1
|
Hope HC, de Sostoa J, Ginefra P, Andreatta M, Chiang YH, Ronet C, Pich-Bavastro C, Corria Osorio J, Kuonen F, Auwerx J, D'Amelio P, Ho PC, Carmona SJ, Coukos G, Migliorini D, Vannini N. Age-associated nicotinamide adenine dinucleotide decline drives CAR-T cell failure. NATURE CANCER 2025:10.1038/s43018-025-00982-7. [PMID: 40394194 DOI: 10.1038/s43018-025-00982-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2024] [Accepted: 04/15/2025] [Indexed: 05/22/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy is one of the most promising cancer treatments. However, different hurdles are limiting its application and efficacy. In this context, how aging influences CAR-T cell outcomes is largely unknown. Here we show that CAR-T cells generated from aged female mice present a mitochondrial dysfunction derived from nicotinamide adenine dinucleotide (NAD) depletion that leads to poor stem-like properties and limited functionality in vivo. Moreover, human data analysis revealed that both age and NAD metabolism determine the responsiveness to CAR-T cell therapy. Targeting NAD pathways, we were able to recover the mitochondrial fitness and functionality of CAR-T cells derived from older adults. Altogether, our study demonstrates that aging is a limiting factor to successful CAR-T cell responses. Repairing metabolic and functional obstacles derived from age, such as NAD decline, is a promising strategy to improve current and future CAR-T cell therapies.
Collapse
Affiliation(s)
- Helen Carrasco Hope
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| | - Jana de Sostoa
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland
| | - Pierpaolo Ginefra
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Massimo Andreatta
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland
| | - Yi-Hsuan Chiang
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Catherine Ronet
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Christine Pich-Bavastro
- Department of Dermatology and Venereology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Jesús Corria Osorio
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland
| | - François Kuonen
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland
- Department of Dermatology and Venereology, Lausanne University Hospital (CHUV) and University of Lausanne, Lausanne, Switzerland
| | - Johan Auwerx
- Laboratory of Integrative and Systems Physiology, Institute of Bioengineering, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Patrizia D'Amelio
- Service of Geriatric Medicine and Geriatric Rehabilitation, Department of Internal Medicine, University of Lausanne Hospital Centre (CHUV), Lausanne, Switzerland
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
| | - Santiago J Carmona
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland
| | - George Coukos
- Department of Oncology, University of Lausanne, Lausanne, Switzerland
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland
- Agora Cancer Research Center, Lausanne, Switzerland
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland
| | - Denis Migliorini
- Department of Oncology, Geneva University Hospitals (HUG), Geneva, Switzerland.
- Agora Cancer Research Center, Lausanne, Switzerland.
- Swiss Cancer Center Léman (SCCL), Lausanne and Geneva, Geneva, Switzerland.
- Center for Translational Research in Onco-Hematology, University of Geneva, Geneva, Switzerland.
| | - Nicola Vannini
- Department of Oncology, University of Lausanne, Lausanne, Switzerland.
- Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| |
Collapse
|
2
|
Cheng J, Zheng J, Ma C, Li Y, Hao H. T-cell senescence: Unlocking the tumor immune "Dark Box" - A multidimensional analysis from mechanism to tumor immunotherapeutic intervention. Semin Cancer Biol 2025; 113:190-209. [PMID: 40381926 DOI: 10.1016/j.semcancer.2025.05.010] [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: 01/04/2025] [Revised: 05/13/2025] [Accepted: 05/14/2025] [Indexed: 05/20/2025]
Abstract
Immunosenescence is the dysfunction of the immune system that occurs with age, a process that is complex and characterized by several features, of which T-cell senescence is one of the key manifestations. In the tumor microenvironment, senescent T cells lead to the inability of tumor cells to be effectively eliminated, triggering immunosuppression, which in turn affects the efficacy of immunotherapy. This is a strong indication that T-cell senescence significantly weakens the immune function of the body, making individuals, especially elderly patients with cancer, more vulnerable to cancer attacks. Despite the many challenges, T-cell senescence is important as a potential therapeutic target. This review provides insights into the molecular mechanisms of T-cell senescence and its research advances in patients with cancer, especially in older adults, and systematically analyzes potential intervention strategies, including molecular mechanism-based interventions, the use of immune checkpoint inhibitors, and CAR-T cell therapy. It is hoped that this will establish a theoretical framework for T-cell senescence in the field of tumor immunology and provide a scientific and prospective reference basis for subsequent in-depth research and clinical practice on senescent T cells.
Collapse
Affiliation(s)
- Jia Cheng
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen 361004, China; Institute of Gastrointestinal Oncology, School of Medicine, Xiamen University, Xiamen 361004, China; Xiamen Municipal Key Laboratory of Gastrointestinal Oncology, Xiamen 361004, China.
| | - Jian Zheng
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China
| | - Chen Ma
- Department of Emergency Internal Medicine, Zibo Central Hospital, Zibo 255024, China
| | - Yongzhang Li
- Department of Urology, Hebei Provincial Hospital of Chinese Medicine, Shijiazhuang 050017, China.
| | - Hua Hao
- Department of Pathology, Yangpu Hospital, School of Medicine, Tongji University, Shanghai 200090, China.
| |
Collapse
|
3
|
de la Iglesia-San Sebastián I, López-Esteban M, Bastos-Oreiro M, Fernández de Córdoba-Oñate S, Gutierrez M, Carbonell D, Bailén R, Gómez-Centurión I, Fernández-Caldas P, Castilla L, Anguita J, Kwon M, García-Sanz R, Buño I, Martínez-Laperche C. Chimeric antigen receptor copies in cell-free DNA predict relapse in aggressive B-cell lymphoma patients treated with CAR T-cell therapy. Br J Haematol 2025; 206:195-203. [PMID: 39668521 DOI: 10.1111/bjh.19916] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 11/11/2024] [Indexed: 12/14/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a transformative treatment for aggressive B-cell lymphomas (ABCL), However, about half of patients relapse, most of them early. This study investigates the detection of CAR copies in circulating cell-free DNA (cfDNA) as a potential predictive biomarker of early relapse (<6 months) to improve patient management. In this research, we have consecutively selected 73 ABCL patients treated with anti-CD19 CAR T-cells, analysing CAR levels in peripheral blood and other clinical variables. Our results indicate that no correlation is present between genomic DNA and cfDNA; moreover, higher levels of CAR-cfDNA on day +14 after infusion (0.44 vs. 0.07; p = 0.019) are associated with improved 6-month progression-free survival rates (74.2% vs. 26%. p < 0.01), suggesting that CAR-cfDNA could be a strong predictor of CAR T-cell therapy short-term outcomes. These findings underscore the potential of integrating CAR-cfDNA analysis into routine clinical practice to enhance the prognostic accuracy and therapeutic strategies for ABCL patients undergoing CAR T-cell therapy.
Collapse
Affiliation(s)
- Ismael de la Iglesia-San Sebastián
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
- Doctorate School, Autonomous University of Madrid, Madrid, Spain
| | - Miguel López-Esteban
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Mariana Bastos-Oreiro
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Sara Fernández de Córdoba-Oñate
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Maravillas Gutierrez
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Diego Carbonell
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Rebeca Bailén
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Ignacio Gómez-Centurión
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Paula Fernández-Caldas
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Lucía Castilla
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Javier Anguita
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Mi Kwon
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Ramón García-Sanz
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Ismael Buño
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
- Department of Cell Biology, Complutense University of Madrid, Madrid, Spain
- Genomics Unit, Hospital General Universitario Gregorio Marañón, Health Research Institute Gregorio Marañón, Madrid, Spain
| | - Carolina Martínez-Laperche
- Department of Hematology, Hospital General Universitario Gregorio Marañón, Madrid, Spain
- Health Research Institute Gregorio Marañón, Madrid, Spain
| |
Collapse
|
4
|
Wenes M, Lepez A, Arinkin V, Maundrell K, Barabas O, Simonetta F, Dutoit V, Romero P, Martinou JC, Migliorini D. A novel mitochondrial pyruvate carrier inhibitor drives stem cell-like memory CAR T cell generation and enhances antitumor efficacy. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200897. [PMID: 39559715 PMCID: PMC11570499 DOI: 10.1016/j.omton.2024.200897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 08/23/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
Adoptive cell transfer with chimeric antigen receptor (CAR)-expressing T cells can induce remarkable complete responses in cancer patients. Therapeutic success has been correlated with central and stem cell-like memory T cell subsets in the infusion product, which are better able to drive efficient CAR T cell in vivo expansion and long-term persistence. We previously reported that inhibition of the mitochondrial pyruvate carrier (MPC) during mouse CAR T cell culture induces a memory phenotype and enhances antitumor efficacy against melanoma. Here, we use a novel MPC inhibitor, MITO-66, which robustly induces a stem cell-like memory phenotype in CD19-CAR T cells generated from healthy donors and patients with relapsed/refractory B cell malignancies. MITO-66-conditioned CAR T cells were superior in controlling human pre-B cell acute lymphoblastic leukemia in mice. Following adoptive cell transfer, MITO-66-conditioned CAR T cells maintained a memory phenotype and protected cured mice against tumor rechallenge. Furthermore, in an in vivo B cell leukemia stress model, CD19-CAR T cells generated in the presence of MITO-66 largely outperformed clinical-stage AKT and PI-3Kδ inhibitors. Thus, we provide compelling preclinical evidence that MPC inhibition with MITO-66 during CAR T cell manufacturing dramatically enhances their antitumor efficacy, thereby paving the way to clinical translation.
Collapse
Affiliation(s)
- Mathias Wenes
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
- MPC Therapeutics, 1206 Geneva, Switzerland
| | - Anouk Lepez
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
| | - Vladimir Arinkin
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Kinsey Maundrell
- MPC Therapeutics, 1206 Geneva, Switzerland
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Orsolya Barabas
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Federico Simonetta
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
- Division of Hematology, Department of Oncology, Geneva University Hospitals (HUG), 1206 Geneva, Switzerland
| | - Valérie Dutoit
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
| | - Pedro Romero
- Faculty of Biology and Medicine, University of Lausanne, 1015 Lausanne, Switzerland
- Novigenix SA, 1066 Epalinges, Switzerland
| | - Jean-Claude Martinou
- MPC Therapeutics, 1206 Geneva, Switzerland
- Department of Molecular and Cellular Biology, University of Geneva, 1206 Geneva, Switzerland
| | - Denis Migliorini
- AGORA Cancer Research Center, 1005 Lausanne, Switzerland
- Swiss Cancer Center Léman, Geneva, Lausanne, Switzerland
- Center for Translational Research in Onco-Hematology, University of Geneva, 1206 Geneva, Switzerland
- Department of Oncology, Geneva University Hospitals (HUG), 1206 Geneva, Switzerland
| |
Collapse
|
5
|
Ullrich F, Bröckelmann PJ, Turki AT, Khan AM, Chiru ED, Vetter M, von Tresckow B, Wirth R, Cordoba R, Ortiz-Maldonado V, Fülöp T, Neuendorff NR. Impact of immunological aging on T cell-mediated therapies in older adults with multiple myeloma and lymphoma. J Immunother Cancer 2024; 12:e009462. [PMID: 39622581 PMCID: PMC11624774 DOI: 10.1136/jitc-2024-009462] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Accepted: 10/24/2024] [Indexed: 12/09/2024] Open
Abstract
The treatment landscape for lymphoma and multiple myeloma, which disproportionally affect older adults, has been transformed by the advent of T cell-mediated immunotherapies, including immune checkpoint inhibition, T cell-engaging bispecific antibodies, and chimeric antigen receptor (CAR) T cell therapy, during the last decade. These treatment modalities re-enable the patient's own immune system to combat malignant cells and offer the potential for sustained remissions and cure for various diseases.Age profoundly affects the physiological function of the immune system. The process of biological aging is largely driven by inflammatory signaling, which is reciprocally fueled by aging-related alterations of physiology and metabolism. In the T cell compartment, aging contributes to T cell senescence and exhaustion, increased abundance of terminally differentiated cells, a corresponding attrition in naïve T cell numbers, and a decrease in the breadth of the receptor repertoire. Furthermore, inflammatory signaling drives aging-related pathologies and contributes to frailty in older individuals. Thus, there is growing evidence of biological aging modulating the efficacy and toxicity of T cell-mediated immunotherapies.Here, we review the available evidence from biological and clinical studies focusing on the relationship between T cell-mediated treatment of hematologic malignancies and age. We discuss biological features potentially impacting clinical outcomes in various scenarios, and potential strategies to improve the safety and efficacy of immune checkpoint inhibitors, T cell-engaging bispecific antibodies, and CAR-T cell therapy in older patients.
Collapse
Affiliation(s)
- Fabian Ullrich
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Paul J Bröckelmann
- Faculty of Medicine and University Hospital of Cologne, Department I of Internal Medicine, Center for Integrated Oncology Aachen Bonn Cologne Düsseldorf (CIO ABCD) and German Hodgkin Study Group (GHSG), University of Cologne, Cologne, Germany
- Max Planck Institute for Biology of Ageing, Cologne, Nordrhein-Westfalen, Germany
| | - Amin T Turki
- Department of Hematology and Oncology, University Hospital Marien Hospital Herne, Herne, Nordrhein-Westfalen, Germany
- Institute for Artificial Intelligence in Medicine, University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Abdullah M Khan
- Division of Hematology, The Ohio State University Comprehensive Cancer Center Arthur G James Cancer Hospital and Richard J Solove Research Institute, Columbus, Ohio, USA
| | - Elena-Diana Chiru
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Marcus Vetter
- Cancer Center Baselland, University of Basel Faculty of Medicine, Basel, Liestal, Switzerland
| | - Bastian von Tresckow
- Department of Hematology and Stem Cell Transplantation, West German Cancer Center and German Cancer Consortium (DKTK partner site Essen), University Duisburg-Essen, University Hospital Essen, Essen, Nordrhein-Westfalen, Germany
| | - Rainer Wirth
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| | - Raul Cordoba
- Department of Hematology, Lymphoma Unit, Hospital Universitario Fundacion Jimenez Diaz, Madrid, Spain
| | - Valentín Ortiz-Maldonado
- Department of Hematology, Oncoimmunotherapy Unit, Hospital Clínic de Barcelona, Barcelona, Spain
| | - Tamas Fülöp
- Department of Medicine, Division of Geriatrics, Research Center on Aging, Université de Sherbrooke, Sherbrooke, Quebec, Canada
| | - Nina Rosa Neuendorff
- Department of Geriatrics, Ruhr University Bochum, University Hospital Marien Hospital Herne, Herne, Germany
| |
Collapse
|
6
|
Penna F, Rubini G, Costelli P. Immunomodulation: A new approach to cancer cachexia, potentially suitable for aging. Mol Aspects Med 2024; 100:101318. [PMID: 39260232 DOI: 10.1016/j.mam.2024.101318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 07/18/2024] [Accepted: 09/05/2024] [Indexed: 09/13/2024]
Abstract
Cancer cachexia is the prototypical example of comorbidity, occurring in most of cancer patients. It is a direct consequence of tumor growth and of the associated inflammatory/immune response. Cachexia can be exacerbated by anti-cancer therapies, frequently resulting in dose limitation and/or treatment delay or discontinuation. The pathogenesis of cancer cachexia is still unclear and includes nutritional, metabolic, hormonal and immunological components. Tumor ability to shape the immune response to its own advantage is now well accepted, while the possibility that such an altered immune response could play a role in the onset of cachexia is still an undefined issue. Indeed, most of the immune-related research on cachexia mainly focused on pro-inflammatory mediators, almost totally disregarding the interactions among immune cells and the homeostasis of peripheral tissues. The present review provides an overview of the immune system dysregulations occurring in cancer cachexia, focusing on the possibility that immunomodulating strategies, mainly developed to stimulate the anti-cancer immune response, could be useful to counteract cachexia as well. Cancer and cachexia are frequent comorbidities of aging. Along this line, cancer- and aging-associated muscle wasting likely coexist in the same patients. Since both conditions share some of the underlying mechanisms, the potential effectiveness of immunomodulation on sarcopenia of aging is discussed.
Collapse
Affiliation(s)
- Fabio Penna
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Giacomo Rubini
- Department of Clinical and Biological Sciences, University of Turin, Italy
| | - Paola Costelli
- Department of Clinical and Biological Sciences, University of Turin, Italy.
| |
Collapse
|
7
|
Zhang X, Liu L. Senescent T Cells: The Silent Culprit in Acute Myeloid Leukemia Progression? Int J Mol Sci 2024; 25:12550. [PMID: 39684260 DOI: 10.3390/ijms252312550] [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: 10/31/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Malignant tumors can evade immune surveillance and elimination through multiple mechanisms, with the induction of immune cell dysfunction serving as a crucial strategy. Mounting evidence indicates that T cell senescence constitutes the primary mechanism underlying T cell dysfunction in acute myeloid leukemia (AML) and represents one of the potential causes of immunotherapy failure. AML usually progresses rapidly and is highly susceptible to drug resistance, thereby resulting in recurrence and patient mortality. Hence, disrupting the immune interface within the bone marrow microenvironment of AML has emerged as a critical objective for synergistically enhancing tumor immunotherapy. In this review, we summarize the general characteristics, distinctive phenotypes, and regulatory signaling networks of senescent T cells and highlight their potential clinical significance in the bone marrow microenvironment of AML. Additionally, we discuss potential therapeutic strategies for alleviating and reversing T cell senescence.
Collapse
Affiliation(s)
- Xiaolan Zhang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lingbo Liu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
8
|
Obajdin J, Larcombe-Young D, Glover M, Kausar F, Hull CM, Flaherty KR, Tan G, Beatson RE, Dunbar P, Mazza R, Bove C, Taylor C, Bille A, Spillane KM, Cozzetto D, Vigilante A, Schurich A, Davies DM, Maher J. Solid tumor immunotherapy using NKG2D-based adaptor CAR T cells. Cell Rep Med 2024; 5:101827. [PMID: 39566469 PMCID: PMC11604534 DOI: 10.1016/j.xcrm.2024.101827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 09/03/2024] [Accepted: 10/21/2024] [Indexed: 11/22/2024]
Abstract
NKG2D ligands (NKG2DLs) are broadly expressed in cancer. To target these, we describe an adaptor chimeric antigen receptor (CAR) termed NKG2D/Dap10-12. Herein, T cells are engineered to co-express NKG2D with a fusion protein that comprises Dap10 joined to a Dap12 endodomain. NKG2D/Dap10-12 T cells elicit compelling efficacy, eradicating or controlling NKG2DL-expressing tumors in several established xenograft models. Importantly, durable responses, long-term survival, and rejection of tumor re-challenge are reproducibly achieved. Efficacy is markedly superior to a clinical stage CAR analog, comprising an NKG2D-CD3ζ fusion. Structure-function analysis using an extended CAR panel demonstrates that potency is dependent on membrane proximity of signaling units, high NKG2D cell surface expression, adaptor structure, provision of exogenous Dap10, and inclusion of one rather than three immune tyrosine activation motifs per signaling unit. Potent therapeutic impact of NKG2D/Dap10-12 T cells is also underpinned by enhanced oxidative phosphorylation, reduced senescence, and transcriptomic re-programming for increased ribosomal biogenesis.
Collapse
Affiliation(s)
- Jana Obajdin
- King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, London SE1 9RT, UK
| | - Daniel Larcombe-Young
- King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, London SE1 9RT, UK
| | - Maya Glover
- Leucid Bio Ltd, Guy's Hospital, London SE1 9RT, UK
| | | | | | - Katie R Flaherty
- King's College London, Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy's Hospital, London SE1 9RT, UK
| | - Ge Tan
- King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, London SE1 9RT, UK
| | - Richard E Beatson
- Department of Respiratory Medicine, Division of Medicinal Sciences, University College London, London, UK
| | | | | | - Camilla Bove
- Leucid Bio Ltd, Guy's Hospital, London SE1 9RT, UK
| | | | - Andrea Bille
- Department of Thoracic Surgery, Guy's and St. Thomas' NHS Trust Foundation, London SE1 9RT, UK
| | | | - Domenico Cozzetto
- Division of Digestive Diseases, Faculty of Medicine, Imperial College London, London W12 0NN, UK
| | - Alessandra Vigilante
- King's College London, Centre for Stem Cells and Regenerative Medicine & Institute for Liver Studies, Guy's Hospital, London SE1 9RT, UK
| | - Anna Schurich
- King's College London, Department of Infectious Diseases, School of Immunology and Microbial Sciences, Guy's Hospital, London SE1 9RT, UK
| | | | - John Maher
- King's College London, School of Cancer and Pharmaceutical Sciences, CAR Mechanics Lab, London SE1 9RT, UK; Leucid Bio Ltd, Guy's Hospital, London SE1 9RT, UK; Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex BN21 2UD, UK.
| |
Collapse
|
9
|
Liu L, Jin F, Fan H. Metabolic tumor volume and the survival of patients with Non-Hodgkin lymphoma treated with chimeric antigen receptor T cell therapy: a meta-analysis. Front Immunol 2024; 15:1433012. [PMID: 39267739 PMCID: PMC11390410 DOI: 10.3389/fimmu.2024.1433012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 08/09/2024] [Indexed: 09/15/2024] Open
Abstract
Background Chimeric antigen receptor T cell (CAR-T) is a promising treatment for aggressive Non-Hodgkin lymphoma (NHL). The aim of the meta-analysis was to determine the association between metabolic tumor volumes (MTV) derived on positron emission tomography before CAR-T infusion and the survival of patients with NHL. Methods Relevant observational studies pertaining to the purpose of the meta-analysis were obtained through a search of PubMed, Web of Science, and Embase from inception of the databases to April 1, 2024. The data was combined using a random-effects model that accounted for the potential influence of between-study heterogeneity. Results Fifteen observational studies were included. Pooled results showed that compared to those with a lower MTV, the NHL patients with a higher MTV before CAR-T infusion were associated with a poor progression-free survival (hazard ratio [HR]: 1.73, 95% confidence interval [CI]: 1.48 to 2.02, p < 0.001; I2 = 20%) and overall survival (HR: 2.11, 95% CI: 1.54 to 2.89, p < 0.001; I2 = 58%). Subgroup analysis showed that the association between MTV and survival of NHL patients after CAR-T was not significantly impacted by study design, methods for determination of MTV cutoff, or analytic models (univariate or multivariate, p for each subgroup all < 0.05). Subgroup analysis suggested a stronger association between MTV and poor survival outcomes in patients with median of lines of previous treatment of 2 or 3 as compared to those of 4 (p for subgroup difference < 0.05). Further meta-regression analyses suggested that the association between MTV and survival was not significantly affected by sample size, age, proportion of men, cutoff value of MTV, follow-up duration, or study quality scores (p all > 0.05). Conclusion A high MTV at baseline is associated with a poor survival of NHL patients after CAR-T. Systematic Review Registration https://inplasy.com/, identifier INPLASY (INPLASY202450069).
Collapse
Affiliation(s)
- Lin Liu
- Department of Hematology, The Forth Affiliated Hospital of China Medical University, Shenyang, China
| | - Feng Jin
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Hua Fan
- Department of Hematology, The Forth Affiliated Hospital of China Medical University, Shenyang, China
| |
Collapse
|
10
|
Nakagawara K, Ando M, Srirat T, Mise-Omata S, Hayakawa T, Ito M, Fukunaga K, Yoshimura A. NR4A ablation improves mitochondrial fitness for long persistence in human CAR-T cells against solid tumors. J Immunother Cancer 2024; 12:e008665. [PMID: 39151930 PMCID: PMC11331892 DOI: 10.1136/jitc-2023-008665] [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: 07/19/2024] [Indexed: 08/19/2024] Open
Abstract
BACKGROUND Antitumor effect of chimeric antigen receptor (CAR)-T cells against solid tumors is limited due to various factors, such as low infiltration rate, poor expansion capacity, and exhaustion of T cells within the tumor. NR4A transcription factors have been shown to play important roles in T-cell exhaustion in mice. However, the precise contribution of each NR4a factor to human T-cell differentiation remains to be clarified. METHODS In this study, we deleted NR4A family factors, NR4A1, NR4A2, and NR4A3, in human CAR-T cells recognizing human epidermal growth factor receptor type 2 (HER2) by using the CRISPR/Cas9 system. We induced T-cell exhaustion in these cells in vitro through repeated co-culturing of CAR-T cells with Her2+A549 lung adenocarcinoma cells and evaluated cell surface markers such as memory and exhaustion phenotypes, proliferative capacity, cytokine production and metabolic activity. We validated the antitumor toxicity of NR4A1/2/3 triple knockout (TKO) CAR-T cells in vivo by transferring CAR-T cells into A549 tumor-bearing immunodeficient mice. RESULTS Human NR4A-TKO CAR-T cells were resistant against exhaustion induced by repeated antigen stimulation in vitro, and maintained higher tumor-killing activity both in vitro and in vivo compared with control CAR-T cells. A comparison of the effectiveness of NR4A single, double, and TKOs demonstrated that triple KO was the most effective in avoiding exhaustion. Furthermore, a strong enhancement of antitumor effects by NR4A TKO was also observed in T cells from various donors including aged persons. Mechanistically, NR4A TKO CAR-T cells showed enhanced mitochondrial oxidative phosphorylation, therefore could persist for longer periods within the tumors. CONCLUSIONS NR4A factors regulate CAR-T cell persistence and stemness through mitochondrial gene expression, therefore NR4A is a highly promising target for the generation of superior CAR-T cells against solid tumors.
Collapse
MESH Headings
- Humans
- Animals
- Mice
- Mitochondria/metabolism
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Nuclear Receptor Subfamily 4, Group A, Member 1/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 1/genetics
- Nuclear Receptor Subfamily 4, Group A, Member 2/metabolism
- Nuclear Receptor Subfamily 4, Group A, Member 2/genetics
- Receptors, Thyroid Hormone/metabolism
- Receptors, Thyroid Hormone/genetics
- Neoplasms/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Xenograft Model Antitumor Assays
- Female
- DNA-Binding Proteins
- Receptors, Steroid
Collapse
Affiliation(s)
- Kensuke Nakagawara
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Ando
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Tanakorn Srirat
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Setsuko Mise-Omata
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| | - Taeko Hayakawa
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Minako Ito
- Division of Allergy and Immunology, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Koichi Fukunaga
- Division of Pulmonary Medicine, Department of Medicine, Keio University School of Medicine, Tokyo, Japan
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
- Research Institute for Biomedical Sciences, Tokyo University of Science, Noda, Chiba, Japan
| |
Collapse
|
11
|
Lelarge V, Capelle R, Oger F, Mathieu T, Le Calvé B. Senolytics: from pharmacological inhibitors to immunotherapies, a promising future for patients' treatment. NPJ AGING 2024; 10:12. [PMID: 38321020 PMCID: PMC10847408 DOI: 10.1038/s41514-024-00138-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/17/2023] [Accepted: 01/17/2024] [Indexed: 02/08/2024]
Abstract
The involvement of cellular senescence in the initiation and propagation of diseases is clearly characterized, making the elimination of senescent cells essential to treat age-related diseases. The development of senolytic drugs demonstrated that targeting these cells limits the deterioration of patients' condition, by inducing apoptosis. Nevertheless, the first generations of senolytics which has been developed displayed their activities through specific mechanisms and demonstrated several limitations during clinical development. However, the rational to eliminate senescent cells remains evident, with the necessity to develop specific therapies in a context of diseases and tissues. The evolutions in the field of drug discovery open the way to a new generation of senolytic therapies, such as immunological approaches (CAR-T cells, Antibody-Drug Conjugated or vaccines), which require preliminary steps of research to identify markers specifically expressed on senescent cells, demonstrating promising specific effects. Currently, the preclinical development of these strategies appears more challenging to avoid strong side effects, but the expected results are commensurate with patients' hopes for treatments. In this review, we highlight the fact that the classical senolytic approach based on drug repurposing display limited efficacy and probably reached its limits in term of clinical development. The recent development of more complex therapies and the extension of interest in the domain of senescence in different fields of research allow to extend the possibility to discover powerful therapies. The future of age-related diseases treatment is linked to the development of new approaches based on cell therapy or immunotherapy to offer the best treatment for patients.
Collapse
Affiliation(s)
- V Lelarge
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - R Capelle
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
| | - F Oger
- University of Lille, Inserm, CHU Lille, Institut Pasteur de Lille, CNRS, U1283 - UMR 8199 - EGID, 59000, Lille, France
| | - T Mathieu
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France
- Synlab, 60/62 Rue d'Hauteville, 75010, Paris, France
| | - B Le Calvé
- StarkAge Therapeutics, Campus de l'Institut Pasteur de Lille, 1 rue du Professeur Calmette, 59800, Lille, France.
| |
Collapse
|