1
|
Ito Y, Suzuki T, Shimomura M, Takenouchi K, Ohnuki K, Shoda K, Kenmochi Y, Yagyu S, Matsuura K, Hayashi R, Nakatsura T. Feasibility of Intratumoral Administration With EPHB4-CAR-T Cells for the Treatment of Oral Squamous Cell Carcinoma. Cancer Sci 2025; 116:1227-1238. [PMID: 40029791 PMCID: PMC12044661 DOI: 10.1111/cas.70023] [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: 06/05/2024] [Revised: 01/13/2025] [Accepted: 02/05/2025] [Indexed: 05/02/2025] Open
Abstract
Oral squamous cell carcinoma (OSCC) represents the most common type of oral cancer, and its prognosis remains poor. In this study, we found that almost OSCC cases showed high Ephrin type-B receptor 4 (EPHB4) expression that was mainly localized on the membrane of tumor cells. Therefore, EPHB4 represents a potential target of chimeric antigen receptor (CAR) T cell therapy for OSCC treatment. Because the oral cavity can be directly accessed, local administration of CAR-T cells is feasible for treating OSCC. In this study, we investigated the efficacy of intratumoral injection of EPHB4-specific CAR-T cells in OSCC using xenograft models. To evaluate the anti-tumor effect, the SAS OSCC cell line or an OSCC patient-derived xenograft (PDX) tumor was subcutaneously implanted into NOD SCID gamma mice, and EPHB4-CAR-T cells were intratumorally injected twice. As expected, administration of CAR-T cells suppressed tumor growth of both SAS cells and PDX tumor. EPHB4 expression in tumor tissues was attenuated by CAR-T cell treatment, which was accompanied by a reduction in tumor area and accumulation of CAR-T cells. Our findings suggest that intratumoral injection of EPHB4-CAR-T cells represents a potential therapeutic strategy for OSCC.
Collapse
MESH Headings
- Animals
- Humans
- Receptor, EphB4/metabolism
- Receptor, EphB4/immunology
- Receptor, EphB4/genetics
- Mouth Neoplasms/therapy
- Mouth Neoplasms/pathology
- Mouth Neoplasms/immunology
- Mouth Neoplasms/metabolism
- Mice
- Cell Line, Tumor
- Xenograft Model Antitumor Assays
- Carcinoma, Squamous Cell/therapy
- Carcinoma, Squamous Cell/pathology
- Carcinoma, Squamous Cell/immunology
- Mice, SCID
- Immunotherapy, Adoptive/methods
- Female
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/metabolism
- Mice, Inbred NOD
- Male
- Injections, Intralesional
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- Feasibility Studies
- Middle Aged
Collapse
Affiliation(s)
- Yusuke Ito
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
- Department of Head and Neck SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Toshihiro Suzuki
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Manami Shimomura
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Kazumasa Takenouchi
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Kazunobu Ohnuki
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Kayoko Shoda
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Yuka Kenmochi
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| | - Shigeki Yagyu
- Innovative Research and Liaison OrganizationShinshu UniversityMatsumotoJapan
- Department of PediatricsKyoto Prefectural University of MedicineKyotoJapan
| | - Kazuto Matsuura
- Department of Head and Neck SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Ryuichi Hayashi
- Department of Head and Neck SurgeryNational Cancer Center Hospital EastKashiwaJapan
| | - Tetsuya Nakatsura
- Division of Cancer ImmunotherapyExploratory Oncology Research and Clinical Trial Center, National Cancer CenterKashiwaJapan
| |
Collapse
|
2
|
Yu B, Kang J, Lei H, Li Z, Yang H, Zhang M. Immunotherapy for colorectal cancer. Front Immunol 2024; 15:1433315. [PMID: 39238638 PMCID: PMC11375682 DOI: 10.3389/fimmu.2024.1433315] [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: 05/16/2024] [Accepted: 07/30/2024] [Indexed: 09/07/2024] Open
Abstract
Colorectal cancer is the third most common cancer and the second most lethal cancer in the world. The main cause of the disease is due to dietary and behavioral factors. The treatment of this complex disease is mainly based on traditional treatments, including surgery, radiotherapy, and chemotherapy. Due to its high prevalence and high morbidity, more effective treatments with fewer side effects are urgently needed. In recent years, immunotherapy has become a potential therapeutic alternative and one of the fastest-developing treatments. Immunotherapy inhibits tumor growth by activating or enhancing the immune system to recognize and attack cancer cells. This review presents the latest immunotherapies for immune checkpoint inhibitors, cell therapy, tumor-infiltrating lymphocytes, and oncolytic viruses. Some of these have shown promising results in clinical trials and are used in clinical treatment.
Collapse
Affiliation(s)
- Bing Yu
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Jian Kang
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Hong Lei
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Zhe Li
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Hao Yang
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| | - Meng Zhang
- Department of the Colorectal Anal Surgery, The Affiliated Taian City Centeral Hospital of Qingdao University, Tai'an, Shandong, China
| |
Collapse
|
3
|
Lee HJ, Hwang SJ, Jeong EH, Chang MH. Genetically Engineered CLDN18.2 CAR-T Cells Expressing Synthetic PD1/CD28 Fusion Receptors Produced Using a Lentiviral Vector. J Microbiol 2024; 62:555-568. [PMID: 38700775 PMCID: PMC11303488 DOI: 10.1007/s12275-024-00133-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/18/2024] [Accepted: 03/24/2024] [Indexed: 08/07/2024]
Abstract
This study aimed to develop synthetic Claudin18.2 (CLDN18.2) chimeric antigen receptor (CAR)-T (CAR-T) cells as a treatment for advanced gastric cancer using lentiviral vector genetic engineering technology that targets the CLDN18.2 antigen and simultaneously overcomes the immunosuppressive environment caused by programmed cell death protein 1 (PD-1). Synthetic CAR T cells are a promising approach in cancer immunotherapy but face many challenges in solid tumors. One of the major problems is immunosuppression caused by PD-1. CLDN18.2, a gastric-specific membrane protein, is considered a potential therapeutic target for gastric and other cancers. In our study, CLDN18.2 CAR was a second-generation CAR with inducible T-cell costimulatory (CD278), and CLDN18.2-PD1/CD28 CAR was a third-generation CAR, wherein the synthetic PD1/CD28 chimeric-switch receptor (CSR) was added to the second-generation CAR. In vitro, we detected the secretion levels of different cytokines and the killing ability of CAR-T cells. We found that the secretion of cytokines such as interferon-gamma (IFN-γ) and tumor necrosis factor-alpha (TNF-α) secreted by three types of CAR-T cells was increased, and the killing ability against CLDN18.2-positive GC cells was enhanced. In vivo, we established a xenograft GC model and observed the antitumor effects and off-target toxicity of CAR-T cells. These results support that synthetic anti-CLDN18.2 CAR-T cells have antitumor effect and anti-CLDN18.2-PD1/CD28 CAR could provide a promising design strategy to improve the efficacy of CAR-T cells in advanced gastric cancer.
Collapse
MESH Headings
- Animals
- Humans
- Mice
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Cell Line, Tumor
- Claudins/genetics
- Claudins/metabolism
- Cytokines/metabolism
- Genetic Engineering
- Genetic Vectors/genetics
- Immunotherapy, Adoptive/methods
- Interferon-gamma/metabolism
- Lentivirus/genetics
- Programmed Cell Death 1 Receptor/genetics
- Programmed Cell Death 1 Receptor/metabolism
- Programmed Cell Death 1 Receptor/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Antigen, T-Cell/metabolism
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/immunology
- Stomach Neoplasms/therapy
- Stomach Neoplasms/immunology
- Stomach Neoplasms/genetics
- T-Lymphocytes/immunology
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Heon Ju Lee
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea.
| | - Seo Jin Hwang
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| | - Eun Hee Jeong
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| | - Mi Hee Chang
- CARBio Therapeutics Co., Ltd., Cheongju, 28160, Republic of Korea
| |
Collapse
|
4
|
Sheykhhasan M, Ahmadieh-Yazdi A, Vicidomini R, Poondla N, Tanzadehpanah H, Dirbaziyan A, Mahaki H, Manoochehri H, Kalhor N, Dama P. CAR T therapies in multiple myeloma: unleashing the future. Cancer Gene Ther 2024; 31:667-686. [PMID: 38438559 PMCID: PMC11101341 DOI: 10.1038/s41417-024-00750-2] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2023] [Revised: 02/10/2024] [Accepted: 02/16/2024] [Indexed: 03/06/2024]
Abstract
In recent years, the field of cancer treatment has witnessed remarkable breakthroughs that have revolutionized the landscape of care for cancer patients. While traditional pillars such as surgery, chemotherapy, and radiation therapy have long been available, a cutting-edge therapeutic approach called CAR T-cell therapy has emerged as a game-changer in treating multiple myeloma (MM). This novel treatment method complements options like autologous stem cell transplants and immunomodulatory medications, such as proteasome inhibitors, by utilizing protein complexes or anti-CD38 antibodies with potent complement-dependent cytotoxic effects. Despite the challenges and obstacles associated with these treatments, the recent approval of the second FDA multiple myeloma CAR T-cell therapy has sparked immense promise in the field. Thus far, the results indicate its potential as a highly effective therapeutic solution. Moreover, ongoing preclinical and clinical trials are exploring the capabilities of CAR T-cells in targeting specific antigens on myeloma cells, offering hope for patients with relapsed/refractory MM (RRMM). These advancements have shown the potential for CAR T cell-based medicines or combination therapies to elicit greater treatment responses and minimize side effects. In this context, it is crucial to delve into the history and functions of CAR T-cells while acknowledging their limitations. We can strategize and develop innovative approaches to overcome these barriers by understanding their challenges. This article aims to provide insights into the application of CAR T-cells in treating MM, shedding light on their potential, limitations, and strategies employed to enhance their efficacy.
Collapse
Affiliation(s)
- Mohsen Sheykhhasan
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
| | - Amirhossein Ahmadieh-Yazdi
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Stem Cell Biology Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi, University of Medical Sciences, Yazd, Iran
| | - Rosario Vicidomini
- Section on Cellular Communication, Eunice Kennedy Shriver National Institute of Child Health and Human Development (NICHD), National Institutes of Health (NIH), Bethesda, MD, USA
| | | | - Hamid Tanzadehpanah
- Antimicrobial Resistance Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Ashkan Dirbaziyan
- Department of Microbiology, Faculty of Medicine, AJA University of Medical Sciences, Tehran, Iran
| | - Hanie Mahaki
- Vascular & Endovascular Surgery Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Hamed Manoochehri
- The Persian Gulf Marine Biotechnology Research Center, The Persian Gulf Biomedical Sciences Research Institute, Bushehr University of Medical Sciences, Bushehr, Iran
| | - Naser Kalhor
- Department of Mesenchymal Stem Cells, Academic Center for Education, Culture and Research, Qom, Iran
| | - Paola Dama
- School of Life Sciences, University of Sussex, Brighton, BN1 9QG, UK.
| |
Collapse
|
5
|
Ma J, Song J, Yi X, Zhang S, Sun L, Huang L, Han C. Enhanced T cell immune activity mediated by Drp1 promotes the efficacy of PD-1 inhibitors in treating lung cancer. Cancer Immunol Immunother 2024; 73:40. [PMID: 38340166 PMCID: PMC10858821 DOI: 10.1007/s00262-023-03582-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 11/10/2023] [Indexed: 02/12/2024]
Abstract
BACKGROUND Dynamin-related protein 1 (Drp1)-mediated mitochondrial fission plays important roles in the activation, proliferation, and migration of T cells. METHODS We investigated the synergistic effect of Drp1-mediated T cell antitumor activities and programmed cell death protein 1 (PD-1) blockade for treating lung cancer through in vitro co-culture experiments and an in vivo nude mouse xenograft model. RESULTS High expression levels of Drp1 positively regulated T cell activation, enhanced T cell-induced suppression of lung cancer cells, promoted CD8+ T cell infiltration in the tumor and spleen, and significantly enhanced the antitumor immune response of the PD-1 inhibitor pembrolizumab. The mechanism of this synergistic antitumor effect involved the secretion of immune killing-related cytokines and the regulation of the PD-1-ERK/Drp1 pathway in T cells. CONCLUSIONS Our findings suggest that modifying Drp1 expression in T cells could serve as a potential therapeutic target for enhancing the antitumor immune response in future immunotherapies.
Collapse
Affiliation(s)
- Jietao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Jun Song
- Department of Oncology, Xiangyang Central Hospital, Affiliated Hospital of Hubei University of Arts and Science, Xiangyang, 441100, China
| | - Xiaofang Yi
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Shuling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Letian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China
| | - Chengbo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|
6
|
Liu Y, Peng C, Ahad F, Ali Zaidi SA, Muluh TA, Fu Q. Advanced Strategies of CAR-T Cell Therapy in Solid Tumors and Hematological Malignancies. Recent Pat Anticancer Drug Discov 2024; 19:557-572. [PMID: 38213150 DOI: 10.2174/0115748928277331231218115402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2023] [Revised: 10/30/2023] [Accepted: 11/10/2023] [Indexed: 01/13/2024]
Abstract
Chimeric antigen receptor T-cells, known as CAR-T cells, represent a promising breakthrough in the realm of adoptive cell therapy. These T-cells are genetically engineered to carry chimeric antigen receptors that specifically target tumors. They have achieved notable success in the treatment of blood-related cancers, breathing new life into this field of medical research. However, numerous obstacles limit chimeric antigen receptors T-cell therapy's efficacy, such as it cannot survive in the body long. It is prone to fatigue and exhaustion, leading to difficult tumor elimination and repeated recurrence, affecting solid tumors and hematological malignancies. The challenges posed by solid tumors, especially in the context of the complex solid-tumor microenvironment, require specific strategies. This review outlines recent advancements in improving chimeric antigen receptors T-cell therapy by focusing on the chimeric antigen receptors protein, modifying T-cells, and optimizing the interaction between T-cells and other components within the tumor microenvironment. This article aims to provide an extensive summary of the latest discoveries regarding CAR-T cell therapy, encompassing its application across various types of human cancers. Moreover, it will delve into the obstacles that have emerged in recent times, offering insights into the challenges faced by this innovative approach. Finally, it highlights novel therapeutic options in treating hematological and solid malignancies with chimeric antigen receptors T-cell therapies.
Collapse
Affiliation(s)
- Yangjie Liu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan, PRC China
| | - Cao Peng
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| | - Faiza Ahad
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Syed Aqib Ali Zaidi
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Tobias Achu Muluh
- Shenzhen University Medical School, Shenzhen University, Shenzhen 518060, China
| | - Qiuxia Fu
- Department of Pharmacy, Luzhou People's Hospital, Luzhou 646000, Sichuan PRC China
| |
Collapse
|
7
|
Drougkas K, Karampinos K, Karavolias I, Koumprentziotis IA, Ploumaki I, Triantafyllou E, Trontzas I, Kotteas E. Comprehensive clinical evaluation of CAR-T cell immunotherapy for solid tumors: a path moving forward or a dead end? J Cancer Res Clin Oncol 2023; 149:2709-2734. [PMID: 36564524 PMCID: PMC10129996 DOI: 10.1007/s00432-022-04547-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Accepted: 12/16/2022] [Indexed: 12/25/2022]
Abstract
INTRODUCTION Chimeric Antigen Receptor (CAR)-T cell therapy is a form of adoptive cell therapy that has demonstrated tremendous results in the treatment of hematopoietic malignancies, leading to the US Food and Drug Administration (FDA) approval of four CD19-targeted CAR-T cell products. With the unprecedented success of CAR-T cell therapy in hematological malignancies, hundreds of preclinical studies and clinical trials are currently undergoing to explore the translation of this treatment to solid tumors. However, the clinical experience in non-hematologic malignancies has been less encouraging, with only a few patients achieving complete responses. Tumor-associated antigen heterogeneity, inefficient CAR-T cell trafficking and the immunosuppressive tumor microenvironment are considered as the most pivotal roadblocks in solid tumor CAR-T cell therapy. MATERIALS AND METHODS We reviewed the relevant literature/clinical trials for CAR-T cell immunotherapy for solid tumors from Pubmed and ClinicalTrials.gov. CONCLUSION Herein, we provide an update on solid tumor CAR-T cell clinical trials, focusing on the studies with published results. We further discuss some of the key hurdles that CAR-T cell therapy is encountering for solid tumor treatment as well as the strategies that are exploited to overcome these obstacles.
Collapse
Affiliation(s)
- Konstantinos Drougkas
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece.
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece.
| | - Konstantinos Karampinos
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioannis Karavolias
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioannis-Alexios Koumprentziotis
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioanna Ploumaki
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Efthymios Triantafyllou
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Medicine, School of Medicine, National and Kapodistrian University of Athens, Athens, Attica, Greece
| | - Ioannis Trontzas
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
- Department of Pathology, Yale University School of Medicine, New Haven, USA, CT
| | - Elias Kotteas
- Oncology Unit, Sotiria General Hospital, National and Kapodistrian University of Athens, 152 Mesogeion Avenue, 11527, Athens, Greece
| |
Collapse
|
8
|
Wang AX, Ong XJ, D’Souza C, Neeson PJ, Zhu JJ. Combining chemotherapy with CAR-T cell therapy in treating solid tumors. Front Immunol 2023; 14:1140541. [PMID: 36949946 PMCID: PMC10026332 DOI: 10.3389/fimmu.2023.1140541] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Accepted: 02/22/2023] [Indexed: 03/08/2023] Open
Abstract
Chemotherapy has long been a standard treatment for a wide range of malignancies, where patients typically undergo multiple rounds of chemotherapy regimens to control tumor growth. In the clinic, the chemotherapy drugs cyclophosphamide and fludarabine are commonly used prior to Chimeric Antigen Receptor T (CAR-T) cell therapy to lymphodeplete and improve CAR-T cell engraftment. In this review, we discuss the use of chemotherapy in combination with CAR-T cell therapy. We also show that chemotherapy can deplete immunosuppressive cells, promote a pro-inflammatory tumor microenvironment, disrupt tumor stroma, and improve CAR-T cell recruitment to the tumor. Although the combination of chemotherapy plus CAR-T cell therapy is promising, certain aspects of chemotherapy also pose a challenge. In addition, the combined therapeutic effect may be heavily dependent on the dose and the treatment schedule. Thus, we also discussed the obstacles to effective clinical outcomes of the combination therapy.
Collapse
Affiliation(s)
- Arthur Xuan Wang
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Xiao Jing Ong
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Criselle D’Souza
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, VIC, Australia
| | - Paul J. Neeson
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, VIC, Australia
| | - Joe Jiang Zhu
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
- Sir Peter MacCallum Department of Oncology, Faculty of Medicine, Dentistry and Health Science, University of Melbourne, Melbourne, VIC, Australia
- *Correspondence: Joe Jiang Zhu,
| |
Collapse
|
9
|
Dubois VP, Sehl OC, Foster PJ, Ronald JA. Visualizing CAR-T cell Immunotherapy Using 3 Tesla Fluorine-19 MRI. Mol Imaging Biol 2022; 24:298-308. [PMID: 34786668 PMCID: PMC8983548 DOI: 10.1007/s11307-021-01672-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 09/02/2021] [Accepted: 10/20/2021] [Indexed: 01/19/2023]
Abstract
PURPOSE Chimeric antigen receptor (CAR) T cell cancer immunotherapies have shown remarkable results in patients with hematological malignancies and represent the first approved genetically modified cellular therapies. However, not all blood cancer patients respond favorably, serious side effects have been reported, and the treatment of solid tumors has been a challenge. An imaging tool for visualizing the variety of CAR-T cell products in use and being explored could provide important patient-specific data on CAR-T cell location to inform on potential success or failure of treatment as well as off-target toxicities. Fluorine-19 (19F) magnetic resonance imaging (MRI) allows for the noninvasive detection of 19F perfluorocarbon (PFC) labeled cells. Our objective was to visualize PFC-labeled (PFC +) CAR-T cells in a mouse model of leukemia using clinical field strength (3 Tesla) 19F MRI and compare the cytotoxicity of PFC + versus unlabeled CAR-T cells. PROCEDURES NSG mice (n = 17) received subcutaneous injections of CD19 + human B cell leukemia cells (NALM6) expressing firefly luciferase in their left hind flank (1 × 106). Twenty-one days later, each mouse received an intratumoral injection of 10 × 106 PFC + CD19-targeted CAR-T cells (n = 6), unlabeled CD19-targeted CAR-T cells (n = 3), PFC + untransduced T cells (n = 5), or an equivalent volume of saline (n = 3). 19F MRI was performed on mice treated with PFC + CAR-T cells days 1, 3, and 7 post-treatment. Bioluminescence imaging (BLI) was performed on all mice days - 1, 5, 10, and 14 post-treatment to monitor tumor response. RESULTS PFC + CAR-T cells were successfully detected in tumors using 19F MRI on days 1, 3, and 7 post-injection. In vivo BLI data revealed that mice treated with PFC + or PFC - CAR-T cells had significantly lower tumor burden by day 14 compared to untreated mice and mice treated with PFC + untransduced T cells (p < 0.05). Importantly, mice treated with PFC + CAR-T cells showed equivalent cytotoxicity compared to mice receiving PFC - CAR-T cells. CONCLUSIONS Our studies demonstrate that clinical field strength 19F MRI can be used to visualize PFC + CAR-T cells for up to 7 days post-intratumoral injection. Importantly, PFC labeling did not significantly affect in vivo CAR-T cell cytotoxicity. These imaging tools may have broad applications for tracking emerging CAR-T cell therapies in preclinical models and may eventually be useful for the detection of CAR-T cells in patients where localized injection of CAR-T cells is being pursued.
Collapse
Affiliation(s)
- Veronica P Dubois
- Robarts Research Institute, London, ON, Canada
- The Department of Medical Biophysics, Western University, London, ON, Canada
| | - Olivia C Sehl
- Robarts Research Institute, London, ON, Canada
- The Department of Medical Biophysics, Western University, London, ON, Canada
| | - Paula J Foster
- Robarts Research Institute, London, ON, Canada
- The Department of Medical Biophysics, Western University, London, ON, Canada
| | - John A Ronald
- Robarts Research Institute, London, ON, Canada.
- The Department of Medical Biophysics, Western University, London, ON, Canada.
- Lawson Health Research Institute, London, ON, Canada.
| |
Collapse
|
10
|
Li M, Li Y, Li S, Jia L, Du C, Li M, Li S, Galons H, Guo N, Yu P. Co-delivery of F7 and crizotinib by thermosensitive liposome for breast cancer treatment. J Liposome Res 2021; 32:265-275. [PMID: 34904521 DOI: 10.1080/08982104.2021.2001499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
In order to enhance the targeting efficiency and reduce the side effects and drug resistance, crizotinib (Cri) and F7 were co-loaded in a thermosensitive liposome (TSL) (F7-Cri-TSL), which showed enhanced permeability and retention in breast cancer model, as well as local controlled release by external hyperthermia. Cri is an inhibitor for cell proliferation and a promoter of apoptosis, by inhibiting the phosphorylation of intracellular ALK and c-Met, but its drug resistance limits its application. F7 is a novel drug candidate with significant resistance to cyclin-dependent kinase, but its use was restricted by its high toxicity. The F7-Cri-TSL was found with excellent particle size (about 108 nm), high entrapment efficiency (>95%), significant thermosensitive property, and good stability. Furthermore, F7-Cri-TSL/H had strongest cell lethality compared with other formulations. On the MCF-7 xenograft mice model, the F7-Cri-TSL also exhibited therapeutic synergism of Cri, F7 and hyperthermia. Meanwhile, it was shown that the TSL reduced the systemic toxicity of the chemotherapy drug. Therefore, the F7-Cri-TSL may serve as a promising system for temperature triggered breast cancer treatment.
Collapse
Affiliation(s)
- Mingyuan Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Yuan Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Shiqin Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Lin Jia
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Chunyang Du
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Meng Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Shuangshuang Li
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Hervé Galons
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China.,Institut Parisien de Chimie Moléculaire, UMR CNRS 8232, Paris, France
| | - Na Guo
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| | - Peng Yu
- China International Science and Technology, Cooperation Base of Food Nutrition/Safety and Medicinal Chemistry, Tianjin International Cooperation Research Centre of Food Nutrition/Safety and Medicinal Chemistry, College of Biotechnology, Tianjin University of Science and Technology/Tianjin Enterprise Key Laboratory for Application Research of Hyaluronic Acid, Tianjin, China
| |
Collapse
|
11
|
Wang J, Zhang Y, Pi J, Xing D, Wang C. Localized delivery of immunotherapeutics: A rising trend in the field. J Control Release 2021; 340:149-167. [PMID: 34699871 DOI: 10.1016/j.jconrel.2021.10.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 10/11/2021] [Indexed: 02/08/2023]
Abstract
Immunotherapy is becoming a new standard of care for multiple cancers, while several limitations are impending its further clinical success. Immunotherapeutic agents often have inappropriate pharmacokinetics on their own and/or exhibit limited specificity to tumor cells, leading to severe immuno-related adverse effects and limited efficacy. Suitable formulating strategies that confer prolonged contact with or efficient proliferation in tumors while reducing exposure to normal tissues are highly worthy to explore. With the assistance of biomaterial carriers, targeted therapy can be achieved artificially by implanting or injecting drug depots into desired sites, about which the wisdoms in literature have been rich. The relevant results have suggested a "local but systemic" effect, that is, local replenishment of immune modulators achieves a high treatment efficacy that also governs distant metastases, thereby building another rationale for localized delivery. Particularly, implantable scaffolds have been further engineered to recruit disseminated tumor cells with an efficiency high enough to reduce tumor burdens at typical metastatic organs, and simultaneously provide diagnostic signals. This review introduces recent advances in this emerging area along with a perspective on the opportunities and challenges in the way to clinical application.
Collapse
Affiliation(s)
- Jie Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| | - Yukun Zhang
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Jiuchan Pi
- Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| | - Chao Wang
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Qingdao Cancer Institute, Qingdao University, Qingdao 266071, China.
| |
Collapse
|
12
|
Globerson Levin A, Rivière I, Eshhar Z, Sadelain M. CAR T cells: Building on the CD19 paradigm. Eur J Immunol 2021; 51:2151-2163. [PMID: 34196410 DOI: 10.1002/eji.202049064] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Accepted: 06/28/2021] [Indexed: 12/11/2022]
Abstract
Spearheaded by the therapeutic use of chimeric antigen receptors (CARs) targeting CD19, synthetic immunology has entered the clinical arena. CARs are recombinant receptors for antigen that engage cell surface molecules through the variable region of an antibody and signal through arrayed T-cell activating and costimulatory domains. CARs allow redirection of T-cell cytotoxicity against any antigen of choice, independent of MHC expression. Patient T cells engineered to express CARs specific for CD19 have yielded remarkable outcomes in subjects with relapsed/refractory B- cell malignancies, setting off unprecedented interest in T-cell engineering and cell-based cancer immunotherapy. In this review, we present the challenges to extend the use of CAR T cells to solid tumors and other pathologies. We further highlight progress in CAR design, cell manufacturing, and genome editing, which in aggregate hold the promise of generating safer and more effective genetically instructed immunity. Novel engineered cell types, including innate T-cell types, natural killer (NK) cells, macrophages, and induced pluripotent stem cell-derived immune cells, are on the horizon, as are applications of CAR T cells to treat autoimmunity, severe infections, and senescence-associated pathologies.
Collapse
Affiliation(s)
| | - Isabelle Rivière
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zelig Eshhar
- Immunology Lab, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| | - Michel Sadelain
- Center for Cell Engineering, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| |
Collapse
|
13
|
Marofi F, Motavalli R, Safonov VA, Thangavelu L, Yumashev AV, Alexander M, Shomali N, Chartrand MS, Pathak Y, Jarahian M, Izadi S, Hassanzadeh A, Shirafkan N, Tahmasebi S, Khiavi FM. CAR T cells in solid tumors: challenges and opportunities. Stem Cell Res Ther 2021; 12:81. [PMID: 33494834 PMCID: PMC7831265 DOI: 10.1186/s13287-020-02128-1] [Citation(s) in RCA: 347] [Impact Index Per Article: 86.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 12/28/2020] [Indexed: 01/11/2023] Open
Abstract
BACKGROUND CARs are simulated receptors containing an extracellular single-chain variable fragment (scFv), a transmembrane domain, as well as an intracellular region of immunoreceptor tyrosine-based activation motifs (ITAMs) in association with a co-stimulatory signal. MAIN BODY Chimeric antigen receptor (CAR) T cells are genetically engineered T cells to express a receptor for the recognition of the particular surface marker that has given rise to advances in the treatment of blood disorders. The CAR T cells obtain supra-physiological properties and conduct as "living drugs" presenting both immediate and steady effects after expression in T cells surface. But, their efficacy in solid tumor treatment has not yet been supported. The pivotal challenges in the field of solid tumor CAR T cell therapy can be summarized in three major parts: recognition, trafficking, and surviving in the tumor. On the other hand, the immunosuppressive tumor microenvironment (TME) interferes with T cell activity in terms of differentiation and exhaustion, and as a result of the combined use of CARs and checkpoint blockade, as well as the suppression of other inhibitor factors in the microenvironment, very promising results were obtained from the reduction of T cell exhaustion. CONCLUSION Nowadays, identifying and defeating the mechanisms associated with CAR T cell dysfunction is crucial to establish CAR T cells that can proliferate and lyse tumor cells severely. In this review, we discuss the CAR signaling and efficacy T in solid tumors and evaluate the most significant barriers in this process and describe the most novel therapeutic methods aiming to the acquirement of the promising therapeutic outcome in non-hematologic malignancies.
Collapse
Affiliation(s)
- Faroogh Marofi
- Department of Hematology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Roza Motavalli
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
- Kidney Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Vladimir A. Safonov
- The Laboratory of Biogeochemistry and Environment, Vernadsky Institute of Geochemistry and Analytical Chemistry of Russian Academy of Sciences, Kosygina 19 Street, Moscow, Russian Federation 119991
| | - Lakshmi Thangavelu
- Department of Pharmacology, Saveetha Dental College and Hospital, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India
| | | | - Markov Alexander
- Tyumen State Medical University, Tyumen Industrial University, Tyumen, Russian Federation
| | - Navid Shomali
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120 Heidelberg, Germany
| | | | - Yashwant Pathak
- Taneja College of Pharmacy, University of South Florida, Tampa, FL USA
| | - Mostafa Jarahian
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Sepideh Izadi
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120 Heidelberg, Germany
| | - Ali Hassanzadeh
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120 Heidelberg, Germany
| | - Naghmeh Shirafkan
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120 Heidelberg, Germany
| | - Safa Tahmasebi
- Toxicology and Chemotherapy Unit (G401), German Cancer Research Center, 69120 Heidelberg, Germany
| | | |
Collapse
|
14
|
Ciccocioppo R, Comoli P, Astori G, Del Bufalo F, Prapa M, Dominici M, Locatelli F. Developing cell therapies as drug products. Br J Pharmacol 2020; 178:262-279. [PMID: 33140850 DOI: 10.1111/bph.15305] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/22/2020] [Accepted: 10/23/2020] [Indexed: 02/06/2023] Open
Abstract
In the last 20 years, the global regulatory frameworks for drug assessment have been managing the challenges posed by using cellular products as new therapeutic tools. Currently, they are defined as "Advanced Therapy Medicinal Products", comprising a large group of cellular types that either alone or in combination with gene and tissue engineering technology. They have the potential to change the natural course of still lethal or highly debilitating diseases, including cancers, opportunistic infections and chronic inflammatory conditions. Globally, more than 50 cell-based products have obtained market authorization. This overview describes the advantages and unsolved challenges on developing cells as innovative therapeutic vehicles. The main cell therapy players and the legal framework are discussed, starting from chimeric antigen receptor T-cells for leukaemia and solid tumours, dealing then with lymphocytes as potent anti-microbiological tools and then focusing on mesenchymal stem/stromal cells whose role covers regenerative medicine, immunology and anti-tumour therapy.
Collapse
Affiliation(s)
- Rachele Ciccocioppo
- Gastroenterology Unit, Department of Medicine, A.O.U.I. Policlinico G.B. Rossi & University of Verona, Verona, Italy
| | - Patrizia Comoli
- Cell Factory and Paediatric Haematology/Oncology Unit, Fondazione I.R.C.C.S. Policlinico San Matteo, Pavia, Italy
| | - Giuseppe Astori
- Laboratory of Advanced Cellular Therapies, Haematology Unit, San Bortolo Hospital, A.U.L.S.S. 8 "Berica", Vicenza, Italy
| | - Francesca Del Bufalo
- Department of Paediatric Haematology and Oncology and Cell and Gene Therapy, I.R.C.C.S. Bambino Gesù Children's Hospital, Rome, Italy
| | - Malvina Prapa
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Franco Locatelli
- Department of Paediatric Haematology and Oncology and Cell and Gene Therapy, I.R.C.C.S. Bambino Gesù Children's Hospital, Rome, Italy.,Department of Paediatrics, Sapienza University of Rome, Rome, Italy
| |
Collapse
|