1
|
Li Q, Yang L, Li S, Zhao W, Xue Y, Lu Z, Tang J, Gao X, Zheng J, Zhang Q, Sun S. Cabozantinib enhances CAIX specific CAR-T cells against renal cancer by improving effector functions and augmenting tumor immune microenvironment. Biochem Biophys Res Commun 2024; 734:150781. [PMID: 39368372 DOI: 10.1016/j.bbrc.2024.150781] [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: 05/29/2024] [Revised: 09/23/2024] [Accepted: 09/30/2024] [Indexed: 10/07/2024]
Abstract
Despite demonstrating promising outcomes in treating hematologic malignancies, the efficacy of chimeric antigen receptor-modified T (CAR-T) cell therapy remains limited when applied to solid tumors due to tumor immune microenvironment (TIME). Strategies to augment CAR-T cell efficacy against solid tumors have been investigated by ameliorating TIME to a certain extent. In this study, Cabozantinib was utilized in combination with CAR-T cells targeting carbonic anhydrase IX (CAIX) for the treatment of renal cancer. Our findings indicate that combination therapy with CAIX-CAR-T and Cabozantinib demonstrated synergistic efficacy against an orthotopic xenograft tumor model and a subcutaneous tumor model of renal cell carcinoma in mice. Mechanistically, it was observed that CAR-T cells combined with Cabozantinib led to an increase in the infiltration of tumor-infiltrating T cells, while reducing tumor-associated macrophages and M2 polarization. Additionally, Cabozantinib blocked the programmed cell death-1 (PD-1)/programmed death-ligand 1 (PD-L1) axis by decreasing the expression of PD-L1 in tumor cells and PD-1 in T cells. Furthermore, Cabozantinib promoted CAR-T cell effector function and reduced T cell exhaustion. This combination therapy represents a novel approach to enhancing CAR-T cell efficacy against solid tumors and holds significant promise for advancing CAR-T cell therapy in clinical settings.
Collapse
Affiliation(s)
- Qihong Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Lin Yang
- General Surgery Department, Xuzhou Mine Hospital, Xuzhou, Jiangsu, PR China
| | - Shuyu Li
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Wanxin Zhao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Ying Xue
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Zhuyu Lu
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Jingwei Tang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Xiaoge Gao
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China
| | - Qing Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| | - Shishuo Sun
- Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, PR China; Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, Jiangsu, PR China.
| |
Collapse
|
2
|
Malik S, Sureka N, Ahuja S, Aden D, Zaheer S, Zaheer S. Tumor-associated macrophages: A sentinel of innate immune system in tumor microenvironment gone haywire. Cell Biol Int 2024; 48:1406-1449. [PMID: 39054741 DOI: 10.1002/cbin.12226] [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: 12/11/2023] [Revised: 06/10/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
The tumor microenvironment (TME) is a critical determinant in the initiation, progression, and treatment outcomes of various cancers. Comprising of cancer-associated fibroblasts (CAF), immune cells, blood vessels, and signaling molecules, the TME is often likened to the soil supporting the seed (tumor). Among its constituents, tumor-associated macrophages (TAMs) play a pivotal role, exhibiting a dual nature as both promoters and inhibitors of tumor growth. This review explores the intricate relationship between TAMs and the TME, emphasizing their diverse functions, from phagocytosis and tissue repair to modulating immune responses. The plasticity of TAMs is highlighted, showcasing their ability to adopt either protumorigenic or anti-tumorigenic phenotypes based on environmental cues. In the context of cancer, TAMs' pro-tumorigenic activities include promoting angiogenesis, inhibiting immune responses, and fostering metastasis. The manuscript delves into therapeutic strategies targeting TAMs, emphasizing the challenges faced in depleting or inhibiting TAMs due to their multifaceted roles. The focus shifts towards reprogramming TAMs to an anti-tumorigenic M1-like phenotype, exploring interventions such as interferons, immune checkpoint inhibitors, and small molecule modulators. Noteworthy advancements include the use of CSF1R inhibitors, CD40 agonists, and CD47 blockade, demonstrating promising results in preclinical and clinical settings. A significant section is dedicated to Chimeric Antigen Receptor (CAR) technology in macrophages (CAR-M cells). While CAR-T cells have shown success in hematological malignancies, their efficacy in solid tumors has been limited. CAR-M cells, engineered to infiltrate solid tumors, are presented as a potential breakthrough, with a focus on their development, challenges, and promising outcomes. The manuscript concludes with the exploration of third-generation CAR-M technology, offering insight into in-vivo reprogramming and nonviral vector approaches. In conclusion, understanding the complex and dynamic role of TAMs in cancer is crucial for developing effective therapeutic strategies. While early-stage TAM-targeted therapies show promise, further extensive research and larger clinical trials are warranted to optimize their targeting and improve overall cancer treatment outcomes.
Collapse
Affiliation(s)
- Shaivy Malik
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Sana Ahuja
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| | - Durre Aden
- Department of Pathology, Hamdard Institute of Medical Science and Research, Jamia Hamdard, New Delhi, New Delhi, India
| | - Samreen Zaheer
- Department of Radiotherapy, Jawaharlal Nehru Medical College, AMU, Aligarh, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, New Delhi, India
| |
Collapse
|
3
|
Holtermann A, Gislon M, Angele M, Subklewe M, von Bergwelt-Baildon M, Lauber K, Kobold S. Prospects of Synergy: Local Interventions and CAR T Cell Therapy in Solid Tumors. BioDrugs 2024; 38:611-637. [PMID: 39080180 PMCID: PMC11358237 DOI: 10.1007/s40259-024-00669-y] [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] [Accepted: 06/17/2024] [Indexed: 08/30/2024]
Abstract
Chimeric antigen receptor T cell therapy has been established in the treatment of various B cell malignancies. However, translating this therapeutic effect to treat solid tumors has been challenging because of their inter-tumoral as well as intratumoral heterogeneity and immunosuppressive microenvironment. Local interventions, such as surgery, radiotherapy, local ablation, and locoregional drug delivery, can enhance chimeric antigen receptor T cell therapy in solid tumors by improving tumor infiltration and reducing systemic toxicities. Additionally, ablation and radiotherapy have proven to (re-)activate systemic immune responses via abscopal effects and reprogram the tumor microenvironment on a physical, cellular, and chemical level. This review highlights the potential synergy of the combined approaches to overcome barriers of chimeric antigen receptor T cell therapy and summarizes recent studies that may pave the way for new treatment regimens.
Collapse
Affiliation(s)
- Anne Holtermann
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Mila Gislon
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany
| | - Martin Angele
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, Germany
| | - Marion Subklewe
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Michael von Bergwelt-Baildon
- Department of Medicine III, University Hospital, Ludwig Maximilian University (LMU) of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany
| | - Kirsten Lauber
- Department of Radiation Oncology, LMU University Hospital, LMU Munich, Munich, Germany
| | - Sebastian Kobold
- Division of Clinical Pharmacology, Department of Medicine IV, University Hospital, Ludwig Maximilian University (LMU) of Munich, Lindwurmstrasse 2a, 80336, Munich, Germany.
- German Cancer Consortium (DKTK), Partner Site Munich, a partnership between the DKFZ Heidelberg and the University Hospital of the LMU, Munich, Germany.
- Einheit für Klinische Pharmakologie (EKLiP), Helmholtz Zentrum München-German Research Center for Environmental Health Neuherberg, Munich, Germany.
| |
Collapse
|
4
|
Sarkar R, Adhikary K, Banerjee A, Ganguly K, Sarkar R, Mohanty S, Dhua R, Bhattacharya K, Ahuja D, Pal S, Maiti R. Novel targets and improved immunotherapeutic techniques with an emphasis on antimycosal drug resistance for the treatment and management of mycosis. Heliyon 2024; 10:e35835. [PMID: 39224344 PMCID: PMC11367498 DOI: 10.1016/j.heliyon.2024.e35835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/04/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Infections due to pathogenic fungi are endemic in particular area with increased morbidity and mortality. More than a thousand people are infected per year and the way of treatment is of high demand having a significant impact on the population health. Medical practitioners confront various troublesome analytic and therapeutical challenges in the administration of immunosuppressed sufferer at high danger of expanding fungal infections. An upgraded antimycosal treatment is fundamental for a fruitful result while treating intrusive mycoses. A collection of antimycosal drugs keeps on developing with their specific antifungal targets including cell membrane, mitochondria, cell wall, and deoxyribonucleic acid (DNA)/ribonucleic acid (RNA) or protein biosynthesis. Some fundamental classes of ordinarily directed medications are the polyenes, amphotericin B, syringomycin, allylamines, honokiol, azoles, flucytosine, echinocandins etc. However, few immunotherapy processes and vaccinations are being developed to mark this need, although one presently can't seem to arrive at the conclusion. In this review article, there has been a trial to give details upgradation about the current immune therapeutic techniques and vaccination strategies against prevention or treatment of mycosis as well as the difficulties related with their turn of events. There has been also a visualization in the mentioned review paper about the various assorted drugs and their specific target analysis along with therapeutic interventions.
Collapse
Affiliation(s)
- Riya Sarkar
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Adhikary
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Arundhati Banerjee
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Krishnendu Ganguly
- Department of Medical Lab Technology and Biotechnology, Paramedical College Durgapur, West Bengal, 713212, India
| | - Riya Sarkar
- Department of Medical Laboratory Technology, Dr. B. C. Roy Academy of Professional Courses, Durgapur, West Bengal, 713206, India
| | - Satyajit Mohanty
- Department of Advanced Pharmacology, Birla Institute of Technology, Mesra, Ranchi, Jharkhand, 835215, India
| | - Rumpa Dhua
- Department of Nutrition, Bankura Sammilani College, Kenduadihi, Bankura, West Bengal, 722102, India
| | - Koushik Bhattacharya
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Deepika Ahuja
- School of Paramedics and Allied Health Sciences, Centurion University of Technology and Management, Jatni, Bhubaneswar, Odisha, 752050, India
| | - Suchandra Pal
- Department of Biotechnology, National Institute of Technology, Durgapur, West Bengal, 713209, India
| | - Rajkumar Maiti
- Department of Physiology, Bankura Christian College, Bankura, West Bengal, 722101, India
| |
Collapse
|
5
|
Garlisi B, Lauks S, Aitken C, Ogilvie LM, Lockington C, Petrik D, Eichhorn JS, Petrik J. The Complex Tumor Microenvironment in Ovarian Cancer: Therapeutic Challenges and Opportunities. Curr Oncol 2024; 31:3826-3844. [PMID: 39057155 PMCID: PMC11275383 DOI: 10.3390/curroncol31070283] [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: 05/15/2024] [Revised: 06/27/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
The tumor microenvironment (TME) in ovarian cancer (OC) has much greater complexity than previously understood. In response to aggressive pro-angiogenic stimulus, blood vessels form rapidly and are dysfunctional, resulting in poor perfusion, tissue hypoxia, and leakiness, which leads to increased interstitial fluid pressure (IFP). Decreased perfusion and high IFP significantly inhibit the uptake of therapies into the tumor. Within the TME, there are numerous inhibitor cells, such as myeloid-derived suppressor cells (MDSCs), tumor association macrophages (TAMs), regulatory T cells (Tregs), and cancer-associated fibroblasts (CAFs) that secrete high numbers of immunosuppressive cytokines. This immunosuppressive environment is thought to contribute to the lack of success of immunotherapies such as immune checkpoint inhibitor (ICI) treatment. This review discusses the components of the TME in OC, how these characteristics impede therapeutic efficacy, and some strategies to alleviate this inhibition.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Jim Petrik
- Department of Biomedical Sciences, University of Guelph, Guelph, ON N1G 2W1, Canada; (B.G.); (S.L.); (C.A.); (L.M.O.); (C.L.); (D.P.); (J.S.E.)
| |
Collapse
|
6
|
Utkarsh K, Srivastava N, Kumar S, Khan A, Dagar G, Kumar M, Singh M, Haque S. CAR-T cell therapy: a game-changer in cancer treatment and beyond. Clin Transl Oncol 2024; 26:1300-1318. [PMID: 38244129 DOI: 10.1007/s12094-023-03368-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Accepted: 12/04/2023] [Indexed: 01/22/2024]
Abstract
In recent years, cancer has become one of the primary causes of mortality, approximately 10 million deaths worldwide each year. The most advanced, chimeric antigen receptor (CAR) T cell immunotherapy has turned out as a promising treatment for cancer. CAR-T cell therapy involves the genetic modification of T cells obtained from the patient's blood, and infusion back to the patients. CAR-T cell immunotherapy has led to a significant improvement in the remission rates of hematological cancers. CAR-T cell therapy presently limited to hematological cancers, there are ongoing efforts to develop additional CAR constructs such as bispecific CAR, tandem CAR, inhibitory CAR, combined antigens, CRISPR gene-editing, and nanoparticle delivery. With these advancements, CAR-T cell therapy holds promise concerning potential to improve upon traditional cancer treatments such as chemotherapy and radiation while reducing associated toxicities. This review covers recent advances and advantages of CAR-T cell immunotherapy.
Collapse
Affiliation(s)
- Kumar Utkarsh
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Namita Srivastava
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Sachin Kumar
- Department of Microbiology and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Azhar Khan
- Faculty of Applied Science and Biotechnology, Shoolini University, Solan, Himachal Pradesh, 173229, India
| | - Gunjan Dagar
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Mukesh Kumar
- Department of Medical Oncology, Thomas Jefferson University, Philadelphia, PA, 19107, USA
| | - Mayank Singh
- Department of Medical Oncology, All India Institute of Medical Sciences, New Delhi, India
| | - Shabirul Haque
- Department of Autoimmune Diseases, Feinstein Institute for Medical Research, Northwell Health, 350, Community Drive, Manhasset, NY, 11030, USA.
| |
Collapse
|
7
|
Zhang G, Wang Y, Lu S, Ding F, Wang X, Zhu C, Wang Y, Wang K. Molecular understanding and clinical outcomes of CAR T cell therapy in the treatment of urological tumors. Cell Death Dis 2024; 15:359. [PMID: 38789450 PMCID: PMC11126652 DOI: 10.1038/s41419-024-06734-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 05/26/2024]
Abstract
Chimeric antigen receptor engineered T (CAR T) cell therapy has developed rapidly in recent years, leading to profound developments in oncology, especially for hematologic malignancies. However, given the pressure of immunosuppressive tumor microenvironments, antigen escape, and diverse other factors, its application in solid tumors is less developed. Urinary system tumors are relatively common, accounting for approximately 24% of all new cancers in the United States. CAR T cells have great potential for urinary system tumors. This review summarizes the latest developments of CAR T cell therapy in urinary system tumors, including kidney cancer, bladder cancer, and prostate cancer, and also outlines the various CAR T cell generations and their pathways and targets that have been developed thus far. Finally, the current advantages, problems, and side effects of CAR T cell therapy are discussed in depth, and potential future developments are proposed in view of current shortcomings.
Collapse
Affiliation(s)
- Gong Zhang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yuan Wang
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shiyang Lu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Fengzhu Ding
- Department of Nursing, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xia Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Chunming Zhu
- Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Yibing Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Kefeng Wang
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
8
|
Huang P, Zhang X, Prabhu JS, Pandey V. Therapeutic vulnerabilities in triple negative breast cancer: Stem-like traits explored within molecular classification. Biomed Pharmacother 2024; 174:116584. [PMID: 38613998 DOI: 10.1016/j.biopha.2024.116584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/05/2024] [Accepted: 04/10/2024] [Indexed: 04/15/2024] Open
Abstract
Triple Negative Breast Cancer (TNBC) is the most aggressive type of breast cancer (BC). Despite advances in the clinical management of TNBC, recurrence-related mortality remains a challenge. The stem-like phenotype of TNBC plays a significant role in the persistence of minimal disease residue after therapy. Individuals exhibiting stem-like characteristics are particularly prone to inducing malignant relapse accompanied by strong resistance. Therefore, stem-like traits have been broadly proposed as therapeutic vulnerabilities to treat TNBC and reduce recurrence. However, heterogeneity within TNBC often generally restricts the stability of the therapeutic efficacy. To understand the heterogeneity and manage TNBC more precisely, multiple TNBC subtyping categories have been reported, providing the basis for profile-according therapeutic regimens. To provide more insight into targeting stem-like traits to ablate TNBC and reduce recurrence in the context of heterogeneity, this paper reviewed the molecular subtyping of TNBC, identified the consensus subtypes with distinct stem-like phenotypes, characterized the stemness hierarchy of TNBC, outlined the biological models for stem-like TNBC subtypes, summarized the therapeutic vulnerabilities in stem-like traits of the subtypes, and proposed potential therapeutic regimens targeting stem-like characteristics to improve TNBC prognosis.
Collapse
Affiliation(s)
- Peng Huang
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xi Zhang
- Shenzhen Bay Laboratory, Shenzhen 518055, China
| | - Jyothi S Prabhu
- Division of Molecular Medicine, St. John's Research Institute, St. John's Medical College, Bangalore, India
| | - Vijay Pandey
- Tsinghua Berkeley Shenzhen Institute, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China; Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China.
| |
Collapse
|
9
|
Shumnalieva R, Velikova T, Monov S. Expanding the role of CAR T-cell therapy: From B-cell hematological malignancies to autoimmune rheumatic diseases. Int J Rheum Dis 2024; 27:e15182. [PMID: 38742463 DOI: 10.1111/1756-185x.15182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 04/04/2024] [Accepted: 04/28/2024] [Indexed: 05/16/2024]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a form of immunotherapy where the lymphocytes, mostly T-cells, are redirected to specifically recognize and eliminate a target antigen by coupling them with CARs. The binding of CAR and target cell surface antigens leads to vigorous T cell activation and robust anti-tumor immune responses. Areas of implication of CAR T-cell therapies include mainly hematological malignancies (i.e., advanced B-cell cancers); however, recent studies have proven the unprecedented success of the new immunotherapy also in autoimmune rheumatic diseases. We aim to review the recent advances in CAR T-cell therapies in rheumatology but also to address the limitations of their use in the real clinical practice based on the data on their efficacy and safety.
Collapse
Affiliation(s)
- Russka Shumnalieva
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| | - Tsvetelina Velikova
- Medical Faculty, Sofia University "St. Kliment Ohridski"- Sofia, Sofia, Bulgaria
| | - Simeon Monov
- Department of Rheumatology, Clinic of Rheumatology, Medical University-Sofia, Faculty of Medicine, Sofia, Bulgaria
| |
Collapse
|
10
|
Capella MP, Esfahani K. A Review of Practice-Changing Therapies in Oncology in the Era of Personalized Medicine. Curr Oncol 2024; 31:1913-1919. [PMID: 38668046 PMCID: PMC11049499 DOI: 10.3390/curroncol31040143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Revised: 02/17/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
In the past decade, a lot of insight was gathered into the composition of the host and tumor factors that promote oncogenesis and treatment resistance. This in turn has led to the ingenious design of multiple new classes of drugs, which have now become the new standards of care in cancer therapy. These include novel antibody-drug conjugates, chimeric antigen receptor T cell therapies (CAR-T), and bispecific T cell engagers (BitTE). Certain host factors, such as the microbiome composition, are also emerging not only as biomarkers for the response and toxicity to anti-cancer therapies but also as potentially useful tools to modulate anti-tumor responses. The field is slowly moving away from one-size-fits-all treatment options to personalized treatments tailored to the host and tumor. This commentary aims to cover the basic concepts associated with these emerging therapies and the promises and challenges to fight cancer.
Collapse
Affiliation(s)
- Mariana Pilon Capella
- Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, QC H3T 1E9, Canada;
| | - Khashayar Esfahani
- Lady Davis Institute and Segal Cancer Centre, Jewish General Hospital, Departments of Medicine and Oncology, McGill University, Montreal, QC H3T 1E9, Canada;
- St Mary’s Hospital, Departments of Medicine and Oncology, McGill University, Montreal, QC H3T 1M5, Canada
| |
Collapse
|
11
|
Yu T, Jiang W, Wang Y, Zhou Y, Jiao J, Wu M. Chimeric antigen receptor T cells in the treatment of osteosarcoma (Review). Int J Oncol 2024; 64:40. [PMID: 38390935 PMCID: PMC10919759 DOI: 10.3892/ijo.2024.5628] [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: 09/12/2023] [Accepted: 01/25/2024] [Indexed: 02/24/2024] Open
Abstract
Osteosarcoma (OS) is a frequently occurring primary bone tumor, mostly affecting children, adolescents and young adults. Before 1970, surgical resection was the main treatment method for OS, but the clinical results were not promising. Subsequently, the advent of chemotherapy has improved the prognosis of patients with OS. However, there is still a high incidence of metastasis or recurrence, and chemotherapy has several side effects, thus making the 5‑year survival rate markedly low. Recently, chimeric antigen receptor T (CAR‑T) cell therapy represents an alternative immunotherapy approach with significant potential for hematologic malignancies. Nevertheless, the application of CAR‑T cells in the treatment of OS faces numerous challenges. The present review focused on the advances in the development of CAR‑T cells to improve their clinical efficacy, and discussed ways to overcome the difficulties faced by CAR T‑cell therapy for OS.
Collapse
Affiliation(s)
- Tong Yu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Weibo Jiang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Yang Wang
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Ying Zhou
- Department of Operating Room, The Third Hospital of Qinhuangdao, Qinhuangdao, Hebei 066000, P.R. China
| | - Jianhang Jiao
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| | - Minfei Wu
- Department of Orthopedics, The Second Norman Bethune Hospital of Jilin University, Changchun, Jilin 130000, P.R. China
| |
Collapse
|
12
|
Lan HR, Chen M, Yao SY, Chen JX, Jin KT. Novel immunotherapies for breast cancer: Focus on 2023 findings. Int Immunopharmacol 2024; 128:111549. [PMID: 38266449 DOI: 10.1016/j.intimp.2024.111549] [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: 11/07/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 01/26/2024]
Abstract
Immunotherapy has emerged as a revolutionary approach in cancer therapy, and recent advancements hold significant promise for breast cancer (BCa) management. Employing the patient's immune system to combat BCa has become a focal point in immunotherapeutic investigations. Strategies such as immune checkpoint inhibitors (ICIs), adoptive cell transfer (ACT), and targeting the tumor microenvironment (TME) have disclosed encouraging clinical outcomes. ICIs, particularly programmed cell death protein 1 (PD-1)/PD-L1 inhibitors, exhibit efficacy in specific BCa subtypes, including triple-negative BCa (TNBC) and human epidermal growth factor receptor 2 (HER2)-positive cancers. ACT approaches, including tumor-infiltrating lymphocytes (TILs) and chimeric antigen receptor (CAR) T-cell therapy, showed promising clinical outcomes in enhancing tumor recognition and elimination. Targeting the TME through immune agonists and oncolytic viruses signifies a burgeoning field of research. While challenges persist in patient selection, resistance mechanisms, and combination therapy optimization, these novel immunotherapies hold transformative potential for BCa treatment. Continued research and clinical trials are imperative to refine and implement these innovative approaches, paving the way for improved outcomes and revolutionizing the management of BCa. This review provides a concise overview of the latest immunotherapies (2023 studies) in BCa, highlighting their potential and current status.
Collapse
Affiliation(s)
- Huan-Rong Lan
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang 310002, China
| | - Min Chen
- Department of Colorectal Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310006, China
| | - Shi-Ya Yao
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China
| | - Jun-Xia Chen
- Department of Gynecology, Shaoxing People's Hospital, Shaoxing, Zhejiang 312000, China.
| | - Ke-Tao Jin
- Department of Gastrointestinal, Colorectal and Anal Surgery, Affiliated Hangzhou First People's Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang 310006, China.
| |
Collapse
|
13
|
Cheng S, Wang H, Kang X, Zhang H. Immunotherapy Innovations in the Fight against Osteosarcoma: Emerging Strategies and Promising Progress. Pharmaceutics 2024; 16:251. [PMID: 38399305 PMCID: PMC10892906 DOI: 10.3390/pharmaceutics16020251] [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: 11/13/2023] [Revised: 01/20/2024] [Accepted: 01/31/2024] [Indexed: 02/25/2024] Open
Abstract
Immunosuppressive elements within the tumor microenvironment are the primary drivers of tumorigenesis and malignant advancement. The presence, as well as the crosstalk between myeloid-derived suppressor cells (MDSCs), osteosarcoma-associated macrophages (OS-Ms), regulatory T cells (Tregs), and endothelial cells (ECs) with osteosarcoma cells cause the poor prognosis of OS. In addition, the consequent immunosuppressive factors favor the loss of treatment potential. Nanoparticles offer a means to dynamically and locally manipulate immuno-nanoparticles, which present a promising strategy for transforming OS-TME. Additionally, chimeric antigen receptor (CAR) technology is effective in combating OS. This review summarizes the essential mechanisms of immunosuppressive cells in the OS-TME and the current immune-associated strategies. The last part highlights the limitations of existing therapies and offers insights into future research directions.
Collapse
Affiliation(s)
- Shigao Cheng
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
- Department of Orthopedics, Hunan Loudi Central Hospital, Loudi 417000, China
| | - Huiyuan Wang
- Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 200031, China
| | - Xuejia Kang
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL 36849, USA
| | - Hui Zhang
- Laboratory of Stem Cell and Tissue Engineering, Orthopedic Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
14
|
Bellino S, La Salvia A, Cometa MF, Botta R. Cell-based medicinal products approved in the European Union: current evidence and perspectives. Front Pharmacol 2023; 14:1200808. [PMID: 37583902 PMCID: PMC10424920 DOI: 10.3389/fphar.2023.1200808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Accepted: 07/18/2023] [Indexed: 08/17/2023] Open
Abstract
Advanced Therapy Medicinal Products (ATMPs) are innovative clinical treatments exploiting the pharmacological, immunological, or metabolic properties of cells and/or gene(s) with the aim to restore, correct, or modify a biological function in the recipient. ATMPs are heterogeneous medicinal products, developed mainly as individualized and patient-specific treatments, and represent new opportunities for diseases characterized by a high-unmet medical need, including rare, genetic and neurodegenerative disorders, haematological malignancies, cancer, autoimmune, inflammatory and orthopaedic conditions. Into the European Union (EU) market, the first ATMP has been launched in 2009 and, to date, a total of 24 ATMPs have been approved. This review aims at reporting on current evidence of cell-based therapies authorized in the EU, including Somatic Cell Therapies, Tissue Engineering Products, and Cell-based Gene Therapy Products as Chimeric Antigen Receptor (CAR) T-cells, focusing on the evaluation of efficacy and safety in clinical trials and real-world settings. Despite cell-based therapy representing a substantial promise for patients with very limited treatment options, some limitations for its widespread use in the clinical setting remain, including restricted indications, highly complex manufacturing processes, elevated production costs, the lability of cellular products over time, and the potential safety concerns related to the intrinsic characteristics of living cells, including the risk of severe or life-threatening toxicities, such as CAR-T induced neurotoxicity and cytokine release syndrome (CRS). Although encouraging findings support the clinical use of ATMPs, additional data, comparative studies with a long-term follow-up, and wider real-world evidences are needed to provide further insights into their efficacy and safety profiles.
Collapse
Affiliation(s)
- Stefania Bellino
- National Center for Drug Research and Evaluation, National Institute of Health (Istituto Superiore di Sanità), Rome, Italy
| | | | | | | |
Collapse
|
15
|
Zhao M, Huang H, He F, Fu X. Current insights into the hepatic microenvironment and advances in immunotherapy for hepatocellular carcinoma. Front Immunol 2023; 14:1188277. [PMID: 37275909 PMCID: PMC10233045 DOI: 10.3389/fimmu.2023.1188277] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Accepted: 05/04/2023] [Indexed: 06/07/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common type of primary liver cancer and shows high global incidence and mortality rates. The liver is an immune-tolerated organ with a specific immune microenvironment that causes traditional therapeutic approaches to HCC, such as chemotherapy, radiotherapy, and molecular targeted therapy, to have limited efficacy. The dramatic advances in immuno-oncology in the past few decades have modified the paradigm of cancer therapy, ushering in the era of immunotherapy. Currently, despite the rapid integration of cancer immunotherapy into clinical practice, some patients still show no response to treatment. Therefore, a rational approach is to target the tumor microenvironment when developing the next generation of immunotherapy. This review aims to provide insights into the hepatic immune microenvironment in HCC and summarize the mechanisms of action and clinical usage of immunotherapeutic options for HCC, including immune checkpoint blockade, adoptive therapy, cytokine therapy, vaccine therapy, and oncolytic virus-based therapy.
Collapse
Affiliation(s)
| | | | - Feng He
- *Correspondence: Feng He, ; Xiangsheng Fu,
| | | |
Collapse
|
16
|
Li YR, Dunn ZS, Yu Y, Li M, Wang P, Yang L. Advancing cell-based cancer immunotherapy through stem cell engineering. Cell Stem Cell 2023; 30:592-610. [PMID: 36948187 PMCID: PMC10164150 DOI: 10.1016/j.stem.2023.02.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 01/04/2023] [Accepted: 02/22/2023] [Indexed: 03/24/2023]
Abstract
Advances in cell-based therapy, particularly CAR-T cell therapy, have transformed the treatment of hematological malignancies. Although an important step forward for the field, autologous CAR-T therapies are hindered by high costs, manufacturing challenges, and limited efficacy against solid tumors. With ongoing progress in gene editing and culture techniques, engineered stem cells and their application in cell therapy are poised to address some of these challenges. Here, we review stem cell-based immunotherapy approaches, stem cell sources, gene engineering and manufacturing strategies, therapeutic platforms, and clinical trials, as well as challenges and future directions for the field.
Collapse
Affiliation(s)
- Yan-Ruide Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Zachary Spencer Dunn
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA
| | - Yanqi Yu
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Miao Li
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Pin Wang
- Mork Family Department of Chemical Engineering and Materials Science, University of Southern California, Los Angeles, CA 90089, USA; Department of Pharmacology and Pharmaceutical Sciences, University of Southern California, Los Angeles, CA 90089, USA.
| | - Lili Yang
- Department of Microbiology, Immunology & Molecular Genetics, University of California, Los Angeles, Los Angeles, CA 90095, USA; Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA 90095, USA; Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA 90095, USA; Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA 90095, USA.
| |
Collapse
|
17
|
Ahmed H, Mahmud AR, Siddiquee MFR, Shahriar A, Biswas P, Shimul MEK, Ahmed SZ, Ema TI, Rahman N, Khan MA, Mizan MFR, Emran TB. Role of T cells in cancer immunotherapy: Opportunities and challenges. CANCER PATHOGENESIS AND THERAPY 2023; 1:116-126. [PMID: 38328405 PMCID: PMC10846312 DOI: 10.1016/j.cpt.2022.12.002] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Revised: 12/11/2022] [Accepted: 12/16/2022] [Indexed: 09/01/2023]
Abstract
Immunotherapies boosting the immune system's ability to target cancer cells are promising for the treatment of various tumor types, yet clinical responses differ among patients and cancers. Recently, there has been increasing interest in novel cancer immunotherapy practices aimed at triggering T cell-mediated anti-tumor responses. Antigen-directed cytotoxicity mediated by T lymphocytes has become a central focal point in the battle against cancer utilizing the immune system. The molecular and cellular mechanisms involved in the actions of T lymphocytes have directed new therapeutic approaches in cancer immunotherapy, including checkpoint blockade, adoptive and chimeric antigen receptor (CAR) T cell therapy, and cancer vaccinology. This review addresses all the strategies targeting tumor pathogenesis, including metabolic pathways, to evaluate the clinical significance of current and future immunotherapies for patients with cancer, which are further engaged in T cell activation, differentiation, and response against tumors.
Collapse
Affiliation(s)
- Hossain Ahmed
- Department of Biotechnology and Genetic Engineering, University of Development Alternative (UODA), 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | - Aar Rafi Mahmud
- Department of Biochemistry and Molecular Biology, Mawlana Bhashani Science and Technology University, Tangail, 1902, Bangladesh
| | | | - Asif Shahriar
- Department of Immunology and Microbiology, School of Medicine, University of Texas Rio Grande Valley, McAllen, TX, 78504, USA
| | - Partha Biswas
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology (JUST), Jashore, 7408, Bangladesh
| | - Md. Ebrahim Khalil Shimul
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Science and Technology, Jashore University of Science and Technology (JUST), Jashore, 7408, Bangladesh
| | - Shahlaa Zernaz Ahmed
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh
| | - Tanzila Ismail Ema
- Department of Biochemistry and Microbiology, North South University, Dhaka, 1229, Bangladesh
| | - Nova Rahman
- Department of Biochemistry and Molecular Biology, Jahangirnagar University, Savar, Dhaka, 1342, Bangladesh
| | - Md. Arif Khan
- Department of Biotechnology and Genetic Engineering, University of Development Alternative (UODA), 4/4B, Block A, Lalmatia, Dhaka, 1209, Bangladesh
| | | | - Talha Bin Emran
- Department of Pharmacy, BGC Trust University Bangladesh, Chittagong, 4381, Bangladesh
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, 1207, Bangladesh
| |
Collapse
|
18
|
Xu H, Li N, Wang G, Cao Y. Predictive short/long-term efficacy biomarkers and resistance mechanisms of CD19-directed CAR-T immunotherapy in relapsed/refractory B-cell lymphomas. Front Immunol 2023; 14:1110028. [PMID: 37051246 PMCID: PMC10083339 DOI: 10.3389/fimmu.2023.1110028] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Accepted: 03/14/2023] [Indexed: 03/28/2023] Open
Abstract
Genetically modified T-cell immunotherapies are revolutionizing the therapeutic options for hematological malignancies, especially those of B-cell origin. Impressive efficacies of CD19-directed chimeric antigen receptor (CAR)-T therapy have been reported in refractory/relapsed (R/R) B-cell non-Hodgkin lymphoma (NHL) patients who were resistant to current standard therapies, with a complete remission (CR) rate of approximately 50%. At the same time, problems of resistance and relapse following CAR-T therapy have drawn growing attention. Recently, great efforts have been made to determine various factors that are connected to the responses and outcomes following CAR-T therapy, which may not only allow us to recognize those with a higher likelihood of responding and who could benefit most from the therapy but also identify those with a high risk of resistance and relapse and to whom further appropriate treatment should be administered following CAR-T therapy. Thus, we concentrate on the biomarkers that can predict responses and outcomes after CD19-directed CAR-T immunotherapy. Furthermore, the mechanisms that may lead to treatment failure are also discussed in this review.
Collapse
Affiliation(s)
- Hao Xu
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Ningwen Li
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
| | - Gaoxiang Wang
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
- *Correspondence: Gaoxiang Wang, ; Yang Cao,
| | - Yang Cao
- Department of Hematology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
- Immunotherapy Research Center for Hematologic Diseases of Hubei Province, Wuhan, Hubei, China
- *Correspondence: Gaoxiang Wang, ; Yang Cao,
| |
Collapse
|
19
|
Zhang XW, Wu YS, Xu TM, Cui MH. CAR-T Cells in the Treatment of Ovarian Cancer: A Promising Cell Therapy. Biomolecules 2023; 13:biom13030465. [PMID: 36979400 PMCID: PMC10046142 DOI: 10.3390/biom13030465] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 02/23/2023] [Accepted: 03/01/2023] [Indexed: 03/06/2023] Open
Abstract
Ovarian cancer (OC) is among the most common gynecologic malignancies with a poor prognosis and a high mortality rate. Most patients are diagnosed at an advanced stage (stage III or IV), with 5-year survival rates ranging from 25% to 47% worldwide. Surgical resection and first-line chemotherapy are the main treatment modalities for OC. However, patients usually relapse within a few years of initial treatment due to resistance to chemotherapy. Cell-based therapies, particularly adoptive T-cell therapy and chimeric antigen receptor T (CAR-T) cell therapy, represent an alternative immunotherapy approach with great potential for hematologic malignancies. However, the use of CAR-T-cell therapy for the treatment of OC is still associated with several difficulties. In this review, we comprehensively discuss recent innovations in CAR-T-cell engineering to improve clinical efficacy, as well as strategies to overcome the limitations of CAR-T-cell therapy in OC.
Collapse
|
20
|
Kacen A, Javitt A, Kramer MP, Morgenstern D, Tsaban T, Shmueli MD, Teo GC, da Veiga Leprevost F, Barnea E, Yu F, Admon A, Eisenbach L, Samuels Y, Schueler-Furman O, Levin Y, Nesvizhskii AI, Merbl Y. Post-translational modifications reshape the antigenic landscape of the MHC I immunopeptidome in tumors. Nat Biotechnol 2023; 41:239-251. [PMID: 36203013 PMCID: PMC11197725 DOI: 10.1038/s41587-022-01464-2] [Citation(s) in RCA: 34] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 08/09/2022] [Indexed: 11/08/2022]
Abstract
Post-translational modification (PTM) of antigens provides an additional source of specificities targeted by immune responses to tumors or pathogens, but identifying antigen PTMs and assessing their role in shaping the immunopeptidome is challenging. Here we describe the Protein Modification Integrated Search Engine (PROMISE), an antigen discovery pipeline that enables the analysis of 29 different PTM combinations from multiple clinical cohorts and cell lines. We expanded the antigen landscape, uncovering human leukocyte antigen class I binding motifs defined by specific PTMs with haplotype-specific binding preferences and revealing disease-specific modified targets, including thousands of new cancer-specific antigens that can be shared between patients and across cancer types. Furthermore, we uncovered a subset of modified peptides that are specific to cancer tissue and driven by post-translational changes that occurred in the tumor proteome. Our findings highlight principles of PTM-driven antigenicity, which may have broad implications for T cell-mediated therapies in cancer and beyond.
Collapse
Affiliation(s)
- Assaf Kacen
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Aaron Javitt
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Matthias P Kramer
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - David Morgenstern
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Tomer Tsaban
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Merav D Shmueli
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Guo Ci Teo
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | | | - Eilon Barnea
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Fengchao Yu
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
| | - Arie Admon
- Faculty of Biology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Lea Eisenbach
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yardena Samuels
- Department of Molecular Cell Biology, Weizmann Institute of Science, Rehovot, Israel
| | - Ora Schueler-Furman
- Department of Microbiology and Molecular Genetics, Institute for Medical Research Israel-Canada, Faculty of Medicine, The Hebrew University, Jerusalem, Israel
| | - Yishai Levin
- De Botton Institute for Protein Profiling, Nancy and Stephen Grand Israel National Center for Personalized Medicine, Weizmann Institute of Science, Rehovot, Israel
| | - Alexey I Nesvizhskii
- Department of Pathology, University of Michigan, Ann Arbor, MI, USA
- Department of Computational Medicine and Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Yifat Merbl
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel.
| |
Collapse
|
21
|
Keshavjee SH, Moy RH, Reiner SL, Ryeom SW, Yoon SS. Gastric Cancer and the Immune System: The Key to Improving Outcomes? Cancers (Basel) 2022; 14:cancers14235940. [PMID: 36497422 PMCID: PMC9739366 DOI: 10.3390/cancers14235940] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/21/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
Gastric adenocarcinoma is by far the most common form of gastric cancer (GC) and is a highly lethal form of cancer arising from the gastric epithelium. GC is an important area of focus of the medical community, given its often late-stage of diagnosis and associated high mortality rate. While surgery and chemotherapy remain the primary treatments, attention has been drawn to the use of immunologic therapies, which have shown promise in the treatment of other malignancies. The role for immune-based therapies has become clearer as we obtain a greater understanding of the role of the immune system in gastric cancer formation and growth. A variety treatment to augment the immune system are under evaluation in clinical trials, and these include immune checkpoint inhibitors, antibody-drug conjugates, and immune cell-based therapies. Here, we review the immune landscape and immune-based therapies for GC.
Collapse
Affiliation(s)
- Sara H. Keshavjee
- Division of Surgical Oncology, Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
| | - Ryan H. Moy
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Division of Hematology/Oncology, Department of Medicine, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Steven L. Reiner
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sandra W. Ryeom
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Division of Surgical Sciences, Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
| | - Sam S. Yoon
- Division of Surgical Oncology, Department of Surgery, Columbia University Irving Medical Center, New York, NY 10032, USA
- Vagelos College of Physicians and Surgeons, Columbia University, New York, NY 10032, USA
- Correspondence:
| |
Collapse
|
22
|
Jin M, Cao W, Chen B, Xiong M, Cao G. Tumor-Derived Lactate Creates a Favorable Niche for Tumor via Supplying Energy Source for Tumor and Modulating the Tumor Microenvironment. Front Cell Dev Biol 2022; 10:808859. [PMID: 35646923 PMCID: PMC9136137 DOI: 10.3389/fcell.2022.808859] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 04/08/2022] [Indexed: 12/05/2022] Open
Abstract
Tumor evolution is influenced by events involving tumor cells and the environment in which they live, known as the tumor microenvironment (TME). TME is a functional and structural niche composed of tumor cells, endothelial cells (ECs), cancer-associated fibroblasts (CAFs), mesenchymal stromal cells (MSCs), and a subset of immune cells (macrophages, dendritic cells, natural killer cells, T cells, B cells). Otto Warburg revealed the Warburg effect in 1923, a characteristic metabolic mechanism of tumor cells that performs high glucose uptake and excessive lactate formation even in abundant oxygen. Tumor tissues excrete a large amount of lactate into the extracellular microenvironment in response to TME’s hypoxic or semi-hypoxic state. High lactate concentrations in tumor biopsies have been linked to metastasis and poor clinical outcome. This indicates that the metabolite may play a role in carcinogenesis and lead to immune escape in TME. Lactate is now recognized as an essential carbon source for cellular metabolism and as a signaling molecule in TME, forming an active niche that influences tumor progression. This review summarized the advanced literature demonstrating the functional role of lactate in TME remodeling, elucidating how lactate shapes the behavior and the phenotype of both tumor cells and tumor-associated cells. We also concluded the intriguing interactions of multiple immune cells in TME. Additionally, we demonstrated how lactate functioned as a novel function factor by being used in a new histone modification, histone lysine lactylation, and to regulate gene expression in TME. Ultimately, because lactate created a favorable niche for tumor progression, we summarized potential anti-tumor strategies targeting lactate metabolism and signaling to investigate better cancer treatment.
Collapse
Affiliation(s)
| | | | - Bo Chen
- *Correspondence: Bo Chen, ; Maoming Xiong, ; Guodong Cao,
| | - Maoming Xiong
- *Correspondence: Bo Chen, ; Maoming Xiong, ; Guodong Cao,
| | - Guodong Cao
- *Correspondence: Bo Chen, ; Maoming Xiong, ; Guodong Cao,
| |
Collapse
|
23
|
Kimura Y, Ghosn M, Cheema W, Adusumilli PS, Solomon SB, Srimathveeralli G. Expanding the role of interventional oncology for advancing precision immunotherapy of solid tumors. Mol Ther Oncolytics 2022; 24:194-204. [PMID: 35036524 PMCID: PMC8752905 DOI: 10.1016/j.omto.2021.12.018] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Adoptive cell therapy with chimeric antigen receptors (CAR) T cells has proven effective for hematologic malignancies, but success in solid tumors has been impeded by poor intratumoral infiltration, exhaustion of effector cells from antigen burden, and an immunosuppressive tumor microenvironment. Results from recent clinical trials and preclinical studies lend promising evidence of locoregional approaches for CAR T cell delivery, priming the tumor microenvironment, and performing adjuvant therapies that sustain T cell activity. Interventional oncology is a subspeciality of interventional radiology where imaging guidance is used to perform percutaneous and catheter-directed procedures for localized, non-surgical therapy or interrogation of solid tumors. Interventional oncology provides unique synergies with immunotherapy, which has been well-studied to improve treatment efficacy while reducing toxicities associated with systemic treatment. Besides aiding in CAR T cell delivery, priming, or the stimulation of the tumor microenvironment to promote effector survival and function, interventional oncology can also aid in the monitoring of treatment response through selective, multiplex tumor sampling and catheter-based venous sampling. This review presents an overview of interventional oncology, its various procedures, and its potential for advancing CAR T cell immunotherapy of solid tumors.
Collapse
Affiliation(s)
- Yasushi Kimura
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA
| | - Mario Ghosn
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Waseem Cheema
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Prasad S. Adusumilli
- Thoracic Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Stephen B. Solomon
- Interventional Radiology Service, Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Govindarajan Srimathveeralli
- Department of Mechanical and Industrial Engineering, University of Massachusetts, Amherst, MA, USA
- Department of Biomedical Engineering, University of Massachusetts at Amherst, Amherst, MA, USA
- Institute for Applied Life Sciences, University of Massachusetts at Amherst, Amherst, MA, USA
| |
Collapse
|
24
|
Genetic Modification of T Cells for the Immunotherapy of Cancer. Vaccines (Basel) 2022; 10:vaccines10030457. [PMID: 35335089 PMCID: PMC8949949 DOI: 10.3390/vaccines10030457] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 03/05/2022] [Accepted: 03/11/2022] [Indexed: 02/01/2023] Open
Abstract
Immunotherapy is a beneficial treatment approach for multiple cancers, however, current therapies are effective only in a small subset of patients. Adoptive cell transfer (ACT) is a facet of immunotherapy where T cells targeting the tumor cells are transferred to the patient with several primary forms, utilizing unmodified or modified T cells: tumor-infiltrating lymphocytes (TIL), genetically modified T cell receptor transduced T cells, and chimeric antigen receptor (CAR) transduced T cells. Many clinical trials are underway investigating the efficacy and safety of these different subsets of ACT, as well as trials that combine one of these subsets with another type of immunotherapy. The main challenges existing with ACT are improving clinical responses and decreasing adverse events. Current research focuses on identifying novel tumor targeting T cell receptors, improving safety and efficacy, and investigating ACT in combination with other immunotherapies.
Collapse
|
25
|
Patel U, Abernathy J, Savani BN, Oluwole O, Sengsayadeth S, Dholaria B. CAR T cell therapy in solid tumors: A review of current clinical trials. EJHAEM 2022; 3:24-31. [PMID: 35844304 PMCID: PMC9175685 DOI: 10.1002/jha2.356] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 10/13/2021] [Indexed: 01/26/2023]
Abstract
Chimeric antigen receptor (CAR) T cell therapy has made tremendous strides in the arena of hematological malignancies with approved therapies in certain leukemias, lymphomas, and recently myeloma with overall highly favorable response rates. While numerous clinical studies are still ongoing for hematological malignancies, research is developing to translate the feasibility of CAR T therapy in solid organ malignancies. Unfortunately, the majority of diagnosed cancers are primarily solid tumors. Thus, a highly unmet clinical need for further research and development exists in this field. This review article highlights currently active clinical trials and a few pertinent preclinical studies involving CAR T cell therapy in solid tumors while briefly discussing study outcomes and potential key targets that may allow for the feasibility of this therapy option. Finally, we mention critical challenges existing in the solid tumor environment and discuss developing strategies that may potentially overcome the existing barriers to CAR T cell progress in solid tumors.
Collapse
Affiliation(s)
- Urvi Patel
- College of PharmacyThe University of Tennessee Health Science CenterNashvilleTennesseeUSA
| | - John Abernathy
- College of MedicineThe University of Tennessee Health Science CenterMemphisTennesseeUSA
| | - Bipin N Savani
- Department of Hematology‐ OncologyVanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Olalekan Oluwole
- Department of Hematology‐ OncologyVanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Salyka Sengsayadeth
- Department of Hematology‐ OncologyVanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| | - Bhagirathbhai Dholaria
- Department of Hematology‐ OncologyVanderbilt‐Ingram Cancer CenterVanderbilt University Medical CenterNashvilleTennesseeUSA
| |
Collapse
|
26
|
Luo Y, Chen Z, Sun M, Li B, Pan F, Ma A, Liao J, Yin T, Tang X, Huang G, Zhang B, Pan H, Zheng M, Cai L. IL-12 nanochaperone-engineered CAR T cell for robust tumor-immunotherapy. Biomaterials 2021; 281:121341. [PMID: 34995901 DOI: 10.1016/j.biomaterials.2021.121341] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 12/17/2021] [Accepted: 12/25/2021] [Indexed: 12/31/2022]
Abstract
Although chimeric antigen receptor T (CAR T) cell immunotherapy has demonstrated remarkable success in clinical, therapeutic effects are still limited in solid tumor due to lack of activated T cell infiltration in immunosuppression of tumor microenvironment. Herein, we develop IL-12 nanostimulant-engineered CAR T cell (INS-CAR T) biohybrids for boosting antitumor immunity of CAR T cells via immunofeedback. As stimulating nanochaperone, IL-12-loaded human serum albumin (HSA) nanoparticles are effectively conjugated onto CAR T cells via bioorthogonal chemistry without influencing their antitumor capabilities. IL-12 is responsively released from INS-CAR T biohybrids in presence of the increased thiol groups on cell-surface triggered by tumor antigens. In return, released IL-12 obviously promotes the secretion of CCL5, CCL2 and CXCL10, which further selectively recruits and expands CD8+ CAR T cells in tumors. Ultimately, the immune-enhancing effects of IL-12 nanochaperone significantly boost CAR T cell antitumor capabilities, dramatically eliminated solid tumor and minimized unwanted side effects. Hence, immunofeedback INS-CAR T biohybrids, which include INS that serves as an intelligent 'nanochaperone', could provide a powerful tool for efficient and safe antitumor immunotherapy.
Collapse
Affiliation(s)
- Yingmei Luo
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China; Key Laboratory for Nanomedicine, Department of Histology and Embryology and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China
| | - Ze Chen
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Mingjian Sun
- School of Information Science and Engineering, Harbin Institute of Technology, Weihai, Shandong, 264209, China
| | - Baohong Li
- Key Laboratory for Nanomedicine, Department of Histology and Embryology and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China
| | - Fan Pan
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Aiqing Ma
- Key Laboratory for Nanomedicine, Department of Histology and Embryology and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China
| | - Jianhong Liao
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Ting Yin
- Key Laboratory for Nanomedicine, Department of Histology and Embryology and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China
| | - Xiaofan Tang
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Guojun Huang
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Baozhen Zhang
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China
| | - Hong Pan
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China.
| | - Mingbin Zheng
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China; National Clinical Research Center for Infectious Disease, Shenzhen Third People's Hospital, The Second Affiliated Hospital, Southern University of Science and Technology Shenzhen, 518112, PR China; Key Laboratory for Nanomedicine, Department of Histology and Embryology and School of Pharmacy, Guangdong Medical University, Dongguan, 523808, PR China; Zhuhai Institute of Advanced Technology Chinese Academy of Sciences, 519000, Zhuhai, PR China.
| | - Lintao Cai
- Guangdong Key Laboratory of Nanomedicine, CAS Key Laboratory of Health Informatics, CAS-HK Joint Lab of Biomaterials, Shenzhen Institutes of Advanced Technology (SIAT), Chinese Academy of Sciences, Shenzhen, 518055, PR China; Zhuhai Institute of Advanced Technology Chinese Academy of Sciences, 519000, Zhuhai, PR China.
| |
Collapse
|
27
|
Roy S, Ghosh J, Ghosh R. Cancer Vaccine in Solid Tumors: Where We Stand. Indian J Med Paediatr Oncol 2021. [DOI: 10.1055/s-0041-1735439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
AbstractCancer immunotherapy has achieved landmark progress in the field of medical oncology in the era of personalized medicine. In the recent past, our knowledge has expanded regarding how tumor cells escape from the immune system, introducing immunosuppressive microenvironments, and developing tolerance. Therapeutic cancer vaccine leads to activation of immune memory that is long-lasting, safe, and effective; hence, it is becoming an attractive method of immunotherapy. Various cancer vaccine trials in the past have taught us the types of target selection, magnitude of immune response, and implementation of appropriate technologies for the development of new successful cancer vaccines. Tumor-associated antigens, cancer germline antigens, oncogenic viral antigens, and tumor-specific antigens, also known as neoantigens, are potential targets for designing therapeutic cancer vaccines. Cancer vaccine could be cell based, viral vector based, peptide based, and nucleic acid based (DNA/RNA). Several preclinical and clinical studies have demonstrated the mechanism of action, safety, efficacy, and toxicities of various types of cancer vaccines. In this article, we review the types of various tumor antigens and types of cancer vaccines tested in clinical trials and discuss the application and importance of this approach toward precision medicine in the field of immuno-oncology.
Collapse
Affiliation(s)
- Somnath Roy
- Department of Medical Oncology, Tata Memorial Centre, Homi Bhabha Cancer Hospital, Varanasi, Uttar Pradesh, India
| | - Joydeep Ghosh
- Department of Medical Oncology, Tata Medical Center, Kolkata, West Bengal, India
| | - Ranti Ghosh
- Deparment of Radiation Oncology, Tata Memorial Centre, Homi Bhabha Cancer Hospital, Varanasi, Uttar Pradesh, India
| |
Collapse
|
28
|
Chen M, Fu M, Wang A, Wu X, Zhen J, Gong M, Zhang X, Yue G, Du Q, Zhao W, Zhao Y, Lu P, Wang H. Cytoplasmic CD79a is a promising biomarker for B lymphoblastic leukemia follow up post CD19 CAR-T therapy. Leuk Lymphoma 2021; 63:426-434. [PMID: 34672246 DOI: 10.1080/10428194.2021.1980214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Minimal residual disease (MRD) detection is an important prognostic parameter in patients with refractory or relapsed B-cell acute lymphoblastic leukemia (R/R B-ALL). CD79a has been reported to exhibit a high degree of linage-specificity for B-cell differentiation, with a specificity of 88% and a sensitivity of 100%. In this study, we investigated the efficiency and prognostic role of cytoplasmic CD79a (cCD79a) antibody-gated multicolor flow cytometry (MFC) in MRD detection in patients with B-ALL who received CD19-targeted chimeric antigen receptor (CAR) T-cell therapy bridging to allogeneic hematopoietic stem cell transplantation (allo-HSCT). The retrospective analysis was carried on to 59 patients who accepted allo-HSCT after CD19-CAR-T infusion from June 2016 to May 2017. The MFC MRD statuses before and after allo-HSCT were both strongly correlated with the transplantation prognosis, the MFC panel with cCD79a gating can effectively monitor MRD after CD19 CAR T-cell therapy and predict the prognosis after allo-HSCT. Trial registration: ClinicalTrials#: ChiCTR-IIh-16008711.gov: NCT03173417. Registered 30 May 2017 - retrospectively registered, https://www.clinicaltrials.gov/.
Collapse
Affiliation(s)
- Man Chen
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Minjing Fu
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Aixian Wang
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Xueying Wu
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Junyi Zhen
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Meiwei Gong
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Xian Zhang
- Department of Haematology, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Guanlan Yue
- Department of Stem Cell Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Qing Du
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Wei Zhao
- Department of Stem Cell Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Yanli Zhao
- Department of Stem Cell Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Peihua Lu
- Department of Stem Cell Transplantation, Hebei Yanda Lu Daopei Hospital, Langfang, China
| | - Hui Wang
- Department of Laboratory Medicine, Hebei Yanda Lu Daopei Hospital, Langfang, China
| |
Collapse
|
29
|
Short Review on Advances in Hydrogel-Based Drug Delivery Strategies for Cancer Immunotherapy. Tissue Eng Regen Med 2021; 19:263-280. [PMID: 34596839 DOI: 10.1007/s13770-021-00369-6] [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] [Received: 06/01/2021] [Revised: 06/10/2021] [Accepted: 06/16/2021] [Indexed: 12/20/2022] Open
Abstract
Cancer immunotherapy has become the new paradigm of cancer treatment. The introduction and discovery of various therapeutic agents have also accelerated the application of immunotherapy in clinical trials. However, despite the significant potency and demonstrated advantages of cancer immunotherapy, its clinical application to patients faces several safety and efficacy issues, including autoimmune reactions, cytokine release syndrome, and vascular leak syndrome-related issues. In addressing these problems, biomaterials traditionally used for tissue engineering and drug delivery are attracting attention. Among them, hydrogels can be easily injected into tumors with drugs, and they can minimize side effects by retaining immune therapeutics at the tumor site for a long time. This article reviews the status of functional hydrogels for effective cancer immunotherapy. First, we describe the basic mechanisms of cancer immunotherapy and the advantages of using hydrogels to apply these mechanisms. Next, we summarize recent advances in the development of functional hydrogels designed to locally release various immunotherapeutic agents, including cytokines, cancer immune vaccines, immune checkpoint inhibitors, and chimeric antigen receptor-T cells. Finally, we briefly discuss the current problems and possible prospects of hydrogels for effective cancer immunotherapy.
Collapse
|
30
|
Jafarzadeh L, Masoumi E, Mirzaei HR, Alishah K, Fallah-Mehrjardi K, Khakpoor-Koosheh M, Rostamian H, Noorbakhsh F, Hadjati J. Targeted knockdown of Tim3 by short hairpin RNAs improves the function of anti-mesothelin CAR T cells. Mol Immunol 2021; 139:1-9. [PMID: 34450537 DOI: 10.1016/j.molimm.2021.06.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/28/2021] [Accepted: 06/13/2021] [Indexed: 12/21/2022]
Abstract
T-cell immunoglobulin mucin 3 (Tim3) is an immune checkpoint receptor that plays a central role in chimeric antigen receptor (CAR) T cell exhaustion within the tumor microenvironment. This study was aimed to evaluate the effects of targeted-knockdown of Tim3 on the antitumor function of anti-mesothelin (MSLN)-CAR T cells. To knockdown Tim3 expression, three different shRNA sequences specific to different segments of the human Tim3 gene were designed and co-inserted with an anti-MSLN-CAR transgene into lentiviral vectors. To investigate the efficacy of Tim3 targeting in T cells, expression of Tim3 was assessed before and after antigen stimulation. Afterwards, cytotoxic effects, proliferative response and cytokine production of MSLN-CAR T cells and Tim3-targeted MSLN-CAR T cells were analyzed. Our results showed that activation of T cells and MSLN-CAR T cells led to up-regulation of Tim3. Tim3 knockdown significantly decreased its expression in different groups of MSLN-CAR T cells. Tim3 knockdown significantly improved cytotoxic function, cytokine production and proliferation capacity of MSLN-CAR T cells. Our findings indicate that targeted knockdown of Tim3 allows tumor-infiltrating CAR T cells that would otherwise be inactivated to continue to expand and carry out effector functions, thereby altering the tumor microenvironment from immunosuppressive to immunosupportive via mitigated Tim3 signaling.
Collapse
Affiliation(s)
- Leila Jafarzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Elham Masoumi
- Department of Immunology, Faculty of Medicine, Ilam University of Medical Sciences, Ilam, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Khadijeh Alishah
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Keyvan Fallah-Mehrjardi
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Khakpoor-Koosheh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hosein Rostamian
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Farshid Noorbakhsh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Jamshid Hadjati
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
31
|
Liang X, Huang Y, Li D, Yang X, Jiang L, Zhou W, Su J, Chen N, Wang W. Distinct functions of CAR-T cells possessing a dectin-1 intracellular signaling domain. Gene Ther 2021; 30:411-420. [PMID: 33953316 DOI: 10.1038/s41434-021-00257-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 03/20/2021] [Accepted: 04/01/2021] [Indexed: 02/05/2023]
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has demonstrated remarkable efficacies in treating hematopoietic malignancies, but not in the solid tumors. Incorporating costimulatory signaling domains, such as ICOS or 4-1BB, can positively influence CAR-T cell functions and then the immune responses. These CAR-engineered T cells have showed their enhanced persistence and effector functions with improved antitumor activities, and provided a new approach for the treatment of solid tumors. Here, we designed novel 2nd generation CARs with a costimulatory signaling molecule, dectin-1. The impacts of dectin-1 signaling domain on CAR-T cells were evaluated in vitro and in vivo. Our data show that in vitro cytokine secretions by HER2 or CD19 specific CAR-T cells increase significantly via incorporating this dectin-1 signaling domain. Additional properties of these novel CAR-T cells are affected by this costimulatory domain. Compared with a popular reference (i.e., anti-HER2 CAR-T cells with 4-1BB), in vitro T cell functions and in vivo antitumor activity of the dectin-1 engineered CAR-T cells are similar to the 4-1BB based, and both are discrete to the mock T cells. Furthermore, we found that the CAR-T cells with dectin-1 show distinct phenotype and exhaustion marker expression. These collective results suggest that the incorporation of this new signaling domain, dectin-1, into the CARs may provide the clinical potential of the CAR-T cells through this signaling domain in treating solid tumors.
Collapse
Affiliation(s)
- Xiao Liang
- Department of Head & Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yong Huang
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dan Li
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiao Yang
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Jiang
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Weilin Zhou
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jinhua Su
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Nianyong Chen
- Department of Head & Neck Oncology, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Wei Wang
- State Key Laboratory of Biotherapy/Collaborative Center for Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| |
Collapse
|
32
|
Dana H, Chalbatani GM, Jalali SA, Mirzaei HR, Grupp SA, Suarez ER, Rapôso C, Webster TJ. CAR-T cells: Early successes in blood cancer and challenges in solid tumors. Acta Pharm Sin B 2021; 11:1129-1147. [PMID: 34094824 PMCID: PMC8144892 DOI: 10.1016/j.apsb.2020.10.020] [Citation(s) in RCA: 53] [Impact Index Per Article: 17.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Revised: 07/20/2020] [Accepted: 08/21/2020] [Indexed: 12/12/2022] Open
Abstract
New approaches to cancer immunotherapy have been developed, showing the ability to harness the immune system to treat and eliminate cancer. For many solid tumors, therapy with checkpoint inhibitors has shown promise. For hematologic malignancies, adoptive and engineered cell therapies are being widely developed, using cells such as T lymphocytes, as well as natural killer (NK) cells, dendritic cells, and potentially others. Among these adoptive cell therapies, the most active and advanced therapy involves chimeric antigen receptor (CAR)-T cells, which are T cells in which a chimeric antigen receptor is used to redirect specificity and allow T cell recognition, activation and killing of cancers, such as leukemia and lymphoma. Two autologous CAR-T products have been approved by several health authorities, starting with the U.S. Food and Drug Administration (FDA) in 2017. These products have shown powerful, inducing, long-lasting effects against B cell cancers in many cases. In distinction to the results seen in hematologic malignancies, the field of using CAR-T products against solid tumors is in its infancy. Targeting solid tumors and trafficking CAR-T cells into an immunosuppressive microenvironment are both significant challenges. The goal of this review is to summarize some of the most recent aspects of CAR-T cell design and manufacturing that have led to successes in hematological malignancies, allowing the reader to appreciate the barriers that must be overcome to extend CAR-T therapies to solid tumors successfully.
Collapse
Affiliation(s)
- Hassan Dana
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 13145-158, Iran
| | - Ghanbar Mahmoodi Chalbatani
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717434, Iran
| | - Seyed Amir Jalali
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran 1985717434, Iran
| | - Hamid Reza Mirzaei
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran 1417613151, Iran
| | - Stephan A. Grupp
- Division of Oncology, Department of Pediatrics, the Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Eloah Rabello Suarez
- Center for Natural and Human Sciences, Federal University of ABC, Santo André, SP 09210-580, Brazil
| | - Catarina Rapôso
- Faculty of Pharmaceutical Sciences, State University of Campinas (UNICAMP), Campinas, SP 13083-871, Brazil
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, Boston, MA 02115, USA
| |
Collapse
|
33
|
Cotechini T, Atallah A, Grossman A. Tissue-Resident and Recruited Macrophages in Primary Tumor and Metastatic Microenvironments: Potential Targets in Cancer Therapy. Cells 2021; 10:960. [PMID: 33924237 PMCID: PMC8074766 DOI: 10.3390/cells10040960] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/16/2021] [Accepted: 04/17/2021] [Indexed: 12/24/2022] Open
Abstract
Macrophages within solid tumors and metastatic sites are heterogenous populations with different developmental origins and substantially contribute to tumor progression. A number of tumor-promoting phenotypes associated with both tumor- and metastasis-associated macrophages are similar to innate programs of embryonic-derived tissue-resident macrophages. In contrast to recruited macrophages originating from marrow precursors, tissue-resident macrophages are seeded before birth and function to coordinate tissue remodeling and maintain tissue integrity and homeostasis. Both recruited and tissue-resident macrophage populations contribute to tumor growth and metastasis and are important mediators of resistance to chemotherapy, radiation therapy, and immune checkpoint blockade. Thus, targeting various macrophage populations and their tumor-promoting phenotypes holds therapeutic promise. Here, we discuss various macrophage populations as regulators of tumor progression, immunity, and immunotherapy. We provide an overview of macrophage targeting strategies, including therapeutics designed to induce macrophage depletion, impair recruitment, and induce repolarization. We also provide a perspective on the therapeutic potential for macrophage-specific acquisition of trained immunity as an anti-cancer agent and discuss the therapeutic potential of exploiting macrophages and their traits to reduce tumor burden.
Collapse
Affiliation(s)
- Tiziana Cotechini
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 3N6, Canada; (A.A.); (A.G.)
| | | | | |
Collapse
|
34
|
Ray SK, Meshram Y, Mukherjee S. Cancer Immunology and CAR-T Cells: A Turning Point Therapeutic Approach in Colorectal Carcinoma with Clinical Insight. Curr Mol Med 2021; 21:221-236. [PMID: 32838717 DOI: 10.2174/1566524020666200824103749] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 07/24/2020] [Accepted: 08/28/2020] [Indexed: 12/24/2022]
Abstract
Cancer immunotherapy endeavours in harnessing the delicate strength and specificity of the immune system for therapy of different malignancies, including colorectal carcinoma. The recent challenge for cancer immunotherapy is to practice and develop molecular immunology tools to create tactics that efficiently and securely boost antitumor reactions. After several attempts of deceptive outcomes, the wave has lastly altered and immunotherapy has become a clinically confirmed treatment for several cancers. Immunotherapeutic methods include the administration of antibodies or modified proteins that either block cellular activity or co-stimulate cells through immune control pathways, cancer vaccines, oncolytic bacteria, ex vivo activated adoptive transfer of T cells and natural killer cells. Engineered T cells are used to produce a chimeric antigen receptor (CAR) to treat different malignancies, including colorectal carcinoma in a recent decade. Despite the considerable early clinical success, CAR-T therapies are associated with some side effects and sometimes display minimal efficacy. It gives special emphasis on the latest clinical evidence with CAR-T technology and also other related immunotherapeutic methods with promising performance, and highlighted how this therapy can affect the therapeutic outcome and next upsurge as a key clinical aspect of colorectal carcinoma. In this review, we recapitulate the current developments produced to improve the efficacy and specificity of CAR-T therapies in colon cancer.
Collapse
Affiliation(s)
- Suman K Ray
- Independent Researcher, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh-462020, India
| | - Yamini Meshram
- Independent Researcher, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh-462020, India
| | - Sukhes Mukherjee
- Department of Biochemistry, All India Institute of Medical Sciences, Bhopal, Madhya Pradesh-462020, India
| |
Collapse
|
35
|
Nukala U, Rodriguez Messan M, Yogurtcu ON, Wang X, Yang H. A Systematic Review of the Efforts and Hindrances of Modeling and Simulation of CAR T-cell Therapy. AAPS JOURNAL 2021; 23:52. [PMID: 33835308 DOI: 10.1208/s12248-021-00579-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 03/06/2021] [Indexed: 01/08/2023]
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is an immunotherapy that has recently become highly instrumental in the fight against life-threatening diseases. A variety of modeling and computational simulation efforts have addressed different aspects of CAR T-cell therapy, including T-cell activation, T- and malignant cell population dynamics, therapeutic cost-effectiveness strategies, and patient survival. In this article, we present a systematic review of those efforts, including mathematical, statistical, and stochastic models employing a wide range of algorithms, from differential equations to machine learning. To the best of our knowledge, this is the first review of all such models studying CAR T-cell therapy. In this review, we provide a detailed summary of the strengths, limitations, methodology, data used, and data gap in currently published models. This information may help in designing and building better models for enhanced prediction and assessment of the benefit-risk balance associated with novel CAR T-cell therapies, as well as with the data need for building such models.
Collapse
Affiliation(s)
- Ujwani Nukala
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, Silver Spring, Maryland, USA
| | - Marisabel Rodriguez Messan
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, Silver Spring, Maryland, USA
| | - Osman N Yogurtcu
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, Silver Spring, Maryland, USA
| | - Xiaofei Wang
- Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, US FDA, Silver Spring, Maryland, USA
| | - Hong Yang
- Office of Biostatistics and Epidemiology, Center for Biologics Evaluation and Research, US FDA, Silver Spring, Maryland, USA.
| |
Collapse
|
36
|
Luo Y, Song G, Liang S, Li F, Liu K. Research advances in chimeric antigen receptor-modified T-cell therapy (Review). Exp Ther Med 2021; 21:484. [PMID: 33790993 PMCID: PMC8005741 DOI: 10.3892/etm.2021.9915] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 01/26/2021] [Indexed: 12/15/2022] Open
Abstract
Chimeric antigen receptor (CAR)-modified T-cells are T-cells that have been genetically engineered to express CAR molecules to target specific surface antigens on tumor cells. CAR T-cell therapy, a novel cancer immunotherapy, has been attracting increasing attention, since it exhibited notable efficacy in the treatment of hematological tumors in clinical trials. However, for this type of therapy, challenges must be overcome in the treatment of solid tumors. Furthermore, certain side effects associated with CAR T-cell therapy, including cytokine release syndrome, immune effector cell-related neurotoxicity syndrome, tumor lysis syndrome and on-target off-tumor toxicity, must be taken into consideration. The present study provides a systematic review of the principle, clinical application, current challenges, possible solutions and future perspectives for CAR T-cell therapy.
Collapse
Affiliation(s)
- Yuxi Luo
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China.,The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Guiqin Song
- Department of Biology, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| | - Shichu Liang
- The First Clinic of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Feifei Li
- Department of Pathophysiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Kang Liu
- Institute of Tissue Engineering and Stem Cells, Nanchong Central Hospital, The Second Clinical Medical College, North Sichuan Medical College, Nanchong, Sichuan 637000, P.R. China
| |
Collapse
|
37
|
Abstract
Chimeric antigen receptor (CAR) T-cell therapy is a revolutionary addition to the burgeoning field of immunotherapy. CAR T-cells are engineered by combining a T-cell receptor with the antigen-binding site of an immunoglobulin that allows the hybrid cell to target antigens of interest. CAR T-cell therapy has been approved to treat various hematologic malignancies, including relapsed or refractory B-cell acute lymphoblastic leukemia and diffuse large B-cell lymphoma. While the treatment efficacy is exciting, challenges remain in understanding the unique spectrum of adverse effects of CAR T-cell therapy, including cytokine release syndrome and neurotoxicity. Innovative research is underway to expand this therapy into solid tumors and fields beyond hematology and oncology. To date, there has been limited research into ophthalmic uses and considerations of CAR T-cell therapy. This review focuses on preclinical investigations into CAR T-cell therapy for retinoblastoma and uveal melanoma, as well as ophthalmic complications of CAR T-cell therapy.
Collapse
Affiliation(s)
- Kevin D Chodnicki
- Massachusetts Eye and Ear, Department of Ophthalmology, Boston, MA, USA.,Harvard Medical School, Boston, MA, USA
| | - Sashank Prasad
- Harvard Medical School, Boston, MA, USA.,Brigham and Women's Hospital, Department of Neurology, Boston, MA, USA
| |
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
|
Sarkar E, Khan A. Erratic journey of CRISPR/Cas9 in oncology from bench-work to successful-clinical therapy. Cancer Treat Res Commun 2021; 27:100289. [PMID: 33667951 DOI: 10.1016/j.ctarc.2020.100289] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2020] [Revised: 12/02/2020] [Accepted: 12/15/2020] [Indexed: 06/12/2023]
Abstract
CRISPR is a customized molecular scissor, comprising genetic guide made of RNA and an enzyme, Cas9 which snips DNA in simpler, cheaper and more precise way than any other gene editing tools. In recent years CRISPR/Cas has taken the research world by storm being go-to genome editor for potential gene therapy to fix cancer as well as several hereditary disorders. This review explores the literature around the mechanism of Nobel winning CRISPR/Cas9 and its journey from its discovery to various pre-clinical and clinical trials in oncology, focusing mostly on PD-1 knockout CAR-T cell therapy. It also discusses the hurdles and ethical dispute associated with CRISPR, such as unintended on-target and off-target cuts, embryonic germ-line editing. Despite the controversies regarding the safety of this technique, many studies reported promising results on targeting cancer and other diseases using CRISPR/Cas9. Outcomes from the first successful clinical trial showed the beneficial long term effect on genetically modified T-cells in targeting cancer cells which opens the door for CRISPR to be the most preferred technique to help treating cancer and other diseases in future. As far as germ-line editing is concerned, further studies are needed to support the safety of this technique in humans fixing genetic disorders and mutations. Therefore till date only somatic cell editing is ethically approved.
Collapse
Affiliation(s)
- Esha Sarkar
- Department of Biochemistry, Era's Lucknow Medical College and hospital, Era University, Lucknow, Uttar Pradesh, India
| | - Afreen Khan
- Department of Biochemistry, Era's Lucknow Medical College and hospital, Era University, Lucknow, Uttar Pradesh, India.
| |
Collapse
|
40
|
Fang Z, Jiang C, Li S. The Potential Regulatory Roles of Circular RNAs in Tumor Immunology and Immunotherapy. Front Immunol 2021; 11:617583. [PMID: 33613544 PMCID: PMC7886782 DOI: 10.3389/fimmu.2020.617583] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2020] [Accepted: 12/14/2020] [Indexed: 12/13/2022] Open
Abstract
Circular RNAs (circRNAs) are covalently closed RNA molecules in eukaryotes with features of high stability, tissue-specific and cell-specific expression. According to their biogenesis, circRNAs are mainly classified into five types, i.e. exonic circRNAs (EciRNAs), exon-intron circRNAs (EIciRNAs), intronic RNAs (CiRNAs), fusion circRNAs (f-circRNAs), and read-through circRNAs (rt-circRNAs). CircRNAs have been emerging as important non-coding regulatory RNAs in a variety of human cancers. CircRNA4s were revealed to exert regulatory function through multiple mechanisms, such as sponges/decoys of miRNAs and proteins, enhancers of protein functions, protein scaffolds, protein recruitment, or protein translation templates. Furthermore, some circRNAs are intensively associated with immune cells in tumor immune microenvironment (TIME), e.g. circARSP91 and natural killer cells. Through regulating immune checkpoint genes, circRNAs are demonstrated to modulate the immune checkpoint blockade immunotherapy, e.g. circCPA4 could up-regulate PD-L1 expression. In summary, we reviewed the molecular features of circRNAs and mechanisms how they exert functions. We further summarized functional implications of circRNA regulations in tumor immunology and immunotherapy. Further understanding of the regulatory roles of circRNAs in tumor immunology and immunotherapy will benefit tumor treatment.
Collapse
Affiliation(s)
- Zhixiao Fang
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chunjie Jiang
- Institute for Diabetes, Obesity, and Metabolism, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
- Division of Endocrinology, Diabetes, and Metabolism, Department of Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, United States
| | - Shengli Li
- Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
41
|
Jin KT, Chen B, Liu YY, Lan HUR, Yan JP. Monoclonal antibodies and chimeric antigen receptor (CAR) T cells in the treatment of colorectal cancer. Cancer Cell Int 2021; 21:83. [PMID: 33522929 PMCID: PMC7851946 DOI: 10.1186/s12935-021-01763-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 01/06/2021] [Indexed: 02/07/2023] Open
Abstract
Colorectal cancer (CRC) is the third most common cancer and the second leading cause of cancer deaths worldwide. Besides common therapeutic approaches, such as surgery, chemotherapy, and radiotherapy, novel therapeutic approaches, including immunotherapy, have been an advent in CRC treatment. The immunotherapy approaches try to elicit patients` immune responses against tumor cells to eradicate the tumor. Monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR) T cells are two branches of cancer immunotherapy. MAbs demonstrate the great ability to completely recognize cancer cell-surface receptors and blockade proliferative or inhibitory pathways. On the other hand, T cell activation by genetically engineered CAR receptor via the TCR/CD3 and costimulatory domains can induce potent immune responses against specific tumor-associated antigens (TAAs). Both of these approaches have beneficial anti-tumor effects on CRC. Herein, we review the different mAbs against various pathways and their applications in clinical trials, the different types of CAR-T cells, various specific CAR-T cells against TAAs, and their clinical use in CRC treatment.
Collapse
Affiliation(s)
- Ke-Tao Jin
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - Bo Chen
- Department of Neurology, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, Hangzhou, 310014, China
| | - Yu-Yao Liu
- Department of Colorectal Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - H Uan-Rong Lan
- Department of Breast and Thyroid Surgery, Affiliated Jinhua Hosptial, Zhejiang University School of Medicine, Zhejiang Province, Jinhua, 312000, P.R. China
| | - Jie-Ping Yan
- Department of Pharmacy, Zhejiang Provincial People's Hospital, People's Hospital of Hangzhou Medical College, No. 158 Shangtang Road, Hangzhou, 310014, China.
| |
Collapse
|
42
|
Bansal D, Reimers MA, Knoche EM, Pachynski RK. Immunotherapy and Immunotherapy Combinations in Metastatic Castration-Resistant Prostate Cancer. Cancers (Basel) 2021; 13:cancers13020334. [PMID: 33477569 PMCID: PMC7831137 DOI: 10.3390/cancers13020334] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2020] [Revised: 01/01/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022] Open
Abstract
Although most prostate cancers are localized, and the majority are curable, recurrences occur in approximately 35% of men. Among patients with prostate-specific antigen (PSA) recurrence and PSA doubling time (PSADT) less than 15 months after radical prostatectomy, prostate cancer accounted for approximately 90% of the deaths by 15 years after recurrence. An immunosuppressive tumor microenvironment (TME) and impaired cellular immunity are likely largely responsible for the limited utility of checkpoint inhibitors (CPIs) in advanced prostate cancer compared with other tumor types. Thus, for immunologically "cold" malignancies such as prostate cancer, clinical trial development has pivoted towards novel approaches to enhance immune responses. Numerous clinical trials are currently evaluating combination immunomodulatory strategies incorporating vaccine-based therapies, checkpoint inhibitors, and chimeric antigen receptor (CAR) T cells. Other trials evaluate the efficacy and safety of these immunomodulatory agents' combinations with standard approaches such as androgen deprivation therapy (ADT), taxane-based chemotherapy, radiotherapy, and targeted therapies such as tyrosine kinase inhibitors (TKI) and poly ADP ribose polymerase (PARP) inhibitors. Here, we will review promising immunotherapies in development and ongoing trials for metastatic castration-resistant prostate cancer (mCRPC). These novel trials will build on past experiences and promise to usher a new era to treat patients with mCRPC.
Collapse
|
43
|
Guo C, Dong E, Lai Q, Zhou S, Zhang G, Wu M, Yue X, Tao Y, Peng Y, Ali J, Lu Y, Fu Y, Lai W, Zhang Z, Ma F, Yao Y, Gou L, Yang H, Yang J. Effective antitumor activity of 5T4-specific CAR-T cells against ovarian cancer cells in vitro and xenotransplanted tumors in vivo. MedComm (Beijing) 2020; 1:338-350. [PMID: 34766126 PMCID: PMC8491242 DOI: 10.1002/mco2.34] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 02/05/2023] Open
Abstract
Ovarian cancer is considered to be the most lethal gynecologic malignancy, and despite the development of conventional therapies and new therapeutic approaches, the patient's survival time remains short because of tumor recurrence and metastasis. Therefore, effective methods to control tumor progression are urgently needed. The oncofetal tumor-associated antigen 5T4 (trophoblast glycoprotein, TPBG) represents an appealing target for adoptive T-cell immunotherapy as it is highly expressed on the surface of various tumor cells, has very limited expression in normal tissues, and spreads widely in malignant tumors throughout their development. In this study, we generated second-generation human chimeric antigen receptor (CAR) T cells with redirected specificity to 5T4 (5T4 CAR-T) and demonstrated that these CAR-T cells can elicit lytic cytotoxicity in targeted tumor cells, in addition to the secretion of cytotoxic cytokines, including IFN-γ, IL-2, and GM-CSF. Furthermore, adoptive transfer of 5T4 CAR-T cells significantly delayed tumor formation in xenografts of peritoneal and subcutaneous animal models. These results demonstrate the potential efficacy and feasibility of 5T4 CAR-T cell immunotherapy and provide a theoretical basis for the clinical study of future immunotherapies targeting 5T4 for ovarian cancer.
Collapse
Affiliation(s)
- Cuiyu Guo
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - E Dong
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Qinhuai Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Shijie Zhou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Guangbing Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Mengdan Wu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Xiaozhu Yue
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yiran Tao
- West China‐California Research Center for Predictive Intervention MedicineWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yujia Peng
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Jamel Ali
- Department of Chemical and Biomedical EngineeringFAMU‐FSU College of EngineeringTallahasseeFlorida
| | - Ying Lu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yuyin Fu
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Weirong Lai
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Zhixiong Zhang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Fanxin Ma
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Yuqin Yao
- Healthy Food Evaluation Research Center/Sichuan UniversityWest China School of Public Health and West China Fourth HospitalChengduPeople's Republic of China
| | - Lantu Gou
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Hanshuo Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| | - Jinliang Yang
- State Key Laboratory of Biotherapy and Cancer Center/Collaborative Innovation Center for BiotherapyWest China HospitalSichuan UniversityChengduSichuanPeople's Republic of China
| |
Collapse
|
44
|
Venetis K, Invernizzi M, Sajjadi E, Curigliano G, Fusco N. Cellular immunotherapy in breast cancer: The quest for consistent biomarkers. Cancer Treat Rev 2020; 90:102089. [PMID: 32889360 DOI: 10.1016/j.ctrv.2020.102089] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Revised: 08/06/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Breast cancer is the most common malignancy in women worldwide, with a relatively high proportion of patients experiencing resistance to standard treatments. Cellular immunotherapy (CI), which is based on the extraction, modification, and re-infusion of the patient's immune cells, is showing promising results in these patients. Among CI possible approaches, adoptive cell therapy (ACT) and dendritic cell (DC) vaccination are the most comprehensively explored in both primary/translational research studies and clinical trials. ACT may include the use of tumor-infiltrating lymphocytes (TILs), T cell receptor (TCR)-, or chimeric antigen receptor (CAR)-engineered T-cells. There are indications suggesting that a biomarker-based approach might be beneficial in effectively selecting breast cancer patients for CI. Here, we sought to provide the current knowledge of CI in breast cancer, focusing on candidate biomarkers, ongoing clinical trials, limitations, and immediate future perspectives.
Collapse
Affiliation(s)
- Konstantinos Venetis
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Ph.D. Program in Translational Medicine, University of Milan, 20133 Milan, Italy; Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Marco Invernizzi
- Department of Health Sciences, University of Eastern Piedmont, 28100 Novara, Italy
| | - Elham Sajjadi
- Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Giuseppe Curigliano
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of Early Drug Development for Innovative Therapies, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy.
| | - Nicola Fusco
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy; Division of Pathology and Laboratory Medicine, IEO, European Institute of Oncology IRCCS, 20141 Milan, Italy.
| |
Collapse
|
45
|
Yuan X, Sun Z, Yuan Q, Hou W, Liang Q, Wang Y, Mo W, Wang H, Yu M. Dual-function chimeric antigen receptor T cells targeting c-Met and PD-1 exhibit potent anti-tumor efficacy in solid tumors. Invest New Drugs 2020; 39:34-51. [PMID: 32772342 DOI: 10.1007/s10637-020-00978-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Accepted: 07/29/2020] [Indexed: 12/31/2022]
Abstract
Purpose Programmed cell death 1 (PD-1), which is upregulated under the continuous induction of the tumor microenvironment, causes chimeric antigen receptor (CAR)-T cell hypofunction via interaction with programmed death ligand 1 (PD-L1). This study aimed to construct CAR-T cells that are resistant to PD-1 inhibition to improve the effect of CAR-T cells in solid tumors. Methods We constructed a type of dual-function CAR-T cell that targets tumor-associated antigen c-Met and blocks the binding of PD-1 with PD-L1. The expression of c-Met, PD-L1, and inhibitory receptors was measured using flow cytometry. The cytotoxicity, cytokine release, and differentiation level of CAR-T cells were determined using lactate dehydrogenase release assay, enzyme-linked immunosorbent assay, and flow cytometry, respectively. The levels of p-Akt, p-MAPK, caspase-3, and Bcl2 were detected by western blot. The in vivo anti-tumor effect was evaluated using tumor xenograft models. Results Dual-function CAR-T cells could mediate enhanced active signals upon encountering target antigens and had targeted cytotoxicity to target cells. However, the cytotoxicity of c-Met-CAR-PD-1+ T cells was impaired due to the interaction of PD-1 with PD-L1. By blocking the binding of PD-1 and PD-L1, the novel dual-function CAR-PD-1+ T cells could maintain cytotoxicity to PD-L1+ tumor cells. In tumor tissue, the dual-function CAR-T cells showed lower inhibitory receptor expression and lower differentiation characteristics, which resulted in potent anti-tumor effects and prolonged survival in PD-L1+ tumor xenograft models compared to single-target CAR-T cells. Conclusion These results confirm that the novel dual-function CAR-T cells exhibit stronger anti-tumor activity against solid tumors than traditional single-target CAR-T cells and present a new approach that enhance the activity of CAR-T cells in solid tumors.
Collapse
Affiliation(s)
- Xingxing Yuan
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China
| | - Zujun Sun
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China.,Department of Clinical Laboratory, Shanghai Tongji Hospital, Tongji University School of Medicine, Shanghai, 200065, China
| | - Qingyun Yuan
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China
| | - Weihua Hou
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China
| | - Qiaoyan Liang
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China
| | - Yuxiong Wang
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China
| | - Wei Mo
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China
| | - Huijie Wang
- Department of Medical Oncology, Fudan University Shanghai Cancer Center; Department of Oncology, Shanghai Medical College, Fudan University, No. 270 Dong An Road, Shanghai, China.
| | - Min Yu
- The Key Laboratory of Metabolism and Molecular Medicine, The Ministry of Education and the Department of Biochemistry and Molecular Biology, School of Basic Medicine, Fudan University, P.O. Box #238, No. 138 Yi Xue Yuan Road, Shanghai, China.
| |
Collapse
|
46
|
Jo Y, Ali LA, Shim JA, Lee BH, Hong C. Innovative CAR-T Cell Therapy for Solid Tumor; Current Duel between CAR-T Spear and Tumor Shield. Cancers (Basel) 2020; 12:cancers12082087. [PMID: 32731404 PMCID: PMC7464778 DOI: 10.3390/cancers12082087] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Revised: 07/21/2020] [Accepted: 07/23/2020] [Indexed: 12/12/2022] Open
Abstract
Novel engineered T cells containing chimeric antigen receptors (CAR-T cells) that combine the benefits of antigen recognition and T cell response have been developed, and their effect in the anti-tumor immunotherapy of patients with relapsed/refractory leukemia has been dramatic. Thus, CAR-T cell immunotherapy is rapidly emerging as a new therapy. However, it has limitations that prevent consistency in therapeutic effects in solid tumors, which accounts for over 90% of all cancer patients. Here, we review the literature regarding various obstacles to CAR-T cell immunotherapy for solid tumors, including those that cause CAR-T cell dysfunction in the immunosuppressive tumor microenvironment, such as reactive oxygen species, pH, O2, immunosuppressive cells, cytokines, and metabolites, as well as those that impair cell trafficking into the tumor microenvironment. Next-generation CAR-T cell therapy is currently undergoing clinical trials to overcome these challenges. Therefore, novel approaches to address the challenges faced by CAR-T cell immunotherapy in solid tumors are also discussed here.
Collapse
Affiliation(s)
- Yuna Jo
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Laraib Amir Ali
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Ju A Shim
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
| | - Byung Ha Lee
- NeoImmuneTech, Inc., 2400 Research Blvd., Suite 250, Rockville, MD 20850, USA;
| | - Changwan Hong
- Department of Anatomy, Pusan National University School of Medicine, Yangsan 50612, Korea; (Y.J.); (L.A.A.); (J.A.S.)
- Correspondence: ; Tel.: +82-51-510-8041
| |
Collapse
|
47
|
Feng D, Sun J. Overview of anti-BCMA CAR-T immunotherapy for multiple myeloma and relapsed/refractory multiple myeloma. Scand J Immunol 2020; 92:e12910. [PMID: 32471019 DOI: 10.1111/sji.12910] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/23/2020] [Accepted: 05/19/2020] [Indexed: 12/14/2022]
Abstract
Multiple myeloma (MM) is a haematological malignancy caused by malignant proliferation of plasma cells in bone marrow. In recent years, MM patients are commonly treated with chemotherapy, autologous stem cell transplantation, protease inhibitors, immunomodulatory drugs and monoclonal antibodies, however most patients eventually relapse. Therefore, more effective therapies are highly needed. Anti-BCMA CAR-T therapy, a novel and efficacious method for treating MM and relapsed/refractory multiple myeloma (RRMM), has been designed and applied in clinics. The CAR-T can specifically recognize the targeted molecule B cell maturation antigen (BCMA) and kill MM cells expressing BCMA and several clinical trials have revealed high response rates in the therapy. Herein, we summarize the developments, the current design and clinical trials, the side effects of anti-BCMA CAR-T therapy and comparison of it with other CAR-T therapies.
Collapse
Affiliation(s)
- Deming Feng
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| | - Jian Sun
- Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Science and Technology, Tianjin University, Tianjin, China
| |
Collapse
|
48
|
Roncati L, Palmieri B. Adoptive cell transfer (ACT) of autologous tumor-infiltrating lymphocytes (TILs) to treat malignant melanoma: the dawn of a chimeric antigen receptor T (CAR-T) cell therapy from autologous donor. Int J Dermatol 2020; 59:763-769. [PMID: 32441324 DOI: 10.1111/ijd.14945] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2019] [Revised: 03/02/2020] [Accepted: 04/21/2020] [Indexed: 12/18/2022]
Abstract
BACKGROUND Tumor-infiltrating lymphocytes (TILs) are B, T-helper, and T-cytotoxic lymphocytes migrated from the blood or lymph stream toward a tumor with the aim to infiltrate and destroy it. They can be histologically graded as brisk, nonbrisk, or absent. Malignant melanoma has been the first malignancy found to be correlated with TILs status, being brisk TILs associated with better clinical outcomes. By the terminology of "adoptive cell transfer" (ACT), the medical oncology refers to the transfer of cells in a tumor-bearing patient from the same recipient or a healthy donor. METHODS A PubMed literature search on the topic has been performed. Additional documents known to the authors and identified from the reference list of cited publications have been included. RESULTS In the past, autologous TILs ACT was successfully tested for the treatment of malignant melanoma and, today, it is a standardized procedure in several centers around the world. It represents the first research step toward the bioengineered chimeric antigen receptor T (CAR-T) cell therapy from autologous donor. CONCLUSIONS Both autologous TILs ACT and CAR-T cell therapy from autologous donor exploit the anticancer power of targeted self-lymphocytes, but CAR-T cell technology also virtually allows treatment of those melanomas devoid of TILs or with so few cytotoxic TILs that are difficult to identify.
Collapse
Affiliation(s)
- Luca Roncati
- Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
| | - Beniamino Palmieri
- Department of Medical and Surgical Sciences, University Hospital of Modena, Modena (MO), Italy
| |
Collapse
|
49
|
Yang M, Liu B, Wang Y, Liu Y, Gong X, Gong B, Xu Y, Mi Y, Wang M, Wang J. Chimeric antigen receptor-modified T-cell therapy for bone marrow and skin relapse Philadelphia chromosome-like acute lymphoblastic leukemia: A case report. Medicine (Baltimore) 2020; 99:e18639. [PMID: 32358340 PMCID: PMC7440224 DOI: 10.1097/md.0000000000018639] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
RATIONALE Chimeric antigen receptor-modified T-cell (CART) therapy has revolutionized the treatment of patients with relapsed or refractory B-cell acute lymphoblastic leukemia (ALL). However, the capacity of CART therapy has not yet been fully elucidated. PATIENT CONCERNS An 18-year-old Chinese male patient presented with multiple firm masses on the skin all over his body following regular chemotherapy. DIAGNOSES Bone marrow smear and skin biopsy confirmed that it was a bone marrow and skin relapse from the initial B-cell ALL. INTERVENTIONS CD19 CART-cell therapy was performed to manage the bone marrow and skin of the relapsed B-cell ALL. OUTCOMES During CART-cell therapy, cytokine release syndrome and central nervous encephalopathy occurred. Eventually, the lesions disappeared, and the bone marrow and skin tested minimal residual disease (MRD) negative. The patient achieved complete remission (CR). Fourteen days after testing MRD negative, he received allogeneic hematopoietic stem-cell transplantation and has remained disease free to date. LESSONS The CR of this patient with leukemia cutis demonstrated that CART exhibited efficacy in this case. While further research is still required, this treatment could potentially be used as a therapy for skin leukemia, lymphoma, and other primary skin cancers.
Collapse
|
50
|
Xu X, Zhao W, Yue Z, Qin M, Jin M, Chang L, Ma X. 4SCAR-GD2-modified T-cell therapy in neuroblastoma with MYCN amplification: A case report with over 4-year follow-up data. Pediatr Investig 2020; 4:55-58. [PMID: 32851343 PMCID: PMC7331334 DOI: 10.1002/ped4.12181] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Accepted: 06/11/2019] [Indexed: 02/03/2023] Open
Abstract
INTRODUCTION Neuroblastoma (NB) is the most common extracranial solid tumor among children. The 5-year event-free survival rate for high-risk (HR) NB is still poor, especially for patients with advanced NB with MYCN gene amplification. Chimeric antigen receptor T (CAR-T) cell therapy is a new treatment for HR-NB. CASE PRESENTATION A 55-month-old boy with stage IV HR-NB received 4th-generation CAR-T cells that target disialoganglioside GD2, as consolidation maintenance treatment after intensive chemotherapy, surgery, and autologous stem-cell transplantation. As of February 2019, his CAR-T follow-up time was 37.5 months, indicating prolonged survival. Cranial MRI and ultrasound showed no mass; 123I-metaiodobenzylguanidine (123I-MIBG) scan was negative. CONCLUSION GD2-CAR-T cells may be an effective treatment option for NB patients with MYCN amplification.
Collapse
Affiliation(s)
- Xiao Xu
- Beijing Key Laboratory of Pediatric Hematology Oncology; National Discipline of Pediatrics, Ministry of Education; MOE Key Laboratory of Major Diseases in Children; Hematology Oncology Center, Beijing Children’s HospitalCapital Medical University, National Center for Children’s HealthBeijing100045China
| | - Wen Zhao
- Beijing Key Laboratory of Pediatric Hematology Oncology; National Discipline of Pediatrics, Ministry of Education; MOE Key Laboratory of Major Diseases in Children; Hematology Oncology Center, Beijing Children’s HospitalCapital Medical University, National Center for Children’s HealthBeijing100045China
| | - Zhixia Yue
- Beijing Key Laboratory of Pediatric Hematology Oncology; National Discipline of Pediatrics, Ministry of Education; MOE Key Laboratory of Major Diseases in Children; Hematology Oncology Center, Beijing Children’s HospitalCapital Medical University, National Center for Children’s HealthBeijing100045China
| | - Maoquan Qin
- Beijing Key Laboratory of Pediatric Hematology Oncology; National Discipline of Pediatrics, Ministry of Education; MOE Key Laboratory of Major Diseases in Children; Hematology Oncology Center, Beijing Children’s HospitalCapital Medical University, National Center for Children’s HealthBeijing100045China
| | - Mei Jin
- Beijing Key Laboratory of Pediatric Hematology Oncology; National Discipline of Pediatrics, Ministry of Education; MOE Key Laboratory of Major Diseases in Children; Hematology Oncology Center, Beijing Children’s HospitalCapital Medical University, National Center for Children’s HealthBeijing100045China
| | - Lung‐Ji Chang
- Shenzhen Geno‐immune Medical Institute and America Yuva Biomed IncShenzhenChina
| | - Xiaoli Ma
- Beijing Key Laboratory of Pediatric Hematology Oncology; National Discipline of Pediatrics, Ministry of Education; MOE Key Laboratory of Major Diseases in Children; Hematology Oncology Center, Beijing Children’s HospitalCapital Medical University, National Center for Children’s HealthBeijing100045China
| |
Collapse
|