1
|
Kushwaha N, Panjwani D, Patel S, Ahlawat P, Yadav MR, Patel AS. Emerging advances in nano-biomaterial assisted amyloid beta chimeric antigen receptor macrophages (CAR-M) therapy: reducing plaque burden in Alzheimer's disease. J Drug Target 2024:1-21. [PMID: 39403775 DOI: 10.1080/1061186x.2024.2417012] [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: 08/04/2024] [Revised: 09/23/2024] [Accepted: 10/08/2024] [Indexed: 10/22/2024]
Abstract
Alzheimer's disease is the most common form, accounting for 60-70% of 55 million dementia cases. Even though the precise pathophysiology of AD is not completely understood, clinical trials focused on antibodies targeting aggregated forms of β amyloid (Aβ) have demonstrated that reducing amyloid plaques can arrest cognitive decline in patients in the early stages of AD. In this study, we provide an overview of current research and innovations for controlled release from nano-biomaterial-assisted chimeric antigen receptor macrophage (CAR-M) therapeutic strategies targeted at AD. Nano-bio materials, such as iron-oxide nanoparticles (IONPs), can be made selectively (Hp-Hb/mannose) to bind and take up Aβ plaques like CAR-M cells. By using nano-bio materials, both the delivery and stability of CAR-M cells in brain tissue can be improved to overcome the barriers of the BBB and enhance therapeutic effects. By enhancing the targeting capabilities and stability of CAR-M cells, mRNA-loaded nano-biomaterials can significantly improve the efficacy of immunotherapy for plaque reduction in AD. This novel strategy holds promise for translating preclinical successes into clinical applications, potentially revolutionising the management of AD.
Collapse
Affiliation(s)
- Nishabh Kushwaha
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Drishti Panjwani
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Shruti Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Priyanka Ahlawat
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| | - Mange Ram Yadav
- Research and Development Cell, Parul University, Vadodara, India
| | - Asha S Patel
- Department of Pharmaceutics, Parul Institute of Pharmacy, Parul University, Vadodara, India
| |
Collapse
|
2
|
Butler SE, Ackerman ME. Challenges and future perspectives for high-throughput chimeric antigen receptor T cell discovery. Curr Opin Biotechnol 2024; 90:103216. [PMID: 39437676 DOI: 10.1016/j.copbio.2024.103216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 08/09/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024]
Abstract
Novel chimeric antigen receptor (CAR) T cell designs are being developed to overcome challenges with tumor recognition, trafficking, on-target but off-tumor binding, cytotoxicity, persistence, and immune suppression within the tumor microenvironment. Whereas traditional CAR engineering is an iterative, hypothesis-driven process in which novel designs are rationally constructed and tested for in vivo efficacy, drawing from the fields of small-molecule and protein-based therapeutic discovery, we consider how high-throughput, functional screening technologies are beginning to be applied for the development of promising CAR candidates. We review how the development of high-throughput screening methods has the potential to streamline the CAR discovery process, ultimately improving efficiency and clinical efficacy.
Collapse
Affiliation(s)
- Savannah E Butler
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
| | - Margaret E Ackerman
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA; Thayer School of Engineering, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
3
|
Cheever A, Kang CC, O’Neill KL, Weber KS. Application of novel CAR technologies to improve treatment of autoimmune disease. Front Immunol 2024; 15:1465191. [PMID: 39445021 PMCID: PMC11496059 DOI: 10.3389/fimmu.2024.1465191] [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: 07/15/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has become an important treatment for hematological cancers, and its success has spurred research into CAR T cell therapies for other diseases, including solid tumor cancers and autoimmune diseases. Notably, the development of CAR-based treatments for autoimmune diseases has shown great progress recently. Clinical trials for anti-CD19 and anti-BCMA CAR T cells in treating severe B cell-mediated autoimmune diseases, like systemic lupus erythematosus (SLE), have shown lasting remission thus far. CAR T cells targeting autoreactive T cells are beginning clinical trials for treating T cell mediated autoimmune diseases. Chimeric autoantigen receptor (CAAR) T cells specifically target and eliminate only autoreactive B cells, and they have shown promise in treating mucosal pemphigus vulgaris and MuSK myasthenia gravis. Regulatory CAR T cells have also been developed, which show potential in altering autoimmune affected areas by creating a protective barrier as well as helping decrease inflammation. These new treatments are only the beginning of potential CAR T cell applications in treating autoimmune disease. Novel CAR technologies have been developed that increase the safety, potency, specificity, and efficacy of CAR T cell therapy. Applying these novel modifications to autoimmune CARs has the potential to enhance the efficacy and applicability of CAR therapies to autoimmune disease. This review will detail several recently developed CAR technologies and discuss how their application to autoimmune disease will improve this emerging field. These include logic-gated CARs, soluble protein-secreting CARs, and modular CARs that enable CAR T cell therapies to be more specific, reach a wider span of target cells, be safer for patients, and give a more potent cytotoxic response. Applying these novel CAR technologies to the treatment of autoimmune diseases has the potential to revolutionize this growing application of CAR T cell therapies.
Collapse
|
4
|
Qi C, Liu D, Liu C, Wei X, Ma M, Lu X, Tao M, Zhang C, Wang X, He T, Li J, Dai F, Ding Y, Shen L. Antigen-independent activation is critical for the durable antitumor effect of GUCY2C-targeted CAR-T cells. J Immunother Cancer 2024; 12:e009960. [PMID: 39366753 PMCID: PMC11459315 DOI: 10.1136/jitc-2024-009960] [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: 09/16/2024] [Indexed: 10/06/2024] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-T cells face many obstacles in solid tumor therapy, including heterogeneous antigen expression and inefficient T cell persistence. Guanylyl cyclase C (GUCY2C) has been identified as a suitable tumor antigen for targeted therapy due to its intestinal-restricted expression pattern in normal tissues and steady overexpression in gastrointestinal tumors, especially colorectal cancer. An antigen-sensitive and long-lasting CAR-T cell targeting GUCY2C was investigated in this study. METHODS Using constructed tumor cell lines with various GUCY2C expression densities, we screened out an antigen-sensitive single chain variable fragment (scFv) that enabled CAR-T cells to efficiently eradicate the GUCY2C lowly expressed tumor cells. CAR-T cells with different compositions of the hinge, transmembrane and costimulatory domains were also constructed for selection of the long-lasting CAR-T format with durable antitumor efficacy in vitro and in tumor-bearing mice. The underlying mechanism was further investigated based on mutation of the hinge and transmembrane domains. RESULTS We found that the composition of the antigen-sensitive scFv, CD8α hinge, CD8α transmembrane, and CD28 costimulatory domains boosted CAR-T cells to rapidly kill tumors, maintain high expansion capacity, and long-term efficacy in various colorectal cancer models. The durable antitumor function was attributed to the optimal CAR tonic signaling that conferred CAR-T cells with autonomous activation, proliferation, survival and cytokine release in the absence of antigen stimulation. The tonic signaling was associated with the length and the cysteine residues in the CD8α hinge and transmembrane domains. CONCLUSIONS This study demonstrated a potent GUCY2C-targeted CAR-T cell for gastrointestinal tumor therapy and highlights the importance of adequate tonic signaling for effective CAR-T cell therapy against solid tumors.
Collapse
Affiliation(s)
- Changsong Qi
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Early Drug Development Centre, Peking University Cancer Hospital, Beijing, China
| | - Dongqun Liu
- Beijing Imunopharm Technology Co Ltd, Beijing, China
| | - Chang Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Early Drug Development Centre, Peking University Cancer Hospital, Beijing, China
| | - Xiaofei Wei
- Beijing Imunopharm Technology Co Ltd, Beijing, China
| | - Mingyang Ma
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Xinan Lu
- Beijing Imunopharm Technology Co Ltd, Beijing, China
| | - Min Tao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Cheng Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Xicheng Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Ting He
- Beijing Imunopharm Technology Co Ltd, Beijing, China
| | - Jian Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| | - Fei Dai
- Beijing Imunopharm Technology Co Ltd, Beijing, China
| | - Yanping Ding
- Beijing Imunopharm Technology Co Ltd, Beijing, China
| | - Lin Shen
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital, Beijing, China
| |
Collapse
|
5
|
Brown MP. CAR spacers: Not just filling space. Mol Ther 2024; 32:3207-3208. [PMID: 39293429 PMCID: PMC11489524 DOI: 10.1016/j.ymthe.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Revised: 09/04/2024] [Accepted: 09/04/2024] [Indexed: 09/20/2024] Open
Affiliation(s)
- Michael P Brown
- Translational Oncology Laboratory, Centre for Cancer Biology, SA Pathology and University of South Australia, Adelaide, SA 5000, Australia; RAH Cancer Clinical Trials Unit, Royal Adelaide Hospital, Adelaide, SA 5000, Australia; Adelaide Medical School, The University of Adelaide, Adelaide, SA 5005, Australia.
| |
Collapse
|
6
|
Kua L, Ng CH, Tan JW, Tan HC, Seh CC, Wong F, Ong R, Rooney CM, Tan J, Chen Q, Horak ID, Tan KW, Low L. Novel OX40 and 4-1BB derived spacers enhance CD30 CAR activity and safety in CD30 positive lymphoma models. Mol Ther 2024; 32:3504-3521. [PMID: 38946142 PMCID: PMC11489532 DOI: 10.1016/j.ymthe.2024.06.037] [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: 06/22/2023] [Revised: 04/26/2024] [Accepted: 06/28/2024] [Indexed: 07/02/2024] Open
Abstract
The chimeric antigen receptor (CAR) derived from the CD30 specific murine antibody, HRS-3, has produced promising clinical efficacy with a favorable safety profile in the treatment of relapsed or refractory CD30-positive lymphomas. However, persistence of the autologous CAR-T cells was brief, and many patients relapsed a year after treatment. The lack of persistence may be attributed to the use of a wild-type immunoglobulin (Ig)G1 spacer that can associate with Fc receptors. We first identified the cysteine-rich domain (CRD) 5 of CD30 as the primary binding epitope of HRS-3 and armed with this insight, attempted to improve the HRS-3 CAR functionality with a panel of novel spacer designs. We demonstrate that HRS-3 CARs with OX40 and 4-1BB derived spacers exhibited similar anti-tumor efficacy, circumvented interactions with Fc receptors, and secreted lower levels of cytokines in vitro than a CAR employing the IgG1 spacer. Humanization of the HRS-3 scFv coupled with the 4-1BB spacer preserved potent on-target, on-tumor efficacy, and on-target, off-tumor safety. In a lymphoma mouse model of high tumor burden, T cells expressing humanized HRS-3 CD30.CARs with the 4-1BB spacer potently killed tumors with low levels of circulating inflammatory cytokines, providing a promising candidate for future clinical development in the treatment of CD30-positive malignancies.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Cell Line, Tumor
- Disease Models, Animal
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Ki-1 Antigen/immunology
- Ki-1 Antigen/metabolism
- Lymphoma/therapy
- Lymphoma/immunology
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, OX40/metabolism
- Receptors, OX40/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/metabolism
- Tumor Necrosis Factor Receptor Superfamily, Member 9/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Lindsay Kua
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Chee Hoe Ng
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Jin Wei Tan
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | | | | | - Fiona Wong
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Richard Ong
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Cliona M Rooney
- Center for Cell and Gene Therapy, Texas Children's Hospital, Houston Methodist Hospital, and Baylor College of Medicine, Houston, TX 77030, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX 77030, USA; Department of Pediatrics, Baylor College of Medicine, Houston, TX 77030, USA; Texas Children's Cancer and Hematology Centers, Baylor College of Medicine, Houston, TX 77030, USA; Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX 77030, USA
| | - Joel Tan
- Institute for Molecular and Cellular Biology, A∗STAR Singapore 138673, Singapore
| | - Qingfeng Chen
- Institute for Molecular and Cellular Biology, A∗STAR Singapore 138673, Singapore
| | - Ivan D Horak
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Kar Wai Tan
- Tessa Therapeutics Ltd, Singapore 138673, Singapore
| | - Lionel Low
- Tessa Therapeutics Ltd, Singapore 138673, Singapore.
| |
Collapse
|
7
|
Pe KCS, Jewmoung S, Rad SAH, Chantarat N, Chanswangphuwana C, Tashiro H, Suppipat K, Tawinwung S. Optimization of anti-TIM3 chimeric antigen receptor with CD8α spacer and TNFR-based costimulation for enhanced efficacy in AML therapy. Biomed Pharmacother 2024; 179:117388. [PMID: 39243430 DOI: 10.1016/j.biopha.2024.117388] [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: 07/04/2024] [Revised: 08/26/2024] [Accepted: 08/30/2024] [Indexed: 09/09/2024] Open
Abstract
CAR T cell therapy for AML remains limited due to the lack of a proper target without on-target off-tumor toxicity. TIM3 is a promising target due to its high expression on AML cells and absence in most normal hematopoietic cells. Previous reports have shown that each CAR component impacts CAR functionality. Here, we optimized TIM-3 targeting CAR T cells for AML therapy. We generated CARs targeting TIM3 with two different non-signaling domains: an IgG2-CH3 spacer with CD28 transmembrane domain (CH3/CD28) and a CD8α spacer with CD8α transmembrane domain (CD8/CD8), and evaluated their characteristics and function. Incorporating the non-signaling CH3/CD28 domain resulted in unstable CAR expression in anti-TIM3 CAR T cells, leading to lower surface CAR expression over time and reduced cytotoxic function compared to anti-TIM3 CARs with the CD8/CD8 domain. Both types of anti-TIM3 CAR T cells transiently exhibited fratricide, which subsided overtime, and both CAR T cells achieved substantial T cell expansion. To further optimize the design, we explored the effects of different costimulatory domains. Compared with CD28 costimulation, 4-1BB and CD27 combined with a CD8/CD8 non-signaling domain showed higher cytokine secretion, superior antitumor activity, and enhanced T-cell persistence after repeated antigen exposure. These findings emphasize the impact of the optimal design of CAR constructs that provide efficient function. In the context of anti-TIM3 CAR T cells, using a CD8α spacer and transmembrane domain with TNFR-based costimulation is a promising CAR design to improve anti-TIM3 CAR T cell function for AML therapy.
Collapse
MESH Headings
- Humans
- Receptors, Chimeric Antigen/immunology
- Leukemia, Myeloid, Acute/therapy
- Leukemia, Myeloid, Acute/immunology
- Animals
- Hepatitis A Virus Cellular Receptor 2/metabolism
- Immunotherapy, Adoptive/methods
- CD8 Antigens/metabolism
- CD8 Antigens/immunology
- Cell Line, Tumor
- Mice
- CD28 Antigens/immunology
- CD28 Antigens/metabolism
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor/metabolism
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Kristine Cate S Pe
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirirut Jewmoung
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | | | - Natthida Chantarat
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Chantiya Chanswangphuwana
- Division of Hematology, Department of Medicine, King Chulalongkorn Memorial Hospital, Bangkok, Thailand
| | - Haruko Tashiro
- Department of Hematology/Oncology, Teikyo University School of Medicine, Tokyo, Japan
| | - Koramit Suppipat
- Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand; Department of Research Affair, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand; Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Bangkok, Thailand
| | - Supannikar Tawinwung
- Department of Pharmacology and Physiology, Faculty of Pharmaceutical Sciences, Chulalongkorn University, Bangkok, Thailand; Cellular Immunotherapy Research Unit, Chulalongkorn University, Bangkok, Thailand; Thailand Hub of Talents in Cancer Immunotherapy (TTCI), Bangkok, Thailand.
| |
Collapse
|
8
|
Sierro-Martínez B, Escamilla-Gómez V, Pérez-Ortega L, Guijarro-Albaladejo B, Hernández-Díaz P, de la Rosa-Garrido M, Lara-Chica M, Rodríguez-Gil A, Reguera-Ortega JL, Sanoja-Flores L, Arribas-Arribas B, Montiel-Aguilera MÁ, Carmona G, Robles MJ, Caballero-Velázquez T, Briones J, Einsele H, Hudecek M, Pérez-Simón JA, García-Guerrero E. Next-generation BCMA-targeted chimeric antigen receptor CARTemis-1: the impact of manufacturing procedure on CAR T-cell features. Cell Oncol (Dordr) 2024:10.1007/s13402-024-00984-0. [PMID: 39192092 DOI: 10.1007/s13402-024-00984-0] [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] [Accepted: 08/09/2024] [Indexed: 08/29/2024] Open
Abstract
PURPOSE CAR therapy targeting BCMA is under investigation as treatment for multiple myeloma. However, given the lack of plateau in most studies, pursuing more effective alternatives is imperative. We present the preclinical and clinical validation of a new optimized anti-BCMA CAR (CARTemis-1). In addition, we explored how the manufacturing process could impact CAR-T cell product quality and fitness. METHODS CARTemis-1 optimizations were evaluated at the preclinical level both, in vitro and in vivo. CARTemis-1 generation was validated under GMP conditions, studying the dynamics of the immunophenotype from leukapheresis to final product. Here, we studied the impact of the manufacturing process on CAR-T cells to define optimal cell culture protocol and expansion time to increase product fitness. RESULTS Two different versions of CARTemis-1 with different spacers were compared. The longer version showed increased cytotoxicity. The incorporation of the safety-gene EGFRt into the CARTemis-1 structure can be used as a monitoring marker. CARTemis-1 showed no inhibition by soluble BCMA and presents potent antitumor effects both in vitro and in vivo. Expansion with IL-2 or IL-7/IL-15 was compared, revealing greater proliferation, less differentiation, and less exhaustion with IL-7/IL-15. Three consecutive batches of CARTemis-1 were produced under GMP guidelines meeting all the required specifications. CARTemis-1 cells manufactured under GMP conditions showed increased memory subpopulations, reduced exhaustion markers and selective antitumor efficacy against MM cell lines and primary myeloma cells. The optimal release time points for obtaining the best fit product were > 6 and < 10 days (days 8-10). CONCLUSIONS CARTemis-1 has been rationally designed to increase antitumor efficacy, overcome sBCMA inhibition, and incorporate the expression of a safety-gene. The generation of CARTemis-1 was successfully validated under GMP standards. A phase I/II clinical trial for patients with multiple myeloma will be conducted (EuCT number 2022-503063-15-00).
Collapse
Affiliation(s)
- Belén Sierro-Martínez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Virginia Escamilla-Gómez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Laura Pérez-Ortega
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Beatriz Guijarro-Albaladejo
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Paola Hernández-Díaz
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - María de la Rosa-Garrido
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Maribel Lara-Chica
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Alfonso Rodríguez-Gil
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Juan Luis Reguera-Ortega
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Luzalba Sanoja-Flores
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Blanca Arribas-Arribas
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC)-Planta CTTC Campus Virgen del Rocío de Sevilla, Red Andaluza de diseño y traslación de Terapias Avanzadas, Seville, Spain
- Programa doctorado Tecnología y Ciencias del Medicamento, Facultad de Farmacia, Universidad de Sevilla, Seville, Spain
| | - Miguel Ángel Montiel-Aguilera
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC)-Planta CTTC Campus Virgen del Rocío de Sevilla, Red Andaluza de diseño y traslación de Terapias Avanzadas, Seville, Spain
| | - Gloria Carmona
- Unidad de Producción y Reprogramación Celular de Sevilla (UPRC)-Planta CTTC Campus Virgen del Rocío de Sevilla, Red Andaluza de diseño y traslación de Terapias Avanzadas, Seville, Spain
| | - Maria Jose Robles
- Unidad de Patología Comparada, Biobanco Virgen del Rocío-IBiS, Unidad de Gestión Clínica de Anatomía Patológica, Hospital Universitario Virgen del Rocío, Sevilla, Spain
| | - Teresa Caballero-Velázquez
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain
| | - Javier Briones
- Servicio de Hematología, Instituto de Investigación Biomédica Sant Pau (IIB-Sant Pau), Barcelona, Spain
| | - Hermann Einsele
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II and Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Michael Hudecek
- Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II and Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, Würzburg, Germany
| | - Jose Antonio Pérez-Simón
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
| | - Estefanía García-Guerrero
- Instituto de Biomedicina de Sevilla, IBiS/Hospital Universitario Virgen del Rocío/CSIC/Universidad de Sevilla, Sevilla, Spain.
| |
Collapse
|
9
|
Hamadeh IS, Friend R, Mailankody S, Atrash S. Chimeric antigen receptor T-cells: a review on current status and future directions for relapsed/refractory multiple myeloma. Front Oncol 2024; 14:1455464. [PMID: 39175472 PMCID: PMC11338754 DOI: 10.3389/fonc.2024.1455464] [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: 06/26/2024] [Accepted: 07/25/2024] [Indexed: 08/24/2024] Open
Abstract
Although multiple myeloma is an incurable disease, the past decade has witnessed significant improvement in patient outcomes. This was brought about by the development of T-cell redirection therapies such as chimeric antigen receptor (CAR) T-cells, which can leverage the natural ability of the immune system to fight myeloma cells. The approval of the B-cell maturation antigen (BCMA)-directed CAR T, idecabtagene vicleucel (ide-cel), and ciltacabtagene autoleucel (cilta-cel) has resulted in a paradigm shift in the treatment of relapsed/refractory multiple myeloma. Overall response rates ranging from 73 to 97% are currently achievable. However, the limitations of KarMMa-1 and CARTITUDE-1 studies spurred the generation of real-world data to provide some insights into the effectiveness of ide-cel and cilta-cel among patients who were excluded from clinical trials, particularly those who received prior BCMA-targeted or other T-cell redirection therapies. Despite their unprecedented clinical efficacy in heavily pretreated patients, responses to CAR T remain non-durable. Although the underlying mechanisms of resistance to these agents haven't been fully elucidated, studies have suggested that resistance patterns could be multifaceted, implicating T-cell exhaustion and tumor intrinsic mechanisms such as BCMA target loss, upregulation of gamma-secretase, and others. Herein, we provide a succinct overview of the development of CAR T-cells, manufacturing process, and associated toxicities/complications. In this review, we also recapitulate the existing literature pertaining MM CAR-T as well as emerging data from some of the ongoing clinical trials designed to mitigate the shortcomings of these agents, and improve the clinical efficacy of CAR T, especially in the relapsed/refractory setting.
Collapse
Affiliation(s)
- Issam S. Hamadeh
- Clinical Pharmacy Services, Pharmacy Department, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Reed Friend
- Plasma Cell Disorders Division, Department of Hematologic Oncology & Blood Disorders Levine Cancer Institute, Atrium Health, Charlotte, NC, United States
| | - Sham Mailankody
- Myeloma Service, Division of Hematologic Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, United States
| | - Shebli Atrash
- Plasma Cell Disorders Division, Department of Hematologic Oncology & Blood Disorders Levine Cancer Institute, Atrium Health, Charlotte, NC, United States
| |
Collapse
|
10
|
Xiong Y, Libby KA, Su X. The physical landscape of CAR-T synapse. Biophys J 2024; 123:2199-2210. [PMID: 37715447 PMCID: PMC11331049 DOI: 10.1016/j.bpj.2023.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 08/30/2023] [Accepted: 09/12/2023] [Indexed: 09/17/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells form dynamic immunological synapses with their cancer cell targets. After a CAR-antigen engagement, the CAR-T synapse forms, matures, and finally disassembles, accompanied by substantial remodeling of cell surface proteins, lipids, and glycans. In this review, we provide perspectives for understanding protein distribution, membrane topology, and force transmission across the CAR-T synapse. We highlight the features of CAR-T synapses that differ from T cell receptor synapses, including the disorganized protein pattern, adjustable synapse width, diverse mechano-responding properties, and resulting signaling consequences. Through a range of examples, we illustrate how revealing the biophysical nature of the CAR-T synapse could guide the design of CAR-Ts with improved anti-tumor function.
Collapse
Affiliation(s)
- Yiwei Xiong
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut
| | - Kendra A Libby
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut; Department of Biology, Massachusetts Institute of Technology, Cambridge, Massachusetts; Ragon Institute of MGH, MIT and Harvard, Cambridge, Massachusetts
| | - Xiaolei Su
- Department of Cell Biology, Yale School of Medicine, New Haven, Connecticut; Yale Cancer Center, Yale University, New Haven, Connecticut; Yale Stem Cell Center, Yale University, New Haven, Connecticut.
| |
Collapse
|
11
|
Ramapriyan R, Vykunta VS, Vandecandelaere G, Richardson LGK, Sun J, Curry WT, Choi BD. Altered cancer metabolism and implications for next-generation CAR T-cell therapies. Pharmacol Ther 2024; 259:108667. [PMID: 38763321 DOI: 10.1016/j.pharmthera.2024.108667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2024] [Revised: 04/30/2024] [Accepted: 05/14/2024] [Indexed: 05/21/2024]
Abstract
This review critically examines the evolving landscape of chimeric antigen receptor (CAR) T-cell therapy in treating solid tumors, with a particular focus on the metabolic challenges within the tumor microenvironment. CAR T-cell therapy has demonstrated remarkable success in hematologic malignancies, yet its efficacy in solid tumors remains limited. A significant barrier is the hostile milieu of the tumor microenvironment, which impairs CAR T-cell survival and function. This review delves into the metabolic adaptations of cancer cells and their impact on immune cells, highlighting the competition for nutrients and the accumulation of immunosuppressive metabolites. It also explores emerging strategies to enhance CAR T-cell metabolic fitness and persistence, including genetic engineering and metabolic reprogramming. An integrated approach, combining metabolic interventions with CAR T-cell therapy, has the potential to overcome these constraints and improve therapeutic outcomes in solid tumors.
Collapse
Affiliation(s)
- Rishab Ramapriyan
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| | - Vivasvan S Vykunta
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94143, USA; ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA; Medical Scientist Training Program, School of Medicine, University of California, San Francisco, San Francisco, CA 94143, USA; Biomedical Sciences Graduate Program, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Gust Vandecandelaere
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leland G K Richardson
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jing Sun
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - William T Curry
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Bryan D Choi
- Brain Tumor Immunotherapy Laboratory, Department of Neurosurgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
12
|
Kennewick KT, Yamaguchi Y, Gibson J, Gerdts EA, Jeang B, Tilakawardane D, Murad JP, Chang WC, Wright SL, Thiel MS, Forman SJ, Stern LA, Priceman SJ. Nonsignaling extracellular spacer regulates tumor antigen selectivity of CAR T cells. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200789. [PMID: 38939825 PMCID: PMC11208731 DOI: 10.1016/j.omton.2024.200789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/13/2024] [Accepted: 02/27/2024] [Indexed: 06/29/2024]
Abstract
Advancing chimeric antigen receptor (CAR)-engineered T cells for the treatment of solid tumors is a major focus in the field of cellular immunotherapy. Several hurdles have hindered similar CAR T cell clinical responses in solid tumors as seen in hematological malignancies. These challenges include on-target off-tumor toxicities, which have inspired efforts to optimize CARs for improved tumor antigen selectivity and overall safety. We recently developed a CAR T cell therapy targeting prostate stem cell antigen (PSCA) for prostate and pancreatic cancers, showing improved preclinical antitumor activity and T cell persistence by optimizing the intracellular co-stimulatory domain. Similar studies were undertaken to optimize HER2-directed CAR T cells with modifications to the intracellular co-stimulatory domain for selective targeting of breast cancer brain metastasis. In the present study, we evaluate various nonsignaling extracellular spacers in these CARs to further improve tumor antigen selectivity. Our findings suggest that length and structure of the extracellular spacer can dictate the ability of CARs to selectively target tumor cells with high antigen density, while sparing cells with low antigen density. This study contributes to CAR construct design considerations and expands our knowledge of tuning solid tumor CAR T cell therapies for improved safety and efficacy.
Collapse
Affiliation(s)
- Kelly T. Kennewick
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Yukiko Yamaguchi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Jackson Gibson
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Ethan A. Gerdts
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Brook Jeang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Dileshni Tilakawardane
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - John P. Murad
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Wen-Chung Chang
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Sarah L. Wright
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Michalina S. Thiel
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
| | - Stephen J. Forman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| | - Lawrence A. Stern
- Department of Chemical, Biological, and Materials Engineering, University of South Florida, Tampa, FL 33620, USA
| | - Saul J. Priceman
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope, Duarte, CA 91010, USA
- Department of Immuno-Oncology, Beckman Research Institute of City of Hope, Duarte, CA 91010, USA
| |
Collapse
|
13
|
Utkarsh K, Srivastava N, Kumar S, Khan A, Dagar G, Kumar M, Singh M, Haque S. CAR-T cell therapy: a game-changer in cancer treatment and beyond. Clin Transl Oncol 2024; 26:1300-1318. [PMID: 38244129 DOI: 10.1007/s12094-023-03368-2] [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: 09/09/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024]
Abstract
In recent years, cancer has become one of the primary causes of mortality, approximately 10 million deaths worldwide each year. The most advanced, chimeric antigen receptor (CAR) T cell immunotherapy has turned out as a promising treatment for cancer. CAR-T cell therapy involves the genetic modification of T cells obtained from the patient's blood, and infusion back to the patients. CAR-T cell immunotherapy has led to a significant improvement in the remission rates of hematological cancers. CAR-T cell therapy presently limited to hematological cancers, there are ongoing efforts to develop additional CAR constructs such as bispecific CAR, tandem CAR, inhibitory CAR, combined antigens, CRISPR gene-editing, and nanoparticle delivery. With these advancements, CAR-T cell therapy holds promise concerning potential to improve upon traditional cancer treatments such as chemotherapy and radiation while reducing associated toxicities. This review covers recent advances and advantages of CAR-T cell immunotherapy.
Collapse
Affiliation(s)
- Kumar Utkarsh
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Namita Srivastava
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sachin Kumar
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Azhar Khan
- Faculty of Applied Science and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Gunjan Dagar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Mukesh Kumar
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mayank Singh
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Shabirul Haque
- Department of Autoimmune Diseases, Feinstein Institute for Medical Research, Northwell Health, 350, Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
14
|
Choudhery MS, Arif T, Mahmood R, Harris DT. CAR-T-Cell-Based Cancer Immunotherapies: Potentials, Limitations, and Future Prospects. J Clin Med 2024; 13:3202. [PMID: 38892913 PMCID: PMC11172642 DOI: 10.3390/jcm13113202] [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: 03/14/2024] [Revised: 05/17/2024] [Accepted: 05/23/2024] [Indexed: 06/21/2024] Open
Abstract
Cancer encompasses various elements occurring at the cellular and genetic levels, necessitating an immunotherapy capable of efficiently addressing both aspects. T cells can combat cancer cells by specifically recognizing antigens on them. This innate capability of T cells has been used to develop cellular immunotherapies, but most of them can only target antigens through major histocompatibility complexes (MHCs). New gene-editing techniques such as clustered regularly interspaced short palindromic repeat (CRISPR)-associated protein 9 (CRISPR-cas9) can precisely edit the DNA sequences. CRISPR-cas9 has made it possible to generate genetically engineered chimeric antigen receptors (CARs) that can overcome the problems associated with old immunotherapies. In chimeric antigen receptor T (CAR-T) cell therapy, the patient's T cells are isolated and genetically modified to exhibit synthetic CAR(s). CAR-T cell treatment has shown remarkably positive clinical outcomes in cancers of various types. Nevertheless, there are various challenges that reduce CAR-T effectiveness in solid tumors. It is required to address these challenges in order to make CAR-T cell therapy a better and safer option. Combining CAR-T treatment with other immunotherapies that target multiple antigens has shown positive outcomes. Moreover, recently generated Boolean logic-gated advanced CARs along with artificial intelligence has expanded its potential to treat solid tumors in addition to blood cancers. This review aims to describe the structure, types, and various methods used to develop CAR-T cells. The clinical applications of CAR-T cells in hematological malignancies and solid tumours have been described in detail. In addition, this discussion has addressed the limitations associated with CAR-T cells, explored potential strategies to mitigate CAR-T-related toxicities, and delved into future perspectives.
Collapse
Affiliation(s)
- Mahmood S. Choudhery
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 54600, Pakistan;
| | - Taqdees Arif
- Department of Human Genetics & Molecular Biology, University of Health Sciences, Lahore 54600, Pakistan;
| | - Ruhma Mahmood
- Jinnah Hospital, Allama Iqbal Medical College, Lahore 54700, Pakistan;
| | - David T. Harris
- Department of Immunobiology, College of Medicine, University of Arizona Health Sciences Biorepository, The University of Arizona, Tucson, AZ 85724-5221, USA;
| |
Collapse
|
15
|
Satapathy BP, Sheoran P, Yadav R, Chettri D, Sonowal D, Dash CP, Dhaka P, Uttam V, Yadav R, Jain M, Jain A. The synergistic immunotherapeutic impact of engineered CAR-T cells with PD-1 blockade in lymphomas and solid tumors: a systematic review. Front Immunol 2024; 15:1389971. [PMID: 38799440 PMCID: PMC11116574 DOI: 10.3389/fimmu.2024.1389971] [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: 02/22/2024] [Accepted: 04/11/2024] [Indexed: 05/29/2024] Open
Abstract
Currently, therapies such as chimeric antigen receptor-T Cell (CAR-T) and immune checkpoint inhibitors like programmed cell death protein-1 (PD-1) blockers are showing promising results for numerous cancer patients. However, significant advancements are required before CAR-T therapies become readily available as off-the-shelf treatments, particularly for solid tumors and lymphomas. In this review, we have systematically analyzed the combination therapy involving engineered CAR-T cells and anti PD-1 agents. This approach aims at overcoming the limitations of current treatments and offers potential advantages such as enhanced tumor inhibition, alleviated T-cell exhaustion, heightened T-cell activation, and minimized toxicity. The integration of CAR-T therapy, which targets tumor-associated antigens, with PD-1 blockade augments T-cell function and mitigates immune suppression within the tumor microenvironment. To assess the impact of combination therapy on various tumors and lymphomas, we categorized them based on six major tumor-associated antigens: mesothelin, disialoganglioside GD-2, CD-19, CD-22, CD-133, and CD-30, which are present in different tumor types. We evaluated the efficacy, complete and partial responses, and progression-free survival in both pre-clinical and clinical models. Additionally, we discussed potential implications, including the feasibility of combination immunotherapies, emphasizing the importance of ongoing research to optimize treatment strategies and improve outcomes for cancer patients. Overall, we believe combining CAR-T therapy with PD-1 blockade holds promise for the next generation of cancer immunotherapy.
Collapse
Affiliation(s)
- Bibhu Prasad Satapathy
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Pooja Sheoran
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Rohit Yadav
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Dewan Chettri
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Dhruba Sonowal
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Chinmayee Priyadarsini Dash
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Prachi Dhaka
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Vivek Uttam
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Ritu Yadav
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| | - Manju Jain
- Department of Biochemistry, School of Basic Sciences, Central University of Punjab, Bathinda, Punjab, India
| | - Aklank Jain
- Department of Zoology, Non-Coding RNA and Cancer Biology Laboratory, Central University of Punjab, Bathinda, Punjab, India
| |
Collapse
|
16
|
Jilani S, Saco JD, Mugarza E, Pujol-Morcillo A, Chokry J, Ng C, Abril-Rodriguez G, Berger-Manerio D, Pant A, Hu J, Gupta R, Vega-Crespo A, Baselga-Carretero I, Chen JM, Shin DS, Scumpia P, Radu RA, Chen Y, Ribas A, Puig-Saus C. CAR-T cell therapy targeting surface expression of TYRP1 to treat cutaneous and rare melanoma subtypes. Nat Commun 2024; 15:1244. [PMID: 38336975 PMCID: PMC10858182 DOI: 10.1038/s41467-024-45221-2] [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: 08/17/2023] [Accepted: 01/18/2024] [Indexed: 02/12/2024] Open
Abstract
A major limitation to developing chimeric antigen receptor (CAR)-T cell therapies for solid tumors is identifying surface proteins highly expressed in tumors but not in normal tissues. Here, we identify Tyrosinase Related Protein 1 (TYRP1) as a CAR-T cell therapy target to treat patients with cutaneous and rare melanoma subtypes unresponsive to immune checkpoint blockade. TYRP1 is primarily located intracellularly in the melanosomes, with a small fraction being trafficked to the cell surface via vesicular transport. We develop a highly sensitive CAR-T cell therapy that detects surface TYRP1 in tumor cells with high TYRP1 overexpression and presents antitumor activity in vitro and in vivo in murine and patient-derived cutaneous, acral and uveal melanoma models. Furthermore, no systemic or off-tumor severe toxicities are observed in an immunocompetent murine model. The efficacy and safety profile of the TYRP1 CAR-T cell therapy supports the ongoing preparation of a phase I clinical trial.
Collapse
Affiliation(s)
- Sameeha Jilani
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Justin D Saco
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Edurne Mugarza
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Aleida Pujol-Morcillo
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jeffrey Chokry
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Clement Ng
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Gabriel Abril-Rodriguez
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
| | - David Berger-Manerio
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ami Pant
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Jane Hu
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Rubi Gupta
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Agustin Vega-Crespo
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Ignacio Baselga-Carretero
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
| | - Jia M Chen
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
| | - Daniel Sanghoon Shin
- Division of Hematology/Oncology, VA Greater Los Angeles Healthcare System, Los Angeles, CA, USA
- Molecular Biology Institute, UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA
| | - Philip Scumpia
- Division of Dermatology, Department of Medicine, UCLA, Los Angeles, CA, USA
- Department of Dermatology, VA Greater Los Angeles Healthcare System-West Los Angeles, Los Angeles, CA, USA
| | - Roxana A Radu
- UCLA Stein Eye Institute and Department of Ophthalmology, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Yvonne Chen
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA
- Department of Microbiology, Immunology, and Molecular Genetics at UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center-UCLA, Los Angeles, CA, USA
| | - Antoni Ribas
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA
- Broad Stem Cell Research Center-UCLA, Los Angeles, CA, USA
| | - Cristina Puig-Saus
- Department of Hematology-Oncology, David Geffen School of Medicine at the University of California Los Angeles (UCLA), Los Angeles, CA, USA.
- Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, CA, USA.
- Jonsson Comprehensive Cancer Center-UCLA, Los Angeles, CA, USA.
- Broad Stem Cell Research Center-UCLA, Los Angeles, CA, USA.
| |
Collapse
|
17
|
Partin AC, Bruno R, Shafaattalab S, Vander Mause E, Winters A, Daris M, Gahrs C, Jette CA, DiAndreth B, Sandberg ML, Hamburger AE, Kamb A, Riley TP. Geometric parameters that affect the behavior of logic-gated CAR T cells. Front Immunol 2024; 15:1304765. [PMID: 38343543 PMCID: PMC10853413 DOI: 10.3389/fimmu.2024.1304765] [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: 09/29/2023] [Accepted: 01/12/2024] [Indexed: 02/15/2024] Open
Abstract
Clinical applications of CAR-T cells are limited by the scarcity of tumor-specific targets and are often afflicted with the same on-target/off-tumor toxicities that plague other cancer treatments. A new promising strategy to enforce tumor selectivity is the use of logic-gated, two-receptor systems. One well-described application is termed Tmod™, which originally utilized a blocking inhibitory receptor directed towards HLA-I target antigens to create a protective NOT gate. Here we show that the function of Tmod blockers targeting non-HLA-I antigens is dependent on the height of the blocker antigen and is generally compatible with small, membrane-proximal targets. We compensate for this apparent limitation by incorporating modular hinge units to artificially extend or retract the ligand-binding domains relative to the effector cell surface, thereby modulating Tmod activator and blocker function. By accounting for structural differences between activator and blocker targets, we developed a set of simple geometric parameters for Tmod receptor design that enables targeting of blocker antigens beyond HLA-I, thereby broadening the applications of logic-gated cell therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Alexander Kamb
- A2 Biotherapeutics, Inc., Agoura Hills, CA, United States
| | | |
Collapse
|
18
|
Taheri FH, Hassani M, Sharifzadeh Z, Behdani M, Abdoli S, Sayadi M, Bagherzadeh K, Arashkia A, Abolhassani M. Tuning spacer length improves the functionality of the nanobody-based VEGFR2 CAR T cell. BMC Biotechnol 2024; 24:1. [PMID: 38178096 PMCID: PMC10768260 DOI: 10.1186/s12896-023-00827-0] [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: 03/09/2023] [Accepted: 12/19/2023] [Indexed: 01/06/2024] Open
Abstract
BACKGROUND The chimeric antigen receptor-expressing T (CAR-T) cells for cancer immunotherapy have obtained considerable clinical importance. CAR T cells need an optimized intracellular signaling domain to get appropriately activated and also for the proper antigen recognition, the length and composition of the extracellular spacer are critical factors. RESULTS We constructed two third-generation nanobody-based VEGFR2-CARs containing either IgG1 hinge-CH2-CH3 region or hinge-only as long or short extracellular spacers, respectively. Both CARs also contained intracellular activating domains of CD28, OX40, and CD3ζ. The T cells from healthy individuals were transduced efficiently with the two CARs, and showed increased secretion of IL-2 and IFN-γ cytokines, and also CD69 and CD25 activation markers along with cytolytic activity after encountering VEGFR2+ cells. The VEGFR2-CAR T cells harboring the long spacer showed higher cytokine release and CD69 and CD25 expression in addition to a more efficient cytolytic effect on VEGFR2+ target cells. CONCLUSIONS The results demonstrated that the third-generation anti-VEGFR2 nanobody-based CAR T cell with a long spacer had a superior function and potentially could be a better candidate for solid tumor treatment.
Collapse
Affiliation(s)
- Fatemeh Hajari Taheri
- Hybridoma Lab, Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
- Food and Drug Laboratory Research Center (FDLRC), Iran Food and Drug Administration (IFDA), MOH & ME, Tehran, Iran
| | - Mahmoud Hassani
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Zahra Sharifzadeh
- Hybridoma Lab, Department of Immunology, Pasteur Institute of Iran, Tehran, Iran
| | - Mahdi Behdani
- Department of Medical Biotechnology, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, Iran
| | - Shahryar Abdoli
- Department of Medical Biotechnology, Golestan University of Medical Science, Gorgān, Iran
| | - Mahtab Sayadi
- Cellular and Molecular Research Center, Birjand University of Medical Sciences, Birjand, Iran
| | - Kowsar Bagherzadeh
- Eye Research Center, Five Senses Health Institute, Rassoul Akram Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Arash Arashkia
- Department of Molecular Virology, Pasteur Institute of Iran, Tehran, Iran.
| | - Mohsen Abolhassani
- Hybridoma Lab, Department of Immunology, Pasteur Institute of Iran, Tehran, Iran.
| |
Collapse
|
19
|
Khodke P, Kumbhar BV. Engineered CAR-T cells: An immunotherapeutic approach for cancer treatment and beyond. ADVANCES IN PROTEIN CHEMISTRY AND STRUCTURAL BIOLOGY 2024; 140:157-198. [PMID: 38762269 DOI: 10.1016/bs.apcsb.2023.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2024]
Abstract
Chimeric Antigen Receptor (CAR) T cell therapy is a type of adoptive immunotherapy that offers a promising avenue for enhancing cancer treatment since traditional cancer treatments like chemotherapy, surgery, and radiation therapy have proven insufficient in completely eradicating tumors, despite the relatively positive outcomes. It has been observed that CAR-T cell therapy has shown promising results in treating the majority of hematological malignancies but also have a wide scope for other cancer types. CAR is an extra receptor on the T-cell that helps to increase and accelerate tumor destruction by efficiently activating the immune system. It is made up of three domains, the ectodomain, transmembrane, and the endodomain. The ectodomain is essential for antigen recognition and binding, whereas the co-stimulatory signal is transduced by the endodomain. To date, the Food and Drug Administration (FDA) has granted approval for six CAR-T cell therapies. However, despite its remarkable success, CAR-T therapy is associated with numerous adverse events and has certain limitations. This chapter focuses on the structure and function of the CAR domain, various generations of CAR, and the process of CAR-T cell development, adverse effects, and challenges in CAR-T therapy. CAR-T cell therapy also has scopes in other disease conditions which include systemic lupus erythematosus, multiple sclerosis, and myocardial fibrosis, etc.
Collapse
Affiliation(s)
- Purva Khodke
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, India
| | - Bajarang Vasant Kumbhar
- Department of Biological Sciences, Sunandan Divatia School of Science, SVKM's Narsee Monjee Institute of Management Studies (NMIMS) Deemed-to-be University, Mumbai, India.
| |
Collapse
|
20
|
Eskandari SK, Daccache A, Azzi JR. Chimeric antigen receptor T reg therapy in transplantation. Trends Immunol 2024; 45:48-61. [PMID: 38123369 DOI: 10.1016/j.it.2023.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/23/2023]
Abstract
In the quest for more precise and effective organ transplantation therapies, chimeric antigen receptor (CAR) regulatory T cell (Treg) therapies represent a potential cutting-edge advance. This review comprehensively analyses CAR Tregs and how they may address important drawbacks of polyclonal Tregs and conventional immunosuppressants. We examine a growing body of preclinical findings of CAR Treg therapy in transplantation, discuss CAR Treg design specifics, and explore established and attractive new targets in transplantation. In addition, we explore present impediments where future studies will be necessary to determine the efficacy of CAR Tregs in reshaping alloimmune responses and transplant microenvironments to reduce reliance on chemical immunosuppressants. Overall, ongoing studies and trials are crucial for understanding the full scope of CAR Treg therapy in transplantation.
Collapse
Affiliation(s)
- Siawosh K Eskandari
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Department of Surgery, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| | - Andrea Daccache
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA; Division of Bioscience Education and Research (UFR Biosciences), Claude Bernard University Lyon 1, Lyon, France
| | - Jamil R Azzi
- Transplantation Research Center, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
21
|
Mc Laughlin AM, Milligan PA, Yee C, Bergstrand M. Model-informed drug development of autologous CAR-T cell therapy: Strategies to optimize CAR-T cell exposure leveraging cell kinetic/dynamic modeling. CPT Pharmacometrics Syst Pharmacol 2023; 12:1577-1590. [PMID: 37448343 PMCID: PMC10681459 DOI: 10.1002/psp4.13011] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 06/23/2023] [Accepted: 07/10/2023] [Indexed: 07/15/2023] Open
Abstract
Autologous Chimeric antigen receptor (CAR-T) cell therapy has been highly successful in the treatment of aggressive hematological malignancies and is also being evaluated for the treatment of solid tumors as well as other therapeutic areas. A challenge, however, is that up to 60% of patients do not sustain a long-term response. Low CAR-T cell exposure has been suggested as an underlying factor for a poor prognosis. CAR-T cell therapy is a novel therapeutic modality with unique kinetic and dynamic properties. Importantly, "clear" dose-exposure relationships do not seem to exist for any of the currently approved CAR-T cell products. In other words, dose increases have not led to a commensurate increase in the measurable in vivo frequency of transferred CAR-T cells. Therefore, alternative approaches beyond dose titration are needed to optimize CAR-T cell exposure. In this paper, we provide examples of actionable variables - design elements in CAR-T cell discovery, development, and clinical practice, which can be modified to optimize autologous CAR-T cell exposure. Most of these actionable variables can be assessed throughout the various stages of discovery and development as part of a well-informed research and development program. Model-informed drug development approaches can enable such study and program design choices from discovery through to clinical practice and can be an important contributor to cell therapy effectiveness and efficiency.
Collapse
Affiliation(s)
| | | | - Cassian Yee
- Department of Melanoma Medical OncologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
- Department of ImmunologyThe University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| | | |
Collapse
|
22
|
Rios X, Pardias O, Morales MA, Bhattacharya P, Chen Y, Guo L, Zhang C, Di Pierro EJ, Tian G, Barragan GA, Sumazin P, Metelitsa LS. Refining chimeric antigen receptors via barcoded protein domain combination pooled screening. Mol Ther 2023; 31:3210-3224. [PMID: 37705245 PMCID: PMC10638030 DOI: 10.1016/j.ymthe.2023.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Revised: 09/06/2023] [Accepted: 09/08/2023] [Indexed: 09/15/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cells represent a promising frontier in cancer immunotherapy. However, the current process for developing new CAR constructs is time consuming and inefficient. To address this challenge and expedite the evaluation and comparison of full-length CAR designs, we have devised a novel cloning strategy. This strategy involves the sequential assembly of individual CAR domains using blunt ligation, with each domain being assigned a unique DNA barcode. Applying this method, we successfully generated 360 CAR constructs that specifically target clinically validated tumor antigens CD19 and GD2. By quantifying changes in barcode frequencies through next-generation sequencing, we characterize CARs that best mediate proliferation and expansion of transduced T cells. The screening revealed a crucial role for the hinge domain in CAR functionality, with CD8a and IgG4 hinges having opposite effects in the surface expression, cytokine production, and antitumor activity in CD19- versus GD2-based CARs. Importantly, we discovered two novel CD19-CAR architectures containing the IgG4 hinge domain that mediate superior in vivo antitumor activity compared with the construct used in Kymriah, a U.S. Food and Drug Administration (FDA)-approved therapy. This novel screening approach represents a major advance in CAR engineering, enabling accelerated development of cell-based cancer immunotherapies.
Collapse
Affiliation(s)
- Xavier Rios
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Osmay Pardias
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Marc A Morales
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Pradyot Bhattacharya
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Yibin Chen
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Linjie Guo
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Chunchao Zhang
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Erica J Di Pierro
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Gengwen Tian
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Gabriel A Barragan
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA
| | - Leonid S Metelitsa
- Center for Advanced Innate Cell Therapy, Texas Children's Cancer Center, Department of Pediatrics, Baylor College of Medicine, Houston, TX, USA; Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX, USA; Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
23
|
Singh N, Maus MV. Synthetic manipulation of the cancer-immunity cycle: CAR-T cell therapy. Immunity 2023; 56:2296-2310. [PMID: 37820585 DOI: 10.1016/j.immuni.2023.09.010] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/15/2023] [Accepted: 09/15/2023] [Indexed: 10/13/2023]
Abstract
Synthetic immunity to cancer has been pioneered by the application of chimeric antigen receptor (CAR) engineering into autologous T cells. CAR T cell therapy is highly amenable to molecular engineering to bypass barriers of the cancer immunity cycle, such as endogenous antigen presentation, immune priming, and natural checkpoints that constrain immune responses. Here, we review CAR T cell design and the mechanisms that drive sustained CAR T cell effector activity and anti-tumor function. We discuss engineering approaches aimed at improving anti-tumor function through a variety of mechanistic interventions for both hematologic and solid tumors. The ability to engineer T cells in such a variety of ways, including by modifying their trafficking, antigen recognition, costimulation, and addition of synthetic genes, circuits, knockouts and base edits to finely tune complex functions, is arguably the most powerful way to manipulate the cancer immunity cycle in patients.
Collapse
Affiliation(s)
- Nathan Singh
- Division of Oncology, Washington University in St Louis School of Medicine, St. Louis, MO 63110, USA.
| | - Marcela V Maus
- Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
24
|
Bandara V, Foeng J, Gundsambuu B, Norton TS, Napoli S, McPeake DJ, Tyllis TS, Rohani-Rad E, Abbott C, Mills SJ, Tan LY, Thompson EJ, Willet VM, Nikitaras VJ, Zheng J, Comerford I, Johnson A, Coombs J, Oehler MK, Ricciardelli C, Cowin AJ, Bonder CS, Jensen M, Sadlon TJ, McColl SR, Barry SC. Pre-clinical validation of a pan-cancer CAR-T cell immunotherapy targeting nfP2X7. Nat Commun 2023; 14:5546. [PMID: 37684239 PMCID: PMC10491676 DOI: 10.1038/s41467-023-41338-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 08/29/2023] [Indexed: 09/10/2023] Open
Abstract
Chimeric antigen receptor (CAR)-T cell immunotherapy is a novel treatment that genetically modifies the patients' own T cells to target and kill malignant cells. However, identification of tumour-specific antigens expressed on multiple solid cancer types, remains a major challenge. P2X purinoceptor 7 (P2X7) is a cell surface expressed ATP gated cation channel, and a dysfunctional version of P2X7, named nfP2X7, has been identified on cancer cells from multiple tissues, while being undetectable on healthy cells. We present a prototype -human CAR-T construct targeting nfP2X7 showing potential antigen-specific cytotoxicity against twelve solid cancer types (breast, prostate, lung, colorectal, brain and skin). In xenograft mouse models of breast and prostate cancer, CAR-T cells targeting nfP2X7 exhibit robust anti-tumour efficacy. These data indicate that nfP2X7 is a suitable immunotherapy target because of its broad expression on human tumours. CAR-T cells targeting nfP2X7 have potential as a wide-spectrum cancer immunotherapy for solid tumours in humans.
Collapse
Affiliation(s)
- Veronika Bandara
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Jade Foeng
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Batjargal Gundsambuu
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Todd S Norton
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Silvana Napoli
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Dylan J McPeake
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Timona S Tyllis
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Elaheh Rohani-Rad
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Caitlin Abbott
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Stuart J Mills
- University of South Australia, STEM (Future Industries Institute) SA, Adelaide, 5095, Australia
| | - Lih Y Tan
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
| | - Emma J Thompson
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
| | - Vasiliki M Willet
- Reproductive Cancer Research Group, Discipline Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Victoria J Nikitaras
- Reproductive Cancer Research Group, Discipline Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Jieren Zheng
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia
| | - Iain Comerford
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Adam Johnson
- Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Justin Coombs
- Carina Biotech, Level 2 Innovation & Collaboration Centre, UniSA Bradley Building, Adelaide, SA, 5001, Australia
| | - Martin K Oehler
- Department of Gynaecological Oncology, Royal Adelaide Hospital, Adelaide, SA, 5005, Australia
| | - Carmela Ricciardelli
- Reproductive Cancer Research Group, Discipline Obstetrics and Gynaecology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5005, Australia
| | - Allison J Cowin
- University of South Australia, STEM (Future Industries Institute) SA, Adelaide, 5095, Australia
| | - Claudine S Bonder
- Centre for Cancer Biology, University of South Australia and SA Pathology, Adelaide, SA, 5001, Australia
- Adelaide Medical School, The University of Adelaide, Adelaide, SA, 5005, Australia
| | - Michael Jensen
- Seattle Children's Research Institute, Seattle, WA, 98101, USA
| | - Timothy J Sadlon
- Department of Gastroenterology, Women's and Children's Health Network, North Adelaide, SA, 5006, Australia
| | - Shaun R McColl
- Chemokine Biology Laboratory, Department of Molecular and Cellular Biology, School of Biological Sciences, University of Adelaide, Adelaide, SA, 5005, Australia
- Carina Biotech, Level 2 Innovation & Collaboration Centre, UniSA Bradley Building, Adelaide, SA, 5001, Australia
| | - Simon C Barry
- Molecular Immunology, Robinson Research Institute, University of Adelaide, Adelaide, SA, 5000, Australia.
- Carina Biotech, Level 2 Innovation & Collaboration Centre, UniSA Bradley Building, Adelaide, SA, 5001, Australia.
- Department of Gastroenterology, Women's and Children's Health Network, North Adelaide, SA, 5006, Australia.
| |
Collapse
|
25
|
Dagar G, Gupta A, Masoodi T, Nisar S, Merhi M, Hashem S, Chauhan R, Dagar M, Mirza S, Bagga P, Kumar R, Akil ASAS, Macha MA, Haris M, Uddin S, Singh M, Bhat AA. Harnessing the potential of CAR-T cell therapy: progress, challenges, and future directions in hematological and solid tumor treatments. J Transl Med 2023; 21:449. [PMID: 37420216 PMCID: PMC10327392 DOI: 10.1186/s12967-023-04292-3] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 06/21/2023] [Indexed: 07/09/2023] Open
Abstract
Traditional cancer treatments use nonspecific drugs and monoclonal antibodies to target tumor cells. Chimeric antigen receptor (CAR)-T cell therapy, however, leverages the immune system's T-cells to recognize and attack tumor cells. T-cells are isolated from patients and modified to target tumor-associated antigens. CAR-T therapy has achieved FDA approval for treating blood cancers like B-cell acute lymphoblastic leukemia, large B-cell lymphoma, and multiple myeloma by targeting CD-19 and B-cell maturation antigens. Bi-specific chimeric antigen receptors may contribute to mitigating tumor antigen escape, but their efficacy could be limited in cases where certain tumor cells do not express the targeted antigens. Despite success in blood cancers, CAR-T technology faces challenges in solid tumors, including lack of reliable tumor-associated antigens, hypoxic cores, immunosuppressive tumor environments, enhanced reactive oxygen species, and decreased T-cell infiltration. To overcome these challenges, current research aims to identify reliable tumor-associated antigens and develop cost-effective, tumor microenvironment-specific CAR-T cells. This review covers the evolution of CAR-T therapy against various tumors, including hematological and solid tumors, highlights challenges faced by CAR-T cell therapy, and suggests strategies to overcome these obstacles, such as utilizing single-cell RNA sequencing and artificial intelligence to optimize clinical-grade CAR-T cells.
Collapse
Affiliation(s)
- Gunjan Dagar
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Ashna Gupta
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Tariq Masoodi
- Laboratory of Cancer Immunology and Genetics, Sidra Medicine, Doha, Qatar
| | - Sabah Nisar
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Maysaloun Merhi
- National Center for Cancer Care and Research, Hamad Medical Corporation, 3050, Doha, Qatar
| | - Sheema Hashem
- Department of Human Genetics, Sidra Medicine, Doha, Qatar
| | - Ravi Chauhan
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India
| | - Manisha Dagar
- Shiley Eye Institute, University of California San Diego, San Diego, CA, USA
| | - Sameer Mirza
- Department of Chemistry, College of Sciences, United Arab Emirates University, Al-Ain, United Arab Emirates
| | - Puneet Bagga
- Department of Diagnostic Imaging, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Rakesh Kumar
- School of Biotechnology, Shri Mata Vaishno Devi University, Katra, Jammu and Kashmir, 182320, India
| | - Ammira S Al-Shabeeb Akil
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar
| | - Muzafar A Macha
- Watson-Crick Centre for Molecular Medicine, Islamic University of Science and Technology, Pulwama, Jammu and Kashmir, India
| | - Mohammad Haris
- Center for Advanced Metabolic Imaging in Precision Medicine, Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, USA
- Laboratory Animal Research Center, Qatar University, Doha, Qatar
| | - Shahab Uddin
- Laboratory Animal Research Center, Qatar University, Doha, Qatar.
- Translational Research Institute, Academic Health System, Hamad Medical Corporation, P.O. Box 3050, Doha, Qatar.
| | - Mayank Singh
- Department of Medical Oncology (Lab.), Dr. BRAIRCH, All India Institute of Medical Sciences (AIIMS), New Delhi, Delhi, 110029, India.
| | - Ajaz A Bhat
- Department of Human Genetics-Precision Medicine in Diabetes, Obesity and Cancer Program, Sidra Medicine, P.O. Box 26999, Doha, Qatar.
| |
Collapse
|
26
|
Pfeifer R, Al Rawashdeh W, Brauner J, Martinez-Osuna M, Lock D, Herbel C, Eckardt D, Assenmacher M, Bosio A, Hardt OT, Johnston ICD. Targeting Stage-Specific Embryonic Antigen 4 (SSEA-4) in Triple Negative Breast Cancer by CAR T Cells Results in Unexpected on Target/off Tumor Toxicities in Mice. Int J Mol Sci 2023; 24:ijms24119184. [PMID: 37298141 DOI: 10.3390/ijms24119184] [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: 04/13/2023] [Revised: 05/19/2023] [Accepted: 05/20/2023] [Indexed: 06/12/2023] Open
Abstract
Due to the paucity of targetable antigens, triple-negative breast cancer (TNBC) remains a challenging subtype of breast cancer to treat. In this study, we developed and evaluated a chimeric antigen receptor (CAR) T cell-based treatment modality for TNBC by targeting stage-specific embryonic antigen 4 (SSEA-4), a glycolipid whose overexpression in TNBC has been correlated with metastasis and chemoresistance. To delineate the optimal CAR configuration, a panel of SSEA-4-specific CARs containing alternative extracellular spacer domains was constructed. The different CAR constructs mediated antigen-specific T cell activation characterized by degranulation of T cells, secretion of inflammatory cytokines, and killing of SSEA-4-expressing target cells, but the extent of this activation differed depending on the length of the spacer region. Adoptive transfer of the CAR-engineered T cells into mice with subcutaneous TNBC xenografts mediated a limited antitumor effect but induced severe toxicity symptoms in the cohort receiving the most bioactive CAR variant. We found that progenitor cells in the lung and bone marrow express SSEA-4 and are likely co-targeted by the CAR T cells. Thus, this study has revealed serious adverse effects that raise safety concerns for SSEA-4-directed CAR therapies because of the risk of eliminating vital cells with stem cell properties.
Collapse
Affiliation(s)
- Rita Pfeifer
- Miltenyi Biotec GmbH, 51429 Bergisch Gladbach, Germany
| | | | | | | | - Dominik Lock
- Miltenyi Biotec GmbH, 51429 Bergisch Gladbach, Germany
| | | | | | | | - Andreas Bosio
- Miltenyi Biotec GmbH, 51429 Bergisch Gladbach, Germany
| | - Olaf T Hardt
- Miltenyi Biotec GmbH, 51429 Bergisch Gladbach, Germany
| | | |
Collapse
|
27
|
Pieper T, Roth KDR, Glaser V, Riet T, Buitrago-Molina LE, Hagedorn M, Lieber M, Hust M, Noyan F, Jaeckel E, Hardtke-Wolenski M. Generation of Chimeric Antigen Receptors against Tetraspanin 7. Cells 2023; 12:1453. [PMID: 37296574 PMCID: PMC10252682 DOI: 10.3390/cells12111453] [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: 03/05/2023] [Revised: 05/10/2023] [Accepted: 05/17/2023] [Indexed: 06/12/2023] Open
Abstract
Adoptive transfer of antigen-specific regulatory T cells (Tregs) has shown promising results in the treatment of autoimmune diseases; however, the use of polyspecific Tregs has limited effects. However, obtaining a sufficient number of antigen-specific Tregs from patients with autoimmune disorders remains challenging. Chimeric antigen receptors (CARs) provide an alternative source of T cells for novel immunotherapies that redirect T cells independently of the MHC. In this study, we aimed to generate antibody-like single-chain variable fragments (scFv) and subsequent CARs against tetraspanin 7 (TSPAN7), a membrane protein highly expressed on the surface of pancreatic beta cells, using phage display technology. We established two methods for generating scFvs against TSPAN7 and other target structures. Moreover, we established novel assays to analyze and quantify their binding abilities. The resulting CARs were functional and activated specifically by the target structure, but could not recognize TSPAN7 on the surface of beta cells. Despite this, this study demonstrates that CAR technology is a powerful tool for generating antigen-specific T cells and provides new approaches for generating functional CARs.
Collapse
Affiliation(s)
- Tom Pieper
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Kristian Daniel Ralph Roth
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Medizinische Biotechnologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Viktor Glaser
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Tobias Riet
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department I of Internal Medicine, Tumor Genetics, University Hospital of Cologne, Center for Molecular Medicine Cologne (CMMC), University of Cologne, 50933 Cologne, Germany
| | - Laura Elisa Buitrago-Molina
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Maike Hagedorn
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Maren Lieber
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Michael Hust
- Institut für Biochemie, Biotechnologie und Bioinformatik, Abteilung Medizinische Biotechnologie, Technische Universität Braunschweig, 38106 Braunschweig, Germany
| | - Fatih Noyan
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
| | - Elmar Jaeckel
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Department of Liver Transplantation, Multi Organ Transplant Program, University Health Network, University of Toronto, Toronto, ON M5T 0S8, Canada
| | - Matthias Hardtke-Wolenski
- Department of Gastroenterology, Hepatology, Infectious Diseases & Endocrinology, Hannover Medical School, 30625 Hannover, Germany
- Institute of Medical Microbiology, University Hospital Essen, University Duisburg-Essen, 47057 Essen, Germany
| |
Collapse
|
28
|
Mavi AK, Gaur S, Gaur G, Babita, Kumar N, Kumar U. CAR T-cell therapy: Reprogramming patient's immune cell to treat cancer. Cell Signal 2023; 105:110638. [PMID: 36822565 DOI: 10.1016/j.cellsig.2023.110638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 02/20/2023] [Indexed: 02/23/2023]
Abstract
Chimeric antigen receptor (CAR)-T cell therapy is a game changer in cancer treatment. Although CAR-T cell therapy has achieved significant clinical responses in specific subgroups of B cell leukaemia or lymphoma, various difficulties restrict CAR-T cell therapy's therapeutic effectiveness in solid tumours and haematological malignancies. Severe life-threatening toxicities, poor anti-tumour effectiveness, antigen escape, restricted trafficking, and limited tumour penetration are all barriers to successful CAR-T cell treatment. Furthermore, CAR-T cell interactions with the host and tumour microenvironment have a significant impact on their activity. Furthermore, developing and implementing these therapies necessitates a complicated staff. Innovative methodologies and tactics to engineering more potent CAR-T cells with greater anti-tumour activity and less toxicity are required to address these important difficulties.
Collapse
Affiliation(s)
- Anil Kumar Mavi
- Department of Pulmonary Medicine, Vallabhbhai Patel Chest Institute, University of Delhi, Delhi 110007, India
| | - Sonal Gaur
- Department of Biosciences and Biotechnology, Banasthali Vidyapith, Jaipur, Rajasthan 304022, India
| | - Gauri Gaur
- Department of Biotechnology, Maharishi Markandeshwar (Deemed to be University), Mullana, Ambala, Haryana 133203, India
| | - Babita
- Department of Pharmacology, Sharda School of Allied Health Sciences, Sharda University, Knowledge Park III, Greater Noida, Uttar Pradesh 201310, India
| | - Neelesh Kumar
- Department of Aquaculture, College of Fisheries, GB Pant University of Agriculture & Technology, Pantnagar, Udham Singh Nagar, Uttarakhand 263145, India
| | - Umesh Kumar
- School of Biosciences, Institute of Management Studies Ghaziabad (University Courses Campus), NH09, Adhyatmik Nagar, Ghaziabad, Uttar Pradesh 201015, India.
| |
Collapse
|
29
|
Li N, Quan A, Li D, Pan J, Ren H, Hoeltzel G, de Val N, Ashworth D, Ni W, Zhou J, Mackay S, Hewitt SM, Cachau R, Ho M. The IgG4 hinge with CD28 transmembrane domain improves V HH-based CAR T cells targeting a membrane-distal epitope of GPC1 in pancreatic cancer. Nat Commun 2023; 14:1986. [PMID: 37031249 PMCID: PMC10082787 DOI: 10.1038/s41467-023-37616-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2022] [Accepted: 03/23/2023] [Indexed: 04/10/2023] Open
Abstract
Heterogeneous antigen expression is a key barrier influencing the activity of chimeric antigen receptor (CAR) T cells in solid tumors. Here, we develop CAR T cells targeting glypican-1 (GPC1), an oncofetal antigen expressed in pancreatic cancer. We report the generation of dromedary camel VHH nanobody (D4)-based CAR T cells targeting GPC1 and the optimization of the hinge (H) and transmembrane domain (TM) to improve activity. We find that a structurally rigid IgG4H and CD28TM domain brings the two D4 fragments in proximity, driving CAR dimerization and leading to enhanced T-cell signaling and tumor regression in pancreatic cancer models with low antigen density in female mice. Furthermore, single-cell-based proteomic and transcriptomic analysis of D4-IgG4H-CD28TM CAR T cells reveals specific genes (e.g., HMGB1) associated with high T-cell polyfunctionality. This study demonstrates the potential of VHH-based CAR T for pancreatic cancer therapy and provides an engineering strategy for developing potent CAR T cells targeting membrane-distal epitopes.
Collapse
Affiliation(s)
- Nan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Alex Quan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Dan Li
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jiajia Pan
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hua Ren
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gerard Hoeltzel
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Natalia de Val
- Center for Molecular Microscopy, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Frederick, MD, 21702, USA
| | | | - Weiming Ni
- IsoPlexis Corporation, Branford, CT, 06405, USA
| | - Jing Zhou
- IsoPlexis Corporation, Branford, CT, 06405, USA
| | - Sean Mackay
- IsoPlexis Corporation, Branford, CT, 06405, USA
| | - Stephen M Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Raul Cachau
- Integrated Data Science Section, Research Technologies Branch, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Mitchell Ho
- Laboratory of Molecular Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
30
|
Lakhani A, Chen X, Chen LC, Khericha M, Chen YY, Park JO. Extracellular Domains of CAR Reprogram T-Cell Metabolism Without Antigen Stimulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.04.03.533021. [PMID: 37066394 PMCID: PMC10103977 DOI: 10.1101/2023.04.03.533021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/02/2023]
Abstract
Metabolism is an indispensable part of T-cell proliferation, activation, and exhaustion, yet the metabolism of chimeric antigen receptor (CAR)-T cells remains incompletely understood. CARs are comprised of extracellular domains that determine cancer specificity, often using single-chain variable fragments (scFvs), and intracellular domains that trigger signaling upon antigen binding. Here we show that CARs differing only in the scFv reprogram T-cell metabolism differently. Even in the absence of antigens, some CARs increase proliferation and nutrient uptake in T cells. Using stable isotope tracers and mass spectrometry, we observe basal metabolic fluxes through glycolysis doubling and amino acid uptake overtaking anaplerosis in CAR-T cells harboring rituximab scFv, unlike other similar anti-CD20 scFvs. Disparate rituximab and 14g2a-based anti-GD2 CAR-T cells are similarly hypermetabolic and channel excess nutrients to nitrogen overflow metabolism. Since CAR-dependent metabolic reprogramming alters cellular energetics, nutrient utilization, and proliferation, metabolic profiling should be an integral part of CAR-T cell development.
Collapse
|
31
|
Tuning charge density of chimeric antigen receptor optimizes tonic signaling and CAR-T cell fitness. Cell Res 2023; 33:341-354. [PMID: 36882513 PMCID: PMC10156745 DOI: 10.1038/s41422-023-00789-0] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 02/10/2023] [Indexed: 03/09/2023] Open
Abstract
Tonic signaling of chimeric antigen receptor (CAR), i.e., the spontaneous CAR activation in the absence of tumor antigen stimulation, is considered to be a pivotal event controlling CAR-T efficacy. However, the molecular mechanism underlying the spontaneous CAR signals remains elusive. Here, we unveil that positively charged patches (PCPs) on the surface of the CAR antigen-binding domain mediate CAR clustering and result in CAR tonic signaling. For CARs with high tonic signaling (e.g., GD2.CAR and CSPG4.CAR), reducing PCPs on CARs or boosting ionic strength in the culture medium during ex vivo CAR-T cell expansion minimizes spontaneous CAR activation and alleviates CAR-T cell exhaustion. In contrast, introducing PCPs into the CAR with weak tonic signaling, such as CD19.CAR, results in improved in vivo persistence and superior antitumor function. These results demonstrate that CAR tonic signaling is induced and maintained by PCP-mediated CAR clustering. Notably, the mutations we generated to alter the PCPs maintain the antigen-binding affinity and specificity of the CAR. Therefore, our findings suggest that the rational tuning of PCPs to optimize tonic signaling and in vivo fitness of CAR-T cells is a promising design strategy for the next-generation CAR.
Collapse
|
32
|
Chimeric antigen receptor T cells therapy in solid tumors. Clin Transl Oncol 2023:10.1007/s12094-023-03122-8. [PMID: 36853399 DOI: 10.1007/s12094-023-03122-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 02/10/2023] [Indexed: 03/01/2023]
Abstract
Chimeric antigen receptor T cells therapy (CAR-T therapy) is a class of ACT therapy. Chimeric antigen receptor (CAR) is an engineered synthetic receptor of CAR-T, which give T cells the ability to recognize tumor antigens in a human leukocyte antigen-independent (HLA-independent) manner and enables them to recognize more extensive target antigens than natural T cell surface receptor (TCR), resulting in tumor destruction. CAR-T is composed of an extracellular single-chain variable fragment (scFv) of antibody, which serves as the targeting moiety, hinge region, transmembrane spacer, and intracellular signaling domain(s). CAR-T has been developing in many generations, which differ according to costimulatory domains. CAR-T therapy has several limitations that reduce its wide availability in immunotherapy which we can summarize in antigen escape that shows either partial or complete loss of target antigen expression, so multiplexing CAR-T cells are promoted to enhance targeting of tumor profiles. In addition, the large diversity in the tumor microenvironment also plays a major role in limiting this kind of treatment. Therefore, engineered CAR-T cells can evoke immunostimulatory signals that rebalance the tumor microenvironment. Using CAR-T therapy in treating the solid tumor is mainly restricted by the difficulty of CAR-T cells infiltrating the tumor site, so local administration was developed to improve the quality of treatment. The most severe toxicity after CAR-T therapy is on-target/on-tumor toxicity, such as cytokine release syndrome (CRS). Another type of toxicity is on-target/off-tumor toxicity which originates from the binding of CAR-T cells to target antigen that has shared expression on normal cells leading to damage in healthy cells and organs. Toxicity management should become a focus of implementation to permit management beyond specialized centers.
Collapse
|
33
|
Ghaedrahmati F, Esmaeil N, Abbaspour M. Targeting immune checkpoints: how to use natural killer cells for fighting against solid tumors. Cancer Commun (Lond) 2022; 43:177-213. [PMID: 36585761 PMCID: PMC9926962 DOI: 10.1002/cac2.12394] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Revised: 10/08/2022] [Accepted: 11/15/2022] [Indexed: 01/01/2023] Open
Abstract
Natural killer (NK) cells are unique innate immune cells that mediate anti-viral and anti-tumor responses. Thus, they might hold great potential for cancer immunotherapy. NK cell adoptive immunotherapy in humans has shown modest efficacy. In particular, it has failed to demonstrate therapeutic efficiency in the treatment of solid tumors, possibly due in part to the immunosuppressive tumor microenvironment (TME), which reduces NK cell immunotherapy's efficiencies. It is known that immune checkpoints play a prominent role in creating an immunosuppressive TME, leading to NK cell exhaustion and tumor immune escape. Therefore, NK cells must be reversed from their dysfunctional status and increased in their effector roles in order to improve the efficiency of cancer immunotherapy. Blockade of immune checkpoints can not only rescue NK cells from exhaustion but also augment their robust anti-tumor activity. In this review, we discussed immune checkpoint blockade strategies with a focus on chimeric antigen receptor (CAR)-NK cells to redirect NK cells to cancer cells in the treatment of solid tumors.
Collapse
Affiliation(s)
- Farhoodeh Ghaedrahmati
- Department of ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran
| | - Nafiseh Esmaeil
- Department of ImmunologySchool of MedicineIsfahan University of Medical SciencesIsfahanIran,Research Institute for Primordial Prevention of Non‐Communicable DiseaseIsfahan University of Medical SciencesIsfahanIran
| | - Maryam Abbaspour
- Department of Pharmaceutical BiotechnologyFaculty of PharmacyIsfahan University of Medical SciencesIsfahanIran
| |
Collapse
|
34
|
Asmamaw Dejenie T, Tiruneh G/Medhin M, Dessie Terefe G, Tadele Admasu F, Wale Tesega W, Chekol Abebe E. Current updates on generations, approvals, and clinical trials of CAR T-cell therapy. Hum Vaccin Immunother 2022; 18:2114254. [PMID: 36094837 DOI: 10.1080/21645515.2022.2114254] [Citation(s) in RCA: 49] [Impact Index Per Article: 24.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a novel, customized immunotherapy that is considered a 'living' and self-replicating drug to treat cancer, sometimes resulting in a complete cure. CAR T-cells are manufactured through genetic engineering of T-cells by equipping them with CARs to detect and target antigen-expressing cancer cells. CAR is designed to have an ectodomain extracellularly, a transmembrane domain spanning the cell membrane, and an endodomain intracellularly. Since its first discovery, the CAR structure has evolved greatly, from the first generation to the fifth generation, to offer new therapeutic alternatives for cancer patients. This treatment has achieved long-term and curative therapeutic efficacy in multiple blood malignancies that nowadays profoundly change the treatment landscape of lymphoma, leukemia, and multiple myeloma. But CART-cell therapy is associated with several hurdles, such as limited therapeutic efficacy, little effect on solid tumors, adverse effects, expensive cost, and feasibility issues, hindering its broader implications.
Collapse
Affiliation(s)
- Tadesse Asmamaw Dejenie
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Markeshaw Tiruneh G/Medhin
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Gashaw Dessie Terefe
- Department of Biochemistry, School of Medicine, College of Medicine and Health Sciences, University of Gondar, Gondar, Ethiopia
| | - Fitalew Tadele Admasu
- Department of Biochemistry, College of Medicine and Health Science Arbaminch University, Arbaminch, Ethiopia
| | - Wondwossen Wale Tesega
- Department of Biochemistry, College of Health Sciences, Debre Tabor University, Debre Tabor, Ethiopia
| | - Endeshaw Chekol Abebe
- Department of Biochemistry, College of Medicine and Health Science Arbaminch University, Arbaminch, Ethiopia
| |
Collapse
|
35
|
Teppert K, Wang X, Anders K, Evaristo C, Lock D, Künkele A. Joining Forces for Cancer Treatment: From "TCR versus CAR" to "TCR and CAR". Int J Mol Sci 2022; 23:14563. [PMID: 36498890 PMCID: PMC9739809 DOI: 10.3390/ijms232314563] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 11/14/2022] [Accepted: 11/19/2022] [Indexed: 11/24/2022] Open
Abstract
T cell-based immunotherapy has demonstrated great therapeutic potential in recent decades, on the one hand, by using tumor-infiltrating lymphocytes (TILs) and, on the other hand, by engineering T cells to obtain anti-tumor specificities through the introduction of either engineered T cell receptors (TCRs) or chimeric antigen receptors (CARs). Given the distinct design of both receptors and the type of antigen that is encountered, the requirements for proper antigen engagement and downstream signal transduction by TCRs and CARs differ. Synapse formation and signal transduction of CAR T cells, despite further refinement of CAR T cell designs, still do not fully recapitulate that of TCR T cells and might limit CAR T cell persistence and functionality. Thus, deep knowledge about the molecular differences in CAR and TCR T cell signaling would greatly advance the further optimization of CAR designs and elucidate under which circumstances a combination of both receptors would improve the functionality of T cells for cancer treatment. Herein, we provide a comprehensive review about similarities and differences by directly comparing the architecture, synapse formation and signaling of TCRs and CARs, highlighting the knowns and unknowns. In the second part of the review, we discuss the current status of combining CAR and TCR technologies, encouraging a change in perspective from "TCR versus CAR" to "TCR and CAR".
Collapse
Affiliation(s)
- Karin Teppert
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Xueting Wang
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Kathleen Anders
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - César Evaristo
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Dominik Lock
- Miltenyi Biotec B.V. & Co. KG, 51429 Bergisch Gladbach, Germany
| | - Annette Künkele
- German Cancer Consortium (DKTK), 10117 Berlin, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Department of Pediatric Oncology and Hematology, Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany
| |
Collapse
|
36
|
Chimeric antigen receptor clustering via cysteines enhances T-cell efficacy against tumor. Cancer Immunol Immunother 2022; 71:2801-2814. [DOI: 10.1007/s00262-022-03195-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/24/2022] [Indexed: 10/18/2022]
|
37
|
Seif M, Kakoschke TK, Ebel F, Bellet MM, Trinks N, Renga G, Pariano M, Romani L, Tappe B, Espie D, Donnadieu E, Hünniger K, Häder A, Sauer M, Damotte D, Alifano M, White PL, Backx M, Nerreter T, Machwirth M, Kurzai O, Prommersberger S, Einsele H, Hudecek M, Löffler J. CAR T cells targeting Aspergillus fumigatus are effective at treating invasive pulmonary aspergillosis in preclinical models. Sci Transl Med 2022; 14:eabh1209. [PMID: 36170447 DOI: 10.1126/scitranslmed.abh1209] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Aspergillus fumigatus is a ubiquitous mold that can cause severe infections in immunocompromised patients, typically manifesting as invasive pulmonary aspergillosis (IPA). Adaptive and innate immune cells that respond to A. fumigatus are present in the endogenous repertoire of patients with IPA but are infrequent and cannot be consistently isolated and expanded for adoptive immunotherapy. Therefore, we gene-engineered A. fumigatus-specific chimeric antigen receptor (Af-CAR) T cells and demonstrate their ability to confer antifungal reactivity in preclinical models in vitro and in vivo. We generated a CAR targeting domain AB90-E8 that recognizes a conserved protein antigen in the cell wall of A. fumigatus hyphae. T cells expressing the Af-CAR recognized A. fumigatus strains and clinical isolates and exerted a direct antifungal effect against A. fumigatus hyphae. In particular, CD8+ Af-CAR T cells released perforin and granzyme B and damaged A. fumigatus hyphae. CD8+ and CD4+ Af-CAR T cells produced cytokines that activated macrophages to potentiate the antifungal effect. In an in vivo model of IPA in immunodeficient mice, CD8+ Af-CAR T cells localized to the site of infection, engaged innate immune cells, and reduced fungal burden in the lung. Adoptive transfer of CD8+ Af-CAR T cells conferred greater antifungal efficacy compared to CD4+ Af-CAR T cells and an improvement in overall survival. Together, our study illustrates the potential of gene-engineered T cells to treat aggressive infectious diseases that are difficult to control with conventional antimicrobial therapy and support the clinical development of Af-CAR T cell therapy to treat IPA.
Collapse
Affiliation(s)
- Michelle Seif
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Tamara Katharina Kakoschke
- Klinik und Poliklinik für Mund-, Kiefer- und Gesichtschirurgie, Klinikum der Universität München, LMU, 80337 München, Germany.,Institut für Infektionsmedizin und Zoonosen, Medizinische Fakultät, LMU, 80539 München, Germany
| | - Frank Ebel
- Institut für Infektionsmedizin und Zoonosen, Medizinische Fakultät, LMU, 80539 München, Germany
| | - Marina Maria Bellet
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Nora Trinks
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum und RVZ - Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Giorgia Renga
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Marilena Pariano
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Luigina Romani
- Dipartimento di Medicina e Chirurgia, Università degli Studi di Perugia, 06132 Perugia, Italy
| | - Beeke Tappe
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - David Espie
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France.,CAR-T Cells Department, Invectys, 75013 Paris, France
| | - Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, CNRS, 75014 Paris, France.,Equipe labellisée Ligue Contre le Cancer, 75014 Paris, France
| | - Kerstin Hünniger
- Institut für Hygiene und Mikrobiologie, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany.,Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Antje Häder
- Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Markus Sauer
- Lehrstuhl für Biotechnologie und Biophysik, Biozentrum und RVZ - Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg, 97074 Würzburg, Germany
| | - Diane Damotte
- Department of Pathology, Paris Centre University Hospitals, AP-HP, 75014 Paris, France.,INSERM U1138, Cordeliers Research Center, Team Cancer, Immune Control and Escape, Paris, France; University Pierre and Marie Curie, 75006 Paris, France
| | - Marco Alifano
- Department of Thoracic Surgery, Paris Centre University Hospitals, AP-HP, Paris, France; University Paris Descartes, 75014 Paris, France
| | - P Lewis White
- Public Health Wales, Microbiology Cardiff, UHW, CF14 4XW Cardiff, UK
| | - Matthijs Backx
- Public Health Wales, Microbiology Cardiff, UHW, CF14 4XW Cardiff, UK
| | - Thomas Nerreter
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Markus Machwirth
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Oliver Kurzai
- Institut für Hygiene und Mikrobiologie, Julius-Maximilians-Universität Würzburg, 97080 Würzburg, Germany.,Fungal Septomics Research Group, Leibniz-Institut für Naturstoff-Forschung und Infektionsbiologie - Hans-Knöll-Institut (HKI), 07743 Jena, Germany
| | - Sabrina Prommersberger
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Hermann Einsele
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Michael Hudecek
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| | - Jürgen Löffler
- Medizinische Klinik und Poliklinik II und Lehrstuhl für Zelluläre Immuntherapie, Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg, 97080 Würzburg, Germany
| |
Collapse
|
38
|
Mazinani M, Rahbarizadeh F. CAR-T cell potency: from structural elements to vector backbone components. Biomark Res 2022; 10:70. [PMID: 36123710 PMCID: PMC9487061 DOI: 10.1186/s40364-022-00417-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2022] [Accepted: 09/07/2022] [Indexed: 12/03/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy, in which a patient’s own T lymphocytes are engineered to recognize and kill cancer cells, has achieved remarkable success in some hematological malignancies in preclinical and clinical trials, resulting in six FDA-approved CAR-T products currently available in the market. Once equipped with a CAR construct, T cells act as living drugs and recognize and eliminate the target tumor cells in an MHC-independent manner. In this review, we first described all structural modular of CAR in detail, focusing on more recent findings. We then pointed out behind-the-scene elements contributing to CAR expression and reviewed how CAR expression can be drastically affected by the elements embedded in the viral vector backbone.
Collapse
Affiliation(s)
- Marzieh Mazinani
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran
| | - Fatemeh Rahbarizadeh
- Department of Medical Biotechnology, Faculty of Medical Sciences, Tarbiat Modares University, P.O. Box 14115-111, Tehran, Iran. .,Research and Development Center of Biotechnology, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
39
|
Honikel MM, Olejniczak SH. Co-Stimulatory Receptor Signaling in CAR-T Cells. Biomolecules 2022; 12:biom12091303. [PMID: 36139142 PMCID: PMC9496564 DOI: 10.3390/biom12091303] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/08/2022] [Accepted: 09/13/2022] [Indexed: 01/28/2023] Open
Abstract
T cell engineering strategies have emerged as successful immunotherapeutic approaches for the treatment of human cancer. Chimeric Antigen Receptor T (CAR-T) cell therapy represents a prominent synthetic biology approach to re-direct the specificity of a patient's autologous T cells toward a desired tumor antigen. CAR-T therapy is currently FDA approved for the treatment of hematological malignancies, including subsets of B cell lymphoma, acute lymphoblastic leukemia (ALL) and multiple myeloma. Mechanistically, CAR-mediated recognition of a tumor antigen results in propagation of T cell activation signals, including a co-stimulatory signal, resulting in CAR-T cell activation, proliferation, evasion of apoptosis, and acquisition of effector functions. The importance of including a co-stimulatory domain in CARs was recognized following limited success of early iteration CAR-T cell designs lacking co-stimulation. Today, all CAR-T cells in clinical use contain either a CD28 or 4-1BB co-stimulatory domain. Preclinical investigations are exploring utility of including additional co-stimulatory molecules such as ICOS, OX40 and CD27 or various combinations of multiple co-stimulatory domains. Clinical and preclinical evidence implicates the co-stimulatory signal in several aspects of CAR-T cell therapy including response kinetics, persistence and durability, and toxicity profiles each of which impact the safety and anti-tumor efficacy of this immunotherapy. Herein we provide an overview of CAR-T cell co-stimulation by the prototypical receptors and discuss current and emerging strategies to modulate co-stimulatory signals to enhance CAR-T cell function.
Collapse
|
40
|
Bister A, Ibach T, Haist C, Gerhorst G, Smorra D, Soldierer M, Roellecke K, Wagenmann M, Scheckenbach K, Gattermann N, Wiek C, Hanenberg H. Optimized NGFR-derived hinges for rapid and efficient enrichment and detection of CAR T cells in vitro and in vivo. Mol Ther Oncolytics 2022; 26:120-134. [PMID: 35795096 PMCID: PMC9240717 DOI: 10.1016/j.omto.2022.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 05/27/2022] [Indexed: 11/25/2022] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has demonstrated unprecedented success with high remission rates for heavily pretreated patients with hematological malignancies. The hinge connecting the extracellular antigen recognition unit to the transmembrane domain provides the length and flexibility of the CAR constructs and ensures that the CAR can reach the target antigen and mediate recognition and killing of target cells. The hinge can also include specific amino acid sequences to improve CAR expression, influence T cell proliferation, and facilitate CAR T cell detection, enrichment, and even elimination. Here, we report the generation of two novel hinge domains derived from the low-affinity p75 chain of the human nerve growth factor receptor (NGFR), termed N3 and N4, which, when incorporated into the CAR backbone, allow detection as well as high-grade enrichment of CAR T cells with GMP-compatible immunomagnetic reagents. After optimizing the MACS protocol for excellent CAR T cell purity and yield, we demonstrated that N3- and N4-hinged CAR T cells are as efficacious as their CD8-hinged counterparts in vitro against hematological blasts and also in vivo in the control of acute monocytic leukemia in an immunodeficient mouse xenograft model. Thus, both hinges could potentially be an integral part of future CAR designs and universally applicable in clinical applications.
Collapse
|
41
|
Immunotherapy and immunoengineering for breast cancer; a comprehensive insight into CAR-T cell therapy advancements, challenges and prospects. Cell Oncol (Dordr) 2022; 45:755-777. [PMID: 35943716 DOI: 10.1007/s13402-022-00700-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/26/2022] [Indexed: 11/03/2022] Open
Abstract
BACKGROUND Breast cancer (BC) is a highly prevalent solid cancer with a high-rise infiltration of immune cells, turning it into a significant candidate for tumor-specific immunotherapies. Chimeric antigen receptor (CAR)-T cells are emerging as immunotherapeutic tools with genetically engineered receptors to efficiently recognize and attack tumor cells that express specific target antigens. Technological advancements in CAR design have provided five generations of CAR-T cells applicable to a wide range of cancer patients while boosting CAR-T cell therapy safety. However, CAR-T cell therapy is ineffective against breast cancer because of the loss of specified antigens, the immunosuppressive nature of the tumor and CAR-T cell-induced toxicities. Next-generation CAR-T cells actively pass through the tumor vascular barriers, persist for extended periods and disrupt the tumor microenvironment (TME) to block immune escape. CONCLUSION CAR-T cell therapy embodies advanced immunotherapy for BC, but further pre-clinical and clinical assessments are recommended to achieve maximized efficiency and safety.
Collapse
|
42
|
Developing ROR1 Targeting CAR-T Cells against Solid Tumors in Preclinical Studies. Cancers (Basel) 2022; 14:cancers14153618. [PMID: 35892876 PMCID: PMC9331269 DOI: 10.3390/cancers14153618] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/18/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells (CAR-T) have demonstrated promising clinical benefits against B-cell malignancies. Yet, its application for solid tumors is still facing challenges. Unlike haematological cancers, solid tumors often lack good targets, which are ideally expressed on the tumor cells, but not by the normal healthy cells. Fortunately, receptor tyrosine kinase-like orphan receptor 1 (ROR1) is among a few good cancer targets that is aberrantly expressed on various tumors but has a low expression on normal tissue, suggesting it as a good candidate for CAR-T therapy. Here, we constructed two ROR1 CARs with the same antigen recognition domain that was derived from Zilovertamab but differing in hinge regions. Both CARs target ROR1+ cancer cells specifically, but CAR with a shorter IgG4 hinge exhibits a higher surface expression and better in vitro functionality. We further tested the ROR1 CAR-T in three human solid tumor xenografted mouse models. Our ROR1 CAR-T cells controlled the solid tumor growth without causing any severe toxicity. Our results demonstrated that ROR1 CAR-T derived from Zilovertamab is efficacious and safe to suppress ROR1+ solid tumors in vitro and in vivo, providing a promising therapeutic option for future clinical application.
Collapse
|
43
|
McComb S, Nguyen T, Shepherd A, Henry KA, Bloemberg D, Marcil A, Maclean S, Zafer A, Gilbert R, Gadoury C, Pon RA, Sulea T, Zhu Q, Weeratna RD. Programmable Attenuation of Antigenic Sensitivity for a Nanobody-Based EGFR Chimeric Antigen Receptor Through Hinge Domain Truncation. Front Immunol 2022; 13:864868. [PMID: 35935988 PMCID: PMC9354126 DOI: 10.3389/fimmu.2022.864868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Epidermal growth factor family receptor (EGFR) is commonly overexpressed in many solid tumors and an attractive target for chimeric antigen receptor (CAR)-T therapy, but as EGFR is also expressed at lower levels in healthy tissues a therapeutic strategy must balance antigenic responsiveness against the risk of on-target off-tumor toxicity. Herein, we identify several camelid single-domain antibodies (also known as nanobodies) that are effective EGFR targeting moieties for CARs (EGFR-sdCARs) with very strong reactivity to EGFR-high and EGFR-low target cells. As a strategy to attenuate their potent antigenic sensitivity, we performed progressive truncation of the human CD8 hinge commonly used as a spacer domain in many CAR constructs. Single amino acid hinge-domain truncation progressively decreased both EGFR-sdCAR-Jurkat cell binding to EGFR-expressing targets and expression of the CD69 activation marker. Attenuated signaling in hinge-truncated EGFR-sdCAR constructs increased selectivity for antigen-dense EGFR-overexpressing cells over an EGFR-low tumor cell line or healthy donor derived EGFR-positive fibroblasts. We also provide evidence that epitope location is critical for determining hinge-domain requirement for CARs, as hinge truncation similarly decreased antigenic sensitivity of a membrane-proximal epitope targeting HER2-CAR but not a membrane-distal EGFRvIII-specific CAR. Hinge-modified EGFR-sdCAR cells showed clear functional attenuation in Jurkat-CAR-T cells and primary human CAR-T cells from multiple donors in vitro and in vivo. Overall, these results indicate that hinge length tuning provides a programmable strategy for throttling antigenic sensitivity in CARs targeting membrane-proximal epitopes, and could be employed for CAR-optimization and improved tumor selectivity.
Collapse
Affiliation(s)
- Scott McComb
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
- Centre for Infection, Immunity and Inflammation, University of Ottawa, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
- *Correspondence: Scott McComb,
| | - Tina Nguyen
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Alex Shepherd
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Kevin A. Henry
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
- Department of Biochemistry, Microbiology, and Immunology, University of Ottawa, Ottawa, ON, Canada
| | - Darin Bloemberg
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Anne Marcil
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Susanne Maclean
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Ahmed Zafer
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Rénald Gilbert
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
- Department of Bioengineering, McGill University, Montréal, QC, Canada
| | - Christine Gadoury
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Robert A. Pon
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Traian Sulea
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
- Institute of Parasitology, McGill University, Sainte-Anne-de-Bellevue, QC, Canada
| | - Qin Zhu
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| | - Risini D. Weeratna
- Human Health Therapeutics Research Centre, National Research Council, Ottawa, ON, Canada
| |
Collapse
|
44
|
Chen L, Chen F, Niu H, Li J, Pu Y, Yang C, Wang Y, Huang R, Li K, Lei Y, Huang Y. Chimeric Antigen Receptor (CAR)-T Cell Immunotherapy Against Thoracic Malignancies: Challenges and Opportunities. Front Immunol 2022; 13:871661. [PMID: 35911706 PMCID: PMC9334018 DOI: 10.3389/fimmu.2022.871661] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2022] [Accepted: 06/14/2022] [Indexed: 11/13/2022] Open
Abstract
Different from surgery, chemical therapy, radio-therapy and target therapy, Chimeric antigen receptor-modified T (CAR-T) cells, a novel adoptive immunotherapy strategy, have been used successfully against both hematological tumors and solid tumors. Although several problems have reduced engineered CAR-T cell therapeutic outcomes in clinical trials for the treatment of thoracic malignancies, including the lack of specific antigens, an immunosuppressive tumor microenvironment, a low level of CAR-T cell infiltration into tumor tissues, off-target toxicity, and other safety issues, CAR-T cell treatment is still full of bright future. In this review, we outline the basic structure and characteristics of CAR-T cells among different period, summarize the common tumor-associated antigens in clinical trials of CAR-T cell therapy for thoracic malignancies, and point out the current challenges and new strategies, aiming to provide new ideas and approaches for preclinical experiments and clinical trials of CAR-T cell therapy for thoracic malignancies.
Collapse
Affiliation(s)
- Long Chen
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Fukun Chen
- Department of Nuclear Medicine, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Huatao Niu
- Department of Neurosurgery, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Jindan Li
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yongzhu Pu
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Conghui Yang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yue Wang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Rong Huang
- Department of PET/CT Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Ke Li
- Department of Cancer Biotherapy Center, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yujie Lei
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| | - Yunchao Huang
- Department of Thoracic Surgery I, Key Laboratory of Lung Cancer of Yunnan Province, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Cancer Center of Yunnan Province, Kunming, China
| |
Collapse
|
45
|
Achkova DY, Beatson RE, Maher J. CAR T-Cell Targeting of Macrophage Colony-Stimulating Factor Receptor. Cells 2022; 11:cells11142190. [PMID: 35883636 PMCID: PMC9323367 DOI: 10.3390/cells11142190] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 07/07/2022] [Accepted: 07/12/2022] [Indexed: 12/18/2022] Open
Abstract
Macrophage colony-stimulating factor receptor (M-CSFR) is found in cells of the mononuclear phagocyte lineage and is aberrantly expressed in a range of tumours, in addition to tumour-associated macrophages. Consequently, a variety of cancer therapies directed against M-CSFR are under development. We set out to engineer chimeric antigen receptors (CARs) that employ the natural ligands of this receptor, namely M-CSF or interleukin (IL)-34, to achieve specificity for M-CSFR-expressing target cells. Both M-CSF and IL-34 bind to overlapping regions of M-CSFR, although affinity of IL-34 is significantly greater than that of M-CSF. Matched second- and third-generation CARs targeted using M-CSF or IL-34 were expressed in human T-cells using the SFG retroviral vector. We found that both M-CSF- and IL-34-containing CARs enable T-cells to mediate selective destruction of tumour cells that express enforced or endogenous M-CSFR, accompanied by production of both IL-2 and interferon (IFN)-γ. Although they contain an additional co-stimulatory module, third-generation CARs did not outperform second-generation CARs. M-CSF-containing CARs mediated enhanced cytokine production and cytolytic activity compared to IL-34-containing CARs. These data demonstrate the feasibility of targeting M-CSFR using ligand-based CARs and raise the possibility that the low picomolar affinity of IL-34 for M-CSFR is detrimental to CAR function.
Collapse
Affiliation(s)
- Daniela Yordanova Achkova
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK; (D.Y.A.); (R.E.B.)
| | - Richard Esmond Beatson
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK; (D.Y.A.); (R.E.B.)
| | - John Maher
- CAR Mechanics Group, Guy’s Cancer Centre, School of Cancer and Pharmaceutical Sciences, King’s College London, Great Maze Pond, London SE1 9RT, UK; (D.Y.A.); (R.E.B.)
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne BN21 2UD, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
- Correspondence: ; Tel.: +44-(0)207188-1468
| |
Collapse
|
46
|
Gao TA, Chen YY. Engineering Next-Generation CAR-T Cells: Overcoming Tumor Hypoxia and Metabolism. Annu Rev Chem Biomol Eng 2022; 13:193-216. [PMID: 35700528 DOI: 10.1146/annurev-chembioeng-092120-092914] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
T cells engineered to express chimeric antigen receptors (CARs) have shown remarkable success in treating B-cell malignancies, reflected by multiple US Food and Drug Administration-approved CAR-T cell products currently on the market. However, various obstacles have thus far limited the use of approved products and constrained the efficacy of CAR-T cell therapy against solid tumors. Overcoming these obstacles will necessitate multidimensional CAR-T cell engineering approaches and better understanding of the intricate tumor microenvironment (TME). Key challenges include treatment-related toxicity, antigen escape and heterogeneity, and the highly immunosuppressive profile of the TME. Notably, the hypoxic and nutrient-deprived nature of the TME severely attenuates CAR-T cell fitness and efficacy, highlighting the need for more sophisticated engineering strategies. In this review, we examine recent advances in protein- and cell-engineering strategies to improve CAR-T cell safety and efficacy, with an emphasis on overcoming immunosuppression induced by tumor metabolism and hypoxia.
Collapse
Affiliation(s)
- Torahito A Gao
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California, USA; ,
| | - Yvonne Y Chen
- Department of Chemical and Biomolecular Engineering, University of California, Los Angeles, California, USA; , .,Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, California, USA.,Parker Institute for Cancer Immunotherapy Center at UCLA, Los Angeles, California, USA
| |
Collapse
|
47
|
Larcombe-Young D, Whilding L, Davies DM, Draper B, Bechman N, Maher J. Generation of human parallel chimeric antigen receptor (pCAR) T cells to achieve synergistic T cell co-stimulation. STAR Protoc 2022; 3:101414. [PMID: 35620078 PMCID: PMC9127424 DOI: 10.1016/j.xpro.2022.101414] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Dual co-stimulation may be harnessed using parallel chimeric antigen receptors (pCARs) in which two distinct co-stimulatory units are adjacently localized on the plasma membrane. This protocol summarizes construct design, human T cell isolation, retroviral transduction, tissue culture expansion, and preclinical testing of pCAR T cells, exemplified by receptors that co-target avb6 integrin and ErbB dimers. For complete details on the use and execution of this protocol, please refer to Muliaditan et al. (2021). A protocol to enable robust generation of pCAR T cells Effective dual co-stimulation is provided by this platform Sustained and enhanced antitumor activity by pCAR T cells Detailed in vitro and in vivo evaluation of pCAR technology
Publisher’s note: Undertaking any experimental protocol requires adherence to local institutional guidelines for laboratory safety and ethics.
Collapse
Affiliation(s)
- Daniel Larcombe-Young
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Campus, London SE1 9RT, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Lynsey Whilding
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Campus, London SE1 9RT, UK
| | - David Marc Davies
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Campus, London SE1 9RT, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
| | - Benjamin Draper
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Campus, London SE1 9RT, UK
| | - Natasha Bechman
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Campus, London SE1 9RT, UK
| | - John Maher
- School of Cancer and Pharmaceutical Sciences, King’s College London, Faculty of Life Sciences and Medicine, Guy’s Campus, London SE1 9RT, UK
- Leucid Bio Ltd., Guy’s Hospital, Great Maze Pond, London SE1 9RT, UK
- Department of Immunology, Eastbourne Hospital, Kings Drive, Eastbourne, East Sussex BN21 2UD, UK
- Corresponding author
| |
Collapse
|
48
|
Labanieh L, Majzner RG, Klysz D, Sotillo E, Fisher CJ, Vilches-Moure JG, Pacheco KZB, Malipatlolla M, Xu P, Hui JH, Murty T, Theruvath J, Mehta N, Yamada-Hunter SA, Weber EW, Heitzeneder S, Parker KR, Satpathy AT, Chang HY, Lin MZ, Cochran JR, Mackall CL. Enhanced safety and efficacy of protease-regulated CAR-T cell receptors. Cell 2022; 185:1745-1763.e22. [PMID: 35483375 PMCID: PMC9467936 DOI: 10.1016/j.cell.2022.03.041] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/04/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
Regulatable CAR platforms could circumvent toxicities associated with CAR-T therapy, but existing systems have shortcomings including leakiness and attenuated activity. Here, we present SNIP CARs, a protease-based platform for regulating CAR activity using an FDA-approved small molecule. Design iterations yielded CAR-T cells that manifest full functional capacity with drug and no leaky activity in the absence of drug. In numerous models, SNIP CAR-T cells were more potent than constitutive CAR-T cells and showed diminished T cell exhaustion and greater stemness. In a ROR1-based CAR lethality model, drug cessation following toxicity onset reversed toxicity, thereby credentialing the platform as a safety switch. In the same model, reduced drug dosing opened a therapeutic window that resulted in tumor eradication in the absence of toxicity. SNIP CARs enable remote tuning of CAR activity, which provides solutions to safety and efficacy barriers that are currently limiting progress in using CAR-T cells to treat solid tumors.
Collapse
Affiliation(s)
- Louai Labanieh
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robbie G Majzner
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dorota Klysz
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chris J Fisher
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - José G Vilches-Moure
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kaithlen Zen B Pacheco
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Meena Malipatlolla
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peng Xu
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jessica H Hui
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tara Murty
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Biophysics Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johanna Theruvath
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nishant Mehta
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Sean A Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Evan W Weber
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sabine Heitzeneder
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin R Parker
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Michael Z Lin
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
49
|
Feng Z, He X, Zhang X, Wu Y, Xing B, Knowles A, Shan Q, Miller S, Hojnacki T, Ma J, Katona BW, Gade TPF, Siegel DL, Schrader J, Metz DC, June CH, Hua X. Potent suppression of neuroendocrine tumors and gastrointestinal cancers by CDH17CAR T cells without toxicity to normal tissues. NATURE CANCER 2022; 3:581-594. [PMID: 35314826 DOI: 10.1038/s43018-022-00344-7] [Citation(s) in RCA: 39] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 02/09/2022] [Indexed: 12/15/2022]
Abstract
Gastrointestinal cancers (GICs) and neuroendocrine tumors (NETs) are often refractory to therapy after metastasis. Adoptive cell therapy using chimeric antigen receptor (CAR) T cells, though remarkably efficacious for treating leukemia, is yet to be developed for solid tumors such as GICs and NETs. Here we isolated a llama-derived nanobody, VHH1, and found that it bound cell surface adhesion protein CDH17 upregulated in GICs and NETs. VHH1-CAR T cells (CDH17CARTs) killed both human and mouse tumor cells in a CDH17-dependent manner. CDH17CARTs eradicated CDH17-expressing NETs and gastric, pancreatic and colorectal cancers in either tumor xenograft or autochthonous mouse models. Notably, CDH17CARTs do not attack normal intestinal epithelial cells, which also express CDH17, to cause toxicity, likely because CDH17 is localized only at the tight junction between normal intestinal epithelial cells. Thus, CDH17 represents a class of previously unappreciated tumor-associated antigens that is 'masked' in healthy tissues from attack by CAR T cells for developing safer cancer immunotherapy.
Collapse
Affiliation(s)
- Zijie Feng
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xin He
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xuyao Zhang
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Yuan Wu
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bowen Xing
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Alison Knowles
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Qiaonan Shan
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Samuel Miller
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Taylor Hojnacki
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jian Ma
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Bryson W Katona
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Division of Gastroenterology and Hepatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Terence P F Gade
- Department of Radiology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Don L Siegel
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Jörg Schrader
- Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - David C Metz
- Division of Gastroenterology and Hepatology, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | - Xianxin Hua
- Department of Cancer Biology, Abramson Family Cancer Research Institute, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
50
|
López-Cantillo G, Urueña C, Camacho BA, Ramírez-Segura C. CAR-T Cell Performance: How to Improve Their Persistence? Front Immunol 2022; 13:878209. [PMID: 35572525 PMCID: PMC9097681 DOI: 10.3389/fimmu.2022.878209] [Citation(s) in RCA: 63] [Impact Index Per Article: 31.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Accepted: 03/25/2022] [Indexed: 01/07/2023] Open
Abstract
Adoptive cell therapy with T cells reprogrammed to express chimeric antigen receptors (CAR-T cells) has been highly successful in patients with hematological neoplasms. However, its therapeutic benefits have been limited in solid tumor cases. Even those patients who respond to this immunotherapy remain at risk of relapse due to the short-term persistence or non-expansion of CAR-T cells; moreover, the hostile tumor microenvironment (TME) leads to the dysfunction of these cells after reinfusion. Some research has shown that, in adoptive T-cell therapies, the presence of less differentiated T-cell subsets within the infusion product is associated with better clinical outcomes. Naive and memory T cells persist longer and exhibit greater antitumor activity than effector T cells. Therefore, new methods are being studied to overcome the limitations of this therapy to generate CAR-T cells with these ideal phenotypes. In this paper, we review the characteristics of T-cell subsets and their implications in the clinical outcomes of adoptive therapy with CAR-T cells. In addition, we describe some strategies developed to overcome the reduced persistence of CAR T-cells and alternatives to improve this therapy by increasing the expansion ability and longevity of modified T cells. These methods include cell culture optimization, incorporating homeostatic cytokines during the expansion phase of manufacturing, modulation of CAR-T cell metabolism, manipulating signaling pathways involved in T-cell differentiation, and strategies related to CAR construct designs.
Collapse
Affiliation(s)
- Gina López-Cantillo
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud (IDCBIS), Bogotá, Colombia
| | - Claudia Urueña
- Grupo de Inmunobiología y Biología Celular, Facultad de Ciencias, Pontificia Universidad Javeriana, Bogotá, Colombia
| | | | - Cesar Ramírez-Segura
- Laboratorio de Investigación en Ingeniería Celular y Molecular, Instituto Distrital de Ciencia Biotecnología e Innovación en Salud (IDCBIS), Bogotá, Colombia
- Instituto Distrital de Ciencia Biotecnología e Innovación en Salud (IDCBIS), Bogotá, Colombia
| |
Collapse
|