1
|
Donzel M, Trecourt A, Ghesquières H, Nguyen T, Dandash S, Denis M, Cros‐Perrial E, Fontaine J, Dumontet C, Traverse‐Glehen A. Netrin-1 and B-cell maturation antigen expression in a large cohort of 361 lymphomas: sensitive and specific staining in plasmablastic lymphomas, and therapeutic perspectives. J Pathol Clin Res 2025; 11:e70027. [PMID: 40235169 PMCID: PMC12000542 DOI: 10.1002/2056-4538.70027] [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: 12/05/2024] [Revised: 02/25/2025] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
Netrin-1 and B-cell maturation antigen (BCMA) are currently being evaluated as therapeutic targets in oncology. However, studies investigating their expression in mature human lymphoid malignancies are sparse. This study aimed to investigate the expression of BCMA and Netrin-1 in a large cohort of lymphomas to determine their potential role as biomarkers or therapeutic targets. BCMA and Netrin-1 expression was investigated comprehensively using immunohistochemistry in a cohort that included 261 B-cell lymphomas, 45 T-cell lymphomas, and 55 classical Hodgkin lymphomas. Netrin-1 displayed a cytoplasmic staining pattern in plasmablastic lymphomas (27/28, 96%) and classical Hodgkin lymphomas (8/55, 15%). BCMA displayed cytoplasmic staining in most plasmablastic lymphomas (17/20, 85%). Among mature B-cell lymphomas, Netrin-1 and BCMA displayed sensitive (96% and 85%, respectively) and specific (100% and 95%, respectively) staining in plasmablastic lymphomas. These results suggest that these proteins may help pathologists in complex diagnoses and reinforce the interest in developing clinical trials assessing Netrin-1 or BCMA-targeted therapies in plasmablastic lymphoma and classical Hodgkin lymphomas, for which our therapeutic arsenal is weak.
Collapse
Affiliation(s)
- Marie Donzel
- Hospices Civils de LyonInstitut de pathologie multisite, Hôpital Lyon SudLyonFrance
- Université Claude Bernard Lyon 1LyonFrance
- Centre International de Recherche en Infectiologie (CIRI)Institut national de la santé et de la recherche médicale (INSERM) U1111, Centre national de la recherche scientifique (CNRS), UMR5308, Ecole normale supérieure de LyonLyonFrance
| | - Alexis Trecourt
- Hospices Civils de LyonInstitut de pathologie multisite, Hôpital Lyon SudLyonFrance
- Université Claude Bernard Lyon 1LyonFrance
| | - Hervé Ghesquières
- Université Claude Bernard Lyon 1LyonFrance
- Hospices Civils de Lyon, service d'hématologieHôpital Lyon SudLyonFrance
| | | | | | - Morgane Denis
- Hospices Civils de Lyon, service d'hématologieHôpital Lyon SudLyonFrance
- Cancer Research Center of Lyon (CRCL)INSERM 1052/CNRS 5286/University of LyonLyonFrance
| | - Emeline Cros‐Perrial
- Cancer Research Center of Lyon (CRCL)INSERM 1052/CNRS 5286/University of LyonLyonFrance
| | - Juliette Fontaine
- Hospices Civils de LyonInstitut de pathologie multisite, Hôpital Lyon SudLyonFrance
| | - Charles Dumontet
- Université Claude Bernard Lyon 1LyonFrance
- Hospices Civils de Lyon, service d'hématologieHôpital Lyon SudLyonFrance
- Cancer Research Center of Lyon (CRCL)INSERM 1052/CNRS 5286/University of LyonLyonFrance
| | - Alexandra Traverse‐Glehen
- Hospices Civils de LyonInstitut de pathologie multisite, Hôpital Lyon SudLyonFrance
- Université Claude Bernard Lyon 1LyonFrance
| |
Collapse
|
2
|
Ikeda H. Cancer immunotherapy in progress-an overview of the past 130 years. Int Immunol 2025; 37:253-260. [PMID: 39792088 PMCID: PMC11975553 DOI: 10.1093/intimm/dxaf002] [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: 05/07/2024] [Accepted: 01/09/2025] [Indexed: 01/12/2025] Open
Abstract
Since the first approval of an immune checkpoint inhibitor, we have witnessed the clinical success of cancer immunotherapy. Adoptive T-cell therapy with chimeric antigen receptor T (CAR-T) cells has shown remarkable efficacy in hematological malignancies. Concurrently with these successes, the cancer immunoediting concept that refined the cancer immunosurveillance concept underpinned the scientific mechanism and reason for past failures, as well as recent breakthroughs in cancer immunotherapy. Now, we face the next step of issues to be solved in this field, such as tumor heterogeneity, the tumor microenvironment, the metabolism of tumors and the immune system, and personalized approaches for patients, aiming to expand the population benefitted by the therapies.
Collapse
Affiliation(s)
- Hiroaki Ikeda
- Department of Oncology, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan
- Leading Medical Research Core Unit, Nagasaki University Graduate School of Biomedical Sciences, 1-12-4, Sakamoto, Nagasaki 852-8523, Japan
| |
Collapse
|
3
|
Leleux J, Rosenberg J, Sonzogni O, Walker RL, Venkitaraman A, Garrison SM, Jin N, Gregory PD, Jarjour J. RESET: A TCR-coupled antigen receptor with superior targeting sensitivity and reversible drug-regulated anti-tumor activity. Mol Ther 2025; 33:1608-1620. [PMID: 39980194 PMCID: PMC11997482 DOI: 10.1016/j.ymthe.2025.02.026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 12/02/2024] [Accepted: 02/17/2025] [Indexed: 02/22/2025] Open
Abstract
Chimeric antigen receptor (CAR) T cells are effective cancer therapies, particularly in indications with high, stable, and tumor-specific antigen expression. Other settings may require improved targeting sensitivity, controllable targeting selectivity, and/or additional potency enhancements to achieve robust efficacy. Here, we describe a novel receptor architecture called RESET (rapamycin-enabled, switchable endogenous T cell receptor) that combines (1) cell surface antigen targeting, (2) small-molecule regulation, and (3) the signaling proficiency and inherent sensitivity of native T cell receptors. RESET-T cells outperformed both constitutive and drug-regulated CAR-T cells and show hallmarks of TCR activation that suggest improved fidelity to native T cell responses. Pharmacological control then increases safety through toggling T cell activation between active and resting states and may mitigate T cell exhaustion caused by continuous antigen exposure. This convergence of drug-regulated targeting and natural immune receptor signal transduction may better replicate the kinetics and physiology of a classical T cell response and potentiate more successful and safer immunotherapies.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Immunotherapy, Adoptive/methods
- Animals
- Lymphocyte Activation/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/drug effects
- Mice
- Signal Transduction
- Neoplasms/therapy
- Neoplasms/immunology
- Neoplasms/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
| | | | | | | | | | | | - Nan Jin
- Regeneron Pharmaceuticals, Tarrytown, NY 10591, USA
| | | | | |
Collapse
|
4
|
Yue T, Sun Y, Dai Y, Jin F. Mechanisms for resistance to BCMA-targeted immunotherapies in multiple myeloma. Blood Rev 2025; 70:101256. [PMID: 39818472 DOI: 10.1016/j.blre.2025.101256] [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: 10/16/2024] [Revised: 01/03/2025] [Accepted: 01/10/2025] [Indexed: 01/18/2025]
Abstract
Multiple myeloma (MM) remains incurable and patients eventually face the relapse/refractory dilemma. B cell maturation antigen (BCMA)-targeted immunotherapeutic approaches have shown great effectiveness in patients with relapsed/refractory MM, mainly including chimeric antigen receptor T cells (CAR-T), bispecific T cell engagers (TCEs), and antibody-drug conjugates (ADCs). However, their impact on long-term survival remains to be determined. Nonetheless, resistance to these novel therapies is still inevitable, raising a challenge that we have never met in both laboratory research and clinical practice. In this scenario, the investigation aiming to enhance and prolong the anti-MM activity of BCMA-targeted therapies has been expanding rapidly. Despite considerable uncertainty in our understanding of the mechanisms for their resistance, they have mainly been attributed to antigen-dependency, T cell-driven factors, and (immune) tumor microenvironment. In this review, we summarize the current understanding of the mechanisms for resistance to BCMA-targeted immunotherapies and discuss potential strategies for overcoming it.
Collapse
Affiliation(s)
- Tingting Yue
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin, China; Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yue Sun
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, First Hospital of Jilin University, Changchun, Jilin, China.
| | - Fengyan Jin
- Department of Hematology, First Hospital of Jilin University, Changchun, Jilin, China.
| |
Collapse
|
5
|
Jamroziak K, Zielonka K, Khwaja J, Wechalekar AD. Update on B-cell maturation antigen-directed therapies in AL amyloidosis. Br J Haematol 2025; 206:817-831. [PMID: 39748220 DOI: 10.1111/bjh.19960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025]
Abstract
Systemic light chain (AL) amyloidosis is a rare clonal plasma cell disorder characterized by the production of amyloidogenic immunoglobulin light chains, which causes the formation and deposition of amyloid fibrils, leading to multi-organ dysfunction. Current treatment is directed at the underlying plasma cell clone to achieve a profound reduction in the monoclonal free light chain production. The standard-of-care first-line therapy is a combination of daratumumab, cyclophosphamide, bortezomib and dexamethasone (D-VCd regimen), resulting in high rates of haematological and organ responses. However, AL amyloidosis remains incurable, and all patients inevitably relapse. Hence, novel treatment options are needed for patients with an inadequate response or relapsed/refractory disease. B-cell maturation antigen (BCMA) is a tumour necrosis factor (TNF receptor superfamily receptor overexpressed on plasma cells in multiple myeloma (MM) and AL amyloidosis. Recently, several novel anti-BCMA immunotherapies have been approved for the treatment of relapsed/refractory MM, including antibody-drug conjugate belantamab mafodotin, bispecific antibodies teclistamab and elranatamab and chimeric antigen receptor T-cell therapies idecabtagene vicleucel and ciltacabtagene autoleucel. Despite lower expression than in MM, BCMA is also a promising target in AL amyloidosis. This review aims to provide up-to-date information on the efficacy and toxicity of anti-BCMA therapy in AL amyloidosis.
Collapse
Affiliation(s)
- Krzysztof Jamroziak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Klaudia Zielonka
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, Warsaw, Poland
| | - Jahanzaib Khwaja
- Department of Haematology, University College London Hospital, London, UK
| | | |
Collapse
|
6
|
Yin H, Wei X. The design of retroviral vectors used in the CAR-T products, risk management, and future perspective. MedComm (Beijing) 2025; 6:e70067. [PMID: 39866836 PMCID: PMC11758153 DOI: 10.1002/mco2.70067] [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: 10/22/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 01/28/2025] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a revolutionary approach in cancer treatment. More than 10 CAR-T products have already approved on market worldly wide, and they use either gamma retroviral vectors or lentiviral vectors to deliver the CAR gene. Both vectors have the ability to effectively and persistently integrate the CAR gene into T cells. Despite the advancements in CAR-T therapy, the potential risks associated with the vectors, particularly the risks of the secondary malignancies, still remain as a concern. This article compares the characteristics of gamma retroviral and lentiviral vectors, discusses the development of vector packaging systems, and examines the design of self-inactivating (SIN) vectors. It also addresses the risks of secondary malignancies that might possibly be associated with the retroviral vectors, and the strategies to decrease the risks and increase the safer clinical use of the vectors. This article also discusses the current regulatory landscape and management approaches aiming to mitigate these risks through stringent safety measures and ongoing monitoring. Future perspectives focus on improving the safety profiles of the vectors and broadening their scope of use. The article provides a thorough overview of the most recent research discoveries and regulatory updates in the field of CAR-T therapy, highlighting the significance of a balanced strategy that strikes a balance between innovation and patient safety in the development and implementation of CAR-T therapy.
Collapse
Affiliation(s)
- Huifang Yin
- Office of Pharmaceutical ScienceYangtze River Delta Center for Drug Evaluation and InspectionNational Medical Products AdministrationShanghaiChina
| | - Xuejing Wei
- Office of Pharmaceutical ScienceYangtze River Delta Center for Drug Evaluation and InspectionNational Medical Products AdministrationShanghaiChina
| |
Collapse
|
7
|
Hu Y, Xie Y, Wang X, Yang L, Geng H, Yi Z, Zhang Y, Ma L, Chen F. Targeting BCMA in multiple myeloma: designs, challenges, and future directions. Cancer Immunol Immunother 2025; 74:77. [PMID: 39891674 PMCID: PMC11787132 DOI: 10.1007/s00262-024-03913-0] [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: 12/01/2024] [Indexed: 02/03/2025]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has emerged as a groundbreaking immunotherapy, demonstrating significant efficacy in treating B cell malignancies. In the context of multiple myeloma (MM), B cell maturation antigen (BCMA) has been identified as a critical target, driving the development of CAR T cell therapies designed to address this plasma cell cancer. Various CAR designs, utilizing different BCMA recognition domains, have yielded promising clinical results, leading to the approval of two BCMA-targeting CAR T cell therapies by the US Food and Drug Administration (FDA) for the treatment of MM. This review uniquely examines the BCMA CAR T cell landscape, emphasizing the design of recognition domains, clinical efficacy, and patient outcomes. It critically addresses emerging challenges such as antigen escape and toxicity profiles, which have surfaced alongside therapeutic advances. Moreover, the review spotlights cutting-edge developments, including dual-targeting CAR T strategies, advancements in CAR T cell manufacturing, and innovative allogeneic CAR T approaches utilizing healthy donor cells. By detailing both the breakthroughs and ongoing challenges in BCMA CAR T cell therapy, this review offers a comprehensive perspective on the current state and future possibilities of CAR T cell therapy for MM and its expanding role in treating hematologic malignancies and beyond.
Collapse
Affiliation(s)
- Yi Hu
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Yuetao Xie
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Xiaodong Wang
- Department of Hematology and Oncology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Lufeng Yang
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - He Geng
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Zugang Yi
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Yao Zhang
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Lin Ma
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China
| | - Fang Chen
- Department of Anesthesiology, Shenzhen Children's Hospital, Shenzhen, 518038, Guangdong, China.
| |
Collapse
|
8
|
Su M, Dey A, Maddah E, Mugundu GM, Singh AP. Quantitative pharmacology of dual-targeted bicistronic CAR-T-cell therapy using multiscale mechanistic modeling. CPT Pharmacometrics Syst Pharmacol 2025; 14:229-245. [PMID: 39508140 PMCID: PMC11812944 DOI: 10.1002/psp4.13259] [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: 07/23/2024] [Revised: 09/28/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
Despite the initial success of single-targeted chimeric-antigen receptor (CAR) T-cell therapy in hematological malignancies, its long-term effectiveness is often hindered by antigen heterogeneity and escape. As a result, there is a growing interest in cell therapies targeting multiple antigens (≥2). However, the dose-exposure-response relationship and specific factors influencing the pharmacology of dual-targeted CAR-T-cell therapy remain unclear. In this study, we have developed a multiscale cellular kinetic-pharmacodynamic (CK-PD) model using case studies from CD19/CD22 and GPRC5D/BCMA autologous CAR-Ts. Initially, an in vitro tumor-killing model characterized the impact of individual binder affinities and their contribution to overall potency across varying (1) effector: target (ET) ratios and (2) tumor-associated antigen (TAA) expressing cell lines. Subsequently, an integrated CK-PD model was developed in pediatric acute lymphoblastic leukemia (ALL) patients, which accounted for CAR-T-cell product composition and relative antigen abundance in patients' tumor burden to characterize patient-level multiphasic cellular kinetics using multiple bioanalytical assays (e.g., flow and qPCR-based readouts). Global sensitivity analysis highlighted relative antigen expression, maximum killing rate constant, and CAR-T expansion rate constant as major determinants for observed exposure of dual-targeted CAR-T-cell therapy. This modeling framework could facilitate dose-optimization and construct refinement for dual-targeted bicistronic CAR-T-cell therapies, serving as a valuable tool for both forward and reverse translation in drug development.
Collapse
Affiliation(s)
- Mei‐Chi Su
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
- Experimental and Clinical Pharmacology, College of PharmacyUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Agnish Dey
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Erfan Maddah
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Ganesh M. Mugundu
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Aman P. Singh
- Cell Therapy Clinical Pharmacology and Modeling, Precision and Translational Medicine, Oncology Cell Therapy and Therapeutic Area UnitTakeda PharmaceuticalsCambridgeMassachusettsUSA
| |
Collapse
|
9
|
Bachiller M, Barceló-Genestar N, Rodriguez-Garcia A, Alserawan L, Dobaño-López C, Giménez-Alejandre M, Castellsagué J, Colell S, Otero-Mateo M, Antoñana-Vildosola A, Español-Rego M, Ferruz N, Pascal M, Martín-Antonio B, Anguela XM, Fillat C, Olesti E, Calvo G, Juan M, Delgado J, Pérez-Galán P, Urbano-Ispizua Á, Guedan S. ARI0003: Co-transduced CD19/BCMA dual-targeting CAR-T cells for the treatment of non-Hodgkin lymphoma. Mol Ther 2025; 33:317-335. [PMID: 39563035 PMCID: PMC11764334 DOI: 10.1016/j.ymthe.2024.11.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/10/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
CD19 CAR-T therapy has achieved remarkable responses in relapsed/refractory non-Hodgkin lymphoma (NHL). However, challenges persist, with refractory responses or relapses after CAR-T administration linked to CD19 loss or downregulation. Given the co-expression of CD19 and BCMA in NHL, we hypothesized that dual targeting could enhance long-term efficacy. We optimized different dual-targeting approaches, including co-transduction of two lentiviral vectors, bicistronic, tandem, and loop and pool strategies, based on our academic anti-CD19 (ARI0001) and anti-BCMA (ARI0002h) CAR-T cells. Comparison with anti-CD19/CD20 or anti-CD19/CD22 dual targeting was also performed. We demonstrate that anti-CD19/BCMA CAR-T cells can be effectively generated through the co-transduction of two lentiviral vectors after optimization to minimize competition for cellular resources. Co-transduced T cells, called ARI0003, effectively targeted NHL tumor cells with high avidity, outperforming anti-CD19 CAR-T cells and other dual-targeting approaches both in vitro and in vivo, particularly in low CD19 antigen density models. ARI0003 maintained effectiveness post-CD19 CAR-T treatment in xenograft models and in spheroids from relapsed CART-treated patients. ARI0003 CAR-T cells were effectively manufactured under Good Manufacturing Practice conditions, with a reduced risk of genotoxicity compared to other dual-targeting approaches. A first-in-human phase 1 clinical trial (CARTD-BG-01; this study was registered at ClinicalTrials.gov [NCT06097455]) has been initiated to evaluate the safety and efficacy of ARI0003 in NHL.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Antigens, CD19/immunology
- Antigens, CD19/metabolism
- Cell Line, Tumor
- Clinical Trials, Phase I as Topic
- Disease Models, Animal
- Genetic Vectors/genetics
- Genetic Vectors/administration & dosage
- Immunotherapy, Adoptive/methods
- Lentivirus/genetics
- Lymphoma, Non-Hodgkin/therapy
- Lymphoma, Non-Hodgkin/genetics
- Randomized Controlled Trials as Topic
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Transduction, Genetic
- Xenograft Model Antitumor Assays
- Male
- Female
Collapse
Affiliation(s)
- Mireia Bachiller
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Nina Barceló-Genestar
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Alba Rodriguez-Garcia
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | | | - Cèlia Dobaño-López
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Marta Giménez-Alejandre
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Joan Castellsagué
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Salut Colell
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Marc Otero-Mateo
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Asier Antoñana-Vildosola
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain
| | - Marta Español-Rego
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Department of Immunology, Hospital Clínic, 08036 Barcelona, Spain
| | - Noelia Ferruz
- Centre for Genomic Regulation (CRG), Barcelona Institute for Science and Technology, 08003 Barcelona, Spain
| | - Mariona Pascal
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Department of Immunology, Hospital Clínic, 08036 Barcelona, Spain; University of Barcelona, 08034 Barcelona, Spain
| | - Beatriz Martín-Antonio
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Instituto de Salud Carlos III, 28029 Madrid, Spain
| | | | - Cristina Fillat
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación en Red-Enfermedades Raras (CIBERER), 08036 Barcelona, Spain
| | - Eulàlia Olesti
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; University of Barcelona, 08034 Barcelona, Spain; Department of Clinical Pharmacology, Hospital Clínic, 08036 Barcelona, Spain
| | - Gonzalo Calvo
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; University of Barcelona, 08034 Barcelona, Spain; Department of Clinical Pharmacology, Hospital Clínic, 08036 Barcelona, Spain
| | - Manel Juan
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Department of Immunology, Hospital Clínic, 08036 Barcelona, Spain
| | - Julio Delgado
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain; University of Barcelona, 08034 Barcelona, Spain; Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain
| | - Patricia Pérez-Galán
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; Centro de Investigación Biomédica en Red-Oncología (CIBERONC), 28029 Madrid, Spain
| | - Álvaro Urbano-Ispizua
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain; University of Barcelona, 08034 Barcelona, Spain; Department of Hematology, Hospital Clínic, 08036 Barcelona, Spain
| | - Sonia Guedan
- Fundació de Recerca Clínic Barcelona-Institut d'Investigacions Biomèdiques August Pi Sunyer (FRCB-IDIBAPS), 08036 Barcelona, Spain.
| |
Collapse
|
10
|
Rampotas A, Gannon I, Roddie C. Choosing the right double-barreled gun: ARI0003 takes aim at lymphoma by targeting both CD19 and BCMA. Mol Ther 2025; 33:23-25. [PMID: 39708800 PMCID: PMC11764553 DOI: 10.1016/j.ymthe.2024.12.034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 12/13/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024] Open
Affiliation(s)
- Alexandros Rampotas
- University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK; University College London Hospital, 235 Euston Road, London NW1 2BU, UK
| | - Isaac Gannon
- University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK
| | - Claire Roddie
- University College London Cancer Institute, 72 Huntley Street, London WC1E 6DD, UK; University College London Hospital, 235 Euston Road, London NW1 2BU, UK.
| |
Collapse
|
11
|
Zhang SC, Ballas LK. Radiation for Multiple Myeloma in the Era of Novel Agents: Indications, Safety, and Dose Selection. Semin Radiat Oncol 2025; 35:87-98. [PMID: 39672645 DOI: 10.1016/j.semradonc.2024.10.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2024]
Abstract
Survival outcomes for multiple myeloma (MM) have drastically improved over the past two decades with the advent of highly effective biologic agents and integration of autologous stem cell transplant (ASCT) for select patients. Despite these advances, MM remains an incurable disease and duration of remission decreases with each relapse. Palliative radiotherapy (RT) for MM, including treatment of pain, relief of compression, and prevention of fracture, is highly effective and generally well tolerated. Though RT can be delivered concurrently with biologic agents, caution should be exercised for potential added hematologic toxicity that may disrupt systemic therapy, especially in heavily pretreated patients, who have limited bone marrow reserve. In this review, we discuss the safety of RT with biologic agents (proteasome inhibitors, immunomodulators, monoclonal antibodies), review indications for palliative RT in MM, and present a framework for how to personalize RT based on goals of treatment, classification of uncomplicated versus complicated lesions, and patient and lesion characteristics. Additionally, we discuss the emerging role of bridging RT prior to chimeric antigen receptor (CAR) T-cell therapy.
Collapse
Affiliation(s)
- Samuel C Zhang
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA
| | - Leslie K Ballas
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles, CA.
| |
Collapse
|
12
|
Mancuso K, Barbato S, Talarico M, Tacchetti P, Zamagni E, Cavo M. Idecabtagene vicleucel (ide-cel) for the treatment of triple-class exposed relapsed and refractory multiple myeloma. Expert Opin Biol Ther 2025; 25:27-46. [PMID: 39651553 DOI: 10.1080/14712598.2024.2433518] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/20/2024] [Indexed: 12/11/2024]
Abstract
INTRODUCTION Modern anti-myeloma therapies have broken new ground in the treatment of the disease, and the incorporation of ide-cel in the treatment landscape represents one of the major scientific and clinical advances. AREAS COVERED Ide-cel was the first cell-based gene therapy approved for the treatment of triple-class exposed relapsed/refractory myeloma patients, showing impressive results, and demonstrating superiority over standard regimens in terms of efficacy, potential treatment-free intervals, and improved quality of life in heavily pretreated patients and in high-risk disease. This review summarizes the state-of-the-art of the most recent updates deriving from the use of ide-cel within ongoing, or upcoming, clinical trials, and from real-life experiences. EXPERT OPINION As the use of chimeric antigen receptor (CAR)-T therapy is likely to progressively increase over time and current indications expand to earlier treatment lines, efforts should be directed toward ameliorating overall management to facilitate proactive planning for treatment sequencing and provide adequate time for logistical planning. Importantly, the potential limited availability of CAR-T therapy highlights the importance of careful patient selection and coordination among centers. Meanwhile, attempts are underway to improve tolerance and reduce toxicity while enhancing anti-myeloma activity.
Collapse
Affiliation(s)
- Katia Mancuso
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Simona Barbato
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Marco Talarico
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Paola Tacchetti
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Elena Zamagni
- IRCCS Azienda Ospedaliero-Universitaria di Bologna, Istituto di Ematologia "Seràgnoli", Bologna, Italy
| | - Michele Cavo
- Dipartimento di Scienze Mediche e Chirurgiche, Università di Bologna, Bologna, Italy
| |
Collapse
|
13
|
Li M, Zheng R, Liu Z, Zhang P, Zhu T, Xin X, Zhao H, Chen W, Zheng B, Zhao A, Gao J. Optimized BCMA/CS1 bispecific TRuC-T cells secreting IL-7 and CCL21 robustly control multiple myeloma. Front Immunol 2024; 15:1502936. [PMID: 39776916 PMCID: PMC11703830 DOI: 10.3389/fimmu.2024.1502936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 12/09/2024] [Indexed: 01/11/2025] Open
Abstract
Introduction Challenges remain in reducing antigen escape and tumor recurrence while CAR-T cell therapy has substantially improved outcomes in the treatment of multiple myeloma. T cell receptor fusion construct (TRuC)-T cells, which utilize intact T cell receptor (TCR)-CD3 complex to eliminate tumor cells in a non-major histocompatibility complex (MHC)-restricted manner, represent a promising strategy. Moreover, interleukin-7 (IL-7) is known to enhance the proliferation and survival of T cells. C-C motif chemokine ligand 21 (CCL21) is a ligand for chemokine C-C motif receptor 7 (CCR7) and exhibits strong chemotaxis against naïve T cells and antigen-presenting cells such as dendritic cells. Methods The bispecific TRuC-T cells simultaneously targeting B cell maturation antigen (BCMA) and CD2 subset 1 (CS1) were constructed by pairing two of five subunits (i.e., TCRαC, TCRβC, CD3γ, CD3δ, and CD3ϵ) in the TCR/CD3 complex and were named C-AC-B-3E, C-BC-B-3E, C-3G-B-3E, C-3D-B-3E, C-3E-B-3E, B-3E-C-3E, B-3G-C-3E, and B-3D-C-3E. Additionally, the BCMA/CS1 bispecific TRuC-T cells secreting IL-7 and CCL21, named BC-7×21 TRuC-T cells, were generated. All of the bispecific TRuC-T cells were characterized and tested in vitro and in vivo. Results Following the optimization of various pairs of two subunits of TCR/CD3 complex, B-3G-C-3E TRuC-T cells, characterized by incorporating CD3γ and CD3ε, exhibited the strongest myeloma-specific cytotoxicity. Furthermore, the bispecific BC-7×21 TRuC-T cells had stronger proliferation, chemotaxis, and cytotoxicity in vitro. Accordingly, the bispecific BC-7×21 TRuC-T cells showed better persistence in vivo so as to effectively suppress tumor growth in the NCG mouse xenograft model of MM.1S multiple myeloma. Discussion This study demonstrated that BC-7×21 TRuC-T cells, engineered through the optimization of the two subunits of TCR/CD3 complex and a co-expression cytokine strategy, may offer a novel and effective therapy for relapsed/refractory multiple myeloma.
Collapse
MESH Headings
- Multiple Myeloma/immunology
- Multiple Myeloma/therapy
- Multiple Myeloma/metabolism
- B-Cell Maturation Antigen/immunology
- B-Cell Maturation Antigen/metabolism
- Humans
- Animals
- Mice
- Interleukin-7/metabolism
- Interleukin-7/immunology
- Chemokine CCL21/metabolism
- Immunotherapy, Adoptive/methods
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Mice, SCID
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Min Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Rong Zheng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- Yicheng County People’s Hospital, Linfen, Shanxi, China
| | - Zairu Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- Ningbo Hangzhou Bay Hospital, Ningbo, Zhejiang, China
| | - Peiyuan Zhang
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Tingwei Zhu
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, Taizhou, Zhejiang, China
| | - Xueyi Xin
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- Taizhou Central Hospital (Taizhou University Hospital), Taizhou, Zhejiang, China
| | - Hongli Zhao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Wenyi Chen
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Binjiao Zheng
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
| | - Ai Zhao
- Affiliated Hangzhou First People’s Hospital, Westlake University School of Medicine, Hangzhou, Zhejiang, China
| | - Jimin Gao
- Key Laboratory of Laboratory Medicine, Ministry of Education, School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, China
- Zhejiang Qixin Biotech, Wenzhou, Zhejiang, China
| |
Collapse
|
14
|
Honaker Y, Gruber D, Jacobs C, Yu-Hong Cheng R, Patel S, Galvan CZ, Khan IF, Zhou K, Sommer K, Astrakhan A, Cook PJ, James RG, Rawlings DJ. Targeting human plasma cells using regulated BCMA CAR T cells eliminates circulating antibodies in humanized mice. Mol Ther 2024:S1525-0016(24)00817-7. [PMID: 39673129 DOI: 10.1016/j.ymthe.2024.12.018] [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: 06/14/2024] [Revised: 10/22/2024] [Accepted: 12/10/2024] [Indexed: 12/16/2024] Open
Abstract
Pathogenic long-lived plasma cells (LLPCs) secrete autoreactive antibodies, exacerbating autoimmune diseases and complicating solid organ transplantation. Targeted elimination of the autoreactive B cell pool represents a promising therapeutic strategy, yet current treatment modalities fall short in depleting mature PCs. Here, we demonstrate that chimeric antigen receptor (CAR) T cells, targeting B cell maturation antigen (BCMA) utilizing a split-receptor design, offer a controlled and effective therapeutic strategy against LLPCs. Dimerizing agent-regulated immune-receptor complex (DARIC) T cells demonstrated robust rapamycin-dependent targeting of tumor and PCs. Notably, in humanized mouse models, DARIC T cells regulated peripheral human immunoglobulin levels through specific elimination of human LLPCs from the bone marrow. Furthermore, DARIC constructs were efficiently integrated into the T cell receptor α constant (TRAC) locus while maintaining potent antigen-specific cytotoxicity. These findings underscore the potential of split-receptor CAR T cells in autoimmune and transplant medicine, highlighting their versatility in applications beyond oncology.
Collapse
Affiliation(s)
- Yuchi Honaker
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - David Gruber
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Chester Jacobs
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Rene Yu-Hong Cheng
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Shivani Patel
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Christopher Zavala Galvan
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Iram F Khan
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Kevin Zhou
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Karen Sommer
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | | | - Peter J Cook
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA
| | - Richard G James
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA
| | - David J Rawlings
- Program for Cell and Gene Therapy and Center for Immunity and Immunotherapies, Seattle Children's Research Institute, Seattle, WA 98101, USA; Department of Pediatrics, University of Washington, Seattle, WA 98105, USA; Department of Immunology, University of Washington, Seattle, WA 98109, USA.
| |
Collapse
|
15
|
Zhou X, Kortuem KM, Rasche L, Einsele H. Bispecific antibody and chimeric antigen receptor (CAR) modified T-cell in the treatment of multiple myeloma: Where do we stand today? Presse Med 2024; 54:104265. [PMID: 39662761 DOI: 10.1016/j.lpm.2024.104265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024] Open
Abstract
Although the prognosis of patients with multiple myeloma (MM) has been significantly improved by the introduction of proteasome inhibitors, immunomodulatory drugs and monoclonal antibodies, MM is still considered an incurable disease in the vast majority of the patients. In recent years, T-cell based immunotherapy represents a novel treatment strategy for relapsed/refractory (RR) MM. So far, chimeric antigen receptor (CAR) modified T-cells and bispecific T-cell engaging antibodies (bsAb) have shown promising anti-MM efficacy and manageable safety profile within clinical trials, and B-cell maturation antigen (BCMA) is the most commonly used immune target for T-cell based immunotherapies in MM. To date, several CAR T-cell and bsAb products have already been approved for the treatment of RRMM, leading to a paradigm shift in the MM therapy and providing a potential curative option. In this review, we provide a summary of mechanisms of action, immune targets, selected clinical data, resistance mechanisms and therapy sequencing of CAR T-cell and bsAb in MM.
Collapse
Affiliation(s)
- Xiang Zhou
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - K Martin Kortuem
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Leo Rasche
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany
| | - Hermann Einsele
- Department of Internal Medicine II, University Hospital of Würzburg, Würzburg, Germany.
| |
Collapse
|
16
|
Reddy ST, Hosoya H, Mikkilineni L. CAR T-cell therapy to treat multiple myeloma: current state and future directions. Cancer Metastasis Rev 2024; 44:14. [PMID: 39625587 DOI: 10.1007/s10555-024-10219-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 11/07/2024] [Indexed: 12/17/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy represents a transformative advancement in treating relapsed or refractory multiple myeloma (MM) in both early- and late-line settings. MM, a plasma cell malignancy, traditionally requires ongoing complex drug regimens, posing significant burdens on patients. In contrast, CAR T-cell therapy offers a one-time treatment option without the need for continuous maintenance therapy. CAR T-cell therapy leverages engineered T-cells to target specific antigens on tumor cells, leading to their elimination. Current approved therapies target B-cell maturation antigen (BCMA); new targets are under investigation, such as G-protein-coupled receptor class C group 5 member D (GPRC5D). Despite its efficacy, CAR T-cell therapy is associated with serious toxicities such as cytokine release syndrome (CRS) and immune-effector cell-associated neurotoxicity syndrome (ICANS), necessitating careful management. The review will provide an overview of the design and manufacturing of CAR T-cells and current FDA indications, as well as challenges and future directions of CAR-T therapy for MM treatment.
Collapse
Affiliation(s)
- Siddhartha Thammineni Reddy
- Division of Bone and Marrow Transplantation & Cellular Therapies, Stanford University, 870 Welch Road, Palo Alto, CA, 94304, USA
- Hackensack University Medical Center, NJ, USA
| | - Hitomi Hosoya
- Division of Bone and Marrow Transplantation & Cellular Therapies, Stanford University, 870 Welch Road, Palo Alto, CA, 94304, USA
| | - Lekha Mikkilineni
- Division of Bone and Marrow Transplantation & Cellular Therapies, Stanford University, 870 Welch Road, Palo Alto, CA, 94304, USA.
| |
Collapse
|
17
|
Arunachalam AK, Grégoire C, Coutinho de Oliveira B, Melenhorst JJ. Advancing CAR T-cell therapies: Preclinical insights and clinical translation for hematological malignancies. Blood Rev 2024; 68:101241. [PMID: 39289094 DOI: 10.1016/j.blre.2024.101241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/19/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has achieved significant success in achieving durable and potentially curative responses in patients with hematological malignancies. CARs are tailored fusion proteins that direct T cells to a specific antigen on tumor cells thereby eliciting a targeted immune response. The approval of several CD19-targeted CAR T-cell therapies has resulted in a notable surge in clinical trials involving CAR T cell therapies for hematological malignancies. Despite advancements in understanding response mechanisms, resistance patterns, and adverse events associated with CAR T-cell therapy, the translation of these insights into robust clinical efficacy has shown modest outcomes in both clinical trials and real-world scenarios. Therefore, the assessment of CAR T-cell functionality through rigorous preclinical studies plays a pivotal role in refining therapeutic strategies for clinical applications. This review provides an overview of the various in vitro and animal models used to assess the functionality of CAR T-cells. We discuss the findings from preclinical research involving approved CAR T-cell products, along with the implications derived from recent preclinical studies aiming to optimize the functionality of CAR T-cells. The review underscores the importance of robust preclinical evaluations and the need for models that accurately replicate human disease to bridge the gap between preclinical success and clinical efficacy.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Hematologic Neoplasms/therapy
- Hematologic Neoplasms/immunology
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Translational Research, Biomedical
- Disease Models, Animal
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
- Arun K Arunachalam
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Céline Grégoire
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Beatriz Coutinho de Oliveira
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America
| | - Jan Joseph Melenhorst
- Cell Therapy & Immuno-Engineering Program, Center for Immunotherapy and Precision Immuno-Oncology, Lerner College of Medicine, Cleveland Clinic, Cleveland, OH 44195, United States of America.
| |
Collapse
|
18
|
Kim SE, Yun S, Doh J, Kim HN. Imaging-Based Efficacy Evaluation of Cancer Immunotherapy in Engineered Tumor Platforms and Tumor Organoids. Adv Healthc Mater 2024; 13:e2400475. [PMID: 38815251 DOI: 10.1002/adhm.202400475] [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: 02/06/2024] [Revised: 05/16/2024] [Indexed: 06/01/2024]
Abstract
Cancer immunotherapy is used to treat tumors by modulating the immune system. Although the anticancer efficacy of cancer immunotherapy has been evaluated prior to clinical trials, conventional in vivo animal and endpoint models inadequately replicate the intricate process of tumor elimination and reflect human-specific immune systems. Therefore, more sophisticated models that mimic the complex tumor-immune microenvironment must be employed to assess the effectiveness of immunotherapy. Additionally, using real-time imaging technology, a step-by-step evaluation can be applied, allowing for a more precise assessment of treatment efficacy. Here, an overview of the various imaging-based evaluation platforms recently developed for cancer immunotherapeutic applications is presented. Specifically, a fundamental technique is discussed for stably observing immune cell-based tumor cell killing using direct imaging, a microwell that reproduces a confined space for spatial observation, a droplet assay that facilitates cell-cell interactions, and a 3D microphysiological system that reconstructs the vascular environment. Furthermore, it is suggested that future evaluation platforms pursue more human-like immune systems.
Collapse
Affiliation(s)
- Seong-Eun Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
| | - Suji Yun
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, South Korea
| | - Junsang Doh
- Interdisciplinary Program for Bioengineering, Seoul National University, Seoul, 08826, South Korea
- Department of Materials Science and Engineering, Research Institute of Advanced Materials, Institute of Engineering Research, Bio-MAX institute, Soft Foundry Institute, Seoul National University, Seoul, 08826, South Korea
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, South Korea
- Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology, Seoul, 02792, Republic of Korea
- School of Mechanical Engineering, Yonsei University, Seoul, 03722, Republic of Korea
- Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul, 03722, Republic of Korea
| |
Collapse
|
19
|
Springer AD, Wang R, Wang J, Du Q, Pi W, Nguyen AQ, Li X, Khasanov A, Zhu T, Yan Z, Hong Y, Zhou H, Zhang Y, Kerwin L, Li L, Ji H, Zhang H. Preclinical Evaluation of STI-8811, a Novel Antibody-Drug Conjugate Targeting BCMA for the Treatment of Multiple Myeloma. CANCER RESEARCH COMMUNICATIONS 2024; 4:2660-2672. [PMID: 39292169 PMCID: PMC11467701 DOI: 10.1158/2767-9764.crc-24-0413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 08/29/2024] [Accepted: 09/13/2024] [Indexed: 09/19/2024]
Abstract
Treatment for patients with multiple myeloma has experienced rapid development and improvement in recent years; however, patients continue to experience relapse, and multiple myeloma remains largely incurable. B-cell maturation antigen (BCMA) has been widely recognized as a promising target for treatment of multiple myeloma due to its exclusive expression in B-cell linage cells and its critical role in the growth and survival of malignant plasma cells. Here, we introduce STI-8811, a BCMA-targeting antibody-drug conjugate (ADC) linked to an auristatin-derived duostatin payload via an enzymatically cleavable peptide linker, using our proprietary C-lock technology. STI-8811 exhibits target-specific binding activity and rapid internalization, leading to G2/M cell-cycle arrest, caspase 3/7 activation, and apoptosis in BCMA-expressing tumor cells in vitro. Soluble BCMA (sBCMA) is shed by multiple myeloma cells into the blood and increases with disease progression, competing for ADC binding and reducing its efficacy. We report enhanced cytotoxic activity in the presence of high levels of sBCMA compared with a belantamab mafodotin biosimilar (J6M0-mcMMAF). STI-8811 demonstrated greater in vivo activity than J6M0-mcMMAF in solid and disseminated multiple myeloma models, including tumor models with low BCMA expression and/or in large solid tumors representing soft-tissue plasmacytomas. In cynomolgus monkeys, STI-8811 was well tolerated, with toxicities consistent with other BCMA-targeting ADCs with auristatin payloads in clinical studies. STI-8811 has the potential to outperform current clinical candidates with lower toxicity and higher activity under conditions found in patients with advanced disease. Significance: STI-8811 is a BCMA-targeting ADC carrying a potent auristatin derivative. We report unique binding properties which maintain potent cytotoxic activity under sBCMA-high conditions that hinder the clinical efficacy of current BCMA-targeting ADC candidates. Beyond disseminated models of multiple myeloma, we observed efficacy in solid tumor models of plasmacytomas with low and heterogenous BCMA expressions at a magnitude and duration of response exceeding that of clinical comparators.
Collapse
Affiliation(s)
| | | | | | - Qinyi Du
- Levena BioPharma, San Diego, California.
| | - Willie Pi
- Levena BioPharma, San Diego, California.
| | | | - Xiaoqing Li
- Levena BioPharma, San Diego, California.
- Sorrento Therapeutics Inc., San Diego, California.
| | | | - Tong Zhu
- Levena BioPharma, San Diego, California.
| | - Zheng Yan
- Levena BioPharma, San Diego, California.
| | | | - Heyue Zhou
- Sorrento Therapeutics Inc., San Diego, California.
| | | | - Lisa Kerwin
- Sorrento Therapeutics Inc., San Diego, California.
| | - Lingna Li
- Levena BioPharma, San Diego, California.
| | - Henry Ji
- Levena BioPharma, San Diego, California.
- Sorrento Therapeutics Inc., San Diego, California.
| | - Hong Zhang
- Levena BioPharma, San Diego, California.
- Sorrento Therapeutics Inc., San Diego, California.
| |
Collapse
|
20
|
Gagelmann N, Merz M. Fast and furious: Changing gears on the road to cure with chimeric antigen receptor T cells in multiple myeloma. Semin Hematol 2024; 61:306-313. [PMID: 39095225 DOI: 10.1053/j.seminhematol.2024.07.002] [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: 05/22/2024] [Revised: 06/28/2024] [Accepted: 07/03/2024] [Indexed: 08/04/2024]
Abstract
Based on the pivotal KarMMa-1 and CARTITUDE-1 studies, Idecabtagene vicleucel (Ide-cel) and Ciltacabtagene autoleucel (Cilta-cel) have been approved to treat multiple myeloma patients, who have been exposed to at least 1 proteasome inhibitor, immunomodulatory drug and anti-CD38 antibody after 4 or 3 lines of therapy, respectively. The unprecedented rates of deep and long-lasting remissions have been meanwhile confirmed in multiple real-world analyses and more recently, the KarMMa-3 and CARTITUDE-4 studies lead to the approval in earlier lines of therapy. It is currently believed that ultimately all patients with relapsed/refractory multiple myeloma experience relapse after anti-BCMA CAR T-cell therapies. There is a plethora of CAR T-cell therapies targeting novel antigens, with the aim to overcome current CAR T-cell resistance. In this review, we will summarize current evidence of novel antigens and their clinical potential. Together with current CAR T-cell therapy and T-cell engagers, these approaches might lead us to the next frontier in multiple myeloma: total immunotherapy and the road to chemotherapy-free cure.
Collapse
Affiliation(s)
- Nico Gagelmann
- Department of Stem Cell Transplantation, University Medical Center Hamburg-Eppendorf, Hamburg, Hamburg, Germany.
| | - Maximilian Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectiology, University Hospital of Leipzig, Leipzig, Saxony, Germany
| |
Collapse
|
21
|
Liu W, Lin S, Zhu X, Yin L, Liu Q, Lei S, Feng B. Safety assessment of anti-B cell maturation antigen chimeric antigen receptor T cell therapy: a real-world study based on the FDA adverse event reporting system database. Front Immunol 2024; 15:1433075. [PMID: 39290710 PMCID: PMC11405296 DOI: 10.3389/fimmu.2024.1433075] [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: 09/19/2024] Open
Abstract
Background On April 18, 2024, the U.S. Food and Drug Administration officially required updating of the "boxed warning" for T cell malignancies for all chimeric antigen receptor T cell (CAR-T) therapies. Given the clinical significance of these therapies, a rigorous safety assessment is paramount. However, comprehensive real-world safety studies have been lacking for the newly marketed CAR-T products idecabtagene vicleucel (ide-cel) and ciltacabtagene autoleucel (cilta-cel), which target B cell maturation antigen, especially regarding the risk of secondary malignancies. Therefore, we aimed to thoroughly analyze the adverse events (AEs) information in the FDA Adverse Event Reporting System (FAERS) database to comprehensively understand the safety risks of ide-cel and cilta-cel. Methods We extracted AE reports related to ide-cel and cilta-cel from the FAERS database (https://fis.fda.gov/extensions/FPD-QDE-FAERS/FPD-QDE-FAERS.html.) from January 1, 2019 to December 31, 2023. Disproportionality analysis and Bayesian analysis were used to identify risk signals across subgroups and specific cases (including for death and secondary malignancies). Weibull distribution analysis was employed to determine the time to AE onset. Results A total of 695 AE reports for ide-cel and 848 for cilta-cel were included in the FAERS database. This analysis identified 81 positive signals for ide-cel and 74 for cilta-cel. Notably, comparisons with the drug labels revealed "unexpected signals," including febrile bone marrow aplasia (reporting odds ratio=69.10; confidence interval 39.12-122.03) and plasma cell myeloma (12.45; 8.18-18.95) for ide-cel, and increased serum ferritin (24.98; 8.0-77.58) and large intestine perforation (18.57; 5.98-57.69) for cilta-cel. Both drugs showed a higher AE incidence among male recipients and patients aged ≥65 years, although female recipients faced a greater risk. Most AEs occurred at the early stage of administration. However, secondary malignancies were detected for both drugs, primarily occurring one-year post-administration. Conclusion This study provides a foundation for understanding the safety profile of CAR-T cell therapy, particularly in relation to the emergence of secondary malignancies. Such insights are helpful for clinical decision-making and the safe and effective utilization of these therapeutic agents.
Collapse
Affiliation(s)
- Wei Liu
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Shuzhi Lin
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Xiaoying Zhu
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Lin Yin
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Qian Liu
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Shuang Lei
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| | - Bianling Feng
- The Department of Pharmacy Administration, School of Pharmacy, Xi'an Jiaotong University, Xi'an, Shaanxi, China
- The Center for Drug Safety and Policy Research, Xi' an Jiaotong University, Xi' an, Shaanxi, China
| |
Collapse
|
22
|
Chen D, Zhu Y, Chen Z, Jiang S, He H, Qiang W, Xiang F, Sun X, Du J. A 5-Year Follow-up Clinical Study of the B-cell Maturation Antigen Chimeric Antigen Receptor T-cell Therapy HDS269B in Patients with Relapsed or Refractory Multiple Myeloma. Clin Cancer Res 2024; 30:3747-3756. [PMID: 38869658 PMCID: PMC11369620 DOI: 10.1158/1078-0432.ccr-24-0414] [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: 02/04/2024] [Revised: 03/27/2024] [Accepted: 06/11/2024] [Indexed: 06/14/2024]
Abstract
PURPOSE This study aimed to report the 5-year clinical outcomes of anti-B-cell maturation antigen chimeric antigen receptor (CAR) T-cell (HDS269B) therapy in patients with relapsed/refractory multiple myeloma (RRMM), including those with poor performance status [Eastern Cooperative Oncology Group (ECOG) scores 3 to 4], and to identify factors influencing long-term outcomes. PATIENTS AND METHODS Forty-nine patients with RRMM enrolled from 2016 to 2020 received HDS269B (9 × 106 cells/kg) after receiving a conditioning chemotherapy consisting of cyclophosphamide and fludarabine. The overall response, long-term outcomes, and safety were assessed, as were their associations with clinical and disease characteristics. RESULTS With a median follow-up of 59.0 months, the overall response rate was 77.55%. The median progression-free survival (PFS) and overall survival (OS) were 9.5 months [95% confidence interval (CI), 5.01-13.99] and 20.0 months (95% CI, 11.26-28.74), respectively. The 5-year PFS and OS rates were 21.3% (95% CI, 12.3%-36.7%) and 34.1% (95% CI, 22.7%-51.3%), respectively. Patients with ECOG 0 to 2 had marked longer survival, with a median PFS of 11.0 months and a median OS of 41.8 months. Early minimal residual disease negativity, higher and persistent CAR T-cell expansion, and the absence of extramedullary disease were associated with better survival outcomes. No new CAR T-cell therapy-associated toxicities were observed. Importantly, ECOG scores 0 to 2, prior therapy lines <4, and CAR T-cell persistence at ≥6 months were independently associated with longer OS. CONCLUSIONS HDS269B is effective and safe, especially for patients with ECOG scores 0 to 2. Early CAR T-cell intervention may improve prognosis in patients with RRMM.
Collapse
Affiliation(s)
- Dongjian Chen
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Shanghai, China.
| | - Yu Zhu
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Shanghai, China.
| | - Zhi Chen
- Department of Hematology, Henan Province Hospital of Traditional Chinese Medicine, Institute of Hematology, Henan University of Traditional Chinese Medicine, Zhengzhou, China.
| | - Songfu Jiang
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
| | - Haiyan He
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Shanghai, China.
| | - Wanting Qiang
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Shanghai, China.
| | - Fang Xiang
- HRAIN Biotechnology Co., Ltd., Shanghai, China.
| | - Xuedong Sun
- HRAIN Biotechnology Co., Ltd., Shanghai, China.
| | - Juan Du
- Department of Hematology, Myeloma & Lymphoma Center, Shanghai Changzheng Hospital, Shanghai, China.
| |
Collapse
|
23
|
Martin KE, Hammer Q, Perica K, Sadelain M, Malmberg KJ. Engineering immune-evasive allogeneic cellular immunotherapies. Nat Rev Immunol 2024; 24:680-693. [PMID: 38658708 DOI: 10.1038/s41577-024-01022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/05/2024] [Indexed: 04/26/2024]
Abstract
Allogeneic cellular immunotherapies hold a great promise for cancer treatment owing to their potential cost-effectiveness, scalability and on-demand availability. However, immune rejection of adoptively transferred allogeneic T and natural killer (NK) cells is a substantial obstacle to achieving clinical responses that are comparable to responses obtained with current autologous chimeric antigen receptor T cell therapies. In this Perspective, we discuss strategies to confer cell-intrinsic, immune-evasive properties to allogeneic T cells and NK cells in order to prevent or delay their immune rejection, thereby widening the therapeutic window. We discuss how common viral and cancer immune escape mechanisms can serve as a blueprint for improving the persistence of off-the-shelf allogeneic cell therapies. The prospects of harnessing genome editing and synthetic biology to design cell-based precision immunotherapies extend beyond programming target specificities and require careful consideration of innate and adaptive responses in the recipient that may curtail the biodistribution, in vivo expansion and persistence of cellular therapeutics.
Collapse
Affiliation(s)
- Karen E Martin
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway
- Department of Cancer Immunology, Institute for Cancer Research Oslo, Oslo University Hospital, Oslo, Norway
| | - Quirin Hammer
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Karlo Perica
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Cell Therapy Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Karl-Johan Malmberg
- Precision Immunotherapy Alliance, The University of Oslo, Oslo, Norway.
- Department of Cancer Immunology, Institute for Cancer Research Oslo, Oslo University Hospital, Oslo, Norway.
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden.
| |
Collapse
|
24
|
Xie L, Chen C, Zhang T, Yang W, Zheng D, Cao L, Yuan J, Xu Y, Zhang Y, Liu L, Liang A, Yu Z, Zheng J. LILRB4 regulates multiple myeloma development through STAT3-PFKFB1 pathway. Cell Death Dis 2024; 15:515. [PMID: 39025844 PMCID: PMC11258265 DOI: 10.1038/s41419-024-06883-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Revised: 06/19/2024] [Accepted: 07/02/2024] [Indexed: 07/20/2024]
Abstract
Although multiple myeloma (MM) responds well to immunotherapeutic treatment, certain portions of MM are still unresponsive or relapse after immunotherapy. Other immune molecules are needed for the immunotherapy of MM. Here, we revealed that leukocyte immunoglobulin-like receptor B4 (LILRB4) was highly expressed in multiple myeloma cell lines and patient samples and that the expression of LILRB4 was adversely correlated with the overall survival of MM patients. Knockdown of LILRB4 efficiently delayed the growth of MM cells both in vitro and in vivo. Mechanistically, IKZF1 transactivated LILRB4 expression to trigger the downstream of STAT3-PFKFB1 pathways to support MM cell proliferation. Blockade of LILRB4 signaling by blocking antibodies can effectively inhibit MM progression. Our data show that targeting LILRB4 is potentially an additional therapeutic strategy for the immunotherapeutic treatment of MM.
Collapse
Affiliation(s)
- Li Xie
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chiqi Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Tinghua Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wenqian Yang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Denghao Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Liyuan Cao
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Jin Yuan
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 201203, China
| | - Yilu Xu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yaping Zhang
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Ligen Liu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Aibin Liang
- Department of Hematology, Shanghai Tongji Hospital, Shanghai Tongji University School of Medicine, Shanghai, 200065, China.
| | - Zhuo Yu
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
25
|
Lee HJ, Hwang SJ, Jeong EH, Chang MH. Genetically Engineered CLDN18.2 CAR-T Cells Expressing Synthetic PD1/CD28 Fusion Receptors Produced Using a Lentiviral Vector. J Microbiol 2024; 62:555-568. [PMID: 38700775 PMCID: PMC11303488 DOI: 10.1007/s12275-024-00133-0] [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: 02/06/2024] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 08/07/2024]
Abstract
This study aimed to develop synthetic Claudin18.2 (CLDN18.2) chimeric antigen receptor (CAR)-T (CAR-T) cells as a treatment for advanced gastric cancer using lentiviral vector genetic engineering technology that targets the CLDN18.2 antigen and simultaneously overcomes the immunosuppressive environment caused by programmed cell death protein 1 (PD-1). Synthetic CAR T cells are a promising approach in cancer immunotherapy but face many challenges in solid tumors. One of the major problems is immunosuppression caused by PD-1. CLDN18.2, a gastric-specific membrane protein, is considered a potential therapeutic target for gastric and other cancers. In our study, CLDN18.2 CAR was a second-generation CAR with inducible T-cell costimulatory (CD278), and CLDN18.2-PD1/CD28 CAR was a third-generation CAR, wherein the synthetic PD1/CD28 chimeric-switch receptor (CSR) was added to the second-generation CAR. In vitro, we detected the secretion levels of different cytokines and the killing ability of CAR-T cells. We found that the secretion of cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) secreted by three types of CAR-T cells was increased, and the killing ability against CLDN18.2-positive GC cells was enhanced. In vivo, we established a xenograft GC model and observed the antitumor effects and off-target toxicity of CAR-T cells. These results support that synthetic anti-CLDN18.2 CAR-T cells have antitumor effect and anti-CLDN18.2-PD1/CD28 CAR could provide a promising design strategy to improve the efficacy of CAR-T cells in advanced gastric cancer.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Cell Line, Tumor
- Claudins/genetics
- Claudins/metabolism
- Cytokines/metabolism
- Genetic Engineering
- Genetic Vectors/genetics
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- Lentivirus/genetics
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Stomach Neoplasms/therapy
- Stomach Neoplasms/immunology
- Stomach Neoplasms/genetics
- T-Lymphocytes/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Heon Ju Lee
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea.
| | - Seo Jin Hwang
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| | - Eun Hee Jeong
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| | - Mi Hee Chang
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| |
Collapse
|
26
|
Paul S, Konig MF, Pardoll DM, Bettegowda C, Papadopoulos N, Wright KM, Gabelli SB, Ho M, van Elsas A, Zhou S. Cancer therapy with antibodies. Nat Rev Cancer 2024; 24:399-426. [PMID: 38740967 PMCID: PMC11180426 DOI: 10.1038/s41568-024-00690-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/29/2024] [Indexed: 05/16/2024]
Abstract
The greatest challenge in cancer therapy is to eradicate cancer cells with minimal damage to normal cells. Targeted therapy has been developed to meet that challenge, showing a substantially increased therapeutic index compared with conventional cancer therapies. Antibodies are important members of the family of targeted therapeutic agents because of their extraordinarily high specificity to the target antigens. Therapeutic antibodies use a range of mechanisms that directly or indirectly kill the cancer cells. Early antibodies were developed to directly antagonize targets on cancer cells. This was followed by advancements in linker technologies that allowed the production of antibody-drug conjugates (ADCs) that guide cytotoxic payloads to the cancer cells. Improvement in our understanding of the biology of T cells led to the production of immune checkpoint-inhibiting antibodies that indirectly kill the cancer cells through activation of the T cells. Even more recently, bispecific antibodies were synthetically designed to redirect the T cells of a patient to kill the cancer cells. In this Review, we summarize the different approaches used by therapeutic antibodies to target cancer cells. We discuss their mechanisms of action, the structural basis for target specificity, clinical applications and the ongoing research to improve efficacy and reduce toxicity.
Collapse
Affiliation(s)
- Suman Paul
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| | - Maximilian F Konig
- Division of Rheumatology, Department of Medicine, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Drew M Pardoll
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Chetan Bettegowda
- Department of Neurosurgery, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Katharine M Wright
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA
| | - Sandra B Gabelli
- Discovery Chemistry, Merck Research Laboratory, Merck and Co, West Point, PA, USA.
| | - Mitchell Ho
- Antibody Engineering Program, Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | | | - Shibin Zhou
- Department of Oncology, Johns Hopkins School of Medicine, Baltimore, MD, USA
| |
Collapse
|
27
|
Asensi Cantó P, Arnao Herraiz M, de la Rubia Comos J. [Immunotherapy in multiple myeloma]. Med Clin (Barc) 2024; 162:485-493. [PMID: 38218655 DOI: 10.1016/j.medcli.2023.11.019] [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: 10/03/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 01/15/2024]
Abstract
Patients with multiple myeloma who present with refractory disease or relapse after receiving the main classes of available drugs -immunomodulators, proteasome inhibitors and antibodies against CD38- do not have satisfactory therapeutic alternatives. New treatments based on the redirection of T lymphocytes to act directly against tumor cells, such as bispecific antibodies and T cells with chimeric antigen receptors, are changing this scenario. The published information confirms unprecedented antitumor activity of these agents in patients with refractory myeloma and they will certainly represent the backbone of the treatment of these patients in the immediate future. However, these therapies also present specific characteristics and medium or long-term toxicities that pose new healthcare challenges. In this review, we address the current results and future challenges of the administration of these treatments in patients with relapsed or refractory multiple myeloma.
Collapse
Affiliation(s)
- Pedro Asensi Cantó
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - Mario Arnao Herraiz
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, España
| | - Javier de la Rubia Comos
- Servicio de Hematología y Hemoterapia, Hospital Universitario y Politécnico La Fe, Valencia, España; Facultad de Medicina y Odontología, Universidad Católica de Valencia, Valencia, España; Instituto de Investigación Sanitaria La Fe, Valencia, España; Ciberonc CB16/12/00284, Valencia, España.
| |
Collapse
|
28
|
Sheykhhasan M, Ahmadieh-Yazdi A, Vicidomini R, Poondla N, Tanzadehpanah H, Dirbaziyan A, Mahaki H, Manoochehri H, Kalhor N, Dama P. CAR T therapies in multiple myeloma: unleashing the future. Cancer Gene Ther 2024; 31:667-686. [PMID: 38438559 PMCID: PMC11101341 DOI: 10.1038/s41417-024-00750-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/06/2024]
Abstract
In recent years, the field of cancer treatment has witnessed remarkable breakthroughs that have revolutionized the landscape of care for cancer patients. While traditional pillars such as surgery, chemotherapy, and radiation therapy have long been available, a cutting-edge therapeutic approach called CAR T-cell therapy has emerged as a game-changer in treating multiple myeloma (MM). This novel treatment method complements options like autologous stem cell transplants and immunomodulatory medications, such as proteasome inhibitors, by utilizing protein complexes or anti-CD38 antibodies with potent complement-dependent cytotoxic effects. Despite the challenges and obstacles associated with these treatments, the recent approval of the second FDA multiple myeloma CAR T-cell therapy has sparked immense promise in the field. Thus far, the results indicate its potential as a highly effective therapeutic solution. Moreover, ongoing preclinical and clinical trials are exploring the capabilities of CAR T-cells in targeting specific antigens on myeloma cells, offering hope for patients with relapsed/refractory MM (RRMM). These advancements have shown the potential for CAR T cell-based medicines or combination therapies to elicit greater treatment responses and minimize side effects. In this context, it is crucial to delve into the history and functions of CAR T-cells while acknowledging their limitations. We can strategize and develop innovative approaches to overcome these barriers by understanding their challenges. This article aims to provide insights into the application of CAR T-cells in treating MM, shedding light on their potential, limitations, and strategies employed to enhance their efficacy.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Amirhossein Ahmadieh-Yazdi
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi, University of Medical Sciences, Yazd, Iran
| | - Rosario Vicidomini
- Section on Cellular Communication, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ashkan Dirbaziyan
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - Paola Dama
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|
29
|
Blüm P, Kayser S. Chimeric Antigen Receptor (CAR) T-Cell Therapy in Hematologic Malignancies: Clinical Implications and Limitations. Cancers (Basel) 2024; 16:1599. [PMID: 38672680 PMCID: PMC11049267 DOI: 10.3390/cancers16081599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 04/16/2024] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has become a powerful treatment option in B-cell and plasma cell malignancies, and many patients have benefited from its use. To date, six CAR T-cell products have been approved by the FDA and EMA, and many more are being developed and investigated in clinical trials. The whole field of adoptive cell transfer has experienced an unbelievable development process, and we are now at the edge of a new era of immune therapies that will have its impact beyond hematologic malignancies. Areas of interest are, e.g., solid oncology, autoimmune diseases, infectious diseases, and others. Although much has been achieved so far, there is still a huge effort needed to overcome significant challenges and difficulties. We are witnessing a rapid expansion of knowledge, induced by new biomedical technologies and CAR designs. The era of CAR T-cell therapy has just begun, and new products will widen the therapeutic landscape in the future. This review provides a comprehensive overview of the clinical applications of CAR T-cells, focusing on the approved products and emphasizing their benefits but also indicating limitations and challenges.
Collapse
Affiliation(s)
- Philipp Blüm
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany;
| | - Sabine Kayser
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, Heidelberg University, German Red Cross Blood Service Baden-Württemberg-Hessen, 68167 Mannheim, Germany;
- NCT Trial Center, National Center of Tumor Diseases, Heidelberg University Hospital and German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Stock S, Fertig L, Gottschlich A, Dörr J, Märkl F, Majed L, Menkhoff VD, Grünmeier R, Rejeski K, Cordas Dos Santos DM, Theurich S, von Bergwelt-Baildon M, Endres S, Subklewe M, Kobold S. Comparative performance of scFv-based anti-BCMA CAR formats for improved T cell therapy in multiple myeloma. Cancer Immunol Immunother 2024; 73:100. [PMID: 38630291 PMCID: PMC11024081 DOI: 10.1007/s00262-024-03688-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 03/22/2024] [Indexed: 04/19/2024]
Abstract
In multiple myeloma (MM), B cell maturation antigen (BCMA)-directed CAR T cells have emerged as a novel therapy with potential for long-term disease control. Anti-BCMA CAR T cells with a CD8-based transmembrane (TM) and CD137 (41BB) as intracellular costimulatory domain are in routine clinical use. As the CAR construct architecture can differentially impact performance and efficacy, the optimal construction of a BCMA-targeting CAR remains to be elucidated. Here, we hypothesized that varying the constituents of the CAR structure known to impact performance could shed light on how to improve established anti-BCMA CAR constructs. CD8TM.41BBIC-based anti-BCMA CAR vectors with either a long linker or a short linker between the light and heavy scFv chain, CD28TM.41BBIC-based and CD28TM.CD28IC-based anti-BCMA CAR vector systems were used in primary human T cells. MM cell lines were used as target cells. The short linker anti-BCMA CAR demonstrated higher cytokine production, whereas in vitro cytotoxicity, T cell differentiation upon activation and proliferation were superior for the CD28TM.CD28IC-based CAR. While CD28TM.CD28IC-based CAR T cells killed MM cells faster, the persistence of 41BBIC-based constructs was superior in vivo. While CD28 and 41BB costimulation come with different in vitro and in vivo advantages, this did not translate into a superior outcome for either tested model. In conclusion, this study showcases the need to study the influence of different CAR architectures based on an identical scFv individually. It indicates that current scFv-based anti-BCMA CAR with clinical utility may already be at their functional optimum regarding the known structural variations of the scFv linker.
Collapse
Affiliation(s)
- Sophia Stock
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany.
| | - Luisa Fertig
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Adrian Gottschlich
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
| | - Janina Dörr
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Florian Märkl
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Lina Majed
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Vivien D Menkhoff
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Ruth Grünmeier
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
| | - Kai Rejeski
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - David M Cordas Dos Santos
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany
- Cancer- and Immunometabolism Research Group, LMU Gene Center, Munich, Germany
| | - Sebastian Theurich
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany
- Cancer- and Immunometabolism Research Group, LMU Gene Center, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany
| | - Stefan Endres
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany
| | - Marion Subklewe
- Department of Medicine III, LMU University Hospital, LMU Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany
- Laboratory of Translational Cancer Immunology, LMU Gene Center, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, LMU University Hospital, LMU Munich, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the LMU University Hospital, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München, German Research Center for Environmental Health (HMGU), Neuherberg, Germany.
| |
Collapse
|
31
|
Casey NP, Kleinmanns K, Forcados C, Gelebart PF, Joaquina S, Lode M, Benard E, Kaveh F, Caulier B, Helgestad Gjerde C, García de Jalón E, Warren DJ, Lindemann K, Rokkones E, Davidson B, Myhre MR, Kvalheim G, Bjørge L, McCormack E, Inderberg EM, Wälchli S. Efficient CAR T cell targeting of the CA125 extracellular repeat domain of MUC16. J Immunother Cancer 2024; 12:e008179. [PMID: 38604812 PMCID: PMC11015285 DOI: 10.1136/jitc-2023-008179] [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: 03/08/2024] [Indexed: 04/13/2024] Open
Abstract
BACKGROUND Ovarian cancer (OC) is the leading cause of death from gynecologic malignancies in the Western world. Contributing factors include a high frequency of late-stage diagnosis, the development of chemoresistance, and the evasion of host immune responses. Currently, debulking surgery and platinum-based chemotherapy are the treatment cornerstones, although recurrence is common. As the clinical efficacy of immune checkpoint blockade is low, new immunotherapeutic strategies are needed. Chimeric antigen receptor (CAR) T cell therapy empowers patients' own T cells to fight and eradicate cancer, and has been tested against various targets in OC. A promising candidate is the MUC16 ectodomain. This ectodomain remains on the cell surface after cleavage of cancer antigen 125 (CA125), the domain distal from the membrane, which is currently used as a serum biomarker for OC. CA125 itself has not been tested as a possible CAR target. In this study, we examined the suitability of the CA125 as a target for CAR T cell therapy. METHODS We tested a series of antibodies raised against the CA125 extracellular repeat domain of MUC16 and adapted them to the CAR format. Comparisons between these candidates, and against an existing CAR targeting the MUC16 ectodomain, identified K101 as having high potency and specificity. The K101CAR was subjected to further biochemical and functional tests, including examination of the effect of soluble CA125 on its activity. Finally, we used cell lines and advanced orthotopic patient-derived xenograft (PDX) models to validate, in vivo, the efficiency of our K101CAR construct. RESULTS We observed a high efficacy of K101CAR T cells against cell lines and patient-derived tumors, in vitro and in vivo. We also demonstrated that K101CAR functionality was not impaired by the soluble antigen. Finally, in direct comparisons, K101CAR, which targets the CA125 extracellular repeat domains, was shown to have similar efficacy to the previously validated 4H11CAR, which targets the MUC16 ectodomain. CONCLUSIONS Our in vitro and in vivo results, including PDX studies, demonstrate that the CA125 domain of MUC16 represents an excellent target for treating MUC16-positive malignancies.
Collapse
Affiliation(s)
- Nicholas P Casey
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Katrin Kleinmanns
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Christopher Forcados
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Pascal F Gelebart
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Sandy Joaquina
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Martine Lode
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Emmanuelle Benard
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Fatemeh Kaveh
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Benjamin Caulier
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
- Department of Molecular Cell Biology, Institute for Cancer Research, Oslo University Hospital, Oslo, Norway
- Center for Cancer Cell Reprogramming (CanCell), Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Christiane Helgestad Gjerde
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Elvira García de Jalón
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - David J Warren
- Department of Medical Biochemistry, Oslo University Hospital, Oslo, Norway
| | - Kristina Lindemann
- Department of Gynecologic Oncology, Oslo University Hospital, Oslo, Norway
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Erik Rokkones
- Department of Gynecologic Oncology, Oslo University Hospital, Oslo, Norway
| | - Ben Davidson
- Institute for Clinical Medicine, Faculty of Medicine, University of Oslo, Oslo, Norway
- Department of Pathology, Division of Laboratory Medicine, Oslo University Hospital, Oslo, Norway
| | - Marit Renee Myhre
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Gunnar Kvalheim
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Line Bjørge
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Emmet McCormack
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Science, University of Bergen, Bergen, Norway
- Centre for Pharmacy, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Internal Medicine, Hematology Section, Haukeland University Hospital, Bergen, Norway
| | - Else Marit Inderberg
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| | - Sébastien Wälchli
- Translational Research Unit, Section of Cellular Therapy, Department of Oncology, Oslo University Hospital, Oslo, Norway
| |
Collapse
|
32
|
Fang J, Zhou F. BCMA-targeting chimeric antigen receptor T cell therapy for relapsed and/or refractory multiple myeloma. Ann Hematol 2024; 103:1069-1083. [PMID: 37704875 DOI: 10.1007/s00277-023-05444-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/06/2023] [Indexed: 09/15/2023]
Abstract
Recently, many new therapies have improved the outcomes of patients with relapsed and/or refractory multiple myeloma (RRMM). Nevertheless, recurrence is still unavoidable, and better treatment choices for RRMM are urgently needed. The clinical success of Chimera antigen receptor (CAR) T cell therapy in many hematological diseases, including leukemia and lymphoma, has drawn considerable attention to RRMM. As CAR T cell therapy continues to mature and challenge traditional therapies, it is gradually changing the treatment paradigm for MM patients. The B cell maturation antigen (BCMA), expressed in malignant plasma cells but not normal ones, is an ideal target for MM treatment, due to its high expression. The US Food and Drug Administration (FDA) and European Medicines Agency (EMA) has approved two BCMA-targeting CAR T cell products, idecabtagene vicleucel (Ide-cel) and ciltacabtagene autoleucel (Cilta-cel), for use in RRMM. In this review, we focus on data from RRMM patients involved in clinical trials of Ide-cel and Cilta-cel and discuss the present situation and future direction of CAR T cell therapy for this condition.
Collapse
Affiliation(s)
- Jiamin Fang
- Department of Hematology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, 430072, China
| | - Fuling Zhou
- Department of Hematology, Zhongnan Hospital of Wuhan University, No.169 Donghu Road, Wuhan, 430072, China.
| |
Collapse
|
33
|
Hughes CFM, Shah GL, Paul BA. Autologous hematopoietic stem cell transplantation for multiple myeloma in the age of CAR T cell therapy. Front Oncol 2024; 14:1373548. [PMID: 38601770 PMCID: PMC11004402 DOI: 10.3389/fonc.2024.1373548] [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: 01/19/2024] [Accepted: 03/15/2024] [Indexed: 04/12/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has revolutionized the management of relapsed and refractory myeloma, with excellent outcomes and a tolerable safety profile. High dose chemotherapy with autologous hematopoietic stem cell transplantation (AHCT) is established as a mainstream of newly diagnosed multiple myeloma (NDMM) management in patients who are young and fit enough to tolerate such intensity. This standard was developed based on randomized trials comparing AHCT to chemotherapy in the era prior to novel agents. More recently, larger studies have primarily shown a progression free survival (PFS) benefit of upfront AHCT, rather than overall survival (OS) benefit. There is debate about the significance of this lack of OS, acknowledging the potential confounders of the chronic nature of the disease, study design and competing harms and benefits of exposure to AHCT. Indeed upfront AHCT may not be as uniquely beneficial as we once thought, and is not without risk. New quadruple-agent regimens are highly active and effective in achieving a deep response as quantified by measurable residual disease (MRD). The high dose chemotherapy administered with AHCT imposes a burden of short and long-term adverse effects, which may alter the disease course and patient's ability to tolerate future therapies. Some high-risk subgroups may have a more valuable benefit from AHCT, though still ultimately suffer poor outcomes. When compared to the outcomes of CAR T cell therapy, the question of whether AHCT can or indeed should be deferred has become an important topic in the field. Deferring AHCT may be a personalized decision in patients who achieve MRD negativity, which is now well established as a key prognostic factor for PFS and OS. Reserving or re-administering AHCT at relapse is feasible in many cases and holds the promise of resetting the T cell compartment and opening up options for immune reengagement. It is likely that personalized MRD-guided decision making will shape how we sequence in the future, though more studies are required to delineate when this is safe and appropriate.
Collapse
Affiliation(s)
- Charlotte F. M. Hughes
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Gunjan L. Shah
- Adult Bone Marrow Transplant Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
- Cellular Therapy Service, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Barry A. Paul
- Department of Hematologic Oncology and Blood Disorders, Levine Cancer Institute, Atrium Health/Wake Forest Baptist, Charlotte, NC, United States
| |
Collapse
|
34
|
Scheller L, Tebuka E, Rambau PF, Einsele H, Hudecek M, Prommersberger SR, Danhof S. BCMA CAR-T cells in multiple myeloma-ready for take-off? Leuk Lymphoma 2024; 65:143-157. [PMID: 37997705 DOI: 10.1080/10428194.2023.2276676] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 10/24/2023] [Indexed: 11/25/2023]
Abstract
Although the approval of new drugs has improved the clinical outcome of multiple myeloma (MM), it was widely regarded as incurable over the past decades. However, recent advancements in groundbreaking immunotherapies, such as chimeric antigen receptor T cells (CAR-T), have yielded remarkable results in heavily pretreated relapse/refractory patients, instilling hope for a potential cure. CAR-T are genetically modified cells armed with a novel receptor to specifically recognize and kill tumor cells. Among the potential targets for MM, the B-cell maturation antigen (BCMA) stands out since it is highly and almost exclusively expressed on plasma cells. Here, we review the currently approved BCMA-directed CAR-T products and ongoing clinical trials in MM. Furthermore, we explore innovative approaches to enhance BCMA-directed CAR-T and overcome potential reasons for treatment failure. Additionally, we explore the side effects associated with these novel therapies and shed light on accessibility of CAR-T therapy around the world.
Collapse
Affiliation(s)
- Lukas Scheller
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
- Interdisziplinäres Zentrum für Klinische Forschung (IZKF), Universitätsklinikum Würzburg, Würzburg, Germany
| | - Erius Tebuka
- Department of Pathology, Catholic University of Health and Allied Sciences (CUHAS), Mwanza, Tanzania
- Else-Kröner-Center Würzburg-Mwanza, Catholic University of Health and Allied Sciences (CUHAS), Mwanza, Tanzania
| | - Peter Fabian Rambau
- Department of Pathology, Catholic University of Health and Allied Sciences (CUHAS), Mwanza, Tanzania
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Sabrina Rebecca Prommersberger
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Sophia Danhof
- Medizinische Klinik und Poliklinik II und Lehrstuhl für zelluläre Immuntherapie, Medizinische Klinik II, Universitätsklinikum Würzburg, Würzburg, Germany
- Mildred Scheel Early Career Center, Universitätsklinikum Würzburg, Würzburg, Germany
| |
Collapse
|
35
|
Fischer L, Grieb N, Platzbecker U, Vucinic V, Merz M. CAR T cell therapy in multiple myeloma, where are we now and where are we heading for? Eur J Haematol 2024; 112:19-27. [PMID: 37547971 DOI: 10.1111/ejh.14051] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/07/2023] [Accepted: 07/10/2023] [Indexed: 08/08/2023]
Abstract
The introduction of chimeric antigen receptor (CAR) T cells revolutionized treatment of relapsed and refractory multiple myeloma (RRMM) in recent years. Currently, two CAR T cell products-idecabtagene vicleucel and ciltacabtagene autoleucel-are approved in the United States and the European Union to treat patients with three prior lines of therapy, including a proteasome inhibitor, an immunomodulatory drug, and an anti-CD38 antibody. Moreover, seminal phase III trials of both agents in earlier lines of therapy have been published recently. Despite unprecedented rates of deep and lasting remissions in RRMM, there are still areas of uncertainty regarding the optimal use and distribution of CAR T cells in multiple myeloma. In the current review, we discuss the available data on approved CAR T cell products as well as unmet clinical needs and ongoing developments to optimize usage of this promising treatment modality in multiple myeloma.
Collapse
Affiliation(s)
- Luise Fischer
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Nora Grieb
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectiology, University Hospital of Leipzig, Leipzig, Germany
- Innovation Center Computer Assisted Surgery (ICCAS), University of Leipzig, Leipzig, Germany
| | - Uwe Platzbecker
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Vladan Vucinic
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| | - Maximilian Merz
- Department of Hematology, Cellular Therapy, Hemostaseology and Infectiology, University Hospital of Leipzig, Leipzig, Germany
| |
Collapse
|
36
|
Mishra AK, Gupta A, Dagar G, Das D, Chakraborty A, Haque S, Prasad CP, Singh A, Bhat AA, Macha MA, Benali M, Saini KS, Previs RA, Saini D, Saha D, Dutta P, Bhatnagar AR, Darswal M, Shankar A, Singh M. CAR-T-Cell Therapy in Multiple Myeloma: B-Cell Maturation Antigen (BCMA) and Beyond. Vaccines (Basel) 2023; 11:1721. [PMID: 38006053 PMCID: PMC10674477 DOI: 10.3390/vaccines11111721] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Revised: 10/19/2023] [Accepted: 11/12/2023] [Indexed: 11/26/2023] Open
Abstract
Significant progress has been achieved in the realm of therapeutic interventions for multiple myeloma (MM), leading to transformative shifts in its clinical management. While conventional modalities such as surgery, radiotherapy, and chemotherapy have improved the clinical outcomes, the overarching challenge of effecting a comprehensive cure for patients afflicted with relapsed and refractory MM (RRMM) endures. Notably, adoptive cellular therapy, especially chimeric antigen receptor T-cell (CAR-T) therapy, has exhibited efficacy in patients with refractory or resistant B-cell malignancies and is now also being tested in patients with MM. Within this context, the B-cell maturation antigen (BCMA) has emerged as a promising candidate for CAR-T-cell antigen targeting in MM. Alternative targets include SLAMF7, CD38, CD19, the signaling lymphocyte activation molecule CS1, NKG2D, and CD138. Numerous clinical studies have demonstrated the clinical efficacy of these CAR-T-cell therapies, although longitudinal follow-up reveals some degree of antigenic escape. The widespread implementation of CAR-T-cell therapy is encumbered by several barriers, including antigenic evasion, uneven intratumoral infiltration in solid cancers, cytokine release syndrome, neurotoxicity, logistical implementation, and financial burden. This article provides an overview of CAR-T-cell therapy in MM and the utilization of BCMA as the target antigen, as well as an overview of other potential target moieties.
Collapse
Affiliation(s)
- Abhinava K. Mishra
- Molecular, Cellular and Developmental Biology Department, University of California Santa Barbara, Santa Barbara, CA 93106, USA;
| | - Ashna Gupta
- Department of Medical Oncology (Lab), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.G.); (G.D.); (C.P.P.)
| | - Gunjan Dagar
- Department of Medical Oncology (Lab), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.G.); (G.D.); (C.P.P.)
| | - Dayasagar Das
- Department of Medicine, NYU Langone Health, New York, NY 10016, USA;
| | - Abhijit Chakraborty
- Department of Investigational Cancer Therapeutics, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA;
| | - Shabirul Haque
- Feinstein Institute of Medical Research, Northwell Health, Manhasset, NY 11030, USA;
| | - Chandra Prakash Prasad
- Department of Medical Oncology (Lab), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.G.); (G.D.); (C.P.P.)
| | - Archana Singh
- Department of Biochemistry, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India;
| | - Ajaz A. Bhat
- Precision Medicine in Diabetes, Obesity and Cancer Program, Department of Human Genetics, Sidra Medicine, Doha P.O. Box 26999, Qatar;
| | - Muzafar A. Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Awantipora 192122, India;
| | - Moez Benali
- Fortrea Inc., Durham, NC 27709, USA; (M.B.); (K.S.S.)
| | - Kamal S. Saini
- Fortrea Inc., Durham, NC 27709, USA; (M.B.); (K.S.S.)
- Addenbrooke’s Hospital, Cambridge University Hospitals NHS Foundation Trust, Cambridge CB2 0QQ, UK
| | - Rebecca Ann Previs
- Labcorp Oncology, Durham, NC 27560, USA;
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, Duke University Medical Center, Durham, NC 27710, USA
| | - Deepak Saini
- Department of Materia Medica, State Lal Bahadur Shastri Homoeopathic Medical College, Prayagraj 211013, India;
| | - Dwaipayan Saha
- Pratap Chandra Memorial Homoeopathic Hospital & College, Kolkata 700011, India; (D.S.); (P.D.)
| | - Preyangsee Dutta
- Pratap Chandra Memorial Homoeopathic Hospital & College, Kolkata 700011, India; (D.S.); (P.D.)
| | - Aseem Rai Bhatnagar
- Department of Radiation Oncology, Henry Ford Cancer Institute, Detroit, MI 48202, USA;
| | - Mrinalini Darswal
- Harvard T.H. Chan School of Public Health, Huntington Ave, Boston, MA 02115, USA;
| | - Abhishek Shankar
- Department of Radiation Oncology, Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Mayank Singh
- Department of Medical Oncology (Lab), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India; (A.G.); (G.D.); (C.P.P.)
| |
Collapse
|
37
|
Miliotou AN, Georgiou-Siafis SK, Ntenti C, Pappas IS, Papadopoulou LC. Recruiting In Vitro Transcribed mRNA against Cancer Immunotherapy: A Contemporary Appraisal of the Current Landscape. Curr Issues Mol Biol 2023; 45:9181-9214. [PMID: 37998753 PMCID: PMC10670245 DOI: 10.3390/cimb45110576] [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: 10/15/2023] [Revised: 11/05/2023] [Accepted: 11/14/2023] [Indexed: 11/25/2023] Open
Abstract
Over 100 innovative in vitro transcribed (IVT)-mRNAs are presently undergoing clinical trials, with a projected substantial impact on the pharmaceutical market in the near future. Τhe idea behind this is that after the successful cellular internalization of IVT-mRNAs, they are subsequently translated into proteins with therapeutic or prophylactic relevance. Simultaneously, cancer immunotherapy employs diverse strategies to mobilize the immune system in the battle against cancer. Therefore, in this review, the fundamental principles of IVT-mRNA to its recruitment in cancer immunotherapy, are discussed and analyzed. More specifically, this review paper focuses on the development of mRNA vaccines, the exploitation of neoantigens, as well as Chimeric Antigen Receptor (CAR) T-Cells, showcasing their clinical applications and the ongoing trials for the development of next-generation immunotherapeutics. Furthermore, this study investigates the synergistic potential of combining the CAR immunotherapy and the IVT-mRNAs by introducing our research group novel, patented delivery method that utilizes the Protein Transduction Domain (PTD) technology to transduce the IVT-mRNAs encoding the CAR of interest into the Natural Killer (NK)-92 cells, highlighting the potential for enhancing the CAR NK cell potency, efficiency, and bioenergetics. While IVT-mRNA technology brings exciting progress to cancer immunotherapy, several challenges and limitations must be acknowledged, such as safety, toxicity, and delivery issues. This comprehensive exploration of IVT-mRNA technology, in line with its applications in cancer therapeutics, offers valuable insights into the opportunities and challenges in the evolving landscape of cancer immunotherapy, setting the stage for future advancements in the field.
Collapse
Affiliation(s)
- Androulla N. Miliotou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- Department of Health Sciences, KES College, 1055 Nicosia, Cyprus
- Faculty of Pharmacy, Department of Health Sciences, University of Nicosia, 1700 Nicosia, Cyprus
| | - Sofia K. Georgiou-Siafis
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Thessaly, Greece;
| | - Charikleia Ntenti
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
- 1st Laboratory of Pharmacology, School of Medicine, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece
| | - Ioannis S. Pappas
- Laboratory of Pharmacology and Toxicology, Faculty of Veterinary Medicine, University of Thessaly, 43100 Karditsa, Thessaly, Greece;
| | - Lefkothea C. Papadopoulou
- Laboratory of Pharmacology, School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, 54124 Thessaloniki, Macedonia, Greece; (A.N.M.); (S.K.G.-S.); (C.N.)
| |
Collapse
|
38
|
Niederlova V, Tsyklauri O, Kovar M, Stepanek O. IL-2-driven CD8 + T cell phenotypes: implications for immunotherapy. Trends Immunol 2023; 44:890-901. [PMID: 37827864 PMCID: PMC7615502 DOI: 10.1016/j.it.2023.09.003] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 09/11/2023] [Accepted: 09/12/2023] [Indexed: 10/14/2023]
Abstract
The therapeutic potential of interleukin (IL)-2 in cancer treatment has been known for decades, yet its widespread adoption in clinical practice remains limited. Recently, chimeric proteins of an anti-PD-1 antibody and suboptimal IL-2 variants were shown to stimulate potent antitumor and antiviral immunity by inducing unique effector CD8+ T cells in mice. A similar subset of cytotoxic T cells is induced by depletion of regulatory T cells (Tregs), suggesting IL-2 sequestration as a major mechanism through which regulatory T cells suppress activated CD8+ T cells. Here, we present our view of how IL-2-based biologicals can boost the antitumor response at a cellular level, and propose that the role of Tregs following such treatments may have been previously overestimated.
Collapse
Affiliation(s)
- Veronika Niederlova
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Oksana Tsyklauri
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic
| | - Marek Kovar
- Laboratory of Tumor Immunology, Institute of Microbiology of the Czech Academy of Sciences, Prague, Czech Republic
| | - Ondrej Stepanek
- Laboratory of Adaptive Immunity, Institute of Molecular Genetics of the Czech Academy of Sciences, Prague, Czech Republic.
| |
Collapse
|
39
|
Wachsmann TLA, Meeuwsen MH, Remst DFG, Buchner K, Wouters AK, Hagedoorn RS, Falkenburg JHF, Heemskerk MHM. Combining BCMA-targeting CAR T cells with TCR-engineered T-cell therapy to prevent immune escape of multiple myeloma. Blood Adv 2023; 7:6178-6183. [PMID: 37567150 PMCID: PMC10582830 DOI: 10.1182/bloodadvances.2023010410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 07/22/2023] [Accepted: 08/04/2023] [Indexed: 08/13/2023] Open
Affiliation(s)
| | - Miranda H. Meeuwsen
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dennis F. G. Remst
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Karen Buchner
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Anne K. Wouters
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Renate S. Hagedoorn
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | |
Collapse
|
40
|
O’Neal J, Cooper ML, Ritchey JK, Gladney S, Niswonger J, González LS, Street E, Haas GJ, Carter A, Amayta PN, Gao F, Lee BH, Choi D, Berrien-Elliott M, Zhou A, Fehniger TA, Rettig MP, DiPersio JF. Anti-myeloma efficacy of CAR-iNKT is enhanced with a long-acting IL-7, rhIL-7-hyFc. Blood Adv 2023; 7:6009-6022. [PMID: 37399471 PMCID: PMC10582278 DOI: 10.1182/bloodadvances.2023010032] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 05/30/2023] [Accepted: 06/19/2023] [Indexed: 07/05/2023] Open
Abstract
Multiple myeloma (MM), a malignancy of mature plasma cells, remains incurable. B-cell maturation antigen (BCMA) is the lead protein target for chimeric antigen receptor (CAR) therapy because of its high expression in most MM, with limited expression in other cell types, resulting in favorable on-target, off tumor toxicity. The response rate to autologous BCMA CAR-T therapy is high; however, it is not curative and is associated with risks of cytokine release syndrome (CRS) and immune effector cell-associated neurotoxicity syndrome. Outcomes in patients treated with BCMA CAR-T cells (CAR-Ts) may improve with allogeneic CAR T-cell therapy, which offer higher cell fitness and reduced time to treatment. However, to prevent the risk of graft-versus-host disease (GVHD), allogenic BCMA CAR-Ts require genetic deletion of the T-cell receptor (TCR), which has potential for unexpected functional or phenotype changes. Invariant natural killer T cells (iNKTs) have an invariant TCR that does not cause GVHD and, as a result, can be used in an allogeneic setting without the need for TCR gene editing. We demonstrate significant anti-myeloma activity of BCMA CAR-iNKTs in a xenograft mouse model of myeloma. We found that a long-acting interleukin-7 (IL-7), rhIL-7-hyFc, significantly prolonged survival and reduced tumor burden in BCMA CAR-iNKT-treated mice in both primary and re-challenge settings. Furthermore, in CRS in vitro assays, CAR-iNKTs induced less IL-6 than CAR-Ts, suggesting a reduced likelihood of CAR-iNKT therapy to induce CRS in patients. These data suggest that BCMA CAR-iNKTs are potentially a safer, effective alternative to BCMA CAR-Ts and that BCMA CAR-iNKT efficacy is further potentiated with rhIL-7-hyFc.
Collapse
Affiliation(s)
- Julie O’Neal
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO
| | - Matthew L. Cooper
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Julie K. Ritchey
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Susan Gladney
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Jessica Niswonger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - L. Sofía González
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Emily Street
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Gabriel J. Haas
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Alun Carter
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Parmeshwar N. Amayta
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
| | - Feng Gao
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine, Saint Louis, MO
| | | | | | - Melissa Berrien-Elliott
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO
| | - Alice Zhou
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO
| | - Todd A. Fehniger
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO
| | - Mike P. Rettig
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO
| | - John F. DiPersio
- Division of Oncology, Department of Medicine, Washington University School of Medicine, Saint Louis, MO
- Siteman Cancer Center, Washington University in St. Louis, St. Louis, MO
| |
Collapse
|
41
|
Zhang X, Ouyang C, Sun G, Liu H, Qi J, Suo X. Anti‑BCMA CAR‑T cell immunotherapy for relapsed or refractory multiple myeloma. Exp Ther Med 2023; 26:471. [PMID: 37664681 PMCID: PMC10468803 DOI: 10.3892/etm.2023.12170] [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: 05/12/2023] [Accepted: 07/20/2023] [Indexed: 09/05/2023] Open
Abstract
The present study aimed to study the efficacy and adverse effects of anti-B-cell maturation antigen (BCMA) chimeric antigen receptor T (CAR-T) cell therapy in relapsed or refractory multiple myeloma. Patients were divided into three dose groups based on cell therapy concentration. After CAR-T cell therapy for 10 patients with recurrent or refractory multiple myeloma, the patients were monitored and evaluated regularly to observe the efficacy and adverse reactions of CAR-T cell therapy. At a median follow-up of 337 (253-504) days, one patient succumbed 24 days due to rapidly progressing disease. The overall response rate of nine patients was 88.9%, including 77.8% (7/9) with minimal residual disease negative complete remission (CR) and 11.1% (1/9) with partial remission. A total of three patients were maintained in remission state for more than a year and eight were maintained for more than six months. Among the three patients with extramedullary invasion, two extramedullary lesions disappeared and one was stable. The highest copy number of CAR-T cells in seven patients with CR was >1x105 copies/µl gDNA, and the best therapeutic effect can be achieved within 30 (7-30) days after the copy number of CAR-T cells reached 1x105 copies/µl genomic DNA. The median onset time in the nine patients was 43 (22-169) days, and the median progression-free survival was 337 (253-504). Among the 10 patients, nine (90%) had cytokine release syndrome, all of which were below grade II. There were nine (90%) patients with hematological adverse reactions, six (60%) patients with severe anemia, five (50%) patients with grade III and above leukopenia, five (50%) patients with granulocytopenia, four (40%) patients with grade III and above thrombocytopenia, and three (30%) patients with grade III and above pancytopenia. It was concluded that anti-BCMA CAR-T cell therapy is a promising treatment method for relapsed or refractory multiple myeloma and extramedullary invasion, with stable efficacy and controllable adverse effects.
Collapse
Affiliation(s)
- Xiaohui Zhang
- Department of Hematology, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Chenxi Ouyang
- Department of Vascular Surgery, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Beijing 100037, P.R. China
| | - Guofeng Sun
- Department of Hematology, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Hongfeng Liu
- Department of Hematology, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| | - Junyuan Qi
- Good Clinic Practice, Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences, Tianjin 300020, P.R. China
| | - Xiaohui Suo
- Department of Hematology, Handan Central Hospital, Handan, Hebei 056001, P.R. China
| |
Collapse
|
42
|
Salem AM, Mugundu GM, Singh AP. Development of a multiscale mechanistic modeling framework integrating differential cellular kinetics of CAR T-cell subsets and immunophenotypes in cancer patients. CPT Pharmacometrics Syst Pharmacol 2023; 12:1285-1304. [PMID: 37448297 PMCID: PMC10508581 DOI: 10.1002/psp4.13009] [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: 10/03/2022] [Revised: 06/26/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Chimeric antigen receptor (CAR) T-cell subsets and immunophenotypic composition of the pre-infusion product, as well as their longitudinal changes following infusion, are expected to affect CAR T-cell expansion, persistence, and clinical outcomes. Herein, we sequentially evolved our previously described cellular kinetic-pharmacodynamic (CK-PD) model to incorporate CAR T-cell product-associated attributes by utilizing published preclinical and clinical datasets from two affinity variants (FMC63 and CAT19 scFv) anti-CD19 CAR T-cells. In step 1, a unified cell-level PD model was used to simultaneously characterize the in vitro killing datasets of two CAR T-cells against CD19+ cell lines at varying effector:target ratios. In step 2, an augmented CK-PD model for anti-CD19 CAR T-cells was developed, by integrating CK dataset(s) from two bioanalytical measurements (quantitative polymerase chain reaction and flow cytometry) in patients with cancer. The model described the differential in vivo expansion properties of CAR T-cell subsets. The estimated expansion rate constant was ~1.12-fold higher for CAR+CD8+ cells in comparison to CAR+CD4+ T-cells. In step 3, the model was extended to characterize the disposition of four immunophenotypic populations of CAR T-cells, including stem-cell memory, central memory, effector memory, and effector cells. The model adequately characterized the longitudinal changes in immunophenotypes post anti-CD19 CAR T-cell infusion in pediatric patients with acute lymphocytic leukemia. Polyclonality in the pre-infusion product was identified as a categorical covariate influencing differentiation of immunophenotypes. In the future, this model could be leveraged a priori toward optimizing the composition of CAR T-cell infusion product, and further understand the CK-PD relationship in patients.
Collapse
Affiliation(s)
- Ahmed M. Salem
- Clinical Pharmacology and Modeling, Precision and Translational MedicineOncology Cell Therapy and Therapeutic Area Unit, Takeda PharmaceuticalsCambridgeMassachusettsUSA
- Center for Translational MedicineUniversity of Maryland School of PharmacyBaltimoreMarylandUSA
| | - Ganesh M. Mugundu
- Clinical Pharmacology and Modeling, Precision and Translational MedicineOncology Cell Therapy and Therapeutic Area Unit, Takeda PharmaceuticalsCambridgeMassachusettsUSA
| | - Aman P. Singh
- Clinical Pharmacology and Modeling, Precision and Translational MedicineOncology Cell Therapy and Therapeutic Area Unit, Takeda PharmaceuticalsCambridgeMassachusettsUSA
| |
Collapse
|
43
|
Lin Y, Raje NS, Berdeja JG, Siegel DS, Jagannath S, Madduri D, Liedtke M, Rosenblatt J, Maus MV, Massaro M, Petrocca F, Yeri A, Finney O, Caia A, Yang Z, Martin N, Campbell TB, Rytlewski J, Fuller J, Hege K, Munshi NC, Kochenderfer JN. Idecabtagene vicleucel for relapsed and refractory multiple myeloma: post hoc 18-month follow-up of a phase 1 trial. Nat Med 2023; 29:2286-2294. [PMID: 37592106 PMCID: PMC10504071 DOI: 10.1038/s41591-023-02496-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 07/11/2023] [Indexed: 08/19/2023]
Abstract
Idecabtagene vicleucel (ide-cel) is a B-cell-maturation antigen (BCMA)-directed chimeric antigen receptor T cell therapy. We performed a post hoc analysis of a single-arm phase 1 multicenter study in relapsed/refractory multiple myeloma (CRB-401) (n = 62; median follow-up, 18.1 months). The primary endpoint was safety outcomes, and secondary endpoints included overall response rate (ORR), complete response (CR) and very good partial response (VGPR). The study met its primary endpoint with low rates of grade 3/grade 4 cytokine release syndrome (6.5%) and neurotoxicity (1.6%). ORR was 75.8%; 64.5% achieved VGPR or better and 38.7% achieved CR or stringent CR. Among exploratory endpoints, median duration of response, progression-free survival (PFS) and overall survival were 10.3, 8.8 and 34.2 months, respectively, and ide-cel expansion in blood and bone marrow correlated with clinical efficacy and postinfusion reduction of soluble BCMA. Patients with PFS ≥ 18 months had more naive and less exhausted T cells in apheresis material and improved functional T cell phenotype in the drug product compared with those with less durable responses. These results confirm ide-cel safety, tolerability and efficacy and describe T cell qualities that correlate with durable response. Clinicaltrials.gov identifier : NCT02658929 .
Collapse
Affiliation(s)
- Yi Lin
- Mayo Clinic, Rochester, MN, USA.
| | - Noopur S Raje
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | - Jesús G Berdeja
- Sarah Cannon Research Institute and Tennessee Oncology, Nashville, TN, USA
| | - David S Siegel
- Hackensack University Medical Center, Hackensack, NJ, USA
| | | | | | | | | | - Marcela V Maus
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | - James N Kochenderfer
- Surgery Branch, National Cancer Institute/National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
44
|
Ahmed N, Wesson W, Mushtaq MU, Bansal R, AbdelHakim H, Bromert S, Appenfeller A, Ghazal BA, Singh A, Abhyankar S, Ganguly S, McGuirk J, Abdallah AO, Shune L. "Waitlist mortality" is high for myeloma patients with limited access to BCMA therapy. Front Oncol 2023; 13:1206715. [PMID: 37601685 PMCID: PMC10436079 DOI: 10.3389/fonc.2023.1206715] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2023] [Accepted: 06/29/2023] [Indexed: 08/22/2023] Open
Abstract
Background The first-in-class approved BCMA CAR-T therapy was idecabtagene vicleucel (ide-cel), approved in March 2021, for RRMM patients who progressed after 4 or more lines of therapy. Despite the promising outcomes, there were limited apheresis/production slots for ide-cel. We report outcomes of patients at our institution who were on the "waitlist" to receive ide-cel in 2021 and who could not secure a slot. Methods We conducted a retrospective review of RRMM patients evaluated at the University of Kansas Cancer Center for ide-cel from 3/2021-7/2021. A retrospective chart review was performed to determine patient and disease characteristics. Descriptive statistics were reported using medians for continuous variables. Survival analysis from initial consult was performed using Kaplan-Meier Survival estimator. Results Forty patients were eligible and were on the "waitlist" for CAR-T. The median follow-up was 14 months (2-25mo). Twenty-four patients (60%) secured a production slot and 16 (40%) did not. The median time from consult to collection was 38 days (8-703). The median time from collection to infusion was 42 days (34-132 days). The median overall survival was higher in the CAR-T group (NR vs 9 mo, p<0.001). Conclusions Many patients who were eligible for ide-cel were not able to secure a timely slot in 2021. Mortality was higher in this group, due to a lack of comparable alternatives. Increasing alternate options as well as improvement in manufacturing and access is an area of high importance to improve RRMM outcomes.
Collapse
Affiliation(s)
- Nausheen Ahmed
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - William Wesson
- School of Medicine, University of Kansas, Kansas, KS, United States
| | - Muhammad Umair Mushtaq
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Rajat Bansal
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Haitham AbdelHakim
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Sarah Bromert
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Allison Appenfeller
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Batool Abu Ghazal
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Anurag Singh
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Sunil Abhyankar
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Siddhartha Ganguly
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
- Mary and Ron Neal Cancer Center, Houston Methodist Hospital, Houston, TX, United States
| | - Joseph McGuirk
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Al-Ola Abdallah
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| | - Leyla Shune
- Division of Hematologic Malignancies & Cellular Therapeutics, University of Kansas Cancer Center, Westwood, KS, United States
| |
Collapse
|
45
|
Di Meo F, Iyer A, Akama K, Cheng R, Yu C, Cesarano A, Kurihara N, Tenshin H, Aljoufi A, Marino S, Soni RK, Roda J, Sissons J, Vu LP, Guzman M, Huang K, Laskowski T, Broxmeyer HE, Roodman DG, Perna F. A target discovery pipeline identified ILT3 as a target for immunotherapy of multiple myeloma. Cell Rep Med 2023; 4:101110. [PMID: 37467717 PMCID: PMC10394163 DOI: 10.1016/j.xcrm.2023.101110] [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: 11/15/2022] [Revised: 02/17/2023] [Accepted: 06/16/2023] [Indexed: 07/21/2023]
Abstract
Multiple myeloma (MM) is an incurable malignancy of plasma cells. To identify targets for MM immunotherapy, we develop an integrated pipeline based on mass spectrometry analysis of seven MM cell lines and RNA sequencing (RNA-seq) from 900+ patients. Starting from 4,000+ candidates, we identify the most highly expressed cell surface proteins. We annotate candidate protein expression in many healthy tissues and validate the expression of promising targets in 30+ patient samples with relapsed/refractory MM, as well as in primary healthy hematopoietic stem cells and T cells by flow cytometry. Six candidates (ILT3, SEMA4A, CCR1, LRRC8D, FCRL3, IL12RB1) and B cell maturation antigen (BCMA) present the most favorable profile in malignant and healthy cells. We develop a bispecific T cell engager targeting ILT3 that shows potent killing effects in vitro and decreased tumor burden and prolonged mice survival in vivo, suggesting therapeutic relevance. Our study uncovers MM-associated antigens that hold great promise for immune-based therapies of MM.
Collapse
Affiliation(s)
- Francesco Di Meo
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - Keith Akama
- NGM Biopharmaceuticals, San Francisco, CA, USA
| | - Rujin Cheng
- NGM Biopharmaceuticals, San Francisco, CA, USA
| | - Christina Yu
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Annamaria Cesarano
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Noriyoshi Kurihara
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Hirofumi Tenshin
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Arafat Aljoufi
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Silvia Marino
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Rajesh K Soni
- Proteomics and Macromolecular Crystallography Shared Resource, Herbert Irving Comprehensive Cancer Center, Columbia University Irving Medical Center, New York, NY, USA
| | - Julie Roda
- NGM Biopharmaceuticals, San Francisco, CA, USA
| | | | - Ly P Vu
- British Columbia Cancer Center, Vancouver, BC, Canada
| | | | - Kun Huang
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Hal E Broxmeyer
- Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA
| | - David G Roodman
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Fabiana Perna
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA; Department of Blood and Marrow Transplant and Cellular Immunotherapy, H. Lee Moffitt Cancer Center, Tampa, FL, USA; Department of Microbiology and Immunology, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
46
|
Jiang D, Huang H, Qin H, Tang K, Shi X, Zhu T, Gao Y, Zhang Y, Tian X, Fu J, Qu W, Cai W, Xu Y, Wu D, Chu J. Chimeric antigen receptor T cells targeting FcRH5 provide robust tumour-specific responses in murine xenograft models of multiple myeloma. Nat Commun 2023; 14:3642. [PMID: 37339964 DOI: 10.1038/s41467-023-39395-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2022] [Accepted: 06/07/2023] [Indexed: 06/22/2023] Open
Abstract
BCMA-targeting chimeric antigen receptor (CAR) T cell therapy demonstrates impressive clinical response in multiple myeloma (MM). However, some patients with BCMA-deficient tumours cannot benefit from this therapy, and others can experience BCMA antigen loss leading to relapse, thus necessitating the identification of additional CAR-T targets. Here, we show that FcRH5 is expressed on multiple myeloma cells and can be targeted with CAR-T cells. FcRH5 CAR-T cells elicited antigen-specific activation, cytokine secretion and cytotoxicity against MM cells. Moreover, FcRH5 CAR-T cells exhibited robust tumoricidal efficacy in murine xenograft models, including one deficient in BCMA expression. We also show that different forms of soluble FcRH5 can interfere with the efficacy of FcRH5 CAR-T cells. Lastly, FcRH5/BCMA-bispecific CAR-T cells efficiently recognized MM cells expressing FcRH5 and/or BCMA and displayed improved efficacy, compared with mono-specific CAR-T cells in vivo. These findings suggest that targeting FcRH5 with CAR-T cells may represent a promising therapeutic avenue for MM.
Collapse
Affiliation(s)
- Dongpeng Jiang
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Haiwen Huang
- Department of hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Huimin Qin
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Koukou Tang
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Xiangru Shi
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Tingting Zhu
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Yuqing Gao
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Ying Zhang
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Xiaopeng Tian
- Department of hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Jianhong Fu
- Department of hematology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Weiwei Qu
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Weilan Cai
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China
| | - Yang Xu
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.
| | - Depei Wu
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.
| | - Jianhong Chu
- Institute of Blood and Marrow Transplantation, National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, Jiangsu, China.
| |
Collapse
|
47
|
Yan X, Liu X, Zhao C, Chen GQ. Applications of synthetic biology in medical and pharmaceutical fields. Signal Transduct Target Ther 2023; 8:199. [PMID: 37169742 PMCID: PMC10173249 DOI: 10.1038/s41392-023-01440-5] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 03/15/2023] [Accepted: 03/24/2023] [Indexed: 05/13/2023] Open
Abstract
Synthetic biology aims to design or assemble existing bioparts or bio-components for useful bioproperties. During the past decades, progresses have been made to build delicate biocircuits, standardized biological building blocks and to develop various genomic/metabolic engineering tools and approaches. Medical and pharmaceutical demands have also pushed the development of synthetic biology, including integration of heterologous pathways into designer cells to efficiently produce medical agents, enhanced yields of natural products in cell growth media to equal or higher than that of the extracts from plants or fungi, constructions of novel genetic circuits for tumor targeting, controllable releases of therapeutic agents in response to specific biomarkers to fight diseases such as diabetes and cancers. Besides, new strategies are developed to treat complex immune diseases, infectious diseases and metabolic disorders that are hard to cure via traditional approaches. In general, synthetic biology brings new capabilities to medical and pharmaceutical researches. This review summarizes the timeline of synthetic biology developments, the past and present of synthetic biology for microbial productions of pharmaceutics, engineered cells equipped with synthetic DNA circuits for diagnosis and therapies, live and auto-assemblied biomaterials for medical treatments, cell-free synthetic biology in medical and pharmaceutical fields, and DNA engineering approaches with potentials for biomedical applications.
Collapse
Affiliation(s)
- Xu Yan
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Xu Liu
- PhaBuilder Biotech Co. Ltd., Shunyi District, Zhaoquan Ying, 101309, Beijing, China
| | - Cuihuan Zhao
- School of Life Sciences, Tsinghua University, 100084, Beijing, China
| | - Guo-Qiang Chen
- School of Life Sciences, Tsinghua University, 100084, Beijing, China.
- Center for Synthetic and Systems Biology, Tsinghua University, 100084, Beijing, China.
- MOE Key Lab for Industrial Biocatalysis, Dept Chemical Engineering, Tsinghua University, 100084, Beijing, China.
| |
Collapse
|
48
|
Cappell KM, Kochenderfer JN. Long-term outcomes following CAR T cell therapy: what we know so far. Nat Rev Clin Oncol 2023; 20:359-371. [PMID: 37055515 PMCID: PMC10100620 DOI: 10.1038/s41571-023-00754-1] [Citation(s) in RCA: 480] [Impact Index Per Article: 240.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/14/2023] [Indexed: 04/15/2023]
Abstract
Chimeric antigen receptors (CAR) are engineered fusion proteins designed to target T cells to antigens expressed on cancer cells. CAR T cells are now an established treatment for patients with relapsed and/or refractory B cell lymphomas, B cell acute lymphoblastic leukaemia and multiple myeloma. At the time of this writing, over a decade of follow-up data are available from the initial patients who received CD19-targeted CAR T cells for B cell malignancies. Data on the outcomes of patients who received B cell maturation antigen (BCMA)-targeted CAR T cells for multiple myeloma are more limited owing to the more recent development of these constructs. In this Review, we summarize long-term follow-up data on efficacy and toxicities from patients treated with CAR T cells targeting CD19 or BCMA. Overall, the data demonstrate that CD19-targeted CAR T cells can induce prolonged remissions in patients with B cell malignancies, often with minimal long-term toxicities, and are probably curative for a subset of patients. By contrast, remissions induced by BCMA-targeted CAR T cells are typically more short-lived but also generally have only limited long-term toxicities. We discuss factors associated with long-term remissions, including the depth of initial response, malignancy characteristics predictive of response, peak circulating CAR levels and the role of lymphodepleting chemotherapy. We also discuss ongoing investigational strategies designed to improve the length of remission following CAR T cell therapy.
Collapse
Affiliation(s)
- Kathryn M Cappell
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA
| | - James N Kochenderfer
- Surgery Branch, Center for Cancer Research, National Cancer Institute (NCI), NIH, Bethesda, MD, USA.
| |
Collapse
|
49
|
Yang J, Zhou W, Li D, Niu T, Wang W. BCMA-targeting chimeric antigen receptor T-cell therapy for multiple myeloma. Cancer Lett 2023; 553:215949. [PMID: 36216149 DOI: 10.1016/j.canlet.2022.215949] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2022] [Revised: 09/29/2022] [Accepted: 10/04/2022] [Indexed: 11/24/2022]
Abstract
Multiple myeloma (MM) remains an incurable hematologic malignancy, despite the development of numerous innovative therapies during the past two decades. Immunotherapies are changing the treatment paradigm of MM and have improved the overall response and survival of patients with relapsed/refractory (RR) MM. B cell maturation antigen (BCMA), selectively expressed in normal and malignant plasma cells, has been targeted by several immunotherapeutic modalities. Chimeric antigen receptor (CAR) T cells, the breakthrough in cancer immunotherapy, have revolutionized the treatment of B cell malignancies and remarkably improved the prognosis of RRMM. BCMA-targeting CAR T cell therapy is the most developed CAR T cell therapy for MM, and the US Food and Drug Administration has already approved idecabtagene vicleucel (Ide-cel) and ciltacabtagene autoleucel (Cilta-cel) for MM. However, the development of novel BCMA-targeting CAR T cell therapies remains in progress. This review focuses on BCMA-targeting CAR T cell therapy, covering all stages of investigational progress, including the innovative preclinical studies, the initial phase I clinical trials, and the more developed phase II clinical trials. It also discusses possible measures to improve the efficacy and safety of this therapy.
Collapse
Affiliation(s)
- Jinrong Yang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Weilin Zhou
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Dan Li
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China
| | - Ting Niu
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| | - Wei Wang
- Department of Hematology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and National Collaborative Innovation Center, Chengdu, 610041, China.
| |
Collapse
|
50
|
Zhang M, Gray F, Cushman I, Wurmser A, Chan H, Couto S, Wang M, Nakayama Y, Hagner P, Al-Masri H, Williams S, Hersey S. A Novel BCMA Immunohistochemistry Assay Reveals a Heterogenous and Dynamic BCMA Expression Profile in Multiple Myeloma. Mod Pathol 2023; 36:100050. [PMID: 36788077 DOI: 10.1016/j.modpat.2022.100050] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Revised: 03/22/2022] [Accepted: 11/08/2022] [Indexed: 01/13/2023]
Abstract
B-cell maturation antigen (BCMA) is a promising target for the treatment of multiple myeloma (MM) because the expression of this protein is largely limited to B-cell sets, plasma cells, MM, and other B-cell malignancies. Early studies assessing BCMA protein expression and localization have used insufficiently qualified immunohistochemistry assays, which have reported broad ranges of BCMA expression. As a result, our understanding of BCMA tissue expression derived from these data is limited, specifically the prevalence of BCMA expression on the cell surface/membrane, which has mechanistic relevance to the antimyeloma activity of several novel biotherapeutics. Here, we report on the qualification and application of a novel anti-BCMA immunohistochemistry antibody, 805G12. This antibody shows robust detection of BCMA in formalin-fixed, decalcified bone marrow tissue and provides key insights into membrane BCMA expression. The clone 805G12, which was raised against an intracellular C-terminal domain peptide of membrane BCMA, exhibited increased sensitivity and superior specificity across healthy and diseased tissue compared with the frequently referenced commercial reagent AF193. The new clone also demonstrated a broad range of expression of BCMA in MM and diffuse large B-cell lymphoma specimens. Additionally, cross-reactivity with closely related tumor necrosis factor receptor family members was observed with AF193 but not with 805G12. Furthermore, via established 805G12 and other independent BCMA assays, it was concluded that proteolytic processing by γ-secretase contributes to the levels of BCMA localized to the plasma membrane. As BCMA-directed therapeutics emerge to address the need for more effective treatment in the relapsed or refractory MM disease setting, the implementation of a qualified assay would ensure that reliable and consistent data on BCMA surface expression are used to inform clinical trial decisions and patient responses.
Collapse
Affiliation(s)
| | - Falon Gray
- Bristol Myers Squibb, Princeton, New Jersey.
| | | | | | - Henry Chan
- Bristol Myers Squibb, Princeton, New Jersey
| | - Suzana Couto
- Formerly Celgene Corporation, a Bristol Myers Squibb Company, Princeton, New Jersey
| | - Maria Wang
- Bristol Myers Squibb, Princeton, New Jersey
| | | | | | | | | | | |
Collapse
|