1
|
Xiao W, Xu L, Wang J, Yu K, Xu B, Que Y, Zhao J, Pan Q, Gao C, Zhou P, Zhang X. FGFR4-specific CAR-T cells with inducible caspase-9 suicide gene as an approach to treat rhabdomyosarcoma. Cancer Gene Ther 2024; 31:1571-1584. [PMID: 39183354 PMCID: PMC11489081 DOI: 10.1038/s41417-024-00823-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Revised: 07/24/2024] [Accepted: 08/12/2024] [Indexed: 08/27/2024]
Abstract
Metastatic rhabdomyosarcoma is associated with poor survival and unsatisfactory treatment outcomes. Therefore, new immunotherapeutic methods are urgently required. Fibroblast growth factor receptor 4 (FGFR4), a new therapeutic target for rhabdomyosarcoma, plays a crucial role in its onset and development. This study aimed to generate FGFR4 single-chain variable fragment-based chimeric antigen receptor (CAR) T cells without causing evident toxicity and incorporating an inducible caspase-9 (iCasp9) suicide gene system to enhance their safety. FGFR4 antigen expression was evaluated in normal murine tissues, normal human tissues, and specimens from patients with rhabdomyosarcoma. Combined with a 4-1BB co-stimulatory domain, a CD3ζ signaling domain, and an iCasp9 suicide gene, CAR-T cells with an FGFR4-specific single-chain variable fragment were developed. The specific cytotoxic effects, T-cell proliferation, cytokine secretion, apoptosis induction by chemical dimerization (AP20187), and toxicity of FGFR4 CAR-T cells were investigated in vitro and in vivo. FGFR4 CAR-T cells generated a variety of immune-promoting cytokines, including tumor necrosis factor α, interleukin 2, and interferon γ, and displayed effective cytotoxic activity against FGFR4-overexpressing rhabdomyosarcoma cells in vitro. FGFR4 CAR-T cells were relatively effective against FGFR4-overexpressing rhabdomyosarcoma, with tumor regression and poor survival in a subcutaneous xenograft model. The iCasp9 gene was incorporated into FGFR4 CAR-T cells and it was demonstrated that effective and reliable suicide gene activity depends on the administration of AP20187. By making use of the cross-reaction of FGFR4 CAR-T cells with murine FGFR4 in a syngeneic tumor model, this study found that FGFR4 CAR-T cells could regulate the growth of tumors without evident toxicity. Our study demonstrates that FGFR4 is a prospective target for CAR-T cell therapy in rhabdomyosarcoma without serious on-target off-tumor toxicity. FGFR4 CAR-T cells with the iCasp9 suicide gene system as a safety switch to limit toxicity may broaden the clinical applications of cellular therapy.
Collapse
MESH Headings
- Rhabdomyosarcoma/therapy
- Rhabdomyosarcoma/genetics
- Animals
- Humans
- Mice
- Receptor, Fibroblast Growth Factor, Type 4/genetics
- Receptor, Fibroblast Growth Factor, Type 4/metabolism
- Genes, Transgenic, Suicide
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Caspase 9/genetics
- Caspase 9/metabolism
- Immunotherapy, Adoptive/methods
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Apoptosis
- Female
Collapse
Affiliation(s)
- Wei Xiao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- Melanoma and Sarcoma Medical Oncology Unit, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, 628 Zhenyuan Road, Guangming District, Shenzhen, Guangdong, 518107, China
| | - Liping Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
| | - Jinghua Wang
- Department of Hematology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, 106 Zhongshan 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Kuai Yu
- Department of Blood Transfusion, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330209, China
- Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330209, China
| | - Bushu Xu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- Melanoma and Sarcoma Medical Oncology Unit, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Yi Que
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- Melanoma and Sarcoma Medical Oncology Unit, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Jingjing Zhao
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- Melanoma and Sarcoma Medical Oncology Unit, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Qiuzhong Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China
- Melanoma and Sarcoma Medical Oncology Unit, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China
| | - Chengqi Gao
- Department of Blood Transfusion, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330209, China
- Key Laboratory of Jiangxi Province for Transfusion Medicine, The First Affiliated Hospital of Nanchang University, 17 Yongwaizheng Street, Nanchang, Jiangxi, 330209, China
| | - Penghui Zhou
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.
- Department of Experimental Research, Sun Yat-Sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.
| | - Xing Zhang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, Guangdong, 510060, China.
- Melanoma and Sarcoma Medical Oncology Unit, Sun Yat-sen University Cancer Center, 651 Dongfeng East Road, Guangzhou, 510060, China.
| |
Collapse
|
2
|
Liu C, Wang Q, Li L, Gao F, Zhang Y, Zhu Y. The peptide-based bispecific CAR T cells target EGFR and tumor stroma for effective cancer therapy. Int J Pharm 2024; 663:124558. [PMID: 39111352 DOI: 10.1016/j.ijpharm.2024.124558] [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: 05/20/2024] [Revised: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND AND PURPOSE The efficacy of chimeric antigen receptor (CAR)-T cell for solid tumors is limited partially because of the lack of tumor-specific antigens and off-target effects. Low molecular weight peptides allowed CAR T cell to display several antigen receptors to reduce off-target effects. Here, we develop a peptide-based bispecific CAR for EGFR and tumor stroma, which are expressed in a variety of tumor types. EXPERIMENTAL APPROACH AND KEY RESULTS The peptide-based CAR T cells show excellent proliferation, cytotoxicity activity and are only activated by tumor cells overexpressing EGFR instead of normal cells with low EGFR expressing. In mouse xenograft models, the peptide bispecific CAR T cells can be delivered into the inner of tumor masses and thus are effective in inhibiting tumor growth. Meanwhile, they show strong expansion capacity and the property of maintaining long-term function in vivo. During treatment, no off-tumor toxicity is observed on healthy organs expressing lower levels of EGFR. CONCLUSIONS & IMPLICATIONS Our findings demonstrate that peptide-based bispecific CAR T holds great potential in solid tumor therapy due to an excellent targeting ability towards tumors and tumor microenvironment.
Collapse
Affiliation(s)
- Cuijuan Liu
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China; The Fifth Medical Center of Chinese PLA General Hospital, Beijing 100071, China
| | - Qianqian Wang
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Lin Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Fan Gao
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei 230026, China; CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China
| | - Yuanyue Zhang
- Department of Oncology, Suzhou BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou, China
| | - Yimin Zhu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, China.
| |
Collapse
|
3
|
Prikhodko IV, Guria GT. The method for assessing the specificity of developing CAR therapies. BIOPHYSICAL REPORTS 2024; 4:100172. [PMID: 39025235 PMCID: PMC11344002 DOI: 10.1016/j.bpr.2024.100172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/10/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
The effectiveness of antitumor chimeric antigen receptor (CAR) therapy mainly dealt with an elevated sensitivity of CAR cells to target cells. However, CAR therapies are associated with nonspecific side effects: on-target off-tumor toxicity. Sensitivity and specificity of CAR cells are the most important properties of the recognition process of target cells among other cells. Current developments are mainly concentrated on exploring molecular biology methods for designing CAR cells with the highest sensitivity, while the problem of the CAR cell specificity is rarely considered. For the assessment of CAR cell specificity, we suggest that, in addition to an elevated level of CAR-antigen affinity, the ability of CARs for clustering should be taken into account. We assume that the CAR cell cytotoxicity is determined by CAR clustering. The latter is treated within the framework of nucleation theory. The master equation for the probability of CAR cell cytotoxicity is derived. The size of a critical CAR cluster is found to be one of two most essential parameters. The conditions for necessary sensitivity and sufficient specificity are explored. Relevant parametric diagrams are derived. Possible applications of the method for assessing the specificity of developing CAR therapies are discussed.
Collapse
Affiliation(s)
- Ivan V Prikhodko
- Laboratory for Mathematical Modelling of Biological Processes, National Medical Research Center for Hematology, Moscow, Russia
| | - Georgy Th Guria
- Laboratory for Mathematical Modelling of Biological Processes, National Medical Research Center for Hematology, Moscow, Russia; Chair of the Living Systems Physics, Moscow Institute of Physics and Technology, Dolgoprudny, Moscow Region, Russia.
| |
Collapse
|
4
|
Ali A, Alamri A, Hajar A. NK/DC crosstalk-modulating antitumor activity via Sema3E/PlexinD1 axis for enhanced cancer immunotherapy. Immunol Res 2024:10.1007/s12026-024-09536-y. [PMID: 39235526 DOI: 10.1007/s12026-024-09536-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Accepted: 08/29/2024] [Indexed: 09/06/2024]
Abstract
The complex relationship between natural killer (NK) cells and dendritic cells (DCs) within the tumor microenvironment significantly impacts the success of cancer immunotherapy. Recent advancements in cancer treatment have sought to bolster innate and adaptive immune responses through diverse modalities, aiming to tilt the immune equilibrium toward tumor elimination. Optimal antitumor immunity entails a multifaceted interplay involving NK cells, T cells and DCs, orchestrating immune effector functions. Although DC-based vaccines and NK cells' cytotoxic capabilities hold substantial therapeutic potential, their interaction is frequently hindered by immunosuppressive elements such as myeloid-derived suppressor cells (MDSCs) and regulatory T cells. Chemokines and cytokines, such as CXCL12, CCL2, interferons, and interleukins, play crucial roles in modulating NK/DC interactions and enhancing immune responses. This review elucidates the mechanisms underlying NK/DC interaction, emphasizing their pivotal roles in augmenting antitumor immune responses and the impediments posed by tumor-induced immunosuppression. Furthermore, it explores the therapeutic prospects of restoring NK/DC crosstalk, highlighting the significance of molecules like Sema3E/PlexinD1 in this context, offering potential avenues for enhancing the effectiveness of current immunotherapeutic strategies and advancing cancer treatment paradigms. Harnessing the dynamic interplay between NK and DC cells, including the modulation of Sema3E/PlexinD1 signaling, holds promise for developing more potent therapies that harness the immune system's full potential in combating cancer.
Collapse
Affiliation(s)
- Awais Ali
- Department of Biochemistry, Abdul Wali Khan University Mardan (AWKUM), Mardan, 23200, Pakistan.
| | - Abdulaziz Alamri
- Department of Biochemistry, College of Science, King Saud University, 11451, Riyadh, Saudi Arabia
| | - Azraida Hajar
- Department of Biology, Faculty of Sciences Semlalia, Cadi Ayyad University, Marrakech, Morocco
| |
Collapse
|
5
|
Ercilla-Rodríguez P, Sánchez-Díez M, Alegría-Aravena N, Quiroz-Troncoso J, Gavira-O'Neill CE, González-Martos R, Ramírez-Castillejo C. CAR-T lymphocyte-based cell therapies; mechanistic substantiation, applications and biosafety enhancement with suicide genes: new opportunities to melt side effects. Front Immunol 2024; 15:1333150. [PMID: 39091493 PMCID: PMC11291200 DOI: 10.3389/fimmu.2024.1333150] [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: 11/04/2023] [Accepted: 06/14/2024] [Indexed: 08/04/2024] Open
Abstract
Immunotherapy has made significant strides in cancer treatment with strategies like checkpoint blockade antibodies and adoptive T cell transfer. Chimeric antigen receptor T cells (CAR-T) have emerged as a promising approach to combine these strategies and overcome their limitations. This review explores CAR-T cells as a living drug for cancer treatment. CAR-T cells are genetically engineered immune cells designed to target and eliminate tumor cells by recognizing specific antigens. The study involves a comprehensive literature review on CAR-T cell technology, covering structure optimization, generations, manufacturing processes, and gene therapy strategies. It examines CAR-T therapy in haematologic cancers and solid tumors, highlighting challenges and proposing a suicide gene-based mechanism to enhance safety. The results show significant advancements in CAR-T technology, particularly in structure optimization and generation. The manufacturing process has improved for broader clinical application. However, a series of inherent challenges and side effects still need to be addressed. In conclusion, CAR-T cells hold great promise for cancer treatment, but ongoing research is crucial to improve efficacy and safety for oncology patients. The proposed suicide gene-based mechanism offers a potential solution to mitigate side effects including cytokine release syndrome (the most common toxic side effect of CAR-T therapy) and the associated neurotoxicity.
Collapse
MESH Headings
- Humans
- Immunotherapy, Adoptive/adverse effects
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Genes, Transgenic, Suicide
- Neoplasms/therapy
- Neoplasms/immunology
- Neoplasms/genetics
- T-Lymphocytes/immunology
- Animals
- Genetic Therapy/adverse effects
- Genetic Therapy/methods
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
Collapse
Affiliation(s)
| | - Marta Sánchez-Díez
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Nicolás Alegría-Aravena
- Grupo de Biología y Producción de Cérvidos, Instituto de Desarrollo Regional, Universidad de Castilla-La Mancha, Albacete, Spain
- Asociación Española Contra el Cáncer (AECC)-Fundación Científica AECC, Albacete, Spain
| | - Josefa Quiroz-Troncoso
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Clara E. Gavira-O'Neill
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Sección de Oncología, Instituto de Investigación Sanitaria San Carlos, Madrid, Spain
| | - Raquel González-Martos
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
| | - Carmen Ramírez-Castillejo
- ETSIAAB, Universidad Politécnica de Madrid, Madrid, Spain
- Laboratorio Cancer Stem Cell, HST group, Centro de Tecnología Biomédica, Universidad Politécnica de Madrid, Madrid, Spain
- Sección de Oncología, Instituto de Investigación Sanitaria San Carlos, Madrid, Spain
| |
Collapse
|
6
|
Farhat-Younis L, Na M, Zarfin A, Khateeb A, Santana-Magal N, Richter A, Gutwillig A, Rasoulouniriana D, Gleiberman A, Beck L, Giger T, Ashkenazi A, Barzel A, Rider P, Carmi Y. Expression of modified FcγRI enables myeloid cells to elicit robust tumor-specific cytotoxicity. eLife 2024; 12:RP91999. [PMID: 38885133 PMCID: PMC11182644 DOI: 10.7554/elife.91999] [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] [Indexed: 06/20/2024] Open
Abstract
Despite the central role of T cells in tumor immunity, attempts to harness their cytotoxic capacity as a therapy have met limited efficacy, partially as a result of the suppressive microenvironment which limits their migration and activation. In contrast, myeloid cells massively infiltrate tumors and are well adapted to survive these harsh conditions. While they are equipped with cell-killing abilities, they often adopt an immunosuppressive phenotype upon migration to tumors. Therefore, the questions of how to modify their activation programming against cancer, and what signaling cascades should be activated in myeloid cells to elicit their cytotoxicity have remained unclear. Here, we found that activation of IgM-induced signaling in murine myeloid cells results in secretion of lytic granules and massive tumor cell death. These findings open venues for designing novel immunotherapy by equipping monocytes with chimeric receptors that target tumor antigens and consequently, signal through IgM receptor. Nonetheless, we found that myeloid cells do not express the antibody-derived portion used to recognize the tumor antigen due to the induction of an ER stress response. To overcome this limitation, we designed chimeric receptors that are based on the high-affinity FcγRI for IgG. Incubation of macrophages expressing these receptors along with tumor-binding IgG induced massive tumor cell killing and secretion of reactive oxygen species and Granzyme B. Overall, this work highlights the challenges involved in genetically reprogramming the signaling in myeloid cells and provides a framework for endowing myeloid cells with antigen-specific cytotoxicity.
Collapse
Affiliation(s)
- Leen Farhat-Younis
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Manho Na
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Amichai Zarfin
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Aseel Khateeb
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | | | - Alon Richter
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Amit Gutwillig
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | | | - Annette Gleiberman
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Lir Beck
- Department of Human Molecular Genetics and Biochemistry, Tel Aviv UniversityTel AvivIsrael
| | - Tamar Giger
- Department of Molecular Cell Biology, Weizmann InstituteRehovotIsrael
| | - Avraham Ashkenazi
- Department of Cell and Developmental Biology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Adi Barzel
- Department of Biochemistry Molecular Biology, George S. Wise Faculty of Life Sciences, Tel Aviv UniversityTel AvivIsrael
| | - Peleg Rider
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| | - Yaron Carmi
- Department of Pathology, School of Medicine, Tel Aviv UniversityTel AvivIsrael
| |
Collapse
|
7
|
Gong N, Han X, Xue L, Billingsley MM, Huang X, El-Mayta R, Qin J, Sheppard NC, June CH, Mitchell MJ. Small-molecule-mediated control of the anti-tumour activity and off-tumour toxicity of a supramolecular bispecific T cell engager. Nat Biomed Eng 2024; 8:513-528. [PMID: 38378820 DOI: 10.1038/s41551-023-01147-6] [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: 01/22/2023] [Accepted: 10/24/2023] [Indexed: 02/22/2024]
Abstract
The broader clinical use of bispecific T cell engagers for inducing anti-tumour toxicity is hindered by their on-target off-tumour toxicity and the associated neurotoxicity and cytokine-release syndrome. Here we show that the off-tumour toxicity of a supramolecular bispecific T cell engager binding to the T cell co-receptor CD3 and to the human epidermal growth factor receptor 2 on breast tumour cells can be halted by disengaging the T cells from the tumour cells via the infusion of the small-molecule drug amantadine, which disassembles the supramolecular aggregate. In mice bearing human epidermal growth factor receptor 2-expressing tumours and with a human immune system, high intravenous doses of such a 'switchable T cell nanoengager' elicited strong tumour-specific adaptive immune responses that prevented tumour relapse, while the infusion of amantadine restricted off-tumour toxicity, cytokine-release syndrome and neurotoxicity. Supramolecular chemistry may be further leveraged to control the anti-tumour activity and off-tumour toxicity of bispecific antibodies.
Collapse
Affiliation(s)
- Ningqiang Gong
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Xuexiang Han
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Lulu Xue
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | | | - Xisha Huang
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Rakan El-Mayta
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Jingya Qin
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Neil C Sheppard
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Carl H June
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Michael J Mitchell
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Cellular Immunotherapies, University of Pennsylvania, Philadelphia, PA, USA.
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Abramson Cancer Center, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Immunology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Cardiovascular Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for Regenerative Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- Institute for RNA Innovation, University of Pennsylvania, Philadelphia, PA, USA.
- Center for Precision Engineering for Health, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
8
|
Zhou Y, Liu X, Gao W, Luo X, Lv J, Wang Y, Liu D. The role of intestinal flora on tumor immunotherapy: recent progress and treatment implications. Heliyon 2024; 10:e23919. [PMID: 38223735 PMCID: PMC10784319 DOI: 10.1016/j.heliyon.2023.e23919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 12/08/2023] [Accepted: 12/15/2023] [Indexed: 01/16/2024] Open
Abstract
Immunotherapy, specifically immune checkpoint inhibitors, has emerged as a promising approach for treating malignant tumors. The gut, housing approximately 70 % of the body's immune cells, is abundantly populated with gut bacteria that actively interact with the host's immune system. Different bacterial species within the intestinal flora are in a delicate equilibrium and mutually regulate each other. However, when this balance is disrupted, pathogenic microorganisms can dominate, adversely affecting the host's metabolism and immunity, ultimately promoting the development of disease. Emerging researches highlight the potential of interventions such as fecal microflora transplantation (FMT) to improve antitumor immune response and reduce the toxicity of immunotherapy. These remarkable findings suggest the major role of intestinal flora in the development of cancer immunotherapy and led us to the hypothesis that intestinal flora transplantation may be a new breakthrough in modifying immunotherapy side effects.
Collapse
Affiliation(s)
- Yimin Zhou
- School of Basic Medical Sciences, Shandong University, Jinan 250011, China
| | - Xiangdong Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Wei Gao
- School of Basic Medical Sciences, Shandong University, Jinan 250011, China
| | - Xin Luo
- School of Basic Medical Sciences, Shandong University, Jinan 250011, China
| | - Junying Lv
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Yunshan Wang
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| | - Duanrui Liu
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong University, Jinan 250021, China
- Department of Clinical Laboratory, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, China
| |
Collapse
|
9
|
Lee IK, Sharma N, Noguera-Ortega E, Liousia M, Baroja ML, Etersque JM, Pham J, Sarkar S, Carreno BM, Linette GP, Puré E, Albelda SM, Sellmyer MA. A genetically encoded protein tag for control and quantitative imaging of CAR T cell therapy. Mol Ther 2023; 31:3564-3578. [PMID: 37919903 PMCID: PMC10727978 DOI: 10.1016/j.ymthe.2023.10.020] [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: 04/01/2023] [Revised: 09/14/2023] [Accepted: 10/31/2023] [Indexed: 11/04/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cell therapy has been successful for hematological malignancies. Still, a lack of efficacy and potential toxicities have slowed its application for other indications. Furthermore, CAR T cells undergo dynamic expansion and contraction in vivo that cannot be easily predicted or controlled. Therefore, the safety and utility of such therapies could be enhanced by engineered mechanisms that engender reversible control and quantitative monitoring. Here, we use a genetic tag based on the enzyme Escherichia coli dihydrofolate reductase (eDHFR), and derivatives of trimethoprim (TMP) to modulate and monitor CAR expression and T cell activity. We fused eDHFR to the CAR C terminus, allowing regulation with TMP-based proteolysis-targeting chimeric small molecules (PROTACs). Fusion of eDHFR to the CAR does not interfere with cell signaling or its cytotoxic function, and the addition of TMP-based PROTACs results in a reversible and dose-dependent inhibition of CAR activity via the proteosome. We show the regulation of CAR expression in vivo and demonstrate imaging of the cells with TMP radiotracers. In vitro immunogenicity assays using primary human immune cells and overlapping peptide fragments of eDHFR showed no memory immune repertoire for eDHFR. Overall, this translationally-orientied approach allows for temporal monitoring and image-guided control of cell-based therapies.
Collapse
Affiliation(s)
- Iris K Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Bioengineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Nitika Sharma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Estela Noguera-Ortega
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria Liousia
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Miren L Baroja
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jean M Etersque
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jonathan Pham
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Swarbhanu Sarkar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beatriz M Carreno
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gerald P Linette
- Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Ellen Puré
- Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Steven M Albelda
- Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Cellular Immunotherapies, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Biochemistry and Biophysics, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Etersque JM, Lee IK, Sharma N, Xu K, Ruff A, Northrup JD, Sarkar S, Nguyen T, Lauman R, Burslem GM, Sellmyer MA. Regulation of eDHFR-tagged proteins with trimethoprim PROTACs. Nat Commun 2023; 14:7071. [PMID: 37923771 PMCID: PMC10624689 DOI: 10.1038/s41467-023-42820-3] [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: 02/07/2023] [Accepted: 10/22/2023] [Indexed: 11/06/2023] Open
Abstract
Temporal control of protein levels in cells and living animals can be used to improve our understanding of protein function. In addition, control of engineered proteins could be used in therapeutic applications. PRoteolysis-TArgeting Chimeras (PROTACs) have emerged as a small-molecule-driven strategy to achieve rapid, post-translational regulation of protein abundance via recruitment of an E3 ligase to the target protein of interest. Here, we develop several PROTAC molecules by covalently linking the antibiotic trimethoprim (TMP) to pomalidomide, a ligand for the E3 ligase, Cereblon. These molecules induce degradation of proteins of interest (POIs) genetically fused to a small protein domain, E. coli dihydrofolate reductase (eDHFR), the molecular target of TMP. We show that various eDHFR-tagged proteins can be robustly degraded to 95% of maximum expression with PROTAC molecule 7c. Moreover, TMP-based PROTACs minimally affect the expression of immunomodulatory imide drug (IMiD)-sensitive neosubstrates using proteomic and biochemical assays. Finally, we show multiplexed regulation with another known degron-PROTAC pair, as well as reversible protein regulation in a rodent model of metastatic cancer, demonstrating the formidable strength of this system. Altogether, TMP PROTACs are a robust approach for selective and reversible degradation of eDHFR-tagged proteins in vitro and in vivo.
Collapse
Affiliation(s)
- Jean M Etersque
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Iris K Lee
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Nitika Sharma
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Kexiang Xu
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Andrew Ruff
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Justin D Northrup
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Swarbhanu Sarkar
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Tommy Nguyen
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Richard Lauman
- The Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - George M Burslem
- The Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
- The Department of Cancer Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Mark A Sellmyer
- Department of Radiology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
- The Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.
| |
Collapse
|
11
|
Golubovskaya V. Editorial on "Cell Therapy, Bispecific Antibodies and Other Immunotherapies against Cancer". Cancers (Basel) 2023; 15:5053. [PMID: 37894420 PMCID: PMC10605091 DOI: 10.3390/cancers15205053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 10/17/2023] [Indexed: 10/29/2023] Open
Abstract
This Special Issue in Cancers, "Cell Therapy, Bispecific Antibodies and other Immunotherapies Against Cancer", includes interesting reports and reviews on cell therapies and bispecific antibodies [...].
Collapse
|
12
|
Sotoudeh M, Mansouri V, Shakeri R, Sharififard B, Sajadi N, Haghpanah V, Naderi M. Decoding the expression pattern of MUC3A in gastric adenocarcinoma: unveiling the key to successful immunotherapy. Expert Opin Ther Targets 2023; 27:1299-1305. [PMID: 38069509 DOI: 10.1080/14728222.2023.2293764] [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: 03/30/2023] [Accepted: 12/08/2023] [Indexed: 12/31/2023]
Abstract
AIMS Despite the promise of immunotherapy for gastric adenocarcinoma, resistance is common, necessitating the validation of new targets. Based on our previous bioinformatics analysis, the MUC3A antigen emerged as a promising candidate for immunotherapy against gastric adenocarcinoma. However, a comprehensive understanding of its expression at protein level remains elusive, despite its crucial role in determining clinical response. We also sought to establish a connection between the expression pattern and relevant clinical variables of the disease, whenever feasible. METHODS Immunohistochemistry was used to determine the percentage of MUC3A-positive tumor cells in primary (PT) and metastatic tumor (MT) sites of 190 gastric adenocarcinoma patients. We also evaluated the association between MUC3A expression and variables such as Lauren classification, history of neoadjuvant chemotherapy and/or radiotherapy, and overall patient survival. RESULTS Median MUC3A expression was 50% in PT and 70% in MT sites, exhibiting a positive correlation. MT intestinal type showed significantly higher MUC3A expression compared to other types. Neoadjuvant therapy history did not affect MUC3A expression. Higher MUC3A expression correlated with improved survival. CONCLUSIONS Based on our previous bioinformatics data and the consistently high expression of MUC3A on gastric tumor cells, we propose advancing experimental aspects of anti-MUC3A immunotherapy for gastric adenocarcinoma.
Collapse
Affiliation(s)
- Masoud Sotoudeh
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Vahid Mansouri
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Shakeri
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Bahareh Sharififard
- Digestive Oncology Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Nasim Sajadi
- Ali-Asghar Children Hospital, Iran University of Medical Sciences, Tehran, Iran
| | - Vahid Haghpanah
- Endocrinology and Metabolism Research Center, Endocrinology and Metabolism Clinical Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Mahmood Naderi
- Digestive Diseases Research Center, Digestive Diseases Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
13
|
Celichowski P, Turi M, Charvátová S, Radhakrishnan D, Feizi N, Chyra Z, Šimíček M, Jelínek T, Bago JR, Hájek R, Hrdinka M. Tuning CARs: recent advances in modulating chimeric antigen receptor (CAR) T cell activity for improved safety, efficacy, and flexibility. J Transl Med 2023; 21:197. [PMID: 36922828 PMCID: PMC10015723 DOI: 10.1186/s12967-023-04041-6] [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: 02/08/2023] [Accepted: 03/07/2023] [Indexed: 03/17/2023] Open
Abstract
Cancer immunotherapies utilizing genetically engineered T cells have emerged as powerful personalized therapeutic agents showing dramatic preclinical and clinical results, particularly in hematological malignancies. Ectopically expressed chimeric antigen receptors (CARs) reprogram immune cells to target and eliminate cancer. However, CAR T cell therapy's success depends on the balance between effective anti-tumor activity and minimizing harmful side effects. To improve CAR T cell therapy outcomes and mitigate associated toxicities, scientists from different fields are cooperating in developing next-generation products using the latest molecular cell biology and synthetic biology tools and technologies. The immunotherapy field is rapidly evolving, with new approaches and strategies being reported at a fast pace. This comprehensive literature review aims to provide an up-to-date overview of the latest developments in controlling CAR T cell activity for improved safety, efficacy, and flexibility.
Collapse
Affiliation(s)
- Piotr Celichowski
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Marcello Turi
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Sandra Charvátová
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Dhwani Radhakrishnan
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
- Faculty of Science, University of Ostrava, Ostrava, Czech Republic
| | - Neda Feizi
- Department of Internal Clinical Sciences, Anesthesiology and Cardiovascular Sciences, Sapienza University of Rome, Rome, Italy
| | - Zuzana Chyra
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Michal Šimíček
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Tomáš Jelínek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Juli Rodriguez Bago
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Roman Hájek
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic
| | - Matouš Hrdinka
- Department of Haematooncology, Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic.
- Department of Haematooncology, University Hospital Ostrava, Ostrava, Czech Republic.
| |
Collapse
|
14
|
Yan T, Zhu L, Chen J. Current advances and challenges in CAR T-Cell therapy for solid tumors: tumor-associated antigens and the tumor microenvironment. Exp Hematol Oncol 2023; 12:14. [PMID: 36707873 PMCID: PMC9883880 DOI: 10.1186/s40164-023-00373-7] [Citation(s) in RCA: 61] [Impact Index Per Article: 61.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Accepted: 01/10/2023] [Indexed: 01/28/2023] Open
Abstract
The past decade has witnessed ongoing progress in immune therapy to ameliorate human health. As an emerging technique, chimeric antigen receptor (CAR) T-cell therapy has the advantages of specific killing of cancer cells, a high remission rate of cancer-induced symptoms, rapid tumor eradication, and long-lasting tumor immunity, opening a new window for tumor treatment. However, challenges remain in CAR T-cell therapy for solid tumors due to target diversity, tumor heterogeneity, and the complex microenvironment. In this review, we have outlined the development of the CAR T-cell technique, summarized the current advances in tumor-associated antigens (TAAs), and highlighted the importance of tumor-specific antigens (TSAs) or neoantigens for solid tumors. We also addressed the challenge of the TAA binding domain in CARs to overcome off-tumor toxicity. Moreover, we illustrated the dominant tumor microenvironment (TME)-induced challenges and new strategies based on TME-associated antigens (TMAs) for solid tumor CAR T-cell therapy.
Collapse
Affiliation(s)
- Ting Yan
- grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| | - Lingfeng Zhu
- grid.443397.e0000 0004 0368 7493Department of Urology, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| | - Jin Chen
- grid.443397.e0000 0004 0368 7493Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China ,grid.443397.e0000 0004 0368 7493Department of Clinical Laboratory, The Second Affiliated Hospital of Hainan Medical University, Haikou, 570311 Hainan China
| |
Collapse
|
15
|
Kinch MS, Kraft Z, Schwartz T. Monoclonal antibodies: Trends in therapeutic success and commercial focus. Drug Discov Today 2023; 28:103415. [PMID: 36280042 DOI: 10.1016/j.drudis.2022.103415] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 07/07/2022] [Accepted: 10/18/2022] [Indexed: 02/02/2023]
Abstract
Monoclonal antibody products have risen from obscurity in the 1990s to a position that increasingly dominates both revenue generation and patient impact. This success has occurred largely over the past two decades, and we have identified factors associated with the remarkable advances that have contributed to the discovery, development and approval of monoclonal antibodies. Although consolidation has increased in recent years, the net number of monoclonal antibody developers continues to grow, bucking the general trend in the biopharmaceutical industry.
Collapse
Affiliation(s)
- Michael S Kinch
- Center for Research Innovation in Biotechnology, Long Island University, Brookville, NY 11548, USA.
| | - Zachary Kraft
- Center for Research Innovation in Biotechnology, Long Island University, Brookville, NY 11548, USA
| | - Tyler Schwartz
- Center for Research Innovation in Biotechnology, Long Island University, Brookville, NY 11548, USA
| |
Collapse
|
16
|
Dölz M, Hasiuk M, Gagnon JD, Kornete M, Marone R, Bantug G, Kageyama R, Hess C, Ansel KM, Seyres D, Roux J, Jeker LT. Forced expression of the non-coding RNA miR-17∼92 restores activation and function in CD28-deficient CD4 + T cells. iScience 2022; 25:105372. [PMID: 36388982 PMCID: PMC9646923 DOI: 10.1016/j.isci.2022.105372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 08/12/2022] [Accepted: 10/12/2022] [Indexed: 11/06/2022] Open
Abstract
CD28 provides the prototypical costimulatory signal required for productive T-cell activation. Known molecular consequences of CD28 costimulation are mostly based on studies of protein signaling molecules. The microRNA cluster miR-17∼92 is induced by T cell receptor stimulation and further enhanced by combined CD28 costimulation. We demonstrate that transgenic miR-17∼92 cell-intrinsically largely overcomes defects caused by CD28 deficiency. Combining genetics, transcriptomics, bioinformatics, and biochemical miRNA:mRNA interaction maps we empirically validate miR-17∼92 target genes that include several negative regulators of T cell activation. CD28-deficient T cells exhibit derepressed miR-17∼92 target genes during activation. CRISPR/Cas9-mediated ablation of the miR-17∼92 targets Pten and Nrbp1 in naive CD28-/- CD4+ T cells differentially increases proliferation and expression of the activation markers CD25 and CD44, respectively. Thus, we propose that miR-17∼92 constitutes a central mediator for T cell activation, integrating signals by the TCR and CD28 costimulation by dampening multiple brakes that prevent T cell activation.
Collapse
Affiliation(s)
- Marianne Dölz
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Marko Hasiuk
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - John D. Gagnon
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Mara Kornete
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Romina Marone
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Glenn Bantug
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
| | - Robin Kageyama
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Christoph Hess
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Department of Medicine – CITIID, University of Cambridge, Puddicombe Way, Cambridge CB2 0AW, UK
| | - K. Mark Ansel
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
- Sandler Asthma Basic Research Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Denis Seyres
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| | - Julien Roux
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Swiss Institute of Bioinformatics, Basel, Switzerland
| | - Lukas T. Jeker
- Department of Biomedicine, Basel University Hospital and University of Basel, Hebelstrasse 20, CH-4031 Basel, Switzerland
- Transplantation Immunology & Nephrology, Basel University Hospital, Petersgraben 4, CH-4031 Basel, Switzerland
| |
Collapse
|
17
|
Zhu L, Liu JL, Yang JT, Wu DW, Xu N, Huo KF, Wang HB. PD-1 engineered cytomembrane cloaked molybdenum nitride for synergistic photothermal and enhanced immunotherapy of breast cancer. J Mater Chem B 2022; 10:9249-9257. [PMID: 36321642 DOI: 10.1039/d2tb01710e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Incomplete tumor ablation and subsequent tumor metastasis usually occur during photothermal anti-tumor processes. The combination of photothermal and immunotherapy has proven to be a promising method to conquer technical challenges. Inhibiting the programmed death ligand-1 (PD-L1)/programmed cell death protein 1 (PD-1) immune pathway represents one of the most successful immunotherapy strategies. Whereas, the PD-L1 expression level significantly differs, leading to a relatively low response rate to the immune checkpoint blockade (ICB) approaches. Therefore, improving the expression level of PD-L1 becomes one potential method to enhance the response rate. Herein, NIH 3T3 cells were educated to steadily express PD-1 protein. Furthermore, the synthesized molybdenum nitride was then coated with PD-1 protein-modified cytomembrane, which endows it with immune checkpoint blocking capability. Moreover, under the irradiation of near-infrared light, the local mild heat released from the molybdenum nitride causes the apoptosis of tumor cells. More importantly, the elevated temperature simultaneously helps elevate the expression level of PD-L1, further enhancing the response rate of ICB. Finally, the PD-1 cytomembrane coatings interact with the upregulated PD-L1, leading to the activation of the immune system. In summary, we confirmed that the PD-1 protein-coated molybdenum nitride could synergistically ablate tumors and avoid metastasis.
Collapse
Affiliation(s)
- Lian Zhu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Jia-Lin Liu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| | - Jiang-Tao Yang
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430070, China.
| | - Ding-Wei Wu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430070, China.
| | - Na Xu
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430070, China.
| | - Kai-Fu Huo
- Wuhan National Lab for Optoelectronics, Huazhong University of Science and Technology, Wuhan 430074, China.
| | - Hai-Bo Wang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China.
| |
Collapse
|
18
|
Wang X, Yang X, Yuan X, Wang W, Wang Y. Chimeric antigen receptor-engineered NK cells: new weapons of cancer immunotherapy with great potential. Exp Hematol Oncol 2022; 11:85. [PMID: 36324149 PMCID: PMC9628181 DOI: 10.1186/s40164-022-00341-7] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Accepted: 09/29/2022] [Indexed: 11/18/2022] Open
Abstract
Chimeric antigen receptor (CAR)-engineered T (CAR-T) cells have obtained prominent achievement in the clinical immunotherapy of hematological malignant tumors, leading to a rapid development of cellular immunotherapy in cancer treatment. Scientists are also aware of the prospective advantages of CAR engineering in cellular immunotherapy. Due to various limitations such as the serious side effects of CAR-T therapy, researchers began to investigate other immune cells for CAR modification. Natural killer (NK) cells are critical innate immune cells with the characteristic of non-specifically recognizing target cells and with the potential to become "off-the-shelf" products. In recent years, many preclinical studies on CAR-engineered NK (CAR-NK) cells have shown their remarkable efficacy in cancer therapy and their superiority over autologous CAR-T cells. In this review, we summarize the generation, mechanisms of anti-tumor activity and unique advantages of CAR-NK cells, and then analyze some challenges and recent clinical trials about CAR-NK cells therapy. We believe that CAR-NK therapy is a promising prospect for cancer immunotherapy in the future.
Collapse
Affiliation(s)
- Xiao Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xuejiao Yang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| | - Xiang Yuan
- grid.13291.380000 0001 0807 1581Department of Thoracic Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, 610041 China
| | - Wenbo Wang
- grid.24516.340000000123704535Department of Oncology, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Yueying Wang
- grid.16821.3c0000 0004 0368 8293Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025 China
| |
Collapse
|
19
|
Keshavarz A, Salehi A, Khosravi S, Shariati Y, Nasrabadi N, Kahrizi MS, Maghsoodi S, Mardi A, Azizi R, Jamali S, Fotovat F. Recent findings on chimeric antigen receptor (CAR)-engineered immune cell therapy in solid tumors and hematological malignancies. Stem Cell Res Ther 2022; 13:482. [PMID: 36153626 PMCID: PMC9509604 DOI: 10.1186/s13287-022-03163-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 08/12/2022] [Indexed: 11/10/2022] Open
Abstract
Advancements in adoptive cell therapy over the last four decades have revealed various new therapeutic strategies, such as chimeric antigen receptors (CARs), which are dedicated immune cells that are engineered and administered to eliminate cancer cells. In this context, CAR T-cells have shown significant promise in the treatment of hematological malignancies. However, many obstacles limit the efficacy of CAR T-cell therapy in both solid tumors and hematological malignancies. Consequently, CAR-NK and CAR-M cell therapies have recently emerged as novel therapeutic options for addressing the challenges associated with CAR T-cell therapies. Currently, many CAR immune cell trials are underway in various human malignancies around the world to improve antitumor activity and reduce the toxicity of CAR immune cell therapy. This review will describe the comprehensive literature of recent findings on CAR immune cell therapy in a wide range of human malignancies, as well as the challenges that have emerged in recent years.
Collapse
Affiliation(s)
- Ali Keshavarz
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ali Salehi
- Department of Oral and Maxillofacial Radiology, School of Dentistry, Islamic Azad University,, Isfahan (Khorasgan) Branch, Isfahan, Iran
| | - Setareh Khosravi
- Department of Orthodontics, School of Dentistry, Alborz University of Medical Sciences, Karaj, Iran
| | - Yasaman Shariati
- Department of General Surgery, School of Medicine, Arak University of Medical Sciences, Arak, Iran
| | - Navid Nasrabadi
- Department of Endodontics, School of Dentistry, Birjand University of Medical Sciences, Birjand, Iran
| | | | - Sairan Maghsoodi
- Department of Paramedical, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Amirhossein Mardi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ramyar Azizi
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Samira Jamali
- Department of Endodontics, College of Stomatology, Stomatological Hospital, Xi’an Jiaotong University, Shaanxi, People’s Republic of China
| | - Farnoush Fotovat
- Department of Prosthodontics, School of Dentistry, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
20
|
Yekehfallah V, Pahlavanneshan S, Sayadmanesh A, Momtahan Z, Ma B, Basiri M. Generation and Functional Characterization of PLAP CAR-T Cells against Cervical Cancer Cells. Biomolecules 2022; 12:biom12091296. [PMID: 36139135 PMCID: PMC9496028 DOI: 10.3390/biom12091296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2022] [Revised: 09/05/2022] [Accepted: 09/11/2022] [Indexed: 11/16/2022] Open
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is one of the cancer treatment modalities that has recently shown promising results in treating hematopoietic malignancies. However, one of the obstacles that need to be addressed in solid tumors is the on-target and off-tumor cytotoxicity due to the lack of specific tumor antigens with low expression in healthy cells. Placental alkaline phosphatase (PLAP) is a shared placenta- and tumor-associated antigen (TAA) that is expressed in ovarian, cervical, colorectal, and prostate cancers and is negligible in normal cells. In this study, we constructed second-generation CAR T cells with a fully human scFv against PLAP antigen andthen evaluated the characteristics of PLAP CAR T cells in terms of tonic signaling and differentiation in comparison with ΔPLAP CAR T cells and CD19 CAR T cells. In addition, by co-culturing PLAP CAR T cells with HeLa and CaSki cells, we analyzed the tumor-killing functions and the secretion of anti-tumor molecules. Results showed that PLAP CAR T cells not only proliferated during co-culture with cancer cells but also eliminated them in vitro. We also observed increased secretion of IL-2, granzyme A, and IFN-γ by PLAP CAR T cells upon exposure to the target cells. In conclusion, PLAP CAR T cells are potential candidates for further investigation in cervical cancer and, potentially, other solid tumors.
Collapse
Affiliation(s)
- Vahid Yekehfallah
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665666311, Iran
| | - Saghar Pahlavanneshan
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, Tehran 1968917313, Iran
| | - Ali Sayadmanesh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665666311, Iran
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz 5166653431, Iran
| | - Zahra Momtahan
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Tehran Medical Sciences, Islamic Azad University, Tehran 1916893813, Iran
| | - Bin Ma
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, China
- Clinical Stem Cell Research Center, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
- Correspondence: (B.M.); (M.B.); Tel.: +86-21-62933631 (B.M.); +98-21-40223417 (M.B.)
| | - Mohsen Basiri
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 1665666311, Iran
- Correspondence: (B.M.); (M.B.); Tel.: +86-21-62933631 (B.M.); +98-21-40223417 (M.B.)
| |
Collapse
|
21
|
Yun CO, Hong J, Yoon AR. Current clinical landscape of oncolytic viruses as novel cancer immunotherapeutic and recent preclinical advancements. Front Immunol 2022; 13:953410. [PMID: 36091031 PMCID: PMC9458317 DOI: 10.3389/fimmu.2022.953410] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/03/2022] [Indexed: 12/12/2022] Open
Abstract
Oncolytic viruses (OVs) have been gaining attention in the pharmaceutical industry as a novel immunotherapeutic and therapeutic adjuvant due to their ability to induce and boost antitumor immunity through multiple mechanisms. First, intrinsic mechanisms of OVs that enable exploitation of the host immune system (e.g., evading immune detection) can nullify the immune escape mechanism of tumors. Second, many types of OVs have been shown to cause direct lysis of tumor cells, resulting in an induction of tumor-specific T cell response mediated by release of tumor-associated antigens and danger signal molecules. Third, armed OV-expressing immune stimulatory therapeutic genes could be highly expressed in tumor tissues to further improve antitumor immunity. Last, these OVs can inflame cold tumors and their microenvironment to be more immunologically favorable for other immunotherapeutics. Due to these unique characteristics, OVs have been tested as an adjuvant of choice in a variety of therapeutics. In light of these promising attributes of OVs in the immune-oncology field, the present review will examine OVs in clinical development and discuss various strategies that are being explored in preclinical stages for the next generation of OVs that are optimized for immunotherapy applications.
Collapse
Affiliation(s)
- Chae-Ok Yun
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
- GeneMedicine CO., Ltd., Seoul, South Korea
| | | | - A-Rum Yoon
- Department of Bioengineering, College of Engineering, Hanyang University, Seoul, South Korea
- Institute of Nano Science and Technology (INST), Hanyang University, Seoul, South Korea
- Hanyang Institute of Bioscience and Biotechnology (HY-IBB), Hanyang University, Seoul, South Korea
| |
Collapse
|
22
|
Greenshpan Y, Sharabi O, Yegodayev KM, Novoplansky O, Elkabets M, Gazit R, Porgador A. The Contribution of the Minimal Promoter Element to the Activity of Synthetic Promoters Mediating CAR Expression in the Tumor Microenvironment. Int J Mol Sci 2022; 23:7431. [PMID: 35806439 PMCID: PMC9266962 DOI: 10.3390/ijms23137431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Revised: 07/01/2022] [Accepted: 07/01/2022] [Indexed: 11/16/2022] Open
Abstract
Harnessing immune effector cells to benefit cancer patients is becoming more and more prevalent in recent years. However, the increasing number of different therapeutic approaches, such as chimeric antigen receptors and armored chimeric antigen receptors, requires constant adjustments of the transgene expression levels. We have previously demonstrated it is possible to achieve spatial and temporal control of transgene expression as well as tailoring the inducing agents using the Chimeric Antigen Receptor Tumor Induced Vector (CARTIV) platform. Here we describe the next level of customization in our promoter platform. We have tested the functionality of three different minimal promoters, representing three different promoters' strengths, leading to varying levels of CAR expression and primary T cell function. This strategy shows yet another level of CARTIV gene regulation that can be easily integrated into existing CAR T systems.
Collapse
Affiliation(s)
- Yariv Greenshpan
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Omri Sharabi
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Ksenia M. Yegodayev
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
| | - Ofra Novoplansky
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
| | - Moshe Elkabets
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
| | - Roi Gazit
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| | - Angel Porgador
- The Shraga Segal Department of Microbiology, Faculty of Health Sciences, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel; (Y.G.); (O.S.); (K.M.Y.); (O.N.); (M.E.)
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva 84105, Israel
| |
Collapse
|
23
|
Liu C, Li L, Gao F, Zhou J, Qin Y, Yuan X, Yang G, Zhu Y. Reforming the Chimeric Antigen Receptor by Peptide Towards Optimized CAR T Cells With Enhanced Anti-Cancer Potency and Safety. Front Bioeng Biotechnol 2022; 10:928169. [PMID: 35782491 PMCID: PMC9247402 DOI: 10.3389/fbioe.2022.928169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 05/25/2022] [Indexed: 11/29/2022] Open
Abstract
The emerging chimeric antigen receptor (CAR) T cell revolutionized the clinic treatment of hematological cancers, but meet its Waterloo in solid tumor therapy. Although there exist many reasons for this limitation, one of the largest challenges is the scarcity of recognition for tumor cells, resulting in the undesirable side effects and the subsequent ineffectiveness. To overcome it, a lung-cancer-cell-targeting peptide termed A1 was used in this work to reform the scFv domain of CAR by genetic manipulation. As a result, this modified A1CAR T exhibited the optimized cancer-cell targeting and cytotoxicity in vitro and in vivo. More importantly, by tuning the sensitivity of CAR to antigen, peptide-based A1CAR T cells could distinguish tumors from normal tissue, thereby eliminating the off-tumor toxicity in healthy organs. Collectively, we herein constructed a genetic peptide-engineered CAR T cells by inserting A1 peptide into the scFv domain. Profitted from the optimized recognition pattern and sensitivity, A1CAR T cells showed the ascendancy in solid tumor treatment. Our findings demonstrate that peptide-based CAR T holds great potential in solid tumor therapy due to an excellent targeting ability towards tumor cells.
Collapse
Affiliation(s)
- Cuijuan Liu
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Lin Li
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Fan Gao
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Jundong Zhou
- Nanjing Medical University, Affiliated Suzhou Hospital, Department Radio Oncology, Suzhou, China
| | - Yingzhou Qin
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Xin Yuan
- School of Nano Technology and Nano Bionics, University of Science and Technology of China, Hefei, China
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
| | - Guang Yang
- Department of Oncology, Suzhou BenQ Medical Center, The Affiliated BenQ Hospital of Nanjing Medical University, Suzhou, China
- *Correspondence: Guang Yang, ; Yimin Zhu,
| | - Yimin Zhu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou, China
- *Correspondence: Guang Yang, ; Yimin Zhu,
| |
Collapse
|
24
|
Labanieh L, Majzner RG, Klysz D, Sotillo E, Fisher CJ, Vilches-Moure JG, Pacheco KZB, Malipatlolla M, Xu P, Hui JH, Murty T, Theruvath J, Mehta N, Yamada-Hunter SA, Weber EW, Heitzeneder S, Parker KR, Satpathy AT, Chang HY, Lin MZ, Cochran JR, Mackall CL. Enhanced safety and efficacy of protease-regulated CAR-T cell receptors. Cell 2022; 185:1745-1763.e22. [PMID: 35483375 PMCID: PMC9467936 DOI: 10.1016/j.cell.2022.03.041] [Citation(s) in RCA: 111] [Impact Index Per Article: 55.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 01/04/2022] [Accepted: 03/29/2022] [Indexed: 11/22/2022]
Abstract
Regulatable CAR platforms could circumvent toxicities associated with CAR-T therapy, but existing systems have shortcomings including leakiness and attenuated activity. Here, we present SNIP CARs, a protease-based platform for regulating CAR activity using an FDA-approved small molecule. Design iterations yielded CAR-T cells that manifest full functional capacity with drug and no leaky activity in the absence of drug. In numerous models, SNIP CAR-T cells were more potent than constitutive CAR-T cells and showed diminished T cell exhaustion and greater stemness. In a ROR1-based CAR lethality model, drug cessation following toxicity onset reversed toxicity, thereby credentialing the platform as a safety switch. In the same model, reduced drug dosing opened a therapeutic window that resulted in tumor eradication in the absence of toxicity. SNIP CARs enable remote tuning of CAR activity, which provides solutions to safety and efficacy barriers that are currently limiting progress in using CAR-T cells to treat solid tumors.
Collapse
Affiliation(s)
- Louai Labanieh
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Robbie G Majzner
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Dorota Klysz
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Elena Sotillo
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Chris J Fisher
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - José G Vilches-Moure
- Department of Comparative Medicine, School of Medicine, Stanford University, Stanford, CA 94305, USA
| | - Kaithlen Zen B Pacheco
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Meena Malipatlolla
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Peng Xu
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jessica H Hui
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Tara Murty
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Medical Scientist Training Program, Stanford University School of Medicine, Stanford, CA 94305, USA; Biophysics Program, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Johanna Theruvath
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Nishant Mehta
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Sean A Yamada-Hunter
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Evan W Weber
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Sabine Heitzeneder
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Kevin R Parker
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA
| | | | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA 94305, USA; Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Michael Z Lin
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Department of Neurobiology, Stanford University, Stanford, CA 94305, USA; Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, CA 94305, USA
| | - Jennifer R Cochran
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Chemical Engineering, Stanford University, Stanford, CA 94305, USA
| | - Crystal L Mackall
- Center for Cancer Cell Therapy, Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Pediatrics, Stanford University School of Medicine, Stanford, CA 94305, USA; Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA 94305, USA; Department of Medicine, Stanford University School of Medicine, Stanford, CA 94305, USA.
| |
Collapse
|
25
|
Wang W, Ouyang D. Opportunities and challenges of physiologically based pharmacokinetic modeling in drug delivery. Drug Discov Today 2022; 27:2100-2120. [PMID: 35452792 DOI: 10.1016/j.drudis.2022.04.015] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 03/03/2022] [Accepted: 04/13/2022] [Indexed: 12/15/2022]
Abstract
Physiologically based pharmacokinetic (PBPK) modeling is an important in silico tool to bridge drug properties and in vivo PK behaviors during drug development. Over the recent decade, the PBPK method has been largely applied to drug delivery systems (DDS), including oral, inhaled, transdermal, ophthalmic, and complex injectable products. The related therapeutic agents have included small-molecule drugs, therapeutic proteins, nucleic acids, and even cells. Simulation results have provided important insights into PK behaviors of new dosage forms, which strongly support drug regulation. In this review, we comprehensively summarize recent progress in PBPK applications in drug delivery, which shows large opportunities for facilitating drug development. In addition, we discuss the challenges of applying this methodology from a practical viewpoint.
Collapse
Affiliation(s)
- Wei Wang
- Institute of Chinese Medical Sciences (ICMS), State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, China; Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macau, China
| | - Defang Ouyang
- Institute of Chinese Medical Sciences (ICMS), State Key Laboratory of Quality Research in Chinese Medicine, University of Macau, Macau, China; Department of Public Health and Medicinal Administration, Faculty of Health Sciences, University of Macau, Macau, China.
| |
Collapse
|
26
|
Oberholtzer N, Quinn KM, Chakraborty P, Mehrotra S. New Developments in T Cell Immunometabolism and Implications for Cancer Immunotherapy. Cells 2022; 11:708. [PMID: 35203357 PMCID: PMC8870179 DOI: 10.3390/cells11040708] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Revised: 02/05/2022] [Accepted: 02/10/2022] [Indexed: 12/12/2022] Open
Abstract
Despite rapid advances in the field of immunotherapy, the elimination of established tumors has not been achieved. Many promising new treatments such as adoptive cell therapy (ACT) fall short, primarily due to the loss of T cell effector function or the failure of long-term T cell persistence. With the availability of new tools and advancements in technology, our understanding of metabolic processes has increased enormously in the last decade. Redundancy in metabolic pathways and overlapping targets that could address the plasticity and heterogenous phenotypes of various T cell subsets have illuminated the need for understanding immunometabolism in the context of multiple disease states, including cancer immunology. Herein, we discuss the developing field of T cell immunometabolism and its crucial relevance to improving immunotherapeutic approaches. This in-depth review details the metabolic pathways and preferences of the antitumor immune system and the state of various metabolism-targeting therapeutic approaches.
Collapse
Affiliation(s)
- Nathaniel Oberholtzer
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.Q.); (P.C.)
| | | | | | - Shikhar Mehrotra
- Department of Surgery, Medical University of South Carolina, Charleston, SC 29425, USA; (K.M.Q.); (P.C.)
| |
Collapse
|
27
|
Abstract
The association of gut microbiota with gastrointestinal carcinogenesis has been heavily investigated since the recent advance in sequencing technology. Accumulating evidence has revealed the critical roles of commensal microbes in cancer progression. Given by its importance, emerging studies have focussed on targeting microbiota to ameliorate therapeutic effectiveness. It is now clear that the microbial community is closely related to the efficacy of chemotherapy, while the correlation of microbiota with immunotherapy is much less studied. Herein, we review the up-to-date findings on the influence of gut microbiota on three common immunotherapies including adoptive cell transfer, immune checkpoint blockade, and CpG-oligodeoxynucleotide therapy. We then explore three microbiota-targeted strategies that may improve treatment efficacy, involving dietary intervention, probiotics supplementation, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Harry Cheuk Hay Lau
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Joseph Jao-Yiu Sung
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Sha Tin, Hong Kong
| | - Jun Yu
- Institute of Digestive Disease, Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, CUHK Shenzhen Research Institute, the Chinese University of Hong Kong, Sha Tin, Hong Kong,CONTACT Jun Yu Institute of Digestive Disease, Department of Medicine and Therapeutics, Prince of Wales Hospital, the Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
28
|
Angelici B, Shen L, Schreiber J, Abraham A, Benenson Y. An AAV gene therapy computes over multiple cellular inputs to enable precise targeting of multifocal hepatocellular carcinoma in mice. Sci Transl Med 2021; 13:eabh4456. [PMID: 34910545 DOI: 10.1126/scitranslmed.abh4456] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
[Figure: see text].
Collapse
Affiliation(s)
- Bartolomeo Angelici
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel 4058, Switzerland
| | - Linling Shen
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel 4058, Switzerland.,Department of Chemistry, University of Basel, Mattenstrasse 24a, Basel 4058, Switzerland
| | - Joerg Schreiber
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel 4058, Switzerland
| | - Anthony Abraham
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel 4058, Switzerland
| | - Yaakov Benenson
- Department of Biosystems Science and Engineering, ETH Zurich, Mattenstrasse 26, Basel 4058, Switzerland
| |
Collapse
|
29
|
Savanur MA, Weinstein-Marom H, Gross G. Implementing Logic Gates for Safer Immunotherapy of Cancer. Front Immunol 2021; 12:780399. [PMID: 34804073 PMCID: PMC8600566 DOI: 10.3389/fimmu.2021.780399] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Accepted: 10/21/2021] [Indexed: 12/14/2022] Open
Abstract
Targeting solid tumors with absolute precision is a long-standing challenge in cancer immunotherapy. The identification of antigens, which are expressed by a large fraction of tumors of a given type and, preferably, across various types, but not by normal cells, holds the key to developing safe, off-the-shelf immunotherapies. Although the quest for widely shared, strictly tumor-specific antigens has been the focus of tremendous effort, only few such candidates have been implicated. Almost all antigens that are currently explored as targets for chimeric antigen receptor (CAR) or T cell receptor (TCR)-T cell therapy are also expressed by healthy cells and the risk of on-target off-tumor toxicity has remained a major concern. Recent studies suggest that this risk could be obviated by targeting instead combinations of two or more antigens, which are co-expressed by tumor but not normal cells and, as such, are tumor-specific. Moreover, the expression of a shared tumor antigen along with the lack of a second antigen that is expressed by normal tissues can also be exploited for precise recognition. Additional cues, antigenic or non-antigenic ones, which characterize the tumor microenvironment, could be harnessed to further increase precision. This review focuses on attempts to define the targetable signatures of tumors and assesses different strategies employing advanced synthetic biology for translating such information into safer modes of immunotherapy, implementing the principles of Boolean logic gates.
Collapse
Affiliation(s)
- Mohammed Azharuddin Savanur
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Hadas Weinstein-Marom
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| | - Gideon Gross
- Laboratory of Immunology, MIGAL - Galilee Research Institute, Kiryat Shmona, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
| |
Collapse
|
30
|
Khorasani ABS, Sanaei MJ, Pourbagheri-Sigaroodi A, Ghaffari SH, Bashash D. CAR T cell therapy in solid tumors; with an extensive focus on obstacles and strategies to overcome the challenges. Int Immunopharmacol 2021; 101:108260. [PMID: 34678690 DOI: 10.1016/j.intimp.2021.108260] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 09/19/2021] [Accepted: 10/11/2021] [Indexed: 12/12/2022]
Abstract
The application of the CAR T cell therapy in hematologic malignancies holds prosperous results that intensified the unprecedented enthusiasm to employ this fascinating strategy in other types of human malignancies. Although the researchers invested a great deal of effort to exploit the utmost efficacy of these cells in the context of solid tumors, few articles reviewed obstacles and opportunities. The current review aims to provide comprehensive literature of recent advances of CAR T cell therapy in a wide range of solid tumors; and also, to discuss the original data obtained from international research laboratories on this topic. Despite promising results, several radical obstacles are on the way of this approach. This review discusses the most important drawbacks and also responds to questions on how the intrinsic features of solid tumors in addition to the tumor microenvironment-related challenges and the immune-relating adverse effects can curb satisfactory outcomes of CAR T cells. The last section allocates a special focus on innovative and contemporary policies which have already been adopted to surmount these challenges. Finally, we comment on the future research aspects in which the efficacy, as well as the safety of CAR T cell therapy, might be improved.
Collapse
Affiliation(s)
| | - Mohammad-Javad Sanaei
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Atieh Pourbagheri-Sigaroodi
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Seyed H Ghaffari
- Hematology, Oncology and Stem Cell Transplantation Research Center, Shariati Hospital, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Davood Bashash
- Department of Hematology and Blood Banking, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Lawal G, Xiao Y, Rahnemai-Azar AA, Tsilimigras DI, Kuang M, Bakopoulos A, Pawlik TM. The Immunology of Hepatocellular Carcinoma. Vaccines (Basel) 2021; 9:vaccines9101184. [PMID: 34696292 PMCID: PMC8538643 DOI: 10.3390/vaccines9101184] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Revised: 09/20/2021] [Accepted: 10/08/2021] [Indexed: 12/13/2022] Open
Abstract
Liver cancer is the third leading cause of cancer death worldwide. Hepatocellular carcinoma (HCC) is the most common primary malignant tumor of the liver. Liver resection or transplantation offer the only potentially curative options for HCC; however, many patients are not candidates for surgical resection, either due to presentation at advanced stages or poor liver function and portal hypertension. Liver transplantation is also limited to patients with certain characteristics, such as those that meet the Milan criteria (one tumor ≤ 5 cm, or up to three tumors no larger than 3 cm, along with the absence of gross vascular invasion or extrahepatic spread). Locoregional therapies, such as ablation (radiofrequency, ethanol, cryoablation, microwave), trans-arterial therapies like chemoembolization (TACE) or radioembolization (TARE), and external beam radiation therapy, have been used mainly as palliative measures with poor prognosis. Therefore, emerging novel systemic treatments, such as immunotherapy, have increasingly become popular. HCC is immunogenic, containing infiltrating tumor-specific T-cell lymphocytes and other immune cells. Immunotherapy may provide a more effective and discriminatory targeting of tumor cells through induction of a tumor-specific immune response in cancer cells and can improve post-surgical recurrence-free survival in HCC. We herein review evidence supporting different immunomodulating cell-based technology relative to cancer therapy in vaccines and targeted therapies, such as immune checkpoint inhibitors, in the management of hepatocellular carcinoma among patients with advanced disease.
Collapse
Affiliation(s)
- Gbemisola Lawal
- Division of Surgical Oncology, Department of Surgery, Arrowhead Regional Cancer Center, California University of Science and Medicine, Colton, CA 92324, USA; (G.L.); (A.A.R.-A.)
| | - Yao Xiao
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (Y.X.); (M.K.)
| | - Amir A. Rahnemai-Azar
- Division of Surgical Oncology, Department of Surgery, Arrowhead Regional Cancer Center, California University of Science and Medicine, Colton, CA 92324, USA; (G.L.); (A.A.R.-A.)
| | - Diamantis I. Tsilimigras
- Department of Surgery, The Ohio State Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
- Correspondence: ; Tel.: +1-215-987-9177
| | - Ming Kuang
- Department of Liver Surgery, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou 510080, China; (Y.X.); (M.K.)
| | - Anargyros Bakopoulos
- Department of Surgery, Attikon University Hospital, University of Athens, 12462 Athens, Greece;
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State Comprehensive Cancer Center, The Ohio State University College of Medicine, Columbus, OH 43210, USA;
| |
Collapse
|
32
|
Novel CS1 CAR-T Cells and Bispecific CS1-BCMA CAR-T Cells Effectively Target Multiple Myeloma. Biomedicines 2021; 9:biomedicines9101422. [PMID: 34680541 PMCID: PMC8533376 DOI: 10.3390/biomedicines9101422] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 09/29/2021] [Accepted: 10/01/2021] [Indexed: 11/26/2022] Open
Abstract
Multiple myeloma (MM) is a hematological cancer caused by abnormal proliferation of plasma cells in the bone marrow, and novel types of treatment are needed for this deadly disease. In this study, we aimed to develop novel CS1 CAR-T cells and bispecific CS1-BCMA CAR-T cells to specifically target multiple myeloma. We generated a new CS1 (CD319, SLAM-7) antibody, clone (7A8D5), which specifically recognized the CS1 antigen, and we applied it for the generation of CS1-CAR. CS1-CAR-T cells caused specific killing of CHO-CS1 target cells with secretion of IFN-gamma and targeted multiple myeloma cells. In addition, bispecific CS1-BCMA-41BB-CD3 CAR-T cells effectively killed CHO-CS1 and CHO-BCMA target cells, killed CS1/BCMA-positive multiple myeloma cells, and secreted IFN-gamma. Moreover, CS1-CAR-T cells and bispecific CS1-BCMA CAR-T cells effectively blocked MM1S multiple myeloma tumor growth in vivo. These data for the first time demonstrate that novel CS1 and bispecific CS1-BCMA-CAR-T cells are effective in targeting MM cells and provide a basis for future clinical trials.
Collapse
|
33
|
Johnson AJ, Wei J, Rosser JM, Künkele A, Chang CA, Reid AN, Jensen MC. Rationally Designed Transgene-Encoded Cell-Surface Polypeptide Tag for Multiplexed Programming of CAR T-cell Synthetic Outputs. Cancer Immunol Res 2021; 9:1047-1060. [PMID: 34244298 DOI: 10.1158/2326-6066.cir-20-0470] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 03/24/2021] [Accepted: 07/02/2021] [Indexed: 11/16/2022]
Abstract
Synthetic immunology, as exemplified by chimeric antigen receptor (CAR) T-cell immunotherapy, has transformed the treatment of relapsed/refractory B cell-lineage malignancies. However, there are substantial barriers-including limited tumor homing, lack of retention of function within a suppressive tumor microenvironment, and antigen heterogeneity/escape-to using this technology to effectively treat solid tumors. A multiplexed engineering approach is needed to equip effector T cells with synthetic countermeasures to overcome these barriers. This, in turn, necessitates combinatorial use of lentiviruses because of the limited payload size of current lentiviral vectors. Accordingly, there is a need for cell-surface human molecular constructs that mark multi-vector cotransduced T cells, to enable their purification ex vivo and their tracking in vivo. To this end, we engineered a cell surface-localizing polypeptide tag based on human HER2, designated HER2t, that was truncated in its extracellular and intracellular domains to eliminate ligand binding and signaling, respectively, and retained the membrane-proximal binding epitope of the HER2-specific mAb trastuzumab. We linked HER2t to CAR coexpression in lentivirally transduced T cells and showed that co-transduction with a second lentivirus expressing our previously described EGFRt tag linked to a second CAR efficiently generated bispecific dual-CAR T cells. Using the same approach, we generated T cells expressing a CAR and a second module, a chimeric cytokine receptor. The HER2txEGFRt multiplexing strategy is now being deployed for the manufacture of CD19xCD22 bispecific CAR T-cell products for the treatment of acute lymphoblastic leukemia (NCT03330691).
Collapse
Affiliation(s)
- Adam J Johnson
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington.,Seattle Children's Therapeutics, Seattle, Washington
| | - Jia Wei
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington.,Seattle Children's Therapeutics, Seattle, Washington
| | - James M Rosser
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington.,Seattle Children's Therapeutics, Seattle, Washington
| | - Annette Künkele
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington
| | - Cindy A Chang
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington
| | - Aquene N Reid
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington.,Seattle Children's Therapeutics, Seattle, Washington
| | - Michael C Jensen
- Ben Towne Center for Childhood Cancer Research, Seattle Children's Research Institute, Seattle, Washington. .,Seattle Children's Therapeutics, Seattle, Washington.,Department of Pediatrics, University of Washington, Seattle, Washington.,Department of Bioengineering, University of Washington, Seattle, Washington
| |
Collapse
|
34
|
Rousso-Noori L, Mastandrea I, Talmor S, Waks T, Globerson Levin A, Haugas M, Teesalu T, Alvarez-Vallina L, Eshhar Z, Friedmann-Morvinski D. P32-specific CAR T cells with dual antitumor and antiangiogenic therapeutic potential in gliomas. Nat Commun 2021; 12:3615. [PMID: 34127674 PMCID: PMC8203650 DOI: 10.1038/s41467-021-23817-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Accepted: 05/12/2021] [Indexed: 12/29/2022] Open
Abstract
Glioblastoma is considered one of the most aggressive malignancies in adult and pediatric patients. Despite decades of research no curative treatment is available and it thus remains associated with a very dismal prognosis. Although recent pre-clinical and clinical studies have demonstrated the feasibility of chimeric antigen receptors (CAR) T cell immunotherapeutic approach in glioblastoma, tumor heterogeneity and antigen loss remain among one of the most important challenges to be addressed. In this study, we identify p32/gC1qR/HABP/C1qBP to be specifically expressed on the surface of glioma cells, making it a suitable tumor associated antigen for redirected CAR T cell therapy. We generate p32 CAR T cells and find them to recognize and specifically eliminate p32 expressing glioma cells and tumor derived endothelial cells in vitro and to control tumor growth in orthotopic syngeneic and xenograft mouse models. Thus, p32 CAR T cells may serve as a therapeutic option for glioblastoma patients. Chimeric antigen receptor (CAR) T cell therapy has been proposed as a promising approach for treating glioblastoma. Here the authors show that p32 is expressed in murine and human glioma and that p32-directed CAR-T cells promote anti-tumor responses in preclinical models by targeting glioma cells and tumor derived endothelial cells.
Collapse
Affiliation(s)
- Liat Rousso-Noori
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Ignacio Mastandrea
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Shauli Talmor
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Tova Waks
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Maarja Haugas
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia
| | - Tambet Teesalu
- Laboratory of Cancer Biology, Institute of Biomedicine, Centre of Excellence for Translational Medicine, University of Tartu, Tartu, Estonia.,Cancer Research Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA.,Center for Nanomedicine and Department of Cell, Molecular and Developmental Biology, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Luis Alvarez-Vallina
- Cancer Immunotherapy Unit (UNICA), Department of Immunology, Hospital Universitario 12 de Octubre, Madrid, Spain.,Immuno-Oncology and Immunotherapy Group, Instituto de Investigación Sanitaria 12 de Octubre (imas12), Madrid, Spain.,Immunotherapy and Cell Engineering Laboratory, Department of Engineering, Aarhus University, Aarhus, Denmark
| | - Zelig Eshhar
- Tel Aviv Sourasky Medical Center (TASMC), Tel Aviv, Israel.,Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dinorah Friedmann-Morvinski
- School of Neurobiology, Biochemistry and Biophysics, The George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel. .,Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
35
|
Kumar ARK, Shou Y, Chan B, L K, Tay A. Materials for Improving Immune Cell Transfection. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2021; 33:e2007421. [PMID: 33860598 DOI: 10.1002/adma.202007421] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/08/2020] [Indexed: 06/12/2023]
Abstract
Chimeric antigen receptor T cell (CAR-T) therapy holds great promise for preventing and treating deadly diseases such as cancer. However, it remains challenging to transfect and engineer primary immune cells for clinical cell manufacturing. Conventional tools using viral vectors and bulk electroporation suffer from low efficiency while posing risks like viral transgene integration and excessive biological perturbations. Emerging techniques using microfluidics, nanoparticles, and high-aspect-ratio nanostructures can overcome these challenges, and on top of that, provide universal and high-throughput cargo delivery. Herein, the strengths and limitations of traditional and emerging materials for immune cell transfection, and commercial development of these tools, are discussed. To enhance the characterization of transfection techniques and uptake by the clinical community, a list of in vitro and in vivo assays to perform, along with relevant protocols, is recommended. The overall aim, herein, is to motivate the development of novel materials to meet rising demand in transfection for clinical CAR-T cell manufacturing.
Collapse
Affiliation(s)
- Arun R K Kumar
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| | - Yufeng Shou
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Brian Chan
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Krishaa L
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
| | - Andy Tay
- Department of Biomedical Engineering, National University of Singapore, Singapore, 117583, Singapore
- Institute for Health Innovation & Technology, National University of Singapore, Singapore, 117599, Singapore
| |
Collapse
|
36
|
Akhoundi M, Mohammadi M, Sahraei SS, Sheykhhasan M, Fayazi N. CAR T cell therapy as a promising approach in cancer immunotherapy: challenges and opportunities. Cell Oncol (Dordr) 2021; 44:495-523. [PMID: 33759063 DOI: 10.1007/s13402-021-00593-1] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Chimeric antigen receptor (CAR)-modified T cell therapy has shown great potential in the immunotherapy of patients with hematologic malignancies. In spite of this striking achievement, there are still major challenges to overcome in CAR T cell therapy of solid tumors, including treatment-related toxicity and specificity. Also, other obstacles may be encountered in tackling solid tumors, such as their immunosuppressive microenvironment, the heterogeneous expression of cell surface markers, and the cumbersome arrival of T cells at the tumor site. Although several strategies have been developed to overcome these challenges, aditional research aimed at enhancing its efficacy with minimum side effects, the design of precise yet simplified work flows and the possibility to scale-up production with reduced costs and related risks is still warranted. CONCLUSIONS Here, we review main strategies to establish a balance between the toxicity and activity of CAR T cells in order to enhance their specificity and surpass immunosuppression. In recent years, many clinical studies have been conducted that eventually led to approved products. To date, the FDA has approved two anti-CD19 CAR T cell products for non-Hodgkin lymphoma therapy, i.e., axicbtagene ciloleucel and tisagenlecleucel. With all the advances that have been made in the field of CAR T cell therapy for hematologic malignancies therapy, ongoing studies are focused on optimizing its efficacy and specificity, as well as reducing the side effects. Also, the efforts are poised to broaden CAR T cell therapeutics for other cancers, especially solid tumors.
Collapse
Affiliation(s)
- Maryam Akhoundi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Seyedeh Saeideh Sahraei
- Department of Reproductive Biology, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran
| | - Mohsen Sheykhhasan
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran. .,Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom Branch, Qom, Iran.
| | - Nashmin Fayazi
- Research Center for Molecular Medicine, Hamadan University of Medical Sciences, Hamadan, Iran
| |
Collapse
|
37
|
Marofi F, Rahman HS, Thangavelu L, Dorofeev A, Bayas-Morejón F, Shirafkan N, Shomali N, Chartrand MS, Jarahian M, Vahedi G, Mohammed RN, Shahrokh S, Akbari M, Khiavi FM. Renaissance of armored immune effector cells, CAR-NK cells, brings the higher hope for successful cancer therapy. Stem Cell Res Ther 2021; 12:200. [PMID: 33752707 PMCID: PMC7983395 DOI: 10.1186/s13287-021-02251-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/28/2021] [Indexed: 02/13/2023] Open
Abstract
In recent decades, a new method of cellular immunotherapy was introduced based on engineering and empowering the immune effector cells. In this type of immunotherapy, the immune effector cells are equipped with chimeric antigen receptor (CAR) to specifically target cancer cells. In much of the trials and experiments, CAR-modified T cell immunotherapy has achieved very promising therapeutic results in the treatment of some types of cancers and infectious diseases. However, there are also some considerable drawbacks in the clinical application of CAR-T cells although much effort is in progress to rectify the issues. In some conditions, CAR-T cells initiate over-activated and strong immune responses, therefore, causing unexpected side-effects such as systemic cytokine toxicity (i.e., cytokine release syndrome), neurotoxicity, on-target, off-tumor toxicity, and graft-versus-host disease (GvHD). To overcome these limitations in CAR-T cell immunotherapy, NK cells as an alternative source of immune effector cells have been utilized for CAR-engineering. Natural killer cells are key players of the innate immune system that can destroy virus-infected cells, tumor cells, or other aberrant cells with their efficient recognizing capability. Compared to T cells, CAR-transduced NK cells (CAR-NK) have several advantages, such as safety in clinical use, non-MHC-restricted recognition of tumor cells, and renewable and easy cell sources for their preparation. In this review, we will discuss the recent preclinical and clinical studies, different sources of NK cells, transduction methods, possible limitations and challenges, and clinical considerations.
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Heshu Sulaiman Rahman
- Department of Physiology, College of Medicine, University of Suleimanyah, Sulaymaniyah, Iraq
| | - Lakshmi Thangavelu
- Associate professor, Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | - Aleksey Dorofeev
- Department of Propaedeutics of Dental Diseases, I.M. Sechenov First Moscow State Medical University (Sechenov University,), Moscow, Russian Federation
| | - Favian Bayas-Morejón
- Center for Research and Biotechnological Development, Research Department, Bolivar State University, Faculty of Agricultural Sciences, Natural Resources and the Environment, CP 020150 Guaranda, Ecuador
| | - Naghmeh Shirafkan
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Navid Shomali
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mostafa Jarahian
- German Cancer Research Center, Toxicology and Chemotherapy Unit (G401), 69120 Heidelberg, Germany
| | - Ghasem Vahedi
- Department of Immunology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | - Rebar N. Mohammed
- College of Veterinary Medicine, University of Sulaimani, Suleimanyah, Iraq
| | - Somayeh Shahrokh
- Department of Pathobiology, Faculty of Veterinary Medicine, University of Shahrekord, Shahrekord, Iran
| | - Morteza Akbari
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | |
Collapse
|
38
|
Anna F, Bole-Richard E, LeMaoult J, Escande M, Lecomte M, Certoux JM, Souque P, Garnache F, Adotevi O, Langlade-Demoyen P, Loustau M, Caumartin J. First immunotherapeutic CAR-T cells against the immune checkpoint protein HLA-G. J Immunother Cancer 2021; 9:e001998. [PMID: 33737343 PMCID: PMC7978334 DOI: 10.1136/jitc-2020-001998] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/15/2021] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND CAR-T cells immunotherapy is a breakthrough in the treatment of hematological malignancies such as acute lymphoblastic leukemia (ALL) and B-cell malignancies. However, CAR-T therapies face major hurdles such as the lack of tumor-specific antigen (TSA), and immunosuppressive tumor microenvironment sometimes caused by the tumorous expression of immune checkpoints (ICPs) such as HLA-G. Indeed, HLA-G is remarkable because it is both a potent ICP and a TSA. HLA-G tumor expression causes immune escape by impairing innate and adaptive immune responses and by inducing a suppressive microenvironment. Yet, to date, no immunotherapy targets it. METHODS We have developed two anti-HLA-G third-generation CARs based on new anti-HLA-G monoclonal antibodies. RESULTS Anti-HLA-G CAR-T cells were specific for immunosuppressive HLA-G isoforms. HLA-G-activated CAR-T cells polarized toward T helper 1, and became cytotoxic against HLA-G+ tumor cells. In vivo, anti-HLA-G CAR-T cells were able to control and eliminate HLA-G+ tumor cells. The interaction of tumor-HLA-G with interleukin (IL)T2-expressing T cells is known to result in effector T cell functional inhibition, but anti-HLA-G CAR-T cells were insensitive to this inhibition and still exerted their function even when expressing ILT2. Lastly, we show that anti-HLA-G CAR-T cells differentiated into long-term memory effector cells, and seemed not to lose function even after repeated stimulation by HLA-G-expressing tumor cells. CONCLUSION We report for the first time that HLA-G, which is both a TSA and an ICP, constitutes a valid target for CAR-T cell therapy to specifically target and eliminate both tumor cells and HLA-G+ suppressive cells.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/genetics
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/metabolism
- Antigens, CD/metabolism
- Cell Differentiation
- Coculture Techniques
- Cytotoxicity, Immunologic
- HLA-G Antigens/immunology
- HLA-G Antigens/metabolism
- Humans
- Immunologic Memory
- Immunotherapy, Adoptive
- K562 Cells
- Leukemia, Erythroblastic, Acute/genetics
- Leukemia, Erythroblastic, Acute/immunology
- Leukemia, Erythroblastic, Acute/metabolism
- Leukemia, Erythroblastic, Acute/therapy
- Leukocyte Immunoglobulin-like Receptor B1/metabolism
- Memory T Cells/immunology
- Memory T Cells/metabolism
- Memory T Cells/transplantation
- Mice, Inbred NOD
- Mice, SCID
- Phenotype
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Time Factors
- Tumor Microenvironment
- Xenograft Model Antitumor Assays
- Mice
Collapse
Affiliation(s)
- François Anna
- Preclinical Department, Invectys, Paris, France
- Molecular Virology and Vaccinology Unit, Virology Department, Pasteur Institute, Paris, Île-de-France, France
| | - Elodie Bole-Richard
- INSERM UMR1098 RIGHT Interactions hôte-greffon-tumeur - Ingénierie Cellulaire et Génique, Besancon, Franche-Comté, France
- Université Bourgogne Franche-Comté, Besançon, France
- Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France
| | - Joel LeMaoult
- Service de Recherche en Hémato-Immunologie (SRHI), CEA, Paris, France
- Université de Paris, Paris, Île-de-France, France
| | | | | | - Jean-Marie Certoux
- INSERM UMR1098 RIGHT Interactions hôte-greffon-tumeur - Ingénierie Cellulaire et Génique, Besancon, Franche-Comté, France
- Université Bourgogne Franche-Comté, Besançon, France
- Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France
| | - Philippe Souque
- Molecular Virology and Vaccinology Unit, Virology Department, Pasteur Institute, Paris, Île-de-France, France
| | - Francine Garnache
- INSERM UMR1098 RIGHT Interactions hôte-greffon-tumeur - Ingénierie Cellulaire et Génique, Besancon, Franche-Comté, France
- Université Bourgogne Franche-Comté, Besançon, France
- Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France
| | - Olivier Adotevi
- INSERM UMR1098 RIGHT Interactions hôte-greffon-tumeur - Ingénierie Cellulaire et Génique, Besancon, Franche-Comté, France
- Université Bourgogne Franche-Comté, Besançon, France
- Etablissement Français du Sang Bourgogne Franche-Comté, Besançon, France
| | | | | | | |
Collapse
|
39
|
CAR-T Therapy, the End of a Chapter or the Beginning of a New One? Cancers (Basel) 2021; 13:cancers13040853. [PMID: 33670515 PMCID: PMC7922383 DOI: 10.3390/cancers13040853] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2021] [Revised: 02/14/2021] [Accepted: 02/14/2021] [Indexed: 12/19/2022] Open
Abstract
Simple Summary CAR-T therapy is a breakthrough treatment in our fight against cancer. It was recently approved for the treatment of advanced diffuse large B-cell lymphoma and acute lymphoblastic leukaemia after the failure of previous multiple therapies. The positive results achieved in the registration studies for those patients were remarkable. Unfortunately, this was not the end of this chapter. Disease relapses occur in the range of 30–60% of patients treated with CAR-T therapy. Cytokine release syndrome represents a major side effect for treatment with CAR-T therapy. Notwithstanding, the high positive results triggered the start of a huge research activity of CAR-T therapy in other haematologic malignancies such as acute myelogenous leukaemia, Hodgkin’s disease, chronic lymphocytic leukaemia, and multiple myeloma. The research is also trying to overcome the hurdles stated above. These activities represent a new chapter in the management of haematologic malignancies with CAR-T therapy. Abstract Chimeric antigen receptor-T (CAR-T) therapy targeting CD19 has revolutionised the treatment of advanced acute lymphoblastic leukaemia (ALL) and diffuse large B-cell lymphoma (DLBCL). The ability to specifically target the cancer cells has shown high positive results as reported in the registration studies. The success of CAR-T therapy in the first two indications led to the initiation of a large number of studies testing CAR-T therapy in different haematologic tumours such as acute myelogenous leukaemia (AML), Hodgkin’s disease (HD), chronic lymphocytic leukaemia (CLL), multiple myeloma (MM), as well as different solid tumours. Unfortunately, relapses occurred in patients treated with CAR-T therapy, calling for the development of effective subsequent therapies. Likewise, this novel mechanism of action was also accompanied by a different toxicity profile, such as cytokine release syndrome (CRS). Patients’ access to the treatment is still limited by its cost. Notwithstanding, this did not prohibit further development of this new therapy to treat other malignancies. This research activity of CAR-T therapy moves it from being used as an end-stage treatment for ALL and DLBCL to a new therapeutic option for a wide range of patients with different haematologic and solid tumours.
Collapse
|
40
|
Zekri L, Vogt F, Osburg L, Müller S, Kauer J, Manz T, Pflügler M, Maurer A, Heitmann JS, Hagelstein I, Märklin M, Hörner S, Todenhöfer T, Calaminus C, Stenzl A, Pichler B, la Fougère C, Schneider MA, Rammensee H, Zender L, Sipos B, Salih HR, Jung G. An IgG-based bispecific antibody for improved dual targeting in PSMA-positive cancer. EMBO Mol Med 2021; 13:e11902. [PMID: 33372710 PMCID: PMC7863392 DOI: 10.15252/emmm.201911902] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 11/26/2020] [Accepted: 12/01/2020] [Indexed: 12/14/2022] Open
Abstract
The prostate-specific membrane antigen (PSMA) has been demonstrated in numerous studies to be expressed specifically on prostate carcinoma cells and on the neovasculature of several other cancer entities. However, the simultaneous expression of PSMA on both, tumor cells as well as tumor vessels remains unclear, even if such "dual" expression would constitute an important asset to facilitate sufficient influx of effector cells to a given tumor site. We report here on the generation of a PSMA antibody, termed 10B3, which exerts superior dual reactivity on sections of prostate carcinoma and squamous cell carcinoma of the lung. 10B3 was used for the construction of T-cell recruiting bispecific PSMAxCD3 antibodies in Fab- and IgG-based formats, designated Fabsc and IgGsc, respectively. In vitro, both molecules exhibited comparable activity. In contrast, only the larger IgGsc molecule induced complete and durable elimination of established tumors in humanized mice due to favorable pharmacokinetic properties. Upon treatment of three patients with metastasized prostate carcinoma with the IgGsc reagent, marked activation of T cells and rapid reduction of elevated PSA levels were observed.
Collapse
|
41
|
Greenshpan Y, Sharabi O, Ottolenghi A, Cahana A, Kundu K, M Yegodayev K, Elkabets M, Gazit R, Porgador A. Synthetic promoters to induce immune-effectors into the tumor microenvironment. Commun Biol 2021; 4:143. [PMID: 33514819 PMCID: PMC7846768 DOI: 10.1038/s42003-021-01664-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Accepted: 11/20/2020] [Indexed: 11/21/2022] Open
Abstract
Harnessing the immune-system to eradicate cancer is becoming a reality in recent years. Engineered immune cells, such as chimeric antigen receptor (CAR) T cells, are facing the danger of an overt life-threatening immune response due to the ON-target OFF-tumor cytotoxicity and Cytokine Release Syndrome. We therefore developed synthetic promoters for regulation of gene expression under the control of inflammation and Hypoxia-induced signals that are associated with the tumor microenvironment (TME). We termed this methodology as chimeric-antigen-receptor-tumor-induced-vector (CARTIV). For proof of concept, we studied synthetic promoters based on promoter-responsive elements (PREs) of IFNγ, TNFα and hypoxia; triple PRE-based CARTIV promoter manifested a synergistic activity in cell-lines and potent activation in human primary T-cells. CARTIV platform can improve safety of CAR T-cells or other engineered immune-cells, providing TME-focused activity and opening a therapeutic window for many tumor-associated antigens that are also expressed by non-tumor healthy tissues.
Collapse
Affiliation(s)
- Yariv Greenshpan
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omri Sharabi
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Aner Ottolenghi
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Avishag Cahana
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Kiran Kundu
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ksenia M Yegodayev
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Moshe Elkabets
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Roi Gazit
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Angel Porgador
- Faculty of Health Sciences, The Shraga Segal Department of Microbiology, Immunology and Genetics, Ben-Gurion University of the Negev, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
42
|
From the T-cell receptor to cancer therapy: an interview with Tak W. Mak. Cell Death Differ 2020; 28:5-14. [PMID: 33335286 PMCID: PMC7745173 DOI: 10.1038/s41418-020-00666-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Accepted: 10/28/2020] [Indexed: 01/22/2023] Open
Abstract
This interview is part of a series of articles to mark the 25th anniversary of the launching of Cell Death and Differentiation.
Collapse
|
43
|
Samadani AA, Keymoradzdeh A, Shams S, Soleymanpour A, Rashidy-Pour A, Hashemian H, Vahidi S, Norollahi SE. CAR T-cells profiling in carcinogenesis and tumorigenesis: An overview of CAR T-cells cancer therapy. Int Immunopharmacol 2020; 90:107201. [PMID: 33249047 DOI: 10.1016/j.intimp.2020.107201] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2020] [Revised: 11/04/2020] [Accepted: 11/09/2020] [Indexed: 12/15/2022]
Abstract
Immunotherapy of cancer by chimeric antigen receptors (CAR) modified T-cell has a remarkable clinical potential for malignancies. Meaningly, it is a suitable cancer therapy to treat different solid tumors. CAR is a special recombinant protein combination with an antibody targeting structure alongside with signaling domain capacity on order to activate T cells. It is confirmed that the CAR-modified T cells have this ability to terminate and remove B cell malignancies. So, methodologies for investigations the pro risks and also strategies for neutralizing possible off-tumor consequences of are great importance successful protocols and strategies of CAR T-cell therapy can improve the efficacy and safety of this type of cancers. In this review article, we try to classify and illustrate main optimized plans in cancer CAR T-cell therapy.
Collapse
Affiliation(s)
- Ali Akbar Samadani
- Healthy Ageing Research Center, Neyshabur University of Medical Sciences, Neyshabur, Iran.
| | - Arman Keymoradzdeh
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Shima Shams
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Armin Soleymanpour
- Student Research Committee, School of Medicine, Guilan University of Medical Sciences, Rasht, Iran
| | - Ali Rashidy-Pour
- Research Center of Physiology, Semnan University of Medical Sciences, Semnan, Iran
| | - Houman Hashemian
- Pediatrics Diseases Research Center, 17 Shahrivar Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Sogand Vahidi
- Clinical Research Development Unit of Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| | - Seyedeh Elham Norollahi
- Clinical Research Development Unit of Poursina Hospital, Guilan University of Medical Sciences, Rasht, Iran
| |
Collapse
|
44
|
Date V, Nair S. Emerging vistas in CAR T-cell therapy: challenges and opportunities in solid tumors. Expert Opin Biol Ther 2020; 21:145-160. [PMID: 32882159 DOI: 10.1080/14712598.2020.1819978] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
INTRODUCTION Despite advances in modern evidence-based medicine, cancer remains a major cause of global disease-associated mortality. CAR T-cell therapy is a major histocompatibility complex (MHC)-independent immunotherapy involving adoptive cell transfer. Cancer immunotherapy witnessed a major breakthrough with the US FDA approval of the first chimeric antigen receptor (CAR) T-cell therapy KymriahTM (tisagenlecleucel) for relapsed or refractory (R/R) acute lymphoblastic leukemia (ALL) in August 2017 followed by approval of Yescarta® (axicabtagene ciloleucel) for R/R non-Hodgkin's lymphoma (NHL) in October 2017. AREAS COVERED We review the potential of CAR T-cell therapy which, despite showing great promise in hematological malignancies, faces significant challenges in targeting solid tumors. We address these challenges and discuss proposed strategies to overcome them in solid tumors. We highlight the potential of CAR T-cell therapy as cancer precision medicine and briefly discuss the 'financial toxicity' of CAR T-cell therapy. EXPERT OPINION Taken together, we discuss various strategies to circumvent the limitations of CAR T-cell therapy in solid tumors. Despite the rapid advances in CAR NK-cell therapies, there is immense scope for CAR T-cell therapy in solid tumors. We provide a synthetic review of CAR T-cell therapy that will drive future research and harness its full potential in cancer precision medicine for solid tumors.
Collapse
Affiliation(s)
- Varada Date
- Shobhaben Pratapbhai Patel School of Pharmacy & Technology Management, SVKM's NMIMS University , Mumbai, India
| | - Sujit Nair
- SVKM's Dr. Bhanuben Nanavati College of Pharmacy, University of Mumbai , Mumbai, India
| |
Collapse
|
45
|
Editor's Pick: Tumour-Associated Hypoxia: Can We Give Chimeric Antigen Receptor T Cells More Breathing Space? EUROPEAN MEDICAL JOURNAL 2020. [DOI: 10.33590/emj/20-00076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022] Open
Abstract
Immunotherapy using chimeric antigen receptor (CAR)-engineered T cells has encountered important limitations in the transition of their use from liquid to solid tumours. Success is dependent upon T-cell trafficking to, and functional persistence within, tumours that often present a metabolically and immunologically hostile microenvironment. Moreover, CAR targets that are tumour specific are extremely scarce. To address these issues, several strategies have been proposed to improve both tumour selectivity and safety. One approach involves the engineering of CAR-T cells that only deploy their effector function at tumour sites. Conceptually, a solution for this exploits the oxygen-limited nature of advanced tumour deposits through the engineering of CAR that are exclusively expressed or activated under conditions of profound hypoxia. T cells have a complex inter-relationship with oxygen, which also needs to be factored into the refinement of these technologies. Ideally, oxygen-sensing CAR should only function when oxygen tension is below 2%, as is commonly the case in solid tumours but rare in healthy tissue. Successful advancement of such technologies presents opportunities for solid tumour immunotherapy because it should broaden the target repertoire that may safely be exploited in this context.
Collapse
|
46
|
Qin D, Li D, Zhang B, Chen Y, Liao X, Li X, Alexander PB, Wang Y, Li QJ. Potential lung attack and lethality generated by EpCAM-specific CAR-T cells in immunocompetent mouse models. Oncoimmunology 2020; 9:1806009. [PMID: 32923168 PMCID: PMC7458607 DOI: 10.1080/2162402x.2020.1806009] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2020] [Accepted: 07/14/2020] [Indexed: 02/06/2023] Open
Abstract
The tumoricidal efficiency of human CAR-T cells is generally evaluated using immune-deficient mouse models; however, due to their immune-incompetency and the species-specific reactivity of a target antigen, these models are problematic to imitate CAR-T-induced adverse effects in the clinic. Epithelial cell adhesion molecule (EpCAM) is a tumor-associated antigen overtly presented on the cell surface of various carcinomas, making it an attractive target for CAR-T therapy. Here, we developed an anti-mouse EpCAM CAR to evaluate its safety and efficacy in immunocompetent mouse models. As previously reported for their human equivalents, murine EpCAM CAR-T cells exhibit promising anti-tumor efficacy in vitro and in vivo. However, after CAR-T infusion, various dose-depended toxicities including body weight loss, cytokine-release syndrome (CRS), and death were observed in both tumor-bearing and tumor-free mice. Pathological examination revealed unexpected and severe pulmonary immunopathology due to basal EpCAM expression in normal lung. While our study validates EpCAM CAR-T's potent anti-tumor efficacy, it also reveals that EpCAM CAR-T cells used for the treatment of solid tumors may cause lethal toxicity and should, therefore, be evaluated in patients with caution.
Collapse
Affiliation(s)
- Diyuan Qin
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| | - Dan Li
- Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, China
| | - Benxia Zhang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Yue Chen
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
| | - Xuelian Liao
- Department of oncology, The First People’s Hospital of Jintang, Chengdu, China
| | - Xiaoyu Li
- Institute of Drug Clinical Trial, West China Hospital, Sichuan University, Chengdu, China
| | | | - Yongsheng Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, China
- Institute of Drug Clinical Trial, West China Hospital, Sichuan University, Chengdu, China
| | - Qi-Jing Li
- Department of Immunology, Duke University Medical Center, Durham, NC, USA
| |
Collapse
|
47
|
Donnadieu E, Dupré L, Pinho LG, Cotta-de-Almeida V. Surmounting the obstacles that impede effective CAR T cell trafficking to solid tumors. J Leukoc Biol 2020; 108:1067-1079. [PMID: 32620049 PMCID: PMC7586996 DOI: 10.1002/jlb.1mr0520-746r] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
Innovative immunotherapies based on immune checkpoint targeting antibodies and engineered T cells are transforming the way we approach cancer treatment. However, although these T cell centered strategies result in marked and durable responses in patients across many different tumor types, they provide therapeutic efficacy only in a proportion of patients. A major challenge of immuno‐oncology is thereby to identify mechanisms responsible for resistance to cancer immunotherapy in order to overcome them via adapted strategies that will ultimately improve intrinsic efficacy and response rates. Here, we focus on the barriers that restrain the trafficking of chimeric antigen receptor (CAR)‐expressing T cells to solid tumors. Upon infusion, CAR T cells need to home into malignant sites, navigate within complex tumor environments, form productive interactions with cancer cells, deliver their cytotoxic activities, and finally persist. We review the accumulating evidence that the microenvironment of solid tumors contains multiple obstacles that hinder CAR T cells in the dynamic steps underlying their trafficking. We focus on how these hurdles may in part account for the failure of CAR T cell clinical trials in human carcinomas. Given the engineered nature of CAR T cells and possibilities to modify the tumor environment, there are ample opportunities to augment CAR T cell ability to efficiently find and combat tumors. We present some of these strategies, which represent a dynamic field of research with high potential for clinical applicability.
Collapse
Affiliation(s)
- Emmanuel Donnadieu
- Université de Paris, Institut Cochin, INSERM, U1016, CNRS, UMR8104, Paris, France
| | - Loïc Dupré
- INSERM, UMR1043, Centre de Physiopathologie de Toulouse Purpan, Toulouse, France.,CNRS, UMR5282, Toulouse, France.,Université Toulouse III Paul-Sabatier, Toulouse, France
| | - Lia Gonçalves Pinho
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| | - Vinicius Cotta-de-Almeida
- Laboratory on Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation (Fiocruz), Rio de Janeiro, Brazil.,National Institute of Science and Technology on Neuroimmunomodulation, Oswaldo Cruz Institute, Rio de Janeiro, Brazil
| |
Collapse
|
48
|
Combined Expression of Genetic Adjuvants Via mRNA Electroporation Exerts Multiple Immunostimulatory Effects on Antitumor T Cells. J Immunother 2020; 42:43-50. [PMID: 30489430 DOI: 10.1097/cji.0000000000000252] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Adoptive transfer of tumor-infiltrating lymphocytes (TILs) or gene-modified T cells expressing antitumor TCRs or chimeric antigen receptors often yields a high rate of clinical response in several types of cancer. New approaches for enhancing the functional properties of antitumor T cells could improve the clinical outcome of these treatments. To this end, we created 3 classes of genes, each designed to operate autonomously upon expression in T cells. We recently reported on the enhancing effects of constitutively active toll-like receptor 4 (caTLR4), membrane (mem) interleukin-2, memIL-12, and memIL-15, and self-oligomerizing, constitutively active CD40 (caCD40). Here, we evaluated their combined effects on peripheral blood CD8 T cells and different antimelanoma TIL cultures following mRNA electroporation. Expression in CD8 T cells induced transient production of interferon-γ and prolonged and robust upregulation of CD25, CD69, 4-1BB, and OX40. The adjuvants enhanced cytolytic activity of TILs and production of interferon-γ and TNF-α in the presence of autologous, but not mismatched, melanoma for at least 3 days after electroporation. Expression of the 3 adjuvants in young TILs from different patients markedly increased the expression of CD25, OX40, 4-1BB, CD127, and CD28 and exhibited cooperative and, at times, synergistic effects. Furthermore, predefined mixtures of mRNA encoding these adjuvants markedly enhanced the specific antitumor response of selected TILs and killing of autologous melanoma cells by young TILs. Our findings suggest that combinations of these new genetic adjuvants can substantially improve the functional properties of antitumor T cells, offering a new tool of unique versatility in adoptive cell therapy.
Collapse
|
49
|
Bai C, Gao S, Hu S, Liu X, Li H, Dong J, Huang A, Zhu L, Zhou P, Li S, Shao N. Self-Assembled Multivalent Aptamer Nanoparticles with Potential CAR-like Characteristics Could Activate T Cells and Inhibit Melanoma Growth. MOLECULAR THERAPY-ONCOLYTICS 2020; 17:9-20. [PMID: 32280743 PMCID: PMC7139145 DOI: 10.1016/j.omto.2020.03.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/09/2020] [Accepted: 03/10/2020] [Indexed: 01/02/2023]
Abstract
In this study, the CAR-like multivalent aptamer nanoparticles (X-polymers) were assembled with the dimer of murine CD28 RNA aptamer (CD28Apt7), the tetramer of CTLA-4 (cytotoxic T-lymphocyte-associated protein 4) RNA aptamer (Del60), and a folic acid labeled ssDNA fragment in a stable nucleic acid three-way junction scaffold (3WJ). Results showed that the X-polymers could recognize both the mCD28 and mCTLA-4 molecules. Confocal imaging and flow cytometry assays showed that the X-polymers could target both T cells and B16 cells in vitro. With the first costimulatory signals provided by the CD3 antibodies, the X-polymers could increase T cell proliferation and reverse the inhibitory effect of interleukin-2 (IL-2) secreting caused by exogenous B7.1 molecules on T cells in vitro. Results of our study also showed that X-polymers could inhibit mouse melanoma B16 cell growth both in vitro and in vivo. Our study demonstrated for the first time that the multivalent aptamer nanoparticle-activated T cells could fulfill the function of CAR-T, which promised a novel approach to developing a multi-functional design of aptamer drugs with potential CAR-like characteristics to enhance the safety of CAR-T cell immunotherapy.
Collapse
Affiliation(s)
- Chenjun Bai
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China.,Institute of Radiation Medicine, Beijing 100850, China
| | - Shanshan Gao
- Institute of Radiation Medicine, Beijing 100850, China
| | - Sai Hu
- Institute of Radiation Medicine, Beijing 100850, China
| | - Xuemei Liu
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Hui Li
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Jie Dong
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Aixue Huang
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Lingling Zhu
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| | - Pingkun Zhou
- Institute of Radiation Medicine, Beijing 100850, China
| | - Shaohua Li
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China.,The Fifth Medical Center of PLA General Hospital, Beijing 100039, China
| | - Ningsheng Shao
- Institute of Military Cognition and Brain Sciences, Beijing 100850, China
| |
Collapse
|
50
|
Long B, Qin L, Zhang B, Li Q, Wang L, Jiang X, Ye H, Zhang G, Yu Z, Jiao Z. CAR T‑cell therapy for gastric cancer: Potential and perspective (Review). Int J Oncol 2020; 56:889-899. [PMID: 32319561 DOI: 10.3892/ijo.2020.4982] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 12/13/2019] [Indexed: 12/24/2022] Open
Abstract
Gastric cancer (GC) is one of the most frequently diagnosed digestive malignancies and is the third leading cause of cancer‑associated death worldwide. Delayed diagnosis and poor prognosis indicate the urgent need for new therapeutic strategies. The success of chimeric antigen receptor (CAR) T‑cell therapy for chemotherapy‑refractory hematological malignancies has inspired the development of a similar strategy for GC treatment. Although using CAR T‑cells against GC is not without difficulty, results from preclinical studies remain encouraging. The current review summarizes relevant preclinical studies and ongoing clinical trials for the use of CAR T‑cells for GC treatment and investigates possible toxicities, as well as current clinical experiences and emerging approaches. With a deeper understanding of the tumor microenvironment, novel target epitopes and scientific‑technical progress, the potential of CAR T‑cell therapy for GC is anticipated in the near future.
Collapse
Affiliation(s)
- Bo Long
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Long Qin
- The Cuiying Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Boya Zhang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Qiong Li
- Department of Endocrinology, The First Hospital of Lanzhou University, Lanzhou, Gansu 730000, P.R. China
| | - Long Wang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Xiangyan Jiang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Huili Ye
- The Cuiying Center, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Genyuan Zhang
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Zeyuan Yu
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| | - Zuoyi Jiao
- Department of First General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730000, P.R. China
| |
Collapse
|