1
|
Ismail AH, Khormi MA, Mawkili W, Albaqami A, Areshi S, Aborasain AM, Hegazy MM, Amin AH, Abo-Zaid MA. Harnessing the potential of gene-editing technology to overcome the current bottlenecks of CAR-T cell therapy in T-cell malignancies. Exp Hematol 2025; 146:104762. [PMID: 40122371 DOI: 10.1016/j.exphem.2025.104762] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/03/2025] [Accepted: 03/05/2025] [Indexed: 03/25/2025]
Abstract
T-cell malignancies (TCMs) include a diverse spectrum of hematologic cancers marked by complex biology and aggressive nature. Treating TCMs remains a critical unmet need in oncology with poor response to standard therapies. Chimeric antigen receptor (CAR)-T cell therapy is one of the most successful types of immunotherapy that has revolutionized cancer treatment, as evidenced by various approved products for CD19 B-cell malignancies and multiple myeloma. Nonetheless, due to some unique hurdles, such as the risk of CAR-T cell fratricide, product contamination with malignant cells, and severe T-cell aplasia, the translation of this treatment approach to TCMs has not been particularly successful. Moreover, irrespective of the type of treated cancer, CAR-T cell therapy can also present some complexities and potential side effects, such as cumbersome and costly manufacturing processes, impaired in vivo function, cytokine release syndrome (CRS), neurotoxicity, and leukemic transformation of CAR-T cells. Recent groundbreaking advances in gene-editing technology and the evolution of precise gene-editing tools such as the CRISPR/Cas9 system and its derivatives have opened a new way to overcoming the mentioned bottlenecks and paving the way for CAR-T cell therapy in TCMs. This review sheds light on how gene editing is being incorporated into CAR-T cell therapy to address current hurdles, enhance therapeutic efficacy, and improve the safety profile of CAR-T cell therapy in TCMs. Ongoing/conducted clinical trials are also discussed to provide a comprehensive view of the evolving landscape of genome-edited CAR-T cell therapy for TCMs.
Collapse
Affiliation(s)
- Ahmed H Ismail
- Department of Biology, College of Science, Jazan University, P.O. Box 114, 45142 Jazan, Kingdom of Saudi Arabia
| | - Mohsen A Khormi
- Department of Biology, College of Science, Jazan University, P.O. Box 114, 45142 Jazan, Kingdom of Saudi Arabia
| | - Wedad Mawkili
- Department of Pharmacology and Toxicology, College of Pharmacy, Jazan University, Jazan 45142, Saudi Arabia
| | - Amirah Albaqami
- Department of Clinical Laboratory Sciences, Turabah University College, Taif University, Taif, 21944, Saudi Arabia
| | - Sultan Areshi
- Department of Biology, College of Science, Jazan University, P.O. Box 114, 45142 Jazan, Kingdom of Saudi Arabia
| | - Ali M Aborasain
- Department of Biology, College of Science, Jazan University, P.O. Box 114, 45142 Jazan, Kingdom of Saudi Arabia
| | - Maysa M Hegazy
- Department of Biology, College of Science, Jazan University, P.O. Box 114, 45142 Jazan, Kingdom of Saudi Arabia
| | - Ali H Amin
- Zoology Department, Faculty of Science, Mansoura University, Mansoura, Egypt
| | - Mabrouk A Abo-Zaid
- Department of Biology, College of Science, Jazan University, P.O. Box 114, 45142 Jazan, Kingdom of Saudi Arabia.
| |
Collapse
|
2
|
Luan Y, Zhang Y, Li S, Gao C, Ying X, Zhao S, Zhang B. CD47 is a tumor cell-derived exosomal signature and regulates tumor immune microenvironment and immunotherapy responses. Transl Oncol 2025; 53:102291. [PMID: 39864342 PMCID: PMC11803903 DOI: 10.1016/j.tranon.2025.102291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/26/2024] [Accepted: 01/16/2025] [Indexed: 01/28/2025] Open
Abstract
BACKGROUND The pathogenesis of ovarian cancer (OvCa) involves a complex interplay of genetic, environmental, and hormonal factors. With the in-depth exploration of tumor ecosystem, exosomes can mediate the immunological status of tumor microenvironment (TME). Therefore, we aimed to recognize the tumor-derived exosomes (TEXs) which can distinguish the immune-hot and cold tumors and reflect the immunotherapeutic responses. METHODS A large set of transcriptomic and single-cell RNA-sequencing (scRNA-seq) datasets were downloaded and used to analyze the expression pattern of CD47 and its immuno-correlations in OvCa and multiple epithelial cell carcinomas such as breast cancers. In addition, a pan-gynecological cancer cohort was used to validate the correlation between CD47 and the inflamed TME. RESULTS In the current study, we found that CD47 was a TEX signature and had no transcriptional differences among patients with different clinicopathological features. Moreover, CD47 expression was positively correlated with the activation of immunological signaling pathways and enrichment of immune cell subpopulations in OvCa. Furthermore, in breast cancer and gynecological cancers, CD47, specially expressed in tumor cells, also showed favorable ability to distinguish the immune-hot and cold carcinomas. Moreover, in immunotherapy cohorts of breast cancer and other epithelial cell carcinomas, patients with CD47-high phenotype were more sensitive to immunotherapy and tended to achieve remission after treatment. Results from the TMA showed that CD47 was upregulated in tumor tissues and positively correlated with CD8 level. CONCLUSION In conclusion, CD47 is associated with an inflammatory TME, immune-hot tumors, and sensitivity of immunotherapy, highlighting the values of CD47 in identifying immunological traits and an immunotherapeutic response.
Collapse
Affiliation(s)
- Yifei Luan
- School of Innovation and Entrepreneurship, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Yinghui Zhang
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China
| | - Shangjin Li
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China
| | - Caiyun Gao
- Market Supervision and Law Enforcement Guarantee Service Center of Xihu District, Hangzhou 310013, PR China
| | - Xinyi Ying
- Department of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, PR China
| | - Shaojie Zhao
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China.
| | - Bing Zhang
- Wuxi Maternal and Child Health Care Hospital, The Affiliated Women's Hospital of Jiangnan University, Wuxi 214002, PR China.
| |
Collapse
|
3
|
Jing Y, Li B, Aierken A, Zhang Z, Han D, Lin Z, Gao J, Tian H, Hua J. Mesenchymal Stem Cells with Simultaneous Overexpression of GPX3 and CD47 for the Treatment of Drug-Induced Acute Liver Injury. Vet Sci 2025; 12:149. [PMID: 40005909 PMCID: PMC11861084 DOI: 10.3390/vetsci12020149] [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: 11/20/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/27/2025] Open
Abstract
The liver, as the largest metabolic and detoxification organ in mammals, metabolizes approximately 80-90% of drugs. However, drug-induced liver injury (DILI) is common and driven by factors such as individual variability, differences in liver metabolism, and improper drug use. Mesenchymal stem cells (MSCs), with their self-renewal and multipotent differentiation capabilities, offer therapeutic potential, but face challenges such as limited proliferation and increased apoptosis during in vitro expansion. Although MSCs exhibit low immunogenicity, they are often cleared by the host immune system, which limits their survival and engraftment. Glutathione peroxidase 3 (GPX3) is a key antioxidant enzyme that reduces reactive oxygen species (ROS), protecting cells from oxidative damage. CD47, also known as integrin-associated protein (IAP), helps cells evade immune clearance by binding to signal regulatory protein alpha (SIRPα) on the immune cells. Here, we used an acetaminophen (APAP)-induced DILI mouse model to evaluate the therapeutic efficacy of intravenously infused MSCs overexpressing GPX3 and CD47. Compared to unmodified MSCs, modified MSCs showed improved survival, reduced liver inflammation, and alleviated oxidative damage, offering enhanced protection against APAP-induced DILI.
Collapse
Affiliation(s)
- Yuanxiang Jing
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Balun Li
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Aili Aierken
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
- Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Xinjiang Medical University, Urumqi 830000, China
| | - Zengyu Zhang
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Dongyao Han
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Zixi Lin
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Jiaqi Gao
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Hongkai Tian
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| | - Jinlian Hua
- College of Veterinary Medicine, Shanxi Centre of Stem Cells Engineering & Technology, Northwest A&F University, Yangling 712100, China; (Y.J.); (B.L.); (A.A.); (Z.Z.); (D.H.); (Z.L.); (J.G.); (H.T.)
| |
Collapse
|
4
|
Zhang Z, Zhao L, Huang T, Chen Z, Zhao Y, Liang J, Ao X, Jia X, Kang L, Kong L, Jing Q, Hu J, Gu L, Pan F, Hu Z, He L, Zhou M, Chen J, Guo Z. A self-activated and protective module enhances the preclinical performance of allogeneic anti-CD70 CAR-T cells. Front Immunol 2025; 15:1531294. [PMID: 39906740 PMCID: PMC11792090 DOI: 10.3389/fimmu.2024.1531294] [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: 11/20/2024] [Accepted: 12/30/2024] [Indexed: 02/06/2025] Open
Abstract
Introduction Allogeneic chimeric antigen receptor T (CAR-T) therapy, also known as universal CAR-T (UCAR-T) therapy, offers broad applicability, high production efficiency, and reduced costs, enabling quicker access for patients. However, clinical application remains limited by challenges such as immune rejection, and issues with potency and durability. Methods We first screened a safe and effective anti-CD70 scFv to construct anti-CD70 CAR-T cells. Anti-CD70 UCAR-T cells were then generated by knocking out TRAC, B2M, and HLA-DRA. To address the limitations of UCAR-T therapy, we developed an 'all-in-one' self-activated and protective (SAP) module, integrated into the CAR scaffold. The SAP module consists of the CD47 extracellular domain, a mutant interleukin 7 receptor alpha (IL7Rα) transmembrane domain, and the IL7Rα intracellular domain, designed to protect UCAR-T cells from host immune attacks and enhance their survival. Results SAP UCAR-T cells demonstrated significantly reduced immune rejection from the innate immune system, as evidenced by enhanced survival and functionality both in vitro and in vivo. The modified UCAR-T cells exhibited improved persistence, with no observed safety concerns. Furthermore, SAP UCAR-T cells maintained process stability during scale-up production, indicating the potential for large-scale manufacturing. Discussion Our findings highlight the SAP module as a promising strategy for the preclinical development of anti-CD70 UCAR-T, paving the way for an 'off-the-shelf' cell therapy product.
Collapse
Affiliation(s)
- Zhao Zhang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lianfeng Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Tinghui Huang
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhengliang Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Yaoyao Zhao
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Junqing Liang
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xudong Ao
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Xiaoqiong Jia
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Lei Kang
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Linghui Kong
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Qi Jing
- Peking University Cancer Hospital (Inner Mongolia Campus), Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot, Inner Mongolia Autonomous Region, China
| | - Jianhua Hu
- Center of Biotherapy, Jiangsu Province Geriatric Hospital, Nanjing, China
| | - Lili Gu
- Department of Research and Development, Nanjing Calmhome Cell & Gene Engineering Institute Co., Ltd., Nanjing, China
| | - Feiyan Pan
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Hu
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Lingfeng He
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Muya Zhou
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Jiannan Chen
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| | - Zhigang Guo
- Jiangsu Key Laboratory for Molecular and Medical Biotechnology, College of Life Sciences, Nanjing Normal University, Nanjing, China
| |
Collapse
|
5
|
Canciani G, Fabozzi F, Pinacchio C, Ceccarelli M, Del Bufalo F. Developing CAR T-Cell Therapies for Pediatric Solid Tumors. Paediatr Drugs 2025; 27:5-18. [PMID: 39382819 DOI: 10.1007/s40272-024-00653-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 10/10/2024]
Abstract
Chimeric antigen receptor (CAR) T cells have revolutionized the treatment of hematological malignancies, inducing notable and durable clinical responses. However, for solid tumors, including but not limited to pediatric tumors, several peculiar biological features posed substantial challenges for achieving comparable results. Despite sound pre-clinical evidence of the ability of CAR T cells to eradicate solid malignancies, their activity remains suboptimal when facing the in vivo complexity of solid tumors, characterized by antigen heterogeneity, scarce T-cell infiltration, and an immunosuppressive microenvironment. Neuroblastoma was amongst the first tumors to be evaluated as a potential candidate for GD2-targeting CAR T cells, which recently documented promising results in high-risk, heavily pre-treated patients. Moreover, innovative engineering strategies for generating more potent and persistent CAR T cells suggest the possibility to reproduce, and potentially improve, these promising results on a larger scale. In the next years, harnessing the full therapeutic potential of CAR T cells and other immunotherapeutic strategies may open new possibilities for effectively treating the most aggressive forms of pediatric tumors.
Collapse
Affiliation(s)
- Gabriele Canciani
- Department of Hematology, Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
- Residency School of Pediatrics, University of Rome Tor Vergata, Rome, Italy
| | - Francesco Fabozzi
- Department of Hematology, Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Claudia Pinacchio
- Department of Hematology, Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Manuela Ceccarelli
- Department of Hematology, Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy
| | - Francesca Del Bufalo
- Department of Hematology, Oncology and Cell and Gene Therapy, IRCCS Bambino Gesù Children's Hospital, Rome, Italy.
| |
Collapse
|
6
|
Yin Z, Huang Y, Zhu Y, Zhong Q, Shen H, Mahati S, Huang J, Li G, Ou R, Liu Z, Zhang Q, Liu S. Identification and analysis of microplastic aggregation in CAR-T cells. JOURNAL OF HAZARDOUS MATERIALS 2024; 480:136351. [PMID: 39488976 DOI: 10.1016/j.jhazmat.2024.136351] [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: 06/12/2024] [Revised: 10/22/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Microplastics (MPs) are increasingly recognized as contaminants present in various environments and are widely acknowledged as potential hazards to the mammalian immune system. In our study of chimeric antigen receptor T cell (CAR-T) therapy, we observed the presence of MP in CAR-T cell products for the first time. It is worth exploring whether MP could enter CAR-T cells and how they might affect CAR-T cells' functionality. Therefore, we analyzed how MP affected CD19 and BCMA-CAR-T cells. Based on flow cytometry, ELISA, and cytotoxicity analysis of in vitro and in vivo experiments, MP suppressed the activity of CAR-T cells. Subsequent investigation revealed that the exposure of CAR-T cells to varying concentrations of MP resulted in a notable increase in apoptosis, ferroptosis, and exhaustion levels. Furthermore, the hyperactivation of the mTOR signaling pathway in MP-treated CAR-T cells was verified. The partial restoration of CAR-T cell function in MP was achieved by inhibiting the mTOR pathway. MP present a threat to CAR-T cell function due to their role in inducing CAR-T cell apoptosis, ferroptosis, and T-cell exhaustion through the hyperactivation of mTOR signaling pathways.
Collapse
Affiliation(s)
- Zhao Yin
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Yizhen Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Yangmin Zhu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Qi Zhong
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Huijuan Shen
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Shaya Mahati
- Department of Tumor center, First Affiliated Hospital of Xinjiang Medical University, State key laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Centra Asia, Xinjiang Province 830054, China
| | - Jing Huang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Guangchao Li
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China
| | - Ruiming Ou
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Zhi Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Qing Zhang
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| | - Shuang Liu
- Department of Hematology, Guangdong Second Provincial General Hospital, Jinan university, Guangzhou, Guangdong Province 510317, China.
| |
Collapse
|
7
|
Christo SN, McDonald KM, Burn TN, Kurd N, Stanfield J, Kaneda MM, Seelige R, Dillon CP, Fisher TS, Baaten B, Mackay LK. Dual CD47 and PD-L1 blockade elicits anti-tumor immunity by intratumoral CD8 + T cells. Clin Transl Immunology 2024; 13:e70014. [PMID: 39584189 PMCID: PMC11583082 DOI: 10.1002/cti2.70014] [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: 07/11/2024] [Revised: 10/10/2024] [Accepted: 11/08/2024] [Indexed: 11/26/2024] Open
Abstract
Objectives Bispecific antibodies targeting CD47 and PD-L1 (CD47 × PD-L1 BisAb) demonstrate efficacy against a range of solid cancers. While dual blockade negates anti-CD47-mediated toxicity, the effect of combined innate and adaptive immune activation on protective tumor-resident CD8+ T cells has yet to be fully elucidated. Methods CD8+ T cell populations were tracked upon CD47 × PD-L1 BisAb treatment in an orthotopic model of murine breast cancer where anti-tumor immunity is mediated by CD8+ T cells. Immune responses were also compared with anti-PD-L1 monotherapy to assess the advantage of dual checkpoint targeting. Results We found that CD47 × PD-L1 BisAb treatment augmented CD8+ T cell responses in tumors, which resulted in enhanced tumor control. Compared with anti-PD-L1 treatment, dual CD47 and PD-L1 blockade promoted greater numbers of antigen-specific tumor-resident CD8+ T cells that exhibited increased cytokine production. Conclusions Engagement of innate and adaptive immune checkpoint molecules via CD47 × PD-L1 BisAb treatment resulted in robust CD8+ T cell responses, including the induction of tumor-resident CD8+ T cells that exhibited functionally superior anti-tumor immunity. These results demonstrate that innate immune activation potentiates anti-tumor adaptive responses, highlighting the use of dual checkpoint blockade as an optimal strategy for promoting CD8+ T cell-mediated protection.
Collapse
Affiliation(s)
- Susan N Christo
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Keely M McDonald
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Thomas N Burn
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| | - Nadia Kurd
- Oncology Research UnitPfizer Inc.San DiegoCAUSA
| | | | | | | | | | | | - Bas Baaten
- Oncology Research UnitPfizer Inc.San DiegoCAUSA
| | - Laura K Mackay
- Department of Microbiology and ImmunologyThe University of Melbourne at the Peter Doherty Institute for Infection and ImmunityMelbourneAustralia
| |
Collapse
|
8
|
Martins TA, Kaymak D, Tatari N, Gerster F, Hogan S, Ritz MF, Sabatino V, Wieboldt R, Bartoszek EM, McDaid M, Gerber A, Buck A, Beshirova A, Heider A, Shekarian T, Mohamed H, Etter MM, Schmassmann P, Abel I, Boulay JL, Saito Y, Mariani L, Guzman R, Snijder B, Weiss T, Läubli H, Hutter G. Enhancing anti-EGFRvIII CAR T cell therapy against glioblastoma with a paracrine SIRPγ-derived CD47 blocker. Nat Commun 2024; 15:9718. [PMID: 39521782 PMCID: PMC11550474 DOI: 10.1038/s41467-024-54129-w] [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: 10/25/2023] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
A significant challenge for chimeric antigen receptor (CAR) T cell therapy against glioblastoma (GBM) is its immunosuppressive microenvironment, which is densely populated by protumoral glioma-associated microglia and macrophages (GAMs). Myeloid immune checkpoint therapy targeting the CD47-signal regulatory protein alpha (SIRPα) axis induces GAM phagocytic function, but CD47 blockade monotherapy is associated with toxicity and low bioavailability in solid tumors. In this work, we engineer a CAR T cell against epidermal growth factor receptor variant III (EGFRvIII), constitutively secreting a signal regulatory protein gamma-related protein (SGRP) with high affinity to CD47. Anti-EGFRvIII-SGRP CAR T cells eradicate orthotopic EGFRvIII-mosaic GBM in vivo, promoting GAM-mediated tumor cell phagocytosis. In a subcutaneous CD19+ lymphoma mouse model, anti-CD19-SGRP CAR T cell therapy is superior to conventional anti-CD19 CAR T. Thus, combination of CAR and SGRP eliminates bystander tumor cells in a manner that could overcome main mechanisms of CAR T cell therapy resistance, including immune suppression and antigen escape.
Collapse
Affiliation(s)
- Tomás A Martins
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Deniz Kaymak
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Nazanin Tatari
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Fiona Gerster
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Sabrina Hogan
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marie-Françoise Ritz
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Valerio Sabatino
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ronja Wieboldt
- Cancer Immunotherapy, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ewelina M Bartoszek
- Microscopy Core Facility, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marta McDaid
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alexandra Gerber
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Alicia Buck
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Aisha Beshirova
- Experimental Immunology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Anja Heider
- Swiss Institute of Allergy and Asthma Research, University of Zurich, Davos Wolfgang, Switzerland
| | - Tala Shekarian
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Hayget Mohamed
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Manina M Etter
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
| | - Philip Schmassmann
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Ines Abel
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Jean-Louis Boulay
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Yasuyuki Saito
- Division of Molecular and Cellular Signaling, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Kobe, Japan
| | - Luigi Mariani
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Raphael Guzman
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland
- Department of Surgery, University Hospital Basel, Basel, Switzerland
| | - Berend Snijder
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Tobias Weiss
- Department of Neurology, Clinical Neuroscience Center, University Hospital Zurich, Zurich, Switzerland
- Department of Neurology, University of Zurich, Zurich, Switzerland
| | - Heinz Läubli
- Cancer Immunotherapy, Department of Biomedicine, University of Basel, Basel, Switzerland
- Department of Oncology, University Hospital Basel, Basel, Switzerland
| | - Gregor Hutter
- Brain Tumor Immunotherapy and Biology, Department of Biomedicine, University of Basel, Basel, Switzerland.
- Department of Neurosurgery, University Hospital Basel, Basel, Switzerland.
- Department of Surgery, University Hospital Basel, Basel, Switzerland.
| |
Collapse
|
9
|
Liu Y, Weng L, Wang Y, Zhang J, Wu Q, Zhao P, Shi Y, Wang P, Fang L. Deciphering the role of CD47 in cancer immunotherapy. J Adv Res 2024; 63:129-158. [PMID: 39167629 PMCID: PMC11380025 DOI: 10.1016/j.jare.2023.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Revised: 10/05/2023] [Accepted: 10/18/2023] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Immunotherapy has emerged as a novel strategy for cancer treatment following surgery, radiotherapy, and chemotherapy. Immune checkpoint blockade and Chimeric antigen receptor (CAR)-T cell therapies have been successful in clinical trials. Cancer cells evade immune surveillance by hijacking inhibitory pathways via overexpression of checkpoint genes. The Cluster of Differentiation 47 (CD47) has emerged as a crucial checkpoint for cancer immunotherapy by working as a "don't eat me" signal and suppressing innate immune signaling. Furthermore, CD47 is highly expressed in many cancer types to protect cancer cells from phagocytosis via binding to SIRPα on phagocytes. Targeting CD47 by either interrupting the CD47-SIRPα axis or combing with other therapies has been demonstrated as an encouraging therapeutic strategy in cancer immunotherapy. Antibodies and small molecules that target CD47 have been explored in pre- and clinical trials. However, formidable challenges such as the anemia and palate aggregation cannot be avoided because of the wide presentation of CD47 on erythrocytes. AIM OF VIEW This review summarizes the current knowledge on the regulation and function of CD47, and provides a new perspective for immunotherapy targeting CD47. It also highlights the clinical progress of targeting CD47 and discusses challenges and potential strategies. KEY SCIENTIFIC CONCEPTS OF REVIEW This review provides a comprehensive understanding of targeting CD47 in cancer immunotherapy, it also augments the concept of combination immunotherapy strategies by employing both innate and adaptive immune responses.
Collapse
Affiliation(s)
- Yu'e Liu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Linjun Weng
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yanjin Wang
- Department of Nephrology, Shanghai East Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Jin Zhang
- Department of Pharmacology and Toxicology, University of Mississippi, Medical Center, 39216 Jackson, MS, USA
| | - Qi Wu
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China
| | - Pengcheng Zhao
- School of Life Sciences and Medicine, Shandong University of Technology, No.266 Xincun West Road, Zibo 255000, Shandong Province, China
| | - Yufeng Shi
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China; Clinical Center for Brain and Spinal Cord Research, Tongji University, Shanghai 200092, China.
| | - Ping Wang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| | - Lan Fang
- Tongji University Cancer Center, Shanghai Tenth People's Hospital of Tongji University, School of Medicine, Tongji University, Shanghai 200092, China.
| |
Collapse
|
10
|
Lee HJ, Hwang SJ, Jeong EH, Chang MH. Genetically Engineered CLDN18.2 CAR-T Cells Expressing Synthetic PD1/CD28 Fusion Receptors Produced Using a Lentiviral Vector. J Microbiol 2024; 62:555-568. [PMID: 38700775 PMCID: PMC11303488 DOI: 10.1007/s12275-024-00133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 08/07/2024]
Abstract
This study aimed to develop synthetic Claudin18.2 (CLDN18.2) chimeric antigen receptor (CAR)-T (CAR-T) cells as a treatment for advanced gastric cancer using lentiviral vector genetic engineering technology that targets the CLDN18.2 antigen and simultaneously overcomes the immunosuppressive environment caused by programmed cell death protein 1 (PD-1). Synthetic CAR T cells are a promising approach in cancer immunotherapy but face many challenges in solid tumors. One of the major problems is immunosuppression caused by PD-1. CLDN18.2, a gastric-specific membrane protein, is considered a potential therapeutic target for gastric and other cancers. In our study, CLDN18.2 CAR was a second-generation CAR with inducible T-cell costimulatory (CD278), and CLDN18.2-PD1/CD28 CAR was a third-generation CAR, wherein the synthetic PD1/CD28 chimeric-switch receptor (CSR) was added to the second-generation CAR. In vitro, we detected the secretion levels of different cytokines and the killing ability of CAR-T cells. We found that the secretion of cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) secreted by three types of CAR-T cells was increased, and the killing ability against CLDN18.2-positive GC cells was enhanced. In vivo, we established a xenograft GC model and observed the antitumor effects and off-target toxicity of CAR-T cells. These results support that synthetic anti-CLDN18.2 CAR-T cells have antitumor effect and anti-CLDN18.2-PD1/CD28 CAR could provide a promising design strategy to improve the efficacy of CAR-T cells in advanced gastric cancer.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Cell Line, Tumor
- Claudins/genetics
- Claudins/metabolism
- Cytokines/metabolism
- Genetic Engineering
- Genetic Vectors/genetics
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- Lentivirus/genetics
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Stomach Neoplasms/therapy
- Stomach Neoplasms/immunology
- Stomach Neoplasms/genetics
- T-Lymphocytes/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Heon Ju Lee
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea.
| | - Seo Jin Hwang
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| | - Eun Hee Jeong
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| | - Mi Hee Chang
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| |
Collapse
|
11
|
Sue M, Tsubaki T, Ishimoto Y, Hayashi S, Ishida S, Otsuka T, Isumi Y, Kawase Y, Yamaguchi J, Nakada T, Ishiguro J, Nakamura K, Kawaida R, Ohtsuka T, Wada T, Agatsuma T, Kawasaki N. Blockade of SIRPα-CD47 axis by anti-SIRPα antibody enhances anti-tumor activity of DXd antibody-drug conjugates. PLoS One 2024; 19:e0304985. [PMID: 38843278 PMCID: PMC11156334 DOI: 10.1371/journal.pone.0304985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/21/2024] [Indexed: 06/09/2024] Open
Abstract
Signal regulatory protein alpha (SIRPα) is an immune inhibitory receptor on myeloid cells including macrophages and dendritic cells, which binds to CD47, a ubiquitous self-associated molecule. SIRPα-CD47 interaction is exploited by cancer cells to suppress anti-tumor activity of myeloid cells, therefore emerging as a novel immune checkpoint for cancer immunotherapy. In blood cancer, several SIRPα-CD47 blockers have shown encouraging monotherapy activity. However, the anti-tumor activity of SIRPα-CD47 blockers in solid tumors seems limited, suggesting the need for combination therapies to fully exploit the myeloid immune checkpoint in solid tumors. Here we tested whether combination of SIRPα-CD47 blocker with antibody-drug conjugate bearing a topoisomerase I inhibitor DXd (DXd-ADC) would enhance anti-tumor activity in solid tumors. To this end, DS-1103a, a newly developed anti-human SIRPα antibody (Ab), was assessed for the potential combination benefit with datopotamab deruxtecan (Dato-DXd) and trastuzumab deruxtecan (T-DXd), DXd-ADCs targeting human trophoblast cell-surface antigen 2 and human epidermal growth factor receptor 2, respectively. DS-1103a inhibited SIRPα-CD47 interaction and enhanced antibody-dependent cellular phagocytosis of Dato-DXd and T-DXd against human cancer cells. In a whole cancer cell vaccination model, vaccination with DXd-treated cancer cells led to activation of tumor-specific T cells when combined with an anti-mouse SIRPα (anti-mSIRPα) Ab, implying the benefit of combining DXd-ADCs with anti-SIRPα Ab on anti-tumor immunity. Furthermore, in syngeneic mouse models, both Dato-DXd and T-DXd combination with anti-mSIRPα Ab showed stronger anti-tumor activity over the monotherapies. Taken together, this study provides a preclinical rationale of novel therapies for solid tumors combining SIRPα-CD47 blockers with DXd-ADCs.
Collapse
Affiliation(s)
- Mayumi Sue
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takuya Tsubaki
- Modality Research Laboratories III, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yoko Ishimoto
- Translational Science Department I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Shinko Hayashi
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Saori Ishida
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takafumi Otsuka
- Research Innovation Planning Department, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yoshitaka Isumi
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Yumi Kawase
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Junko Yamaguchi
- Discovery Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Takashi Nakada
- Modality Research Laboratories I, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Jun Ishiguro
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Kensuke Nakamura
- Modality Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Reimi Kawaida
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Toshiaki Ohtsuka
- Discovery Research Laboratories V, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | - Teiji Wada
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| | | | - Norihito Kawasaki
- Discovery Research Laboratories II, Daiichi Sankyo Co., Ltd., Tokyo, Japan
| |
Collapse
|
12
|
Yamada-Hunter SA, Theruvath J, McIntosh BJ, Freitas KA, Lin F, Radosevich MT, Leruste A, Dhingra S, Martinez-Velez N, Xu P, Huang J, Delaidelli A, Desai MH, Good Z, Polak R, May A, Labanieh L, Bjelajac J, Murty T, Ehlinger Z, Mount CW, Chen Y, Heitzeneder S, Marjon KD, Banuelos A, Khan O, Wasserman SL, Spiegel JY, Fernandez-Pol S, Kuo CJ, Sorensen PH, Monje M, Majzner RG, Weissman IL, Sahaf B, Sotillo E, Cochran JR, Mackall CL. Engineered CD47 protects T cells for enhanced antitumour immunity. Nature 2024; 630:457-465. [PMID: 38750365 PMCID: PMC11168929 DOI: 10.1038/s41586-024-07443-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 04/18/2024] [Indexed: 06/14/2024]
Abstract
Adoptively transferred T cells and agents designed to block the CD47-SIRPα axis are promising cancer therapeutics that activate distinct arms of the immune system1,2. Here we administered anti-CD47 antibodies in combination with adoptively transferred T cells with the goal of enhancing antitumour efficacy but observed abrogated therapeutic benefit due to rapid macrophage-mediated clearance of T cells expressing chimeric antigen receptors (CARs) or engineered T cell receptors. Anti-CD47-antibody-mediated CAR T cell clearance was potent and rapid enough to serve as an effective safety switch. To overcome this challenge, we engineered the CD47 variant CD47(Q31P) (47E), which engages SIRPα and provides a 'don't eat me' signal that is not blocked by anti-CD47 antibodies. TCR or CAR T cells expressing 47E are resistant to clearance by macrophages after treatment with anti-CD47 antibodies, and mediate substantial, sustained macrophage recruitment to the tumour microenvironment. Although many of the recruited macrophages manifested an M2-like profile3, the combined therapy synergistically enhanced antitumour efficacy. Our study identifies macrophages as major regulators of T cell persistence and illustrates the fundamental challenge of combining T-cell-directed therapeutics with those designed to activate macrophages. It delivers a therapeutic approach that is capable of simultaneously harnessing the antitumour effects of T cells and macrophages, offering enhanced potency against solid tumours.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Antigens, Differentiation/immunology
- Antigens, Differentiation/metabolism
- CD47 Antigen/genetics
- CD47 Antigen/immunology
- CD47 Antigen/metabolism
- Cell Line, Tumor
- Immunotherapy, Adoptive/methods
- Macrophages/cytology
- Macrophages/immunology
- Neoplasms/immunology
- Neoplasms/metabolism
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes/transplantation
- Tumor Microenvironment/immunology
- Antibodies/immunology
- Antibodies/therapeutic use
- Macrophage Activation
Collapse
Affiliation(s)
- Sean A Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
| | - Johanna Theruvath
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Brianna J McIntosh
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Katherine A Freitas
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Frank Lin
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Masters in Translational Research and Applied Medicine Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Molly T Radosevich
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Amaury Leruste
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Shaurya Dhingra
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Naiara Martinez-Velez
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Peng Xu
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jing Huang
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Moksha H Desai
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Zinaida Good
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Biomedical Data Science, Stanford University School of Medicine, Stanford, CA, USA
| | - Roel Polak
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, The Netherlands
| | - Audre May
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
| | - Louai Labanieh
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Jeremy Bjelajac
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
| | - Tara Murty
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Program in Biophysics, Stanford University, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
| | - Zach Ehlinger
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher W Mount
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
- Neurosciences Program, Stanford University, Stanford, CA, USA
| | - Yiyun Chen
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Sabine Heitzeneder
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Kristopher D Marjon
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Allison Banuelos
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Omair Khan
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Savannah L Wasserman
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jay Y Spiegel
- Sylvester Comprehensive Cancer Center, University of Miami, Miami, FL, USA
| | | | - Calvin J Kuo
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Poul H Sorensen
- British Columbia Cancer Agency, Vancouver, British Columbia, Canada
| | - Michelle Monje
- Medical Scientist Training Program, Stanford University, Stanford, CA, USA
- Department of Neurology, Stanford University School of Medicine, Stanford, CA, USA
- Neurosciences Program, Stanford University, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Robbie G Majzner
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA
| | - Irving L Weissman
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, USA
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Bita Sahaf
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Jennifer R Cochran
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA
- Immunology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Parker Institute for Cancer Immunotherapy, San Francisco, CA, USA.
- Cancer Biology Program, Stanford University School of Medicine, Stanford, CA, USA.
- Immunology Graduate Program, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA.
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, USA.
- Ludwig Center for Cancer Stem Cell Research and Medicine, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
13
|
Wu F, Pang H, Li F, Hua M, Song C, Tang J. Progress in cancer research on the regulator of phagocytosis CD47, which determines the fate of tumor cells (Review). Oncol Lett 2024; 27:256. [PMID: 38646501 PMCID: PMC11027102 DOI: 10.3892/ol.2024.14389] [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: 12/14/2023] [Accepted: 03/21/2024] [Indexed: 04/23/2024] Open
Abstract
Cluster of differentiation 47 (CD47) is a transmembrane protein that is widely and moderately expressed on the surface of various cells and can have an essential role in mediating cell proliferation, migration, phagocytosis, apoptosis, immune homeostasis and other related responses by binding to its ligands, integrins, thrombospondin-1 and signal regulatory protein α. The poor prognosis of cancer patients is closely associated with high expression of CD47 in glioblastoma, ovarian cancer, breast cancer, bladder cancer, colon cancer and hepatocellular carcinoma. Upregulation of CD47 expression facilitates the growth of numerous types of tumor cells, while downregulation of its expression promotes phagocytosis of tumor cells by macrophages, thereby limiting tumor growth. In addition, blocking CD47 activates the cyclic GMP-AMP (cGAMP) synthase/cGAMP/interferon gene stimulating factor signaling pathway and initiates an adaptive immune response that kills tumor cells. The present review describes the structure, function and interactions of CD47 with its ligands, as well as its regulation of phagocytosis and tumor cell fate. It summarizes the therapeutics, mechanisms of action, research advances and challenges of targeting CD47. In addition, this paper provides an overview of the latest therapeutic options for targeting CD47, such as chimeric antigen receptor (CAR) T-cells, CAR macrophages and nanotechnology-based delivery systems, which are essential for future clinical research on targeting CD47.
Collapse
Affiliation(s)
- Fan Wu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Hongyuan Pang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Fan Li
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Mengqing Hua
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Chuanwang Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| | - Jie Tang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Department of Immunology, School of Laboratory Medicine, Bengbu Medical College, Bengbu, Anhui 233030, P.R. China
| |
Collapse
|
14
|
Stefanidis E, Semilietof A, Pujol J, Seijo B, Scholten K, Zoete V, Michielin O, Sandaltzopoulos R, Coukos G, Irving M. Combining SiRPα decoy-coengineered T cells and antibodies augments macrophage-mediated phagocytosis of tumor cells. J Clin Invest 2024; 134:e161660. [PMID: 38828721 PMCID: PMC11142748 DOI: 10.1172/jci161660] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 04/16/2024] [Indexed: 06/05/2024] Open
Abstract
The adoptive transfer of T cell receptor-engineered (TCR-engineered) T cells (ACT) targeting the HLA-A2-restricted cancer-testis epitope NY-ESO-1157-165 (A2/NY) has yielded favorable clinical responses against several cancers. Two approaches to improve ACT are TCR affinity optimization and T cell coengineering to express immunomodulatory molecules that can exploit endogenous immunity. By computational design we previously developed a panel of binding-enhanced A2/NY-TCRs including A97L, which augmented the in vitro function of gene-modified T cells as compared with WT. Here, we demonstrated higher persistence and improved tumor control by A97L-T cells. In order to harness macrophages in tumors, we further coengineered A97L-T cells to secrete a high-affinity signal regulatory protein α (SiRPα) decoy (CV1) that blocks CD47. While CV1-Fc-coengineered A97L-T cells mediated significantly better control of tumor outgrowth and survival in Winn assays, in subcutaneous xenograft models the T cells, coated by CV1-Fc, were depleted. Importantly, there was no phagocytosis of CV1 monomer-coengineered T cells by human macrophages. Moreover, avelumab and cetuximab enhanced macrophage-mediated phagocytosis of tumor cells in vitro in the presence of CV1 and improved tumor control upon coadministration with A97L-T cells. Taken together, our study indicates important clinical promise for harnessing macrophages by combining CV1-coengineered TCR-T cells with targeted antibodies to direct phagocytosis against tumor cells.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- Antigens, Differentiation/immunology
- Antigens, Neoplasm/immunology
- CD47 Antigen/immunology
- Cell Line, Tumor
- HLA-A2 Antigen/immunology
- HLA-A2 Antigen/genetics
- Immunotherapy, Adoptive
- Macrophages/immunology
- Macrophages/metabolism
- Phagocytosis
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Immunologic/immunology
- Receptors, Immunologic/metabolism
- Receptors, Immunologic/genetics
- T-Lymphocytes/immunology
- Xenograft Model Antitumor Assays
- Male
- Female
Collapse
Affiliation(s)
- Evangelos Stefanidis
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - Aikaterini Semilietof
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Julien Pujol
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Bili Seijo
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Kirsten Scholten
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Vincent Zoete
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
| | - Olivier Michielin
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
- Swiss Institute of Bioinformatics, Lausanne, Switzerland
- Precision Oncology, University Hospital of Geneva (HUG), Geneva, Switzerland
| | - Raphael Sandaltzopoulos
- Department of Molecular Biology and Genetics, Democritus University of Thrace, Alexandroupolis, Greece
| | - George Coukos
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| | - Melita Irving
- Ludwig Institute for Cancer Research, Department of Oncology, University of Lausanne (UNIL) and University Hospital of Lausanne (CHUV), Lausanne, Switzerland
| |
Collapse
|
15
|
Strzelec A, Helbig G. Are we ready for personalized CAR-T therapy? Eur J Haematol 2024; 112:174-183. [PMID: 37431655 DOI: 10.1111/ejh.14039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 06/29/2023] [Accepted: 06/30/2023] [Indexed: 07/12/2023]
Abstract
The future of chimeric antigen receptor T (CAR-T) therapy remains unclear. New studies are constantly being published confirming the efficacy and favorable safety profile of its innovative enhancements. Currently approved CAR-T drugs are manufactured exclusively for a specific patient from the recipient's own cells. This does not close the door to further modifications with subsequent personalization and better adaptation to the individual needs. Bringing such a drug to market would involve raising the already high costs, so it is necessary to lower the existing ones. On the other hand, so-called universal CAR-T are also getting closer to the patient's bed, but its implementation may struggle with multiple challenges, including development of graft-versus-host disease (GvHD) and alloimmunity. However, that off-the-shelf therapy could prove useful as a quick solution for patients in very poor condition or excluded from current therapy due to manufacturing limitations. The introduction of currently tested solutions may undoubtedly change the current paradigm of treatment.
Collapse
Affiliation(s)
- Anna Strzelec
- Department of Hematology and Bone Marrow Transplantation, Faculty of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| | - Grzegorz Helbig
- Department of Hematology and Bone Marrow Transplantation, Faculty of Medicine in Katowice, Medical University of Silesia, Katowice, Poland
| |
Collapse
|
16
|
Olejarz W, Basak G. Emerging Therapeutic Targets and Drug Resistance Mechanisms in Immunotherapy of Hematological Malignancies. Cancers (Basel) 2023; 15:5765. [PMID: 38136311 PMCID: PMC10741639 DOI: 10.3390/cancers15245765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 11/22/2023] [Accepted: 12/06/2023] [Indexed: 12/24/2023] Open
Abstract
CAR-T cell therapy has revolutionized the treatment of hematological malignancies with high remission rates in the case of ALL and NHL. This therapy has some limitations such as long manufacturing periods, persistent restricted cell sources and high costs. Moreover, combination regimens increase the risk of immune-related adverse events, so the identification new therapeutic targets is important to minimize the risk of toxicities and to guide more effective approaches. Cancer cells employ several mechanisms to evade immunosurveillance, which causes resistance to immunotherapy; therefore, a very important therapeutic approach is to focus on the development of rational combinations of targeted therapies with non-overlapping toxicities. Recent progress in the development of new inhibitory clusters of differentiation (CDs), signaling pathway molecules, checkpoint inhibitors, and immunosuppressive cell subsets and factors in the tumor microenvironment (TME) has significantly improved anticancer responses. Novel strategies regarding combination immunotherapies with CAR-T cells are the most promising approach to cure cancer.
Collapse
Affiliation(s)
- Wioletta Olejarz
- Department of Biochemistry and Pharmacogenomics, Faculty of Pharmacy, Medical University of Warsaw, 02-091 Warsaw, Poland
- Centre for Preclinical Research, Medical University of Warsaw, 02-091 Warsaw, Poland
| | - Grzegorz Basak
- Department of Hematology, Transplantation and Internal Medicine, Medical University of Warsaw, 02-091 Warsaw, Poland;
| |
Collapse
|
17
|
CHEN QIUQIANG, GUO XUEJUN, MA WENXUE. Opportunities and challenges of CD47-targeted therapy in cancer immunotherapy. Oncol Res 2023; 32:49-60. [PMID: 38188674 PMCID: PMC10767231 DOI: 10.32604/or.2023.042383] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2023] [Accepted: 08/09/2023] [Indexed: 01/09/2024] Open
Abstract
Cancer immunotherapy has emerged as a promising strategy for the treatment of cancer, with the tumor microenvironment (TME) playing a pivotal role in modulating the immune response. CD47, a cell surface protein, has been identified as a crucial regulator of the TME and a potential therapeutic target for cancer therapy. However, the precise functions and implications of CD47 in the TME during immunotherapy for cancer patients remain incompletely understood. This comprehensive review aims to provide an overview of CD47's multifaced role in TME regulation and immune evasion, elucidating its impact on various types of immunotherapy outcomes, including checkpoint inhibitors and CAR T-cell therapy. Notably, CD47-targeted therapies offer a promising avenue for improving cancer treatment outcomes, especially when combined with other immunotherapeutic approaches. The review also discusses current and potential CD47-targeted therapies being explored for cancer treatment and delves into the associated challenges and opportunities inherent in targeting CD47. Despite the demonstrated effectiveness of CD47-targeted therapies, there are potential problems, including unintended effects on healthy cells, hematological toxicities, and the development if resistance. Consequently, further research efforts are warranted to fully understand the underlying mechanisms of resistance and to optimize CD47-targeted therapies through innovative combination approaches, ultimately improving cancer treatment outcomes. Overall, this comprehensive review highlights the significance of CD47 as a promising target for cancer immunotherapy and provides valuable insight into the challenges and opportunities in developing effective CD47-targeted therapies for cancer treatment.
Collapse
Affiliation(s)
- QIUQIANG CHEN
- Key Laboratory for Translational Medicine, The First Affiliated Hospital, Huzhou University School of Medicine, Huzhou, 313000, China
| | - XUEJUN GUO
- Department of Hematology, Puyang Youtian General Hospital, Puyang, 457001, China
| | - WENXUE MA
- Department of Medicine, Moores Cancer Center, Sanford Stem Cell Institute, University of California San Diego, La Jolla, San Diego, 92093, USA
| |
Collapse
|