by a group of international experts to define a set of clinically meaningful end-points in MDS. These criteria have been adopted in many MDS clinical trials, allowing for comparisons of response across trials. However, clinical experience has also revealed some limitations of these criteria, and most of the end-points proposed by the IWG require further validation. In this review, we present a critical analysis of the current MDS response criteria from both a practical standpoint and based on currently available clinical trial data. Potential areas for improvement in the criteria are highlighted, which may be considered in future iterations of the response criteria.
Collapse
Affiliation(s)
- Nina Kim
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven Pavletic
- National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Kelly J Norsworthy
- Center for Drug Evaluation and Research, U.S. Food and Drug Administration, Silver Spring, MD, USA
| |
Collapse
|