1
|
Tasis A, Spyropoulos T, Mitroulis I. The Emerging Role of CD8 + T Cells in Shaping Treatment Outcomes of Patients with MDS and AML. Cancers (Basel) 2025; 17:749. [PMID: 40075597 PMCID: PMC11898900 DOI: 10.3390/cancers17050749] [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: 01/13/2025] [Revised: 02/13/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
CD8+ T cells are critical players in anti-tumor immunity against solid tumors, targeted by immunotherapies. Emerging evidence suggests that CD8+ T cells also play a crucial role in anti-tumor responses and determining treatment outcomes in hematologic malignancies like myelodysplastic neoplasms (MDS) and acute myeloid leukemia (AML). In this review, we focus on the implication of CD8+ T cells in the treatment response of patients with MDS and AML. First, we review reported studies of aberrant functionality and clonality of CD8+ T cells in MDS and AML, often driven by the immunosuppressive bone marrow microenvironment, which can hinder effective antitumor immunity. Additionally, we discuss the potential use of CD8+ T cell subpopulations, including memory and senescent-like subsets, as predictive biomarkers for treatment response to a variety of treatment regimens, such as hypomethylating agents, which is the standard of care for patients with higher-risk MDS, and chemotherapy which is the main treatment of patients with AML. Understanding the multifaceted role of CD8+ T cells and their interaction with malignant cells in MDS and AML will provide useful insights into their potential as prognostic/predictive biomarkers, but also uncover alternative approaches to novel treatment strategies that could reshape the therapeutic landscape, thus improving treatment efficacy, aiding in overcoming treatment resistance and improving patient survival in these challenging myeloid neoplasms.
Collapse
Affiliation(s)
- Athanasios Tasis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Theodoros Spyropoulos
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| | - Ioannis Mitroulis
- Translational Research and Laboratory Medicine Unit, First Department of Internal Medicine, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
- Department of Hematology, University Hospital of Alexandroupolis, Democritus University of Thrace, 68100 Alexandroupolis, Greece;
| |
Collapse
|
2
|
Patel SB, Moskop DR, Jordan CT, Pietras EM. Understanding MDS stem cells: Advances and limitations. Semin Hematol 2024; 61:409-419. [PMID: 39472255 DOI: 10.1053/j.seminhematol.2024.09.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 11/10/2024]
Abstract
In work spanning several decades, extensive studies have focused on the properties of malignant stem cells that drive the pathogenesis of acute myeloid leukemia (AML). However, relatively little attention has been devoted to several serious myeloid malignancies that occur prior to the onset of frank leukemia, including myelodysplastic syndrome (MDS). Like leukemia, MDS is hypothesized to arise from a pool of immature malignant stem and progenitor cells (MDS-SCs) that serve as a reservoir for disease evolution and progression1. While multiple studies have sought to identify and characterize the biology and vulnerabilities of MDS-SCs, yet translation of scientific concepts to therapeutically impactful regimens has been limited. Here, we evaluate the currently known properties of MDS-SCs as well as the post-transcriptional mechanisms that drive MDS pathogenesis at a stem and progenitor level. We highlight limits and gaps in our characterization and understanding of MDS-SCs and address the extent to which the properties of MDS-SC are (and can be) inferred from the characterization of LSCs.
Collapse
Affiliation(s)
- Sweta B Patel
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Daniel R Moskop
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO
| | - Craig T Jordan
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| | - Eric M Pietras
- Division of Hematology, University of Colorado Anschutz Medical Campus, Aurora CO.
| |
Collapse
|
3
|
Kapor S, Radojković M, Santibanez JF. Myeloid-derived suppressor cells: Implication in myeloid malignancies and immunotherapy. Acta Histochem 2024; 126:152183. [PMID: 39029317 DOI: 10.1016/j.acthis.2024.152183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/21/2024]
Abstract
Myeloid malignancies stem from a modified hematopoietic stem cell and predominantly include acute myeloid leukemia, myelodysplastic neoplasms, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid-derived suppressor cells (MDSCs) exhibit immunoregulatory properties by governing the innate and adaptive immune systems, creating a permissive and supportive environment for neoplasm growth. This review examines the key characteristics of MDSCs in myeloid malignancies, highlighting that an increased MDSC count corresponds to heightened immunosuppressive capabilities, fostering an immune-tolerant neoplasm microenvironment. Also, this review analyzes and describes the potential of combined cancer therapies, focusing on targeting MDSC generation, expansion, and their inherent immunosuppressive activities to enhance the efficacy of current cancer immunotherapies. A comprehensive understanding of the implications of myeloid malignancies may enhance the exploration of immunotherapeutic strategies for their potential application.
Collapse
Affiliation(s)
- Suncica Kapor
- Department of Hematology, Clinical, and Hospital Center "Dr. Dragiša Mišović-Dedinje,", Heroja Milana Tepića 1, Belgrade 11020, Serbia
| | - Milica Radojković
- Department of Hematology, Clinical, and Hospital Center "Dr. Dragiša Mišović-Dedinje,", Heroja Milana Tepića 1, Belgrade 11020, Serbia; Faculty of Medicine, University of Belgrade, Dr. Subotića Starijeg 8, Belgrade 11000, Serbia
| | - Juan F Santibanez
- Molecular Oncology group, Institute for Medical Research, National Institute of the Republic of Serbia, University of Belgrade, Dr. Subotica 4, POB 102, Belgrade 11129, Serbia; Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O Higgins, General Gana 1780, Santiago 8370854, Chile.
| |
Collapse
|
4
|
Gavrilova T, Schulz E, Mina A. Breaking Boundaries: Immunotherapy for Myeloid Malignancies. Cancers (Basel) 2024; 16:2780. [PMID: 39199554 PMCID: PMC11352449 DOI: 10.3390/cancers16162780] [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: 05/30/2024] [Revised: 07/24/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
Immunotherapy has revolutionized the treatment of myeloid oncologic diseases, particularly for patients resistant to chemotherapy or ineligible for allogeneic stem cell transplantation due to age or fitness constraints. As our understanding of the immunopathogenesis of myeloid malignancies expands, so too do the treatment options available to patients. Immunotherapy in myeloid malignancies, however, faces numerous challenges due to the dynamic nature of the disease, immune dysregulation, and the development of immune evasion mechanisms. This review outlines the progress made in the field of immunotherapy for myeloid malignancies, addresses its challenges, and provides insights into future directions in the field.
Collapse
Affiliation(s)
- Tatyana Gavrilova
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | - Eduard Schulz
- Immune Deficiency—Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.S.); (A.M.)
- NIH Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD 20892, USA
| | - Alain Mina
- Immune Deficiency—Cellular Therapy Program, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892, USA; (E.S.); (A.M.)
- NIH Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD 20892, USA
| |
Collapse
|
5
|
Fredon M, Poussard M, Biichlé S, Bonnefoy F, Mantion CF, Seffar E, Renosi F, Bôle-Richard E, Boidot R, Chevrier S, Anna F, Loustau M, Caumartin J, Gonçalves-Venturelli M, Robinet E, Saas P, Deconinck E, Daguidau E, Roussel X, Godet Y, Adotévi O, Angelot-Delettre F, Galaine J, Garnache-Ottou F. Impact of scFv on Functionality and Safety of Third-Generation CD123 CAR T Cells. Cancer Immunol Res 2024; 12:1090-1107. [PMID: 38819256 DOI: 10.1158/2326-6066.cir-23-0548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2023] [Revised: 12/01/2023] [Accepted: 05/29/2024] [Indexed: 06/01/2024]
Abstract
Chimeric antigen receptor (CAR) T cells express an extracellular domain consisting of a single-chain fragment variable (scFv) targeting a surface tumor-associated antigen. scFv selection should involve safety profiling with evaluation of the efficacy/toxicity balance, especially when the target antigen also is expressed on healthy cells. Here, to assess differences in terms of efficacy and on-target/off-tumor effects, we generated five different CARs targeting CD123 by substituting only the scFv. In in vitro models, T cells engineered to express three of these five CD123 CARs were effectively cytotoxic on leukemic cells without increasing lysis of monocytes or endothelial cells. Using the IncuCyte system, we confirmed the low cytotoxicity of CD123 CAR T cells on endothelial cells. Hematotoxicity evaluation using progenitor culture and CD34 cell lysis showed that two of the five CD123 CAR T cells were less cytotoxic on hematopoietic stem cells. Using a humanized mouse model, we confirmed that CD123- cells were not eliminated by the CD123 CAR T cells. Two CD123 CAR T cells reduced tumor infiltration and increased the overall survival of mice in three in vivo models of blastic plasmacytoid dendritic cell neoplasm. In an aggressive version of this model, bulk RNA sequencing analysis showed that these CD123 CAR T cells upregulated genes associated with cytotoxicity and activation/exhaustion a few days after the injection. Together, these results emphasize the importance of screening different scFvs for the development of CAR constructs to support selection of cells with the optimal risk-benefit ratio for clinical development.
Collapse
Affiliation(s)
- Maxime Fredon
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | - Margaux Poussard
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | - Sabeha Biichlé
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | - Francis Bonnefoy
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | | | - Evan Seffar
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Medical Oncology Department, CHU, Besançon, France
| | - Florian Renosi
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Molecular Onco-Hematology Laboratory, CHU, Besançon, France
| | | | - Romain Boidot
- Department of Tumor Biology and Pathology, Molecular Biology Unit, Georges-François Leclerc Center, Dijon, France
- ICMUB UMR CNRS 6302, Dijon, France
| | - Sandrine Chevrier
- Department of Tumor Biology and Pathology, Molecular Biology Unit, Georges-François Leclerc Center, Dijon, France
| | - François Anna
- Preclinical Department, Invectys, Paris, France
- Molecular Virology and Vaccinology Unit, Pasteur Institute, Paris, France
| | | | | | - Mathieu Gonçalves-Venturelli
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Lymphobank S.A.S.U, Besançon, France
| | | | - Philippe Saas
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | - Eric Deconinck
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Hematology Department, CHU, Besançon, France
| | - Etienne Daguidau
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Hematology Department, CHU, Besançon, France
| | - Xavier Roussel
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Hematology Department, CHU, Besançon, France
| | - Yann Godet
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | - Olivier Adotévi
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Medical Oncology Department, CHU, Besançon, France
| | | | - Jeanne Galaine
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
| | - Francine Garnache-Ottou
- INSERM, EFS BFC, UMR1098-RIGHT, University of Franche-Comté, Besançon, France
- Hematology and Cellular Immunology Laboratory, CHU, Besançon, France
| |
Collapse
|
6
|
Rodriguez-Sevilla JJ, Colla S. T-cell dysfunctions in myelodysplastic syndromes. Blood 2024; 143:1329-1343. [PMID: 38237139 DOI: 10.1182/blood.2023023166] [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: 11/03/2023] [Revised: 12/22/2023] [Accepted: 01/12/2024] [Indexed: 03/25/2024] Open
Abstract
ABSTRACT Escape from immune surveillance is a hallmark of cancer. Immune deregulation caused by intrinsic and extrinsic cellular factors, such as altered T-cell functions, leads to immune exhaustion, loss of immune surveillance, and clonal proliferation of tumoral cells. The T-cell immune system contributes to the pathogenesis, maintenance, and progression of myelodysplastic syndrome (MDS). Here, we comprehensively reviewed our current biological knowledge of the T-cell compartment in MDS and recent advances in the development of immunotherapeutic strategies, such as immune checkpoint inhibitors and T-cell- and antibody-based adoptive therapies that hold promise to improve the outcome of patients with MDS.
Collapse
Affiliation(s)
| | - Simona Colla
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
7
|
Mina A, Greenberg PL, Deeg HJ. How I reduce and treat posttransplant relapse of MDS. Blood 2024; 143:1344-1354. [PMID: 38306658 PMCID: PMC11443576 DOI: 10.1182/blood.2023023005] [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: 10/23/2023] [Revised: 01/12/2024] [Accepted: 01/28/2024] [Indexed: 02/04/2024] Open
Abstract
ABSTRACT Allogeneic hematopoietic stem cell transplantation (HSCT) is the only potentially curative option for patients with high-risk myelodysplastic syndromes (MDS). Advances in conditioning regimens and supportive measures have reduced treatment-related mortality and increased the role of transplantation, leading to more patients undergoing HSCT. However, posttransplant relapse of MDS remains a leading cause of morbidity and mortality for this procedure, necessitating expert management and ongoing results analysis. In this article, we review treatment options and our institutional approaches to managing MDS relapse after HSCT, using illustrative clinical cases that exemplify different clinical manifestations and management of relapse. We address areas of controversy relating to conditioning regimen intensity, chemotherapeutic bridging, and donor selection. In addition, we discuss future directions for advancing the field, including (1) the need for prospective clinical trials separating MDS from acute myeloid leukemia and focusing on posttransplant relapse, as well as (2) the validation of measurable residual disease methodologies to guide timely interventions.
Collapse
Affiliation(s)
- Alain Mina
- Myeloid Malignancies Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD
| | - Peter L. Greenberg
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
8
|
Liu Z, Lei W, Wang H, Liu X, Fu R. Challenges and strategies associated with CAR-T cell therapy in blood malignancies. Exp Hematol Oncol 2024; 13:22. [PMID: 38402232 PMCID: PMC10893672 DOI: 10.1186/s40164-024-00490-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
Cellular immunotherapy, particularly CAR-T cells, has shown potential in the improvement of outcomes in patients with refractory and recurrent malignancies of the blood. However, achieving sustainable long-term complete remission for blood cancer remains a challenge, with resistance and relapse being expected outcomes for many patients. Although many studies have attempted to clarify the mechanisms of CAR-T cell therapy failure, the mechanism remains unclear. In this article, we discuss and describe the current state of knowledge regarding these factors, which include elements that influence the CAR-T cell, cancer cells as a whole, and the microenvironment surrounding the tumor. In addition, we propose prospective approaches to overcome these obstacles in an effort to decrease recurrence rates and extend patient survival subsequent to CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China.
| | - Wenhui Lei
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
- Department of Nephrology, Lishui Municipal Central Hospital, Lishui, Zhejiang, 323000, People's Republic of China
| | - Hao Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
| | - Xiaohan Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China.
| |
Collapse
|
9
|
Vukotić M, Kapor S, Simon F, Cokic V, Santibanez JF. Mesenchymal stromal cells in myeloid malignancies: Immunotherapeutic opportunities. Heliyon 2024; 10:e25081. [PMID: 38314300 PMCID: PMC10837636 DOI: 10.1016/j.heliyon.2024.e25081] [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: 10/05/2023] [Revised: 01/18/2024] [Accepted: 01/19/2024] [Indexed: 02/06/2024] Open
Abstract
Myeloid malignancies are clonal disorders of the progenitor cells or hematopoietic stem cells, including acute myeloid leukemia, myelodysplastic syndromes, myeloproliferative malignancies, and chronic myelomonocytic leukemia. Myeloid neoplastic cells affect the proliferation and differentiation of other hematopoietic lineages in the bone marrow and peripheral blood, leading to severe and life-threatening complications. Mesenchymal stromal cells (MSCs) residing in the bone marrow exert immunosuppressive functions by suppressing innate and adaptive immune systems, thus creating a supportive and tolerant microenvironment for myeloid malignancy progression. This review summarizes the significant features of MSCs in myeloid malignancies, including their role in regulating cell growth, cell death, and antineoplastic resistance, in addition to their immunosuppressive contributions. Understanding the implications of MSCs in myeloid malignancies could pave the path for potential use in immunotherapy.
Collapse
Affiliation(s)
- Milica Vukotić
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Suncica Kapor
- Department of Hematology, Clinical Hospital Center “Dr. Dragisa Misovic-Dedinje,” University of Belgrade, Serbia
| | - Felipe Simon
- Laboratory of Integrative Physiopathology, Faculty of Life Sciences, Universidad Andres Bello, Santiago, Chile
- Millennium Institute on Immunology and Immunotherapy, Santiago, Chile
- Millennium Nucleus of Ion Channel-Associated Diseases, Universidad de Chile, Santiago, Chile
| | - Vladan Cokic
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
| | - Juan F. Santibanez
- Molecular Oncology Group, Institute for Medical Research, University of Belgrade, Belgrade, Serbia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O'Higgins, Santiago, Chile
| |
Collapse
|
10
|
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
|
11
|
Zhang X, Yang X, Ma L, Zhang Y, Wei J. Immune dysregulation and potential targeted therapy in myelodysplastic syndrome. Ther Adv Hematol 2023; 14:20406207231183330. [PMID: 37547364 PMCID: PMC10399277 DOI: 10.1177/20406207231183330] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2022] [Accepted: 06/02/2023] [Indexed: 08/08/2023] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous group of clonal hematological diseases and a high risk for transformation to acute myeloid leukemia (AML). The identification of key genetic alterations in MDS has enhanced our understanding of the pathogenesis and evolution. In recent years, it has been found that both innate and adaptive immune signaling are activated in the hematopoietic niche of MDS with aberrant cytokine secretion in the bone marrow microenvironment. It is also clear that immune dysregulation plays an important role in the occurrence and progression of MDS, especially the destruction of the bone marrow microenvironment, including hematopoiesis and stromal components. The purpose of this review is to explore the role of immune cells, the immune microenvironment, and cytokines in the pathogenesis of MDS. Insights into the mechanisms of these variants may facilitate the development of novel effective treatments to prevent disease progression.
Collapse
Affiliation(s)
- Xiaoying Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xingcheng Yang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Ling Ma
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Yicheng Zhang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education
- National Health Commission (NHC)
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, China
| | - Jia Wei
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, Hubei 430030, China
- Key Laboratory of Organ Transplantation, Ministry of Education
- National Health Commission (NHC)
- Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan, Hubei 430030, China
- Department of Hematology, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, and Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, Shanxi 030032, China
- Sino-German Joint Oncological Research Laboratory, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Taiyuan, Shanxi 030032, China
| |
Collapse
|
12
|
Peng X, Zhu X, Di T, Tang F, Guo X, Liu Y, Bai J, Li Y, Li L, Zhang L. The yin-yang of immunity: Immune dysregulation in myelodysplastic syndrome with different risk stratification. Front Immunol 2022; 13:994053. [PMID: 36211357 PMCID: PMC9537682 DOI: 10.3389/fimmu.2022.994053] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2022] [Accepted: 09/07/2022] [Indexed: 11/13/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous group of myeloid clonal diseases with diverse clinical courses, and immune dysregulation plays an important role in the pathogenesis of MDS. However, immune dysregulation is complex and heterogeneous in the development of MDS. Lower-risk MDS (LR-MDS) is mainly characterized by immune hyperfunction and increased apoptosis, and the immunosuppressive therapy shows a good response. Instead, higher-risk MDS (HR-MDS) is characterized by immune suppression and immune escape, and the immune activation therapy may improve the survival of HR-MDS. Furthermore, the immune dysregulation of some MDS changes dynamically which is characterized by the coexistence and mutual transformation of immune hyperfunction and immune suppression. Taken together, the authors think that the immune dysregulation in MDS with different risk stratification can be summarized by an advanced philosophical thought “Yin-Yang theory” in ancient China, meaning that the opposing forces may actually be interdependent and interconvertible. Clarifying the mechanism of immune dysregulation in MDS with different risk stratification can provide the new basis for diagnosis and clinical treatment. This review focuses on the manifestations and roles of immune dysregulation in the different risk MDS, and summarizes the latest progress of immunotherapy in MDS.
Collapse
Affiliation(s)
- Xiaohuan Peng
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xiaofeng Zhu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Tianning Di
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Futian Tang
- Key Laboratory of the Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Xiaojia Guo
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yang Liu
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Jun Bai
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Yanhong Li
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
| | - Lijuan Li
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| | - Liansheng Zhang
- Department of Hematology, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- Key Laboratory of the Hematology of Gansu Province, Lanzhou University Second Hospital, Lanzhou University, Lanzhou, China
- *Correspondence: Lijuan Li, ; Liansheng Zhang,
| |
Collapse
|
13
|
Laszlo GS, Orozco JJ, Kehret AR, Lunn MC, Huo J, Hamlin DK, Wilbur DS, Dexter SL, Comstock ML, O’Steen S, Sandmaier BM, Green DJ, Walter RB. Development of [ 211At]astatine-based anti-CD123 radioimmunotherapy for acute leukemias and other CD123+ malignancies. Leukemia 2022; 36:1485-1491. [PMID: 35474099 PMCID: PMC9177726 DOI: 10.1038/s41375-022-01580-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 04/14/2022] [Accepted: 04/19/2022] [Indexed: 11/09/2022]
Abstract
Radioimmunotherapy (RIT) has long been pursued to improve outcomes in acute leukemia and higher-risk myelodysplastic syndrome (MDS). Of increasing interest are alpha-particle-emitting radionuclides such as astatine-211 (211At) as they deliver large amounts of radiation over just a few cell diameters, enabling efficient and selective target cell kill. Here, we developed 211At-based RIT targeting CD123, an antigen widely displayed on acute leukemia and MDS cells including underlying neoplastic stem cells. We generated and characterized new murine monoclonal antibodies (mAbs) specific for human CD123 and selected four, all of which were internalized by CD123+ target cells, for further characterization. All mAbs could be conjugated to a boron cage, isothiocyanatophenethyl-ureido-closo-decaborate(2-) (B10), and labeled with 211At. CD123+ cell targeting studies in immunodeficient mice demonstrated specific uptake of 211At-labeled anti-CD123 mAbs in human CD123+ MOLM-13 cell tumors in the flank. In mice injected intravenously with MOLM-13 cells or a CD123NULL MOLM-13 subline, a single dose of up to 40 µCi of 211At delivered via anti-CD123 mAb decreased tumor burdens and substantially prolonged survival dose dependently in mice bearing CD123+ but not CD123- leukemia xenografts, demonstrating potent and target-specific in vivo anti-leukemia efficacy. These data support the further development of 211At-CD123 RIT toward clinical application.
Collapse
Affiliation(s)
- George S. Laszlo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Johnnie J. Orozco
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA
| | - Allie R. Kehret
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Margaret C. Lunn
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Jenny Huo
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Donald K. Hamlin
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | - D. Scott Wilbur
- Department of Radiation Oncology, University of Washington, Seattle, WA
| | - Shannon L. Dexter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Melissa L. Comstock
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Shyril O’Steen
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| | - Brenda M. Sandmaier
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA
| | - Damian J. Green
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, Division of Medical Oncology, University of Washington, Seattle, WA
| | - Roland B. Walter
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, WA, USA,Department of Medicine, Division of Hematology, University of Washington, Seattle, WA,Department of Laboratory Medicine & Pathology, University of Washington, Seattle, WA,Department of Epidemiology, University of Washington, Seattle, WA
| |
Collapse
|
14
|
Zou R, Zhao W, Xiao S, Lu Y. A Signature of Three Apoptosis-Related Genes Predicts Overall Survival in Breast Cancer. Front Surg 2022; 9:863035. [PMID: 35769153 PMCID: PMC9235836 DOI: 10.3389/fsurg.2022.863035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Accepted: 04/25/2022] [Indexed: 12/17/2022] Open
Abstract
Background The commonest malignancy in women is known as breast cancer (BC). Numerous studies demonstrated that apoptosis appears to be critical to the management and clinical outcome of BC patients. The purpose of this study is to explore the potential connection between apoptosis and BC and establish the apoptosis-associated gene signature in BC. Methods The data of BC patient transcripts and related clinical information comes from the Cancer Genome Atlas Database (TCGA), and the genes related to apoptosis come from the Molecular Characterization Database (MSigDB). We identified the abnormally expressed apoptosis-related genes in BC samples. The optimal apoptosis-related genes screened by Cox regression analysis were designed to construct a prognostic model for predicting BC patients. Using the Nom Chart to Predict 1-Year, 3-Year, and 5-Year overall survival for BC patients. The gene signature-related functional pathways were explored by gene set enrichment analysis (GSEA). Results Three genes [alpha subunit of the interleukin 3 receptor (IL3RA), apoptosis-inducing factor mitochondrial-associated 1 (AIFM1), and phosphatidylinositol-3 kinase catalytic alpha (PIK3CA)] correlated with apoptosis were shown to be strongly linked to the overall survival of BC. Survival analysis shows that the risk score is directly proportional to the poor prognosis of BC patients. Risk assessment based on three genetic characteristics (age, pathological stage N, and pathological stage M) can independently predict the prognosis of patients with BC. The Nom chart is most suitable for assessing the long-term survival rate of BC patients. The results of GSEA demonstrated that numerous cell cycle-related pathways were abundant in the high-risk group. Conclusion We constructed an apoptosis-associated gene signature in BC, which had a potential clinical application prospect for BC patients.
Collapse
|
15
|
Mann M, Brunner AM. Emerging immuno-oncology targets in Myelodysplastic Syndromes (MDS). Curr Probl Cancer 2022; 46:100824. [PMID: 34980485 PMCID: PMC8860886 DOI: 10.1016/j.currproblcancer.2021.100824] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 12/03/2021] [Indexed: 02/03/2023]
Abstract
Approaches to immunologic therapies in myelodysplastic syndromes (MDS) have generally fallen into 2 categories: therapies that target immune effector cells and enhance or direct an antileukemic effect, and therapies which target immunological markers on MDS progenitors themselves. Examples of the former include immune checkpoint inhibitors, immunomodulatory therapies, and vaccines, among others, while examples of the latter include antibody-drug conjugate therapies and naked antibodies; while bispecific antibodies and modified T-cells (such as CAR-T therapies) bridge both therapeutic modalities. In this review, we will discuss the rationale for the above therapies, clinical results to date, and potential future directions for investigation.
Collapse
Affiliation(s)
| | - Andrew M. Brunner
- Massachusetts General Hospital Cancer Center, Harvard Medical School
| |
Collapse
|
16
|
Zhang W, Gaikwad H, Groman EV, Purev E, Simberg D, Wang G. Highly aminated iron oxide nanoworms for simultaneous manufacturing and labeling of chimeric antigen receptor T cells. JOURNAL OF MAGNETISM AND MAGNETIC MATERIALS 2022; 541:168480. [PMID: 34720339 PMCID: PMC8553019 DOI: 10.1016/j.jmmm.2021.168480] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2023]
Abstract
Cell based therapies including chimeric antigen receptor (CAR) T cells are promising for treating leukemias and solid cancers. At the same time, there is interest in enhancing the functionality of these cells via surface decoration with nanoparticles (backpacking). Magnetic nanoparticle cell labeling is of particular interest due to opportunities for magnetic separation, in vivo manipulation, drug delivery and magnetic resonance imaging (MRI). While modification of T cells with magnetic nanoparticles (MNPs) was explored before, we questioned whether MNPs are compatible with CAR-T cells when introduced during the manufacturing process. We chose highly aminated 120 nm crosslinked iron oxide nanoworms (CLIO NWs, ~36,000 amines per NW) that could efficiently label different adherent cell lines and we used CD123 CAR-T cells as the labeling model. The CD123 CAR-T cells were produced in the presence of CLIO NWs, CLIO NWs plus protamine sulfate (PS), or PS only. The transduction efficiency of lentiviral CD123 CAR with only NWs was ~23% lower than NW+PS and PS groups (~33% and 35%, respectively). The cell viability from these three transduction conditions was not reduced within CAR-T cell groups, though lower compared to non-transduced T cells (mock T). Use of CLIO NWs instead of, or together with cationic protamine sulfate for enhancement of lentiviral transduction resulted in comparable levels of CAR expression and viability but decreased the proportion of CD8+ cells and increased the proportion of CD4+ cells. CD123 CAR-T transduced in the presence of CLIO NWs, CLIO NWs plus PS, or PS only, showed similar level of cytotoxicity against leukemic cell lines. Furthermore, fluorescence microscopy imaging demonstrated that CD123 CAR-T cells labeled with CLIO NW formed rosettes with CD123+ leukemic cells as the non-labeled CAR-T cells, indicating that the CAR-T targeting to tumor cells has maintained after CLIO NW labeling. The in vivo trafficking of the NW labeled CAR-T cells showed the accumulation of CAR-T labeled with NWs primarily in the bone marrow and spleen. CAR-T cells can be magnetically labeled during their production while maintaining functionality using the positively charged iron oxide NWs, which enable the in vivo biodistribution and tracking of CAR-T cells.
Collapse
Affiliation(s)
- Wei Zhang
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Hanmant Gaikwad
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Ernest V. Groman
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Enkhtsetseg Purev
- Division of Hematology, Department of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
| | - Dmitri Simberg
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Corresponding Authors: (Dmitri Simberg), (Guankui Wang)
| | - Guankui Wang
- Translational Bio-Nanosciences Laboratory, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Colorado Center for Nanomedicine and Nanosafety, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, University of Colorado Anschutz Medical Campus, Aurora, CO, 80045, USA
- Corresponding Authors: (Dmitri Simberg), (Guankui Wang)
| |
Collapse
|
17
|
Hopkins CR, Fraietta JA. Genome Editing as a Vehicle to Drive Successful Chimeric Antigen Receptor T Cell Therapies to the Clinic. EUROPEAN MEDICAL JOURNAL 2021. [DOI: 10.33590/emj/21-000981] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cells have emerged as an effective therapy for patients with relapsed and refractory haematological malignancies. However, there are many challenges preventing clinical efficacy and thus broader translation of this approach. These hurdles include poor autologous T cell fitness, manufacturing issues and lack of conserved tumour-restricted antigens to target. Recent efforts have been directed toward incorporating genome editing technologies to address these challenges and develop potent CAR T cell therapies for a diverse array of haematopoietic cancers. In this review, the authors discuss gene editing strategies that have been employed to augment CAR T cell fitness, generate allogeneic ‘off-the-shelf’ CAR T cell products, and safely target elusive myeloid and T cell cancers that often lack appropriate tumour-specific antigens.
Collapse
Affiliation(s)
- Caitlin R Hopkins
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| | - Joseph A Fraietta
- Department of Microbiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA; Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
| |
Collapse
|
18
|
Comont T, Treiner E, Vergez F. From Immune Dysregulations to Therapeutic Perspectives in Myelodysplastic Syndromes: A Review. Diagnostics (Basel) 2021; 11:diagnostics11111982. [PMID: 34829329 PMCID: PMC8620222 DOI: 10.3390/diagnostics11111982] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/21/2021] [Accepted: 10/22/2021] [Indexed: 12/12/2022] Open
Abstract
The pathophysiology of myelodysplastic syndromes (MDSs) is complex and often includes immune dysregulation of both the innate and adaptive immune systems. Whereas clonal selection mainly involves smoldering inflammation, a cellular immunity dysfunction leads to increased apoptosis and blast proliferation. Addressing immune dysregulations in MDS is a recent concept that has allowed the identification of new therapeutic targets. Several approaches targeting the different actors of the immune system have therefore been developed. However, the results are very heterogeneous, indicating the need to improve our understanding of the disease and interactions between chronic inflammation, adaptive dysfunction, and somatic mutations. This review highlights current knowledge of the role of immune dysregulation in MDS pathophysiology and the field of new drugs.
Collapse
Affiliation(s)
- Thibault Comont
- Department of Internal Medicine, IUCT-Oncopole, Toulouse University Hospital (CHU-Toulouse), 31300 Toulouse, France
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, 31100 Toulouse, France;
- School of Medicine, Université Toulouse III—Paul Sabatier, 31062 Toulouse, France;
- Correspondence: ; Tel.: +33-531-15-62-66; Fax: +33-531-15-62-58
| | - Emmanuel Treiner
- School of Medicine, Université Toulouse III—Paul Sabatier, 31062 Toulouse, France;
- Laboratory of Immunology, Toulouse University Hospital (CHU-Toulouse), 31300 Toulouse, France
- Infinity, Inserm UMR1291, 31000 Toulouse, France
| | - François Vergez
- Cancer Research Center of Toulouse, Unité Mixte de Recherche (UMR) 1037 INSERM, ERL5294 Centre National de La Recherche Scientifique, 31100 Toulouse, France;
- School of Medicine, Université Toulouse III—Paul Sabatier, 31062 Toulouse, France;
- Laboratory of Hematology, IUCT-Oncopole, Toulouse University Hospital (CHU-Toulouse), 31300 Toulouse, France
| |
Collapse
|
19
|
New targets for CAR T therapy in hematologic malignancies. Best Pract Res Clin Haematol 2021; 34:101277. [PMID: 34625226 DOI: 10.1016/j.beha.2021.101277] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 05/30/2021] [Indexed: 12/31/2022]
Abstract
As we expand our acumen of the intricacies of hematological malignancies at a genetic and cellular level, we have paved the way in advancing novel targeted therapeutic avenues such as chimeric antigen receptor T-cell therapies (CAR T). Engineering cells to target a specific antigen has led to dramatic remission rates in cases of relapsed/refractory non-Hodgkin lymphoma, acute lymphoblastic leukemia as well as multiple myeloma thus far with trials in place to further advance targeted therapies in other hematological malignancies. Most currently available CAR T therapies target CD19 antigen. Studies are underway exploring novel CAR T products aimed at other tumor-specific antigens with potential to improve the efficacy and reduce the toxicities. Early studies have confirmed safety and efficacy of CD22 and BCMA targeted CAR T therapies. Moreover, various other targets including CD20, CD30, CD123, kappa, and lambda light chains among others are under clinical investigation as potential avenues of targeted therapy. This review highlights the shift in the treatment paradigm in pursuing diverse antigen targets while addressing the challenges in terms of the efficacy and toxicity of current CAR T-cell therapies.
Collapse
|
20
|
Kapoor S, Champion G, Basu A, Mariampillai A, Olnes MJ. Immune Therapies for Myelodysplastic Syndromes and Acute Myeloid Leukemia. Cancers (Basel) 2021; 13:5026. [PMID: 34638510 PMCID: PMC8507987 DOI: 10.3390/cancers13195026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 09/13/2021] [Accepted: 09/17/2021] [Indexed: 11/16/2022] Open
Abstract
Myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML) are hematologic malignancies arising from the bone marrow. Despite recent advances in treating these diseases, patients with higher-risk MDS and AML continue to have a poor prognosis with limited survival. It has long been recognized that there is an immune component to the pathogenesis of MDS and AML, but until recently, immune therapies have played a limited role in treating these diseases. Immune suppressive therapy exhibits durable clinical responses in selected patients with MDS, but the question of which patients are most suitable for this treatment remains unclear. Over the past decade, there has been remarkable progress in identifying genomic features of MDS and AML, which has led to an improved discernment of the molecular pathogenesis of these diseases. An improved understanding of immune and inflammatory molecular mechanisms of MDS and AML have also recently revealed novel therapeutic targets. Emerging treatments for MDS and AML include monoclonal antibodies such as immune checkpoint inhibitors, bispecific T-cell-engaging antibodies, antibody drug conjugates, vaccine therapies, and cellular therapeutics including chimeric antigen receptor T-cells and NK cells. In this review, we provide an overview of the current understanding of immune dysregulation in MDS and AML and an update on novel immune therapies for these bone marrow malignancies.
Collapse
Affiliation(s)
- Sargam Kapoor
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Grace Champion
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Aparna Basu
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
| | - Anu Mariampillai
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
| | - Matthew J. Olnes
- Hematology and Medical Oncology, Alaska Native Tribal Health Consortium, 3900 Ambassador Dr., Anchorage, AK 99508, USA; (S.K.); (A.B.); (A.M.)
- School of Medicine, University of Washington, 1959 NE Pacific St., Seattle, WA 98195, USA;
- WWAMI School of Medical Education, University of Alaska Anchorage, 3211 Providence Drive, Anchorage, AK 99508, USA
| |
Collapse
|
21
|
Lee P, Yim R, Yung Y, Chu HT, Yip PK, Gill H. Molecular Targeted Therapy and Immunotherapy for Myelodysplastic Syndrome. Int J Mol Sci 2021; 22:10232. [PMID: 34638574 PMCID: PMC8508686 DOI: 10.3390/ijms221910232] [Citation(s) in RCA: 20] [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: 08/31/2021] [Revised: 09/17/2021] [Accepted: 09/21/2021] [Indexed: 12/22/2022] Open
Abstract
Myelodysplastic syndrome (MDS) is a heterogeneous, clonal hematological disorder characterized by ineffective hematopoiesis, cytopenia, morphologic dysplasia, and predisposition to acute myeloid leukemia (AML). Stem cell genomic instability, microenvironmental aberrations, and somatic mutations contribute to leukemic transformation. The hypomethylating agents (HMAs), azacitidine and decitabine are the standard of care for patients with higher-risk MDS. Although these agents induce responses in up to 40-60% of patients, primary or secondary drug resistance is relatively common. To improve the treatment outcome, combinational therapies comprising HMA with targeted therapy or immunotherapy are being evaluated and are under continuous development. This review provides a comprehensive update of the molecular pathogenesis and immune-dysregulations involved in MDS, mechanisms of resistance to HMA, and strategies to overcome HMA resistance.
Collapse
Affiliation(s)
| | | | | | | | | | - Harinder Gill
- Division of Haematology, Medical Oncology and Haemopoietic Stem Cell Transplantation, Department of Medicine, LKS Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (P.L.); (R.Y.); (Y.Y.); (H.-T.C.); (P.-K.Y.)
| |
Collapse
|
22
|
Myelodysplastic Syndromes in the Postgenomic Era and Future Perspectives for Precision Medicine. Cancers (Basel) 2021; 13:cancers13133296. [PMID: 34209457 PMCID: PMC8267785 DOI: 10.3390/cancers13133296] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/18/2021] [Accepted: 06/21/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary With demographic ageing, improved cancer survivorship and increased diagnostic sensitivity, incident cases of patients with Myelodysplastic Syndromes (MDS) are continuously rising, leading to a relevant impact on health care resources. Disease heterogeneity and various comorbidities are challenges for the management of the generally elderly patients. Therefore, experienced physicians and multidisciplinary teams should be involved in the establishment of the correct diagnosis, risk-assessment and personalized treatment plan. Next-generation sequencing allows for early detection of clonal hematopoiesis and monitoring of clonal evolution, but also poses new challenges for its appropriate use. At present, allogeneic hematopoietic stem cell transplantation remains the only curative treatment option for a minority of fit MDS patients. All others receive palliative treatment and will eventually progress, having an unmet need for novel therapies. Targeting compounds are in prospect for precision medicine, however, abrogation of clonal evolution to acute myeloid leukemia remains actually out of reach. Abstract Myelodysplastic syndromes (MDS) represent a heterogeneous group of clonal disorders caused by sequential accumulation of somatic driver mutations in hematopoietic stem and progenitor cells (HSPCs). MDS is characterized by ineffective hematopoiesis with cytopenia, dysplasia, inflammation, and a variable risk of transformation into secondary acute myeloid leukemia. The advent of next-generation sequencing has revolutionized our understanding of the genetic basis of the disease. Nevertheless, the biology of clonal evolution remains poorly understood, and the stochastic genetic drift with sequential accumulation of genetic hits in HSPCs is individual, highly dynamic and hardly predictable. These continuously moving genetic targets pose substantial challenges for the implementation of precision medicine, which aims to maximize efficacy with minimal toxicity of treatments. In the current postgenomic era, allogeneic hematopoietic stem cell transplantation remains the only curative option for younger and fit MDS patients. For all unfit patients, regeneration of HSPCs stays out of reach and all available therapies remain palliative, which will eventually lead to refractoriness and progression. In this review, we summarize the recent advances in our understanding of MDS pathophysiology and its impact on diagnosis, risk-assessment and disease monitoring. Moreover, we present ongoing clinical trials with targeting compounds and highlight future perspectives for precision medicine.
Collapse
|
23
|
Tian Z, Liu M, Zhang Y, Wang X. Bispecific T cell engagers: an emerging therapy for management of hematologic malignancies. J Hematol Oncol 2021; 14:75. [PMID: 33941237 PMCID: PMC8091790 DOI: 10.1186/s13045-021-01084-4] [Citation(s) in RCA: 149] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
Harnessing the power of immune cells, especially T cells, to enhance anti-tumor activities has become a promising strategy in clinical management of hematologic malignancies. The emerging bispecific antibodies (BsAbs), which recruit T cells to tumor cells, exemplified by bispecific T cell engagers (BiTEs), have facilitated the development of tumor immunotherapy. Here we discussed the advances and challenges in BiTE therapy developed for the treatment of hematologic malignancies. Blinatumomab, the first BiTE approved for the treatment of acute lymphocytic leukemia (ALL), is appreciated for its high efficacy and safety. Recent studies have focused on improving the efficacy of BiTEs by optimizing treatment regimens and refining the molecular structures of BiTEs. A considerable number of bispecific T cell-recruiting antibodies which are potentially effective in hematologic malignancies have been derived from BiTEs. The elucidation of mechanisms of BiTE action and neonatal techniques used for the construction of BsAbs can improve the treatment of hematological malignancies. This review summarized the features of bispecific T cell-recruiting antibodies for the treatment of hematologic malignancies with special focus on preclinical experiments and clinical studies.
Collapse
Affiliation(s)
- Zheng Tian
- School of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Ming Liu
- Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China.,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China.,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China
| | - Ya Zhang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| | - Xin Wang
- Department of Hematology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250021, Shandong, China. .,Department of Hematology, Shandong Provincial Hospital Affiliated To Shandong University, Shandong First Medical University, No.324, Jingwu Road, Jinan, 250021, Shandong, China. .,School of Medicine, Shandong University, Jinan, 250012, Shandong, China. .,Shandong Provincial Engineering Research Center of Lymphoma, Jinan, 250021, Shandong, China. .,Branch of National Clinical Research Center for Hematologic Diseases, Jinan, 250021, Shandong, China. .,National Clinical Research Center for Hematologic Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 251006, China.
| |
Collapse
|
24
|
Challenges and Solutions to Bringing Chimeric Antigen Receptor T-Cell Therapy to Myeloid Malignancies. ACTA ACUST UNITED AC 2021; 27:143-150. [PMID: 33750074 DOI: 10.1097/ppo.0000000000000512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
ABSTRACT Myeloid malignancies including myelodysplastic syndromes and acute myeloid leukemia are a group of clonal hematopoietic stem progenitor cell disorders mainly effecting the elderly. Chemotherapeutic approaches improved the outcome in majority of the patients, but it is generally associated with severe toxicities and relapse and does not benefit all the patients. With the success of adoptive cell therapies including chimeric antigen receptor T-cell therapy in treating certain B-cell malignancies, these therapeutic approaches are also being tested for myeloid malignancies, but the preclinical and limited clinical trial data suggest there are significant challenges. The principal hurdle to efficient targeted immunotherapy approaches is the lack of a unique targetable antigen on cancer cells leading to off-target effects including myelosuppression due to depletion of normal myeloid cells. Advanced age of the patients, comorbidities, immunosuppressive bone marrow microenvironment, and cytokine release syndrome are some other challenges that are not unique to myeloid malignancies but pose significant challenge for the successful adaptation of this approach for treatment. In this review, we highlight the challenges and solutions to adopt chimeric antigen receptor T-cell therapies to treat myeloid malignancies.
Collapse
|
25
|
Perriello VM, Gionfriddo I, Rossi R, Milano F, Mezzasoma F, Marra A, Spinelli O, Rambaldi A, Annibali O, Avvisati G, Di Raimondo F, Ascani S, Falini B, Martelli MP, Brunetti L. CD123 Is Consistently Expressed on NPM1-Mutated AML Cells. Cancers (Basel) 2021; 13:cancers13030496. [PMID: 33525388 PMCID: PMC7865228 DOI: 10.3390/cancers13030496] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Revised: 01/20/2021] [Accepted: 01/22/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary One-third of adult acute myeloid leukemia (AML) harbors NPM1 mutations. A deep knowledge of the distribution of selected antigens on the surface of NPM1-mutated AML cells may help optimizing new therapies for this frequent AML subtype. CD123 is known to be expressed on leukemic cells but also on healthy hematopoietic and endothelial cells, although at lower levels. Differences in antigen densities between AML and healthy cells may enlighten therapeutic windows, where targeting CD123 could be effective without triggering “on-target off-tumor” toxicities. Here, we perform a thorough analysis of CD123 expression demonstrating high expression of this antigen on both NPM1-mutated bulk leukemic cells and CD34+CD38− cells. Abstract NPM1-mutated (NPM1mut) acute myeloid leukemia (AML) comprises about 30% of newly diagnosed AML in adults. Despite notable advances in the treatment of this frequent AML subtype, about 50% of NPM1mut AML patients treated with conventional treatment die due to disease progression. CD123 has been identified as potential target for immunotherapy in AML, and several anti-CD123 therapeutic approaches have been developed for AML resistant to conventional therapies. As this antigen has been previously reported to be expressed by NPM1mut cells, we performed a deep flow cytometry analysis of CD123 expression in a large cohort of NPM1mut and wild-type samples, examining the whole blastic population, as well as CD34+CD38− leukemic cells. We demonstrate that CD123 is highly expressed on NPM1mut cells, with particularly high expression levels showed by CD34+CD38− leukemic cells. Additionally, CD123 expression was further enhanced by FLT3 mutations, which frequently co-occur with NPM1 mutations. Our results identify NPM1-mutated and particularly NPM1/FLT3 double-mutated AML as disease subsets that may benefit from anti-CD123 targeted therapies.
Collapse
Affiliation(s)
- Vincenzo Maria Perriello
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Ilaria Gionfriddo
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Roberta Rossi
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Francesca Milano
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Federica Mezzasoma
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Andrea Marra
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
| | - Orietta Spinelli
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy; (O.S.); (A.R.)
| | - Alessandro Rambaldi
- Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, 24127 Bergamo, Italy; (O.S.); (A.R.)
- Department of Oncology and Hematology, University of Milan, 20122 Milan, Italy
| | - Ombretta Annibali
- Hematology and Stem Cell Transplant Unit, Campus Biomedico University Hospital, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Giuseppe Avvisati
- Hematology and Stem Cell Transplant Unit, Campus Biomedico University Hospital, 00128 Rome, Italy; (O.A.); (G.A.)
| | - Francesco Di Raimondo
- Hematology and Bone Marrow Transplant Unit, Catania University Hospital, 95125 Catania, Italy;
| | - Stefano Ascani
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Pathology, Santa Maria Hospital, 05100 Terni, Italy
| | - Brunangelo Falini
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
| | - Maria Paola Martelli
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Correspondence: (M.P.M.); (L.B.)
| | - Lorenzo Brunetti
- Department of Medicine and Surgery, University of Perugia, 06131 Perugia, Italy; (V.M.P.); (I.G.); (R.R.); (F.M.); (F.M.); (A.M.); (S.A.); (B.F.)
- Hematology and Bone Marrow Transplant Unit, Santa Maria della Misericordia Hospital, 06131 Perugia, Italy
- Correspondence: (M.P.M.); (L.B.)
| |
Collapse
|
26
|
El Achi H, Dupont E, Paul S, Khoury JD. CD123 as a Biomarker in Hematolymphoid Malignancies: Principles of Detection and Targeted Therapies. Cancers (Basel) 2020; 12:cancers12113087. [PMID: 33113953 PMCID: PMC7690688 DOI: 10.3390/cancers12113087] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/10/2020] [Accepted: 10/19/2020] [Indexed: 12/15/2022] Open
Abstract
Simple Summary CD123 is overexpressed in multiple hematologic malignancies. Advances in CD123-targeted therapies over the past decade have positioned this molecule as an integral biomarker in current practice. This review provides an overview of CD123 biology and in-depth discussion of clinical laboratory techniques used to determine CD123 expression in various hematolymphoid neoplasms. In addition, we describe various pharmacologic strategies and agents that are available or under evaluation for targeting CD123. Abstract CD123, the α chain of the interleukin 3 receptor, is a cytokine receptor that is overexpressed in multiple hematolymphoid neoplasms, including acute myeloid leukemia, blastic plasmacytoid dendritic cell neoplasm, acute lymphoblastic leukemia, hairy cell leukemia, and systemic mastocytosis. Importantly, CD123 expression is upregulated in leukemic stem cells relative to non-neoplastic hematopoietic stem cells, which makes it a useful diagnostic and therapeutic biomarker in hematologic malignancies. Varying levels of evidence have shown that CD123-targeted therapy represents a promising therapeutic approach in several cancers. Tagraxofusp, an anti-CD123 antibody conjugated to a diphtheria toxin, has been approved for use in patients with blastic plasmacytoid dendritic cell neoplasm. Multiple clinical trials are investigating the use of various CD123-targeting agents, including chimeric antigen receptor-modified T cells (expressing CD123, monoclonal antibodies, combined CD3-CD123 dual-affinity retargeting antibody therapy, recombinant fusion proteins, and CD123-engager T cells. In this review, we provide an overview of laboratory techniques used to evaluate and monitor CD123 expression, describe the strengths and limitations of detecting this biomarker in guiding therapy decisions, and provide an overview of the pharmacologic principles and strategies used in CD123-targeted therapies.
Collapse
Affiliation(s)
- Hanadi El Achi
- Department of Pathology and Laboratory Medicine, The University of Texas at Houston, Houston, TX 77030, USA;
| | - Edouard Dupont
- Faculty of Pharmacy of Paris, Paris Descartes University, 75270 Paris, France;
| | - Shilpa Paul
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Joseph D. Khoury
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
- Correspondence:
| |
Collapse
|
27
|
Pavlovic K, Tristán-Manzano M, Maldonado-Pérez N, Cortijo-Gutierrez M, Sánchez-Hernández S, Justicia-Lirio P, Carmona MD, Herrera C, Martin F, Benabdellah K. Using Gene Editing Approaches to Fine-Tune the Immune System. Front Immunol 2020; 11:570672. [PMID: 33117361 PMCID: PMC7553077 DOI: 10.3389/fimmu.2020.570672] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 08/20/2020] [Indexed: 12/26/2022] Open
Abstract
Genome editing technologies not only provide unprecedented opportunities to study basic cellular system functionality but also improve the outcomes of several clinical applications. In this review, we analyze various gene editing techniques used to fine-tune immune systems from a basic research and clinical perspective. We discuss recent advances in the development of programmable nucleases, such as zinc-finger nucleases (ZFNs), transcription activator-like effector nucleases (TALENs), and clustered regularly interspaced short palindromic repeat (CRISPR)-Cas-associated nucleases. We also discuss the use of programmable nucleases and their derivative reagents such as base editing tools to engineer immune cells via gene disruption, insertion, and rewriting of T cells and other immune components, such natural killers (NKs) and hematopoietic stem and progenitor cells (HSPCs). In addition, with regard to chimeric antigen receptors (CARs), we describe how different gene editing tools enable healthy donor cells to be used in CAR T therapy instead of autologous cells without risking graft-versus-host disease or rejection, leading to reduced adoptive cell therapy costs and instant treatment availability for patients. We pay particular attention to the delivery of therapeutic transgenes, such as CARs, to endogenous loci which prevents collateral damage and increases therapeutic effectiveness. Finally, we review creative innovations, including immune system repurposing, that facilitate safe and efficient genome surgery within the framework of clinical cancer immunotherapies.
Collapse
Affiliation(s)
- Kristina Pavlovic
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - María Tristán-Manzano
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Noelia Maldonado-Pérez
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Marina Cortijo-Gutierrez
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Sabina Sánchez-Hernández
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Pedro Justicia-Lirio
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
- LentiStem Biotech, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - M. Dolores Carmona
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
| | - Concha Herrera
- Maimonides Institute of Biomedical Research in Cordoba (IMIBIC), Cellular Therapy Unit, Reina Sofia University Hospital, University of Cordoba, Cordoba, Spain
- Department of Hematology, Reina Sofía University Hospital, Córdoba, Spain
| | - Francisco Martin
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| | - Karim Benabdellah
- Genomic Medicine Department, GENYO, Centre for Genomics and Oncological Research, Pfizer-University of Granada (Andalusian Regional Government), Health Sciences Technology Park, Granada, Spain
| |
Collapse
|
28
|
Baroni ML, Sanchez Martinez D, Gutierrez Aguera F, Roca Ho H, Castella M, Zanetti SR, Velasco Hernandez T, Diaz de la Guardia R, Castaño J, Anguita E, Vives S, Nomdedeu J, Lapillone H, Bras AE, van der Velden VHJ, Junca J, Marin P, Bataller A, Esteve J, Vick B, Jeremias I, Lopez A, Sorigue M, Bueno C, Menendez P. 41BB-based and CD28-based CD123-redirected T-cells ablate human normal hematopoiesis in vivo. J Immunother Cancer 2020; 8:e000845. [PMID: 32527933 PMCID: PMC7292050 DOI: 10.1136/jitc-2020-000845] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/07/2020] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Acute myeloid leukemia (AML) is a hematopoietic malignancy which is biologically, phenotypically and genetically very heterogeneous. Outcome of patients with AML remains dismal, highlighting the need for improved, less toxic therapies. Chimeric antigen receptor T-cell (CART) immunotherapies for patients with refractory or relapse (R/R) AML are challenging because of the absence of a universal pan-AML target antigen and the shared expression of target antigens with normal hematopoietic stem/progenitor cells (HSPCs), which may lead to life-threating on-target/off-tumor cytotoxicity. CD33-redirected and CD123-redirected CARTs for AML are in advanced preclinical and clinical development, and they exhibit robust antileukemic activity. However, preclinical and clinical controversy exists on whether such CARTs are myeloablative. METHODS We set out to comparatively characterize in vitro and in vivo the efficacy and safety of 41BB-based and CD28-based CARCD123. We analyzed 97 diagnostic and relapse AML primary samples to investigate whether CD123 is a suitable immunotherapeutic target, and we used several xenograft models and in vitro assays to assess the myeloablative potential of our second-generation CD123 CARTs. RESULTS Here, we show that CD123 represents a bona fide target for AML and show that both 41BB-based and CD28-based CD123 CARTs are very efficient in eliminating both AML cell lines and primary cells in vitro and in vivo. However, both 41BB-based and CD28-based CD123 CARTs ablate normal human hematopoiesis and prevent the establishment of de novo hematopoietic reconstitution by targeting both immature and myeloid HSPCs. CONCLUSIONS This study calls for caution when clinically implementing CD123 CARTs, encouraging its preferential use as a bridge to allo-HSCT in patients with R/R AML.
Collapse
Affiliation(s)
- Matteo Libero Baroni
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Diego Sanchez Martinez
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | | | - Heleia Roca Ho
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Maria Castella
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Samanta Romina Zanetti
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Talia Velasco Hernandez
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | | | - Julio Castaño
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
| | - Eduardo Anguita
- Hematology and Hemotherapy Department, Hospital Clinico Universitario San Carlos Instituto Cardiovascular, Madrid, Comunidad de Madrid, Spain
| | - Susana Vives
- Hematology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Catalunya, Spain
| | - Josep Nomdedeu
- Hematology Department, Hospital de la Santa Creu i Sant Pau, Barcelona, Catalunya, Spain
| | - Helene Lapillone
- Centre de Recherce Saint-Antoine, Armand-Trousseau Childrens Hospital, Paris, Île-de-France, France
| | - Anne E Bras
- Immunology Department, Erasmus Medical Center, Rotterdam, Zuid-Holland, Netherlands
| | | | - Jordi Junca
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Hematology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Catalunya, Spain
| | - Pedro Marin
- Hematology Department, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Alex Bataller
- Hematology Department, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Jordi Esteve
- Hematology Department, Hospital Clinic de Barcelona, Barcelona, Catalunya, Spain
| | - Binje Vick
- Helmholtz Center, Munich German Research Center for Environmental Health, Neuherberg, Bayern, Germany
| | - Irmela Jeremias
- Helmholtz Center, Munich German Research Center for Environmental Health, Neuherberg, Bayern, Germany
- Pediatrics Department, Munich University Hospital Dr von Hauner Children's Hospital, Munchen, Bayern, Germany
| | - Angel Lopez
- Human Immunology Department, Centre for Cancer Biology, Adelaide, South Australia, Australia
| | - Marc Sorigue
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Hematology Department, Hospital Universitari Germans Trias i Pujol, Badalona, Catalunya, Spain
| | - Clara Bueno
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Centro de investigación en Red-Oncología, CIBERONC, Comunidad de Madrid, Madrid, Spain
| | - Pablo Menendez
- Biomedicine, Research Institute Against Leukemia Josep Carreras, Barcelona, Catalunya, Spain
- Centro de investigación en Red-Oncología, CIBERONC, Comunidad de Madrid, Madrid, Spain
- Instituciò Catalana de Recerca i Estudis Avançats, ICREA, Barcelona, Catalunya, Spain
| |
Collapse
|
29
|
Bernasconi P, Borsani O. Immune Escape after Hematopoietic Stem Cell Transplantation (HSCT): From Mechanisms to Novel Therapies. Cancers (Basel) 2019; 12:cancers12010069. [PMID: 31881776 PMCID: PMC7016529 DOI: 10.3390/cancers12010069] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2019] [Revised: 12/21/2019] [Accepted: 12/24/2019] [Indexed: 12/21/2022] Open
Abstract
Acute myeloid leukemia (AML) is the most common type of acute leukemia in adults. Recent advances in understanding its molecular basis have opened the way to new therapeutic strategies, including targeted therapies. However, despite an improvement in prognosis it has been documented in recent years (especially in younger patients) that allogenic hematopoietic stem cell transplantation (allo-HSCT) remains the only curative treatment in AML and the first therapeutic option for high-risk patients. After allo-HSCT, relapse is still a major complication, and is observed in about 50% of patients. Current evidence suggests that relapse is not due to clonal evolution, but instead to the ability of the AML cell population to escape immune control by a variety of mechanisms including the altered expression of HLA-molecules, production of anti-inflammatory cytokines, relevant metabolic changes and expression of immune checkpoint (ICP) inhibitors capable of “switching-off” the immune response against leukemic cells. Here, we review the main mechanisms of immune escape and identify potential strategies to overcome these mechanisms.
Collapse
Affiliation(s)
- Paolo Bernasconi
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Hematology Department, Fondazione IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Oscar Borsani
- Department of Molecular Medicine, University of Pavia, 27100 Pavia, Italy
- Correspondence: ; Tel.: +39-340-656-3988
| |
Collapse
|