1
|
Tao Y, Wang S, Li X, Jin L, Liu C, Jiao K, Li X, Cheng Y, Xu K, Zhou X, Wei X. Identification of disulfidptosis-related genes and subgroups in spinal cord injury. Spinal Cord 2025:10.1038/s41393-025-01081-1. [PMID: 40319145 DOI: 10.1038/s41393-025-01081-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 04/03/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
STUDY DESIGN Bioinformatics analysis and experimental validation study. OBJECTIVES To investigate the role and expression patterns of disulfidptosis-related genes in spinal cord injury (SCI), identify potential pivotal genes, and explore possible therapeutic targets. SETTING Shanghai, China. METHODS Data acquisition and pre-processing: Screened 27 disulfidptosis-related genes based on literature and downloaded RNA-sequencing data of ASCI patients from GEO database (GSE151371); Identification of differentially expressed genes (DEGs): Used R package "limma" for differential gene expression analysis between ASCI samples and normal controls; Evaluating immune cell infiltration: Employed ssGSEA algorithm and CIBERSORT to determine immune cell abundance; Identification and functional verification of key genes: Intersected disulfidptosis-related genes with DEGs, and used machine learning techniques (Random Forest, Lasso, Support Vector Machine) to identify hub genes. Validated hub genes expression by real-time PCR; Construction of a diagnostic model: Developed a backpropagation neural network clinical prediction model based on hub genes and clinical features, and evaluated its performance using ROC curve. 6. Subcluster analysis: Performed consensus cluster analysis of ASCI samples and hub genes, and used GSVA to elucidate functional differences between subgroups. RESULTS Identified 7764 DEGs in ASCI, with GO and KEGG enrichment in inflammation and autophagy-related pathways; Found differences in immune cell infiltration between ASCI and control groups, and correlation between immune cells and DRGs; Determined seven hub genes (MYL6, NUBPL, CYFIP1, IQGAP1, FLNB, SLC7A11, CD2AP) through machine learning; Validated the expression of hub genes by qRT-PCR; Constructed a clinical diagnostic model with good predictive accuracy (overall dataset accuracy of 83.3%); Identified two subtypes of ASCI based on hub genes, with different immune infiltration and pathway activity. CONCLUSION Disulfidptosis is closely related to spinal cord injury. The identified hub genes and subtypes provide new insights for biomarker and therapeutic target research. The diagnostic model has potential for clinical application, but further studies are needed due to limitations such as small sample size. SPONSORSHIP This study was supported in part by the project of Youth Scientific and Technological Talents of PLA (2020QN06125), Changhong Talent Project in First affiliated hospital of Navy Medical University (Wei Xianzhao) and Basic Medical Research Project in First affiliated hospital of Navy Medical University (2023PY17). I want to reiterate that there is no prior publication of figures or tables and no conflict of interest in the submission of this manuscript. The graphical abstract is divided into two parts. The upper section sequentially illustrates the occurrence of disulfidptosis and changes in the immune microenvironment in the human body after SCI. The lower section displays the construction of a diagnostic model for SCI through the detection of changes in disulfidptosis-related genes, combined with patient clinical information.
Collapse
Affiliation(s)
- Ye Tao
- Naval Medical University, Shanghai, China
| | | | - Xiongfei Li
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China
| | - Letian Jin
- Hangzhou Medical College, Hangzhou, China
| | - Chen Liu
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China
| | - Kun Jiao
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China
| | - Xiaoyu Li
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China
| | - Yajun Cheng
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China
| | - Kehan Xu
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China.
| | - Xiaoyi Zhou
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China.
| | - Xianzhao Wei
- Department of Orthopaedic Surgery, Changhai Hospital, Shanghai, China.
| |
Collapse
|
2
|
Wang L, Li J, Xuan Y, Zhang J, Wang X, Hu W, Xiu L. Prospects for γδ T cells and chimeric antigen receptor γδ T cells in cancer immunotherapy. Front Immunol 2025; 16:1554541. [PMID: 40370457 PMCID: PMC12075525 DOI: 10.3389/fimmu.2025.1554541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 04/08/2025] [Indexed: 05/16/2025] Open
Abstract
γδ T cells, a type of specialized T cell, differ from alpha-beta T cells due to the presence of γ and δ chain surface T cell receptors. These receptors allow them to directly recognize and bind antigenic molecules without the requirement of attachment to MHC or APC antigen presentation. Given their intrinsic properties and functional versatility, γδ T cells are under intensive investigation as carriers for chimeric antigen receptor (CAR) in the context of cancer therapy. In this regard, γδ CAR-T cells have demonstrated great potential to overcome the limitations of antigen recognition with the help of dual antigen identification mechanisms. However, there are still technological challenges that need to be addressed. This discussion focuses on the research status and future development prospects of γδ T cells and γδ CAR-T cells, aiming to provide valuable insights for the follow-up research and practical application of γδ CAR-T cells.
Collapse
MESH Headings
- Humans
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Neoplasms/therapy
- Neoplasms/immunology
- Immunotherapy, Adoptive/methods
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Receptors, Chimeric Antigen/metabolism
- Animals
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Antigens, Neoplasm/immunology
Collapse
Affiliation(s)
- Lu Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jiaqi Li
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Yaping Xuan
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Jinrui Zhang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Xiao Wang
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| | - Wei Hu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
- State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Department of Infectious Diseases, Shanghai Key Laboratory of Infectious Diseases and Biosafety Emergency Response, National Medical Center for Infectious Diseases, Huashan Hospital, Fudan University, Shanghai, China
| | - Lei Xiu
- State Key Laboratory of Reproductive Regulation and Breeding of Grassland Livestock, Institutes of Biomedical Sciences, School of Life Sciences, Inner Mongolia University, Hohhot, China
| |
Collapse
|
3
|
Zhan C, Peng C, Wei H, Wei K, Ou Y, Zhang Z. Diverse Subsets of γδT Cells and Their Specific Functions Across Liver Diseases. Int J Mol Sci 2025; 26:2778. [PMID: 40141420 PMCID: PMC11943347 DOI: 10.3390/ijms26062778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/15/2025] [Accepted: 03/17/2025] [Indexed: 03/28/2025] Open
Abstract
γδT cells, a distinct group of T lymphocytes, serve as a link between innate and adaptive immune responses. They are pivotal in the pathogenesis of various liver disorders, such as viral hepatitis, nonalcoholic fatty liver disease (NAFLD), alcoholic liver disease (ALD), liver fibrosis, autoimmune liver diseases, and hepatocellular carcinoma (HCC). Despite their importance, the functional diversity and regulatory mechanisms of γδT cells remain incompletely understood. Recent advances in high-throughput single-cell sequencing and spatial transcriptomics have revealed significant heterogeneity among γδT cell subsets, particularly Vδ1+ and Vδ2+, which exhibit distinct immunological roles. Vδ1+ T cells are mainly tissue-resident and contribute to tumor immunity and chronic inflammation, while Vδ2+ T cells, predominantly found in peripheral blood, play roles in systemic immune surveillance but may undergo dysfunction in chronic liver diseases. Additionally, γδT17 cells exacerbate inflammation in NAFLD and ALD, whereas IFN-γ-secreting γδT cells contribute to antiviral and antifibrotic responses. These discoveries have laid the foundation for the creation of innovative solutions. γδT cell-based immunotherapeutic approaches, such as adoptive cell transfer, immune checkpoint inhibition, and strategies targeting metabolic pathways. Future research should focus on harnessing γδT cells' therapeutic potential through targeted interventions, offering promising prospects for precision immunotherapy in liver diseases.
Collapse
Affiliation(s)
- Chenjie Zhan
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Chunxiu Peng
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Huaxiu Wei
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Ke Wei
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Yangzhi Ou
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
| | - Zhiyong Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Bio-Targeting Theranostics, Guangxi Key Laboratory of Bio-Targeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning 530021, China; (C.Z.); (C.P.)
- Department of Surgery, Robert-Wood-Johnson Medical School University Hospital, Rutgers University, New Brunswick, NJ 08901-8554, USA
| |
Collapse
|
4
|
Yan Y, Zhang Y, Tang X, Zhuoya Z, Linyu G, Lingyun S. Vγ6 +γδT Cells Participate in Lupus Nephritis in MRL/Lpr Mice. Int J Rheum Dis 2025; 28:e70040. [PMID: 39740062 DOI: 10.1111/1756-185x.70040] [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: 07/16/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND γδT cells have been implicated in the pathogenesis of autoimmune diseases. The study aims to investigate the abundance of γδT cells in MRL/lpr mice. METHODS MRL/lpr mice were used as lupus models, while C3H/HeJ mice served as normal controls. The abundance of γδT cells in different organs was examined by flow cytometry. Plasma double-stranded DNA antibody levels, blood urea nitrogen, creatinine, and urinary protein levels were measured. Renal histopathology was observed via H&E staining. The correlations between the abundance of γδT cells and lupus manifestations were analyzed. RESULTS Compared with C3H/HeJ mice, the number of γδT cells and Vγ6+γδT cell subset in the peripheral blood of MRL/lpr mice was significantly reduced. However, in the kidney, the number of γδT cells and Vγ6+γδT cell subset was significantly increased. Additionally, the number of Vγ6+γδT cells in the kidney was positively correlated with the urinary protein level. The number of IFN-γ+Vγ6+γδT cells in the kidney was positively correlated with urinary protein level. CONCLUSION In MRL/lpr mice, it is likely that peripheral γδT cells, especially the Vγ6 subset, infiltrate the kidney and secrete IFN-γ, which contributes to the development of lupus nephritis.
Collapse
Affiliation(s)
- Yunxia Yan
- Department of Rheumatology and Immunology, The Drum Tower Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Yue Zhang
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital Clinical College of Jiangsu University, Zhenjiang, China
| | - Xiaojun Tang
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Zhang Zhuoya
- Department of Rheumatology and Immunology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Geng Linyu
- Department of Rheumatology and Immunology, The Drum Tower Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| | - Sun Lingyun
- Department of Rheumatology and Immunology, The Drum Tower Clinical Medical School of Nanjing Medical University, Nanjing, Jiangsu, China
| |
Collapse
|
5
|
Liu K, Hoover AR, Wang L, Sun Y, Valerio TI, Furrer C, Adams J, Yang J, Li M, Chen WR. Localized ablative immunotherapy enhances antitumor immunity by modulating the transcriptome of tumor-infiltrating Gamma delta T cells. Cancer Lett 2024; 604:217267. [PMID: 39307410 PMCID: PMC11471373 DOI: 10.1016/j.canlet.2024.217267] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/10/2024] [Accepted: 09/18/2024] [Indexed: 09/26/2024]
Abstract
Gamma delta T cells (γδT cells) play crucial roles in the immune response against tumors, yet their functional dynamics under different cancer therapies remain poorly understood. Laser Ablative Immunotherapy (LAIT) is a novel cancer treatment modality combining local photothermal therapy (PTT) and intratumoral injection of an immunostimulant, N-dihydrogalactochitosan (glycated chitosan, GC). LAIT has been shown to induce systemic antitumor immune responses in pre-clinical studies and clinical trials, eradicating both treated local tumors and untreated distant metastases. In this study, we used LAIT to treat breast tumors in a mouse model and investigated the effects of LAIT on tumor-infiltrating γδT cells using single-cell RNA sequencing (scRNAseq). We characterized the γδT cells from tumors in control, PTT, GC, and LAIT (PTT + GC) groups, by identifying six distinct subtypes: activated, cytotoxic, cycling cytotoxic, IFN-enriched, antigen-presenting, and IL17-producing γδT cells. Differential gene expression analysis revealed that LAIT significantly upregulated genes associated with T cell activation, leukocyte adhesion, and interferon signaling in treated tumor tissues while downregulating genes involved in protein folding and stress responses. LAIT also uniquely increased the proportion of IL17-producing γδT cells, which correlated with prolonged survival in breast cancer patients, as analyzed using TCGA data. Furthermore, the transcriptomic profiles of γδT cells in LAIT-treated tumors closely resembled those in immune checkpoint inhibitor (ICI)-treated patients, suggesting potential synergistic effects. Our findings indicate that LAIT modulates the γδT cell transcriptome, enhancing their antitumor capabilities and providing a basis for combining LAIT with ICI therapy to improve cancer treatment outcomes.
Collapse
Affiliation(s)
- Kaili Liu
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Ashley R Hoover
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Arthritis and Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Lin Wang
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Yuanhong Sun
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Trisha I Valerio
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Coline Furrer
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jacob Adams
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA
| | - Jingxuan Yang
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Min Li
- Department of Medicine, Department of Surgery, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA
| | - Wei R Chen
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, OK, USA; Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
6
|
Feng X, Xu Y. The recent progress of γδ T cells and its targeted therapies in rheumatoid arthritis. Int J Rheum Dis 2024; 27:e15381. [PMID: 39467001 DOI: 10.1111/1756-185x.15381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Revised: 10/03/2024] [Accepted: 10/08/2024] [Indexed: 10/30/2024]
Abstract
Rheumatoid arthritis (RA) is an autoimmune condition that mostly impacts the joints. During the advanced phases of the disorder, it may be accompanied by other problems. While the precise cause of RA is uncertain, various research has been conducted to gain a better understanding of the immunological processes involved in the development of RA. T cells are acknowledged as significant contributors to the progression of RA because of their roles in cytokine secretion, antigen presentation, and facilitating B cells in the manufacture of antibodies. γδ T cells are a small subset of T cells that have significant functions in the context of infection and diseases linked with tumors. γδ T cells have been the subject of investigation in autoimmune disorders in recent years. This review focused on the involvement of γδ T lymphocytes in the development of RA. In this article, we provide an analysis of the immunological capabilities of γδ T cells, intending to comprehend their significance in RA, which could be pivotal in the creation of innovative clinical treatments.
Collapse
Affiliation(s)
- Xue Feng
- Department of Bone and Joint Surgery, The First Bethune Hospital of Jilin University, Changchun, China
| | - Yan Xu
- Department of Bone and Joint Surgery, The First Bethune Hospital of Jilin University, Changchun, China
| |
Collapse
|
7
|
Xia J, Wang C, Li B. Hepatocellular carcinoma cells induce γδ T cells through metabolic reprogramming into tumor-progressive subpopulation. Front Oncol 2024; 14:1451650. [PMID: 39309735 PMCID: PMC11412793 DOI: 10.3389/fonc.2024.1451650] [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: 06/21/2024] [Accepted: 08/14/2024] [Indexed: 09/11/2024] Open
Abstract
Tumor immune microenvironment (TIME) is a tiny structure that contains multiple immune cell components around tumor cells, which plays an important role in tumorigenesis, and is also the potential core area of activated immunotherapy. How immune cells with tumor-killing capacity in TIME are hijacked by tumor cells during the progression of tumorigenesis and transformed into subpopulations that facilitate cancer advancement is a question that needs to be urgently addressed nowadays. γδ T cells (their T cell receptors are composed of γ and δ chains), a unique T cell subpopulation distinguished from conventional αβ T cells, are involved in a variety of immune response processes through direct tumor-killing effects and/or indirectly influencing the activity of other immune cells. However, the presence of γδ T cells in the tumor microenvironment (TME) has been reported to be associated with poor prognosis in some tumors, suggesting that certain γδ T cell subsets may also have pro-tumorigenic effects. Recent studies have revealed that metabolic pathways such as activation of glycolysis, increase of lipid metabolism, enhancement of mitochondrial biosynthesis, alterations of fatty acid metabolism reshape the local TME, and immune cells trigger metabolic adaptation through metabolic reprogramming to meet their own needs and play the role of anti-tumor or immunosuppression. Combining previous studies and our bioinformatics results, we hypothesize that γδT cells compete for resources with hepatocellular carcinoma (HCC) cells by means of fatty acid metabolic regulation in the TME, which results in the weakening or loss of their ability to recognize and kill HCC cells through genetic and epigenetic alterations, thus allowing γδT cells to be hijacked by HCC cells as a subpopulation that promotes HCC progression.
Collapse
|
8
|
Gheitasi M, Safdel S, Kumar Patra S, Zandvakili R, Nemati M, Saha B, Jafarzadeh A. Generation of immune cells from induced pluripotent stem cells (iPSCs): Their potential for adoptive cell therapy. Hum Immunol 2024; 85:110836. [PMID: 38981248 DOI: 10.1016/j.humimm.2024.110836] [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: 12/01/2023] [Revised: 06/03/2024] [Accepted: 06/24/2024] [Indexed: 07/11/2024]
Abstract
Advances in human stem cell technologies enable induced pluripotent stem cells (iPSCs) to be explored as potent candidates for treating various diseases, such as malignancies, autoimmunity, immunodeficiencies, and allergic reactions. iPSCs with infinite self-renewal ability can be derived from different types of somatic cells without the ethical issues associated with embryonic stem cells. To date, numerous cell types, including various immune cell subsets [CD4+ and CD8+ T cells, gamma delta T (γδ T) cells, regulatory T cells, dendritic cells, natural killer cells, macrophages, and neutrophils] have successfully been generated from iPSCs paving the way for effective adoptive cell transfer therapy, drug development, and disease modeling. Herein, we review various iPSC-derived immune cells and their possible application in immunotherapy.
Collapse
Affiliation(s)
- Mahsa Gheitasi
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Sepeher Safdel
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | | | - Raziyeh Zandvakili
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Bhaskar Saha
- National Centre for Cell Science, Ganeshkhind, Pune 411007, India
| | - Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran; Applied Cellular and Molecular Research Center, Kerman University of Medical Sciences, Kerman, Iran.
| |
Collapse
|
9
|
Al Agrafi F, Gaballa A, Hahn P, Arruda LCM, Jaramillo AC, Witsen M, Lehmann S, Önfelt B, Uhlin M, Stikvoort A. Selective lysis of acute myeloid leukemia cells by CD34/CD3 bispecific antibody through the activation of γδ T-cells. Oncoimmunology 2024; 13:2379063. [PMID: 39076247 PMCID: PMC11285226 DOI: 10.1080/2162402x.2024.2379063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 07/05/2024] [Accepted: 07/08/2024] [Indexed: 07/31/2024] Open
Abstract
Despite the considerable progress in acute myeloid leukemia (AML) treatment, relapse after allogeneic hematopoietic stem cell transplantation (HSCT) is still frequent and associated with a poor prognosis. Relapse has been shown to be correlated with an incomplete eradication of CD34+ leukemic stem cells prior to HSCT. Previously, we have shown that a novel CD34-directed, bispecific T-cell engager (BTE) can efficiently redirect the T-cell effector function toward cancer cells, thus eliminating leukemic cells in vitro and in vivo. However, its impact on γδ T-cells is still unclear. In this study, we tested the efficacy of the CD34-specific BTE using in vitro expanded γδ T-cells as effectors. We showed that the BTEs bind to γδ T-cells and CD34+ leukemic cell lines and induce target cell killing in a dose-dependent manner. Additionally, γδ T-cell mediated killing was found to be superior to αβ T-cell mediated cytotoxicity. Furthermore, we observed that only in the presence of BTE the γδ T-cells induced primary AML blast killing in vitro. Importantly, our results show that γδ T-cells did not target the healthy CD34intermediate endothelial blood-brain barrier cell line (hCMEC/D3) nor lysed CD34+ HSCs from healthy bone marrow samples.
Collapse
MESH Headings
- Antibodies, Bispecific/immunology
- Antibody Specificity
- Lymphocyte Activation
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/pathology
- Humans
- Cell Line, Tumor
- Antigens, CD34/immunology
- CD3 Complex/immunology
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Cell Proliferation
- Receptors, Antigen, T-Cell, alpha-beta/immunology
- Cytokines/immunology
- Cell Death
- T-Lymphocytes/immunology
- Cytotoxicity, Immunologic
Collapse
Affiliation(s)
- Faisal Al Agrafi
- Healthy Aging Research Institute, King Abdulaziz City for Science and Technology (KACST), Riyadh, Kingdom of Saudi Arabia
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Ahmed Gaballa
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
- Department of Biochemistry and Molecular Biology, National Liver Institute, Menoufia University, Shebeen El-Kom, Egypt
| | - Paula Hahn
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Lucas C. M. Arruda
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Adrian C. Jaramillo
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Maartje Witsen
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| | - Sören Lehmann
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
- Department of Medical Sciences, Hematology, Uppsala University Hospital, Uppsala, Sweden
| | - Björn Önfelt
- Department of Applied Physics, Science for Life Laboratory, KTH Royal Institute of Technology, Stockholm, Sweden
| | - Michael Uhlin
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
- Department of Immunology and Transfusion Medicine, Karolinska University Hospital, Stockholm, Sweden
| | - Arwen Stikvoort
- Department of Medicine Huddinge, Karolinska Institutet, Center for Hematology and Regenerative Medicine, Stockholm, Sweden
| |
Collapse
|
10
|
Mehdikhani F, Bahar A, Bashi M, Mohammadlou M, Yousefi B. From immunomodulation to therapeutic prospects: Unveiling the biology of butyrophilins in cancer. Cell Biochem Funct 2024; 42:e4081. [PMID: 38934382 DOI: 10.1002/cbf.4081] [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: 04/09/2024] [Revised: 06/12/2024] [Accepted: 06/17/2024] [Indexed: 06/28/2024]
Abstract
Butyrophilin (BTN) proteins are a type of membrane protein that belongs to the Ig superfamily. They exhibit a high degree of structural similarity to molecules in the B7 family. They fulfill a complex function in regulating immune responses, including immunomodulatory roles, as they influence γδ T cells. The biology of BTN molecules indicates that they are capable of inhibiting the immune system's ability to detect antigens within tumors. A dynamic association between BTN molecules and cellular surfaces is also recognized in specific contexts, influencing their biology. Notably, the dynamism of BTN3A1 is associated with the immunosuppression of T cells or the activation of Vγ9Vδ2 T cells. Cancer immunotherapy relies heavily on T cells to modulate immune function within the intricate interaction of the tumor microenvironment (TME). A significant interaction between the TME and antitumor immunity involves the presence of BTN, which should be taken into account when developing immunotherapy. This review explores potential therapeutic applications of BTN molecules, based on the current understanding of their biology.
Collapse
Affiliation(s)
- Fatemeh Mehdikhani
- Department of Clinical Biochemistry, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Aysa Bahar
- Department of Biochemistry, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Marzieh Bashi
- Cancer Research Center, Semnan University of Medical, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Maryam Mohammadlou
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Bahman Yousefi
- Cancer Research Center, Semnan University of Medical, Semnan, Iran
- Department of Immunology, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
11
|
Li W, Zhao X, Ren C, Gao S, Han Q, Lu M, Li X. The therapeutic role of γδT cells in TNBC. Front Immunol 2024; 15:1420107. [PMID: 38933280 PMCID: PMC11199784 DOI: 10.3389/fimmu.2024.1420107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer that presents significant therapeutic challenges due to the absence of estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) expression. As a result, conventional hormonal and targeted therapies are largely ineffective, underscoring the urgent need for novel treatment strategies. γδT cells, known for their robust anti-tumor properties, show considerable potential in TNBC treatment as they can identify and eliminate tumor cells without reliance on MHC restrictions. These cells demonstrate extensive proliferation both in vitro and in vivo, and can directly target tumors through cytotoxic effects or indirectly by promoting other immune responses. Studies suggest that expansion and adoptive transfer strategies targeting Vδ2 and Vδ1 γδT cell subtypes have shown promise in preclinical TNBC models. This review compiles and discusses the existing literature on the primary subgroups of γδT cells, their roles in cancer therapy, their contributions to tumor cell cytotoxicity and immune modulation, and proposes potential strategies for future γδT cell-based immunotherapies in TNBC.
Collapse
Affiliation(s)
- Wenjing Li
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Xian Zhao
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Chuanxin Ren
- Department of The First Clinical Medical School, Shandong University of Traditional Chinese Medicine, Jinan, Shandong, China
| | - Shang Gao
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Qinyu Han
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Min Lu
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Xiangqi Li
- Department of Breast Center, The Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| |
Collapse
|
12
|
Hu H, Zhu H, Zhan W, Hao B, Yan T, Zhang J, Wang S, Xu X, Zhang T. Integration of multiomics analyses reveals unique insights into CD24-mediated immunosuppressive tumor microenvironment of breast cancer. Inflamm Res 2024; 73:1047-1068. [PMID: 38622285 DOI: 10.1007/s00011-024-01882-9] [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: 12/21/2023] [Revised: 02/19/2024] [Accepted: 04/07/2024] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND Tumor immunotherapy brings new light and vitality to breast cancer patients, but low response rate and limitations of therapeutic targets become major obstacles to its clinical application. Recent studies have shown that CD24 is involved in an important process of tumor immune regulation in breast cancer and is a promising target for immunotherapy. METHODS In this study, singleR was used to annotate each cell subpopulation after t-distributed stochastic neighbor embedding (t-SNE) methods. Pseudo-time trace analysis and cell communication were analyzed by Monocle2 package and CellChat, respectively. A prognostic model based on CD24-related genes was constructed using several machine learning methods. Multiple quantitative immunofluorescence (MQIF) was used to evaluate the spatial relationship between CD24+PANCK+cells and exhausted CD8+T cells. RESULTS Based on the scRNA-seq analysis, 1488 CD24-related differential genes were identified, and a risk model consisting of 15 prognostic characteristic genes was constructed by combining the bulk RNA-seq data. Patients were divided into high- and low-risk groups based on the median risk score. Immune landscape analysis showed that the low-risk group showed higher infiltration of immune-promoting cells and stronger immune reactivity. The results of cell communication demonstrated a strong interaction between CD24+epithelial cells and CD8+T cells. Subsequent MQIF demonstrated a strong interaction between CD24+PANCK+ and exhausted CD8+T cells with FOXP3+ in breast cancer. Additionally, CD24+PANCK+ and CD8+FOXP3+T cells were positively associated with lower survival rates. CONCLUSION This study highlights the importance of CD24+breast cancer cells in clinical prognosis and immunosuppressive microenvironment, which may provide a new direction for improving patient outcomes.
Collapse
Affiliation(s)
- Haihong Hu
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
- Phase I Clinical Trial Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hongxia Zhu
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Wendi Zhan
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Bo Hao
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Ting Yan
- Department of Breast and Thyroid Surgery, The First Affiliated HospitalH, engyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Jingdi Zhang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China
| | - Siyu Wang
- Department of Medical Oncology,The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xuefeng Xu
- Department of Function, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Taolan Zhang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- School of Pharmacy, Hengyang Medical College, University of South China, Hengyang, 421001, Hunan, China.
- Phase I Clinical Trial Center, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
13
|
Minina EP, Dianov DV, Sheetikov SA, Bogolyubova AV. CAR Cells beyond Classical CAR T Cells: Functional Properties and Prospects of Application. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:765-783. [PMID: 38880641 DOI: 10.1134/s0006297924050018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/23/2023] [Accepted: 12/02/2023] [Indexed: 06/18/2024]
Abstract
Chimeric antigen receptors (CARs) are genetically engineered receptors that recognize antigens and activate signaling cascades in a cell. Signal recognition and transmission are mediated by the CAR domains derived from different proteins. T cells carrying CARs against tumor-associated antigens have been used in the development of the CAR T cell therapy, a new approach to fighting malignant neoplasms. Despite its high efficacy in the treatment of oncohematological diseases, CAR T cell therapy has a number of disadvantages that could be avoided by using other types of leukocytes as effector cells. CARs can be expressed in a wide range of cells of adaptive and innate immunity with the emergence or improvement of cytotoxic properties. This review discusses the features of CAR function in different types of immune cells, with a particular focus on the results of preclinical and clinical efficacy studies and the safety of potential CAR cell products.
Collapse
Affiliation(s)
- Elizaveta P Minina
- National Medical Research Centre for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Dmitry V Dianov
- National Medical Research Centre for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Saveliy A Sheetikov
- National Medical Research Centre for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia
| | - Apollinariya V Bogolyubova
- National Medical Research Centre for Hematology, Ministry of Health of the Russian Federation, Moscow, 125167, Russia.
| |
Collapse
|
14
|
Wei XY, Tan YQ, Zhou G. γδ T cells in oral diseases. Inflamm Res 2024; 73:867-876. [PMID: 38563967 DOI: 10.1007/s00011-024-01870-z] [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: 12/25/2023] [Revised: 03/03/2024] [Accepted: 03/05/2024] [Indexed: 04/04/2024] Open
Abstract
OBJECTIVE γδ T cells are a distinct subset of unconventional T cells, which link innate and adaptive immunity by secreting cytokines and interacting with other immune cells, thereby modulating immune responses. As the first line of host defense, γδ T cells are essential for mucosal homeostasis and immune surveillance. When abnormally activated or impaired, γδ T cells can contribute to pathogenic processes. Accumulating evidence has revealed substantial impacts of γδ T cells on the pathogenesis of cancers, infections, and immune-inflammatory diseases. γδ T cells exhibit dual roles in cancers, promoting or inhibiting tumor growth, depending on their phenotypes and the clinical stage of cancers. During infections, γδ T cells exert high cytotoxic activity in infectious diseases, which is essential for combating bacterial and viral infections by recognizing foreign antigens and activating other immune cells. γδ T cells are also implicated in the onset and progression of immune-inflammatory diseases. However, the specific involvement and underlying mechanisms of γδ T cells in oral diseases have not been systematically discussed. METHODS We conducted a systematic literature review using the PubMed/MEDLINE databases to identify and analyze relevant literature on the roles of γδ T cells in oral diseases. RESULTS The literature review revealed that γδ T cells play a pivotal role in maintaining oral mucosal homeostasis and are involved in the pathogenesis of oral cancers, periodontal diseases, graft-versus-host disease (GVHD), oral lichen planus (OLP), and oral candidiasis. γδ T cells mainly influence various pathophysiological processes, such as anti-tumor activity, eradication of infection, and immune response regulation. CONCLUSION This review focuses on the involvement of γδ T cells in oral diseases, with a particular emphasis on the main functions and underlying mechanisms by which γδ T cells influence the pathogenesis and progression of these conditions. This review underscores the potential of γδ T cells as therapeutic targets in managing oral health issues.
Collapse
Affiliation(s)
- Xin-Yi Wei
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Ya-Qin Tan
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| | - Gang Zhou
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, China.
- Department of Oral Medicine, School and Hospital of Stomatology, Wuhan University, Wuhan, China.
| |
Collapse
|
15
|
Di Simone M, Corsale AM, Toia F, Shekarkar Azgomi M, Di Stefano AB, Lo Presti E, Cordova A, Montesano L, Dieli F, Meraviglia S. Tumor-infiltrating γδ T cells as targets of immune checkpoint blockade in melanoma. J Leukoc Biol 2024; 115:760-770. [PMID: 38324004 DOI: 10.1093/jleuko/qiae023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 11/21/2023] [Accepted: 12/31/2023] [Indexed: 02/08/2024] Open
Abstract
Melanoma is one of the most sensitive tumors to immune modulation, and the major challenge for melanoma patients' survival is immune checkpoint inhibitor (ICI) therapy. γδ T lymphocytes play an antitumoral role in a broad variety of tumors including melanoma and they are optimal candidates for cellular immunotherapy. Thus, a comprehensive analysis of the correlation between γδ T cells and immune checkpoint receptors in the context of melanoma was conducted, with the aim of devising an innovative combined immunotherapeutic strategy. In this study, using the GEPIA2.0 database, a significant positive correlation was observed between the expression of γδ T cell-related genes (TRGC1, TRGC2, TCRD) and immune checkpoint genes (PDCD1, HAVCR2, LAG3), highlighting the potential role of γδ T cells in the immune response within melanoma. Moreover, flow cytometry analysis unveiled a significant augmentation in the population of γδ T cells within melanoma lesions, which exhibited the expression of immune checkpoint receptors including LAG3, TIM3, and PD1. Analysis of single-cell RNA sequencing data revealed a significant enrichment and functional reprogramming of γδ T cell clusters in response to ICIs. Interestingly, the effects of ICI therapy varied between Vδ1 and Vδ2 γδ T cell subsets, with distinct changes in gene expression patterns. Last, a correlation analysis between γδ T cell abundance, immune checkpoint gene expression, and clinical outcomes in melanoma patients showed that low expression of immune checkpoint genes, including LAG3, HAVCR2, and PDCD1, was associated with improved 1-year overall survival, emphasizing the significance of these genes in predicting patient outcomes, potentially outweighing the impact of γδ T cell abundance. This study offers critical insights into the dynamic interaction between γδ T cells, immune checkpoint receptors, and melanoma, providing valuable perspectives for potential therapeutic avenues and predictive markers in this intricate interplay.
Collapse
Affiliation(s)
- Marta Di Simone
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Anna Maria Corsale
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Francesca Toia
- Laboratory of Biology and Regenerative Medicine-Plastic Surgery, Plastic and Reconstructive Surgery, Department of Surgical Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Mojtaba Shekarkar Azgomi
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Anna Barbara Di Stefano
- Laboratory of Biology and Regenerative Medicine-Plastic Surgery, Plastic and Reconstructive Surgery, Department of Surgical Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Elena Lo Presti
- National Research Council Institute for Biomedical Research and Innovation, Via Ugo La Malfa 153, 90146, Palermo, Italy
| | - Adriana Cordova
- Laboratory of Biology and Regenerative Medicine-Plastic Surgery, Plastic and Reconstructive Surgery, Department of Surgical Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Luigi Montesano
- Laboratory of Biology and Regenerative Medicine-Plastic Surgery, Plastic and Reconstructive Surgery, Department of Surgical Oncological and Oral Sciences, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Francesco Dieli
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Serena Meraviglia
- Central Laboratory of Advanced Diagnosis and Biomedical Research, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Department of Biomedicine, Neuroscience and Advanced Diagnosis, University of Palermo, Via del Vespro 129, 90127, Palermo, Italy
| |
Collapse
|
16
|
Yuan M, Wang W, Hawes I, Han J, Yao Z, Bertaina A. Advancements in γδT cell engineering: paving the way for enhanced cancer immunotherapy. Front Immunol 2024; 15:1360237. [PMID: 38576617 PMCID: PMC10991697 DOI: 10.3389/fimmu.2024.1360237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/07/2024] [Indexed: 04/06/2024] Open
Abstract
Comprising only 1-10% of the circulating T cell population, γδT cells play a pivotal role in cancer immunotherapy due to their unique amalgamation of innate and adaptive immune features. These cells can secrete cytokines, including interferon-γ (IFN-γ) and tumor necrosis factor-α (TNF-α), and can directly eliminate tumor cells through mechanisms like Fas/FasL and antibody-dependent cell-mediated cytotoxicity (ADCC). Unlike conventional αβT cells, γδT cells can target a wide variety of cancer cells independently of major histocompatibility complex (MHC) presentation and function as antigen-presenting cells (APCs). Their ability of recognizing antigens in a non-MHC restricted manner makes them an ideal candidate for allogeneic immunotherapy. Additionally, γδT cells exhibit specific tissue tropism, and rapid responsiveness upon reaching cellular targets, indicating a high level of cellular precision and adaptability. Despite these capabilities, the therapeutic potential of γδT cells has been hindered by some limitations, including their restricted abundance, unsatisfactory expansion, limited persistence, and complex biology and plasticity. To address these issues, gene-engineering strategies like the use of chimeric antigen receptor (CAR) T therapy, T cell receptor (TCR) gene transfer, and the combination with γδT cell engagers are being explored. This review will outline the progress in various engineering strategies, discuss their implications and challenges that lie ahead, and the future directions for engineered γδT cells in both monotherapy and combination immunotherapy.
Collapse
Affiliation(s)
| | - Wenjun Wang
- *Correspondence: Wenjun Wang, ; Alice Bertaina,
| | | | | | | | - Alice Bertaina
- Division of Hematology, Oncology, Stem Cell Transplantation and Regenerative Medicine, Department of Pediatrics, Stanford University, School of Medicine, Stanford, CA, United States
| |
Collapse
|
17
|
Rao A, Agrawal A, Borthakur G, Battula VL, Maiti A. Gamma delta T cells in acute myeloid leukemia: biology and emerging therapeutic strategies. J Immunother Cancer 2024; 12:e007981. [PMID: 38417915 PMCID: PMC10900322 DOI: 10.1136/jitc-2023-007981] [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] [Accepted: 02/04/2024] [Indexed: 03/01/2024] Open
Abstract
γδ T cells play an important role in disease control in acute myeloid leukemia (AML) and have become an emerging area of therapeutic interest. These cells represent a minor population of T lymphocytes with intrinsic abilities to recognize antigens in a major histocompatibility complex-independent manner and functionally straddle the innate and adaptive immunity interface. AML shows high expression of phosphoantigens and UL-16 binding proteins that activate the Vδ2 and Vδ1 subtypes of γδ T cells, respectively, leading to γδ T cell-mediated cytotoxicity. Insights from murine models and clinical data in humans show improved overall survival, leukemia-free survival, reduced risk of relapse, enhanced graft-versus-leukemia effect, and decreased graft-versus-host disease in patients with AML who have higher reconstitution of γδ T cells following allogeneic hematopoietic stem cell transplantation. Clinical trials leveraging γδ T cell biology have used unmodified and modified allogeneic cells as well as bispecific engagers and monoclonal antibodies. In this review, we discuss γδ T cells' biology, roles in cancer and AML, and mechanisms of immune escape and antileukemia effect; we also discuss recent clinical advances related to γδ T cells in the field of AML therapeutics.
Collapse
Affiliation(s)
- Adishwar Rao
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Akriti Agrawal
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Gautam Borthakur
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Venkata Lokesh Battula
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Abhishek Maiti
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| |
Collapse
|
18
|
Revesz IA, Joyce P, Ebert LM, Prestidge CA. Effective γδ T-cell clinical therapies: current limitations and future perspectives for cancer immunotherapy. Clin Transl Immunology 2024; 13:e1492. [PMID: 38375329 PMCID: PMC10875631 DOI: 10.1002/cti2.1492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 01/24/2024] [Accepted: 02/05/2024] [Indexed: 02/21/2024] Open
Abstract
γδ T cells are a unique subset of T lymphocytes, exhibiting features of both innate and adaptive immune cells and are involved with cancer immunosurveillance. They present an attractive alternative to conventional T cell-based immunotherapy due, in large part, to their lack of major histocompatibility (MHC) restriction and ability to secrete high levels of cytokines with well-known anti-tumour functions. To date, clinical trials using γδ T cell-based immunotherapy for a range of haematological and solid cancers have yielded limited success compared with in vitro studies. This inability to translate the efficacy of γδ T-cell therapies from preclinical to clinical trials is attributed to a combination of several factors, e.g. γδ T-cell agonists that are commonly used to stimulate populations of these cells have limited cellular uptake yet rely on intracellular mechanisms; administered γδ T cells display low levels of tumour-infiltration; and there is a gap in the understanding of γδ T-cell inhibitory receptors. This review explores the discrepancy between γδ T-cell clinical and preclinical performance and offers viable avenues to overcome these obstacles. Using more direct γδ T-cell agonists, encapsulating these agonists into lipid nanocarriers to improve their pharmacokinetic and pharmacodynamic profiles and the use of combination therapies to overcome checkpoint inhibition and T-cell exhaustion are ways to bridge the gap between preclinical and clinical success. Given the ability to overcome these limitations, the development of a more targeted γδ T-cell agonist-checkpoint blockade combination therapy has the potential for success in clinical trials which has to date remained elusive.
Collapse
Affiliation(s)
- Isabella A Revesz
- Clinical Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Paul Joyce
- Clinical Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| | - Lisa M Ebert
- Centre for Cancer BiologySA Pathology and University of South AustraliaAdelaideSAAustralia
- Cancer Clinical Trials UnitRoyal Adelaide HospitalAdelaideSAAustralia
- School of MedicineThe University of AdelaideAdelaideSAAustralia
| | - Clive A Prestidge
- Clinical Health SciencesUniversity of South AustraliaAdelaideSAAustralia
| |
Collapse
|
19
|
Kaur K, Jewett A. Osteoclasts and Probiotics Mediate Significant Expansion, Functional Activation and Supercharging in NK, γδ T, and CD3+ T Cells: Use in Cancer Immunotherapy. Cells 2024; 13:213. [PMID: 38334605 PMCID: PMC10854567 DOI: 10.3390/cells13030213] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 01/17/2024] [Accepted: 01/22/2024] [Indexed: 02/10/2024] Open
Abstract
Our previous studies have introduced osteoclasts (OCs) as major activators of NK cells. It was found that OCs exhibit the capabilities of inducing cell expansion as well as increasing the cytotoxic activity of NK cells by granule release and increasing the secretion of TNF-α and TRAIL, leading to increased lysis of tumors in short-term as well as long-term periods, respectively. OC- induced expanded NK cells were named supercharged NK cells (sNK) due to their significantly high functional activity as well as their significantly higher cell expansion rate. It is, however, unclear whether the OC-mediated effect in NK cells is specific or whether other cytotoxic immune cells can also be expanded and activated by OCs. We chose to focus on γδ T cells and pan T cells, which also include CD8+ T cells. In this paper, we report that OCs are capable of expanding and functionally activating both γδ T cells and pan T cells. Expanded γδ T and pan T cells were capable of secreting high levels of INF-γ, albeit with different dynamics to those of NK cells, and, moreover, they are unable to kill NK-specific targets. Since we used humanized-BLT (hu-BLT) mice as a model of human disease, we next determined whether NK and T cell activation through OCs is also evident in cells obtained from hu-BLT mice. Similar to humans, OCs were capable of increasing the cell expansion and secretion of IFN-γ in the culture of either NK or T cells from hu-BLT mice, providing yet further evidence that these mice are appropriate models to study human disease. Therefore, these studies indicated that CD3+ T or γδ T cells can proliferate and be supercharged by OCs similar to the NK cells; thus, they can be used individually or in combination in the cell therapy of cancers.
Collapse
Affiliation(s)
- Kawaljit Kaur
- Division of Oral Biology and Medicine, School of Dentistry and Medicine, University of California, Los Angeles, CA 90095, USA;
| | - Anahid Jewett
- Division of Oral Biology and Medicine, School of Dentistry and Medicine, University of California, Los Angeles, CA 90095, USA;
- The Jonsson Comprehensive Cancer Center, School of Dentistry and Medicine, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
20
|
Zhao Y, Dong P, He W, Zhang J, Chen H. γδ T cells: Major advances in basic and clinical research in tumor immunotherapy. Chin Med J (Engl) 2024; 137:21-33. [PMID: 37592858 PMCID: PMC10766231 DOI: 10.1097/cm9.0000000000002781] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Indexed: 08/19/2023] Open
Abstract
ABSTRACT γδ T cells are a kind of innate immune T cell. They have not attracted sufficient attention because they account for only a small proportion of all immune cells, and many basic factors related to these cells remain unclear. However, in recent years, with the rapid development of tumor immunotherapy, γδ T cells have attracted increasing attention because of their ability to exert cytotoxic effects on most tumor cells without major histocompatibility complex (MHC) restriction. An increasing number of basic studies have focused on the development, antigen recognition, activation, and antitumor immune response of γδ T cells. Additionally, γδ T cell-based immunotherapeutic strategies are being developed, and the number of clinical trials investigating such strategies is increasing. This review mainly summarizes the progress of basic research and the clinical application of γδ T cells in tumor immunotherapy to provide a theoretical basis for further the development of γδ T cell-based strategies in the future.
Collapse
Affiliation(s)
- Yueqi Zhao
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Peng Dong
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, Jiangsu 213000, China
| | - Wei He
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
| | - Jianmin Zhang
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, Jiangsu 213000, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| | - Hui Chen
- Department of Immunology, CAMS Key Laboratory for T Cell and Immunotherapy, State Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and School of Basic Medicine, Peking Union Medical College, Beijing 100005, China
- Changzhou Xitaihu Institute for Frontier Technology of Cell Therapy, Changzhou, Jiangsu 213000, China
- Haihe Laboratory of Cell Ecosystem, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300020, China
| |
Collapse
|
21
|
Zhang M, Wu Y, Qin Y, Shen J, Cui Z, Lei F, Zhang K, Li B, Liang S, Peng M. Dual regulation effect and mechanism of human myeloid-derived suppressor cells on anticolorectal cancer cells activity of Vγ9Vδ2 T cells. Cell Biochem Funct 2024; 42:e3929. [PMID: 38269504 DOI: 10.1002/cbf.3929] [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: 09/12/2023] [Revised: 12/14/2023] [Accepted: 01/05/2024] [Indexed: 01/26/2024]
Abstract
Myeloid-derived suppressor cells (MDSC) are a group of immature inhibitory cells of bone marrow origin. Human γδ T cells (mainly Vγ9Vδ2 T cells) have emerged as dominant candidates for cancer immunotherapy because of their unique recognition pattern and broad killing activity against tumor cells. Intestinal mucosal intraepithelial lymphocytes are almost exclusively γδ T cells, so it plays an important role in inhibiting the development of colorectal cancer. In this study, we investigated the effects and molecular mechanism of human MDSC on anticolorectal cancer cells activity of Vγ9Vδ2 T cells. Our results suggested that MDSC can reduce the NKG2D expression of Vγ9Vδ2 T cells through direct cell-cell contact, which is associated with membrane-type transforming growth factor-β. In contrast, MDSC can increase Vγ9Vδ2 T cells activation and production of IFN-γ, perforin, Granzyme B through direct cell-cell contact. This may be related to the upregulation of T-bet in Vγ9Vδ2 T cells by MDSC. However, MDSC had a dominant negative regulatory effect on the anticolorectal cancer cells activity of Vγ9Vδ2 T cells. Our study provides a theoretical basis for the immune regulatory function of human MDSC on γδ T cells. This will be conducive to the clinical development of a new antitumor therapy strategy.
Collapse
Affiliation(s)
- Mengyu Zhang
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Yuanyuan Wu
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Yahan Qin
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Jie Shen
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Zhao Cui
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Fan Lei
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Ke Zhang
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Baiqing Li
- Department of Immunology, Bengbu Medical College, Bengbu, China
| | - Shujuan Liang
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| | - Meiyu Peng
- Key Laboratory of Immune Microenvironment and Inflammatory Disease Research in Universities of Shandong Province, School of Basic Medical Sciences, Shandong Second Medical University, Weifang, China
| |
Collapse
|
22
|
Chamorro DF, Somes LK, Hoyos V. Engineered Adoptive T-Cell Therapies for Breast Cancer: Current Progress, Challenges, and Potential. Cancers (Basel) 2023; 16:124. [PMID: 38201551 PMCID: PMC10778447 DOI: 10.3390/cancers16010124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/19/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
Breast cancer remains a significant health challenge, and novel treatment approaches are critically needed. This review presents an in-depth analysis of engineered adoptive T-cell therapies (E-ACTs), an innovative frontier in cancer immunotherapy, focusing on their application in breast cancer. We explore the evolving landscape of chimeric antigen receptor (CAR) and T-cell receptor (TCR) T-cell therapies, highlighting their potential and challenges in targeting breast cancer. The review addresses key obstacles such as target antigen selection, the complex breast cancer tumor microenvironment, and the persistence of engineered T-cells. We discuss the advances in overcoming these barriers, including strategies to enhance T-cell efficacy. Finally, our comprehensive analysis of the current clinical trials in this area provides insights into the future possibilities and directions of E-ACTs in breast cancer treatment.
Collapse
Affiliation(s)
- Diego F. Chamorro
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Lauren K. Somes
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
| | - Valentina Hoyos
- Center for Cell and Gene Therapy, Baylor College of Medicine, Houston, TX 77030, USA; (D.F.C.); (L.K.S.)
- Dan L. Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX 77030, USA
| |
Collapse
|
23
|
Chen Z, Hu Y, Mei H. Advances in CAR-Engineered Immune Cell Generation: Engineering Approaches and Sourcing Strategies. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2303215. [PMID: 37906032 PMCID: PMC10724421 DOI: 10.1002/advs.202303215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 09/03/2023] [Indexed: 11/02/2023]
Abstract
Chimeric antigen receptor T-cell (CAR-T) therapy has emerged as a highly efficacious treatment modality for refractory and relapsed hematopoietic malignancies in recent years. Furthermore, CAR technologies for cancer immunotherapy have expanded from CAR-T to CAR-natural killer cell (CAR-NK), CAR-cytokine-induced killer cell (CAR-CIK), and CAR-macrophage (CAR-MΦ) therapy. Nevertheless, the high cost and complex manufacturing processes of ex vivo generation of autologous CAR products have hampered broader application. There is an urgent need to develop an efficient and economical paradigm shift for exploring new sourcing strategies and engineering approaches toward generating CAR-engineered immune cells to benefit cancer patients. Currently, researchers are actively investigating various strategies to optimize the preparation and sourcing of these potent immunotherapeutic agents. In this work, the latest research progress is summarized. Perspectives on the future of CAR-engineered immune cell manufacturing are provided, and the engineering approaches, and diverse sources used for their development are focused upon.
Collapse
Affiliation(s)
- Zhaozhao Chen
- Institute of HematologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic DiseaseWuhan430022China
| | - Yu Hu
- Institute of HematologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic DiseaseWuhan430022China
| | - Heng Mei
- Institute of HematologyUnion HospitalTongji Medical CollegeHuazhong University of Science and Technology1277 Jiefang AvenueWuhanHubei430022China
- Hubei Clinical Medical Center of Cell Therapy for Neoplastic DiseaseWuhan430022China
| |
Collapse
|
24
|
Qie Y, Sun X, Yang Y, Yan T. Emerging functions and applications of exosomes in oral squamous cell carcinoma. J Oral Pathol Med 2023; 52:886-894. [PMID: 37701945 DOI: 10.1111/jop.13479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 08/09/2023] [Accepted: 08/30/2023] [Indexed: 09/14/2023]
Abstract
Oral squamous cell carcinoma is the most common phenotype in pathology, which accounts for 80% of all oral cancers. The therapeutic methods of oral squamous cell carcinoma include surgical excision, chemotherapy, and radiotherapy. Whereas, the high recurrence rate and poor prognosis lead to a 5-year survival rate less than 50%. In order to explore more therapeutic strategies of oral squamous cell carcinoma, the relevant risk factors, mechanisms, and diagnostics are widely detected. The various exosome-mediated biological effects on the development of oral squamous cell carcinoma have drawn lots of attention. Exosomes, a kind of extracellular vesicles secreted from host cells and transferred to other cells, show great potential in the regulations of tumorigenesis, progression, and metastasis on oral squamous cell carcinoma. Moreover, some studies reported that the exosomes could interact with tumor microenvironment and be applied to diagnosis or therapy of oral squamous cell carcinoma. In this work, we will summarize the frontier studies of exosomes in the tumor growth, tumor-associated angiogenesis, invasion, and metastasis of oral squamous cell carcinoma, and then probe the current biological functions and applications of exosomes and exosome-derived materials for the therapeutic strategies of oral squamous cell carcinoma, which would help us to update the understanding of exosomes in oral squamous cell carcinoma.
Collapse
Affiliation(s)
- Yingchun Qie
- Stomatology Department, Zibo First Hospital, Zibo, Shandong Province, China
| | - Xia Sun
- Stomatology Department, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Yongqiang Yang
- Stomatology Department, Yidu Central Hospital of Weifang, Qingzhou, Shandong Province, China
| | - Tao Yan
- Intensive Care Unit, Zibo First Hospital, Zibo, Shandong Province, China
| |
Collapse
|
25
|
Ishihara M, Miwa H, Fujiwara H, Akahori Y, Kato T, Tanaka Y, Tawara I, Shiku H. αβ-T cell receptor transduction gives superior mitochondrial function to γδ-T cells with promising persistence. iScience 2023; 26:107802. [PMID: 37720098 PMCID: PMC10502403 DOI: 10.1016/j.isci.2023.107802] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Revised: 07/25/2023] [Accepted: 08/29/2023] [Indexed: 09/19/2023] Open
Abstract
Adoptive cell therapy using allogeneic γδ-T cells is a promising option for off-the-shelf T cell products with a low risk of graft-versus-host disease (GVHD). Long-term persistence may boost the clinical development of γδ-T cell products. In this study, we found that genetically modified Vγ9+Vδ2+ T cells expressing a tumor antigen-specific αβ-TCR and CD8 coreceptor (GMC) showed target-specific killing and excellent persistence. To determine the mechanisms underlying these promising effects, we investigated metabolic characteristics. Cytokine secretion by γδ-TCR-stimulated nongene-modified γδ-T cells (NGMCs) and αβ-TCR-stimulated GMCs was equally suppressed by a glycolysis inhibitor, although the cytokine secretion of αβ-TCR-stimulated GMCs was more strongly inhibited by ATP synthase inhibitors than that of γδ-TCR-stimulated NGMCs. Metabolomic and transcriptomic analyses, flow cytometry analysis using mitochondria-labeling dyes and extracellular flux analysis consistently suggest that αβ-TCR-transduced γδ-T cells acquire superior mitochondrial function. In conclusion, αβ-TCR-transduced γδ-T cells acquire superior mitochondrial function with promising persistence.
Collapse
Affiliation(s)
- Mikiya Ishihara
- Department of Medical Oncology, Mie University Hospital, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Hiroshi Miwa
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, 1577 Kurimamachiya-cho, Tsu, Mie 514-8507, Japan
| | - Hiroshi Fujiwara
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, 1577 Kurimamachiya-cho, Tsu, Mie 514-8507, Japan
| | - Yasushi Akahori
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, 1577 Kurimamachiya-cho, Tsu, Mie 514-8507, Japan
| | - Takuma Kato
- Department of Cellular and Molecular Immunology, Mie University Graduate School of Medicine, Tsu, Mie 514-8507, Japan
| | - Yoshimasa Tanaka
- Center for Medical Innovation, Nagasaki University, 1-7-1 Sakamoto, Nagasaki 852-8588, Japan
| | - Isao Tawara
- Department of Hematology and Oncology, Mie University Graduate School of Medicine, 2-174 Edobashi, Tsu, Mie 514-8507, Japan
| | - Hiroshi Shiku
- Department of Personalized Cancer Immunotherapy, Mie University Graduate School of Medicine, 1577 Kurimamachiya-cho, Tsu, Mie 514-8507, Japan
| |
Collapse
|
26
|
Gao X, Zuo S. Immune landscape and immunotherapy of hepatocellular carcinoma: focus on innate and adaptive immune cells. Clin Exp Med 2023; 23:1881-1899. [PMID: 36773210 PMCID: PMC10543580 DOI: 10.1007/s10238-023-01015-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2023] [Accepted: 01/27/2023] [Indexed: 02/12/2023]
Abstract
Hepatocellular carcinoma (HCC) is responsible for roughly 90% of all cases of primary liver cancer, and the cases are on the rise. The treatment of advanced HCC is a serious challenge. Immune checkpoint inhibitor (ICI) therapy has marked a watershed moment in the history of HCC systemic treatment. Atezolizumab in combination with bevacizumab has been approved as a first-line treatment for advanced HCC since 2020; however, the combination therapy is only effective in a limited percentage of patients. Considering that the tumor immune microenvironment (TIME) has a great impact on immunotherapies for HCC, an in-depth understanding of the immune landscape in tumors and the current immunotherapeutic approaches is extremely necessary. We elaborate on the features, functions, and cross talk of the innate and adaptive immune cells in HCC and highlight the benefits and drawbacks of various immunotherapies for advanced HCC, as well as future projections. HCC consists of a heterogeneous group of cancers with distinct etiologies and immune microenvironments. Almost all the components of innate and adaptive immune cells in HCC have altered, showing a decreasing trend in the number of tumor suppressor cells and an increasing trend in the pro-cancer cells, and there is also cross talk between various cell types. Various immunotherapies for HCC have also shown promising efficacy and application prospect. There are multilayered interwoven webs among various immune cell types in HCC, and emerging evidence demonstrates the promising prospect of immunotherapeutic approaches for HCC.
Collapse
Affiliation(s)
- Xiaoqiang Gao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550000, Guizhou, China
- Guizhou Medical University, Guiyang, Guizhou, China
| | - Shi Zuo
- Department of Hepatobiliary Surgery, Affiliated Hospital of Guizhou Medical University, No. 28, Guiyi Street, Guiyang, 550000, Guizhou, China.
- Guizhou Medical University, Guiyang, Guizhou, China.
| |
Collapse
|
27
|
Tai R, Leng J, Li W, Wu Y, Yang J. Construction of the metabolic reprogramming-associated gene signature for clear cell renal cell carcinoma prognosis prediction. BMC Urol 2023; 23:147. [PMID: 37715154 PMCID: PMC10503121 DOI: 10.1186/s12894-023-01317-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Accepted: 09/04/2023] [Indexed: 09/17/2023] Open
Abstract
BACKGROUND Metabolism reprogramming is a hallmark that associates tumor growth, metastasis, progressive, and poor prognosis. However, the metabolism-related molecular patterns and mechanism in clear cell renal cell carcinoma (ccRCC) remain unclear. Herein, the purpose of this study was to identify metabolism-related molecular pattern and to investigate the characteristics and prognostic values of the metabolism-related clustering. METHODS We comprehensively analyzed the differentially expressed genes (DEGs), and metabolism-related genes (MAGs) in ccRCC based on the TCGA database. Consensus clustering was used to construct a metabolism-related molecular pattern. Then, the biological function, molecular characteristics, Estimate/immune/stomal scores, immune cell infiltration, response to immunotherapy, and chemotherapy were analyzed. We also identified the DEGs between subclusters and constructed a poor signature and risk model based on LASSO regression cox analysis and univariable and multivariable cox regression analyses. Then, a predictive nomogram was constructed and validated by calibration curves. RESULTS A total of 1942 DEGs (1004 upregulated and 838 downregulated) between ccRCC tumor and normal samples were identified, and 254 MRGs were screened out from those DEGs. Then, 526 ccRCC patients were divided into two subclusters. The 7 metabolism-related pathways enriched in cluster 2. And cluster 2 with high Estimate/immune/stomal scores and poor survival. While, cluster 1 with higher immune cell infiltrating, expression of the immune checkpoint, IFN, HLA, immune activation-related genes, response to anti-CTLA4 treatment, and chemotherapy. Moreover, we identified 295 DEGs between two metabolism-related subclusters and constructed a 15-gene signature and 9 risk factors. Then, a risk score was calculated and the patients into high- and low-risk groups in TCGA-KIRC and E-MTAB-1980 datasets. And the prediction viability of the risk score was validated by ROC curves. Finally, the clinicopathological characteristics (age and stage), risk score, and molecular clustering, were identified as independent prognostic variables, and were used to construct a nomogram for 1-, 3-, 5-year overall survival predicting. The calibration curves were used to verify the performance of the predicted ability of the nomogram. CONCLUSION Our finding identified two metabolism-related molecular subclusters for ccRCC, which facilitates the estimation of response to immunotherapy and chemotherapy, and prognosis after treatment.
Collapse
Affiliation(s)
- Rongfen Tai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
- Department of Urology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Jinjun Leng
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China
- Department of Urology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Wei Li
- Department of Urology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Yuerong Wu
- Department of Urology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China
| | - Junfeng Yang
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and Technology, Kunming, Yunnan, 650500, China.
- Department of Urology, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, Yunnan, 650032, China.
| |
Collapse
|
28
|
Zhang P, Zhang G, Wan X. Challenges and new technologies in adoptive cell therapy. J Hematol Oncol 2023; 16:97. [PMID: 37596653 PMCID: PMC10439661 DOI: 10.1186/s13045-023-01492-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 30.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
Adoptive cell therapies (ACTs) have existed for decades. From the initial infusion of tumor-infiltrating lymphocytes to the subsequent specific enhanced T cell receptor (TCR)-T and chimeric antigen receptor (CAR)-T cell therapies, many novel strategies for cancer treatment have been developed. Owing to its promising outcomes, CAR-T cell therapy has revolutionized the field of ACTs, particularly for hematologic malignancies. Despite these advances, CAR-T cell therapy still has limitations in both autologous and allogeneic settings, including practicality and toxicity issues. To overcome these challenges, researchers have focused on the application of CAR engineering technology to other types of immune cell engineering. Consequently, several new cell therapies based on CAR technology have been developed, including CAR-NK, CAR-macrophage, CAR-γδT, and CAR-NKT. In this review, we describe the development, advantages, and possible challenges of the aforementioned ACTs and discuss current strategies aimed at maximizing the therapeutic potential of ACTs. We also provide an overview of the various gene transduction strategies employed in immunotherapy given their importance in immune cell engineering. Furthermore, we discuss the possibility that strategies capable of creating a positive feedback immune circuit, as healthy immune systems do, could address the flaw of a single type of ACT, and thus serve as key players in future cancer immunotherapy.
Collapse
Affiliation(s)
- Pengchao Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China
- University of Chinese Academy of Sciences, Beijing, 100049, People's Republic of China
| | - Guizhong Zhang
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China.
| | - Xiaochun Wan
- Center for Protein and Cell-based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 1068 Xueyuan Avenue, Nanshan District, Shenzhen, 518055, People's Republic of China.
| |
Collapse
|
29
|
Yang Z, Yuan ZZ, Ma XL. Identification of a potential novel biomarker in intervertebral disk degeneration by bioinformatics analysis and experimental validation. Front Immunol 2023; 14:1136727. [PMID: 37325631 PMCID: PMC10266279 DOI: 10.3389/fimmu.2023.1136727] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Accepted: 05/10/2023] [Indexed: 06/17/2023] Open
Abstract
Background Intervertebral disk degeneration (IVDD) is a major cause of low back pain and one of the most common health problems all over the world. However, the early diagnosis of IVDD is still restricted. The purpose of this study is to identify and validate the key characteristic gene of IVDD and analyze its correlation with immune cell infiltration. Methods 3 IVDD-related gene expression profiles were downloaded from the Gene Expression Omnibus database to screen for differentially expressed genes (DEGs). Gene Ontology (GO) and gene set enrichment analysis (GSEA) were conducted to explore the biological functions. Two machine learning algorithms were used to identify characteristic genes, which were tested to further find the key characteristic gene. The receiver operating characteristic curve was performed to estimate the clinical diagnostic value of the key characteristic gene. The excised human intervertebral disks were obtained, and the normal nucleus pulposus (NP) and degenerative NP were carefully separated and cultured in vitro. The expression of the key characteristic gene was validated by real-time quantitative PCR (qRT-PCR). The related protein expression in NP cells was detected by Western blot. Finally, the correlation was investigated between the key characteristic gene and immune cell infiltration. Results A total of 5 DEGs, including 3 upregulated genes and 2 downregulated genes, were screened between IVDD and control samples. GO enrichment analysis showed that DEGs were enriched to 4 items in BP, 6 items in CC, and 13 items in MF. They mainly included the regulation of ion transmembrane transport, transporter complex, and channel activity. GSEA suggested that the cell cycle, DNA replication, graft versus host disease, and nucleotide excision repair were enriched in control samples, while complement and coagulation cascades, Fc γ R-mediated phagocytosis, neuroactive ligand-receptor interaction, the NOD-like receptor signaling pathway, gap junctions, etc., were enriched in IVDD samples. Furthermore, ZNF542P was identified and tested as key characteristic gene in IVDD samples through machine learning algorithms and showed a good diagnostic value. The results of qRT-PCR showed that compared with normal NP cells, the expression of ZNF542P gene was decreased in degenerated NP cells. The results of Western blot suggested that compared with normal NP cells, the expression of NLRP3 and pro Caspase-1 was increased in degenerated NP cells. Finally, we found that the expression of ZNF542P was positively related to the proportions of T cells gamma delta (γδT cells). Conclusion ZNF542P is a potential biomarker in the early diagnosis of IVDD and may be associated with the NOD-like receptor signaling pathway and the infiltration of γδT cells.
Collapse
Affiliation(s)
- Zhao Yang
- *Correspondence: Zhao Yang, ; Xin-Long Ma,
| | | | - Xin-Long Ma
- Department of Orthopedics, Tianjin Hospital, Tianjin, China
| |
Collapse
|
30
|
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: 31] [Impact Index Per Article: 15.5] [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
|
31
|
Giannotta C, Autino F, Massaia M. Vγ9Vδ2 T-cell immunotherapy in blood cancers: ready for prime time? Front Immunol 2023; 14:1167443. [PMID: 37143664 PMCID: PMC10153673 DOI: 10.3389/fimmu.2023.1167443] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2023] [Accepted: 03/31/2023] [Indexed: 05/06/2023] Open
Abstract
In the last years, the tumor microenvironment (TME) has emerged as a promising target for therapeutic interventions in cancer. Cancer cells are highly dependent on the TME to growth and evade the immune system. Three major cell subpopulations are facing each other in the TME: cancer cells, immune suppressor cells, and immune effector cells. These interactions are influenced by the tumor stroma which is composed of extracellular matrix, bystander cells, cytokines, and soluble factors. The TME can be very different depending on the tissue where cancer arises as in solid tumors vs blood cancers. Several studies have shown correlations between the clinical outcome and specific patterns of TME immune cell infiltration. In the recent years, a growing body of evidence suggests that unconventional T cells like natural killer T (NKT) cells, mucosal-associated invariant T (MAIT) cells, and γδ T cells are key players in the protumor or antitumor TME commitment in solid tumors and blood cancers. In this review, we will focus on γδ T cells, especially Vγ9Vδ2 T cells, to discuss their peculiarities, pros, and cons as potential targets of therapeutic interventions in blood cancers.
Collapse
Affiliation(s)
- Claudia Giannotta
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
| | - Federica Autino
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
| | - Massimo Massaia
- Laboratorio di Immunologia dei Tumori del Sangue (LITS), Centro Interdipartimentale di Biotecnologie Molecolari “Guido Tarone”, Dipartimento di Biotecnologie Molecolari e Scienze per la Salute, Università Degli Studi di Torino, Torino, Italy
- Struttura Complessa (SC) Ematologia, Azienda Ospedaliera (AO) S. Croce e Carle, Cuneo, Italy
- *Correspondence: Massimo Massaia,
| |
Collapse
|
32
|
Padmaraju V, Sankla Y, Malla RR. Role of γδ T Cells in Cancer Progression and Therapy. Crit Rev Oncog 2023; 28:59-70. [PMID: 38050982 DOI: 10.1615/critrevoncog.2023050067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
γδ T cells signify a foundational group of immune cells that infiltrate tumors early on, engaging in combat against cancer cells. The buildup of γδ T cells as cancer advances underscores their significance. Initially, these cells infiltrate and enact cytotoxic effects within the tumor tissue. However, in later stages, the predominant phenotype of γδ T cells undergoes changes in numerous cancers, fostering tumor growth and metastasis. Different mechanisms induced by cancer cell suppress effector action of γδ T cells and even sometimes promote cancer progression. In the early stages, stopping this mechanism clears this challenge and enables γδ T cells to effectively remove cancer cells. Given this context, it becomes imperative to delve into the mechanisms of how γδ T cells function in tumor microenvironment. This review discusses γδ T cells' role across different cancer types.
Collapse
Affiliation(s)
- Vasudevaraju Padmaraju
- Department of Biochemistry and Bioinformatics, GITAM School of Science, Department of Biochemistry and Bioinformatics, GITAM School of Science (GSS), GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Yogitha Sankla
- Department of Biochemistry and Bioinformatics, GITAM School of Science, Department of Biochemistry and Bioinformatics, GITAM School of Science (GSS), GITAM (Deemed to be University), Visakhapatnam, Andhra Pradesh, 530045, India
| | - Rama Rao Malla
- Cancer Biology Laboratory, Department of Biochemistry and Bioinformatics, School of Science, Gandhi Institute of Technology and Management (GITAM) (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India; Department of Biochemistry and Bioinformatics, School of Science, GITAM (Deemed to be University), Visakhapatnam-530045, Andhra Pradesh, India
| |
Collapse
|
33
|
γδ T Lymphocytes as a Double-Edged Sword-State of the Art in Gynecological Diseases. Int J Mol Sci 2022; 23:ijms232314797. [PMID: 36499125 PMCID: PMC9740168 DOI: 10.3390/ijms232314797] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Revised: 11/21/2022] [Accepted: 11/22/2022] [Indexed: 12/03/2022] Open
Abstract
Human gamma-delta (γδ) T cells are a heterogeneous cell population that bridges the gap between innate and acquired immunity. They are involved in a variety of immunological processes, including tumor escape mechanisms. However, by being prolific cytokine producers, these lymphocytes also participate in antitumor cytotoxicity. Which one of the two possibilities takes place depends on the tumor microenvironment (TME) and the subpopulation of γδ T lymphocytes. The aim of this paper is to summarize existing knowledge about the phenotype and dual role of γδ T cells in cancers, including ovarian cancer (OC). OC is the third most common gynecological cancer and the most lethal gynecological malignancy. Anticancer immunity in OC is modulated by the TME, including by immunosuppressive cells, cytokines, and soluble factors. Immune cells are exposed in the TME to many signals that determine their immunophenotype and can manipulate their functions. The significance of γδ T cells in the pathophysiology of OC is enigmatic and remains to be investigated.
Collapse
|
34
|
Bustos X, Snedal S, Tordesillas L, Pelle E, Abate-Daga D. γδ T Cell-Based Adoptive Cell Therapies Against Solid Epithelial Tumors. Cancer J 2022; 28:270-277. [PMID: 35880936 PMCID: PMC9335899 DOI: 10.1097/ppo.0000000000000606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
ABSTRACT Conventionally, adoptive cell therapies have been developed and optimized using αβ T cells. However, the understudied and less abundant γδ T cells offer unique advantages to the immunotherapy field especially for therapies against solid tumors. Recently, γδ T-cell potential against a broad spectrum of malignant cells has been demonstrated in the preclinical setting. In the clinic, γδ T-cell-based immunotherapies have proven to be safe; however, their efficacy needs improvement. Considering the growing body of literature reflecting the increasing interest in γδ T cells, we sought to capture the current topics of discussion in the field, pertaining to their use in adoptive immunotherapy. We aimed to compile information about γδ T-cell enhancement in terms of expansion, phenotype, and inhibitory receptors, in addition to the latest advances in preclinical and clinical research using γδ T cells specifically against solid epithelial tumors.
Collapse
|
35
|
Hull CM, Maher J. 3D modelling of γδ T-cell immunotherapy. Clin Transl Med 2022; 12:e877. [PMID: 35538922 PMCID: PMC9091998 DOI: 10.1002/ctm2.877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 04/02/2022] [Accepted: 04/28/2022] [Indexed: 11/07/2022] Open
Affiliation(s)
| | - John Maher
- Leucid Bio Ltd.Guy's HospitalGreat Maze PondLondonUK
- CAR Mechanics LabGuy's Cancer CentreSchool of Cancer and Pharmaceutical SciencesKing's College LondonGreat Maze PondLondonUK
- Department of ImmunologyEastbourne HospitalEastbourneEast SussexUK
| |
Collapse
|
36
|
Immune Cell Metabolic Fitness for Life. Antibodies (Basel) 2022; 11:antib11020032. [PMID: 35645205 PMCID: PMC9149842 DOI: 10.3390/antib11020032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2022] [Revised: 04/07/2022] [Accepted: 04/12/2022] [Indexed: 12/10/2022] Open
Abstract
Adoptive cell therapy holds great promise for treating a myriad of diseases, especially cancer. Within the last decade, immunotherapy has provided a significant leap in the successful treatment of leukemia. The research conducted throughout this period to understand the interrelationships between cancer cells and infiltrating immune cells winds up having one very common feature, bioenergetics. Cancer cells and immune cells both need ATP to perform their individual functions and cancer cells have adopted means to limit immune cell activity via changes in immune cell bioenergetics that redirect immune cell behavior to encourage tumor growth. Current leading strategies for cancer treatment super-charge an individual’s own immune cells against cancer. Successful Chimeric Antigen Receptor T Cells (CAR T) target pathways that ultimately influence bioenergetics. In the last decade, scientists identified that mitochondria play a crucial role in T cell physiology. When modifying T cells to create chimeras, a unique mitochondrial fitness emerges that establishes stemness and persistence. This review highlights many of the key findings leading to this generation’s CAR T treatments and the work currently being done to advance immunotherapy, to empower not just T cells but other immune cells as well against a variety of cancers.
Collapse
|