1
|
Zhao J, Wang X, Zhang H, Zhang Q, Bo D, Zhong H, Jiao L, Yuan H, Lu G. Tumor Cell Membrane Biomimetic Mesoporous Silicon Materials in Combination with PD-L1 Knockout Achieved through the CRISPR/Cas9 System for Targeted and Immunotherapeutic Purposes. Bioconjug Chem 2025. [PMID: 40256973 DOI: 10.1021/acs.bioconjchem.5c00001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/22/2025]
Abstract
Nanoparticle-based drug delivery systems, which enable the effective and targeted delivery of chemotherapeutic drugs to tumors, have revolutionized cancer therapy. Mesoporous silicon materials (MSN) have emerged as promising candidates for drug delivery due to their unique properties. The therapeutic efficacy can be significantly enhanced when treatments exhibit both targeting and antiphagocytic properties. In this study, cell membranes extracted from B16-F10 cells were used to encapsulate carboplatin (CBP)-loaded MSN via physical extrusion. Additionally, we intratumorally injected a plasmid containing the CRISPR/Cas9 system to achieve PD-L1 knockout, thereby reactivating the immune system. The cell membrane coating endowed the CBP@MSN with excellent slow-release capability and cytocompatibility. Enhanced tumor cell uptake of CBP@MSN@M was observed due to homologous targeting by cancer cell membranes. Moreover, CBP@MSN@M demonstrated enhanced antitumor efficacy in vivo and promoted the proliferation of immune cells. Finally, the antitumor effect was further improved by the knockout of PD-L1 within the tumor microenvironment. These results suggest that the newly prepared CBP@MSN@M, combined with PD-L1 knockout, holds significant potential as an effective therapeutic approach for treating tumors.
Collapse
Affiliation(s)
- Jinjin Zhao
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Xiaorou Wang
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Haiguang Zhang
- Department of Gynecology and Obstetrics, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Qunmei Zhang
- Department of Blood Transfusion, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - DeYing Bo
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Hua Zhong
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Luyang Jiao
- Department of Blood Transfusion, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Hongchang Yuan
- Department of Pharmacy, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| | - Guangjian Lu
- Clinical Laboratory, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, Henan 453100, China
| |
Collapse
|
2
|
Zhang W, Wang J, Ji J, Wang P, Yuan G, Fang S, Liu F, Jin G, Zhang J. Glioblastoma cells secrete ICAM1 via FASN signaling to promote glioma-associated macrophage infiltration. Cell Signal 2025; 132:111823. [PMID: 40252818 DOI: 10.1016/j.cellsig.2025.111823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 04/14/2025] [Accepted: 04/16/2025] [Indexed: 04/21/2025]
Abstract
Glioma-associated macrophages (GAMs) constitute the most abundant subset of immune cells in the glioblastoma (GBM) microenvironment, but the underlying mechanism of intense infiltration needs to be elucidated. In this study, we found that GBM cells secrete ICAM1 via FASN signaling to promote GAM infiltration. FASN expression is correlated with GAM density in GBM patients. In vitro experiments revealed that FASN regulates the type-I interferon pathway, particularly STAT1 expression. Moreover, disrupting FASN-STAT1 signaling through the overexpression or inhibition of FASN or STAT1 in GBM cells strongly influences microglial recruitment. Additionally, ICAM1 acts as a direct transcriptional candidate of FASN-STAT1 and a paracrine soluble factor, recruiting microglia to GBM tumors. This study revealed crosstalk between GBM cells and GAMs through FASN-STAT1-ICAM1 signaling to promote microglial infiltration, suggesting potential strategies for treating GBM patients.
Collapse
Affiliation(s)
- Wenxin Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jialin Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Jiayu Ji
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, School of Rehabilitation, Capital Medical University, Beijing, PR China
| | - Peiwen Wang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Guiqiang Yuan
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Sheng Fang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China
| | - Fusheng Liu
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China; Department of Neurosurgery, Beijing Tiantan, Capital Medical University, Beijing, China
| | - Guishan Jin
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| | - Junwen Zhang
- Brain Tumor Research Center, Beijing Neurosurgical Institute, Capital Medical University, Beijing, China.
| |
Collapse
|
3
|
Pherez-Farah A, Boncompagni G, Chudnovskiy A, Pasqual G. The Bidirectional Interplay between T Cell-Based Immunotherapies and the Tumor Microenvironment. Cancer Immunol Res 2025; 13:463-475. [PMID: 39786986 PMCID: PMC7617322 DOI: 10.1158/2326-6066.cir-24-0857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/06/2024] [Accepted: 01/07/2025] [Indexed: 01/12/2025]
Abstract
T cell-based therapies, including tumor-infiltrating lymphocyte therapy, T-cell receptor-engineered T cells, and chimeric antigen receptor T cells, are powerful therapeutic approaches for cancer treatment. Whereas these therapies are primarily known for their direct cytotoxic effects on cancer cells, accumulating evidence indicates that they also influence the tumor microenvironment (TME) by altering the cytokine milieu and recruiting additional effector populations to help orchestrate the antitumor immune response. Conversely, the TME itself can modulate the behavior of these therapies within the host by either supporting or inhibiting their activity. In this review, we provide an overview of clinical and preclinical data on the bidirectional influences between T-cell therapies and the TME. Unraveling the interactions between T cell-based therapies and the TME is critical for a better understanding of their mechanisms of action, resistance, and toxicity, with the goal of optimizing efficacy and safety.
Collapse
Affiliation(s)
- Alfredo Pherez-Farah
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | - Gioia Boncompagni
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
| | | | - Giulia Pasqual
- Laboratory of Synthetic Immunology, Department of Surgery, Oncology and Gastroenterology, University of Padua, Padua, Italy
- Veneto Institute of Oncology IOV IRCCS, Padua, Italy
| |
Collapse
|
4
|
Jiang S, Zheng S, Yao C, Ning D, Zou S, Zhan J, Lan T, Yi T, Jin Z, Wu X. Heterogeneity of γδ T-cell subsets and their clinical correlation in patients with AML. Front Immunol 2025; 16:1552235. [PMID: 40236710 PMCID: PMC11996841 DOI: 10.3389/fimmu.2025.1552235] [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: 12/27/2024] [Accepted: 03/10/2025] [Indexed: 04/17/2025] Open
Abstract
Background γδ T cells are integral elements of the immune system and have shown therapeutic potential in the treatment of acute myeloid leukemia (AML). Nevertheless, the influence of distinct functional subsets, including the activating marker NKG2D, the immune exhaustion marker TIGIT, and the regulatory marker Foxp3, on therapeutic outcomes in AML patients remains unknown. Methods First, we analyzed RNA-seq data from 167 patients in The Cancer Genome Atlas (TCGA) database, concentrating on the correlations between NKG2D, TIGIT, and Foxp3 gene expressions and their association with prognosis in AML. We employed flow cytometry to assess the expression of these molecular markers on γδ T cells and the Vδ1/Vδ2 subsets in the peripheral blood of 25 de novo AML (AML-DN) patients, 15 patients in complete remission (CR), and 27 healthy controls (HCs). We also analyzed the relationship between the expression frequencies of NKG2D, TIGIT, and Foxp3 on γδ T cells and their subsets, and their clinical outcomes. Results Based on data from TCGA database, we found that a high expression level of NKG2D in combination with a low expression level of TIGIT was significantly associated with longer overall survival (OS) in AML patients. Clinical data revealed that γδ T cells from AML-DN patients exhibited higher expression levels of TIGIT and Foxp3, whereas NKG2D expression was lower compared to that of HCs. Notably, the expression of the NKG2D+TIGIT- Vδ1 subset was significantly reduced in AML-DN patients compared to CR patients. Univariate logistic regression and Cox regression analyses further indicated that a high expression of the NKG2D+TIGIT- Vδ1 subset was associated with better clinical prognosis. Conclusion This study indicates that NKG2D+TIGIT- Vδ1 T cells are strongly correlated with improved prognosis in AML, and future research should investigate their potential in adoptive immunotherapy to advance more personalized and precise treatment strategies.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/immunology
- Leukemia, Myeloid, Acute/mortality
- Leukemia, Myeloid, Acute/genetics
- Female
- Male
- Middle Aged
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Adult
- Aged
- Receptors, Immunologic/genetics
- Receptors, Immunologic/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- NK Cell Lectin-Like Receptor Subfamily K/genetics
- NK Cell Lectin-Like Receptor Subfamily K/metabolism
- Prognosis
- Forkhead Transcription Factors/genetics
- Forkhead Transcription Factors/metabolism
- Young Adult
- Intraepithelial Lymphocytes/immunology
Collapse
Affiliation(s)
- Siyuan Jiang
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Shiyu Zheng
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Chao Yao
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Dengchong Ning
- Youjiang Medical University for Nationalities, Baise, China
| | - Shaoyun Zou
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Jiannan Zhan
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
| | - Tianbi Lan
- Dongguan Institute of Clinical Cancer Research, Dongguan Key Laboratory of Precision Diagnosis and Treatment for Tumors, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People’s Hospital), Dongguan, China
| | - Tingzhuang Yi
- Department of Oncology, Affiliated Hospital of YouJiang Medical University for Nationalities/Key Laboratory of Molecular Pathology in Tumors of Guangxi Higher Education Institutions, Baise, China
| | - Zhenyi Jin
- Department of Pathology, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
- Jinan-Puhua Joint Laboratory, Guangzhou, China
| | - Xiuli Wu
- Institute of Hematology, Medical Laboratory Center, School of Medicine, Jinan University, Guangzhou, China
- Key Laboratory of Viral Pathogenesis and Infection Prevention and Control (Jinan University), Ministry of Education, Guangzhou, China
- Jinan-Puhua Joint Laboratory, Guangzhou, China
| |
Collapse
|
5
|
Maul LV, Ramelyte E, Dummer R, Mangana J. Management of metastatic melanoma with combinations including PD-1 inhibitors. Expert Opin Biol Ther 2025:1-12. [PMID: 40159098 DOI: 10.1080/14712598.2025.2485315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
INTRODUCTION Melanoma is among the most immunogenic malignancies. The advent of immune checkpoint inhibitors (ICIs) has revolutionized the landscape of melanoma treatment. Long-term durable cancer control is possible in nearly 50% of non-resectable, metastatic melanoma patients with anti-CTLA4 and anti-PD-1 antibodies. AREAS COVERED This review provides a critical overview of the current data and future research directions on the management of metastatic melanoma with ICIs. We reviewed the efficacy and safety of combinations with PD-1 inhibitors through PubMed database research (Nov 2024-Mar 2025). EXPERT OPINION A decade after ipilimumab's approval, challenges remain. To cure more patients, the development of combinations is warranted. Combinations with a limited number of ipilimumab applications improve the overall survival outcome by approximately 10%, with a dramatic increase in adverse events including fatal events. Anti-LAG3/nivolumab is a promising alternative, offering similar efficacy to ipilimumab/nivolumab with better tolerability. In our opinion, ipilimumab/nivolumab combination should be the first-line therapy for high-risk patients (high LDH, brain or liver metastasis), while nivolumab/relatlimab or PD-1 monotherapy may be preferable for lower-risk cases. However, treatment decisions are increasingly complex, since most patients nowadays are pretreated in the (neo)-adjuvant setting. The key limitation today is the lack of biomarkers to guide individualized treatment strategies.
Collapse
Affiliation(s)
- Lara Valeska Maul
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Egle Ramelyte
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Reinhard Dummer
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| | - Joanna Mangana
- Department of Dermatology, University Hospital Zurich, Zurich, Switzerland
- Faculty of Medicine, University of Zurich, Zurich, Switzerland
| |
Collapse
|
6
|
Lv J, Liu Z, Ren X, Song S, Zhang Y, Wang Y. γδT cells, a key subset of T cell for cancer immunotherapy. Front Immunol 2025; 16:1562188. [PMID: 40226616 PMCID: PMC11985848 DOI: 10.3389/fimmu.2025.1562188] [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: 01/17/2025] [Accepted: 03/14/2025] [Indexed: 04/15/2025] Open
Abstract
γδT cells represent a unique and versatile subset of T cells characterized by the expression of T-cell receptors (TCRs) composed of γ and δ chains. Unlike conventional αβT cells, γδT cells do not require major histocompatibility complex (MHC)-dependent antigen presentation for activation, enabling them to recognize and respond to a wide array of antigens, including phosphoantigens, stress-induced ligands, and tumor-associated antigens. While γδT cells are relatively rare in peripheral blood, they are enriched in peripheral tissues such as the skin, intestine, and lung. These cells play a crucial role in tumor immunotherapy by exerting direct cytotoxicity through the production of inflammatory cytokines (e.g., interferon-gamma (IFN-γ), tumor necrosis factor-alpha (TNF-α), and interleukin-17 (IL-17)) and cytotoxic molecules (e.g., perforin and granzyme). Recent advances in γδT cell research have elucidated their mechanisms of tumor recognition, including the detection of phosphoantigens and stress-induced ligands like MICA (MHC class I polypeptide-related sequence A), MICB (MHC class I polypeptide-related sequence B), and ULBP (UL16-binding protein). Furthermore, various strategies to enhance γδT cell-based tumor immunotherapy have been developed, such as in vitro expansion using phosphoantigen-based therapies, cytokine stimulation, and chimeric antigen receptor (CAR)-γδT cell engineering. These advancements have shown promising results in both preclinical and clinical settings, paving the way for γδT cells to become a powerful tool in cancer immunotherapy. This review highlights the key mechanisms, functions, and strategies to harness the potential of γδT cells for effective tumor immunotherapy.
Collapse
Affiliation(s)
- Jianzhen Lv
- Guangxi Key Laboratory of Efficacy Study on Chinese Materia Medica, Institute of Traditional Chinese and Zhuang-Yao Ethnic Medicine, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Zheng Liu
- Pathology Department, University of Texas MD Anderson Cancer Center, Houston, TX, United States
| | - Xiangting Ren
- Medical School, Guangxi University of Chinese Medicine, Nanning, Guangxi, China
| | - Siyuan Song
- Department of Neuroscience, Baylor College of Medicine, Houston, TX, United States
| | - Yan Zhang
- Department of Geriatrics, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, Sichuan, China
- Clinical Immunology Translational Medicine Key Laboratory of Sichuan Province, Center of Organ Transplantation, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, Chengdu, Sichuan, China
| |
Collapse
|
7
|
Cieslak SG, Shahbazi R. Gamma delta T cells and their immunotherapeutic potential in cancer. Biomark Res 2025; 13:51. [PMID: 40148988 PMCID: PMC11951843 DOI: 10.1186/s40364-025-00762-6] [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: 12/18/2024] [Accepted: 03/07/2025] [Indexed: 03/29/2025] Open
Abstract
Gamma-delta (γδ) T cells are a unique subset of T lymphocytes that play diverse roles in immune responses, bridging innate and adaptive immunity. With growing interest in their potential for cancer immunotherapy, a comprehensive and inclusive exploration of γδ T cell families, their development, activation mechanisms, functions, therapeutic implications, and current treatments is essential. This review aims to provide an inclusive and thorough discussion of these topics. Through our discussion, we seek to uncover insights that may harbinger innovative immunotherapeutic strategies. Beginning with an overview of γδ T cell families including Vδ1, Vδ2, and Vδ3, this review highlights their distinct functional properties and contributions to anti-tumor immunity. Despite γδ T cells exhibiting both anti-tumor and pro-tumor activities, our review elucidates strategies to harness the anti-tumor potential of γδ T cells for therapeutic benefit. Moreover, our paper discusses the structural intricacies of the γδ T cell receptor and its significance in tumor recognition. Additionally, this review examines conventional and emerging γδ T cell therapies, encompassing both non-engineered and engineered approaches, with a focus on their efficacy and safety profiles in clinical trials. From multifunctional capabilities to diverse tissue distribution, γδ T cells play a pivotal role in immune regulation and surveillance. By analyzing current research findings, this paper offers insights into the dynamic landscape of γδ T cell-based immunotherapies, underscoring their promise as a potent armamentarium against cancer. Furthermore, by dissecting the complex biology of γδ T cells, we learn valuable information about the anti-cancer contributions of γδ T cells, as well as potential targets for immunotherapeutic interventions.
Collapse
Affiliation(s)
- Stephen G Cieslak
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA
| | - Reza Shahbazi
- Division of Hematology/Oncology, Department of Medicine, Indiana University, Indianapolis, IN, USA.
- Department of Biochemistry and Molecular Biology, Indiana University, Indianapolis, IN, USA.
- Tumor Microenvironment & Metastasis, Indiana University Melvin and Bren Simon Comprehensive Cancer Center, Indianapolis, IN, USA.
- Brown Center for Immunotherapy, Indiana University, Indianapolis, IN, USA.
| |
Collapse
|
8
|
Zhang L, Zhao J, Su C, Wu J, Jiang L, Chi H, Wang Q. Organoid models of ovarian cancer: resolving immune mechanisms of metabolic reprogramming and drug resistance. Front Immunol 2025; 16:1573686. [PMID: 40191206 PMCID: PMC11968360 DOI: 10.3389/fimmu.2025.1573686] [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: 02/09/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Metabolic reprogramming is a hallmark of ovarian cancer, enabling tumor progression, immune evasion and drug resistance. The tumor microenvironment (TME) further shapes metabolic adaptations, enabling cancer cells to withstand hypoxia and nutrient deprivation. While organoid models provide a physiologically relevant platform for studying these processes, they still lack immune and vascular components, limiting their ability to fully recapitulate tumor metabolism and drug responses. In this study, we investigated the key metabolic mechanisms involved in ovarian cancer progression, focusing on glycolysis, lipid metabolism and amino acid metabolism. We integrated metabolomic analyses and drug sensitivity assays to explore metabolic-TME interactions using patient-derived, adult stem cell-derived and iPSC-derived organ tissues. Among these, we found that glycolysis, lipid metabolism and amino acid metabolism play a central role in tumor progression and chemotherapy resistance. We identified methylglyoxal (MGO)-mediated BRCA2 dysfunction as a driver of immune escape, a role for sphingolipid signaling in tumor proliferation and a role for kynurenine metabolism in CD8+ T cell suppression. In addition, PI3K/AKT/mTOR and Wnt/β-catenin pathways promote chemoresistance through metabolic adaptation. By elucidating the link between metabolic reprogramming and immune evasion, this study identifies key metabolic vulnerabilities and potential drug targets in ovarian cancer. Our findings support the development of metabolically targeted therapies and increase the utility of organoid-based precision medicine models.
Collapse
Affiliation(s)
- Lanyue Zhang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jiangnan Zhao
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Chunyu Su
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Jianxi Wu
- Department of Preventive Medicine, Southwest Medical University, Luzhou, China
| | - Lai Jiang
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Hao Chi
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Qin Wang
- Sichuan Provincial Center for Gynecology and Breast Diseases (Gynecology), Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
9
|
Li Y, Chen D, Xu Y, Ding Q, Xu X, Li Y, Mi Y, Chen Y. Prognostic implications, genomic and immune characteristics of lung adenocarcinoma with lepidic growth pattern. J Clin Pathol 2025; 78:277-284. [PMID: 39097406 DOI: 10.1136/jcp-2024-209603] [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: 04/24/2024] [Accepted: 07/17/2024] [Indexed: 08/05/2024]
Abstract
AIMS Conflicting data were provided regarding the prognostic impact and genomic features of lung adenocarcinoma (LUAD) with lepidic growth pattern (LP+A). Delineation of the genomic and immune characteristics of LP+A could provide deeper insights into its prognostic implications and treatment determination. METHODS We conducted a search of articles in PubMed, EMBASE and the Cochrane Library from inception to January 2024. A domestic cohort consisting of 52 LUAD samples was subjected to whole-exome sequencing as internal validation. Data from The Cancer Genomic Atlas and the Gene Expression Omnibus datasets were obtained to characterise the genomic and immune profiles of LP+A. Pooled HRs and rates were calculated. RESULTS The pooled results indicated that lepidic growth pattern was either predominant (0.35, 95% CI 0.22 to 0.56, p<0.01) or minor (HR 0.50, 95% CI 0.36 to 0.70, p<0.01) histological subtype was associated with favourable disease-free survival. Pooled gene mutation rates suggested higher EGFR mutation (0.55, 95% CI 0.46 to 0.64, p<0.01) and lower KRAS mutation (0.14, 95% CI 0.02 to 0.25, p=0.02) in lepidic-predominant LUAD. Lepidic-predominant LUAD had lower tumour mutation burden and pooled positive rate of PD-L1 expression compared with other subtypes. LP+A was characterised by abundance in resting CD4+memory T cells, monocytes and γδ T cells, as well as scarcity of cancer-associated fibroblasts. CONCLUSIONS LP+A was a unique histological subtype with a higher EGFR mutation rate, lower tumour mutation burden and immune checkpoint expression levels. Our findings suggested potential benefits from targeted therapy over immunotherapy in LP+A.
Collapse
Affiliation(s)
- Yue Li
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Donglai Chen
- Department of Thoracic Surgery, Zhongshan Hospital Fudan University, Shanghai, Shanghai, China
| | - Yi Xu
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Qifeng Ding
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Xuejun Xu
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yongzhong Li
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yedong Mi
- Department of Thoracic Surgery, Jiangyin People's Hospital, Jiangyin, Jiangsu, China
| | - Yongbing Chen
- Department of Thoracic Surgery, Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| |
Collapse
|
10
|
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] [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
|
11
|
Zarobkiewicz M, Lehman N, Morawska-Michalska I, Michalski A, Kowalska W, Szymańska A, Tomczak W, Bojarska-Junak A. Characterisation of Cytotoxicity-Related Receptors on γδ T Cells in Chronic Lymphocytic Leukaemia. Cells 2025; 14:451. [PMID: 40136700 PMCID: PMC11941621 DOI: 10.3390/cells14060451] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/14/2025] [Accepted: 03/16/2025] [Indexed: 03/27/2025] Open
Abstract
Chronic lymphocytic leukaemia (CLL) is a haematological malignancy primarily affecting older adults, characterised by the proliferation of functionally impaired B lymphocytes with abnormal expression of CD5, a typical T cell marker. The current study investigates the expression of cytotoxicity-related receptors (CD16, CD56, CD57, CD69) and a checkpoint (LAG-3) on γδ T cells in CLL patients. Sixty-nine treatment-naive CLL patients and fourteen healthy controls were recruited. Flow cytometry analysis revealed that the CLL patients had higher expressions of CD56 and LAG-3 and lower CD16 on their γδ T cells compared to the healthy controls. Subgroup analysis showed that ZAP-70-negative patients exhibited increased CD69, while CD38-negative patients showed higher CD16 expression. Additionally, CD16 expression was inversely correlated with serum LDH levels, a marker of disease progression. Bioinformatic analysis of the LAG-3 ligand mRNA in a CLL dataset indicated higher expression of HLA-DQA2 and HLA-DRB5 in patients with unmutated IGVH. Our findings highlight the altered expression of key cytotoxicity markers on γδ T cells in CLL, suggesting their potential role in disease progression and as a therapeutic target. In particular, the use of anti-LAG-3 antibodies seems promising.
Collapse
MESH Headings
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Lymphocytic, Chronic, B-Cell/blood
- Male
- Female
- Aged
- Middle Aged
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Aged, 80 and over
- Antigens, CD/metabolism
- Antigens, CD/genetics
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Case-Control Studies
Collapse
Affiliation(s)
- Michał Zarobkiewicz
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| | - Natalia Lehman
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| | - Izabela Morawska-Michalska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| | - Adam Michalski
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| | - Wioleta Kowalska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| | - Agata Szymańska
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| | - Waldemar Tomczak
- Department of Haematooncology and Bone Marrow Transplantation, Medical University of Lublin, 20-080 Lublin, Poland;
| | - Agnieszka Bojarska-Junak
- Department of Clinical Immunology, Medical University of Lublin, 20-093 Lublin, Poland; (N.L.); (I.M.-M.); (A.M.); (W.K.); (A.S.); (A.B.-J.)
| |
Collapse
|
12
|
Yan B, Chen Q, Wang D, Ding L, Qu J, Du R, Shi W, Kahlert UD, Yu Z. Artificial intelligence-based radiogenomics reveals the potential immunoregulatory role of COL22A1 in glioma and its induced autoimmune encephalitis. Front Immunol 2025; 16:1562070. [PMID: 40114922 PMCID: PMC11922723 DOI: 10.3389/fimmu.2025.1562070] [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: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Background The tumor microenvironment plays a crucial role in the progression of both glioma and glioma-induced autoimmune encephalitis. However, there remains a significant lack of effective therapeutic targets for these diseases. Method We collected 54 CT images of glioma patients and 54 glioma-induced autoimmune encephalitis patients, respectively. Radiomics features were extracted from tumors and encephalitis regions using Python, followed by dimensionality reduction via random forest and lasso regression, and construction of radiomics-based risk scores. Genomic data matched with clinical information were analyzed to identify key prognostic genes significantly associated with risk scores. Gene expression was validated by immunohistochemistry using our clinical samples. Immune infiltration was evaluated using five algorithms (MCP-counter, EPIC, TIMER, QUANT and IPS). The association between hub genes and immune checkpoint markers as well as immunoregulation-related genes was also analyzed using Spearman correlation. Results We identified 980 radiomics features both in glioma and encephalitis patient images and selected four key features through lasso regression to build a radiomics-based risk score. COL22A1 was strongly correlated with the risk score and identified as the hub prognostic gene. COL22A1 expression was higher in glioblastoma tissues and cell lines, and correlated with clinical factors such as higher age, WHO grade, and IDH mutation status. Immune infiltration analysis indicated associations with diverse immune and stromal cell populations, including CD8+T cells, macrophages, and CAFs. COL22A1 was also positively correlated with immune checkpoints and immune-regulated genes. Conclusion Our study highlights the critical role of COL22A1 in gliomas and glioma-Induced Autoimmune Encephalitis, demonstrating its strong association with poor prognosis and its significant involvement in tumor immune regulation.
Collapse
Affiliation(s)
- Bingchao Yan
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
- Department of Neurosurgery, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, China
| | - Qian Chen
- Department of Neurosurgery, The Second Affiliated Hospital of Guilin Medical University, Guangxi, China
| | - Dacheng Wang
- Department of Neurosurgery, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, China
| | - Leili Ding
- Molecular and Experimental Surgery, Clinic for General-, Visceral -, Vascular- and Transplantation Surgery, Medical Faculty and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
- Nantong University, Nantong, China
| | - Jingfeng Qu
- Department of Neurosurgery, The Affiliated Xuzhou Municipal Hospital of Xuzhou Medical University, Xuzhou, China
| | - Renfei Du
- Molecular and Experimental Surgery, Clinic for General-, Visceral -, Vascular- and Transplantation Surgery, Medical Faculty and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
- Chifeng Municipal Hospital, Chifeng, China
| | - Wenjie Shi
- Molecular and Experimental Surgery, Clinic for General-, Visceral -, Vascular- and Transplantation Surgery, Medical Faculty and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - Ulf D Kahlert
- Molecular and Experimental Surgery, Clinic for General-, Visceral -, Vascular- and Transplantation Surgery, Medical Faculty and University Hospital Magdeburg, Otto-von-Guericke University, Magdeburg, Germany
| | - Zhengquan Yu
- Department of Neurosurgery, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
13
|
Ma X, Xu J, Wang Y, Fleishman JS, Bing H, Yu B, Li Y, Bo L, Zhang S, Chen ZS, Zhao L. Research progress on gene mutations and drug resistance in leukemia. Drug Resist Updat 2025; 79:101195. [PMID: 39740374 DOI: 10.1016/j.drup.2024.101195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/05/2024] [Accepted: 12/20/2024] [Indexed: 01/02/2025]
Abstract
Leukemia is a type of blood cancer characterized by the uncontrolled growth of abnormal cells in the bone marrow, which replace normal blood cells and disrupt normal blood cell function. Timely and personalized interventions are crucial for disease management and improving survival rates. However, many patients experience relapse following conventional chemotherapy, and increasing treatment intensity often fails to improve outcomes due to mutated gene-induced drug resistance in leukemia cells. This article analyzes the association of gene mutations and drug resistance in leukemia. It explores genetic abnormalities in leukemia, highlighting recently identified mutations affecting signaling pathways, cell apoptosis, epigenetic regulation, histone modification, and splicing mechanisms. Additionally, the article discusses therapeutic strategies such as molecular targeting of gene mutations, alternative pathway targeting, and immunotherapy in leukemia. These approaches aim to combat specific drug-resistant mutations, providing potential avenues to mitigate leukemia relapse. Future research with these strategies holds promise for advancing leukemia treatment and addressing the challenges of drug-resistant mutations to improve patient outcomes.
Collapse
Affiliation(s)
- Xiangyu Ma
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Jiamin Xu
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Yanan Wang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Joshua S Fleishman
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Hao Bing
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Boran Yu
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Yanming Li
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China
| | - Letao Bo
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA
| | - Shaolong Zhang
- Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, New York, NY 11439, USA.
| | - Libo Zhao
- Department of Pharmacy, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, 100045, China; Department of Pharmacy, Peking University Third Hospital, Beijing 100191, China.
| |
Collapse
|
14
|
Zeng M, Lin A, Jiang A, Qiu Z, Zhang H, Chen S, Xu M, Liu Z, Cheng Q, Zhang J, Luo P. Decoding the mechanisms behind second primary cancers. J Transl Med 2025; 23:115. [PMID: 39856672 PMCID: PMC11762917 DOI: 10.1186/s12967-025-06151-9] [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: 08/19/2024] [Accepted: 01/19/2025] [Indexed: 01/27/2025] Open
Abstract
Second Primary Cancers (SPCs) are defined as cancers that develop either simultaneously or metachronously in the same individual who has been diagnosed with and survived one primary cancer. SPCs exhibit a high incidence rate and represent the primary cause of mortality among survivors of first primary cancers. There is growing concern about the dangers of SPCs. This review summarizes recent studies on the mechanisms of SPCs, including the roles of genomic changes after first primary cancer (FPC) treatments, stromal cell phenotypic and metabolic changes, hormone levels and receptor expression, immunosuppression, aberrant gene methylation, EGFR signaling, and cell-free DNA in SPC development. This comprehensive analysis contributes to elucidating current research trends in SPC mechanisms and enhances our understanding of the underlying pathophysiology. Furthermore, potential applications of intratumoral microbes, single-cell multi-omics, and metabolomics in investigating SPC mechanisms are also discussed, providing new ideas for follow-up studies.
Collapse
Affiliation(s)
- Meiyuan Zeng
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhengang Qiu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Hongman Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China
| | - Shifu Chen
- HaploX Biotechnology, Shenzhen, China
- Faculty of Data Science, City University of Macau, Macau, China
| | | | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100730, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, People's Republic of China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Hunan, China.
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, 253 Industrial Avenue, Guangzhou, 510282, Guangdong, China.
| |
Collapse
|
15
|
Zhang C, Zhang X, Dai S, Yang W. Exploring prognosis and therapeutic strategies for HBV-HCC patients based on disulfidptosis-related genes. Front Genet 2025; 15:1522484. [PMID: 39882072 PMCID: PMC11774838 DOI: 10.3389/fgene.2024.1522484] [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: 11/04/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) accounts for over 80% of primary liver cancers and is the third leading cause of cancer-related deaths worldwide. Hepatitis B virus (HBV) infection is the primary etiological factor. Disulfidptosis is a newly discovered form of regulated cell death. This study aims to develop a novel HBV-HCC prognostic signature related to disulfidptosis and explore potential therapeutic approaches through risk stratification based on disulfidptosis. Methods Transcriptomic data from HBV-HCC patients were analyzed to identify BHDRGs. A prognostic model was established and validated using machine learning, with internal datasets and external datasets for verification. We then performed immune cell infiltration analysis, tumor microenvironment (TME) analysis, and immunotherapy-related analysis based on the prognostic signature. Besides, RT-qPCR and immunohistochemistry were conducted. Results A prognostic model was constructed using five genes (DLAT, STC2, POF1B, S100A9, and CPS1). A corresponding prognostic nomogram was developed based on riskScores, age, stage. Stratification by median risk score revealed a significant correlation between the prognostic signature and TME, tumor immune cell infiltration, immunotherapy efficacy, and drug sensitivity. The results of the experiments indicate that DLAT expression is higher in tumor tissues compared to adjacent tissues. DLAT expression is higher in HBV-HCC tumor tissues compared to normal tissues. Conclusion This study stratifies HBV-HCC patients into distinct subgroups based on BHDRGs, establishing a prognostic model with significant implications for prognosis assessment, TME remodeling, and personalized therapy in HBV-HCC patients.
Collapse
Affiliation(s)
| | | | - Shengjie Dai
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Wenjun Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
16
|
Wang J, Zhang H, Li J, Ni X, Yan W, Chen Y, Shi T. Exosome-derived proteins in gastric cancer progression, drug resistance, and immune response. Cell Mol Biol Lett 2024; 29:157. [PMID: 39719600 PMCID: PMC11667977 DOI: 10.1186/s11658-024-00676-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 12/09/2024] [Indexed: 12/26/2024] Open
Abstract
Gastric cancer (GC) represents a prevalent malignancy globally, often diagnosed at advanced stages owing to subtle early symptoms, resulting in a poor prognosis. Exosomes are extracellular nano-sized vesicles and are secreted by various cells. Mounting evidence indicates that exosomes contain a wide range of molecules, such as DNA, RNA, lipids, and proteins, and play crucial roles in multiple cancers including GC. Recently, with the rapid development of mass spectrometry-based detection technology, researchers have paid increasing attention to exosomal cargo proteins. In this review, we discussed the origin of exosomes and the diagnostic and prognostic roles of exosomal proteins in GC. Moreover, we summarized the biological functions of exosomal proteins in GC processes, such as proliferation, metastasis, drug resistance, stemness, immune response, angiogenesis, and traditional Chinese medicine therapy. In summary, this review synthesizes current advancements in exosomal proteins associated with GC, offering insights that could pave the way for novel diagnostic and therapeutic strategies for GC in the foreseeable future.
Collapse
Affiliation(s)
- Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Huan Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
| | - Juntao Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xiangyu Ni
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenying Yan
- Department of Bioinformatics, School of Biology and Basic Medical Sciences, Suzhou Medical College of Soochow University, 199 Renai Road, Suzhou, 215123, China.
- Center for Systems Biology, Soochow University, Suzhou, China.
- Jiangsu Province Engineering Research Center of Precision Diagnostics and Therapeutics Development, Suzhou, China.
| | - Yueqiu Chen
- Department of Cardiovascular Surgery of The First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou, 215007, China.
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, 178 East Ganjiang Road, Suzhou, 215000, China.
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
17
|
Zhu J, Meganathan I, MacAruthur R, Kassiri Z. Inflammation in Abdominal Aortic Aneurysm: Cause or Comorbidity? Can J Cardiol 2024; 40:2378-2391. [PMID: 39181326 DOI: 10.1016/j.cjca.2024.08.274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024] Open
Abstract
Aortic aneurysm is a potentially deadly disease. It is chronic degeneration of the aortic wall that involves an inflammatory response and the immune system, aberrant remodelling of the extracellular matrix, and maladaptive transformation of the aortic cells. This review article focuses on the role of the inflammatory cells in abdominal aortic aneurysm. Studies in human aneurysmal specimens and animal models have identified various inflammatory cell types that could contribute to formation or expansion of aneurysms. These include the commonly studied leukocytes (neutrophils and macrophages) as well as the less commonly explored natural killer cells, dendritic cells, T cells, and B cells. Despite the well-demonstrated contribution of inflammatory cells and the related signalling pathways to development and expansion of aneurysms, anti-inflammatory therapy approaches have demonstrated limitations and may require additional considerations such as a combinational approach in targeting multiple pathways for significant beneficial outcomes.
Collapse
Affiliation(s)
- Jiechun Zhu
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Ilamaran Meganathan
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada
| | - Roderick MacAruthur
- Department of Cardiac Surgery, Mazankowski Alberta Heart Institute, University of Alberta Hospital, Edmonton, Alberta, Canada
| | - Zamaneh Kassiri
- Department of Physiology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| |
Collapse
|
18
|
Yu Z, Tang X, Chen Z, Hu Y, Zhang S, Guo C, Gu J, Shi Y, Gong Y. Role of ADAM10/17-Mediated Cleavage of LAG3 in the Impairment of Immunosuppression in Psoriasis. J Invest Dermatol 2024:S0022-202X(24)02948-8. [PMID: 39571889 DOI: 10.1016/j.jid.2024.10.606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 12/25/2024]
Abstract
Despite extensive research on immune activation regulatory mechanisms, studies on immune suppression in psoriasis are limited. LAG3, a newly identified immune checkpoint, plays a crucial role in modulating immune responses and maintaining T-regulatory cell function. However, its involvement in psoriasis is unclear. We show that psoriasis is associated with reduced LAG3 expression in CD4 T cells and T-regulatory cells. Further analysis revealed that the decline in LAG3 levels was linked to ADAM10/17-mediated proteolytic cleavage, which was upregulated in psoriasis. Clinical utilization of the IL-17A antagonist secukinumab, along with the in vivo and in vitro IL-17A-induced models, supported the potential of IL-17A to induce ADAM10/17 expression and trigger LAG3 cleavage. Through the Jurkat cell model, IL-17A was found to regulate ADAM10/17 expression by activating FOXM1. In addition, treatment with the ADAM10/17 inhibitor GW280264X showed ameliorative effects on psoriasis-like mouse models and lipopolysaccharide-induced inflammation. Collectively, the findings of this study uncover the immune regulatory role of the ADAM10/17-LAG3 axis in psoriasis and highlight the therapeutic potential of targeting ADAM10/17 for psoriasis treatment.
Collapse
Affiliation(s)
- Zengyang Yu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Xinyi Tang
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Zeyu Chen
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yifan Hu
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Shuqin Zhang
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Chunyuan Guo
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Jun Gu
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China
| | - Yuling Shi
- Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China; Department of Dermatology, Shanghai Skin Disease Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China.
| | - Yu Gong
- Department of Dermatology, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, People's Republic of China; Institute of Psoriasis, Tongji University School of Medicine, Shanghai, People's Republic of China.
| |
Collapse
|
19
|
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
|
20
|
Liu J, Wu M, Yang Y, Mei X, Wang L, Wang J, Wang Z, He S, Liu H, Jiang H, Qu S, Zhang Y, Chen Y, Tian X, Huang Y, Wang H. BTN3A1 expressed in cervical cancer cells promotes Vγ9Vδ2 T cells exhaustion through upregulating transcription factors NR4A2/3 downstream of TCR signaling. Cell Commun Signal 2024; 22:459. [PMID: 39342337 PMCID: PMC11439235 DOI: 10.1186/s12964-024-01834-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/17/2024] [Indexed: 10/01/2024] Open
Abstract
BACKGROUND Clinical trials have shown that immunotherapy based on Vγ9Vδ2 T cells (Vδ2 T cells) is safe and well-tolerated for various cancers including cervical cancer (CC), but its overall treatment efficacy remains limited. Therefore, exploring the mechanisms underlying the suboptimal efficacy of Vδ2 T cell-based cancer immunotherapy is crucial for enabling its successful clinical translation. METHODS Tumor samples from CC patients and CC cell line-derived xenograft (CDX) mice were analyzed using flow cytometry to examine the exhausted phenotype of tumor-infiltrating Vδ2 T cells. The interrelationship between BTN3A1 expression and Vδ2 T cells in CC, along with their correlation with patient prognosis, was analyzed using data from The Cancer Genome Atlas (TCGA) database. CC cell lines with BTN3A1 knockout (KO) and overexpression (OE) were constructed through lentivirus transduction, which were then co-cultured with expanded Vδ2 T cells, followed by detecting the function of Vδ2 T cells using flow cytometry. The pathways and transcription factors (TFs) related to BTN3A1-induced Vδ2 T cells exhaustion and the factors affecting BTN3A1 expression were identified by RNA-seq analysis, which was confirmed by flow cytometry, Western Blot, and gene manipulation. RESULTS Tumor-infiltrating Vδ2 T cells exhibited an exhausted phenotype in both CC patients and CDX mice. BTN3A1 expressed in CC is highly enhancing exhaustion markers, while reducing the secretion of effector molecules in Vδ2 T cells. Blocking TCR or knocking down nuclear receptor subfamily 4 group A (NR4A) 2/3 can reverse BTN3A1-induced exhaustion in Vδ2 T cells. On the other hand, IFN-γ secreted by Vδ2 T cells promoted the expression of BTN3A1 and PD-L1. CONCLUSIONS Through binding γδ TCRs, BTN3A1 expressed on tumor cells, which is induced by IFN-γ, can promote Vδ2 T cells to upregulate the expression of TFs NR4A2/3, thereby affecting their activation and expression of exhaustion-related molecules in the tumor microenvironment (TME). Therefore, targeting BTN3A1 might overcome the immunosuppressive effect of the TME on Vδ2 T cells in CC.
Collapse
MESH Headings
- Humans
- Uterine Cervical Neoplasms/genetics
- Uterine Cervical Neoplasms/pathology
- Uterine Cervical Neoplasms/immunology
- Uterine Cervical Neoplasms/metabolism
- Female
- Animals
- Up-Regulation
- Signal Transduction
- Mice
- Cell Line, Tumor
- Butyrophilins/genetics
- Butyrophilins/metabolism
- DNA-Binding Proteins/genetics
- DNA-Binding Proteins/metabolism
- Receptors, Thyroid Hormone/genetics
- Receptors, Thyroid Hormone/metabolism
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Gene Expression Regulation, Neoplastic
- Receptors, Steroid
Collapse
Affiliation(s)
- Jian Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xinyu Mei
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Liming Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jingyu Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixuan Wang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan He
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hangyu Liu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Han Jiang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shen Qu
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwei Zhang
- School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ying Chen
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xun Tian
- Department of Obstetrics and Gynecology, Academician Expert Workstation, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, 430014, China.
| | - Yafei Huang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Huazhong University of Science and Technology, Wuhan, China.
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Zhejiang Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
21
|
Liu R, Zhao H, Lu Z, Zeng L, Shi H, Wu L, Wang J, Zhong F, Liu C, Zhang Y, Qiu Z. Toxicity profiles of immune checkpoint inhibitors in nervous system cancer: a comprehensive disproportionality analysis using FDA adverse event reporting system. Clin Exp Med 2024; 24:216. [PMID: 39249163 PMCID: PMC11383843 DOI: 10.1007/s10238-024-01403-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 06/12/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Immune-related adverse events (irAEs) always occur during treatment with immune checkpoint inhibitors (ICIs). Patients with nervous system cancer (NSC) may gain clinical benefit from ICIs, but irAEs in NSC patients are rarely examined. Therefore, our study systematically summarized reports of irAEs in NSC. METHODS We obtained information from the FDA adverse event reporting system from the first quarter (Q1) of 2013 to the fourth quarter (Q4) of 2022. We examined use of a combination of ICIs and chemotherapy (ICI_Chemo) or chemotherapy only (ICI_Chemo) for patients with NSC. Multiple disproportionality analyses were applied to assess irAEs. Multiomics data from the gene expression omnibus (GEO) database were analyzed to explore potential molecular mechanisms associated with irAEs in NSC patients. RESULTS Fourteen irAEs were identified in 8,357 NSC patients after removing duplicates; the top five events were seizure, confused state, encephalopathy, muscular weakness and gait disturbance. Older patients were more likely to develop irAEs than were younger patients. From the start of ICIs_Chemo to irAE occurrence, there was a significant difference in the time to onset of irAEs between age groups. irAEs may occur via mechanisms involving the inflammatory response, secretion of inflammatory mediators, and aberrant activation of pathologic pathways. CONCLUSIONS This study helps to characterize irAEs in NSC patients treated with ICIs. We combined GEO database analysis to explore the potential molecular mechanisms of irAEs. The results of this study provide a basis for improving the toxic effects of ICIs in NSC patients.
Collapse
Affiliation(s)
- Rongrong Liu
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Hui Zhao
- Department of Sleep Medicine, Ganzhou People's Hospital, Ganzhou, Jiangxi, China
| | - Zenghong Lu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Lingshuai Zeng
- Major of Rehabilitation, Faculty of Medicine, Jinggangshan University, Ji'an, Jiangxi, China
| | - Huaqiu Shi
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Longqiu Wu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Jing Wang
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Fangjun Zhong
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Chuanjian Liu
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China
| | - Yu Zhang
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
| | - Zhengang Qiu
- Department of Neurology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
- Department of Oncology, The First Affiliated Hospital of Gannan Medical University, Ganzhou, Jiangxi, China.
| |
Collapse
|
22
|
Kang K, Lin X, Chen P, Liu H, Liu F, Xiong W, Li G, Yi M, Li X, Wang H, Xiang B. T cell exhaustion in human cancers. Biochim Biophys Acta Rev Cancer 2024; 1879:189162. [PMID: 39089484 DOI: 10.1016/j.bbcan.2024.189162] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 07/23/2024] [Accepted: 07/24/2024] [Indexed: 08/04/2024]
Abstract
T cell exhaustion refers to a progressive state in which T cells become functionally impaired due to sustained antigenic stimulation, which is characterized by increased expression of immune inhibitory receptors, but weakened effector functions, reduced self-renewal capacity, altered epigenetics, transcriptional programme and metabolism. T cell exhaustion is one of the major causes leading to immune escape of cancer, creating an environment that supports tumor development and metastatic spread. In addition, T cell exhaustion plays a pivotal role to the efficacy of current immunotherapies for cancer. This review aims to provide a comprehensive view of roles of T cell exhaustion in cancer development and progression. We summerized the regulatory mechanisms that involved in T cell exhaustion, including transcription factors, epigenetic and metabolic reprogramming events, and various microenvironmental factors such as cytokines, microorganisms, and tumor autocrine substances. The paper also discussed the challenges posed by T cell exhaustion to cancer immunotherapies, including immune checkpoint blockade (ICB) therapies and chimeric antigen receptor T cell (CAR-T) therapy, highlightsing the obstacles encountered in ICB therapies and CAR-T therapies due to T cell exhaustion. Finally, the article provides an overview of current therapeutic options aimed to reversing or alleviating T cell exhaustion in ICB and CAR-T therapies. These therapeutic approaches seek to overcome T cell exhaustion and enhance the effectiveness of immunotherapies in treating tumors.
Collapse
Affiliation(s)
- Kuan Kang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Xin Lin
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Pan Chen
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China
| | - Huai Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Feng Liu
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China
| | - Wei Xiong
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Guiyuan Li
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China
| | - Mei Yi
- Department of Dermatology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, China
| | - Xiayu Li
- Hunan Key Laboratory of Nonresolving Infammation and Cancer, The Third Xiangya Hospital, Central South University, Changsha 410013, Hunan, China.
| | - Hui Wang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; Department of Radiation Oncology, Hunan Cancer Hospital and The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan 410013, China.
| | - Bo Xiang
- Hunan Key Laboratory of Translational Radiation Oncology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha 410013, Hunan, China; The Key Laboratory of Carcinogenesis of the Chinese Ministry of Health, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410008, Hunan, China; The Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medical Sciences, Central South University, Changsha 410078, Hunan, China; FuRong Laboratory, Changsha 410078, Hunan, China.
| |
Collapse
|
23
|
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
|
24
|
Zhu J, Zhang J, Lou Y, Zheng Y, Zheng X, Cen W, Ye L, Zhang Q. Developing a machine learning-based prognosis and immunotherapeutic response signature in colorectal cancer: insights from ferroptosis, fatty acid dynamics, and the tumor microenvironment. Front Immunol 2024; 15:1416443. [PMID: 39076986 PMCID: PMC11284049 DOI: 10.3389/fimmu.2024.1416443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 06/28/2024] [Indexed: 07/31/2024] Open
Abstract
Instruction Colorectal cancer (CRC) poses a challenge to public health and is characterized by a high incidence rate. This study explored the relationship between ferroptosis and fatty acid metabolism in the tumor microenvironment (TME) of patients with CRC to identify how these interactions impact the prognosis and effectiveness of immunotherapy, focusing on patient outcomes and the potential for predicting treatment response. Methods Using datasets from multiple cohorts, including The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO), we conducted an in-depth multi-omics study to uncover the relationship between ferroptosis regulators and fatty acid metabolism in CRC. Through unsupervised clustering, we discovered unique patterns that link ferroptosis and fatty acid metabolism, and further investigated them in the context of immune cell infiltration and pathway analysis. We developed the FeFAMscore, a prognostic model created using a combination of machine learning algorithms, and assessed its predictive power for patient outcomes and responsiveness to treatment. The FeFAMscore signature expression level was confirmed using RT-PCR, and ACAA2 progression in cancer was further verified. Results This study revealed significant correlations between ferroptosis regulators and fatty acid metabolism-related genes with respect to tumor progression. Three distinct patient clusters with varied prognoses and immune cell infiltration were identified. The FeFAMscore demonstrated superior prognostic accuracy over existing models, with a C-index of 0.689 in the training cohort and values ranging from 0.648 to 0.720 in four independent validation cohorts. It also responses to immunotherapy and chemotherapy, indicating a sensitive response of special therapies (e.g., anti-PD-1, anti-CTLA4, osimertinib) in high FeFAMscore patients. Conclusion Ferroptosis regulators and fatty acid metabolism-related genes not only enhance immune activation, but also contribute to immune escape. Thus, the FeFAMscore, a novel prognostic tool, is promising for predicting both the prognosis and efficacy of immunotherapeutic strategies in patients with CRC.
Collapse
Affiliation(s)
- Junchang Zhu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jinyuan Zhang
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yunwei Lou
- Department of Gastroenterology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yijie Zheng
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Xuzhi Zheng
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Wei Cen
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Lechi Ye
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qiongying Zhang
- Department of Pathology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
25
|
Wang Y, Tsukamoto Y, Hori M, Iha H. Disulfidptosis: A Novel Prognostic Criterion and Potential Treatment Strategy for Diffuse Large B-Cell Lymphoma (DLBCL). Int J Mol Sci 2024; 25:7156. [PMID: 39000261 PMCID: PMC11241771 DOI: 10.3390/ijms25137156] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 06/24/2024] [Accepted: 06/24/2024] [Indexed: 07/16/2024] Open
Abstract
Diffuse Large B-cell Lymphoma (DLBCL), with its intrinsic genetic and epigenetic heterogeneity, exhibits significantly variable clinical outcomes among patients treated with the current standard regimen. Disulfidptosis, a novel form of regulatory cell death triggered by disulfide stress, is characterized by the collapse of cytoskeleton proteins and F-actin due to intracellular accumulation of disulfides. We investigated the expression variations of disulfidptosis-related genes (DRGs) in DLBCL using two publicly available gene expression datasets. The initial analysis of DRGs in DLBCL (GSE12453) revealed differences in gene expression patterns between various normal B cells and DLBCL. Subsequent analysis (GSE31312) identified DRGs strongly associated with prognostic outcomes, revealing eight characteristic DRGs (CAPZB, DSTN, GYS1, IQGAP1, MYH9, NDUFA11, NDUFS1, OXSM). Based on these DRGs, DLBCL patients were stratified into three groups, indicating that (1) DRGs can predict prognosis, and (2) DRGs can help identify novel therapeutic candidates. This study underscores the significant role of DRGs in various biological processes within DLBCL. Assessing the risk scores of individual DRGs allows for more precise stratification of prognosis and treatment strategies for DLBCL patients, thereby enhancing the effectiveness of clinical practice.
Collapse
Affiliation(s)
- Yu Wang
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan;
| | - Yoshiyuki Tsukamoto
- Department of Molecular Pathology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan;
| | - Mitsuo Hori
- Department of Hematology, Ibaraki Prefectural Central Hospital, Kasama 309-1703, Japan;
| | - Hidekatsu Iha
- Department of Microbiology, Faculty of Medicine, Oita University, Yufu 879-5593, Japan;
- Division of Pathophysiology, The Research Center for GLOBAL and LOCAL Infectious Diseases (RCGLID), Oita University, Yufu 879-5503, Japan
| |
Collapse
|
26
|
Liu J, Wu M, Yang Y, Wang Z, He S, Tian X, Wang H. γδ T cells and the PD-1/PD-L1 axis: a love-hate relationship in the tumor microenvironment. J Transl Med 2024; 22:553. [PMID: 38858763 PMCID: PMC11163710 DOI: 10.1186/s12967-024-05327-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: 03/04/2024] [Accepted: 05/20/2024] [Indexed: 06/12/2024] Open
Abstract
Gamma delta (γδ) T cells demonstrate strong cytotoxicity against diverse cancer cell types in an MHC-independent manner, rendering them promising contenders for cancer therapy. Although amplification and adoptive transfer of γδ T cells are being evaluated in the clinic, their therapeutic efficacy remains unsatisfactory, primarily due to the influence of the immunosuppressive tumor microenvironment (TME). Currently, the utilization of targeted therapeutic antibodies against inhibitory immune checkpoint (ICP) molecules is a viable approach to counteract the immunosuppressive consequences of the TME. Notably, PD-1/PD-L1 checkpoint inhibitors are considered primary treatment options for diverse malignancies, with the objective of preserving the response of αβ T cells. However, γδ T cells also infiltrate various human cancers and are important participants in cancer immunity, thereby influencing patient prognosis. Hence, it is imperative to comprehend the reciprocal impact of the PD-1/PD-L1 axis on γδ T cells. This understanding can serve as a therapeutic foundation for improving γδ T cells adoptive transfer therapy and may offer a novel avenue for future combined immunotherapeutic approaches.
Collapse
Affiliation(s)
- Jian Liu
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Min Wu
- Department of Gynecologic Oncology, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yifan Yang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zixuan Wang
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shan He
- Department of Pathogen Biology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xun Tian
- Department of Obstetrics and Gynecology, Academician Expert Workstation, The Central Hospital of Wuhan, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430014, Hubei, China.
| | - Hui Wang
- Department of Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Cancer Biology Research Center (Key Laboratory of the Ministry of Education), Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
- Department of Gynecologic Oncology, Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
- Zhejiang Provincial Key Laboratory of Precision Diagnosis and Therapy for Major Gynecological Diseases, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| |
Collapse
|
27
|
Huang F, Su Z, Huang Y, Huang Y, Zhou C, Feng S, Qin X, Xie X, Liu C, Yu C. Exploration of the combined role of immune checkpoints and immune cells in the diagnosis and treatment of ankylosing spondylitis: a preliminary study immune checkpoints in ankylosing spondylitis. Arthritis Res Ther 2024; 26:115. [PMID: 38835033 DOI: 10.1186/s13075-024-03341-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 05/12/2024] [Indexed: 06/06/2024] Open
Abstract
OBJECTIVE Immune checkpoints have emerged as promising therapeutic targets for autoimmune diseases. However, the specific roles of immune checkpoints in the pathophysiology of ankylosing spondylitis (AS) remain unclear. METHODS Hip ligament samples were obtained from two patient groups: those with AS and femoral head deformity, and those with femoral head necrosis but without AS, undergoing hip arthroplasty. Label-Free Quantification (LFQ) Protein Park Analysis was used to identify the protein composition of the ligaments. Peripheral blood samples of 104 AS patients from public database were used to validate the expression of key proteins. KEGG, GO, and GSVA were employed to explore potential pathways regulated by immune checkpoints in AS progression. xCell was used to calculate cell infiltration levels, LASSO regression was applied to select key cells, and the correlation between immune checkpoints and immune cells was analyzed. Drug sensitivity analysis was conducted to identify potential therapeutic drugs targeting immune checkpoints in AS. The expression of key genes was validated through immunohistochemistry (IHC). RESULTS HLA-DMB and HLA-DPA1 were downregulated in the ligaments of AS and this has been validated through peripheral blood datasets and IHC. Significant differences in expression were observed in CD8 + Tcm, CD8 + T cells, CD8 + Tem, osteoblasts, Th1 cells, and CD8 + naive T cells in AS. The infiltration levels of CD8 + Tcm and CD8 + naive T cells were significantly positively correlated with the expression levels of HLA-DMB and HLA-DPA1. Immune cell selection using LASSO regression showed good predictive ability for AS, with AUC values of 0.98, 0.81, and 0.75 for the three prediction models, respectively. Furthermore, this study found that HLA-DMB and HLA-DPA1 are involved in Th17 cell differentiation, and both Th17 cell differentiation and the NF-kappa B signaling pathway are activated in the AS group. Drug sensitivity analysis showed that AS patients are more sensitive to drugs such as doramapimod and GSK269962A. CONCLUSION Immune checkpoints and immune cells could serve as avenues for exploring diagnostic and therapeutic strategies for AS.
Collapse
Affiliation(s)
- Feihong Huang
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Zhiping Su
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
- Department of Radiation Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yibin Huang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Yuxiang Huang
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chengyu Zhou
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Sitan Feng
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xiong Qin
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Xi Xie
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China
| | - Chong Liu
- Spine and Osteopathy Ward, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| | - Chaojie Yu
- Department of Bone and Soft Tissue Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
- Guangxi Key Laboratory of Regenerative Medicine, Orthopaedic Department, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi Zhuang Autonomous Region, 530021, China.
| |
Collapse
|
28
|
Ye Z, Cheng P, Huang Q, Hu J, Huang L, Hu G. Immunocytes interact directly with cancer cells in the tumor microenvironment: one coin with two sides and future perspectives. Front Immunol 2024; 15:1388176. [PMID: 38840908 PMCID: PMC11150710 DOI: 10.3389/fimmu.2024.1388176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 05/07/2024] [Indexed: 06/07/2024] Open
Abstract
The tumor microenvironment is closely linked to the initiation, promotion, and progression of solid tumors. Among its constitutions, immunologic cells emerge as critical players, facilitating immune evasion and tumor progression. Apart from their indirect impact on anti-tumor immunity, immunocytes directly influence neoplastic cells, either bolstering or impeding tumor advancement. However, current therapeutic modalities aimed at alleviating immunosuppression from regulatory cells on effector immune cell populations may not consistently yield satisfactory results in various solid tumors, such as breast carcinoma, colorectal cancer, etc. Therefore, this review outlines and summarizes the direct, dualistic effects of immunocytes such as T cells, innate lymphoid cells, B cells, eosinophils, and tumor-associated macrophages on tumor cells within the tumor microenvironment. The review also delves into the underlying mechanisms involved and presents the outcomes of clinical trials based on these direct effects, aiming to propose innovative and efficacious therapeutic strategies for addressing solid tumors.
Collapse
Affiliation(s)
- Zhiyi Ye
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Pu Cheng
- Department of Gynecology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Huang
- Department of Oncology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Department of Oncology, Anhui Medical University, Hefei, Anhui, China
| | - Jingjing Hu
- School of Medicine, Shaoxing University, Zhejiang, China
| | - Liming Huang
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital; Shaoxing Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Guoming Hu
- Department of General Surgery (Breast and Thyroid Surgery), Shaoxing People’s Hospital, Shaoxing Hospital, Zhejiang University School of Medicine, Shaoxing, Zhejiang, China
- Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, Hangzhou, Zhejiang, China
| |
Collapse
|
29
|
Wei J, Guo F, Song Y, Feng T, Wang Y, Xu K, Song J, Kaysar E, Abdukayyum R, Lin F, Li K, Li B, Qian Z, Wang X, Wang H, Xu T. Analysis of the components of Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) and its regulation of γδ T-cell function. Cell Mol Biol Lett 2024; 29:70. [PMID: 38741147 PMCID: PMC11089708 DOI: 10.1186/s11658-024-00585-7] [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: 11/07/2023] [Accepted: 04/26/2024] [Indexed: 05/16/2024] Open
Abstract
BACKGROUND Mycobacterium tuberculosis heat-resistant antigen (Mtb-HAg) is a peptide antigen released from the mycobacterial cytoplasm into the supernatant of Mycobacterium tuberculosis (Mtb) attenuated H37Ra strain after autoclaving at 121 °C for 20 min. Mtb-HAg can specifically induce γδ T-cell proliferation in vitro. However, the exact composition of Mtb-HAg and the protein antigens that are responsible for its function are currently unknown. METHODS Mtb-HAg extracted from the Mtb H37Ra strain was subjected to LC‒MS mass spectrometry. Twelve of the identified protein fractions were recombinantly expressed in Escherichia coli by genetic engineering technology using pET-28a as a plasmid and purified by Ni-NTA agarose resin to stimulate peripheral blood mononuclear cells (PBMCs) from different healthy individuals. The proliferation of γδ T cells and major γδ T-cell subset types as well as the production of TNF-α and IFN-γ were determined by flow cytometry. Their proliferating γδ T cells were isolated and purified using MACS separation columns, and Mtb H37Ra-infected THP-1 was co-cultured with isolated and purified γδ T cells to quantify Mycobacterium viability by counting CFUs. RESULTS In this study, Mtb-HAg from the attenuated Mtb H37Ra strain was analysed by LC‒MS mass spectrometry, and a total of 564 proteins were identified. Analysis of the identified protein fractions revealed that the major protein components included heat shock proteins and Mtb-specific antigenic proteins. Recombinant expression of 10 of these proteins in by Escherichia coli genetic engineering technology was used to successfully stimulate PBMCs from different healthy individuals, but 2 of the proteins, EsxJ and EsxA, were not expressed. Flow cytometry results showed that, compared with the IL-2 control, HspX, GroEL1, and GroES specifically induced γδ T-cell expansion, with Vγ2δ2 T cells as the main subset, and the secretion of the antimicrobial cytokines TNF-α and IFN-γ. In contrast, HtpG, DnaK, GroEL2, HbhA, Mpt63, EsxB, and EsxN were unable to promote γδ T-cell proliferation and the secretion of TNF-α and IFN-γ. None of the above recombinant proteins were able to induce the secretion of TNF-α and IFN-γ by αβ T cells. In addition, TNF-α, IFN-γ-producing γδ T cells inhibit the growth of intracellular Mtb. CONCLUSION Activated γδ T cells induced by Mtb-HAg components HspX, GroES, GroEL1 to produce TNF-α, IFN-γ modulate macrophages to inhibit intracellular Mtb growth. These data lay the foundation for subsequent studies on the mechanism by which Mtb-HAg induces γδ T-cell proliferation in vitro, as well as the development of preventive and therapeutic vaccines and rapid diagnostic reagents.
Collapse
MESH Headings
- Humans
- Antigens, Bacterial/immunology
- Antigens, Bacterial/metabolism
- Antigens, Bacterial/genetics
- Mycobacterium tuberculosis/immunology
- Mycobacterium tuberculosis/genetics
- Cell Proliferation
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- Interferon-gamma/metabolism
- Interferon-gamma/immunology
- Receptors, Antigen, T-Cell, gamma-delta/metabolism
- Receptors, Antigen, T-Cell, gamma-delta/immunology
- Receptors, Antigen, T-Cell, gamma-delta/genetics
- Tumor Necrosis Factor-alpha/metabolism
- Leukocytes, Mononuclear/metabolism
- Leukocytes, Mononuclear/immunology
- Bacterial Proteins/metabolism
- Bacterial Proteins/genetics
- Bacterial Proteins/immunology
Collapse
Affiliation(s)
- Jing Wei
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Fangzheng Guo
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Yamin Song
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Tong Feng
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Ying Wang
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Kun Xu
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Jianhan Song
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Eldana Kaysar
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan, 848099, China
| | - Reyima Abdukayyum
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan, 848099, China
| | - Feiyang Lin
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Kangsheng Li
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Baiqing Li
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Zhongqing Qian
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China
| | - Xiaojing Wang
- Anhui Province Key Laboratory of Clinical and Preclinical Research in Respiratory Disease, Bengbu Medical University, Bengbu, 233000, China
| | - Hongtao Wang
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China.
- Xinjiang Key Laboratory of Hotan Characteristic Chinese Traditional Medicine Research, College of Xinjiang Uyghur Medicine, Hotan, 848099, China.
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China.
| | - Tao Xu
- Laboratory Medicine Experimental Center, Laboratory Medicine College, Bengbu Medical University, Bengbu, 233000, China.
- Anhui Province Key Laboratory of Immunology in Chronic Diseases, Bengbu Medical University, Bengbu, 233000, China.
| |
Collapse
|
30
|
Zhang Q, Liu X, He J. Applications and prospects of microneedles in tumor drug delivery. J Mater Chem B 2024; 12:3336-3355. [PMID: 38501172 DOI: 10.1039/d3tb02646a] [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: 03/20/2024]
Abstract
As drug delivery devices, microneedles are used widely in the local administration of various drugs. Such drug-loaded microneedles are minimally invasive, almost painless, and have high drug delivery efficiency. In recent decades, with advancements in microneedle technology, an increasing number of adaptive, engineered, and intelligent microneedles have been designed to meet increasing clinical needs. This article summarizes the types, preparation materials, and preparation methods of microneedles, as well as the latest research progress in the application of microneedles in tumor drug delivery. This article also discusses the current challenges and improvement strategies in the use of microneedles for tumor drug delivery.
Collapse
Affiliation(s)
- Qiang Zhang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
| | - Jian He
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi, 530021, China.
- School of Pharmacy, Guangxi Medical University, Nanning 530021, China
| |
Collapse
|
31
|
Hao Q, Li R, Li H, Rui S, You L, Zhang L, Zhao Y, Li P, Li Y, Kong X, Chen H, Zou X, Liu F, Wang X, Zhou J, Zhang W, Huang L, Shu Y, Liu J, Sun R, Li C, Zhu J, Jiang Y, Wei T, Qian K, Bai B, Hu Y, Peng Y, Dai L, Caulin C, Xu H, Li Z, Park J, Luo H, Ying B. Dynamics of The Γδtcr Repertoires During The Dedifferentiation Process and Pilot Implications for Immunotherapy of Thyroid Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306364. [PMID: 38286670 PMCID: PMC10987121 DOI: 10.1002/advs.202306364] [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: 09/04/2023] [Revised: 12/29/2023] [Indexed: 01/31/2024]
Abstract
γδ T cells are evolutionarily conserved T lymphocytes that manifest unique antitumor efficacy independent of tumor mutation burden (TMB) and conventional human leukocyte antigen (HLA) recognition. However, the dynamic changes in their T cell receptor (TCR) repertoire during cancer progression and treatment courses remain unclear. Here, a comprehensive characterization of γδTCR repertoires are performed in thyroid cancers with divergent differentiation states through cross-sectional studies. The findings revealed a significant correlation between the differentiation states and TCR repertoire diversity. Notably, highly expanded clones are prominently enriched in γδ T cell compartment of dedifferentiated patients. Moreover, by longitudinal investigations of the γδ T cell response to various antitumor therapies, it is found that the emergence and expansion of the Vδ2neg subset may be potentially associated with favorable clinical outcomes after post-radiotherapeutic immunotherapy. These findings are further validated at single-cell resolution in both advanced thyroid cancer patients and a murine model, underlining the importance of further investigations into the role of γδTCR in cancer immunity and therapeutic strategies.
Collapse
Affiliation(s)
- Qing Hao
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduSichuan610041China
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Ruicen Li
- Health Promotion CenterWest China Hospital, Sichuan UniversityChengduSichuan610041China
| | - Hancong Li
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Shu Rui
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Liting You
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduSichuan610041China
| | - Lingyun Zhang
- School of Biomedical SciencesThe Chinese University of Hong KongHong Kong SAR999077China
| | - Yue Zhao
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Peiheng Li
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Yuanmin Li
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Key Laboratory of Transplant Engineering and Immunology, Frontiers Science Center for Disease Related Molecular Network, West China HospitalSichuan UniversityChengdu610041China
| | - Xinagyu Kong
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Haining Chen
- Colorectal Cancer Center, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Xiuhe Zou
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Feng Liu
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Xiaofei Wang
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Juan Zhou
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduSichuan610041China
| | - Weihan Zhang
- Gastric Cancer Center, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Libing Huang
- Division of Gastrointestinal Surgery, State Key Laboratory of Biotherapy, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Yang Shu
- Gastric Cancer Center, West China HospitalSichuan UniversityChengduSichuan610041China
| | - JiaYe Liu
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Ronghao Sun
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Institute, Sichuan Cancer Prevention and Treatment CenterCancer Hospital of University of Electronic Science and Technology School of MedicineChengdu610041China
| | - Chao Li
- Department of Head and Neck Surgery, Sichuan Cancer Hospital, Sichuan Cancer Institute, Sichuan Cancer Prevention and Treatment CenterCancer Hospital of University of Electronic Science and Technology School of MedicineChengdu610041China
| | - Jingqiang Zhu
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Yong Jiang
- Division of Pathology, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Tao Wei
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Kun Qian
- State Key Laboratory of Systems Medicine for Cancer, School of Biomedical Engineering, Institute of Medical Robotics and Med‐X Research InstituteShanghai Jiao Tong UniversityShanghai200230China
| | - Bing Bai
- State Key Laboratory of Primate Biomedical Research, Institute of Primate Translational Medicine, Kunming University of Science and TechnologyYunnan Key Laboratory of Primate Biomedical ResearchKunmingYunnan650500China
| | - Yiguo Hu
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
| | - Yong Peng
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Lunzhi Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Carlos Caulin
- Department of Otolaryngology – Head & Neck Surgery and University of Arizona Cancer CenterUniversity of ArizonaTucsonAZ85721USA
| | - Heng Xu
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduSichuan610041China
- State Key Laboratory of Biotherapy and Cancer Center, West China HospitalSichuan University and Collaborative Innovation CenterChengduSichuan610041China
| | - Zhihui Li
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
| | - Jihwan Park
- School of Life SciencesGwangju Institute of Science and Technology (GIST)Gwangju61005Republic of Korea
| | - Han Luo
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduSichuan610041China
- Division of Thyroid Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Laboratory of Thyroid and Parathyroid DiseaseFrontiers Science Center for Disease‐Related Molecular NetworkChengdu610041China
- Department of General Surgery, West China HospitalSichuan UniversityChengduSichuan610041China
- Sichuan Clinical Research Center for laboratory medicineChengduSichuan610041China
| | - Binwu Ying
- Department of Laboratory MedicineWest China Hospital, Sichuan UniversityChengduSichuan610041China
- Sichuan Clinical Research Center for laboratory medicineChengduSichuan610041China
| |
Collapse
|
32
|
Nörenberg J, Vida P, Bösmeier I, Forró B, Nörenberg A, Buda Á, Simon D, Erdő-Bonyár S, Jáksó P, Kovács K, Mikó É, Berki T, Mezősi E, Barakonyi A. Decidual γδT cells of early human pregnancy produce angiogenic and immunomodulatory proteins while also possessing cytotoxic potential. Front Immunol 2024; 15:1382424. [PMID: 38601161 PMCID: PMC11004470 DOI: 10.3389/fimmu.2024.1382424] [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: 02/05/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024] Open
Abstract
During pregnancy, the maternal immune system must allow and support the growth of the developing placenta while maintaining the integrity of the mother's body. The trophoblast's unique HLA signature is a key factor in this physiological process. This study focuses on decidual γδT cell populations and examines their expression of receptors that bind to non-classical HLA molecules, HLA-E and HLA-G. We demonstrate that decidual γδT cell subsets, including Vδ1, Vδ2, and double-negative (DN) Vδ1-/Vδ2- cells express HLA-specific regulatory receptors, such as NKG2C, NKG2A, ILT2, and KIR2DL4, each with varying dominance. Furthermore, decidual γδT cells produce cytokines (G-CSF, FGF2) and cytotoxic mediators (Granulysin, IFN-γ), suggesting functions in placental growth and pathogen defense. However, these processes seem to be controlled by factors other than trophoblast-derived non-classical HLA molecules. These findings indicate that decidual γδT cells have the potential to actively contribute to the maintenance of healthy human pregnancy.
Collapse
Affiliation(s)
- Jasper Nörenberg
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Péter Vida
- Department of Obstetrics and Gynaecology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Isabell Bösmeier
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Barbara Forró
- Department of Pathology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Anna Nörenberg
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Ágnes Buda
- Department of Obstetrics and Gynaecology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Diana Simon
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Immunology and Biotechnology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Szabina Erdő-Bonyár
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Immunology and Biotechnology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Pál Jáksó
- Department of Pathology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Kálmán Kovács
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Obstetrics and Gynaecology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Éva Mikó
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| | - Tímea Berki
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Department of Immunology and Biotechnology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Emese Mezősi
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- First Department of Internal Medicine, University of Pécs Medical School, Clinical Center, Pécs, Hungary
| | - Alíz Barakonyi
- Department of Medical Microbiology and Immunology, University of Pécs Medical School, Clinical Center, Pécs, Hungary
- National Laboratory on Human Reproduction, University of Pécs, Pécs, Hungary
- Janos Szentagothai Research Centre, University of Pécs, Pécs, Hungary
| |
Collapse
|
33
|
Peng S, Lin A, Jiang A, Zhang C, Zhang J, Cheng Q, Luo P, Bai Y. CTLs heterogeneity and plasticity: implications for cancer immunotherapy. Mol Cancer 2024; 23:58. [PMID: 38515134 PMCID: PMC10956324 DOI: 10.1186/s12943-024-01972-6] [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: 12/25/2023] [Accepted: 02/26/2024] [Indexed: 03/23/2024] Open
Abstract
Cytotoxic T lymphocytes (CTLs) play critical antitumor roles, encompassing diverse subsets including CD4+, NK, and γδ T cells beyond conventional CD8+ CTLs. However, definitive CTLs biomarkers remain elusive, as cytotoxicity-molecule expression does not necessarily confer cytotoxic capacity. CTLs differentiation involves transcriptional regulation by factors such as T-bet and Blimp-1, although epigenetic regulation of CTLs is less clear. CTLs promote tumor killing through cytotoxic granules and death receptor pathways, but may also stimulate tumorigenesis in some contexts. Given that CTLs cytotoxicity varies across tumors, enhancing this function is critical. This review summarizes current knowledge on CTLs subsets, biomarkers, differentiation mechanisms, cancer-related functions, and strategies for improving cytotoxicity. Key outstanding questions include refining the CTLs definition, characterizing subtype diversity, elucidating differentiation and senescence pathways, delineating CTL-microbe relationships, and enabling multi-omics profiling. A more comprehensive understanding of CTLs biology will facilitate optimization of their immunotherapy applications. Overall, this review synthesizes the heterogeneity, regulation, functional roles, and enhancement strategies of CTLs in antitumor immunity, highlighting gaps in our knowledge of subtype diversity, definitive biomarkers, epigenetic control, microbial interactions, and multi-omics characterization. Addressing these questions will refine our understanding of CTLs immunology to better leverage cytotoxic functions against cancer.
Collapse
Affiliation(s)
- Shengkun Peng
- Department of Radiology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Anqi Lin
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Aimin Jiang
- Department of Urology, Changhai hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and ImmunologySchool of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Jian Zhang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya HospitalCentral South University, Hunan, China.
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510282, Guangdong, China.
| | - Yifeng Bai
- Department of Oncology, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
34
|
Li Y, Zhang C, Jiang A, Lin A, Liu Z, Cheng X, Wang W, Cheng Q, Zhang J, Wei T, Luo P. Potential anti-tumor effects of regulatory T cells in the tumor microenvironment: a review. J Transl Med 2024; 22:293. [PMID: 38509593 PMCID: PMC10953261 DOI: 10.1186/s12967-024-05104-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Accepted: 03/15/2024] [Indexed: 03/22/2024] Open
Abstract
Regulatory T cells (Tregs) expressing the transcription factor FoxP3 are essential for maintaining immunological balance and are a significant component of the immunosuppressive tumor microenvironment (TME). Single-cell RNA sequencing (ScRNA-seq) technology has shown that Tregs exhibit significant plasticity and functional diversity in various tumors within the TME. This results in Tregs playing a dual role in the TME, which is not always centered around supporting tumor progression as typically believed. Abundant data confirms the anti-tumor activities of Tregs and their correlation with enhanced patient prognosis in specific types of malignancies. In this review, we summarize the potential anti-tumor actions of Tregs, including suppressing tumor-promoting inflammatory responses and boosting anti-tumor immunity. In addition, this study outlines the spatial and temporal variations in Tregs function to emphasize that their predictive significance in malignancies may change. It is essential to comprehend the functional diversity and potential anti-tumor effects of Tregs to improve tumor therapy strategies.
Collapse
Affiliation(s)
- Yu Li
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Cangang Zhang
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, 710061, Shaanxi, China
| | - Aimin Jiang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Anqi Lin
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Zaoqu Liu
- Key Laboratory of Proteomics, Beijing Proteome Research Center, National Center for Protein Sciences (Beijing), Beijing, China
- Key Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Department of Pathophysiology, Peking Union Medical College, Beijing, 100730, China
| | - Xiangshu Cheng
- College of Bioinformatics Science and Technology, Harbin Medical University, 157 Baojian Road. Nangang District, Harbin, Heilongiiang, China
| | - Wanting Wang
- Institute of Molecular and Translational Medicine, and Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, 710061, Shaanxi, China
| | - Quan Cheng
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Jian Zhang
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Ting Wei
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| | - Peng Luo
- The Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, China.
| |
Collapse
|
35
|
Fan B, Fan B, Sun N, Zou H, Gu X. A radiomics model to predict γδ T-cell abundance and overall survival in head and neck squamous cell carcinoma. FASEB J 2024; 38:e23529. [PMID: 38441524 DOI: 10.1096/fj.202301353rr] [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: 07/05/2023] [Revised: 02/01/2024] [Accepted: 02/19/2024] [Indexed: 03/07/2024]
Abstract
γδ T cells are becoming increasingly popular because of their attractive potential for antitumor immunotherapy. However, the role and assessment of γδ T cells in head and neck squamous cell carcinoma (HNSCC) are not well understood. We aimed to explore the prognostic value of γδ T cell and predict its abundance using a radiomics model. Computer tomography images with corresponding gene expression data and clinicopathological data were obtained from online databases. After outlining the volumes of interest manually, the radiomic features were screened using maximum melevance minimum redundancy and recursive feature elimination algorithms. A radiomics model was developed to predict γδ T-cell abundance using gradient boosting machine. Kaplan-Meier survival curves and univariate and multivariate Cox regression analyses were used for the survival analysis. In this study, we confirmed that γδ T-cell abundance was an independent predictor of favorable overall survival (OS) in patients with HNSCC. Moreover, a radiomics model was built to predict the γδ T-cell abundance level (the areas under the operating characteristic curves of 0.847 and 0.798 in the training and validation sets, respectively). The calibration and decision curves analysis demonstrated the fitness of the model. The high radiomic score was an independent protective factor for OS. Our results indicated that γδ T-cell abundance was a promising prognostic predictor in HNSCC, and the radiomics model could discriminate its abundance levels and predict OS. The noninvasive radiomics model provided a potentially powerful prediction tool to aid clinical judgment and antitumor immunotherapy.
Collapse
Affiliation(s)
- Binna Fan
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of Nursing, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Binting Fan
- Department of Nursing, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
- Department of General Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Na Sun
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Huawei Zou
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiao Gu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, China
| |
Collapse
|
36
|
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
|
37
|
Wang CQ, Lim PY, Tan AHM. Gamma/delta T cells as cellular vehicles for anti-tumor immunity. Front Immunol 2024; 14:1282758. [PMID: 38274800 PMCID: PMC10808317 DOI: 10.3389/fimmu.2023.1282758] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/26/2023] [Indexed: 01/27/2024] Open
Abstract
Adoptive cellular immunotherapy as a new paradigm to treat cancers is exemplified by the FDA approval of six chimeric antigen receptor-T cell therapies targeting hematological malignancies in recent years. Conventional αβ T cells applied in these therapies have proven efficacy but are confined almost exclusively to autologous use. When infused into patients with mismatched human leukocyte antigen, αβ T cells recognize tissues of such patients as foreign and elicit devastating graft-versus-host disease. Therefore, one way to overcome this challenge is to use naturally allogeneic immune cell types, such as γδ T cells. γδ T cells occupy the interface between innate and adaptive immunity and possess the capacity to detect a wide variety of ligands on transformed host cells. In this article, we review the fundamental biology of γδ T cells, including their subtypes, expression of ligands, contrasting roles in and association with cancer prognosis or survival, as well as discuss the gaps in knowledge pertaining to this cell type which we currently endeavor to elucidate. In addition, we propose how to harness the unique properties of γδ T cells for cellular immunotherapy based on lessons gleaned from past clinical trials and provide an update on ongoing trials involving these cells. Lastly, we elaborate strategies that have been tested or can be explored to improve the anti-tumor activity and durability of γδ T cells in vivo.
Collapse
Affiliation(s)
- Chelsia Qiuxia Wang
- Immune Cell Manufacturing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Pei Yu Lim
- Immune Cell Manufacturing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Andy Hee-Meng Tan
- Immune Cell Manufacturing, Bioprocessing Technology Institute (BTI), Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Food, Chemical and Biotechnology Cluster, Singapore Institute of Technology (SIT), Singapore, Singapore
| |
Collapse
|
38
|
Sun Y, Li Y, Jiang C, Liu C, Song Y. SLC7A2-Mediated Lysine Catabolism Inhibits Immunosuppression in Triple Negative Breast Cancer. Crit Rev Eukaryot Gene Expr 2024; 34:31-43. [PMID: 38842202 DOI: 10.1615/critreveukaryotgeneexpr.2024052503] [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: 06/07/2024]
Abstract
Breast cancer is one of the most common malignant tumors worldwide. SLC7A2 is abnormally expressed in multiple cancers. However, its potential in triple negative breast cancer (TNBC) is still unclear. The purpose of this study was to investigate the roles of SLC7A2 and its underlying molecular mechanisms in TNBC. mRNA expression was detected by RT-qPCR. Protein expression was detected by western blot. Co-localization of ACOX1 and TCF1 was determined using FISH assay. Histone crotonylation was performed using in vitro histone crotonylation assay. Functional analysis was performed using CCK-8 and flow cytometry assays. Xenograft assay was conducted to further verify the role of SLC7A2 in TNBC. CD8A expression was detected using immunohistochemistry. We found that SLC7A2 is downregulated in TNBC tumors. Low levels are associated with advanced stages and lymph node metastasis. SLC7A2 expression is positively correlated with CD8A. SLC7A2-mediated lysine catabolism drives the activation of CD8+ T cells. Moreover, SLC7A2 promotes histone crotonylation via upregulating ACOX1. It also promotes interaction between ACOX1 and TCF1, thus promoting antitumor T cell immunity. Additionally, overexpression of SLC7A2 activates CD8+ T cells and enhances the chemosensitivity of anti-PD-1 therapies in vivo. In conclusion, SLC7A2 may function as an antitumor gene in TNBC by activating antitumor immunity, suggesting SLC7A2/ACOX1/TCF1 signaling as a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yuanyuan Sun
- Tianjin Medical University Cancer Institute & Hospital
| | - Yaqing Li
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Chengying Jiang
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Chenying Liu
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| | - Yuanming Song
- Department of Breast Pathology and Lab, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Tianjin's Clinical Research Center for Cancer; Key Laboratory of Breast Cancer Prevention and Therapy; Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China
| |
Collapse
|
39
|
Li Y, Mo XP, Yao H, Xiong QX. Research Progress of γδT Cells in Tumor Immunotherapy. Cancer Control 2024; 31:10732748241284863. [PMID: 39348473 PMCID: PMC11459529 DOI: 10.1177/10732748241284863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Revised: 08/20/2024] [Accepted: 09/03/2024] [Indexed: 10/02/2024] Open
Abstract
Background: γδT cells are special innate lymphoid cells, which are not restricted by major histocompatibility complex (MHC). γδT cells mainly exist in human epidermis and mucosal epithelium. They can secrete a variety of cytokines and chemokines involved in immune regulation, and produce effective cytotoxic responses to cancer cells. Purpose: To investigate the role of γδT cells in tumor immunotherapy, to understand its anti-tumor mechanism, and to explore the synergistic effect with other treatment modalities. This therapy is expected to become an important means of cancer treatment. Research Design: In this review presents a comprehensive analysis of the existing literature, focusing on the efficacy of γδT cells in a variety of tumor types. Results: The mechanism of γδT cells recognizing tumor antigens and killing tumor was clarified. The tumor immunotherapy based on γδT cells and its application in clinical practice were summarized. Conclusions: γδT cells have shown promising potential in tumor immunotherapy, but the therapeutic effect varies according to the type of tumor, and some patients have poor response. There are still some challenges in the treatment of this disease, such as non-standard expansion regimens and different responses of patients, indicating that the existing treatment methods are not complete. Future research should focus on perfecting γδT cell expansion protocols, gaining a deeper understanding of its anti-tumor mechanisms, and exploring synergies with other treatment modalities. This multifaceted study will promote the development of γδT cells in the field of cancer immunotherapy.
Collapse
Affiliation(s)
- Yan Li
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Xin-pei Mo
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| | - Hong Yao
- Cancer Biotherapy Center, The Third Affiliated Hospital of Kunming Medical University (Tumor Hospital of Yunnan Province), Kunming, China
| | - Qiu-xia Xiong
- Department of Clinical Laboratory, The First Affiliated Hospital of Kunming Medical University, Kunming, China
- Yunnan Key Laboratory of Laboratory Medicine, Kunming, China
| |
Collapse
|
40
|
Wang X, Jin Y, Xu L, Tao S, Wu Y, Ao C. Integrating Single-Cell RNA-Seq and Bulk RNA-Seq to Construct a Novel γδT Cell-Related Prognostic Signature for Human Papillomavirus-Infected Cervical Cancer. Cancer Control 2024; 31:10732748241274228. [PMID: 39206965 PMCID: PMC11363054 DOI: 10.1177/10732748241274228] [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: 04/26/2024] [Revised: 06/11/2024] [Accepted: 07/17/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Gamma delta (γδ) T cells play dual roles in human tumors, with both antitumor and tumor-promoting functions. However, the role of γδT cells in HPV-infected cervical cancer is still undetermined. Therefore, we aimed to identify γδT cell-related prognostic signatures in the cervical tumor microenvironment. METHODS Single-cell RNA-sequencing (scRNA-seq) data, bulk RNA-seq data, and corresponding clinical information of cervical cancer patients were obtained from the TCGA and GEO databases. The Seurat R package was used for single-cell analysis, and machine learning algorithms were used to screen and construct a γδT cell-related prognostic signature. Real-time quantitative PCR (RT-qPCR) was performed to detect the expression of prognostic signature genes. RESULTS Single-cell analysis indicated distinct populations of γδT cells between HPV-positive (HPV+) and HPV-negative (HPV-) cervical cancers. A trajectory analysis indicated γδT cells clustered into differential clusters with the pseudotime. High-dimensional Weighted Gene Co-expression Network Analysis (hdWGCNA) identified the key γδT cell-related gene modules. Bulk RNA-seq analysis also demonstrated the heterogeneity of immune cells, and the γδT-score was positively associated with inflammatory response and negatively associated with MYC stemness. Eight γδT cell-related hub genes (GTRGs), including ITGAE, IKZF3, LSP1, NEDD9, CLEC2D, RBPJ, TRBC2, and OXNAD1, were selected and validated as a prognostic signature for cervical cancer. CONCLUSION We identified γδT cell-related prognostic signatures that can be considered independent factors for survival prediction in cervical cancer.
Collapse
Affiliation(s)
- Xiaochuan Wang
- Department of Dermatology, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, China
| | - Yichao Jin
- Department of Dermatology, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, China
| | - Liangheng Xu
- Department of Dermatology, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, China
| | - Sizhen Tao
- Department of Dermatology, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, China
| | - Yifei Wu
- Department of Dermatology, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, China
| | - Chunping Ao
- Department of Dermatology, The First People’s Hospital of Yunnan Province, the Affiliated Hospital of Kunming University of Science and Technology, Yunnan Provincial Key Laboratory of Clinical Virology, Kunming, China
| |
Collapse
|
41
|
Wang Y, Jin S, Zhuang Q, Liu N, Chen R, Adam SA, Jin J, Sun J. Chimeric antigen receptor natural killer cells: a promising antitumor immunotherapy. MedComm (Beijing) 2023; 4:e422. [PMID: 38045827 PMCID: PMC10691297 DOI: 10.1002/mco2.422] [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: 05/09/2023] [Revised: 10/05/2023] [Accepted: 10/12/2023] [Indexed: 12/05/2023] Open
Abstract
Chimeric antigen receptor (CAR) T cells have been successfully used in adoptive cell therapy for malignancies. However, some obstacles, including side effects such as graft-versus-host disease and cytokine release syndrome, therapy resistance, limited sources, as well as high cost, limited the application of CAR T cells. Recently, CAR natural killer (NK) cells have been pursued as the effector cells for adoptive immunotherapy for their attractive merits of strong intrinsic antitumor activity and relatively mild side effects. Additionally, CAR NK cells can be available from various sources and do not require strict human leukocyte antigen matching, which suggests them as promising "off-the-shelf" products for clinical application. Although the use of CAR NK cells is restrained by the limited proliferation and impaired efficiency within the immunosuppressive tumor microenvironment, further investigation in optimizing CAR structure and combination therapies will overcome these challenges. This review will summarize the advancement of CAR NK cells, CAR NK cell manufacture, the clinical outcomes of CAR NK therapy, the challenges in the field, and prospective solutions. Besides, we will discuss the emerging application of other immune cells for CAR engineering. Collectively, this comprehensive review will provide a valuable and informative summary of current progress and evaluate challenges and future opportunities of CAR NK cells in tumor treatment.
Collapse
Affiliation(s)
- Yan Wang
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Shengjie Jin
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Qiqi Zhuang
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Na Liu
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Department of OncologyAffiliated Hospital of Weifang Medical UniversitySchool of Clinical MedicineWeifang Medical UniversityWeifangShandongChina
| | - Ruyi Chen
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Sofia Abdulkadir Adam
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
| | - Jie Jin
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Zhejiang University Cancer CenterHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouZhejiangChina
| | - Jie Sun
- Department of HematologyThe First Affiliated HospitalZhejiang University School of MedicineHangzhouZhejiangChina
- Key Laboratory of Hematologic MalignanciesDiagnosis, and TreatmentHangzhouZhejiangChina
- Zhejiang Provincial Clinical Research Center for Hematological DisordersHangzhouZhejiangChina
| |
Collapse
|
42
|
Patra S, Ghosal S, Shand H, Mondal R, Rath A, Kumar Jana S, Ghorai S. Function of gamma delta (γδ) T cell in cancer with special emphasis on cervical cancer. Hum Immunol 2023; 84:110724. [PMID: 37932183 DOI: 10.1016/j.humimm.2023.110724] [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: 07/03/2023] [Revised: 10/04/2023] [Accepted: 10/24/2023] [Indexed: 11/08/2023]
Abstract
Cervical cancer is the second-most prevalent gynecologic cancer in India. It is typically detected in women between the ages of 35 and 44. Cervical cancer is mainly associated with the human papillomavirus (HPV). The report shows that 70 % of cervical cancer is caused by HPV 16 and 18. There are few therapeutic options and vaccines available for cervical cancer treatment and γδ T cell therapy is one of them. This therapy can kill various types of cancers, including cervical cancer. The major γδ T cell subset is the Vγ9Vδ2 T cell, mainly distributed in peripheral blood which recognize non-MHC peptide antigens and can eliminate MHC-downregulated cancer. Moreover, γδ T cells can express different types of receptors that bind to the molecules of stressed cells, often produced on cancerous cells but absent from healthy tissue. γδ T cells possess both direct and indirect cytotoxic capabilities against malignancies and show potential antitumoral responses. However, γδ T cells also encourage the progression of cancer. Cancer immunotherapy using γδ T cells will be a potential cancer treatment, as well as cervical cancer. This review focused on the γδ T cell and its function in cancer, with special emphasis on cervical cancer. It also focused on the ligand recognition site of γδ T cells, galectin-mediated therapy and pamidronate-treated therapy for cervical cancer. Instead of the great potential of γδ T cell for the eradication of cervical cancer, no comprehensive in-depth review is available to date, so there is a need to jot down the various roles and modes of action and different applications of γδ T cells for cancer research, which we believe will be a handy tool for the researchers and the readers.
Collapse
Affiliation(s)
- Soumendu Patra
- Department of Microbiology, Raiganj University, North Dinajpur, West Bengal 733134, India
| | - Sayan Ghosal
- Department of Microbiology, Raiganj University, North Dinajpur, West Bengal 733134, India
| | - Harshita Shand
- Department of Microbiology, Raiganj University, North Dinajpur, West Bengal 733134, India
| | - Rittick Mondal
- Department of Sericulture, Raiganj University, North Dinajpur, West Bengal 733134, India
| | - Ankita Rath
- Department of Biotechnology, SRM Institute of Science & Technology, Kattankulathur, Tamil Nadu 603203, India
| | - Saikat Kumar Jana
- Department of Biotechnology, National Institute of Technology, Yupia, Arunachal Pradesh 791112, India
| | - Suvankar Ghorai
- Department of Microbiology, Raiganj University, North Dinajpur, West Bengal 733134, India.
| |
Collapse
|
43
|
Wang K, Zhou M, Si H, Ma J. Gut microbiota-mediated IL-22 alleviates metabolic inflammation. Life Sci 2023; 334:122229. [PMID: 37922980 DOI: 10.1016/j.lfs.2023.122229] [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: 08/28/2023] [Revised: 10/25/2023] [Accepted: 10/29/2023] [Indexed: 11/06/2023]
Abstract
Low-grade chronic inflammation, also known as metabolic inflammation, promotes the development of metabolic diseases. Increasing evidence suggests that changes in gut microbes and metabolites disrupt the integrity of the gut barrier and exert significant effects on the metabolism of various tissues, including the liver and adipose tissue, thereby contributing to metabolic inflammation. We observed that IL-22 is a key signaling molecule that serves as a bridge between intestinal microbes and the host, effectively alleviating metabolic inflammation by modulating the host immunomodulatory network. Here, we focused on elucidating the underlying mechanisms by which the gut microbiota and their metabolites reduce inflammation via IL-22, highlighting the favorable impact of IL-22 on metabolic inflammation. Furthermore, we discuss the potential of IL-22 as a therapeutic target for the management of metabolic inflammation and related diseases.
Collapse
Affiliation(s)
- Kaijun Wang
- College of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, China; Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Miao Zhou
- Animal Nutritional Genome and Germplasm Innovation Research Center, College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, Hunan, China
| | - Hongbin Si
- College of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, China
| | - Jie Ma
- College of Animal Science and Technology, Guangxi University, Nanning 530004, Guangxi, China.
| |
Collapse
|
44
|
Fotie J, Matherne CM, Mather JB, Wroblewski JE, Johnson K, Boudreaux LG, Perez AA. The Fundamental Role of Oxime and Oxime Ether Moieties in Improving the Physicochemical and Anticancer Properties of Structurally Diverse Scaffolds. Int J Mol Sci 2023; 24:16854. [PMID: 38069175 PMCID: PMC10705934 DOI: 10.3390/ijms242316854] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 11/22/2023] [Accepted: 11/24/2023] [Indexed: 12/18/2023] Open
Abstract
The present review explores the critical role of oxime and oxime ether moieties in enhancing the physicochemical and anticancer properties of structurally diverse molecular frameworks. Specific examples are carefully selected to illustrate the distinct contributions of these functional groups to general strategies for molecular design, modulation of biological activities, computational modeling, and structure-activity relationship studies. An extensive literature search was conducted across three databases, including PubMed, Google Scholar, and Scifinder, enabling us to create one of the most comprehensive overviews of how oximes and oxime ethers impact antitumor activities within a wide range of structural frameworks. This search focused on various combinations of keywords or their synonyms, related to the anticancer activity of oximes and oxime ethers, structure-activity relationships, mechanism of action, as well as molecular dynamics and docking studies. Each article was evaluated based on its scientific merit and the depth of the study, resulting in 268 cited references and more than 336 illustrative chemical structures carefully selected to support this analysis. As many previous reviews focus on one subclass of this extensive family of compounds, this report represents one of the rare and fully comprehensive assessments of the anticancer potential of this group of molecules across diverse molecular scaffolds.
Collapse
Affiliation(s)
- Jean Fotie
- Department of Chemistry and Physics, Southeastern Louisiana University, SLU 10878, Hammond, LA 70402-0878, USA; (C.M.M.); (J.B.M.); (J.E.W.); (K.J.); (L.G.B.); (A.A.P.)
| | | | | | | | | | | | | |
Collapse
|
45
|
Karsten H, Matrisch L, Cichutek S, Fiedler W, Alsdorf W, Block A. Broadening the horizon: potential applications of CAR-T cells beyond current indications. Front Immunol 2023; 14:1285406. [PMID: 38090582 PMCID: PMC10711079 DOI: 10.3389/fimmu.2023.1285406] [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: 08/29/2023] [Accepted: 11/10/2023] [Indexed: 12/18/2023] Open
Abstract
Engineering immune cells to treat hematological malignancies has been a major focus of research since the first resounding successes of CAR-T-cell therapies in B-ALL. Several diseases can now be treated in highly therapy-refractory or relapsed conditions. Currently, a number of CD19- or BCMA-specific CAR-T-cell therapies are approved for acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL), mantle cell lymphoma (MCL), multiple myeloma (MM), and follicular lymphoma (FL). The implementation of these therapies has significantly improved patient outcome and survival even in cases with previously very poor prognosis. In this comprehensive review, we present the current state of research, recent innovations, and the applications of CAR-T-cell therapy in a selected group of hematologic malignancies. We focus on B- and T-cell malignancies, including the entities of cutaneous and peripheral T-cell lymphoma (T-ALL, PTCL, CTCL), acute myeloid leukemia (AML), chronic myeloid leukemia (CML), chronic lymphocytic leukemia (CLL), classical Hodgkin-Lymphoma (HL), Burkitt-Lymphoma (BL), hairy cell leukemia (HCL), and Waldenström's macroglobulinemia (WM). While these diseases are highly heterogenous, we highlight several similarly used approaches (combination with established therapeutics, target depletion on healthy cells), targets used in multiple diseases (CD30, CD38, TRBC1/2), and unique features that require individualized approaches. Furthermore, we focus on current limitations of CAR-T-cell therapy in individual diseases and entities such as immunocompromising tumor microenvironment (TME), risk of on-target-off-tumor effects, and differences in the occurrence of adverse events. Finally, we present an outlook into novel innovations in CAR-T-cell engineering like the use of artificial intelligence and the future role of CAR-T cells in therapy regimens in everyday clinical practice.
Collapse
Affiliation(s)
- Hendrik Karsten
- Faculty of Medicine, University of Hamburg, Hamburg, Germany
| | - Ludwig Matrisch
- Department of Rheumatology and Clinical Immunology, University Medical Center Schleswig-Holstein, Lübeck, Germany
- Faculty of Medicine, University of Lübeck, Lübeck, Germany
| | - Sophia Cichutek
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Walter Fiedler
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Winfried Alsdorf
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| | - Andreas Block
- Department of Oncology, Hematology and Bone Marrow Transplantation with Division of Pneumology, University Medical Center Eppendorf, Hamburg, Germany
| |
Collapse
|
46
|
Yan W, Dunmall LSC, Lemoine NR, Wang Y, Wang Y, Wang P. The capability of heterogeneous γδ T cells in cancer treatment. Front Immunol 2023; 14:1285801. [PMID: 38077392 PMCID: PMC10704246 DOI: 10.3389/fimmu.2023.1285801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 11/06/2023] [Indexed: 12/18/2023] Open
Abstract
γδ T cells, a specialized subset of T lymphocytes, have garnered significant attention within the realm of cancer immunotherapy. Operating at the nexus between adaptive and innate immunological paradigms, these cells showcase a profound tumor discernment repertoire, hinting at novel immunotherapeutic strategies. Significantly, these cells possess the capability to directly identify and eliminate tumor cells without reliance on HLA-antigen presentation. Furthermore, γδ T cells have the faculty to present tumor antigens to αβ T cells, amplifying their anti-tumoral efficacy.Within the diverse and heterogeneous subpopulations of γδ T cells, distinct immune functionalities emerge, manifesting either anti-tumor or pro-tumor roles within the tumor microenvironment. Grasping and strategically harnessing these heterogeneous γδ T cell cohorts is pivotal to their integration in tumor-specific immunotherapeutic modalities. The aim of this review is to describe the heterogeneity of the γδ T cell lineage and the functional plasticity it generates in the treatment of malignant tumors. This review endeavors to elucidate the intricate heterogeneity inherent to the γδ T cell lineage, the consequential functional dynamics in combating malignancies, the latest advancements from clinical trials, and the evolving landscape of γδ T cell-based oncological interventions, while addressing the challenges impeding the field.
Collapse
Affiliation(s)
- Wenyi Yan
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Louisa S. Chard Dunmall
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Nicholas R. Lemoine
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Yaohe Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
- Centre for Biomarkers & Biotherapeutics, Barts Cancer Institute, Queen Mary University of London, London, United Kingdom
| | - Yafeng Wang
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Pharmaceutical Sciences, Zhengzhou University, Zhengzhou, China
| | - Pengju Wang
- Sino-British Research Centre for Molecular Oncology, National Centre for International Research in Cell and Gene Therapy, State Key Laboratory of Esophageal Cancer Prevention & Treatment, School of Basic Medical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
47
|
Hu Y, Hu Q, Li Y, Lu L, Xiang Z, Yin Z, Kabelitz D, Wu Y. γδ T cells: origin and fate, subsets, diseases and immunotherapy. Signal Transduct Target Ther 2023; 8:434. [PMID: 37989744 PMCID: PMC10663641 DOI: 10.1038/s41392-023-01653-8] [Citation(s) in RCA: 90] [Impact Index Per Article: 45.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 09/07/2023] [Accepted: 09/12/2023] [Indexed: 11/23/2023] Open
Abstract
The intricacy of diseases, shaped by intrinsic processes like immune system exhaustion and hyperactivation, highlights the potential of immune renormalization as a promising strategy in disease treatment. In recent years, our primary focus has centered on γδ T cell-based immunotherapy, particularly pioneering the use of allogeneic Vδ2+ γδ T cells for treating late-stage solid tumors and tuberculosis patients. However, we recognize untapped potential and optimization opportunities to fully harness γδ T cell effector functions in immunotherapy. This review aims to thoroughly examine γδ T cell immunology and its role in diseases. Initially, we elucidate functional differences between γδ T cells and their αβ T cell counterparts. We also provide an overview of major milestones in γδ T cell research since their discovery in 1984. Furthermore, we delve into the intricate biological processes governing their origin, development, fate decisions, and T cell receptor (TCR) rearrangement within the thymus. By examining the mechanisms underlying the anti-tumor functions of distinct γδ T cell subtypes based on γδTCR structure or cytokine release, we emphasize the importance of accurate subtyping in understanding γδ T cell function. We also explore the microenvironment-dependent functions of γδ T cell subsets, particularly in infectious diseases, autoimmune conditions, hematological malignancies, and solid tumors. Finally, we propose future strategies for utilizing allogeneic γδ T cells in tumor immunotherapy. Through this comprehensive review, we aim to provide readers with a holistic understanding of the molecular fundamentals and translational research frontiers of γδ T cells, ultimately contributing to further advancements in harnessing the therapeutic potential of γδ T cells.
Collapse
Affiliation(s)
- Yi Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Qinglin Hu
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Yongsheng Li
- Department of Medical Oncology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China
| | - Zheng Xiang
- Microbiology and Immunology Department, School of Medicine, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhinan Yin
- Biomedical Translational Research Institute, Jinan University, Guangzhou, Guangdong, 510632, China.
| | - Dieter Kabelitz
- Institute of Immunology, Christian-Albrechts-University Kiel, Kiel, Germany.
| | - Yangzhe Wu
- Guangdong Provincial Key Laboratory of Tumour Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital Affiliated with Jinan University, Jinan University, Zhuhai, Guangdong, 519000, China.
| |
Collapse
|
48
|
Chen Z, He Y, Ding C, Chen J, Gu Y, Xiao M, Li Q. Safety and Efficacy Analysis of PD-1 Inhibitors in Combination with Gemcitabine Plus Nab-Paclitaxel for Advanced Pancreatic Cancer: A Real-World, Single-Center Study. Onco Targets Ther 2023; 16:923-935. [PMID: 37965584 PMCID: PMC10642393 DOI: 10.2147/ott.s427942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2023] [Accepted: 10/20/2023] [Indexed: 11/16/2023] Open
Abstract
Background Pancreatic cancer is a deadly disease with a low five years survival rate, and chemotherapy remains the standard treatment for advanced cases. However, the efficacy of chemotherapy alone is limited, and there is a need for new treatment options. Recently, immune checkpoint inhibitors (ICIs), particularly programmed death-1 (PD-1) inhibitors, have shown promising results in various cancers, including pancreatic cancer. In this study, we explore the safety and efficacy of PD-1 inhibitors in combination with chemotherapy for advanced pancreatic cancer. Materials and Methods A retrospective analysis was conducted on clinical data from 27 patients with advanced pancreatic cancer who were administered a combination of anti-PD-1 antibody and gemcitabine plus nab-paclitaxel (GnP) regimen. The study evaluated the safety of the treatment as well as the objective response rate (ORR), disease control rate (DCR), progression-free survival (PFS), and overall survival (OS). Results In this study, treatment with a combination of anti-PD-1 antibody and GnP regimen for pancreatic cancer resulted in partial response (PR) for 10 out of 27 (37.04%) patients, stable disease (SD) for 10 (37.04%) patients, and progressive disease (PD) for 7 (25.92%) patients. The study found that the median OS (mOS) for these patients was 16.4 months [standard error (SE) = 1.117, 95% confidence interval (CI) 14.211-18.589], while the median PFS (mPFS) was 6.4 months (SE = 1.217, 95% CI 3.981-8.752). Subgroup analysis revealed that pancreatic cancer patients' Eastern Cooperative Oncology Group (ECOG) performance status (PS) (0 vs 1) and treatment cycles (≤6 cycles vs >6 cycles) significantly affected OS and PFS. Patients experienced mostly grade 1-2 adverse events (AEs), which were relieved through clinical treatment. Conclusion The combination of GnP with anti-PD-1 antibodies shows promise as a potential treatment option for advanced pancreatic cancer.
Collapse
Affiliation(s)
- Zhitao Chen
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Yahui He
- School of Medicine, Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, 310003, People’s Republic of China
| | - Chenchen Ding
- Child and Adolescent Psychology, Affiliated Mental Health Centre & Hangzhou Seventh People’s Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310013, People’s Republic of China
| | - Jun Chen
- School of Medicine, Zhejiang Chinese Medical University Zhejiang Shuren College, Hangzhou, 310003, People’s Republic of China
| | - Yangjun Gu
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Min Xiao
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| | - Qiyong Li
- Hepatobiliary and Pancreatic Surgery, Shulan (Hangzhou) Hospital Affiliated to Zhejiang Shuren University Shulan International Medical College, Hangzhou, 310003, People’s Republic of China
| |
Collapse
|
49
|
Choi H, Kim TG, Jeun SS, Ahn S. Human gamma-delta (γδ) T cell therapy for glioblastoma: A novel alternative to overcome challenges of adoptive immune cell therapy. Cancer Lett 2023; 571:216335. [PMID: 37544475 DOI: 10.1016/j.canlet.2023.216335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 05/01/2023] [Accepted: 08/03/2023] [Indexed: 08/08/2023]
Abstract
Glioblastoma is the most common brain malignancy with devastating prognosis. Numerous clinical trials using various target therapeutic agents have failed and recent clinical trials using check point inhibitors also failed to provide survival benefits for glioblastoma patients. Adoptive T cell transfer is suggested as a novel therapeutic approach that has exhibited promise in preliminary clinical studies. However, the clinical outcomes are inconsistent, and there are several limitations of current adoptive T cell transfer strategies for glioblastoma treatment. As an alternative cell therapy, gamma-delta (γδ) T cells have been recently introduced for several cancers including glioblastoma. Since the leading role of γδ T cells is immune surveillance by recognizing a broad range of ligands including stress molecules, phosphoantigens, or lipid antigens, recent studies have suggested the potential benefits of γδ T cell transfer against glioblastomas. However, γδ T cells, as a small subset (1-5%) of T cells in human peripheral blood, are relatively unknown compared to conventional alpha-beta (αβ) T cells. In this context, our study introduced γδ T cells as an alternative and novel option to overcome several challenges regarding immune cell therapy in glioblastoma treatment. We described the unique characteristics and advantages of γδ T cells compared to conventional αβ T cells and summarize several recent preclinical studies using human gamma-delta T cell therapy for glioblastomas. Finally, we suggested future direction of human γδ T cell therapy for glioblastomas.
Collapse
Affiliation(s)
- Haeyoun Choi
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Rebpulic of Korea; Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Tai-Gyu Kim
- Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Rebpulic of Korea; Catholic Hematopoietic Stem Cell Bank, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sin-Soo Jeun
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
| | - Stephen Ahn
- Department of Neurosurgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Gong W, Tian Y, Li L. T cells in abdominal aortic aneurysm: immunomodulation and clinical application. Front Immunol 2023; 14:1240132. [PMID: 37662948 PMCID: PMC10471798 DOI: 10.3389/fimmu.2023.1240132] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Accepted: 08/07/2023] [Indexed: 09/05/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is characterized by inflammatory cell infiltration, extracellular matrix (ECM) degradation, and vascular smooth muscle cell (SMC) dysfunction. The inflammatory cells involved in AAA mainly include immune cells including macrophages, neutrophils, T-lymphocytes and B lymphocytes and endothelial cells. As the blood vessel wall expands, more and more lymphocytes infiltrate into the outer membrane. It was found that more than 50% of lymphocytes in AAA tissues were CD3+ T cells, including CD4+, CD8+T cells, γδ T cells and regulatory T cells (Tregs). Due to the important role of T cells in inflammatory response, an increasing number of researchers have paid attention to the role of T cells in AAA and dug into the relevant mechanism. Therefore, this paper focuses on reviewing the immunoregulatory role of T cells in AAA and their role in immunotherapy, seeking potential targets for immunotherapy and putting forward future research directions.
Collapse
Affiliation(s)
| | | | - Lei Li
- Department of Vascular Surgery, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
| |
Collapse
|