1
|
Wu A, Zhang T, Yu H, Cao Y, Zhang R, Shao R, Liu B, Chen L, Xu K, Chen W, Ho J, Shi X. Mechanisms underlying resistance to CAR-T cell therapy and strategies for enhancement. Cytokine Growth Factor Rev 2025; 83:66-76. [PMID: 40340171 DOI: 10.1016/j.cytogfr.2025.04.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2025] [Revised: 04/19/2025] [Accepted: 04/19/2025] [Indexed: 05/10/2025]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy has emerged as a revolutionary approach in the treatment of hematological malignancies, including acute lymphoblastic leukemia, B-cell lymphoma, and multiple myeloma. Despite its promise, the clinical efficacy is often hampered by transient efficacy and subsequent relapse, which curtail the long-term success of this treatment. Current research focuses on overcoming these obstacles by exploring multitarget strategies and optimizing CAR-T cell design. This review summarizes recent insights into the resistance mechanisms associated with CAR-T cell therapy, and delineates emerging strategies for optimized CAR construction, including targeting multiple antigens, improving CAR design, and enhancing T-cell persistence. The goal is to provide a comprehensive overview of the field's current landscape to guide future research and the clinical application of CAR-T cell therapies.
Collapse
Affiliation(s)
- Anran Wu
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; The Second Clinical Medical School of Nanjing Medical University, Nanjing 210011, China
| | - Tingying Zhang
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; The Second Clinical Medical School of Nanjing Medical University, Nanjing 210011, China
| | - Hongkai Yu
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; The Second Clinical Medical School of Nanjing Medical University, Nanjing 210011, China
| | - Yuyue Cao
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; The Second Clinical Medical School of Nanjing Medical University, Nanjing 210011, China
| | - Rui Zhang
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; The Second Clinical Medical School of Nanjing Medical University, Nanjing 210011, China
| | - Ruonan Shao
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China
| | - Bofeng Liu
- School of Life Science, Beijing Institute of Technology, Beijing 100081, China
| | - Liting Chen
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kailin Xu
- Department of Hematology, Blood Diseases Institute, Key Laboratory of Bone Marrow Stem Cells, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, China
| | - Wei Chen
- Department of Hematology, Blood Diseases Institute, Key Laboratory of Bone Marrow Stem Cells, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu 221004, China.
| | - Jinyuan Ho
- Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, Jiangsu 215123, China.
| | - Xiaofeng Shi
- Department of Hematology, the Second Affiliated Hospital of Nanjing Medical University, Nanjing 210003, China; The Second Clinical Medical School of Nanjing Medical University, Nanjing 210011, China.
| |
Collapse
|
2
|
Zhao Z, Li M, Zheng X, Gao P, Huang C, Yu Q, Jin L, Zhang L, Zhu D, Li F. mRNA lipid nanoparticles in CAR-T therapy: a novel strategy to improve efficacy. NANOTECHNOLOGY 2025; 36:222003. [PMID: 40376884 DOI: 10.1088/1361-6528/add482] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Accepted: 05/06/2025] [Indexed: 05/18/2025]
Abstract
Chimeric antigen receptor T cells (CAR-T) immunotherapy has achieved remarkable progress in the treatment of hematological malignancies. However, it encounters challenges including complex manufacturing processes, high cost, and safety issues. Lipid nanoparticle (LNP) technology, as an advanced gene delivery platform, offers significant advancements to CAR-T therapy through its high efficiency, low immunogenicity, and safety. LNP enablein vivoproduction of CAR-T cells, thereby improving delivery efficiency, reducing the risks of immunogenicity and insertional mutations, simplifying the production process and reducing costs. The scalability and rapid optimization ability of LNP position them as promising candidates for CAR-T cell production. LNP technology is expected to further promote the development of CAR-T immunotherapy and provide safer and more economical treatment options. Therefore, this paper aims to provide a comprehensive and systematic review of the application of LNP in CAR-T therapy. In this review, we initially outline the fundamental design, process, and current challenges of CAR-T therapy. Subsequently, we present the characteristics of LNP, their advantages as a gene delivery vectors, and how they improve the efficacy of CAR-T therapy. Finally, we summarize the current research landscape of LNP applications in CAR-T therapy. This includes enhancingin vitrotransfection of T cells, programming T cellsin situ, facilitating T-cell activation, alleviating the side effects of CAR-T therapy, and combining CAR-T therapy with other immunotherapies. These advancements will aid in the design of mRNA delivery systems based on LNP, thereby promoting the development of CAR-T therapy.
Collapse
Affiliation(s)
- Zengkai Zhao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Mingmei Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Xiang Zheng
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Pengli Gao
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Chenlu Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Qingyu Yu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Limin Jin
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Linhua Zhang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Dunwan Zhu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| | - Fangzhou Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Biomedical Materials, Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, People's Republic of China
| |
Collapse
|
3
|
Muthuvel M, Ganapathy T, Spencer T, Raikar SS, Thangavel S, Srivastava A, Martin S. Engineering safe anti-CD19-CD28ζ CAR T cells with CD8a hinge domain in serum-free media for adoptive immunotherapy. Front Immunol 2025; 16:1545549. [PMID: 40416968 PMCID: PMC12098533 DOI: 10.3389/fimmu.2025.1545549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/08/2025] [Indexed: 05/27/2025] Open
Abstract
Background Despite the curative potential, high cost of manufacturing and the toxicities limits the wider access of Chimeric Antigen Receptor (CAR) T cell therapy in global medicine. CARs are modular synthetic antigen receptors integrating the single-chain variable fragment (scFv) of an immunoglobulin molecule to the TCR signaling. CARs allow HLA independent, T cell mediated destruction of tumor cells independent of tumor associated-HLA downregulation and survive within the patient as 'living drug.' Here we report a safer approach for engineering alpha beta T cells with anti- CD19-CD28ζ CAR using self-inactivating (SIN) lentiviral vectors for adoptive immunotherapy. Method αβ T cells from the peripheral blood (PB) were lentivirally transduced with CAR construct containing hinge domain from CD8α, transmembrane and co-stimulatory domain from CD28 along with signaling domain from CD3ζ and driven by human UBC promoter. The cells were pre-stimulated through CD3/CD28 beads before lentiviral transduction. Transduction efficiency, fold expansion and phenotype were monitored for the CAR T cells expanded for 10-12 days. The antigen-specific tumor-killing capacity of CD19 CAR T cells was assessed against a standard CD19 expressing NALM6 cell lines with a flow cytometry-based assay optimized in the lab. Results and conclusion We have generated high titer lentiviral vectors of CAR with a titer of 9.85 ± 2.2×107 TU/ml (mean ± SEM; n=9) generating a transduction efficiency of 27.57 ± 2.4%. (n=7) at an MOI of 10 in total T cells. The product got higher CD8+ to CD4+ CAR T cell ratio with preponderance of an effector memory phenotype on day 07 and day 12. The CAR-T cells expanded (148.4 ± 29 fold; n=7) in serum free media with very high viability (87.8 ± 2.2%; n=7) on day 12. The antitumor functions of CD19 CAR T cells as gauged against percentage lysis of NALM6 cells at a 1:1 ratio is 27.68 ± 6.87% drawing up to the release criteria. CAR T cells produced IFNγ (11.23 ± 1.5%; n=6) and degranulation marker CD107α (34.82 ± 2.08%; n=5) in an antigen-specific manner. Furthermore, the sequences of WPRE, GFP, and P2A were removed from the CAR construct to enhance safety. These CAR T cells expanded up to 21.7 ± 5.53 fold with 82.7±5.43% viability (n=4). Conclusion We have generated, validated, and characterized a reproducible indigenous workflow for generating anti-CD19 CAR T cells in vitro. This approach can be used for targeting cancer and autoimmune diseases in which CD19+ B lineage cells cause host damage.
Collapse
Affiliation(s)
- Muthuganesh Muthuvel
- Laboratory of Synthetic Immunology, Cancer Research Division, Biotechnology Research Innovation Council- Rajiv Gandhi Centre for Biotechnology (BRIC - RGCB), Department of Biotechnology, Thiruvananthapuram, India
- Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, India
| | - Thamizhselvi Ganapathy
- Center for Stem Cell Research (CSCR), Christian Medical College (CMC) Vellore, Velllore, India
| | - Trent Spencer
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, United States
| | - Sunil S. Raikar
- Cell and Gene Therapy Program, Aflac Cancer and Blood Disorders Center, Department of Pediatrics, Children’s Healthcare of Atlanta and Emory University School of Medicine, Atlanta, GA, United States
| | | | - Alok Srivastava
- Center for Stem Cell Research (CSCR), Christian Medical College (CMC) Vellore, Velllore, India
- Department of Hematology, Christian Medical College (CMC) Vellore, Velllore, India
- Haematology Research Unit, St. John’s Research Institute, St. John’s National Academy of Health Sciences, Bengaluru, Karnataka, India
| | - Sunil Martin
- Laboratory of Synthetic Immunology, Cancer Research Division, Biotechnology Research Innovation Council- Rajiv Gandhi Centre for Biotechnology (BRIC - RGCB), Department of Biotechnology, Thiruvananthapuram, India
| |
Collapse
|
4
|
Mulvey A, Trueb L, Coukos G, Arber C. Novel strategies to manage CAR-T cell toxicity. Nat Rev Drug Discov 2025; 24:379-397. [PMID: 39901030 DOI: 10.1038/s41573-024-01100-5] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 02/05/2025]
Abstract
The immune-related adverse events associated with chimeric antigen receptor (CAR)-T cell therapy result in substantial morbidity as well as considerable cost to the health-care system, and can limit the use of these treatments. Current therapeutic strategies to manage immune-related adverse events include interleukin-6 receptor (IL-6R) blockade and corticosteroids. However, because these interventions do not always address the side effects, nor prevent progression to higher grades of adverse events, new approaches are needed. A deeper understanding of the cell types involved, and their associated signalling pathways, cellular metabolism and differentiation states, should provide the basis for alternative strategies. To preserve treatment efficacy, cytokine-mediated toxicity needs to be uncoupled from CAR-T cell function, expansion, long-term persistence and memory formation. This may be achieved by targeting CAR or independent cytokine signalling axes transiently, and through novel T cell engineering strategies, such as low-affinity CAR-T cells, reversible on-off switches and versatile adaptor systems. We summarize the current management of cytokine release syndrome and immune effector cell-associated neurotoxicity syndrome, and review T cell- and myeloid cell-intrinsic druggable targets and cellular engineering strategies to develop safer CAR-T cells.
Collapse
Affiliation(s)
- Arthur Mulvey
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Lionel Trueb
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
| | - George Coukos
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland
| | - Caroline Arber
- Department of Oncology UNIL-CHUV, Service of Immuno-Oncology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
- Ludwig Institute for Cancer Research Lausanne, Lausanne, Switzerland.
- Departments of Oncology UNIL-CHUV and Laboratory Medicine and Pathology, Service and Central Laboratory of Hematology, Lausanne University Hospital (CHUV) and University of Lausanne (UNIL), Lausanne, Switzerland.
| |
Collapse
|
5
|
Zhao T, You J, Wang C, Li B, Liu Y, Shao M, Zhao W, Zhou C. Cell-based immunotherapies for solid tumors: advances, challenges, and future directions. Front Oncol 2025; 15:1551583. [PMID: 40356763 PMCID: PMC12066282 DOI: 10.3389/fonc.2025.1551583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Cell-based immunotherapies, including CAR-T, CAR-NK, and TCR-T therapies, represent a transformative approach to cancer treatment by offering precise targeting of tumor cells. Despite their success in hematologic malignancies, these therapies encounter significant challenges in treating solid tumors, such as antigen heterogeneity, immunosuppressive tumor microenvironments, limited cellular infiltration, off-target toxicity, and difficulties in manufacturing scalability. CAR-T cells have demonstrated exceptional efficacy in blood cancers but face obstacles in solid tumors, whereas CAR-NK cells offer reduced graft-versus-host disease but encounter similar barriers. TCR-T cells expand the range of treatable cancers by targeting intracellular antigens but require meticulous antigen selection to prevent off-target effects. Alternative therapies like TIL, NK, and CIK cells show promise but require further optimization to enhance persistence and overcome immunosuppressive barriers. Manufacturing complexity, high costs, and ensuring safety and efficacy remain critical challenges. Future advancements in gene editing, multi-antigen targeting, synthetic biology, off-the-shelf products, and personalized medicine hold the potential to address these issues and expand the use of cell-based therapies. Continued research and innovation are essential to improving safety, efficacy, and scalability, ultimately leading to better patient outcomes.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Jinping You
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Congyue Wang
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Bo Li
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Yuhan Liu
- Department of Medical Oncology, Anshan Cancer Hospital, Anshan, China
| | - Mingjia Shao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Wuyang Zhao
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| | - Chuang Zhou
- Department of Oncology, Ansteel Group General Hospital, Anshan, China
| |
Collapse
|
6
|
Teng X, Li S, Zhang C, Ding H, Tian Z, Zhu Y, Liu T, Zhang G, Sun K, Xie H, Tu J, Lu Z. NKG2D/CD28 chimeric receptor boosts cytotoxicity and durability of CAR-T cells for solid and hematological tumors. Exp Hematol Oncol 2025; 14:52. [PMID: 40181405 PMCID: PMC11967049 DOI: 10.1186/s40164-025-00646-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 03/21/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND CAR-T cell therapy faces challenges in solid tumor treatment and hematologic malignancy relapse, among which the limited persistence of CAR-T cells and target antigen downregulation are prominent factors. Therefore, we engineered an NKG2D/CD28 chimeric co-stimulatory receptor (CCR), leveraging its broad ligand expression on tumors to enhance the antitumor activity of MSLN CAR and CD19 CAR-T cells. METHODS We generated MSLN CAR-T and CD19 CAR-T cells co-expressing the NKG2D/CD28 CCR and assessed their antitumor efficacy in vitro and in vivo. CAR-T cell activation, differentiation, and exhaustion were analyzed over time following tumor antigen stimulation. Furthermore, a chronic antigen stimulation model was established using tumor cells with low antigen density to simulate the sustained antigenic pressure encountered in vivo treatment conditions. RESULTS Our study shows that NKG2D/CD28&CAR-T cells exhibit enhanced cytotoxicity against tumor cells, especially those with low antigen density, both in vitro and in vivo. Compared to conventional second-generation MSLN CAR or CD19 CAR-T cells, these dual-targeted NKG2D/CD28&CAR-T cells demonstrate superior sensitivity in recognizing and lysing low-density antigen-expressing lung cancer and leukemia cells, and they are capable of eradicating tumors with low-density antigen expression in vivo. Furthermore, the complementary co-stimulation provided by the 4-1BB and CD28 intracellular domains in the CAR and NKG2D/CD28 promotes cytokine secretion, reduces CAR-T cell exhaustion, and enhances the in vivo persistence of CAR-T cells, significantly improving their antitumor efficacy. CONCLUSION The combination of CAR and NKG2D/CD28 offers a potent strategy to enhance the cytotoxicity and durability of CAR-T cells. This approach is promising for improving therapeutic outcomes in solid and hematological tumors and preventing recurrence in tumors with low target antigen density.
Collapse
Affiliation(s)
- Xia Teng
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Shance Li
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Chaoting Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huirong Ding
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zhihua Tian
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Central Laboratory, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Yuge Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Thoracic Surgery II, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Ting Liu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Guanyu Zhang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Kang Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Huimin Xie
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Jiaxin Tu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Zheming Lu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Laboratory of Biochemistry and Molecular Biology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
7
|
Li S, Zhou Y, Wang H, Qu G, Zhao X, Wang X, Hou R, Guan Z, Liu D, Zheng J, Shi M. Advances in CAR optimization strategies based on CD28. Front Immunol 2025; 16:1548772. [PMID: 40181986 PMCID: PMC11966486 DOI: 10.3389/fimmu.2025.1548772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 02/26/2025] [Indexed: 04/05/2025] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy, which utilizes genetic engineering techniques to modify T-cells to achieve specific targeting of cancer cells, has made significant breakthroughs in cancer treatment in recent years. All marketed CAR-T products are second-generation CAR-T cells containing co-stimulatory structural domains, and co-stimulatory molecules are critical for CAR-T cell activation and function. Although CD28-based co-stimulatory molecules have demonstrated potent cytotoxicity in the clinical application of CAR-T cells, they still suffer from high post-treatment relapse rates, poor efficacy durability, and accompanying severe adverse reactions. In recent years, researchers have achieved specific results in enhancing the anti-tumor function of CD28 by mutating its signaling motifs, combining the co-stimulatory structural domains, and modifying other CAR components besides co-stimulation. This paper reviewed the characteristics and roles of CD28 in CAR-T cell-mediated anti-tumor signaling and activation. We explored potential strategies to enhance CAR-T cell efficacy and reduce side effects by optimizing CD28 motifs and CAR structures, aiming to provide a theoretical basis for further clinical CAR-T cell therapy development.
Collapse
Affiliation(s)
- Sijin Li
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Yusi Zhou
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Hairong Wang
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Gexi Qu
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Xuan Zhao
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Xu Wang
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Rui Hou
- College of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Zhangchun Guan
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Dan Liu
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| | - Ming Shi
- Country Cancer Institute, Xuzhou Medical University, Xuzhou, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, China
| |
Collapse
|
8
|
Li R, Grosskopf AK, Joslyn LR, Stefanich EG, Shivva V. Cellular Kinetics and Biodistribution of Adoptive T Cell Therapies: from Biological Principles to Effects on Patient Outcomes. AAPS J 2025; 27:55. [PMID: 40032717 DOI: 10.1208/s12248-025-01017-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/06/2025] [Indexed: 03/05/2025] Open
Abstract
Cell-based immunotherapy has revolutionized cancer treatment in recent years and is rapidly expanding as one of the major therapeutic options in immuno-oncology. So far ten adoptive T cell therapies (TCTs) have been approved by the health authorities for cancer treatment, and they have shown remarkable anti-tumor efficacy with potent and durable responses. While adoptive T cell therapies have shown success in treating hematological malignancies, they are lagging behind in establishing promising efficacy in treating solid tumors, partially due to our incomplete understanding of the cellular kinetics (CK) and biodistribution (including tumoral penetration) of cell therapy products. Indeed, recent clinical studies have provided ample evidence that CK of TCTs can influence clinical outcomes in both hematological malignancies and solid tumors. In this review, we will discuss the current knowledge on the CK and biodistribution of anti-tumor TCTs. We will first describe the typical CK and biodistribution characteristics of these "living" drugs, and the biological factors that influence these characteristics. We will then review the relationships between CK and pharmacological responses of TCT, and potential strategies in enhancing the persistence and tumoral penetration of TCTs in the clinic. Finally, we will also summarize bioanalytical methods, preclinical in vitro and in vivo tools, and in silico modeling approaches used to assess the CK and biodistribution of TCTs.
Collapse
Affiliation(s)
- Ran Li
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| | - Abigail K Grosskopf
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Louis R Joslyn
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Eric Gary Stefanich
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA
| | - Vittal Shivva
- Translational Pharmacokinetics and Pharmacodynamics, Genentech Inc, 1 DNA Way, South San Francisco, California, 94080, USA.
| |
Collapse
|
9
|
Losi G, Mussetti A, Peña M, Lopez-Pereira P, Sureda A, Novelli S. Anti-CD19 CAR T-cell therapy for primary and secondary CNS lymphomas. Bone Marrow Transplant 2025; 60:259-269. [PMID: 39674850 DOI: 10.1038/s41409-024-02496-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/16/2024]
Abstract
Central nervous system lymphomas (CNSL) are a heterogeneous group of generally aggressive tumors whose prognosis varies significantly, being more favorable in patients with primary disease and poorer in those with secondary lymphoma. Current treatments typically involve intensive chemotherapy followed by consolidation with autologous stem cell transplantation or whole-brain radiotherapy. However, if the disease relapses, there is no established standard of care. The recent approval of anti-CD19 chimeric antigen receptor (CAR) T-cell therapy for systemic B-cell lymphomas has shifted the treatment landscape for previously incurable patients. Even though this therapy was initially underexplored in the setting of CNSL due to safety and efficacy concerns, it could offer a new therapeutic avenue for these patients. In this review, we will provide a concise overview of the current treatment strategies for CNSL, highlighting their key limitations, including relapse rates and long-term toxicity. Following this, we will explore the most important studies and clinical trials on CNSL, focusing on recent advancements in anti-CD19 CAR T-cell therapy. This comprehensive analysis will offer insights into the successes and challenges of treating CNSL effectively.
Collapse
Affiliation(s)
- Giulia Losi
- Clinical Hematology Department, Institut Català d'Oncologia-L' Hospitalet, IDIBELL, Barcelona, Spain.
- Department of Molecular Medicine, University of Pavia, Pavia, Italy.
| | - Alberto Mussetti
- Clinical Hematology Department, Institut Català d'Oncologia-L' Hospitalet, IDIBELL, Barcelona, Spain
| | - Marta Peña
- Clinical Hematology Department, Institut Català d'Oncologia-L' Hospitalet, IDIBELL, Barcelona, Spain
| | - Patricia Lopez-Pereira
- Clinical Hematology Department, Institut Català d'Oncologia-L' Hospitalet, IDIBELL, Barcelona, Spain
| | - Anna Sureda
- Clinical Hematology Department, Institut Català d'Oncologia-L' Hospitalet, IDIBELL, Barcelona, Spain
| | - Silvana Novelli
- Clinical Hematology Department, Institut Català d'Oncologia-L' Hospitalet, IDIBELL, Barcelona, Spain.
| |
Collapse
|
10
|
Lu Y, Ma N, Cheng K, Liu G, Liang J, Xu C, Li D, Cao C, Gao X, Chen L, Wang X, Wang Y, Zhao X, Jiang K. An OMV-Based Nanovaccine as Antigen Presentation Signal Enhancer for Cancer Immunotherapy. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2413392. [PMID: 39811977 DOI: 10.1002/adma.202413392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/07/2025] [Indexed: 01/16/2025]
Abstract
Antigen-presenting cells (APCs) process tumor vaccines and present tumor antigens as the first signals to T cells to activate anti-tumor immunity, which process requires the assistance of co-stimulatory second signals on APCs. The immune checkpoint programmed death ligand 1 (PD-L1) not only mediates the immune escape of tumor cells but also acts as a co-inhibitory second signal on APCs. The serious dysfunction of second signals due to the high expression of PD-L1 on APCs in the tumor body results in the inefficiency of tumor vaccines. To overcome this challenge, a previously established Plug-and-Display tumor vaccine platform based on bacterial outer membrane vesicles (OMVs) is developed into an "Antigen Presentation Signal Enhancer" (APSE) by surface-modifying PD-L1 antibodies (αPD-L1). While delivering tumor antigens, APSE can activate the expression of co-stimulatory second signals in APCs due to the high immunogenicity of OMVs. More importantly, the surface-modified αPD-L1 binds to the co-inhibitory signals PD-L1, potentially restoring CD80 function and ensuring efficient co-stimulatory second signals and activation of anti-tumor immunity. The results reveal the importance of PD-L1 blockage in the initiation process of anti-tumor immunity, and the second signal modulation capability of APSE can expand the application potential of cancer vaccines to less immunogenic malignancies.
Collapse
Affiliation(s)
- Yichao Lu
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Nana Ma
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Keman Cheng
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Guangna Liu
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Jie Liang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chen Xu
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, China
| | - Danrui Li
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Cheng Cao
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Xiaoyu Gao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Liting Chen
- Department of Surgical Oncology and General Surgery, Key Laboratory of Precision Diagnosis and Treatment of Gastrointestinal Tumors, Ministry of Education, The First Affiliated Hospital of China Medical University, 155 North Nanjing Street, Shenyang, 110001, China
| | - Xinwei Wang
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yazhou Wang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| | - Xiao Zhao
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology, No.11 Zhongguancun Beiyitiao, Beijing, 100190, China
- IGDB-NCNST Joint Research Center, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Kuirong Jiang
- Pancreas Center, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, Jiangsu Province, 210029, China
| |
Collapse
|
11
|
Zhang X, Xu K, Gale RP, Pan B. Strategies following failure of CAR-T-cell therapy in non-Hodgkin lymphoma. Bone Marrow Transplant 2025; 60:182-190. [PMID: 39533016 DOI: 10.1038/s41409-024-02463-4] [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: 07/04/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024]
Abstract
Several CD19 CAR-T-cell drugs are approved for safety and efficacy in advanced B-cell cancers with encouraging results. However, primary refractory and relapse are common. We critically analyze long-term data on efficacy of CD19 CAR-T-cell therapies in B-cell non-Hodgkin lymphomas from clinical trials with those of so-called real world data. We identify co-variates associated with efficacy, discuss mechanisms of relapse, summarize the data on the results of post-failure therapy including allotransplants, monoclonal and bi-specific antibodies, antibody-drug conjugates, immune checkpoint-inhibitors and repeat infusions of CAR-T-cells. We conclude, save for allotransplants, there are few data strongly supporting any of these interventions. Most trial are with few heterogeneously-treated subjects with diverse interventions and brief follow-up. Interventions need to be tailored to the cause(s) of CAR-T-cell failure. Prestly, there is not a convincingly safe and effective therapy of people failing initial CAR-T-cell therapy of B-cell non-Hodgkin lymphoma.
Collapse
Affiliation(s)
- Xiaotian Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China
| | - Kailin Xu
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| | - Robert Peter Gale
- Centre for Haematology, Department of Immunology and Inflammation, Imperial College London, London, England
| | - Bin Pan
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, China.
- Blood Diseases Institute, Xuzhou Medical University, Xuzhou, China.
| |
Collapse
|
12
|
Hu Q, Xuan J, Wang L, Shen K, Gao Z, Zhou Y, Wei C, Gu J. Application of adoptive cell therapy in malignant melanoma. J Transl Med 2025; 23:102. [PMID: 39844295 PMCID: PMC11752767 DOI: 10.1186/s12967-025-06093-2] [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: 11/05/2024] [Accepted: 01/07/2025] [Indexed: 01/24/2025] Open
Abstract
Cutaneous melanoma is one of the most aggressive skin cancers originating from skin pigment cells. Patients with advanced melanoma suffer a poor prognosis and generally cannot benefit well from surgical resection and chemo/target therapy due to metastasis and drug resistance. Thus, adoptive cell therapy (ACT), employing immune cells with specific tumor-recognizing receptors, has emerged as a promising therapeutic approach to display on-tumor toxicity. This review discusses the application, efficacy, limitations, as well as future prospects of four commonly utilized approaches -including tumor-infiltrating lymphocytes, chimeric antigen receptor (CAR) T cell, engineered T-cell receptor T cells, and chimeric antigen receptor NK cells- in the context of malignant melanoma.
Collapse
Affiliation(s)
- Qianrong Hu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Jiangying Xuan
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Lu Wang
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Kangjie Shen
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Zixu Gao
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China
| | - Yuhong Zhou
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chuanyuan Wei
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Jianying Gu
- Department of Plastic and Reconstructive Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, People's Republic of China.
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
13
|
Chen Y, Xin Q, Zhu M, Qiu J, Luo Y, Li R, Wei W, Tu J. Exploring CAR-macrophages in non-tumor diseases: Therapeutic potential beyond cancer. J Adv Res 2025:S2090-1232(25)00004-9. [PMID: 39756574 DOI: 10.1016/j.jare.2025.01.004] [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: 09/26/2024] [Revised: 01/01/2025] [Accepted: 01/02/2025] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND After significant advancements in tumor treatment, personalized cell therapy based on chimeric antigen receptors (CAR) holds promise for transforming the management of various diseases. CAR-T therapy, the first approved CAR cell therapy product, has demonstrated therapeutic potential in treating infectious diseases, autoimmune disorders, and fibrosis. CAR-macrophages (CAR-Ms) are emerging as a promising approach in CAR immune cell therapy, particularly for solid tumor treatment, highlighting the feasibility of using macrophages to eliminate pathogens and abnormal cells. AIM OF REVIEW This review summarizes the progress of CAR-M therapy in non-tumor diseases and discusses various CAR intracellular activation domain designs and their potential to optimize therapeutic effects by modulating interactions between cellular components in the tissue microenvironment and CAR-M. Additionally, we discuss the characteristics and advantages of CAR-M therapy compared to traditional medicine and CAR-T/NK therapy, as well as the challenges and prospects for the clinical translation of CAR-M. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of CAR-M for the treatment of non-tumor diseases, analyzes the advantages and characteristics of CAR-M therapy, and highlights the important impact of CAR intracellular domain design on therapeutic efficacy. In addition, the challenges and clinical translation prospects of developing CAR-M as a new cell therapy are discussed.
Collapse
Affiliation(s)
- Yizhao Chen
- Department of Pharmacy, The Third Affiliated Hospital of Anhui Medical University, Hefei First People's Hospital, Hefei, China; Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Qianling Xin
- Anhui Women and Children's Medical Center, Hefei Maternal and Child Health Hospital, Hefei, China
| | - Mengjuan Zhu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Jiaqi Qiu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China
| | - Yan Luo
- Department of Abdominal Radiotherapy, Hubei Provincial Cancer Hospital, Wuhan, China.
| | - Ruilin Li
- Department of Pharmacy, The Third Affiliated Hospital of Anhui Medical University, Hefei First People's Hospital, Hefei, China.
| | - Wei Wei
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| | - Jiajie Tu
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Institute of Clinical Pharmacology, Anhui Medical University, Hefei, China.
| |
Collapse
|
14
|
Rangel-Peláez C, Martínez-Gutiérrez L, Tristán-Manzano M, Callejas JL, Ortego-Centeno N, Martín F, Martín J. CD19 CAR-T cell therapy: a new dawn for autoimmune rheumatic diseases? Front Immunol 2024; 15:1502712. [PMID: 39742256 PMCID: PMC11685126 DOI: 10.3389/fimmu.2024.1502712] [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: 09/27/2024] [Accepted: 11/25/2024] [Indexed: 01/03/2025] Open
Abstract
Autoimmune rheumatic diseases (ARDs), such as rheumatoid arthritis, systemic lupus erythematosus, and systemic sclerosis, involve dysregulated immune responses causing chronic inflammation and tissue damage. Despite advancements in clinical management, many patients do not respond to current treatments, which often show limited efficacy due to the persistence of autoreactive B cells. Chimeric antigen receptor (CAR)-T cell therapy, which has shown success in oncology for B cell malignancies, targets specific antigens and involves the adoptive transfer of genetically engineered T cells. CD19 CAR-T cells, in particular, have shown promise in depleting circulating B cells and achieving clinical remission. This review discusses the potential of CD19 CAR-T cells in ARDs, highlighting clinical achievements and addressing key considerations such as optimal target cell populations, CAR construct design, acceptable toxicities, and the potential for lasting immune reset, crucial for the safe and effective adoption of CAR-T cell therapy in autoimmune treatments.
Collapse
Affiliation(s)
- Carlos Rangel-Peláez
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - Laura Martínez-Gutiérrez
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| | - María Tristán-Manzano
- LentiStem Biotech, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
| | - José Luis Callejas
- Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | - Norberto Ortego-Centeno
- Systemic Autoimmune Disease Unit, Hospital Clínico San Cecilio, Instituto de Investigación Biosanitaria Ibs, Granada, Spain
- Department of Medicine, University of Granada, Granada, Spain
| | - Francisco Martín
- Department of Biochemistry and Molecular Biology III and Immunology, Faculty of Medicine, Instituto Biosanitario de Granada (ibs.GRANADA), University of Granada, Granada, Spain
- Department of Genomic Medicine, Pfizer-University of Granada-Andalusian Regional Government Centre for Genomics and Oncological Research (GENYO), Granada, Spain
- Instituto Biosanitario de Granada (ibs.GRANADA), University of Granada, Granada, Spain
| | - Javier Martín
- Institute of Parasitology and Biomedicine López-Neyra, Consejo Superior de Investigaciones Científicas (CSIC), Granada, Spain
| |
Collapse
|
15
|
Sima H, Shao W. Advancements in the design and function of bispecific CAR-T cells targeting B Cell-Associated tumor antigens. Int Immunopharmacol 2024; 142:113166. [PMID: 39298818 DOI: 10.1016/j.intimp.2024.113166] [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: 07/18/2024] [Revised: 09/11/2024] [Accepted: 09/11/2024] [Indexed: 09/22/2024]
Abstract
Single-targeted CAR-T has exhibited notable success in treating B-cell tumors, effectively improving patient outcomes. However, the recurrence rate among patients remains above fifty percent, primarily attributed to antigen escape and the diminished immune persistence of CAR-T cells. Over recent years, there has been a surge of interest in bispecific CAR-T cell therapies, marked by an increasing number of research articles and clinical applications annually. This paper undertakes a comprehensive review of influential studies on the design of bispecific CAR-T in recent years, examining their impact on bispecific CAR-T efficacy concerning disease classification, targeted antigens, and CAR design. Notable distinctions in antigen targeting within B-ALL, NHL, and MM are explored, along with an analysis of how CAR scFv, transmembrane region, hinge region, and co-stimulatory region design influence Bi-CAR-T efficacy across different tumors. The summary provided aims to serve as a reference for designing novel and improved CAR-Ts, facilitating more efficient treatment for B-cell malignant tumors.
Collapse
Affiliation(s)
- Helin Sima
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China
| | - Wenwei Shao
- Academy of Medical Engineering and Translational Medicine, Tianjin University, Tianjin, China; Medical School of Tianjin University, Tianjin, China; State Key Laboratory of Advanced Medical Materials and Devices, Tianjin University, Tianjin, China.
| |
Collapse
|
16
|
Sainatham C, Yadav D, Dilli Babu A, Tallapalli JR, Kanagala SG, Filippov E, Murillo Chavez F, Ahmed N, Lutfi F. The current socioeconomic and regulatory landscape of immune effector cell therapies. Front Med (Lausanne) 2024; 11:1462307. [PMID: 39697210 PMCID: PMC11652178 DOI: 10.3389/fmed.2024.1462307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/18/2024] [Indexed: 12/20/2024] Open
Abstract
Immune cell effector therapies, including chimeric antigen receptor (CAR)-T cells, T-cell receptor (TCR) T cells, natural killer (NK) cells, and macrophage-based therapies, represent a transformative approach to cancer treatment, harnessing the immune system to target and eradicate malignant cells. CAR-T cell therapy, the most established among these, involves engineering T cells to express CARs specific to cancer cell antigens, showing remarkable efficacy in hematologic malignancies like leukemias, B-cell lymphomas, and multiple myeloma. Similarly, TCR-modified therapies, which reprogram T cells to recognize intracellular tumor antigens presented by major histocompatibility complex (MHC) molecules, offer promise for a range of solid tumors. NK-cell therapies leverage NK cells' innate cytotoxicity, providing an allogeneic approach that avoids some of the immune-related complications associated with T-cell-based therapies. Macrophage-based therapies, still in early stages of the development, focus on reprogramming macrophages to stimulate an immune response against cancer cells in the tumor microenvironment. Despite their promise, socioeconomic and regulatory challenges hinder the accessibility and scalability of immune cell effector therapies. These treatments are costly, with CAR-T therapies currently exceeding $400,000 per patient, creating significant disparities in access based on socioeconomic status and geographic location. The high manufacturing costs stem from the personalized, labor-intensive processes of harvesting, modifying, and expanding patients' cells. Moreover, complex logistics for manufacturing and delivering these therapies limit their reach, particularly in low-resource settings. Regulatory pathways further complicate the landscape. In the United States., the Food and Drug Administrations' (FDA) accelerated approval processes for cell-based therapies facilitate innovation but do not address cost-related barriers. In Europe, the European Medicines Agency (EMA) offers adaptive pathways, yet decentralized reimbursement systems create uneven access across member states. Additionally, differing regulatory standards for manufacturing and quality control worldwide pose hurdles for global harmonization and access. To expand the reach of immune effector cell therapies, a multipronged approach is needed-streamlined regulatory frameworks, policies to reduce treatment costs, and international collaborations to standardize manufacturing. Addressing these socioeconomic and regulatory obstacles is essential to make these life-saving therapies accessible to a broader patient population worldwide. We present a literature review on the current landscape of immune effector cell therapies and barriers of access to currently approved standard of care therapy at various levels.
Collapse
Affiliation(s)
- Chiranjeevi Sainatham
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Devvrat Yadav
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Aravind Dilli Babu
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Jayanth Reddy Tallapalli
- Division of Infectious Diseases, Department of Internal Medicine, University of South Florida, Tampa, FL, United States
| | - Sai Gautham Kanagala
- Department of Internal Medicine, New York Medical College/Metropolitan Hospital Center, New York, NY, United States
| | - Evgenii Filippov
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Franco Murillo Chavez
- Department of Internal Medicine, Sinai Hospital of Baltimore, Baltimore, MD, United States
| | - Nausheen Ahmed
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| | - Forat Lutfi
- Department of Hematologic Malignancies and Cellular Therapeutics, University of Kansas Medical Center, Kansas City, KS, United States
| |
Collapse
|
17
|
Franzese O. Tumor Microenvironment Drives the Cross-Talk Between Co-Stimulatory and Inhibitory Molecules in Tumor-Infiltrating Lymphocytes: Implications for Optimizing Immunotherapy Outcomes. Int J Mol Sci 2024; 25:12848. [PMID: 39684559 DOI: 10.3390/ijms252312848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024] Open
Abstract
This review explores some of the complex mechanisms underlying antitumor T-cell response, with a specific focus on the balance and cross-talk between selected co-stimulatory and inhibitory pathways. The tumor microenvironment (TME) fosters both T-cell activation and exhaustion, a dual role influenced by the local presence of inhibitory immune checkpoints (ICs), which are exploited by cancer cells to evade immune surveillance. Recent advancements in IC blockade (ICB) therapies have transformed cancer treatment. However, only a fraction of patients respond favorably, highlighting the need for predictive biomarkers and combination therapies to overcome ICB resistance. A crucial aspect is represented by the complexity of the TME, which encompasses diverse cell types that either enhance or suppress immune responses. This review underscores the importance of identifying the most critical cross-talk between inhibitory and co-stimulatory molecules for developing approaches tailored to patient-specific molecular and immune profiles to maximize the therapeutic efficacy of IC inhibitors and enhance clinical outcomes.
Collapse
Affiliation(s)
- Ornella Franzese
- Department of Systems Medicine, University of Rome "Tor Vergata", Via Montpellier 1, 00133 Rome, Italy
| |
Collapse
|
18
|
Zhou Y, Wei S, Xu M, Wu X, Dou W, Li H, Zhang Z, Zhang S. CAR-T cell therapy for hepatocellular carcinoma: current trends and challenges. Front Immunol 2024; 15:1489649. [PMID: 39569202 PMCID: PMC11576447 DOI: 10.3389/fimmu.2024.1489649] [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: 09/01/2024] [Accepted: 10/18/2024] [Indexed: 11/22/2024] Open
Abstract
Hepatocellular carcinoma (HCC) ranks among the most prevalent cancers worldwide, highlighting the urgent need for improved diagnostic and therapeutic methodologies. The standard treatment regimen generally involves surgical intervention followed by systemic therapies; however, the median survival rates for patients remain unsatisfactory. Chimeric antigen receptor (CAR) T-cell therapy has emerged as a pivotal advancement in cancer treatment. Both clinical and preclinical studies emphasize the notable efficacy of CAR T cells in targeting HCC. Various molecules, such as GPC3, c-Met, and NKG2D, show significant promise as potential immunotherapeutic targets in liver cancer. Despite this, employing CAR T cells to treat solid tumors like HCC poses considerable challenges within the discipline. Numerous innovations have significant potential to enhance the efficacy of CAR T-cell therapy for HCC, including improvements in T cell trafficking, strategies to counteract the immunosuppressive tumor microenvironment, and enhanced safety protocols. Ongoing efforts to discover therapeutic targets for CAR T cells highlight the need for the development of more practical manufacturing strategies for CAR-modified cells. This review synthesizes recent findings and clinical advancements in the use of CAR T-cell therapies for HCC treatment. We elucidate the therapeutic benefits of CAR T cells in HCC and identify the primary barriers to their broader application. Our analysis aims to provide a comprehensive overview of the current status and future prospects of CAR T-cell immunotherapy for HCC.
Collapse
Affiliation(s)
- Yexin Zhou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
- The General Hospital of Western Theater Command, Chengdu, China
| | - Shanshan Wei
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Menghui Xu
- The General Hospital of Western Theater Command, Chengdu, China
| | - Xinhui Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Wenbo Dou
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Huakang Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Zhonglin Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| | - Shuo Zhang
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, Sichuan, China
| |
Collapse
|
19
|
Ai K, Liu B, Chen X, Huang C, Yang L, Zhang W, Weng J, Du X, Wu K, Lai P. Optimizing CAR-T cell therapy for solid tumors: current challenges and potential strategies. J Hematol Oncol 2024; 17:105. [PMID: 39501358 PMCID: PMC11539560 DOI: 10.1186/s13045-024-01625-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Accepted: 10/18/2024] [Indexed: 11/08/2024] Open
Abstract
Chimeric antigen receptor (CAR)-T cell therapy demonstrates substantial efficacy in various hematological malignancies. However, its application in solid tumors is still limited. Clinical studies report suboptimal outcomes such as reduced cytotoxicity of CAR-T cells and tumor evasion, underscoring the need to address the challenges of sliding cytotoxicity in CAR-T cells. Despite improvements from fourth and next-generation CAR-T cells, new challenges include systemic toxicity from continuously secreted proteins, low productivity, and elevated costs. Recent research targets genetic modifications to boost killing potential, metabolic interventions to hinder tumor progression, and diverse combination strategies to enhance CAR-T cell therapy. Efforts to reduce the duration and cost of CAR-T cell therapy include developing allogenic and in-vivo approaches, promising significant future advancements. Concurrently, innovative technologies and platforms enhance the potential of CAR-T cell therapy to overcome limitations in treating solid tumors. This review explores strategies to optimize CAR-T cell therapies for solid tumors, focusing on enhancing cytotoxicity and overcoming application restrictions. We summarize recent advances in T cell subset selection, CAR-T structural modifications, infiltration enhancement, genetic and metabolic interventions, production optimization, and the integration of novel technologies, presenting therapeutic approaches that could improve CAR-T cell therapy's efficacy and applicability in solid tumors.
Collapse
Affiliation(s)
- Kexin Ai
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Bowen Liu
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xiaomei Chen
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Chuxin Huang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Liping Yang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, Guangdong, China
| | - Weiya Zhang
- Princess Máxima Center for Pediatric Oncology, 3584 CS, Utrecht, The Netherlands
| | - Jianyu Weng
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Xin Du
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China
| | - Kongming Wu
- Cancer Center, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, 030032, China.
- Cancer Center, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, 1095 Jiefang Avenue, Wuhan, 430030, China.
| | - Peilong Lai
- Department of Hematology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, 106 Zhongshan Er Road, Guangzhou, 510280, Guangdong, China.
| |
Collapse
|
20
|
Derigs P, Schubert ML, Dreger P, Schmitt A, Yousefian S, Haas S, Röthemeier C, Neuber B, Hückelhoven-Krauss A, Brüggemann M, Bernhard H, Kobbe G, Lindemann A, Rummel M, Michels B, Korell F, Ho AD, Müller-Tidow C, Schmitt M. Third-generation anti-CD19 CAR T cells for relapsed/refractory chronic lymphocytic leukemia: a phase 1/2 study. Leukemia 2024; 38:2419-2428. [PMID: 39192036 PMCID: PMC11519001 DOI: 10.1038/s41375-024-02392-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Third-generation chimeric antigen receptor T cells (CARTs) for relapsed or refractory (r/r) chronic lymphocytic leukemia (CLL) may improve efficacy compared to second-generation CARTs due to their enhanced CAR design. We performed the first phase 1/2 investigator-initiated trial evaluating escalating doses of third-generation CARTs (HD-CAR-1) targeting CD19 in patients with r/r CLL and B-cell lymphoma. CLL eligibility criteria were failure to two therapy lines including at least one pathway inhibitor and/or allogeneic hematopoietic cell transplantation. Nine heavily pretreated patients received HD-CAR-1 at dose levels ranging from 1 × 106 to 200 × 106 CART/m2. In-house HD-CAR-1 manufacturing was successful for all patients. While neurotoxicity was absent, one case of grade 3 cytokine release syndrome was observed. By day 90, six patients (67%) attained a CR, five of these (83%) with undetectable MRD. With a median follow-up of 27 months, 2-year PFS and OS were 30% and 69%, respectively. HD-CAR-1 products of responders contained significantly more CD4 + T cells compared to non-responders. In non-responders, a strong enrichment of effector memory-like CD8 + T cells with high expression of CD39 and/or CD197 was observed. HD-CAR-1 demonstrated encouraging efficacy and exceptionally low treatment-specific toxicity, presenting new treatment options for patients with r/r CLL. Trial registration: #NCT03676504.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Male
- Immunotherapy, Adoptive/methods
- Immunotherapy, Adoptive/adverse effects
- Antigens, CD19/immunology
- Middle Aged
- Female
- Aged
- Receptors, Chimeric Antigen/immunology
- Neoplasm Recurrence, Local/pathology
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/therapy
- Adult
- Follow-Up Studies
Collapse
Affiliation(s)
- Patrick Derigs
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany.
| | - Maria-Luisa Schubert
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Peter Dreger
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anita Schmitt
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Schayan Yousefian
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Simon Haas
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Precision Healthcare University Research Institute, Queen Mary University of London, London, UK
| | - Caroline Röthemeier
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Brigitte Neuber
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Angela Hückelhoven-Krauss
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Helga Bernhard
- Department of Internal Medicine V, Klinikum Darmstadt, Darmstadt, Germany
| | - Guido Kobbe
- Department of Hematology, Oncology and Clinical Immunology, University Hospital Düsseldorf, Düsseldorf, Germany
| | | | - Mathias Rummel
- Department of Internal Medicine IV, University Hospital Giessen, Giessen, Germany
| | - Birgit Michels
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Korell
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
| | - Anthony D Ho
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Internal Medicine V, Hematology, Oncology and Rheumatology, Heidelberg University Hospital, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| |
Collapse
|
21
|
Acharya S, Basar R, Daher M, Rafei H, Li P, Uprety N, Ensley E, Shanley M, Kumar B, Banerjee PP, Melo Garcia L, Lin P, Mohanty V, Kim KH, Jiang X, Pan Y, Li Y, Liu B, Nunez Cortes AK, Zhang C, Fathi M, Rezvan A, Montalvo MJ, Cha SL, Reyes-Silva F, Shrestha R, Guo X, Kundu K, Biederstadt A, Muniz-Feliciano L, Deyter GM, Kaplan M, Jiang XR, Liu E, Jain A, Roszik J, Fowlkes NW, Solis Soto LM, Raso MG, Khoury JD, Lin P, Vega F, Varadarajan N, Chen K, Marin D, Shpall EJ, Rezvani K. CD28 Costimulation Augments CAR Signaling in NK Cells via the LCK/CD3ζ/ZAP70 Signaling Axis. Cancer Discov 2024; 14:1879-1900. [PMID: 38900051 PMCID: PMC11452288 DOI: 10.1158/2159-8290.cd-24-0096] [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: 01/18/2024] [Revised: 04/16/2024] [Accepted: 05/24/2024] [Indexed: 06/21/2024]
Abstract
Multiple factors in the design of a chimeric antigen receptor (CAR) influence CAR T-cell activity, with costimulatory signals being a key component. Yet, the impact of costimulatory domains on the downstream signaling and subsequent functionality of CAR-engineered natural killer (NK) cells remains largely unexplored. Here, we evaluated the impact of various costimulatory domains on CAR-NK cell activity, using a CD70-targeting CAR. We found that CD28, a costimulatory molecule not inherently present in mature NK cells, significantly enhanced the antitumor efficacy and long-term cytotoxicity of CAR-NK cells both in vitro and in multiple xenograft models of hematologic and solid tumors. Mechanistically, we showed that CD28 linked to CD3ζ creates a platform that recruits critical kinases, such as lymphocyte-specific protein tyrosine kinase (LCK) and zeta-chain-associated protein kinase 70 (ZAP70), initiating a signaling cascade that enhances CAR-NK cell function. Our study provides insights into how CD28 costimulation enhances CAR-NK cell function and supports its incorporation in NK-based CARs for cancer immunotherapy. Significance: We demonstrated that incorporation of the T-cell-centric costimulatory molecule CD28, which is normally absent in mature natural killer (NK) cells, into the chimeric antigen receptor (CAR) construct recruits key kinases including lymphocyte-specific protein tyrosine kinase and zeta-chain-associated protein kinase 70 and results in enhanced CAR-NK cell persistence and sustained antitumor cytotoxicity.
Collapse
Affiliation(s)
- Sunil Acharya
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rafet Basar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - May Daher
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Hind Rafei
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ping Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Nadima Uprety
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Emily Ensley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mayra Shanley
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bijender Kumar
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Pinaki P. Banerjee
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luciana Melo Garcia
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Paul Lin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Vakul Mohanty
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kun Hee Kim
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xianli Jiang
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yuchen Pan
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ye Li
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bin Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ana Karen Nunez Cortes
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Chenyu Zhang
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mohsen Fathi
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
- CellChorus, Inc., Houston, TX, USA
| | - Ali Rezvan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Melisa J. Montalvo
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Sophia L Cha
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francia Reyes-Silva
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Rejeena Shrestha
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xingliang Guo
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Kiran Kundu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander Biederstadt
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Medicine III: Hematology and Oncology, School of Medicine, Technical University of Munich, Munich, Germany
| | - Luis Muniz-Feliciano
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Gary M. Deyter
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Mecit Kaplan
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Ru Jiang
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Enli Liu
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antrix Jain
- Mass Spectrometry Proteomics Core, Baylor College of Medicine, Houston, TX, USA
| | - Janos Roszik
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Natalie W. Fowlkes
- Department of Veterinary Medicine & Surgery, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Luisa M. Solis Soto
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Maria Gabriela Raso
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Joseph D. Khoury
- Department of Pathology, Microbiology, and Immunology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Pei Lin
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Francisco Vega
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Navin Varadarajan
- William A. Brookshire Department of Chemical and Biomolecular Engineering, University of Houston, Houston, TX, USA
| | - Ken Chen
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - David Marin
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Elizabeth J. Shpall
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Katayoun Rezvani
- Department of Stem Cell Transplantation and Cellular Therapy, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
22
|
Zhou S, Yang Y, Jing Y, Zhu X. Generating advanced CAR-based therapy for hematological malignancies in clinical practice: targets to cell sources to combinational strategies. Front Immunol 2024; 15:1435635. [PMID: 39372412 PMCID: PMC11449748 DOI: 10.3389/fimmu.2024.1435635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 09/03/2024] [Indexed: 10/08/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has been a milestone breakthrough in the treatment of hematological malignancies, offering an effective therapeutic option for multi-line therapy-refractory patients. So far, abundant CAR-T products have been approved by the United States Food and Drug Administration or China National Medical Products Administration to treat relapsed or refractory hematological malignancies and exhibited unprecedented clinical efficiency. However, there were still several significant unmet needs to be progressed, such as the life-threatening toxicities, the high cost, the labor-intensive manufacturing process and the poor long-term therapeutic efficacy. According to the demands, many researches, relating to notable technical progress and the replenishment of alternative targets or cells, have been performed with promising results. In this review, we will summarize the current research progress in CAR-T eras from the "targets" to "alternative cells", to "combinational drugs" in preclinical studies and clinical trials.
Collapse
Affiliation(s)
- Shu Zhou
- Department of Hematology, Zhongnan Hospital, Wuhan University, Wuhan, China
| | - Yuhang Yang
- The First Clinical Medical College, Wuhan University, Wuhan, China
| | - Yulu Jing
- The Second Clinical Medical College, Wuhan University, Wuhan, China
| | - Xiaoying Zhu
- National Clinical Research Center for Hematologic Diseases, Jiangsu Institute of Hematology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Blood and Marrow Transplantation, Collaborative Innovation Center of Hematology, Soochow University, Suzhou, China
| |
Collapse
|
23
|
Hou R, Zhang X, Wang X, Zhao X, Li S, Guan Z, Cao J, Liu D, Zheng J, Shi M. In vivo manufacture and manipulation of CAR-T cells for better druggability. Cancer Metastasis Rev 2024; 43:1075-1093. [PMID: 38592427 DOI: 10.1007/s10555-024-10185-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 03/28/2024] [Indexed: 04/10/2024]
Abstract
The current CAR-T cell therapy products have been hampered in their druggability due to the personalized preparation required, unclear pharmacokinetic characteristics, and unpredictable adverse reactions. Enabling standardized manufacturing and having clear efficacy and pharmacokinetic characteristics are prerequisites for ensuring the effective practicality of CAR-T cell therapy drugs. This review provides a broad overview of the different approaches for controlling behaviors of CAR-T cells in vivo. The utilization of genetically modified vectors enables in vivo production of CAR-T cells, thereby abbreviating or skipping the lengthy in vitro expansion process. By equipping CAR-T cells with intricately designed control elements, using molecule switches or small-molecule inhibitors, the control of CAR-T cell activity can be achieved. Moreover, the on-off control of CAR-T cell activity would yield potential gains in phenotypic remodeling. These methods provide beneficial references for the future development of safe, controllable, convenient, and suitable for standardized production of CAR-T cell therapy products.
Collapse
Affiliation(s)
- Rui Hou
- College of Pharmacy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xiaoxue Zhang
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xu Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Xuan Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Sijin Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Zhangchun Guan
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Jiang Cao
- Department of Hematology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Dan Liu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Junnian Zheng
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| | - Ming Shi
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Center of Clinical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China.
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Xuzhou Medical University, Xuzhou, Jiangsu, China.
| |
Collapse
|
24
|
Gao X, Liu J, Sun R, Zhang J, Cao X, Zhang Y, Zhao M. Alliance between titans: combination strategies of CAR-T cell therapy and oncolytic virus for the treatment of hematological malignancies. Ann Hematol 2024; 103:2569-2589. [PMID: 37853078 DOI: 10.1007/s00277-023-05488-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 09/28/2023] [Indexed: 10/20/2023]
Abstract
There have been several clinical studies using chimeric antigen receptor (CAR)-T cell therapy for different hematological malignancies. It has transformed the therapy landscape for hematologic malignancies dramatically. Nonetheless, in acute myeloid leukemia (AML) and T cell malignancies, it still has a dismal prognosis. Even in the most promising locations, recurrence with CAR-T treatment remains a big concern. Oncolytic viruses (OVs) can directly lyse tumor cells or cause immune responses, and they can be manipulated to create therapeutic proteins, increasing anticancer efficacy. Oncolytic viruses have been proven in a rising number of studies to be beneficial in hematological malignancies. There are limitations that cannot be avoided by using either treatment alone, and the combination of CAR-T cell therapy and oncolytic virus therapy may complement the disadvantages of individual application, enhance the advantages of their respective treatment methods and improve the treatment effect. The alternatives for combining two therapies in hematological malignancies are discussed in this article.
Collapse
Affiliation(s)
- Xuejin Gao
- Emergency, Tianjin First Central Hospital, Tianjin, 300192, China
| | - Jile Liu
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Rui Sun
- Nankai University School of Medicine, Tianjin, 300192, China
| | - Jingkun Zhang
- Cardiovascular Research Institute, University of California San Francisco, San Francisco, CA, USA
| | - Xinping Cao
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Yi Zhang
- First Center Clinic College of Tianjin Medical University, Tianjin, 300192, China
| | - Mingfeng Zhao
- Department of Hematology, Tianjin First Central Hospital, Tianjin, 300192, China.
| |
Collapse
|
25
|
Du G, Dou C, Sun P, Wang S, Liu J, Ma L. Regulatory T cells and immune escape in HCC: understanding the tumor microenvironment and advancing CAR-T cell therapy. Front Immunol 2024; 15:1431211. [PMID: 39136031 PMCID: PMC11317284 DOI: 10.3389/fimmu.2024.1431211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Liver cancer, which most commonly manifests as hepatocellular carcinoma (HCC), is the sixth most common cancer in the world. In HCC, the immune system plays a crucial role in the growth and proliferation of tumor cells. HCC achieve immune escape through the tumor microenvironment, which significantly promotes the development of this cancer. Here, this article introduces and summarizes the functions and effects of regulatory T cells (Tregs) in the tumor microenvironment, highlighting how Tregs inhibit and regulate the functions of immune and tumor cells, cytokines, ligands and receptors, etc, thereby promoting tumor immune escape. In addition, it discusses the mechanism of CAR-T therapy for HCC and elaborate on the relationship between CAR-T and Tregs.
Collapse
Affiliation(s)
- Guangtan Du
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Cunmiao Dou
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Peng Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Leina Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
26
|
Tian M, Wei JS, Cheuk ATC, Milewski D, Zhang Z, Kim YY, Chou HC, Liu C, Badr S, Pope EG, Rahmy A, Wu JT, Kelly MC, Wen X, Khan J. CAR T-cells targeting FGFR4 and CD276 simultaneously show potent antitumor effect against childhood rhabdomyosarcoma. Nat Commun 2024; 15:6222. [PMID: 39043633 PMCID: PMC11266617 DOI: 10.1038/s41467-024-50251-x] [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: 10/24/2023] [Accepted: 07/02/2024] [Indexed: 07/25/2024] Open
Abstract
Chimeric antigen receptor (CAR) T-cells targeting Fibroblast Growth Factor Receptor 4 (FGFR4), a highly expressed surface tyrosine receptor in rhabdomyosarcoma (RMS), are already in the clinical phase of development, but tumour heterogeneity and suboptimal activation might hamper their potency. Here we report an optimization strategy of the co-stimulatory and targeting properties of a FGFR4 CAR. We replace the CD8 hinge and transmembrane domain and the 4-1BB co-stimulatory domain with those of CD28. The resulting CARs display enhanced anti-tumor activity in several RMS xenograft models except for an aggressive tumour cell line, RMS559. By searching for a direct target of the RMS core-regulatory transcription factor MYOD1, we identify another surface protein, CD276, as a potential target. Bicistronic CARs (BiCisCAR) targeting both FGFR4 and CD276, containing two distinct co-stimulatory domains, have superior prolonged persistent and invigorated anti-tumor activities compared to the optimized FGFR4-specific CAR and the other BiCisCAR with the same 4-1BB co-stimulatory domain. Our study thus lays down the proof-of-principle for a CAR T-cell therapy targeting both FGFR4 and CD276 in RMS.
Collapse
MESH Headings
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Rhabdomyosarcoma/therapy
- Rhabdomyosarcoma/immunology
- Rhabdomyosarcoma/genetics
- Humans
- Animals
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Mice
- Immunotherapy, Adoptive/methods
- B7 Antigens/metabolism
- B7 Antigens/immunology
- B7 Antigens/genetics
- MyoD Protein/metabolism
- MyoD Protein/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Child
- Female
- Mice, SCID
- Mice, Inbred NOD
Collapse
Affiliation(s)
- Meijie Tian
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jun S Wei
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Adam Tai-Chi Cheuk
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - David Milewski
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Zhongmei Zhang
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Yong Yean Kim
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Hsien-Chao Chou
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Can Liu
- Multiscale Systems Biology Section, Laboratory of Immune System Biology, NIAID, NIH, Bethesda, MD, 20892, USA
| | - Sherif Badr
- Experimental Immunology Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Eleanor G Pope
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Abdelrahman Rahmy
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jerry T Wu
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Michael C Kelly
- Single Cell Analysis Facility, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Xinyu Wen
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Javed Khan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
27
|
Olejarz W, Sadowski K, Szulczyk D, Basak G. Advancements in Personalized CAR-T Therapy: Comprehensive Overview of Biomarkers and Therapeutic Targets in Hematological Malignancies. Int J Mol Sci 2024; 25:7743. [PMID: 39062986 PMCID: PMC11276786 DOI: 10.3390/ijms25147743] [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: 06/30/2024] [Revised: 07/12/2024] [Accepted: 07/12/2024] [Indexed: 07/28/2024] Open
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy is a novel anticancer therapy using autologous or allogeneic T-cells. To date, six CAR-T therapies for specific B-cell acute lymphoblastic leukemia (B-ALL), non-Hodgkin lymphomas (NHL), and multiple myeloma (MM) have been approved by the Food and Drug Administration (FDA). Significant barriers to the effectiveness of CAR-T therapy include cytokine release syndrome (CRS), neurotoxicity in the case of Allogeneic Stem Cell Transplantation (Allo-SCT) graft-versus-host-disease (GVHD), antigen escape, modest antitumor activity, restricted trafficking, limited persistence, the immunosuppressive microenvironment, and senescence and exhaustion of CAR-Ts. Furthermore, cancer drug resistance remains a major problem in clinical practice. CAR-T therapy, in combination with checkpoint blockades and bispecific T-cell engagers (BiTEs) or other drugs, appears to be an appealing anticancer strategy. Many of these agents have shown impressive results, combining efficacy with tolerability. Biomarkers like extracellular vesicles (EVs), cell-free DNA (cfDNA), circulating tumor (ctDNA) and miRNAs may play an important role in toxicity, relapse assessment, and efficacy prediction, and can be implicated in clinical applications of CAR-T therapy and in establishing safe and efficacious personalized medicine. However, further research is required to fully comprehend the particular side effects of immunomodulation, to ascertain the best order and combination of this medication with conventional chemotherapy and targeted therapies, and to find reliable predictive biomarkers.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
| | - Karol Sadowski
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-097 Warsaw, Poland;
- Centre for Preclinical Research, Medical University of Warsaw, 02-097 Warsaw, Poland
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Daniel Szulczyk
- Chair and Department of Biochemistry, The Medical University of Warsaw, 02-097 Warsaw, Poland;
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-097 Warsaw, Poland;
| |
Collapse
|
28
|
Yu Y, Yu M, Luo L, Zhang Z, Zeng H, Chen Y, Lin Z, Chen M, Wang W. Molecular characteristics and immune microenvironment of gastrointestinal stromal tumours: targets for therapeutic strategies. Front Oncol 2024; 14:1405727. [PMID: 39070147 PMCID: PMC11272528 DOI: 10.3389/fonc.2024.1405727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2024] [Accepted: 06/24/2024] [Indexed: 07/30/2024] Open
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal tumours, arising mainly from the interstitial cells of Cajal (ICCs) of the gastrointestinal tract. As radiotherapy and chemotherapy are generally ineffective for GISTs, the current primary treatment is surgical resection. However, surgical resection is not choice for most patients. Therefore, new therapeutic strategies are urgently needed. Targeted therapy, represented by tyrosine kinase inhibitors (TKIs), and immunotherapy, represented by immune checkpoint inhibitor therapies and chimeric antigen receptor T-cell immunotherapy (CAR-T), offer new therapeutic options in GISTs and have shown promising treatment responses. In this review, we summarize the molecular classification and immune microenvironment of GISTs and discuss the corresponding targeted therapy and immunotherapy options. This updated knowledge may provide more options for future therapeutic strategies and applications in GISTs.
Collapse
Affiliation(s)
- Yang Yu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengdie Yu
- Guangzhou KingMed Diagnostics Group Co., Ltd., Guangzhou, Guangdong, China
| | - Lijie Luo
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Zijing Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Haiping Zeng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Yan Chen
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Zeyu Lin
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Mengnan Chen
- Department of Thyroid and Breast Surgery, Baiyun Hospital, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| | - Wei Wang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Guangzhou University of Traditional Chinese Medicine, Guangzhou, Guangdong, China
| |
Collapse
|
29
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [Citation(s) in RCA: 74] [Impact Index Per Article: 74.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
30
|
Zhou Z, Wang J, Wang J, Yang S, Wang R, Zhang G, Li Z, Shi R, Wang Z, Lu Q. Deciphering the tumor immune microenvironment from a multidimensional omics perspective: insight into next-generation CAR-T cell immunotherapy and beyond. Mol Cancer 2024; 23:131. [PMID: 38918817 PMCID: PMC11201788 DOI: 10.1186/s12943-024-02047-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Accepted: 06/17/2024] [Indexed: 06/27/2024] Open
Abstract
Tumor immune microenvironment (TIME) consists of intra-tumor immunological components and plays a significant role in tumor initiation, progression, metastasis, and response to therapy. Chimeric antigen receptor (CAR)-T cell immunotherapy has revolutionized the cancer treatment paradigm. Although CAR-T cell immunotherapy has emerged as a successful treatment for hematologic malignancies, it remains a conundrum for solid tumors. The heterogeneity of TIME is responsible for poor outcomes in CAR-T cell immunotherapy against solid tumors. The advancement of highly sophisticated technology enhances our exploration in TIME from a multi-omics perspective. In the era of machine learning, multi-omics studies could reveal the characteristics of TIME and its immune resistance mechanism. Therefore, the clinical efficacy of CAR-T cell immunotherapy in solid tumors could be further improved with strategies that target unfavorable conditions in TIME. Herein, this review seeks to investigate the factors influencing TIME formation and propose strategies for improving the effectiveness of CAR-T cell immunotherapy through a multi-omics perspective, with the ultimate goal of developing personalized therapeutic approaches.
Collapse
Affiliation(s)
- Zhaokai Zhou
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Jiahui Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Nephrology, Union Medical College Hospital, Chinese Academy of Medical Sciences, PekingBeijing, 100730, China
| | - Jiaojiao Wang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
| | - Shuai Yang
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ruizhi Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Ge Zhang
- Department of Cardiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Zhengrui Li
- Department of Oral and Maxillofacial-Head and Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Run Shi
- Department of Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zhan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, 450052, China
| | - Qiong Lu
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, Hunan, 410011, China.
| |
Collapse
|
31
|
Zhang G, Wang Y, Lu S, Ding F, Wang X, Zhu C, Wang Y, Wang K. Molecular understanding and clinical outcomes of CAR T cell therapy in the treatment of urological tumors. Cell Death Dis 2024; 15:359. [PMID: 38789450 PMCID: PMC11126652 DOI: 10.1038/s41419-024-06734-2] [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: 01/15/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
Chimeric antigen receptor engineered T (CAR T) cell therapy has developed rapidly in recent years, leading to profound developments in oncology, especially for hematologic malignancies. However, given the pressure of immunosuppressive tumor microenvironments, antigen escape, and diverse other factors, its application in solid tumors is less developed. Urinary system tumors are relatively common, accounting for approximately 24% of all new cancers in the United States. CAR T cells have great potential for urinary system tumors. This review summarizes the latest developments of CAR T cell therapy in urinary system tumors, including kidney cancer, bladder cancer, and prostate cancer, and also outlines the various CAR T cell generations and their pathways and targets that have been developed thus far. Finally, the current advantages, problems, and side effects of CAR T cell therapy are discussed in depth, and potential future developments are proposed in view of current shortcomings.
Collapse
Affiliation(s)
- Gong Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yuan Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shiyang Lu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Fengzhu Ding
- Department of Nursing, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Yibing Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
32
|
Zhang PF, Xie D. Targeting the gut microbiota to enhance the antitumor efficacy and attenuate the toxicity of CAR-T cell therapy: a new hope? Front Immunol 2024; 15:1362133. [PMID: 38558812 PMCID: PMC10978602 DOI: 10.3389/fimmu.2024.1362133] [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: 12/27/2023] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
Chimeric antigen receptor (CAR) -T cell therapy has achieved tremendous efficacy in the treatment of hematologic malignancies and represents a promising treatment regimen for cancer. Despite the striking response in patients with hematologic malignancies, most patients with solid tumors treated with CAR-T cells have a low response rate and experience major adverse effects, which indicates the need for biomarkers that can predict and improve clinical outcomes with future CAR-T cell treatments. Recently, the role of the gut microbiota in cancer therapy has been established, and growing evidence has suggested that gut microbiota signatures may be harnessed to personally predict therapeutic response or adverse effects in optimizing CAR-T cell therapy. In this review, we discuss current understanding of CAR-T cell therapy and the gut microbiota, and the interplay between the gut microbiota and CAR-T cell therapy. Above all, we highlight potential strategies and challenges in harnessing the gut microbiota as a predictor and modifier of CAR-T cell therapy efficacy while attenuating toxicity.
Collapse
Affiliation(s)
- Peng-Fei Zhang
- Gastric Cancer Center, Division of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Dan Xie
- Department of Medical Genetics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, Chengdu, China
| |
Collapse
|
33
|
Rees MJ, Kumar S. BCMA-directed therapy, new treatments in the myeloma toolbox, and how to use them. Leuk Lymphoma 2024; 65:287-300. [PMID: 38354090 DOI: 10.1080/10428194.2023.2284088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2023] [Accepted: 11/11/2023] [Indexed: 02/16/2024]
Abstract
To address the dearth of therapeutic options available for relapsed-refractory multiple myeloma (RRMM), attention has shifted to immunotherapeutic strategies, with most products in development targeting the B-cell maturation antigen (BCMA). BCMA is a transmembrane receptor of the tumor necrosis factor receptor superfamily, essential for plasma cell survival and minimally expressed on non-hematopoietic tissues; it represents an ideal therapeutic target. Three categories of BCMA-directed therapies exist, with distinct strengths and weaknesses. Antibody-drug conjugates (ADCs) are immediately available with modest single-agent efficacy in RRMM, but deliverability is hampered by corneal toxicity. CAR T-cells are the most effective class but face significant logistical and financial barriers. Bispecific antibodies offer superior efficacy and tolerability compared to ADCs, but prolonged exposure causes significant cumulative infectious risk. In this review, we will examine the role of BCMA in MM biology, the approved and emerging therapies targeting this antigen, and how these agents can be optimally sequenced.
Collapse
Affiliation(s)
| | - Shaji Kumar
- Division of Hematology, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
34
|
Sun D, Shi X, Li S, Wang X, Yang X, Wan M. CAR‑T cell therapy: A breakthrough in traditional cancer treatment strategies (Review). Mol Med Rep 2024; 29:47. [PMID: 38275119 PMCID: PMC10835665 DOI: 10.3892/mmr.2024.13171] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2023] [Accepted: 12/18/2023] [Indexed: 01/27/2024] Open
Abstract
Chimeric antigen receptor (CAR)‑T cell therapy is an innovative approach to immune cell therapy that works by modifying the T cells of a patient to express the CAR protein on their surface, and thus induce their recognition and destruction of cancer cells. CAR‑T cell therapy has shown some success in treating hematological tumors, but it still faces a number of challenges in the treatment of solid tumors, such as antigen selection, tolerability and safety. In response to these issues, studies continue to improve the design of CAR‑T cells in pursuit of improved therapeutic efficacy and safety. In the future, CAR‑T cell therapy is expected to become an important cancer treatment, and may provide new ideas and strategies for individualized immunotherapy. The present review provides a comprehensive overview of the principles, clinical applications, therapeutic efficacy and challenges of CAR‑T cell therapy.
Collapse
Affiliation(s)
- Dahua Sun
- Department of General Surgery, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Xiang Shi
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Sanyan Li
- Department of Pathology, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Xiaohua Wang
- Department of Obstetrics, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Xiao Yang
- Department of General Surgery, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| | - Meiping Wan
- Department of Traditional Chinese Medicine, Qianjiang Central Hospital, Qianjiang, Hubei 433100, P.R. China
| |
Collapse
|
35
|
Liu Z, Lei W, Wang H, Liu X, Fu R. Challenges and strategies associated with CAR-T cell therapy in blood malignancies. Exp Hematol Oncol 2024; 13:22. [PMID: 38402232 PMCID: PMC10893672 DOI: 10.1186/s40164-024-00490-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2023] [Accepted: 02/19/2024] [Indexed: 02/26/2024] Open
Abstract
Cellular immunotherapy, particularly CAR-T cells, has shown potential in the improvement of outcomes in patients with refractory and recurrent malignancies of the blood. However, achieving sustainable long-term complete remission for blood cancer remains a challenge, with resistance and relapse being expected outcomes for many patients. Although many studies have attempted to clarify the mechanisms of CAR-T cell therapy failure, the mechanism remains unclear. In this article, we discuss and describe the current state of knowledge regarding these factors, which include elements that influence the CAR-T cell, cancer cells as a whole, and the microenvironment surrounding the tumor. In addition, we propose prospective approaches to overcome these obstacles in an effort to decrease recurrence rates and extend patient survival subsequent to CAR-T cell therapy.
Collapse
Affiliation(s)
- Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China.
| | - Wenhui Lei
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
- Department of Nephrology, Lishui Municipal Central Hospital, Lishui, Zhejiang, 323000, People's Republic of China
| | - Hao Wang
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
| | - Xiaohan Liu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China
| | - Rong Fu
- Department of Hematology, Tianjin Medical University General Hospital, 154 Anshan Street, Heping District, Tianjin, 300052, PR China.
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone46Control, Tianjin, 300052, P. R. China.
| |
Collapse
|
36
|
Singh R, Kim YH, Lee SJ, Eom HS, Choi BK. 4-1BB immunotherapy: advances and hurdles. Exp Mol Med 2024; 56:32-39. [PMID: 38172595 PMCID: PMC10834507 DOI: 10.1038/s12276-023-01136-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Revised: 09/22/2023] [Accepted: 10/09/2023] [Indexed: 01/05/2024] Open
Abstract
Since its initial description 35 years ago as an inducible molecule expressed in cytotoxic and helper T cells, 4-1BB has emerged as a crucial receptor in T-cell-mediated immune functions. Numerous studies have demonstrated the involvement of 4-1BB in infection and tumor immunity. However, the clinical development of 4-1BB agonist antibodies has been impeded by the occurrence of strong adverse events, notably hepatotoxicity, even though these antibodies have exhibited tremendous promise in in vivo tumor models. Efforts are currently underway to develop a new generation of agonist antibodies and recombinant proteins with modified effector functions that can harness the potent T-cell modulation properties of 4-1BB while mitigating adverse effects. In this review, we briefly examine the role of 4-1BB in T-cell biology, explore its clinical applications, and discuss future prospects in the field of 4-1BB agonist immunotherapy.
Collapse
Affiliation(s)
- Rohit Singh
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Young-Ho Kim
- Diagnostics and Therapeutics Technology Branch, Division of Technology Convergence, Research Institute, National Cancer Center, Goyang, 10408, Republic of Korea.
| | - Sang-Jin Lee
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Hyeon-Seok Eom
- Hematological Malignancy Center, National Cancer Center, Goyang, 10408, Republic of Korea
| | - Beom K Choi
- Immuno-oncology Branch, Division of Rare and Refractory Cancer, National Cancer Center, Goyang, 10408, Republic of Korea.
- Innobationbio, Co., Ltd., Mapo-gu, Seoul, 03929, Republic of Korea.
| |
Collapse
|
37
|
Schreiber B, Tripathi S, Nikiforow S, Chandraker A. Adoptive Immune Effector Cell Therapies in Cancer and Solid Organ Transplantation: A Review. Semin Nephrol 2024; 44:151498. [PMID: 38555223 DOI: 10.1016/j.semnephrol.2024.151498] [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] [Indexed: 04/02/2024]
Abstract
Cancer is one of the most devastating complications of kidney transplantation and constitutes one of the leading causes of morbidity and mortality among solid organ transplantation (SOT) recipients. Immunosuppression, although effective in preventing allograft rejection, inherently inhibits immune surveillance against oncogenic viral infections and malignancy. Adoptive cell therapy, particularly immune effector cell therapy, has long been a modality of interest in both cancer and transplantation, though has only recently stepped into the spotlight with the development of virus-specific T-cell therapy and chimeric antigen receptor T-cell therapy. Although these modalities are best described in hematopoietic cell transplantation and hematologic malignancies, their potential application in the SOT setting may hold tremendous promise for those with limited therapeutic options. In this review, we provide a brief overview of the development of adoptive cell therapies with a focus on virus-specific T-cell therapy and chimeric antigen receptor T-cell therapy. We also describe the current experience of these therapies in the SOT setting as well as the challenges in their application and future directions in their development.
Collapse
Affiliation(s)
- Brittany Schreiber
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sudipta Tripathi
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Sarah Nikiforow
- Division of Medical Oncology, Department of Medicine, Dana-Farber Cancer Institute, Brigham and Women's Hospital, Harvard Medical School, Boston, MA
| | - Anil Chandraker
- Division of Renal Medicine, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA; Division of Renal Medicine, Department of Medicine, University of Massachusetts Chan Medical School, Worcester, MA.
| |
Collapse
|
38
|
Folliero V, Dell’Annunziata F, Chianese A, Morone MV, Mensitieri F, Di Spirito F, Mollo A, Amato M, Galdiero M, Dal Piaz F, Pagliano P, Rinaldi L, Franci G. Epigenetic and Genetic Keys to Fight HPV-Related Cancers. Cancers (Basel) 2023; 15:5583. [PMID: 38067286 PMCID: PMC10705756 DOI: 10.3390/cancers15235583] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 12/07/2024] Open
Abstract
Cervical cancer ranks as the fourth most prevalent cancer among women globally, with approximately 600,000 new cases being diagnosed each year. The principal driver of cervical cancer is the human papillomavirus (HPV), where viral oncoproteins E6 and E7 undertake the role of driving its carcinogenic potential. Despite extensive investigative efforts, numerous facets concerning HPV infection, replication, and pathogenesis remain shrouded in uncertainty. The virus operates through a variety of epigenetic mechanisms, and the epigenetic signature of HPV-related tumors is a major bottleneck in our understanding of the disease. Recent investigations have unveiled the capacity of viral oncoproteins to influence epigenetic changes within HPV-related tumors, and conversely, these tumors exert an influence on the surrounding epigenetic landscape. Given the escalating occurrence of HPV-triggered tumors and the deficiency of efficacious treatments, substantial challenges emerge. A promising avenue to address this challenge lies in epigenetic modulators. This review aggregates and dissects potential epigenetic modulators capable of combatting HPV-associated infections and diseases. By delving into these modulators, novel avenues for therapeutic interventions against HPV-linked cancers have come to the fore.
Collapse
Affiliation(s)
- Veronica Folliero
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Federica Dell’Annunziata
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.C.); (M.V.M.); (M.G.)
| | - Annalisa Chianese
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.C.); (M.V.M.); (M.G.)
| | - Maria Vittoria Morone
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.C.); (M.V.M.); (M.G.)
| | - Francesca Mensitieri
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Federica Di Spirito
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Antonio Mollo
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Massimo Amato
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Massimiliano Galdiero
- Department of Experimental Medicine, University of Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (A.C.); (M.V.M.); (M.G.)
| | - Fabrizio Dal Piaz
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Pasquale Pagliano
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| | - Luca Rinaldi
- Department of Medicine and Health Sciences “Vincenzo Tiberio”, Università degli Studi del Molise, 86100 Campobasso, Italy
| | - Gianluigi Franci
- Department of Medicine, Surgery and Dentistry “Scuola Medica Salernitana”, University of Salerno, 84081 Baronissi, Italy; (V.F.); (F.D.); (F.M.); (F.D.S.); (A.M.); (M.A.); (F.D.P.); (P.P.)
| |
Collapse
|
39
|
Ma HY, Das J, Prendergast C, De Jong D, Braumuller B, Paily J, Huang S, Liou C, Giarratana A, Hosseini M, Yeh R, Capaccione KM. Advances in CAR T Cell Therapy for Non-Small Cell Lung Cancer. Curr Issues Mol Biol 2023; 45:9019-9038. [PMID: 37998743 PMCID: PMC10670348 DOI: 10.3390/cimb45110566] [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: 10/20/2023] [Revised: 11/08/2023] [Accepted: 11/10/2023] [Indexed: 11/25/2023] Open
Abstract
Since its first approval by the FDA in 2017, tremendous progress has been made in chimeric antigen receptor (CAR) T cell therapy, the adoptive transfer of engineered, CAR-expressing T lymphocyte. CAR T cells are all composed of three main elements: an extracellular antigen-binding domain, an intracellular signaling domain responsible for T cell activation, and a hinge that joins these two domains. Continuous improvement has been made in CARs, now in their fifth generation, particularly in the intracellular signaling domain responsible for T cell activation. CAR T cell therapy has revolutionized the treatment of hematologic malignancies. Nonetheless, the use of CAR T cell therapy for solid tumors has not attained comparable levels of success. Here we review the challenges in achieving effective CAR T cell therapy in solid tumors, and emerging CAR T cells that have shown great promise for non-small cell lung cancer (NSCLC). A growing number of clinical trials have been conducted to study the effect of CAR T cell therapy on NSCLC, targeting different types of surface antigens. They include epidermal growth factor receptor (EGFR), mesothelin (MSLN), prostate stem cell antigen (PSCA), and mucin 1 (MUC1). Potential new targets such as erythropoietin-producing hepatocellular carcinoma A2 (EphA2), tissue factor (TF), and protein tyrosine kinase 7 (PTK7) are currently under investigation in clinical trials. The challenges in developing CAR T for NSCLC therapy and other approaches for enhancing CAR T efficacy are discussed. Finally, we provide our perspective on imaging CAR T cell action by reviewing the two main radionuclide-based CAR T cell imaging techniques, the direct labeling of CAR T cells or indirect labeling via a reporter gene.
Collapse
Affiliation(s)
- Hong Yun Ma
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Jeeban Das
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Conor Prendergast
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | | | - Brian Braumuller
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Jacienta Paily
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Sophia Huang
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Connie Liou
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Anna Giarratana
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Mahdie Hosseini
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| | - Randy Yeh
- Department of Radiology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY 10065, USA
| | - Kathleen M. Capaccione
- Department of Radiology, Columbia University Irving Medica Center, 622 W 168th St., New York, NY 10032, USA; (H.Y.M.); (J.P.); (M.H.)
| |
Collapse
|
40
|
Zheng R, Chen Y, Zhang Y, Liang S, Zhao X, Wang Y, Wang P, Meng R, Yang A, Yan B. Humanized single-domain antibody targeting HER2 enhances function of chimeric antigen receptor T cells. Front Immunol 2023; 14:1258156. [PMID: 38022548 PMCID: PMC10661930 DOI: 10.3389/fimmu.2023.1258156] [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: 07/13/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Introduction Chimeric antigen receptors (CARs) can redirect T cells against antigen-expressing tumors, and each component plays an important role in the function and anti-tumor efficacy. It has been reported that using human sequences or a low affinity of CAR single-chain variable fragments (scFvs) in the CAR binding domains is a potential way to enhance the function of CAR-T cells. However, it remains largely unknown how a lower affinity of CARs using humanized scFvs affects the function of CAR-T cells until recently. Methods We used different humanized anti-HER2 antibodies as the extracellular domain of CARs and further constructed a series of the CAR-T cells with different affinity. Results We have observed that moderately reducing the affinity of CARs (light chain variable domain (VL)-based CAR-T) could maintain the anti-tumor efficacy, and improved the safety of CAR therapy both in vitro and in vivo compared with high-affinity CAR-T cells. Moreover, T cells expressing the VL domain only antibody exhibited long-lasting tumor elimination capability after multiple challenges in vitro, longer persistence and lower cytokine levels in vivo. Discussion Our findings provide an alternative option for CAR-T optimization with the potential to widen the use of CAR T cells.
Collapse
Affiliation(s)
- Rui Zheng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yuankun Chen
- College of Life Science, Shaanxi Normal University, Xi’an, Shaanxi, China
| | - Yiting Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Sixin Liang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
- School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, Henan, China
| | - Xiaojuan Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Yiyi Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
- State Key Laboratory of Cellular Stress Biology, Innovation Center for Cell Signaling Network, School of Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Pengju Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
- Department of Immunology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Ruotong Meng
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
- College of Life Science, Yan’an University, Yan’an, Shaanxi, China
| | - Angang Yang
- Department of Immunology, Fourth Military Medical University, Xi’an, Shaanxi, China
| | - Bo Yan
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi’an, Shaanxi, China
| |
Collapse
|
41
|
Wu H, Yu Y, Zhao Y, Liu W, Liu Z, Zhang G, Chen Z. A C H2C H3 hinge region enhances the cytotoxicity of anti-CD5 CAR-T cells targeting T cell acute lymphoblastic leukemia. Int Immunopharmacol 2023; 124:110904. [PMID: 37690234 DOI: 10.1016/j.intimp.2023.110904] [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] [Received: 06/25/2023] [Revised: 08/26/2023] [Accepted: 09/04/2023] [Indexed: 09/12/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapies show considerable clinical efficacy in patients with B cell malignancies, but their efficacy is limited in patients with T cell acute lymphoblastic leukemia (T-ALL). CD5 is expressed on ∼85 % of malignant T cells, and CD5-targeting CAR-T cells can exhibit potent antitumor activity against T-ALL. However, optimization of CAR costimulatory endo-, hinge, and transmembrane domains could further increase their expansion and persistence, thereby enhancing their efficacy following exposure to tumor cells. Here we designed CD5-specific CARs with different molecular structures to generate CAR-T cells and investigated their anti-tumor efficacy in vitro and in vivo. CD5 CARs with a 4-1BB costimulatory domain (BB.z) or a CD28 costimulatory domain (28.z) exhibited specific cytotoxicity against CD5+ malignant cells in vitro. However, both failed to prolong the survival of T-ALL xenograft mice. Subsequently, we substituted the 28.z CAR hinge region with CH2CH3, which enhanced the ability of CH2CH3-CD5 CAR-T cells to specifically eradicate T-ALL cells in vitro and in vivo. Furthermore, patient-derived CH2CH3-CD5 CAR-T cells were generated which showed a marked killing effect of CD5-positive acute T-ALL cells in vitro. The anti-tumor activity of CD5 CAR-T cells with a CD28 co-stimulation domain and CH2CH3 hinge region was superior to those with BB.z and 28.z domains. These preclinical data provided new insights into the factors dictating efficacy in T-ALL treatment with CAR-T cells and hold promise for clinical translation.
Collapse
Affiliation(s)
- Huantong Wu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Yajie Yu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Yu Zhao
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Weihua Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Zhongfeng Liu
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Guangji Zhang
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China
| | - Zhiguo Chen
- Cell Therapy Center, Beijing Institute of Geriatrics, Xuanwu Hospital Capital Medical University, National Clinical Research Center for Geriatric Diseases, and Key Laboratory of Neurodegenerative Diseases, Ministry of Education, Beijing 100053, China; Center of Neural Injury and Repair, Beijing Institute for Brain Disorders, Beijing 100069, China.
| |
Collapse
|
42
|
Meng L, Yang Y, Mortazavi A, Zhang J. Emerging Immunotherapy Approaches for Treating Prostate Cancer. Int J Mol Sci 2023; 24:14347. [PMID: 37762648 PMCID: PMC10531627 DOI: 10.3390/ijms241814347] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 09/16/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023] Open
Abstract
Immunotherapy has emerged as an important approach for cancer treatment, but its clinical efficacy has been limited in prostate cancer compared to other malignancies. This review summarizes key immunotherapy strategies under evaluation for prostate cancer, including immune checkpoint inhibitors, bispecific T cell-engaging antibodies, chimeric antigen receptor (CAR) T cells, therapeutic vaccines, and cytokines. For each modality, the rationale stemming from preclinical studies is discussed along with outcomes from completed clinical trials and strategies to improve clinical efficacy that are being tested in ongoing clinical trials. Imperative endeavors include biomarker discovery for patient selection, deciphering resistance mechanisms, refining cellular therapies such as CAR T cells, and early-stage intervention were reviewed. These ongoing efforts instill optimism that immunotherapy may eventually deliver significant clinical benefits and expand treatment options for patients with advanced prostate cancer.
Collapse
Affiliation(s)
- Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (L.M.); (Y.Y.); (A.M.)
| | - Yuanquan Yang
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (L.M.); (Y.Y.); (A.M.)
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (L.M.); (Y.Y.); (A.M.)
| | - Jingsong Zhang
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center & Research Institute, University of South Florida, 12902 USF Magnolia Drive, Tampa, FL 33612, USA
| |
Collapse
|
43
|
Tang L, Huang Z, Mei H, Hu Y. Immunotherapy in hematologic malignancies: achievements, challenges and future prospects. Signal Transduct Target Ther 2023; 8:306. [PMID: 37591844 PMCID: PMC10435569 DOI: 10.1038/s41392-023-01521-5] [Citation(s) in RCA: 51] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 05/31/2023] [Accepted: 06/04/2023] [Indexed: 08/19/2023] Open
Abstract
The immune-cell origin of hematologic malignancies provides a unique avenue for the understanding of both the mechanisms of immune responsiveness and immune escape, which has accelerated the progress of immunotherapy. Several categories of immunotherapies have been developed and are being further evaluated in clinical trials for the treatment of blood cancers, including stem cell transplantation, immune checkpoint inhibitors, antigen-targeted antibodies, antibody-drug conjugates, tumor vaccines, and adoptive cell therapies. These immunotherapies have shown the potential to induce long-term remission in refractory or relapsed patients and have led to a paradigm shift in cancer treatment with great clinical success. Different immunotherapeutic approaches have their advantages but also shortcomings that need to be addressed. To provide clinicians with timely information on these revolutionary therapeutic approaches, the comprehensive review provides historical perspectives on the applications and clinical considerations of the immunotherapy. Here, we first outline the recent advances that have been made in the understanding of the various categories of immunotherapies in the treatment of hematologic malignancies. We further discuss the specific mechanisms of action, summarize the clinical trials and outcomes of immunotherapies in hematologic malignancies, as well as the adverse effects and toxicity management and then provide novel insights into challenges and future directions.
Collapse
Affiliation(s)
- Lu Tang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Zhongpei Huang
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China
| | - Heng Mei
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| | - Yu Hu
- Institute of Hematology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic Disease, 430022, Wuhan, China.
- Key Laboratory of Biological Targeted Therapy, the Ministry of Education, 430022, Wuhan, China.
- Hubei Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430022, Wuhan, China.
| |
Collapse
|
44
|
Schubert ML, Schmitt A, Hückelhoven-Krauss A, Neuber B, Kunz A, Waldhoff P, Vonficht D, Yousefian S, Jopp-Saile L, Wang L, Korell F, Keib A, Michels B, Haas D, Sauer T, Derigs P, Kulozik A, Kunz J, Pavel P, Laier S, Wuchter P, Schmier J, Bug G, Lang F, Gökbuget N, Casper J, Görner M, Finke J, Neubauer A, Ringhoffer M, Wolleschak D, Brüggemann M, Haas S, Ho AD, Müller-Tidow C, Dreger P, Schmitt M. Treatment of adult ALL patients with third-generation CD19-directed CAR T cells: results of a pivotal trial. J Hematol Oncol 2023; 16:79. [PMID: 37481608 PMCID: PMC10363324 DOI: 10.1186/s13045-023-01470-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Accepted: 06/20/2023] [Indexed: 07/24/2023] Open
Abstract
BACKGROUND Third-generation chimeric antigen receptor (CAR)-engineered T cells (CARTs) might improve clinical outcome of patients with B cell malignancies. This is the first report on a third-generation CART dose-escalating, phase-1/2 investigator-initiated trial treating adult patients with refractory and/or relapsed (r/r) acute lymphoblastic leukemia (ALL). METHODS Thirteen patients were treated with escalating doses of CD19-directed CARTs between 1 × 106 and 50 × 106 CARTs/m2. Leukapheresis, manufacturing and administration of CARTs were performed in-house. RESULTS For all patients, CART manufacturing was feasible. None of the patients developed any grade of Immune effector cell-associated neurotoxicity syndrome (ICANS) or a higher-grade (≥ grade III) catokine release syndrome (CRS). CART expansion and long-term CART persistence were evident in the peripheral blood (PB) of evaluable patients. At end of study on day 90 after CARTs, ten patients were evaluable for response: Eight patients (80%) achieved a complete remission (CR), including five patients (50%) with minimal residual disease (MRD)-negative CR. Response and outcome were associated with the administered CART dose. At 1-year follow-up, median overall survival was not reached and progression-free survival (PFS) was 38%. Median PFS was reached on day 120. Lack of CD39-expression on memory-like T cells was more frequent in CART products of responders when compared to CART products of non-responders. After CART administration, higher CD8 + and γδ-T cell frequencies, a physiological pattern of immune cells and lower monocyte counts in the PB were associated with response. CONCLUSION In conclusion, third-generation CARTs were associated with promising clinical efficacy and remarkably low procedure-specific toxicity, thereby opening new therapeutic perspectives for patients with r/r ALL. Trial registration This trial was registered at www. CLINICALTRIALS gov as NCT03676504.
Collapse
Affiliation(s)
- Maria-Luisa Schubert
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Anita Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Angela Hückelhoven-Krauss
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Brigitte Neuber
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Alexander Kunz
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Philip Waldhoff
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Dominik Vonficht
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Schayan Yousefian
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
| | - Lea Jopp-Saile
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Faculty of Biosciences, Heidelberg University, Heidelberg, Germany
| | - Lei Wang
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Felix Korell
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Anna Keib
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Birgit Michels
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Dominik Haas
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Tim Sauer
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Patrick Derigs
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
| | - Andreas Kulozik
- Department of Pediatric Hematology, Oncology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Joachim Kunz
- Department of Pediatric Hematology, Oncology and Immunology, University Hospital Heidelberg, Heidelberg, Germany
| | - Petra Pavel
- Institute for Clinical Transfusion Medicine and Cell Therapy (IKTZ), German Red Cross Blood Service Baden-Württemberg-Hessen, Heidelberg, Germany
| | - Sascha Laier
- Institute for Clinical Transfusion Medicine and Cell Therapy (IKTZ), German Red Cross Blood Service Baden-Württemberg-Hessen, Heidelberg, Germany
| | - Patrick Wuchter
- Institute of Transfusion Medicine and Immunology, Medical Faculty Mannheim, of the Heidelberg University, German Red Cross Blood Service Baden-Württemberg - Hessen, Mannheim, Germany
| | | | - Gesine Bug
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Fabian Lang
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Nicola Gökbuget
- Department of Internal Medicine II, University Hospital Frankfurt, Frankfurt, Germany
| | - Jochen Casper
- Department of Hematology and Oncology, University Hospital Oldenburg, Oldenburg, Germany
| | - Martin Görner
- Department of Hematology and Oncology, Hospital Bielefeld, Bielefeld, Germany
| | - Jürgen Finke
- Department of Internal Medicine I, University Hospital Freiburg, Freiburg, Germany
| | - Andreas Neubauer
- Department of Hematology, Oncology and Immunology, University Hospital Giessen und Marburg, Marburg, Germany
| | | | - Denise Wolleschak
- Department of Hematology and Oncology, Center of Internal Medicine, Otto-von-Guericke University Medical Center, Magdeburg, Germany
| | - Monika Brüggemann
- Department of Internal Medicine II, University Hospital Kiel, Kiel, Germany
| | - Simon Haas
- Heidelberg Institute for Stem Cell Technology and Experimental Medicine (HI-STEM gGmbH), Heidelberg, Germany
- Division of Stem Cells and Cancer, Deutsches Krebsforschungszentrum (DKFZ) and DKFZ-ZMBH Alliance, Heidelberg, Germany
- Berlin Institute of Health (BIH) at Charité - Universitätsmedizin Berlin, Berlin, Germany
- Berlin Institute for Medical Systems Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association, Berlin, Germany
- Department of Hematology, Oncology and Tumor Immunology, Charité University Medicine, Berlin, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Anthony D Ho
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Carsten Müller-Tidow
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Peter Dreger
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany
| | - Michael Schmitt
- Department of Internal Medicine V, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ)/National Center for Tumor Diseases (NCT), Heidelberg, Germany.
| |
Collapse
|
45
|
Hosoya H, Rodriguez-Otero P, Sidana S, Borrello IM. Embracing Myeloma Chimeric Antigen Receptor-T: From Scientific Design to Clinical Impact. Am Soc Clin Oncol Educ Book 2023; 43:e389860. [PMID: 37290016 DOI: 10.1200/edbk_389860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Despite recent advancement of treatment strategies in multiple myeloma (MM), patients with relapsed/refractory MM disease, particularly after triple-class refractoriness, continue to have poor prognosis. Chimeric antigen receptor (CAR-T) cells were developed and applied to improve outcomes in this setting, and two products, idecabtagene vicleucel and ciltacabtagene autoleucel, both targeting B-cell maturation antigen, have been approved by the Food and Drug Administration in the United States and European Medicines Agency in Europe. Both have shown unprecedented clinical outcomes with high response rate and prolonged progression-free survival and overall survival in this patient population with grim prognosis. Currently, further investigations are ongoing for CAR-T targeting different tumor antigens such as G protein-coupled receptor, class C, group 5, member D or with different combinations of intracellular signaling domains, as well as fourth-generation CAR-T with antigen-unrestricted inducible cytokines. Although CAR-T therapies hold hopes and enthusiasm from the myeloma community, several hurdles remain before these treatments become available for all patients in need. These barriers include CAR-T-cell manufacturing availability, access to administering centers, financial cost, caregivers' availability, and socioeconomic and racial disparities. Expanding clinical trial eligibility criteria and real-world data collection and analysis is crucial to understand the efficacy and safety of CAR-T in the patient cohort who tends to be excluded from current trials.
Collapse
Affiliation(s)
- Hitomi Hosoya
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
| | - Paula Rodriguez-Otero
- Clínica Universidad de Navarra, CCUN, Centro de investigación médica aplicada (Cima), IDISNA, CIBERONC, Pamplona, Spain
| | - Surbhi Sidana
- Division of Blood and Marrow Transplantation and Cellular Therapy, Stanford University School of Medicine, Stanford, CA
| | | |
Collapse
|
46
|
Wang C, Li Y, Gu L, Chen R, Zhu H, Zhang X, Zhang Y, Feng S, Qiu S, Jian Z, Xiong X. Gene Targets of CAR-T Cell Therapy for Glioblastoma. Cancers (Basel) 2023; 15:cancers15082351. [PMID: 37190280 DOI: 10.3390/cancers15082351] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2023] [Revised: 04/05/2023] [Accepted: 04/15/2023] [Indexed: 05/17/2023] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor with a poor prognosis following conventional therapeutic interventions. Moreover, the blood-brain barrier (BBB) severely impedes the permeation of chemotherapy drugs, thereby reducing their efficacy. Consequently, it is essential to develop novel GBM treatment methods. A novel kind of pericyte immunotherapy known as chimeric antigen receptor T (CAR-T) cell treatment uses CAR-T cells to target and destroy tumor cells without the aid of the antigen with great specificity and in a manner that is not major histocompatibility complex (MHC)-restricted. It has emerged as one of the most promising therapy techniques with positive clinical outcomes in hematological cancers, particularly leukemia. Due to its efficacy in hematologic cancers, CAR-T cell therapy could potentially treat solid tumors, including GBM. On the other hand, CAR-T cell treatment has not been as therapeutically effective in treating GBM as it has in treating other hematologic malignancies. CAR-T cell treatments for GBM have several challenges. This paper reviewed the use of CAR-T cell therapy in hematologic tumors and the selection of targets, difficulties, and challenges in GBM.
Collapse
Affiliation(s)
- Chaoqun Wang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
| | - Yuntao Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
| | - Lijuan Gu
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Ran Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Hua Zhu
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xu Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Yonggang Zhang
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Shi Feng
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sheng Qiu
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
- Huzhou Key Laboratory of Basic Research and Clinical Translation for Neuromodulation, Huzhou 313003, China
| | - Zhihong Jian
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiaoxing Xiong
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Department of Neurosurgery, Huzhou Central Hospital, Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou 310009, China
| |
Collapse
|
47
|
Pei K, Xu H, Wang P, Gan W, Hu Z, Su X, Zhang H, He Y. Anti-CLL1-based CAR T-cells with 4-1-BB or CD28/CD27 stimulatory domains in treating childhood refractory/relapsed acute myeloid leukemia. Cancer Med 2023; 12:9655-9661. [PMID: 37031462 PMCID: PMC10166968 DOI: 10.1002/cam4.5916] [Citation(s) in RCA: 23] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2022] [Revised: 03/06/2023] [Accepted: 03/27/2023] [Indexed: 04/11/2023] Open
Abstract
BACKGROUND Though the efficacy of anti C-type lectin-like molecule-1 (CLL1) CAR T-cells in refractory/relapsed acute myeloid leukemia (R/R-AML) have been occasionally reported, the influence of co-stimulatory domain CAR T-cells is not investigated so far. METHOD Seven R/R-AML children treated with anti-CLL1 CAR T-cells were enrolled onto this preliminary comparison study. Among these seven patients, four received CD28/CD27-based CAR T-cells therapy, and three received 4-1BB-based CAR T-cells therapy. RESULT The overall response rates were 75% and 66.7% in CD28/CD27 and 4-1BB group respectively. All patients experienced grade 1 to 2 cytokine release syndrome, with only one patient experiencing grade 2 immune effector cell-associated neurotoxicity syndrome. The maximum CAR T-cells durations were 156 and 274 days for CD28/CD27 group and 4-1BB group respectively. The 1-yr overall survival rate was 57.1%. CONCLUSIONS A preliminary similar efficacy/safety index was observed in anti-CLL1-based CAR T-cells with 4-1BB or CD28/CD27 co-stimulatory elements in treating pediatric R/R-AML.
Collapse
Affiliation(s)
- Kunlin Pei
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Guangzhou Medical University, Guangzhou, 511495, Guangdong, China
| | - Haoyu Xu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
- Guangzhou Medical University, Guangzhou, 511495, Guangdong, China
| | - Pengfei Wang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Wening Gan
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Zhengbin Hu
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Xiaoling Su
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Hui Zhang
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| | - Yingyi He
- Department of Hematology/Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, 510623, Guangdong, China
| |
Collapse
|
48
|
Zhang Y, Xu Y, Dang X, Zhu Z, Qian W, Liang A, Han W. Challenges and optimal strategies of CAR T therapy for hematological malignancies. Chin Med J (Engl) 2023; 136:269-279. [PMID: 36848181 PMCID: PMC10106177 DOI: 10.1097/cm9.0000000000002476] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Indexed: 03/01/2023] Open
Abstract
ABSTRACT Remarkable improvement relative to traditional approaches in the treatment of hematological malignancies by chimeric antigen receptor (CAR) T-cell therapy has promoted sequential approvals of eight commercial CAR T products within last 5 years. Although CAR T cells' productization is now rapidly boosting their extensive clinical application in real-world patients, the limitation of their clinical efficacy and related toxicities inspire further optimization of CAR structure and substantial development of innovative trials in various scenarios. Herein, we first summarized the current status and major progress in CAR T therapy for hematological malignancies, then described crucial factors which possibly compromise the clinical efficacies of CAR T cells, such as CAR T cell exhaustion and loss of antigen, and finally, we discussed the potential optimization strategies to tackle the challenges in the field of CAR T therapy.
Collapse
Affiliation(s)
- Yajing Zhang
- Department of Bio-Therapeutics, The First Medical Centre, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| | - Yang Xu
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Xiuyong Dang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai 200065, China
| | - Zeyu Zhu
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai 200065, China
| | - Wenbin Qian
- Department of Hematology, The Second Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310009, China
| | - Aibin Liang
- Department of Hematology, Tongji Hospital of Tongji University, Shanghai 200065, China
| | - Weidong Han
- Department of Bio-Therapeutics, The First Medical Centre, The General Hospital of Chinese People's Liberation Army, Beijing 100853, China
| |
Collapse
|
49
|
Dada R. Understanding the differences in outcome between CART studies as second-line treatment in aggressive lymphoma. J Oncol Pharm Pract 2023; 29:183-190. [PMID: 35786102 DOI: 10.1177/10781552221110806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Patients with refractory/early relapsed aggressive lymphomas belong to the most difficult-to-treat patients with hematological neoplasia. The prognosis of such patients is poor and novel treatment approches are urgently needed. Autologous chimeric antigen receptor T-cell therapy is a promising new therapy option that is approved after the failure of two lines of chemotherapy. Recently, the results of three different CART studies (ZUMA-7, TRANSFORM, and BELINDA) were published. Two of them were positive and one was negative, which created a mix of disappointment, confusion, and irritation. In this article, we are analyzing the data of all three trials and shed light on the differences between the studies which may facilitate an easier understanding of the results and relevance of CART in aggressive large B-cell lymphoma.
Collapse
Affiliation(s)
- Reyad Dada
- Department of Oncology, King Faisal Specialist Hospital and Research Centre, Jeddah, Kingdom of Saudi Arabia.,College of Medicine, Al-Faisal University, Riyadh, Kingdom of Saudi Arabia
| |
Collapse
|
50
|
Seng MS, Meierhofer AC, Lim FL, Soh SY, Hwang WYK. A Review of CAR-T Therapy in Pediatric and Young Adult B-Lineage Acute Leukemia: Clinical Perspectives in Singapore. Onco Targets Ther 2023; 16:165-176. [PMID: 36941828 PMCID: PMC10024535 DOI: 10.2147/ott.s271373] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Accepted: 01/07/2023] [Indexed: 03/15/2023] Open
Abstract
Approximately 10-15% of pediatric B-cell acute lymphoblastic leukemia (B-ALL) are high risk at diagnosis or relapsed/ refractory. Prior to the availability of chimeric antigen receptor T-cell (CAR-T) in Singapore and the region, the treatment options for these paediatric and young adults are conventional salvage chemotherapy or chemo-immunotherapy regimens as a bridge to allogeneic total body irradiation-based hematopoietic stem cell transplantation (allo-HSCT). This results in significant acute and long-term toxicities, with suboptimal survival outcomes. Finding a curative salvage therapy with fewer long-term toxicities would translate to improved quality-adjusted life years in these children and young adults. In this review, we focus on the burden of relapsed/refractory pediatric B-ALL, the limitations of current strategies, the emerging paradigms for the role of CAR-T in r/r B-ALL, our local perspectives on the health economics and future direction of CAR-T therapies in pediatric patients.
Collapse
Affiliation(s)
- Michaela S Seng
- Department of Paediatric Hematology and Oncology, KK Women’s and Children’s Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | | | - Francesca L Lim
- Duke-NUS Medical School, Singapore
- Department of Hematology, Singapore General Hospital, Singapore
| | - Shui Yen Soh
- Department of Paediatric Hematology and Oncology, KK Women’s and Children’s Hospital, Singapore
- Duke-NUS Medical School, Singapore
| | - William Y K Hwang
- Duke-NUS Medical School, Singapore
- Department of Hematology, Singapore General Hospital, Singapore
- National Cancer Centre Singapore, Singapore
- Correspondence: William YK Hwang, Department of Haematology, Singapore General Hospital, 31 Third Hospital Ave, 168753, Singapore, Tel +65 62223322, Email
| |
Collapse
|