1
|
Getz TM, Bewersdorf JP, Kewan T, Stempel JM, Bidikian A, Shallis RM, Stahl M, Zeidan AM. Beyond HMAs: Novel Targets and Therapeutic Approaches. Semin Hematol 2024; 61:358-369. [PMID: 39389839 DOI: 10.1053/j.seminhematol.2024.08.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 08/19/2024] [Indexed: 10/12/2024]
Abstract
Myelodysplastic syndromes/neoplasms (MDS) constitute a heterogeneous group of clonal hematopoietic disorders with extremely variable clinical features and outcomes. Management of MDS is largely based on risk stratification of patients into either lower-risk or higher-risk categories using the International Prognostic Scoring System-Revised and, more recently, on the Molecular International Prognostic Scoring System. Lower-risk MDS is often managed with the goal of ameliorating cytopenias and improving quality of life, while higher-risk MDS is treated with therapies aimed at extending survival and delaying progression to acute myeloid leukemia (AML). Therapeutic strategies in lower-risk MDS patients may consist of erythropoiesis stimulating agents, luspatercept, and lenalidomide for selected patients. Furthermore, imetelstat has recently been added to the FDA-approved therapeutic armamentarium for lower-risk MDS. In higher-risk MDS, monotherapy with hypomethylating agents continues to be the standard of care. While several novel hypomethylating agent combinations have and are being studied in large randomized phase 3 clinical trials, including the combination of azacitidine and venetoclax, no combination to date have improved overall survival to azacitidine monotherapy. Moreover, biomarker-directed therapies as well as immonotherapeutic approaches are currently being evaluated in early phase trials. Despite recent advancements, the lack of therapeutic agents, particularly after the failure of first line therapy in higher risk MDS, continues to be a major hurdle in the management of MDS. In this review, we discuss the current treatment landscape of MDS and provide an overview of novel agents currently in clinical development that have the potential to alter our current treatment paradigms.
Collapse
Affiliation(s)
- Ted M Getz
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut.
| | - Jan P Bewersdorf
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut; Department of Medicine, Memorial Sloan Kettering Cancer Center, Leukemia Service, New York, New York
| | - Tariq Kewan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Jessica M Stempel
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Aram Bidikian
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Rory M Shallis
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Amer M Zeidan
- Department of Internal Medicine, Section of Hematology, Yale University and Yale Comprehensive Cancer Center, New Haven, Connecticut
| |
Collapse
|
2
|
Stubbins RJ, Cherniawsky H, Karsan A. Cellular and immunotherapies for myelodysplastic syndromes. Semin Hematol 2024; 61:397-408. [PMID: 39426936 DOI: 10.1053/j.seminhematol.2024.09.006] [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: 08/21/2024] [Accepted: 09/17/2024] [Indexed: 10/21/2024]
Abstract
In this review article, we outline the current landscape of immune and cell therapy-based approaches for patients with myelodysplastic syndromes (MDS). Given the well characterized graft-versus-leukemia (GVL) effect observed with allogeneic hematopoietic cell transplantation, and the known immune escape mechanisms observed in MDS cells, significant interest exists in developing immune-based approaches to treat MDS. These attempts have included antibody-based drugs that block immune escape molecules, such as inhibitors of the PD-1/PD-L1 and TIM-3/galectin-9 axes that mediate interactions between MDS cells and T-lymphocytes, as well as antibodies that block the CD47/SIRPα interaction, which mediates macrophage phagocytosis. Unfortunately, these approaches have been largely unsuccessful. There is significant potential for T-cell engaging therapies and chimeric antigen receptor T (CAR-T) cells, but there are also several limitations to these approaches that are unique to MDS. However, many of these limitations may be overcome by the next generation of cellular therapies, including those with engineered T-cell receptors or natural killer (NK)-cell based platforms. Regardless of the approach, all these immune cells are subject to the complex bone marrow microenvironment in MDS, which harbours a variable and heterogeneous mix of pro-inflammatory cytokines and immunosuppressive elements. Understanding this interaction will be paramount to ensuring the success of immune and cellular therapies in MDS.
Collapse
Affiliation(s)
- Ryan J Stubbins
- Leukemia/BMT Program of BC, BC Cancer, Vancouver V5Z 1M9, BC, Canada; Division of Hematology, Department of Medicine, University of British Columbia, Vancouver V5Z 1M9, BC, Canada.
| | - Hannah Cherniawsky
- Leukemia/BMT Program of BC, BC Cancer, Vancouver V5Z 1M9, BC, Canada; Division of Hematology, Department of Medicine, University of British Columbia, Vancouver V5Z 1M9, BC, Canada
| | - Aly Karsan
- Michael Smith Genome Sciences Centre, BC Cancer, Vancouver, BC V5Z 1L3, Canada.
| |
Collapse
|
3
|
Galassi C, Esteller M, Vitale I, Galluzzi L. Epigenetic control of immunoevasion in cancer stem cells. Trends Cancer 2024; 10:1052-1071. [PMID: 39244477 DOI: 10.1016/j.trecan.2024.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2024] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 09/09/2024]
Abstract
Cancer stem cells (CSCs) are a poorly differentiated population of malignant cells that (at least in some neoplasms) is responsible for tumor progression, resistance to therapy, and disease relapse. According to a widely accepted model, all stages of cancer progression involve the ability of neoplastic cells to evade recognition or elimination by the host immune system. In line with this notion, CSCs are not only able to cope with environmental and therapy-elicited stress better than their more differentiated counterparts but also appear to better evade tumor-targeting immune responses. We summarize epigenetic modifications of DNA and histones through which CSCs evade immune recognition or elimination, and propose that such alterations constitute promising therapeutic targets to increase the sensitivity of some malignancies to immunotherapy.
Collapse
Affiliation(s)
- Claudia Galassi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA
| | - Manel Esteller
- Cancer Epigenetics Group, Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain; Centro de Investigacion Biomedica en Red Cancer (CIBERONC), Madrid, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain; Physiological Sciences Department, School of Medicine and Health Sciences, University of Barcelona (UB), Barcelona, Catalonia, Spain
| | - Ilio Vitale
- Italian Institute for Genomic Medicine, Istituto di Ricovero e Cura a Carattere Scientifico (IRCSS) Candiolo, Torino, Italy; Candiolo Cancer Institute, FPO - IRCCS, Candiolo, Italy.
| | - Lorenzo Galluzzi
- Department of Radiation Oncology, Weill Cornell Medical College, New York, NY, USA; Sandra and Edward Meyer Cancer Center, New York, NY, USA; Caryl and Israel Englander Institute for Precision Medicine, New York, NY, USA.
| |
Collapse
|
4
|
Heidari-Foroozan M, Rezalotfi A, Rezaei N. The molecular landscape of T cell exhaustion in the tumor microenvironment and reinvigoration strategies. Int Rev Immunol 2024; 43:419-440. [PMID: 39257319 DOI: 10.1080/08830185.2024.2401352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/31/2023] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
Immunotherapy has emerged as a promising therapeutic approach for cancer treatment by harnessing the immune system to target cancer cells. However, the efficacy of immunotherapy is hindered by the tumor microenvironment (TME), comprising regulatory T cells (Tregs), macrophages, myeloid-derived suppressor cells (MDSCs), neutrophils, soluble factors (TGF-β, IL-35, IL-10), and hypoxia. These components interact with inhibitory receptors (IRs) on T cells, leading to alterations in T cell transcriptomes, epigenomes, and metabolism, ultimately resulting in T cell exhaustion and compromising the effectiveness of immunotherapy. T cell exhaustion occurs in two phases: pre-exhaustion and exhaustion. Pre-exhausted T cells exhibit reversibility and distinct molecular properties compared to terminally exhausted T cells. Understanding these differences is crucial for designing effective interventions. This comprehensive review summarizes the characteristics of pre-exhausted and exhausted T cells and elucidates the influence of TME components on T cell activity, transcriptomes, epigenomes, and metabolism, ultimately driving T cell exhaustion in cancer. Additionally, potential intervention strategies for reversing exhaustion are discussed. By gaining insights into the mechanisms underlying T cell exhaustion and the impact of the TME, this review aims to inform the development of innovative approaches for combating T cell exhaustion and enhancing the efficacy of immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Mahsa Heidari-Foroozan
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alaleh Rezalotfi
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
5
|
Merz AMA, Platzbecker U. Beyond the horizon: emerging therapeutic approaches in myelodysplastic neoplasms. Exp Hematol 2024; 130:104130. [PMID: 38036096 DOI: 10.1016/j.exphem.2023.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/28/2023] [Accepted: 11/06/2023] [Indexed: 12/02/2023]
Abstract
Management of myelodysplastic neoplasms (MDS) requires a personalized approach, with a focus on improving quality of life and extending lifespan. The International Prognostic Scoring System-Revised and the molecular International Prognostic Scoring System are key tools for risk stratification and management of MDS. They provide a framework for predicting survival and the risk of transformation to acute myeloid leukemia. However, a major challenge in MDS management remains the limited therapeutic options available, especially after the failure of first-line therapies. In lower-risk MDS, the failure of erythropoietin-stimulating agents often leaves few alternatives, although in higher-risk MDS, the prognosis after hypomethylating agent failure is dismal. This highlights the urgent need for novel, more personalized therapeutic approaches. In this review, we discuss emerging novel therapeutic approaches in the treatment of MDS. Several new therapeutic targets are currently being evaluated, offering hope for improved management of MDS in the future.
Collapse
Affiliation(s)
- Almuth Maria Anni Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, University of Leipzig Faculty of Medicine Leipzig, Germany.
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, University of Leipzig Faculty of Medicine Leipzig, Germany.
| |
Collapse
|
6
|
Merz AMA, Sébert M, Sonntag J, Kubasch AS, Platzbecker U, Adès L. Phase to phase: Navigating drug combinations with hypomethylating agents in higher-risk MDS trials for optimal outcomes. Cancer Treat Rev 2024; 123:102673. [PMID: 38176221 DOI: 10.1016/j.ctrv.2023.102673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 12/14/2023] [Accepted: 12/17/2023] [Indexed: 01/06/2024]
Abstract
Recent developments in high-risk Myelodysplastic Neoplasms (HR MDS) treatment are confronted with challenges in study design due to evolving drug combinations with Hypomethylating Agents (HMAs). The shift from the International Prognostic Scoring System (IPSS) to its molecular revision (IPSS-M) has notably influenced research and clinical practice. Introducing concepts like the MDS/AML overlap complicate classifications and including chronic myelomonocytic leukemia (CMML) in MDS studies introduces another layer of complexity. The International Consortium for MDS emphasizes aligning HR MDS criteria with the 2022 ELN criteria for AML. Differences in advancements between AML and MDS treatments and hematological toxicity in HR MDS underline the importance of detailed trial designs. Effective therapeutic strategies require accurate reporting of adverse events, highlighting the need for clarity in criteria like the Common Terminology Criteria for Adverse Events (CTCAE). We provide an overview on negative clinical trials in HR MDS, analyze possible reasons and explore possibilities to optimize future clinical trials in this challenging patient population.
Collapse
Affiliation(s)
- Almuth Maria Anni Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany
| | - Marie Sébert
- Service Hématologie Séniors, Hôpital Saint-Louis (AP-HP), Paris Cité University and INSERM U944, Paris, France
| | - Jan Sonntag
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany
| | - Anne Sophie Kubasch
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectious Disease, University Hospital of Leipzig, Leipzig, Germany.
| | - Lionel Adès
- Service Hématologie Séniors, Hôpital Saint-Louis (AP-HP), Paris Cité University and INSERM U944, Paris, France.
| |
Collapse
|
7
|
Galle P, Finn RS, Mitchell CR, Ndirangu K, Ramji Z, Redhead GS, Pinato DJ. Treatment-emergent antidrug antibodies related to PD-1, PD-L1, or CTLA-4 inhibitors across tumor types: a systematic review. J Immunother Cancer 2024; 12:e008266. [PMID: 38238030 PMCID: PMC10806538 DOI: 10.1136/jitc-2023-008266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/27/2023] [Indexed: 01/23/2024] Open
Abstract
BACKGROUND Increased understanding of how the immune system regulates tumor growth has innovated the use of immunotherapeutics to treat various cancers. The impact of such therapies, including programmed cell death protein 1 (PD-1)/programmed death-ligand 1 (PD-L1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) inhibitors, on the production of antidrug antibodies (ADAs) and their impact on outcomes, is poorly understood. This study aims to evaluate the clinical trial evidence on ADA incidence associated with PD-1, PD-L1, and CTLA-4 inhibitors in the treatment of cancer and to assess associations between treatment administered, ADA incidence, and treatment outcomes. METHODS Embase®, Medline®, and EBM Reviews were searched via the OVID® platform on February 15, 2022. Conference proceedings, clinical trial registries, and global regulatory and reimbursement body websites were also searched. Eligible publications included clinical trials enrolling patients receiving cancer treatment with either PD-1, PD-L1, or CTLA-4 reporting outcomes including incidence or prevalence of ADAs and the impact of immunogenicity on treatment safety and efficacy. Reference lists of eligible publications were also searched. The review was conducted and reported according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses and evidence quality assessment was conducted using the appropriate Joanna Briggs Institute Critical Appraisal tool. RESULTS After screening 4160 records and reviewing 97 full publications, a total of 34 publications reporting on 68 trials were included. A further 41 relevant clinical trials were identified on ClinicalTrials.gov and a further 32 from searches of packaging inserts. In total, 141 relevant trials covering 15 different checkpoint inhibitors and 16 different tumor types were included. Across the included trials, atezolizumab was associated with the highest incidence of ADAs (29.6% of 639 patients), followed by nivolumab (11.2% of 2,085 patients). Combination checkpoint inhibitor treatment appeared to increase the rate of ADAs versus monotherapy. Only 17 trials reported on the impact of ADAs on treatment outcomes with mixed results for the impact of ADAs on treatment efficacy, safety, and pharmacokinetics. CONCLUSIONS Checkpoint inhibitors for the treatment of cancer are immunogenic, with the incidence of treatment-emergent ADAs varying between individual therapies. It remains unclear what impact ADAs have on treatment outcomes.
Collapse
Affiliation(s)
- Peter Galle
- University Medical Centre of the Johannes Gutenberg University Mainz, Mainz, Germany
| | - Richard S Finn
- University of California Los Angeles, Los Angeles, California, USA
| | | | | | | | | | - David J Pinato
- Surgery and Cancer, Imperial College London, London, UK
- Division of Oncology, Department of Translational Medicine, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
8
|
Bilgihan MT, Eryigit AN, Ciftciler R. Efficacy and Safety of Immune Checkpoint Inhibitors in Hematologic Malignancies. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2024; 24:23-31. [PMID: 37863681 DOI: 10.1016/j.clml.2023.09.002] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2023] [Revised: 09/01/2023] [Accepted: 09/05/2023] [Indexed: 10/22/2023]
Abstract
The emergence of immune checkpoint inhibitors (ICIs) has led to a dramatic paradigm shift within the landscape of cancer treatment, igniting significant interest in their potential application in treating hematologic malignancies. This comprehensive review critically has examined the existing body of literature to shed light on the evolving understanding of the efficacy and safety of ICIs, both as a single agent and in combination regimens in hematologic malignancies. Across distinct lymphoma subtypes, the observed treatment responses exhibit diversity, and conflicts. Notably, Hodgkin lymphoma and certain non-Hodgkin lymphomas such as primary mediastinal B-cell lymphoma, emerge as remarkable cases, showing encouraging response rates and outcomes. However, the efficacy of ICIs reveals variations among subtypes such as chronic lymphocytic leukemia and multiple myeloma. Combination therapies consistently demonstrated superior outcomes compared to monotherapy in several malignancies. While the potential benefits of ICIs in hematologic malignancies are evident, the safety profile warrants careful consideration. Immune-related and other adverse events, though generally tolerable and manageable, highlight the necessity of meticulous monitoring and appropriate intervention. The discussions prompted by these findings underscore the need for tailored treatment approaches, driven by disease subtype, patient characteristics, and potential biomarkers. Moreover, the emerging realm of combination therapies involving immune checkpoint inhibitors holds promise for enhanced treatment outcomes, and ongoing research endeavors aim to unravel the optimal strategies.
Collapse
Affiliation(s)
| | | | - Rafiye Ciftciler
- Department of Hematology, Selcuk University Faculty of Medicine, Konya, Turkey.
| |
Collapse
|
9
|
Spillane DR, Assouline S. Immunotherapy for myelodysplastic syndrome and acute myeloid leukemia: where do we stand? Expert Rev Hematol 2023; 16:819-834. [PMID: 37819154 DOI: 10.1080/17474086.2023.2268273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 10/04/2023] [Indexed: 10/13/2023]
Abstract
INTRODUCTION Myelodysplastic syndrome (MDS) and acute myeloid leukemia (AML) are generally characterized by a poor prognosis with currently available therapies. Immunotherapies have already seen success in treating a variety of malignant disorders, and their role in managing myeloid cancers is evolving rapidly. AREAS COVERED This is a review of the immunotherapies tested in MDS and AML, including immune checkpoint inhibitors, bispecific antibodies, and cell therapies such as chimeric antigen receptor (CAR) T cell therapy, T cell receptor (TCR) engineered T cells, and natural killer (NK) cells, with a focus on clinical trials conducted to date and future directions. EXPERT OPINION Initial clinical trials exploring checkpoint inhibitors in MDS and AML have demonstrated high toxicity and disappointing efficacy. However, ongoing trials adding novel checkpoint inhibitors to standard therapy are more promising. Technological advances are improving the outlook for bispecific antibodies, and cellular therapies like adoptive NK cell infusion have favorable efficacy and tolerability in early trials. As our understanding of the immune microenvironment in MDS and AML improves, the role for immunotherapy in the treatment of these diseases will become clearer.
Collapse
Affiliation(s)
- David R Spillane
- Jewish General Hospital, McGill University, Montreal, Québec, Canada
| | - Sarit Assouline
- Jewish General Hospital, McGill University, Montreal, Québec, Canada
| |
Collapse
|
10
|
Wang C, Sallman DA. Therapeutic approaches for the management of higher risk myelodysplastic syndromes. Leuk Lymphoma 2023; 64:511-524. [PMID: 36433645 DOI: 10.1080/10428194.2022.2140287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The heterogeneous nature of myelodysplastic syndromes (MDS) demands a risk-adapted therapeutic approach, and higher risk MDS, characterized by an increased risk of transformation into acute myeloid leukemia and inferior survival, is typically defined based on an integrated assessment of cytopenias, bone marrow blast percentage, and cytogenetic findings using the revised International Prognostic Scoring System. Incorporating mutational data could further refine the risk assessment and identify those with higher-than-expected disease risk. The principal therapeutic goal in this disease subset is to modify the natural history and prolong survival. Allogeneic stem cell transplant, the only potentially curative treatment, should be offered to eligible patients. Hypomethylating agents are the only approved treatment with unsatisfactory response rates and duration, and patients who failed prior hypomethylating agents unfortunately have dismal outcomes with urgent need of novel therapeutic agents. In this review, we provide the therapeutic landscape in higher risk MDS based on the current evidence and discuss the investigational treatment options under development.
Collapse
Affiliation(s)
- Chen Wang
- Department of Internal Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, USA
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
11
|
Abstract
PURPOSE OF REVIEW Therapies that target the immune system are increasingly used across oncology, including in hematologic malignancies such as myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). While allogeneic transplant has been a key therapy in these cancers, new approaches that target the immune system are being explored including immune checkpoint therapies, antibody-drug conjugates, and cellular therapies. RECENT FINDINGS This review outlines updates in the preclinical rationale for immune directed therapies in MDS and AML, as well as recent clinical trials exploring these therapies. This manuscript summarizes the development of therapies targeting T cell immunoglobulin and mucin domain-containing protein 3 (TIM-3) and CD47, which are being evaluated in late phase studies in MDS and AML. It also reviews the landscape of other immune based therapies including antibody-drug conjugates, chimeric antigen receptor-T cells, bispecific antibodies, and tumor vaccines. SUMMARY The treatment landscape in MDS and AML is rapidly changing; with a goal of improving the quality and duration of responses, a number of immune based therapies are under investigation. This review outlines recent advances with these therapies as well as some of the challenges that remain to incorporate them into leukemia care.
Collapse
Affiliation(s)
- Andrew M Brunner
- Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts, USA
| |
Collapse
|
12
|
Rodriguez-Sevilla JJ, Adema V, Garcia-Manero G, Colla S. Emerging treatments for myelodysplastic syndromes: Biological rationales and clinical translation. Cell Rep Med 2023; 4:100940. [PMID: 36787738 PMCID: PMC9975331 DOI: 10.1016/j.xcrm.2023.100940] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/10/2023] [Accepted: 01/20/2023] [Indexed: 02/16/2023]
Abstract
Myelodysplastic syndromes (MDSs) are a heterogeneous group of clonal hematopoietic stem cell disorders characterized by myeloid dysplasia, peripheral blood cytopenias, and increased risk of progression to acute myeloid leukemia (AML). The standard of care for patients with MDS is hypomethylating agent (HMA)-based therapy; however, nearly 50% of patients have no response to the treatment. Patients with MDS in whom HMA therapy has failed have a dismal prognosis and no approved second-line therapy options, so enrollment in clinical trials of experimental agents represents these patients' only chance for improved outcomes. A better understanding of the molecular and biological mechanisms underpinning MDS pathogenesis has enabled the development of new agents that target molecular alterations, cell death regulators, signaling pathways, and immune regulatory proteins in MDS. Here, we review novel therapies for patients with MDS in whom HMA therapy has failed, with an emphasis on the biological rationale for these therapies' development.
Collapse
Affiliation(s)
| | - Vera Adema
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Guillermo Garcia-Manero
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA.
| |
Collapse
|
13
|
Extracellular Vesicles and MicroRNA in Myelodysplastic Syndromes. Cells 2023; 12:cells12040658. [PMID: 36831325 PMCID: PMC9955013 DOI: 10.3390/cells12040658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 02/09/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
The bone marrow niche plays an increasing role in the pathophysiogenesis of myelodysplastic syndromes. More specifically, mesenchymal stromal cells, which can secrete extracellular vesicles and their miRNA contents, modulate the fate of hematopoietic stem cells leading to leukemogenesis. Extracellular vesicles can mediate their miRNA and protein contents between nearby cells but also in the plasma of the patients, being potent tools for diagnosis and prognostic markers in MDS. They can be targeted by antisense miRNA or by modulators of the secretion of extracellular vesicles and could lead to future therapeutic directions in MDS.
Collapse
|
14
|
O’Connell CL, Baer MR, Ørskov AD, Saini SK, Duong VH, Kropf P, Hansen JW, Tsao-Wei D, Jang HS, Emadi A, Holmberg-Thyden S, Cowland J, Brinker BT, Horwood K, Burgos R, Hostetter G, Youngblood BA, Hadrup SR, Issa JP, Jones P, Baylin SB, Siddiqi I, Grønbaek K. Safety, Outcomes, and T-Cell Characteristics in Patients with Relapsed or Refractory MDS or CMML Treated with Atezolizumab in Combination with Guadecitabine. Clin Cancer Res 2022; 28:5306-5316. [PMID: 36222848 PMCID: PMC9772102 DOI: 10.1158/1078-0432.ccr-22-1810] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Revised: 08/17/2022] [Accepted: 10/10/2022] [Indexed: 01/24/2023]
Abstract
PURPOSE We hypothesized that resistance to hypomethylating agents (HMA) among patients with myelodysplastic syndrome (MDS) and chronic myelomonocytic leukemia (CMML) would be overcome by combining a programmed death-ligand 1 antibody with an HMA. PATIENTS AND METHODS We conducted a Phase I/II, multicenter clinical trial for patients with MDS not achieving an International Working Group response after at least 4 cycles of an HMA ("refractory") or progressing after a response ("relapsed") with 3+ or higher risk MDS by the revised International Prognostic Scoring System (IPSS-R) and CMML-1 or -2. Phase I consisted of a 3+3 dose-escalation design beginning with guadecitabine at 30 mg/m2 and escalating to 60 mg/m2 Days 1 to 5 with fixed-dose atezolizumab: 840 mg intravenously Days 8 and 22 of a 28-day cycle. Primary endpoints were safety and tolerability; secondary endpoints were overall response rate (ORR) and survival. RESULTS Thirty-three patients, median age 73 (range 54-85), were treated. Thirty patients had MDS and 3 had CMML, with 30% relapsed and 70% refractory. No dose-limiting toxicities were observed in Phase I. There were 3 (9%) deaths in ≤ 30 days. Five patients (16%) came off study for drug-related toxicity. Immune-related adverse events (IRAE) occurred in 12 (36%) patients (4 grade 3, 3 grade 2, and 5 grade1). ORR was 33% [95% confidence interval (CI), 19%-52%] with 2 complete remission (CR), 3 hematologic improvement, 5 marrow CR, and 1 partial remission. Median overall survival was 15.1 (95% CI, 8.5-25.3) months. CONCLUSIONS Guadecitabine with atezolizumab has modest efficacy with manageable IRAEs and typical cytopenia-related safety concerns for patients with relapsed or refractory MDS and CMML.
Collapse
Affiliation(s)
- Casey L O’Connell
- Jane Anne Nohl Division of Hematology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Maria R Baer
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA,University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Andreas Due Ørskov
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark,Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sunil Kumar Saini
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Vu H. Duong
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA,University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | | | - Jakob Werner Hansen
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark,Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Denice Tsao-Wei
- Department of Preventive Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Hyo Sik Jang
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Ashkan Emadi
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, USA,University of Maryland Greenebaum Comprehensive Cancer Center, Baltimore, MD, USA
| | - Staffan Holmberg-Thyden
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Jack Cowland
- Department of Clinical Genetics, Rigshospitalet, Copenhagen, Denmark
| | - Brett T. Brinker
- Medical Oncology, Cancer and Hematology Centers of West Michigan, Grand Rapids, MI, USA
| | - Kristin Horwood
- Jane Anne Nohl Division of Hematology, Department of Medicine, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Ryan Burgos
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Galen Hostetter
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | | | - Sine Reker Hadrup
- Department of Health Technology, Section of Experimental and Translational Immunology, Technical University of Denmark, Kongens Lyngby, Denmark
| | | | - Peter Jones
- Department of Epigenetics, Van Andel Institute, Grand Rapids, MI, USA
| | - Stephen B Baylin
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Imran Siddiqi
- Department of Pathology, Keck School of Medicine, University of Southern California, Los Angeles, CA, USA
| | - Kirsten Grønbaek
- Department of Hematology, Rigshospitalet, Copenhagen, Denmark,Biotech Research and Innovation Centre, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,The Danish Stem Cell Center (Danstem), Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
15
|
Aubrey BJ, Brunner AM. SOHO State of the Art and Next Questions: Treatment of Higher-Risk Myelodysplastic Syndromes. CLINICAL LYMPHOMA, MYELOMA & LEUKEMIA 2022; 22:869-877. [PMID: 36030175 DOI: 10.1016/j.clml.2022.07.012] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 01/26/2023]
Abstract
Higher-risk myelodysplastic syndromes (MDS) carry a dismal prognosis with rapid disease progression, disease-related complications that impact quality of life, high risk of transformation to acute myeloid leukemia (AML), and poor long-term survival. Higher-risk disease is determined by a number of factors including the depth and type of cytopenias, percentage of myeloblasts occupying the bone marrow, cytogenetic abnormalities, and increasingly also by the presence of higher-risk molecular alterations. In addition to disease characteristics, a patient's performance status and degree of co-morbidity strongly influence treatment decisions and clinical outcomes. A critical first step in the management of patients with higher-risk MDS is evaluating eligibility for allogeneic hematopoietic stem cell transplant (HCT), which currently remains the only curative therapy, and is available to an ever-increasing number of patients. Outside of stem cell transplant, treatment with hypomethylating agent chemotherapy, azacitidine or decitabine, remains the cornerstone of therapy with improvements in overall survival and reduced transformation to AML; however, these approaches are palliative in nature and outcomes remain very poor overall. With a deepening understanding of disease pathophysiology has come a burgeoning array of novel targeted therapies that are currently in pre-clinical and early phase clinical trials offering hope for new treatment options for this malignancy.
Collapse
Affiliation(s)
- Brandon J Aubrey
- Harvard Medical School, Massachusetts General Hospital, Boston, MA
| | - Andrew M Brunner
- Harvard Medical School, Massachusetts General Hospital, Boston, MA.
| |
Collapse
|
16
|
Abaza Y, Zeidan AM. Immune Checkpoint Inhibition in Acute Myeloid Leukemia and Myelodysplastic Syndromes. Cells 2022; 11:cells11142249. [PMID: 35883692 PMCID: PMC9318025 DOI: 10.3390/cells11142249] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/13/2022] [Accepted: 07/15/2022] [Indexed: 11/24/2022] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many solid tumors, with limited progress made in the area of myeloid malignancies. The low mutational burden of acute myeloid leukemia (AML) is one potential reason behind the lack of activity of T-cell harnessing ICIs, particularly CTLA-4 and PD-1 inhibitors. Innate immune checkpoints play a critical role in the immune escape of AML and myelodysplastic syndromes (MDS). The CD47 targeting agent, magrolimab, has shown promising activity when combined with azacitidine in early phase trials conducted in AML and higher-risk MDS, especially among patients harboring a TP53 mutation. Similarly, sabatolimab (an anti-TIM-3 monoclonal antibody) plus hypomethylating agents have shown durable responses in higher-risk MDS and AML in early clinical trials. Randomized trials are currently ongoing to confirm the efficacy of these agents. In this review, we will present the current progress and future directions of immune checkpoint inhibition in AML and MDS.
Collapse
Affiliation(s)
- Yasmin Abaza
- Department of Hematology and Oncology, Northwestern University, Robert H. Lurie Comprehensive Cancer Center, Chicago, IL 60611, USA;
| | - Amer M. Zeidan
- Section of Hematology, Department of Medicine, Smilow Cancer Center, Yale University, New Haven, CT 06511, USA
- Correspondence:
| |
Collapse
|
17
|
Kantarcı EN, Eşkazan AE. Gilteritinib in the management of acute myeloid leukemia: Current evidence and future directions. Leuk Res 2022; 114:106808. [DOI: 10.1016/j.leukres.2022.106808] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 01/29/2022] [Accepted: 02/07/2022] [Indexed: 12/22/2022]
|