1
|
Sureka N, Zaheer S. Regulatory T Cells in Tumor Microenvironment: Therapeutic Approaches and Clinical Implications. Cell Biol Int 2025. [PMID: 40365758 DOI: 10.1002/cbin.70031] [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: 03/10/2025] [Revised: 04/19/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025]
Abstract
Regulatory T cells (Tregs), previously referred to as suppressor T cells, represent a distinct subset of CD4+ T cells that are uniquely specialized for immune suppression. They are characterized by the constitutive expression of the transcription factor FoxP3 in their nuclei, along with CD25 (the IL-2 receptor α-chain) and CTLA-4 on their cell surface. Tregs not only restrict natural killer cell-mediated cytotoxicity but also inhibit the proliferation of CD4+ and CD8+ T-cells and suppress interferon-γ secretion by immune cells, ultimately impairing an effective antitumor immune response. Treg cells are widely recognized as a significant barrier to the effectiveness of tumor immunotherapy in clinical settings. Extensive research has consistently shown that Treg cells play a pivotal role in facilitating tumor initiation and progression. Conversely, the depletion of Treg cells has been linked to a marked delay in tumor growth and development.
Collapse
Affiliation(s)
- Niti Sureka
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| |
Collapse
|
2
|
Tian B, Wang Z, Cao M, Wang N, Jia X, Zhang Y, Zhou J, Liu S, Zhang W, Dong X, Li Z, Xue J, Wang J, Fan GH, Li Q. CCR8 antagonist suppresses liver cancer progression via turning tumor-infiltrating Tregs into less immunosuppressive phenotype. J Exp Clin Cancer Res 2025; 44:113. [PMID: 40186298 PMCID: PMC11969927 DOI: 10.1186/s13046-025-03286-x] [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: 09/25/2024] [Accepted: 01/12/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) are the main immunosuppressive cells in tumor immune microenvironment (TIME). However, systemic Treg depletion is not favored due to the crucial role of Tregs in the maintenance of immune homeostasis and prevention of autoimmunity. Recently, CCR8 has been identified as a key chemokine receptor expressed on tumor-infiltrating Tregs and targeted blockade of CCR8 exerts anticancer effect in several cancer types, but whether this pathway is involved in the progression of hepatocellular carcinoma (HCC) remains unclear. METHODS We determined the involvement of CCR8+ Tregs in HCC using human HCC tissues and TCGA database, and examined the anticancer effect and the underlying molecular mechanisms of the CCR8 antagonist, IPG0521m, which was developed in house, in murine liver cancer model with flow cytometry, bulk and single-cell RNA sequencing and Real-Time PCR. RESULTS Remarkable increase in CCR8+ Tregs was observed in human HCC tissues. Treatment of syngeneic liver cancer model with IPG0521m resulted in dramatic inhibition of tumor growth, associated with increased CD8+ T cells in tumor tissues. Bulk RNA sequencing analysis indicated that IPG0521m treatment resulted in remarkable increase in antitumor immunity. Furthermore, single-cell RNA sequencing analysis demonstrated that IPG0521m treatment resulted in a switch of Tregs from high immunosuppression to low immunosuppression phenotype, associated with elevated CD8+ T and NK cell proliferation and cytotoxicity, and decreased myeloid-derived suppressor cells and tumor-associated macrophages in the tumor tissues. CONCLUSIONS IPG0521m inhibited liver cancer growth via reducing the immunosuppressive function of Tregs, thereby boosting anti-cancer immunity. Our study paves the way for the clinical study of CCR8 antagonist in HCC and other cancers.
Collapse
MESH Headings
- Liver Neoplasms/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/genetics
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- T-Lymphocytes, Regulatory/metabolism
- Animals
- Mice
- Humans
- Receptors, CCR8/antagonists & inhibitors
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- Lymphocytes, Tumor-Infiltrating/metabolism
- Disease Progression
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Tumor Microenvironment/drug effects
- Phenotype
- Disease Models, Animal
- Cell Line, Tumor
- Immune Tolerance
Collapse
Affiliation(s)
- Binle Tian
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zhilong Wang
- Department of Oncology, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China
| | - Mei Cao
- Department of Gynecology and Obstetrics, Taihe Hospital, Hubei University of Medicine, Shiyan, 442000, China
| | - Na Wang
- Department of Antibody Development, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China
| | - Xuebing Jia
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yuanyuan Zhang
- Department of Oncology, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China
| | - Jingyi Zhou
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Sijia Liu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, 200433, China
| | - Wen Zhang
- Department of Oncology, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China
| | - Xiao Dong
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Zheng Li
- Department of Autoimmune Disease, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China
| | - Junli Xue
- Department of Oncology, Shanghai East Hospital, Tongji University School of Medicine, Shanghai, 200123, China.
| | - JianFei Wang
- Excecutive Office, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China.
- Shanghai Laboratory Animal Research Center, Shanghai, 201203, China.
| | - Guo-Huang Fan
- Excecutive Office, Immunophage Biotech Co., Ltd., 10 Lv Zhouhuang Road, Shanghai, 201114, China.
| | - Qi Li
- Cancer Center, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
- Shanghai Key Laboratory of Pancreatic Disease, Institute of Pancreatic Disease, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
3
|
Wen Y, Xia Y, Yang X, Li H, Gao Q. CCR8: a promising therapeutic target against tumor-infiltrating regulatory T cells. Trends Immunol 2025; 46:153-165. [PMID: 39890548 DOI: 10.1016/j.it.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/27/2024] [Accepted: 01/06/2025] [Indexed: 02/03/2025]
Abstract
Tumor-infiltrating regulatory T (TI-Treg) cells constitute key components within the tumor microenvironment (TME) to suppress antitumor immunity and facilitate tumor progression. Although multiple therapies have been developed to eliminate TI-Treg cells, most of them exhibit only modest efficacy and harbor risks of inducing immune-related adverse events (irAEs). Recent studies demonstrate that CC chemokine receptor (CCR)8 is highly and specifically expressed on effector TI-Treg cells in mice and humans, highlighting CCR8 as a promising target for selective TI-Treg cell depletion in the treatment of various cancers. Here, we concentrate on the latest understanding of CCR8 regarding its expression, functions, and regulation, and summarize the current landscape of CCR8-targeted therapies. With favorable efficacy and safety, the latter represent an important class of next-generation putative cancer immunotherapies.
Collapse
Affiliation(s)
- Yuanjia Wen
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yu Xia
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangping Yang
- The Second Affiliated Hospital of Guangzhou Medical University, Guangdong Provincial Key Laboratory of Allergy and Clinical Immunology, Guangzhou, China
| | - Huayi Li
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Qinglei Gao
- Department of Obstetrics and Gynecology, National Clinical Research Center for Obstetrics and Gynecology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China; Key Laboratory of Cancer Invasion and Metastasis (Ministry of Education), Hubei Key Laboratory of Tumor Invasion and Metastasis, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| |
Collapse
|
4
|
Fusco C, Di Rella F, Liotti A, Colamatteo A, Ferrara AL, Gigantino V, Collina F, Esposito E, Donzelli I, Porcellini A, Feola A, Micillo T, Perna F, Garziano F, Maniscalco GT, Varricchi G, Mottola M, Zuccarelli B, De Simone B, di Bonito M, Matarese G, Accurso A, Pontillo M, Russo D, Insabato L, Spaziano A, Cantone I, Pezone A, De Rosa V. CD4 +FOXP3Exon2 + regulatory T cell frequency predicts breast cancer prognosis and survival. SCIENCE ADVANCES 2025; 11:eadr7934. [PMID: 39813341 PMCID: PMC11734725 DOI: 10.1126/sciadv.adr7934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Accepted: 12/13/2024] [Indexed: 01/18/2025]
Abstract
CD4+FOXP3+ regulatory T cells (Tregs) suppress immune responses to tumors, and their accumulation in the tumor microenvironment (TME) correlates with poor clinical outcome in several cancers, including breast cancer (BC). However, the properties of intratumoral Tregs remain largely unknown. Here, we found that a functionally distinct subpopulation of Tregs, expressing the FOXP3 Exon2 splicing variants, is prominent in patients with hormone receptor-positive BC with poor prognosis. Notably, a comprehensive examination of the TCGA validated FOXP3E2 as an independent prognostic marker in all other BC subtypes. We found that FOXP3E2 expression underlies BCs with defective mismatch repair and a stem-like signature and highlights pathways involved in tumor survival. Last, we found that the TME induces FOXP3E2 through the CXCL12/CXCR4 axis and confirmed the higher immunosuppressive capacity of FOXP3E2+ Tregs derived from patients with BC. Our study suggests that FOXP3E2+ Tregs might be used as an independent biomarker to predict BC prognosis and survival and to develop super-targeted immunotherapies.
Collapse
Affiliation(s)
- Clorinda Fusco
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Francesca Di Rella
- Oncologia Clinica Sperimentale di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Antonietta Liotti
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Alessandra Colamatteo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Anne Lise Ferrara
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Vincenzo Gigantino
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Francesca Collina
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Emanuela Esposito
- Chirurgia Oncologica di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Ivana Donzelli
- Chirurgia Oncologica di Senologia, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Antonio Porcellini
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Antonia Feola
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Teresa Micillo
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Francesco Perna
- Dipartimento di Medicina Clinica e Chirurgia, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Federica Garziano
- U.O.C Biochimica Clinica Azienda Ospedaliera Specialistica dei Colli Monaldi-Cotugno-C.T.O. Presidio Monaldi, Napoli, Italy
| | - Giorgia Teresa Maniscalco
- Clinica Neurologica e Unità Stroke, Centro Sclerosi Multipla, Ospedale “A.Cardarelli”, Napoli 80131, Italy
| | - Gilda Varricchi
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
- Dipartimento di Scienze Mediche Traslazionali, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Maria Mottola
- U.O.C Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli, Napoli 80131, Italy
| | - Bruno Zuccarelli
- U.O.C Medicina Trasfusionale, Azienda Ospedaliera Specialistica dei Colli, Napoli 80131, Italy
| | - Bruna De Simone
- Dipartimento di Sanità Pubblica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Maurizio di Bonito
- Unità di Anatomia Patologica, Istituto Nazionale Tumori, IRCCS, Fondazione Pascale, Napoli, Italy
| | - Giuseppe Matarese
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| | - Antonello Accurso
- Dipartimento di Chirurgia Generale, Oncologica, Bariatrica e Metabolica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Martina Pontillo
- Dipartimento di Chirurgia Generale, Oncologica, Bariatrica e Metabolica, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Daniela Russo
- Unità di Anatomia Patologica, Dipartimento di Scienze Biomediche Avanzate, Facoltà di Medicina, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Luigi Insabato
- Unità di Anatomia Patologica, Dipartimento di Scienze Biomediche Avanzate, Facoltà di Medicina, Università degli Studi di Napoli “Federico II”, Napoli 80131, Italy
| | - Alessandra Spaziano
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
| | - Irene Cantone
- Dipartimento di Medicina Molecolare e Biotecnologie Mediche, Università degli Studi di Napoli “Federico II”, Napoli, Italy
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| | - Antonio Pezone
- Dipartimento di Biologia, Complesso Universitario di Monte Sant’Angelo, Università di Napoli “Federico II”, Napoli 80126, Italy
| | - Veronica De Rosa
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “G. Salvatore”, IEOS-CNR, Napoli, Italy
| |
Collapse
|
5
|
Zhang A, Fan T, Liu Y, Yu G, Li C, Jiang Z. Regulatory T cells in immune checkpoint blockade antitumor therapy. Mol Cancer 2024; 23:251. [PMID: 39516941 PMCID: PMC11545879 DOI: 10.1186/s12943-024-02156-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024] Open
Abstract
Regulatory T cells (Tregs), an essential component of the human immune system, are a heterogeneous group of T lymphocytes with the ability to suppress immune responses and maintain immune homeostasis. Recent evidence indicates that Tregs may impair antitumor immunity and facilitate cancer progression by weakening functions of effector T cells (Teffs). Consequently, targeting Tregs to eliminate them from tumor microenvironments to improve Teffs' activity could emerge as an effective strategy for cancer immunotherapy. This review outlines the biology of Tregs, detailing their origins, classification, and crucial markers. Our focus lies on the complex role of Tregs in cancer's development, progression and treatment, particularly on their suppressive role upon antitumor responses via multiple mechanisms. We delve into Tregs' involvement in immune checkpoint blockade (ICB) therapy, their dual effect on cancer immunotherapy and their potential biomarkers for ICB therapy effectiveness. We also summarize advances in the therapies that adjust Tregs to optimize ICB therapy, which may be crucial for devising innovative cancer treatment strategies.
Collapse
Affiliation(s)
- An Zhang
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Tao Fan
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Yixiao Liu
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Guanhua Yu
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Chunxiang Li
- Department of Thoracic Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Zheng Jiang
- Department of Colorectal Surgery, National Clinical Research Center for Cancer/Cancer Hospital, National Cancer Center, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
6
|
Wang P, Jiang N, Zhong J, Chen Q, Huang R, Liu C, Xu P. IFI27 enhances bladder cancer immunotherapy response by modulating regulatory T cell enrichment. J Cancer 2024; 15:6616-6630. [PMID: 39668835 PMCID: PMC11632990 DOI: 10.7150/jca.99014] [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: 05/29/2024] [Accepted: 10/12/2024] [Indexed: 12/14/2024] Open
Abstract
Bladder cancer (BCa) is the 10th most prevalent cancer globally. Neoadjuvant therapy has become the standard treatment for muscle-invasive bladder cancer, yet the pathologic complete response rate for patients is only approximately 35%. However, the mechanisms underlying neoadjuvant therapy resistance in bladder cancer patients remain unclear. We collected two sets of paired bladder cancer specimens before and after neoadjuvant therapy, and performed RNA sequencing. The findings revealed a significant decrease in IFI27 expression levels in the post-neoadjuvant therapy group compared to samples collected before treatment, suggesting that IFI27 may play a role in resistance to neoadjuvant combination therapy. IFI27, a member of the interferon-alpha (IFN-α) inducible gene family, influences the efficacy of immune checkpoint blockade therapy. Further analysis demonstrated that IFI27 is predominantly expressed in the cytoplasm of bladder cancer cells and exhibited low expression levels in bladder cancer tissues and cell lines. Subsequently, we investigated the inhibitory effects of IFI27 on bladder cancer proliferation, migration, epithelial-mesenchymal transition, and lymph node metastasis. Additionally, in a mouse model, PD-1Ab immunotherapy was found to upregulate IFI27 while downregulating the protein level of FOXP3, a key transcription factor for regulatory T cells. Flow cytometric analysis further demonstrated that IFI27 inhibits bladder cancer progression by suppressing regulatory T cell infiltration and enhancing anti-tumor immune responses. In conclusion, these findings establish IFI27 as a promising molecular marker for improving the efficacy of immunotherapy in bladder cancer and offer valuable insights into strategies for enhancing immunotherapy sensitivity.
Collapse
Affiliation(s)
- Peng Wang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Ning Jiang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Jianye Zhong
- Department of Urology, South China Hospital, Medical School, Shenzhen University, Shenzhen, China
| | - Qiwei Chen
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Renliang Huang
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Chunxiao Liu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| | - Peng Xu
- Department of Urology, Zhujiang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Zhao Y, Tang G, Li J, Bian X, Zhou X, Feng J. Integrative transcriptome analysis reveals the molecular events underlying impaired T-cell responses in EGFR-mutant lung cancer. Sci Rep 2024; 14:18366. [PMID: 39112565 PMCID: PMC11306370 DOI: 10.1038/s41598-024-69020-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 07/30/2024] [Indexed: 08/10/2024] Open
Abstract
EGFR mutations are critical oncogenic drivers in lung adenocarcinoma (LUAD). However, the mechanisms by which they impact the tumor microenvironment (TME) and tumor immunity are unclear. Furthermore, the reasons underlying the poor response of EGFR-mutant (EGFR-MU) LUADs to immunotherapy with PD-1/PD-L1 inhibitors are unknown. Utilizing single-cell RNA (sc-RNA) and bulk RNA sequencing datasets, we conducted high-dimensional weighted gene coexpression network analysis to identify key genes and immune-related pathways contributing to the immunosuppressive TME. EGFR-MU cancer cells downregulated MHC class I genes to evade CD8+ cytotoxic T cells, expressed substantial levels of MHC class II molecules, and engaged with CD4+ regulatory T cells (Tregs). EGFR-MU tumors may recruit Tregs primarily through the CCL17/CCL22/CCR4 axis, leading to a Treg-enriched TME. High levels of MHC class II-positive cancer-associated fibroblasts and tumor endothelial cells were found within EGFR-MU tumors. Owing to the absence of costimulatory factors, they may inhibit rather than activate the tumor antigen-specific CD4+ T-cell response, contributing further to immune suppression. Multiplex immunohistochemistry analyses in a LUAD cohort confirmed increased expression of MHC class II molecules in cancer cells and fibroblasts in EGFR-MU tumors. Our research elucidates the highly immunosuppressive TME in EGFR-MU LUAD and suggests potential targets for effective immunotherapy.
Collapse
Affiliation(s)
- Yu Zhao
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Gu Tang
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Jun Li
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
| | - Xiaonan Bian
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China
- Department of Clinical Laboratory, The Sixth Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Xiaorong Zhou
- Department of Immunology, Medical School of Nantong University, Nantong, Jiangsu, China.
| | - Jian Feng
- Department of Respiratory and Critical Care Medicine, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China.
| |
Collapse
|
8
|
Xin S, Zhang Y, Zhang Z, Li Z, Sun X, Liu X, Jin L, Li W, Tang C, Mei W, Cao Q, Wang H, Wei Z, Zhou Z, Li R, Wen X, Yang G, Chen W, Zheng J, Ye L. ScRNA-seq revealed the tumor microenvironment heterogeneity related to the occurrence and metastasis in upper urinary tract urothelial carcinoma. Cancer Gene Ther 2024; 31:1201-1220. [PMID: 38877164 DOI: 10.1038/s41417-024-00779-3] [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: 12/13/2023] [Revised: 04/15/2024] [Accepted: 04/22/2024] [Indexed: 06/16/2024]
Abstract
Metastasis is the greatest clinical challenge for UTUCs, which may have distinct molecular and cellular characteristics from earlier cancers. Herein, we provide single-cell transcriptome profiles of UTUC para cancer normal tissue, primary tumor lesions, and lymphatic metastases to explore possible mechanisms associated with UTUC occurrence and metastasis. From 28,315 cells obtained from normal and tumor tissues of 3 high-grade UTUC patients, we revealed the origin of UTUC tumor cells and the homology between metastatic and primary tumor cells. Unlike the immunomicroenvironment suppression of other tumors, we found no immunosuppression in the tumor microenvironment of UTUC. Moreover, it is imperative to note that stromal cells are pivotal in the advancement of UTUC. This comprehensive single-cell exploration enhances our comprehension of the molecular and cellular dynamics of metastatic UTUCs and discloses promising diagnostic and therapeutic targets in cancer-microenvironment interactions.
Collapse
Affiliation(s)
- Shiyong Xin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
- Department of Urology, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471000, China
| | - Yanwei Zhang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Zhenhua Zhang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Ziyao Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xianchao Sun
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiang Liu
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Liang Jin
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Weiyi Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Chaozhi Tang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Wangli Mei
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Qiong Cao
- Department of Pathology, The Third Affiliated Hospital of Henan University of Science and Technology, Luoyang, 471003, China
| | - Haojie Wang
- Department of Central Laboratory, Zhengzhou University, Luoyang Central Hospital, Luoyang, 471003, China
| | - Zhihao Wei
- Department of Pathology, Yiluo Hospital of Luoyang, The Teaching Hospital of Henan University of Science and Technology, Luoyang, China
| | - Zhen Zhou
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Rongbing Li
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Xiaofei Wen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Guosheng Yang
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China
| | - Weihua Chen
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200127, China.
| | - Lin Ye
- Department of Urology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, 200120, China.
| |
Collapse
|
9
|
Song X, Chen R, Li J, Zhu Y, Jiao J, Liu H, Chen Z, Geng J. Fragile Treg cells: Traitors in immune homeostasis? Pharmacol Res 2024; 206:107297. [PMID: 38977207 DOI: 10.1016/j.phrs.2024.107297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 06/18/2024] [Accepted: 07/04/2024] [Indexed: 07/10/2024]
Abstract
Regulatory T (Treg) cells play a key role in maintaining immune tolerance and tissue homeostasis. However, in some disease microenvironments, Treg cells exhibit fragility, which manifests as preserved FoxP3 expression accompanied by inflammation and loss of immunosuppression. Fragile Treg cells are formatively, phenotypically and functionally diverse in various diseases, further complicating the role of Treg cells in the immunotherapeutic response and offering novel targets for disease treatment by modulating specific Treg subsets. In this review, we summarize findings on fragile Treg cells to provide a framework for characterizing the formation and role of fragile Treg cells in different diseases, and we discuss how this information may guide the development of more specific Treg-targeted immunotherapies.
Collapse
Affiliation(s)
- Xiyu Song
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Ruo Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Jiaxin Li
- Student Brigade of Basic Medicine School, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Yumeng Zhu
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Jianhua Jiao
- Department of Urology, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Hongjiao Liu
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Zhinan Chen
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China.
| | - Jiejie Geng
- National Translational Science Center for Molecular Medicine & Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, PR China; State Key Laboratory of New Targets Discovery and Drug Development for Major Diseases, PR China.
| |
Collapse
|
10
|
Chen Q, Shen M, Yan M, Han X, Mu S, Li Y, Li L, Wang Y, Li S, Li T, Wang Y, Wang W, Wei Z, Hu C, Jin A. Targeting tumor-infiltrating CCR8 + regulatory T cells induces antitumor immunity through functional restoration of CD4 + T convs and CD8 + T cells in colorectal cancer. J Transl Med 2024; 22:709. [PMID: 39080766 PMCID: PMC11290082 DOI: 10.1186/s12967-024-05518-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Accepted: 07/18/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Chemokine (C-C motif) receptor 8 (CCR8) is a chemokine receptor selectively expressed on tumor-infiltrating regulatory T cells (Tregs). Strong immunosuppression mediated by CCR8+ Tregs observed in breast and lung malignancies suggest for their functional significance in cancer therapy. To date, detailed characterization of tumor-infiltrating CCR8+ Tregs cells in colorectal cancer (CRC) is limited. METHODS To study the presence and functional involvement of CCR8+ Tregs in CRC, we analyzed the proportions of CCR8-expressing T cells in different T cell subsets in tumor and adjacent normal tissues and peripheral blood mononuclear cells (PBMCs) from CRC patients by Flow cytometry. Also, we compared the distribution of CCR8+ T cells in malignant tissues and peripheral lymphoid organs from a subcutaneous CRC murine model. Bioinformatic analysis was performed to address the significance of CCR8 expression levels in CRC prognosis, immune regulatory gene expression profiles and potential molecular mechanisms associated with CCR8+ Tregs in CRC tumors. Further, we administrated an anti-CCR8 monoclonal antibody to CT26 tumor-bearing mice and examined the antitumor activity of CCR8-targeted therapy both in vivo and in an ex vivo confirmative model. RESULTS Here, we showed that Tregs was predominantly presented in the tumors of CRC patients (13.4 ± 5.8, p < 0.0001) and the CRC subcutaneous murine model (35.0 ± 2.6, p < 0.0001). CCR8 was found to be preferentially expressed on these tumor-infiltrating Tregs (CRC patients: 63.6 ± 16.0, p < 0.0001; CRC murine model: 65.3 ± 9.5, p < 0.0001), which correlated with poor survival. We found that majority of the CCR8+ Tregs expressed activation markers and exhibited strong suppressive functions. Treatment with anti-CCR8 antibody hampered the growth of subcutaneous CRC tumor through effectively restoring the anti-tumor immunity of CD4+ conventional T cells (CD4+ Tconvs) and CD8+ T cells, which was confirmed in the ex vivo examinations. CONCLUSIONS Collectively, these findings illustrate the importance of CCR8+ Tregs for an immunosuppressive microenvironment in CRC tumors by functional inhibition of CD4+ Tconvs and CD8+ T cells, and suggest for the applicable value of CCR8-targeted therapy for CRC.
Collapse
Affiliation(s)
- Qian Chen
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
- Department of Obstetrics, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Meiying Shen
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Min Yan
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Xiaojian Han
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Song Mu
- Department of Colorectal Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang, 550004, Guizhou, China
| | - Ya Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Luo Li
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
- Department of Clinical Laboratory, Women and Children's Hospital of Chongqing Medical University, Chongqing, 400010, China
| | - Yingming Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Shenglong Li
- School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
| | - Tingting Li
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Yingying Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Wang Wang
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China
| | - Zhengqiang Wei
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Chao Hu
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China.
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China.
| | - Aishun Jin
- Department of Immunology, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, 400010, China.
- Chongqing Key Laboratory of Tumor Immune Regulation and Immune Intervention, Chongqing Medical University, Chongqing, 400010, China.
| |
Collapse
|
11
|
Roider HG, Hoff S, Tseng SY, Berndt S, Trautwein M, Filarsky K, Gritzan U, Camps J, Nadler WM, Grudzinska-Goebel J, Ellinger P, Pesch T, Soon CF, Geyer M, Gluske K, Stelte-Ludwig B, Gorjánácz M. Selective depletion of tumor-infiltrating regulatory T cells with BAY 3375968, a novel Fc-optimized anti-CCR8 antibody. Clin Exp Med 2024; 24:122. [PMID: 38856863 PMCID: PMC11164760 DOI: 10.1007/s10238-024-01362-8] [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: 03/15/2024] [Accepted: 04/28/2024] [Indexed: 06/11/2024]
Abstract
Regulatory T cells (Tregs) are known to facilitate tumor progression by suppressing CD8+ T cells within the tumor microenvironment (TME), thereby also hampering the effectiveness of immune checkpoint inhibitors (ICIs). While systemic depletion of Tregs can enhance antitumor immunity, it also triggers undesirable autoimmune responses. Therefore, there is a need for therapeutic agents that selectively target Tregs within the TME without affecting systemic Tregs. In this study, as shown also by others, the chemokine (C-C motif) receptor 8 (CCR8) was found to be predominantly expressed on Tregs within the TME of both humans and mice, representing a unique target for selective depletion of tumor-residing Tregs. Based on this, we developed BAY 3375968, a novel anti-human CCR8 antibody, along with respective surrogate anti-mouse CCR8 antibodies, and demonstrated their in vitro mode-of-action through induction of potent antibody-dependent cellular cytotoxicity (ADCC) and phagocytosis (ADCP) activities. In vivo, anti-mouse CCR8 antibodies effectively depleted Tregs within the TME primarily via ADCP, leading to increased CD8+ T cell infiltration and subsequent tumor growth inhibition across various cancer models. This monotherapeutic efficacy was significantly enhanced in combination with ICIs. Collectively, these findings suggest that CCR8 targeting represents a promising strategy for Treg depletion in cancer therapies. BAY 3375968 is currently under investigation in a Phase I clinical trial (NCT05537740).
Collapse
MESH Headings
- T-Lymphocytes, Regulatory/immunology
- T-Lymphocytes, Regulatory/drug effects
- Receptors, CCR8/immunology
- Receptors, CCR8/antagonists & inhibitors
- Animals
- Mice
- Humans
- Tumor Microenvironment/immunology
- Tumor Microenvironment/drug effects
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/drug effects
- CD8-Positive T-Lymphocytes/immunology
- Female
- Antibody-Dependent Cell Cytotoxicity
- Lymphocyte Depletion
- Cell Line, Tumor
- Phagocytosis/drug effects
- Antibodies, Monoclonal/pharmacology
- Antibodies, Monoclonal/immunology
- Antibodies, Monoclonal/therapeutic use
Collapse
Affiliation(s)
| | | | - Su-Yi Tseng
- Bayer AG, Pharmaceuticals, San Francisco, USA
| | | | | | - Katharina Filarsky
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Roche Diagnostics GmbH, Penzberg, Germany
| | - Uwe Gritzan
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Memorial Sloan Kettering Cancer Center, New York, USA
| | | | | | | | | | | | | | | | | | - Beatrix Stelte-Ludwig
- Bayer AG, Pharmaceuticals, Wuppertal, Germany
- Current address: Vincerx Pharma, Monheim am Rhein, Germany
| | | |
Collapse
|
12
|
Yi H, Qin L, Ye X, Song J, Ji J, Ye T, Li J, Li L. Progression of radio-labeled molecular imaging probes targeting chemokine receptors. Crit Rev Oncol Hematol 2024; 195:104266. [PMID: 38232861 DOI: 10.1016/j.critrevonc.2024.104266] [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: 03/13/2023] [Revised: 12/31/2023] [Accepted: 01/11/2024] [Indexed: 01/19/2024] Open
Abstract
Chemokine receptors are significantly expressed in the surface of most inflammatory cells and tumor cells. Guided by chemokines, inflammatory cells which express the relevant chemokine receptors migrate to inflammatory lesions and participate in the evolution of inflammation diseases. Similarly, driven by chemokines, immune cells infiltrate into tumor lesions not only induces alterations in the tumor microenvironment, disrupting the efficacy of tumor therapies, but also has the potential to selectively target tumoral cells and diminish tumor progression. Chemokine receptors, which are significantly expressed on the surface of tumor cell membranes, are regulated by chemokines and initiate tumor-associated signaling pathways within tumor cells, playing a complex role in tumor progression. Based on the antagonists targeting chemokine receptors, radionuclide-labeled molecular imaging probes have been developed for the emerging application of molecular imaging in diseases such as tumors and inflammation. The value and limitations of molecular probes in disease imaging are worth reviewing.
Collapse
Affiliation(s)
- Heqing Yi
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China; Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang Province, Hangzhou 310022, China
| | - Lilin Qin
- Second Clinical Medical College of Zhejiang Chinese Medical University, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Xuemei Ye
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Jinling Song
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Jianfeng Ji
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Ting Ye
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China
| | - Juan Li
- Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Dongfang Street 150, Hangzhou, Zhejiang 310022, China.
| | - Linfa Li
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Banshan Street 1, Hangzhou, Zhejiang 310022, China.
| |
Collapse
|
13
|
Liu J, Zhang B, Zhang G, Shang D. Reprogramming of regulatory T cells in inflammatory tumor microenvironment: can it become immunotherapy turning point? Front Immunol 2024; 15:1345838. [PMID: 38449875 PMCID: PMC10915070 DOI: 10.3389/fimmu.2024.1345838] [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: 11/28/2023] [Accepted: 01/29/2024] [Indexed: 03/08/2024] Open
Abstract
Overcoming the immunosuppressive tumor microenvironment and identifying widely used immunosuppressants with minimal side effects are two major challenges currently hampering cancer immunotherapy. Regulatory T cells (Tregs) are present in almost all cancer tissues and play an important role in preserving autoimmune tolerance and tissue homeostasis. The tumor inflammatory microenvironment causes the reprogramming of Tregs, resulting in the conversion of Tregs to immunosuppressive phenotypes. This process ultimately facilitates tumor immune escape or tumor progression. However, current systemic Treg depletion therapies may lead to severe autoimmune toxicity. Therefore, it is crucial to understand the mechanism of Treg reprogramming and develop immunotherapies that selectively target Tregs within tumors. This article provides a comprehensive review of the potential mechanisms involved in Treg cell reprogramming and explores the application of Treg cell immunotherapy. The interference with reprogramming pathways has shown promise in reducing the number of tumor-associated Tregs or impairing their function during immunotherapy, thereby improving anti-tumor immune responses. Furthermore, a deeper understanding of the mechanisms that drive Treg cell reprogramming could reveal new molecular targets for future treatments.
Collapse
Affiliation(s)
- Jinming Liu
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Biao Zhang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Guolin Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, China
| | - Dong Shang
- Department of General Surgery, Clinical Laboratory of Integrative Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian, China
- Institute (College) of Integrative Medicine, Dalian Medical University, Dalian, China
| |
Collapse
|
14
|
Liao X, Yang Y, Wang L, Kong Z, Li W. CC chemokine receptors are prognostic indicators of gastric cancer and are associated with immune infiltration. BMC Med Genomics 2024; 17:1. [PMID: 38169378 PMCID: PMC10763316 DOI: 10.1186/s12920-023-01690-w] [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: 10/22/2022] [Accepted: 10/05/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND CC chemokine receptors are responsible for regulating the tumor microenvironment (TME) and participating in carcinogenesis and tumor advancement. However, no functional study has investigated CC chemokine receptors in gastric cancer (GC) prognosis, risk, immunotherapy, or other treatments. METHODS We conducted a bioinformatics analysis on GC data using online databases, including the Human Protein Atlas (HPA), Kaplan-Meier (KM) plotter, GeneMANIA, MethSurv, the University of ALabama at Birmingham CANcer (UALCAN) Data Analysis Portal, Gene Set Cancer Analysis (GSCA), cBioportal, and Tumor IMmune Estimation Resource (TIMER). RESULTS We noted that CC chemokine receptor expression correlated with survival in GC. CC chemokine receptor expression was also strongly linked to different tumor-infiltrating immune cells. Additionally, CC chemokine receptors were found to be broadly drug-resistant in GC. CONCLUSION Our study identifed CC chemokine receptor expression helped in predicting the prognosis of patients diagnosed with GC. The expression level of the CC chemokine receptors was also positively related to multiple tumor-infiltrating lymphocytes (TILs). These findings provide evidence to monitor patients with GC using CC chemokine receptors, which can be used as an effective biomarker for predicting the disease prognosis and be regarded as a therapeutic target for modulating the tumor immune microenvironment.
Collapse
Affiliation(s)
- Xinghe Liao
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yong Yang
- Department of General Surgery, the First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Lihuan Wang
- Department of Radiology, the First people's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang City, 215400, Jiangsu Province, China
| | - Zhiyuan Kong
- Department of Gastrointestinal Surgery, the First people's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang City, 215400, Jiangsu Province, China
| | - Weiping Li
- Department of Gastrointestinal Surgery, the First people's Hospital of Taicang City, Taicang Affiliated Hospital of Soochow University, Taicang City, 215400, Jiangsu Province, China.
| |
Collapse
|
15
|
Peng M, Chu X, Peng Y, Li D, Zhang Z, Wang W, Zhou X, Xiao D, Yang X. Targeted therapies in bladder cancer: signaling pathways, applications, and challenges. MedComm (Beijing) 2023; 4:e455. [PMID: 38107059 PMCID: PMC10724512 DOI: 10.1002/mco2.455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 11/28/2023] [Accepted: 11/29/2023] [Indexed: 12/19/2023] Open
Abstract
Bladder cancer (BC) is one of the most prevalent malignancies in men. Understanding molecular characteristics via studying signaling pathways has made tremendous breakthroughs in BC therapies. Thus, targeted therapies including immune checkpoint inhibitors (ICIs), antibody-drug conjugates (ADCs), and tyrosine kinase inhibitor (TKI) have markedly improved advanced BC outcomes over the last few years. However, the considerable patients still progress after a period of treatment with current therapeutic regimens. Therefore, it is crucial to guide future drug development to improve BC survival, based on the molecular characteristics of BC and clinical outcomes of existing drugs. In this perspective, we summarize the applications and benefits of these targeted drugs and highlight our understanding of mechanisms of low response rates and immune escape of ICIs, ADCs toxicity, and TKI resistance. We also discuss potential solutions to these problems. In addition, we underscore the future drug development of targeting metabolic reprogramming and cancer stem cells (CSCs) with a deep understanding of their signaling pathways features. We expect that finding biomarkers, developing novo drugs and designing clinical trials with precisely selected patients and rationalized drugs will dramatically improve the quality of life and survival of patients with advanced BC.
Collapse
Affiliation(s)
- Mei Peng
- Department of PharmacyXiangya HospitalCentral South UniversityChangshaHunanChina
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xuetong Chu
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Yan Peng
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Duo Li
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Zhirong Zhang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Weifan Wang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xiaochen Zhou
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Di Xiao
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| | - Xiaoping Yang
- Key Laboratory of Study and Discovery of Small Targeted Molecules of Hunan ProvinceThe Research Center of Reproduction and Translational Medicine of Hunan ProvinceKey Laboratory of Chemical Biology & Traditional Chinese Medicine Research of Ministry of EducationDepartment of PharmacySchool of MedicineHunan Normal UniversityChangshaHunanChina
| |
Collapse
|
16
|
Riaz F, Huang Z, Pan F. Targeting post-translational modifications of Foxp3: a new paradigm for regulatory T cell-specific therapy. Front Immunol 2023; 14:1280741. [PMID: 37936703 PMCID: PMC10626496 DOI: 10.3389/fimmu.2023.1280741] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 10/09/2023] [Indexed: 11/09/2023] Open
Abstract
A healthy immune system is pivotal for the hosts to resist external pathogens and maintain homeostasis; however, the immunosuppressive tumor microenvironment (TME) damages the anti-tumor immunity and promotes tumor progression, invasion, and metastasis. Recently, many studies have found that Foxp3+ regulatory T (Treg) cells are the major immunosuppressive cells that facilitate the formation of TME by promoting the development of various tumor-associated cells and suppressing the activity of effector immune cells. Considering the role of Tregs in tumor progression, it is pivotal to identify new therapeutic drugs to target and deplete Tregs in tumors. Although several studies have developed strategies for targeted deletion of Treg to reduce the TME and support the accumulation of effector T cells in tumors, Treg-targeted therapy systematically affects the Treg population and may lead to the progression of autoimmune diseases. It has been understood that, nevertheless, in disease conditions, Foxp3 undergoes several definite post-translational modifications (PTMs), including acetylation, glycosylation, phosphorylation, ubiquitylation, and methylation. These PTMs not only elevate or mitigate the transcriptional activity of Foxp3 but also affect the stability and immunosuppressive function of Tregs. Various studies have shown that pharmacological targeting of enzymes involved in PTMs can significantly influence the PTMs of Foxp3; thus, it may influence the progression of cancers and/or autoimmune diseases. Overall, this review will help researchers to understand the advances in the immune-suppressive mechanisms of Tregs, the post-translational regulations of Foxp3, and the potential therapeutic targets and strategies to target the Tregs in TME to improve anti-tumor immunity.
Collapse
Affiliation(s)
| | | | - Fan Pan
- Shenzhen Institute of Advanced Technology (SIAT), Chinese Academy of Sciences (CAS), Shenzhen, China
| |
Collapse
|
17
|
Nagira Y, Nagira M, Nagai R, Nogami W, Hirata M, Ueyama A, Yoshida T, Yoshikawa M, Shinonome S, Yoshida H, Haruna M, Miwa H, Chatani N, Ohkura N, Wada H, Tanaka H. S-531011, a Novel Anti-Human CCR8 Antibody, Induces Potent Antitumor Responses through Depletion of Tumor-Infiltrating CCR8-Expressing Regulatory T Cells. Mol Cancer Ther 2023; 22:1063-1072. [PMID: 37420296 PMCID: PMC10477828 DOI: 10.1158/1535-7163.mct-22-0570] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 05/17/2023] [Accepted: 06/30/2023] [Indexed: 07/09/2023]
Abstract
Although regulatory T cells (Treg) are inhibitory immune cells that are essential for maintaining immune homeostasis, Tregs that infiltrate tumor tissue promote tumor growth by suppressing antitumor immunity. Selective reduction of tumor-infiltrating Tregs is, therefore, expected to activate antitumor immunity without affecting immune homeostasis. We previously reported that selective Treg depletion targeted by a C-C motif chemokine receptor 8 (CCR8) resulted in induction of strong antitumor immunity without any obvious autoimmunity in mouse models. Thus, herein, we developed a novel humanized anti-CCR8 monoclonal antibody, S-531011, aimed as a cancer immunotherapy strategy for patients with cancer. S-531011 exclusively recognized human CCR8 among all chemokine receptors and showed potent antibody-dependent cell-mediated cytotoxicity activity toward CCR8+ cells and neutralization activity against CCR8-mediated signaling. We observed that S-531011 reduced tumor-infiltrating CCR8+ Tregs and induced potent antitumor activity in a tumor-bearing human-CCR8 knock-in mouse model. Moreover, combination therapy with S-531011 and anti-mouse programmed cell death 1 (PD-1) antibody strongly suppressed tumor growth compared with anti-PD-1 antibody alone with no observable adverse effects. S-531011 also depleted human tumor-infiltrating Tregs, but not Tregs derived from human peripheral blood mononuclear cells. These results suggest that S-531011 is a promising drug for inducing antitumor immunity without severe side effects in the clinical setting.
Collapse
Affiliation(s)
- Yoji Nagira
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Morio Nagira
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Ryohei Nagai
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Wataru Nogami
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Michinari Hirata
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Azumi Ueyama
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tetsuya Yoshida
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Mai Yoshikawa
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Satomi Shinonome
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Hiroshi Yoshida
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Miya Haruna
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hiroto Miwa
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Natsumi Chatani
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| | - Naganari Ohkura
- Department of Basic Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
- Department of Experimental Immunology, Immunology Frontier Research Center, Osaka University, Osaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hidekazu Tanaka
- Pharmaceutical Research Division, Shionogi & Co., Ltd., Osaka, Japan
| |
Collapse
|
18
|
Ueyama A, Nogami W, Nashiki K, Haruna M, Miwa H, Hagiwara M, Nagira M, Wada H, Nagira Y. Immunotherapy Targeting CCR8+ Regulatory T Cells Induces Antitumor Effects via Dramatic Changes to the Intratumor CD8+ T Cell Profile. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2023; 211:673-682. [PMID: 37350632 DOI: 10.4049/jimmunol.2300067] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Accepted: 06/02/2023] [Indexed: 06/24/2023]
Abstract
Regulatory T cells (Tregs) contribute to the formation of a tumor-immunosuppressive microenvironment. CCR8 is reportedly selectively expressed in tumor Tregs, and an anti-CCR8 Ab can exert potent antitumor effects by eliminating intratumor Tregs in murine tumor models. In this study, we analyzed changes to intratumor immunity after anti-CCR8 Ab administration, especially in CD8+ T cells, which are involved in cancer cell killing, using the CT26 colorectal carcinoma mouse model. Immunophenotyping of tumor-infiltrating cells by mass cytometry after Ab administration on day 5 of tumor inoculation revealed that CD8+ T cell subsets were dramatically altered in the CCR8 Ab-treated group, with an increase in naive cells and nonexhausted effector cells and a decrease in exhausted cells with high expression levels of TOX. These results were corroborated with flow cytometry analysis. Delayed administration of the anti-CCR8 Ab on day 9 or 12, when the amount of CCR8+ Tregs and CD8+ T cell exhaustion were more progressed, also resulted in a decrease in exhausted CD8+ T cells, leading to tumor regression. Finally, we confirmed that high CCR8+ Treg infiltration was associated with high TOX expression in CD8+ T cells in human cancer patients. In conclusion, administration of an anti-CCR8 Ab can dramatically alter the activation and exhaustion state of intratumor CD8+ T cells, resulting in strong antitumor effects. In cancer patients with an advanced tumor-immunosuppressive environment, CD8+ T cell exhaustion has progressed along with CCR8+ Treg induction. Therefore, targeted depletion of CCR8+ Tregs is expected to be effective in these patients.
Collapse
Affiliation(s)
- Azumi Ueyama
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Wataru Nogami
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Kunitaka Nashiki
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Miya Haruna
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Hiroto Miwa
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Masaki Hagiwara
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Morio Nagira
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| | - Hisashi Wada
- Department of Clinical Research in Tumor Immunology, Osaka University, Suita, Japan
| | - Yoji Nagira
- Laboratory for Bio-Drug Discovery, Shionogi & Co., Ltd., Toyonaka, Japan
| |
Collapse
|
19
|
Zhang Z, Wang G, Shao X, Wu H, Su X, Zhu L, Ji Z. A Novel Prognostic Biomarker CCR8 for Gastric Cancer and Anti-CCR8 Blockade Attenuate the Immunosuppressive Capacity of Tregs In Vitro. Cancer Biother Radiopharm 2023; 38:415-424. [PMID: 37102694 DOI: 10.1089/cbr.2022.0095] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/28/2023] Open
Abstract
Objective: To investigate the immunotherapeutic roles and functions of C-C Motif Chemokine Receptor 8 (CCR8) molecule in gastric cancer (GC). Materials and Methods: Clinicopathological features of 95 GC cases were collected by a follow-up survey. The expression level of CCR8 was measured by immunohistochemistry (IHC) staining and analyzed with the cancer genome atlas database. The relationship between CCR8 expression and Clinicopathological features of GC cases was evaluated by univariate and multivariate analysis. Flow cytometry was used to determine the expression of cytokines and the proliferation of CD4+ regulator T cells (Tregs) and CD8+ T cells. Results: An upregulated expression of CCR8 in GC tissues was associated with tumor grade, nodal metastasis, and overall survival (OS). Tumor-infiltrated Tregs with higher expression of CCR8 produced more IL10 molecules in vitro. In addition, anti-CCR8 blocking downregulated IL10 expression produced by CD4+ Tregs, and reversed the suppression by Tregs on the secretion and proliferation of CD8+ T cells. Conclusion: CCR8 molecule could be a prognostic biomarker for GC cases and a therapeutic target for immune treatments.
Collapse
Affiliation(s)
- Zhigang Zhang
- Medical School of Southeast University, Nanjing, China
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Guoqing Wang
- Department of Pathology, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Xiangyu Shao
- Medical School of Southeast University, Nanjing, China
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| | - Hailu Wu
- Medical School of Southeast University, Nanjing, China
| | - Xiangyu Su
- Medical School of Southeast University, Nanjing, China
| | - Long Zhu
- Medical School of Southeast University, Nanjing, China
| | - Zhenling Ji
- Department of General Surgery, Zhongda Hospital Affiliated to Southeast University, Nanjing, China
| |
Collapse
|
20
|
Moreno Ayala MA, Campbell TF, Zhang C, Dahan N, Bockman A, Prakash V, Feng L, Sher T, DuPage M. CXCR3 expression in regulatory T cells drives interactions with type I dendritic cells in tumors to restrict CD8 + T cell antitumor immunity. Immunity 2023; 56:1613-1630.e5. [PMID: 37392735 PMCID: PMC10752240 DOI: 10.1016/j.immuni.2023.06.003] [Citation(s) in RCA: 92] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Revised: 03/07/2023] [Accepted: 06/06/2023] [Indexed: 07/03/2023]
Abstract
Infiltration of regulatory T (Treg) cells, an immunosuppressive population of CD4+ T cells, into solid cancers represents a barrier to cancer immunotherapy. Chemokine receptors are critical for Treg cell recruitment and cell-cell interactions in inflamed tissues, including cancer, and thus are an ideal therapeutic target. Here, we show in multiple cancer models that CXCR3+ Treg cells were increased in tumors compared with lymphoid tissues, exhibited an activated phenotype, and interacted preferentially with CXCL9-producing BATF3+ dendritic cells (DCs). Genetic ablation of CXCR3 in Treg cells disrupted DC1-Treg cell interactions and concomitantly increased DC-CD8+ T cell interactions. Mechanistically, CXCR3 ablation in Treg cells increased tumor antigen-specific cross-presentation by DC1s, increasing CD8+ T cell priming and reactivation in tumors. This ultimately impaired tumor progression, especially in combination with anti-PD-1 checkpoint blockade immunotherapy. Overall, CXCR3 is shown to be a critical chemokine receptor for Treg cell accumulation and immune suppression in tumors.
Collapse
Affiliation(s)
- Mariela A Moreno Ayala
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Timothy F Campbell
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Chenyu Zhang
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Noa Dahan
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Alissa Bockman
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Varsha Prakash
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Lawrence Feng
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Theo Sher
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA
| | - Michel DuPage
- Division of Immunology and Molecular Medicine, Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA 94720, USA.
| |
Collapse
|
21
|
Li B, Chen H, Yang S, Chen F, Xu L, Li Y, Li M, Zhu C, Shao F, Zhang X, Deng C, Zeng L, He Y, Zhang C. Advances in immunology and immunotherapy for mesenchymal gastrointestinal cancers. Mol Cancer 2023; 22:71. [PMID: 37072770 PMCID: PMC10111719 DOI: 10.1186/s12943-023-01770-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 03/29/2023] [Indexed: 04/20/2023] Open
Abstract
Mesenchymal gastrointestinal cancers are represented by the gastrointestinal stromal tumors (GISTs) which occur throughout the whole gastrointestinal tract, and affect human health and economy globally. Curative surgical resections and tyrosine kinase inhibitors (TKIs) are the main managements for localized GISTs and recurrent/metastatic GISTs, respectively. Despite multi-lines of TKIs treatments prolonged the survival time of recurrent/metastatic GISTs by delaying the relapse and metastasis of the tumor, drug resistance developed quickly and inevitably, and became the huge obstacle for stopping disease progression. Immunotherapy, which is typically represented by immune checkpoint inhibitors (ICIs), has achieved great success in several solid tumors by reactivating the host immune system, and been proposed as an alternative choice for GIST treatment. Substantial efforts have been devoted to the research of immunology and immunotherapy for GIST, and great achievements have been made. Generally, the intratumoral immune cell level and the immune-related gene expressions are influenced by metastasis status, anatomical locations, driver gene mutations of the tumor, and modulated by imatinib therapy. Systemic inflammatory biomarkers are regarded as prognostic indicators of GIST and closely associated with its clinicopathological features. The efficacy of immunotherapy strategies for GIST has been widely explored in pre-clinical cell and mouse models and clinical experiments in human, and some patients did benefit from ICIs. This review comprehensively summarizes the up-to-date advancements of immunology, immunotherapy and research models for GIST, and provides new insights and perspectives for future studies.
Collapse
Affiliation(s)
- Bo Li
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Hui Chen
- Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Shaohua Yang
- Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Feng Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Liangliang Xu
- Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, Peking University Shenzhen Hospital, Shenzhen, 518036, China
| | - Yan Li
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Mingzhe Li
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Chengming Zhu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China
| | - Fangyuan Shao
- MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, Institute of Translational Medicine, Cancer Center, University of Macau, Macau SAR, 999078, China
| | - Xinhua Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan Road, Guangzhou, 510080, China
| | - Chuxia Deng
- MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, Institute of Translational Medicine, Cancer Center, University of Macau, Macau SAR, 999078, China.
| | - Leli Zeng
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Yulong He
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| | - Changhua Zhang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
22
|
Wu Y, Xu Y, He S, Li Y, Feng N, Fan J, Gong Y, Li X, Zhou L. Cytoskeleton regulator RNA expression on cancer-associated fibroblasts is associated with prognosis and immunotherapy response in bladder cancer. Heliyon 2023; 9:e13707. [PMID: 36873531 PMCID: PMC9976329 DOI: 10.1016/j.heliyon.2023.e13707] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 01/01/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Background Dysregulation of long noncoding RNAs (lncRNAs) has been reported to be associated with multiple tumors where they act as tumor suppressors or accelerators. The lncRNA CYTOR was identified as an oncogene involved in many cancers, such as gastric cancer, colorectal cancer, hepatocellular carcinoma, and renal cell carcinoma. However, the role of CYTOR in bladder cancer (BCa) has rarely been reported. Methods Using cancer datasets from The Cancer Genome Atlas (TCGA) program, we analyzed the association between CYTOR expression and prognostic value, oncogenic pathways, antitumor immunity and immunotherapy response in BCa. The influence of CYTOR on the immune infiltration pattern in the urothelial carcinoma microenvironment was further verified in our dataset. Single-cell analysis revealed the role of CYTOR in the tumor microenvironment (TME) of BCa. Finally, we evaluated the expression of CYTOR in BCa in the Peking University First Hospital (PKU-BCa) dataset and its correlation with the malignant phenotype of BCa in vitro and in vivo. Results The results indicated that CYTOR was highly expressed in multiple cancer samples, including BCa, and increased CYTOR expression contributed to poor overall survival (OS). Additionally, elevated CYTOR expression was significantly correlated with clinicopathological features of BCa, such as female sex, advanced TNM stage, high histological grade and non-papillary subtype. Functional characterization revealed that CYTOR may be involved in immune-related pathways and the epithelial mesenchymal transformation (EMT) process. Moreover, CYTOR had a significant association with infiltrating immune cells, including M2 macrophages and regulatory T cells (Tregs). CYTOR facilitates the crosstalk between cancer-associated fibroblasts (CAFs) and macrophages, and mediates M2 polarization of macrophages. Correlation analysis revealed a positive correlation between CYTOR expression and programmed cell death-1 (PD-1)/programmed death ligand 1 (PD-L1)/expression and other targets for specific immunotherapy in BCa, which are recognized to predict the efficacy of immunotherapy. Conclusions These results suggest that CYTOR serves as a potential biomarker for predicting survival outcome, TME cell infiltration characteristics and immunotherapy response in BCa.
Collapse
Key Words
- BCa, Bladder cancer
- Bladder cancer
- CAFs, Cancer-associated fibroblasts
- CIBERSOFT, Cell-type Identification By Estimating Relative Subsets Of RNA Transcripts
- CYTOR
- CYTOR, Cytoskeleton regulator RNA
- EMT, Epithelial mesenchymal transformation
- Immune infiltration
- Immunotherapy
- LncRNAs, Long non-coding RNAs
- MIBC, Muscle-invasive bladder cancer
- OS, Overall survival
- PCA, Principal component analysis
- PD-1, Programmed cell death-1
- PD-L1, Programmed death ligand 1
- RT-qPCR, Reverse transcription-quantitative polymerase chain reaction
- Survival
- TCGA, The Cancer Genome Atlas
- TME, Tumor microenvironment
- UMI, Unique molecular identifier
- UTUC, Upper-tract urothelial carcinoma
Collapse
Affiliation(s)
- Yucai Wu
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Yangyang Xu
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Shiming He
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Yifan Li
- Department of Urology, The Affiliated Hospital of Yangzhou University, Yangzhou University, Jiangsu, China
| | | | - Jian Fan
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Yanqing Gong
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Xuesong Li
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| | - Liqun Zhou
- Department of Urology, Peking University First Hospital, Beijing, China.,Institute of Urology, Peking University, Beijing, China.,National Urological Cancer Center, Beijing, China.,Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Peking University, Beijing, China
| |
Collapse
|
23
|
Yang H, Cai H, Shan S, Chen T, Zou J, Abudurufu M, Luo H, Lei Y, Ke Z, Zhu Y. Methylation of N6 adenosine-related long noncoding RNA: effects on prognosis and treatment in 'driver-gene-negative' lung adenocarcinoma. Mol Oncol 2023; 17:365-377. [PMID: 36221911 PMCID: PMC9892826 DOI: 10.1002/1878-0261.13323] [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: 02/18/2022] [Revised: 08/25/2022] [Accepted: 10/11/2022] [Indexed: 02/04/2023] Open
Abstract
The improvement of treatment for patients with 'driver-gene-negative' lung adenocarcinoma (LUAD) remains a critical problem to be solved. We aimed to explore the role of methylation of N6 adenosine (m6A)-related long noncoding RNA (lncRNA) in stratifying 'driver-gene-negative' LUAD risk. Patients negative for mutations in EGFR, KRAS, BRAF, HER2, MET, ALK, RET, and ROS1 were identified as 'driver-gene-negative' cases. RNA sequencing was performed in 46 paired tumors and adjacent normal tissues from patients with 'driver-gene-negative' LUAD. Twenty-three m6A regulators and relevant lncRNAs were identified using Pearson's correlation analysis. K-means cluster analysis was used to stratify patients, and a prognostic nomogram was developed. The CIBERSORT and pRRophetic algorithms were employed to quantify the immune microenvironment and chemosensitivity. We identified two clusters highly consistent with the prognosis based on their unique expression profiles for 46 m6AlncRNAs. A risk model constructed from nine m6A lncRNAs could stratify patients into high- and low-risk groups with promising predictive power (C-index = 0.824), and the risk score was an independent prognostic factor. The clusters and risk models were closely related to immune characteristics and chemosensitivity. Additional pan-cancer analysis using the nine m6AlncRNAs showed that the expression of DIO3 opposite strand upstream RNA (DIO3OS) is closely related to the immune/stromal score and tumor stemness in a variety of cancers. Our results show that m6AlncRNAs are a reliable prognostic tool and can aid treatment decision-making in 'driver-gene-negative' LUAD. DIO3OS is associated with the development of various cancers and has potential clinical applications.
Collapse
Affiliation(s)
- Hao‐Shuai Yang
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - He‐Yuan Cai
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Shi‐Chao Shan
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Ting‐Fei Chen
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Jian‐Yong Zou
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Maimaiti Abudurufu
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Hong‐He Luo
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Yi‐Yan Lei
- Department of Thoracic SurgeryThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zun‐Fu Ke
- Department of Pathology & Institution of Precision MedicineThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Ying Zhu
- Department of RadiologyThe First Affiliated Hospital, Sun Yat‐sen UniversityGuangzhouChina
| |
Collapse
|
24
|
Saito M, Suzuki H, Tanaka T, Asano T, Kaneko MK, Kato Y. Development of an Anti-Mouse CCR8 Monoclonal Antibody (C 8Mab-1) for Flow Cytometry and Immunocytochemistry. Monoclon Antib Immunodiagn Immunother 2022; 41:333-338. [PMID: 35483056 DOI: 10.1089/mab.2021.0069] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
It has been widely accepted that monoclonal antibody (mAb) is an effective tool for cancer immunotherapy. The C-C motif chemokine receptor 8 (CCR8) is highly expressed in regulatory T cells and many cancers and is associated with the progression of the cancers. However, its role in cancer progression remains unclear. Thus, the development of mAbs for CCR8 leads to cancer immunotherapy and elucidation of unknown mechanisms of CCR8-dependent cancer progression. In this study, we have developed an anti-mouse CCR8 (mCCR8) mAb (clone C8Mab-1, rat IgG2a, kappa) using the Cell-Based Immunization and Screening (CBIS) method. We showed that C8Mab-1 and its recombinant antibody (recC8Mab-1) bind to mCCR8-overexpressed Chinese hamster ovary (CHO)-K1 cells (CHO/mCCR8), but not to the parental CHO-K1 cells, in flow cytometry and immunofluorescence. Moreover, C8Mab-1 and recC8Mab-1 specifically reacted to P388 (a mouse lymphocyte-like cells) and J774-1 (a mouse macrophage-like cells), which express endogenous mCCR8, in both applications. These results suggest that C8Mab-1, developed using the CBIS method, is useful for flow cytometry and immunocytochemistry against exogenous and endogenous mCCR8.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Japan.,Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
25
|
Shen X, Mo X, Tan W, Mo X, Li L, Yu F, He J, Deng Z, Xing S, Chen Z, Yang J. KIAA1199 Correlates With Tumor Microenvironment and Immune Infiltration in Lung Adenocarcinoma as a Potential Prognostic Biomarker. Pathol Oncol Res 2022; 28:1610754. [PMID: 36419650 PMCID: PMC9676226 DOI: 10.3389/pore.2022.1610754] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 10/25/2022] [Indexed: 09/05/2023]
Abstract
Background: KIAA1199 has been considered a key regulator of carcinogenesis. However, the relationship between KIAA1199 and immune infiltrates, as well as its prognostic value in lung adenocarcinoma (LUAD) remains unclear. Methods: The expression of KIAA1199 and its influence on tumor prognosis were analyzed using a series of databases, comprising TIMER, GEPIA, UALCAN, LCE, Prognoscan and Kaplan-Meier Plotter. Further, immunohistochemistry (IHC), western blot (WB) and receiver operating characteristic (ROC) curve analyses were performed to verify our findings. The cBioPortal was used to investigate the genomic alterations of KIAA1199. Prediction of candidate microRNA (miRNAs) and transcription factor (TF) targeting KIAA1199, as well as GO and KEGG analyses, were performed based on LinkedOmics. TIMER and TISIDB databases were used to explore the relationship between KIAA1199 and tumor immune infiltration. Results: High expression of KIAA1199 was identified in LUAD and Lung squamous cell carcinoma (LUSC) patients. High expression of KIAA1199 indicated a worse prognosis in LUAD patients. The results of IHC and WB analyses showed that the expression level of KIAA1199 in tumor tissues was higher than that in adjacent tissues. GO and KEGG analyses indicated KIAA1199 was mainly involved in extracellular matrix (ECM)-receptor interaction and extracellular matrix structure constituent. KIAA1199 was positively correlated with infiltrating levels of CD4+ T cells, macrophages, neutrophil cells, dendritic cells, and showed positive relationship with immune marker subsets expression of a variety of immunosuppressive cells. Conclusion: High expression of KIAA1199 predicts a poor prognosis of LUAD patients. KIAA1199 might exert its carcinogenic role in the tumor microenvironment via participating in the extracellular matrix formation and regulating the infiltration of immune cells in LUAD. The results indicate that KIAA1199 might be a novel biomarker for evaluating prognosis and immune cell infiltration in LUAD.
Collapse
Affiliation(s)
- Xiaoju Shen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
- Department of Pharmacy, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Xiaocheng Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Weidan Tan
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Xiaoxiang Mo
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Li Li
- Department of Pharmacology, Guangxi Institute of Chinese Medicine and Pharmaceutical Science, Nanning, China
| | - Fei Yu
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Jingchuan He
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhihua Deng
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Shangping Xing
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Zhiquan Chen
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| | - Jie Yang
- Department of Pharmacology, School of Pharmacy, Guangxi Medical University, Nanning, China
| |
Collapse
|
26
|
Pan S, Li S, Zhan Y, Chen X, Sun M, Liu X, Wu B, Li Z, Liu B. Immune status for monitoring and treatment of bladder cancer. Front Immunol 2022; 13:963877. [PMID: 36159866 PMCID: PMC9492838 DOI: 10.3389/fimmu.2022.963877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2022] [Accepted: 08/22/2022] [Indexed: 11/24/2022] Open
Abstract
The high recurrence rate of non-muscle invasive bladder cancer (BC) and poor prognosis of advanced BC are therapeutic challenges that need to be solved. Bacillus Calmette-Guerin (BCG) perfusion was the pioneer immunotherapy for early BC, and the discovery of immune checkpoint inhibitors has created a new chapter in the treatment of advanced BC. The benefit of immunotherapy is highly anticipated, but its effectiveness still needs to be improved. In this review, we collated and analysed the currently available information and explored the mechaisms by which the internal immune imbalance of BC leads to tumour progression. The relationship between immunity and progression and the prognosis of BC has been explored through tests using body fluids such as blood and urine. These analytical tests have attempted to identify specific immuyne cells and cytokines to predict treatment outcomes and recurrence. The diversity and proportion of immune and matrix cells in BC determine the heterogeneity and immune status of tumours. The role and classification of immune cells have also been redefined, e.g., CD4 cells having recognised cytotoxicity in BC. Type 2 immunity, including that mediated by M2 macrophages, Th2 cells, and interleukin (IL)-13, plays an important role in the recurrence and progression of BC. Pathological fibrosis, activated by type 2 immunity and cancer cells, enhances the rate of cancer progression and irreversibility. Elucidating the immune status of BC and clarifying the mechanisms of action of different cells in the tumour microenvironment is the research direction to be explored in the future.
Collapse
Affiliation(s)
- Shen Pan
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Shijie Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yunhong Zhan
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xiaonan Chen
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Ming Sun
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Xuefeng Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bin Wu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhenhua Li
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Bitian Liu
- Department of Urology, Shengjing Hospital of China Medical University, Shenyang, China
- *Correspondence: Bitian Liu, ;
| |
Collapse
|
27
|
Friedrich V, Choi HW. The Urinary Microbiome: Role in Bladder Cancer and Treatment. Diagnostics (Basel) 2022; 12:diagnostics12092068. [PMID: 36140470 PMCID: PMC9497549 DOI: 10.3390/diagnostics12092068] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Revised: 08/20/2022] [Accepted: 08/24/2022] [Indexed: 11/24/2022] Open
Abstract
Commensal microbes have increasingly been found to be involved in the development and progression of cancer. The recent discovery of the urinary microbiome bolstered the notion that microbes might play a role in bladder cancer. Although microbial involvement in bladder neoplastic transformation and metastatic progression, except schisto somiasis, has not been established, accumulating research suggests that dysbiosis of the urinary microbiome can produce a chronically inflammatory urothelial microenvironment and lead to bladder cancer. In this review, we describe how the urinary microbiome might facilitate the development of bladder cancer by altering the host immune system and the kind of cytokines that are directly involved in these responses. We investigated the therapeutic possibilities of modulating the urinary microbiome, including immune checkpoint therapy. The responsiveness of patients to intravesical Bacillus Calmette-Guerin therapy was evaluated with respect to microbiome composition. We conclude by noting that the application of microbes to orchestrate the inflammatory response in the bladder may facilitate the development of treatments for bladder cancer.
Collapse
|
28
|
Jiang Y, Zeng Z, Xiong S, Jiang M, Huang G, Zhang C, Xi X. New Prognostic Gene Signature and Immune Escape Mechanisms of Bladder Cancer. Front Cell Dev Biol 2022; 10:775417. [PMID: 35646934 PMCID: PMC9133907 DOI: 10.3389/fcell.2022.775417] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Accepted: 04/14/2022] [Indexed: 11/13/2022] Open
Abstract
Background: The immune microenvironment profoundly affects tumor prognosis and therapy. The present study aimed to reveal potential immune escape mechanisms and construct a novel prognostic signature via systematic bioinformatic analysis of the bladder cancer (BLCA) immune microenvironment. Patients and Methods: The transcriptomic data and clinicopathological information for patients with BLCA were obtained from The Cancer Genome Atlas (TCGA). Consensus clustering analysis based on the CIBERSORT and ESTIMATE algorithms was performed with patients with BLCA, which divided them into two clusters. Subsequently, the differentially expressed genes (DEGs) in the two were subjected to univariate Cox and least absolute shrinkage and selection operator (LASSO) regression analyses to identify prognostic genes, which were used to construct a prognostic model. The predictive performance of the model was verified by receiver operating characteristic (ROC) and Kaplan–Meier (K-M) analyses. In addition, we analyzed the differentially altered immune cells, mutation burden, neoantigen load, and subclonal genome fraction between the two clusters to reveal the immune escape mechanism. Results: Based on the ESTIMATE and clustering analyses, patients with BLCA were classified into two heterogeneous clusters: ImmuneScoreH and ImmuneScoreL. Univariate Cox and LASSO regression analyses identified CD96 (HR = 0.83) and IBSP (HR = 1.09), which were used to construct a prognostic gene signature with significant predictive accuracy. Regarding potential immune escape mechanisms, ImmuneScoreH and ImmuneScoreL were characterized by inactivation of innate immune cell chemotaxis. In ImmuneScoreL, a low tumor antigen load might contribute to immune escape. ImmuneScoreH featured high expression of immune checkpoint molecules. Conclusion: CD96 and IBSP were considered prognostic factors for BLCA. Innate immune inactivation and a low tumor antigen load may be associated with immune escape mechanisms in both clusters. Our research complements the exploration of the immune microenvironment in BLCA.
Collapse
Affiliation(s)
- Yi Jiang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Zhenhao Zeng
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Situ Xiong
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Ming Jiang
- Department of Urology, The First Affiliated Hospital of Nanchang University, Nanchang, China
| | - Gaomin Huang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Chiyu Zhang
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Xiaoqing Xi
- Department of Urology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- *Correspondence: Xiaoqing Xi,
| |
Collapse
|
29
|
Saito M, Tanaka T, Asano T, Nakamura T, Yanaka M, Handa S, Komatsu Y, Harigae Y, Tateyama N, Nanamiya R, Li G, Suzuki H, Kaneko MK, Kato Y. C 8Mab-2: An Anti-Mouse C-C Motif Chemokine Receptor 8 Monoclonal Antibody for Immunocytochemistry. Monoclon Antib Immunodiagn Immunother 2022; 41:115-119. [PMID: 35471052 DOI: 10.1089/mab.2021.0045] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
C-C motif chemokine receptor 8 (CCR8) is a G protein-coupled receptor predominantly expressed in regulatory T (Treg) and T helper 2 cells. The evidence that CCR8 expression in Treg is increased in cancers, CCR8 increases migration activity of Treg, and CCR8 induces the anti-apoptotic activity in T cell leukemia and lymphoma suggests that CCR8 is associated with cancer development. Thus, developing a specific monoclonal antibody (mAb) for CCR8 is useful for diagnostic and therapeutic purposes and the anti-CCR8 mAb becomes a remarkable experimental tool for basic research. We previously developed an anti-mouse CCR8 (mCCR8) mAb called C8Mab-2 (rat IgG2b, kappa) that was applicable to flow cytometric analysis for both endogenous and exogenous mCCR8. This study showed that C8Mab-2 and recombinant C8Mab-2 (recC8Mab-2) were specifically bound to exogenously expressed mCCR8 in mCCR8-overexpressed Chinese hamster ovary-K1 cells. In addition, we found that C8Mab-2 and recC8Mab-2 recognized endogenous mCCR8 in P388 (a mouse lymphocyte-like cell line) and J774-1 cells (a mouse macrophage-like cell line). These data demonstrate that C8Mab-2 and recC8Mab-2 are useful for immunocytochemical analysis.
Collapse
Affiliation(s)
- Masaki Saito
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Tomohiro Tanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Teizo Asano
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Takuro Nakamura
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Miyuki Yanaka
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Saori Handa
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yu Komatsu
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yasuhiro Harigae
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Nami Tateyama
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Ren Nanamiya
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Guanjie Li
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Hiroyuki Suzuki
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Mika K Kaneko
- Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| | - Yukinari Kato
- Department of Molecular Pharmacology, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan.,Department of Antibody Drug Development, Tohoku University Graduate School of Medicine, Sendai, Miyagi, Japan
| |
Collapse
|
30
|
Haruna M, Ueyama A, Yamamoto Y, Hirata M, Goto K, Yoshida H, Higuchi N, Yoshida T, Kidani Y, Nakamura Y, Nagira M, Kawashima A, Iwahori K, Shintani Y, Ohkura N, Wada H. The impact of CCR8+ regulatory T cells on cytotoxic T cell function in human lung cancer. Sci Rep 2022; 12:5377. [PMID: 35354899 PMCID: PMC8967908 DOI: 10.1038/s41598-022-09458-5] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 03/23/2022] [Indexed: 12/02/2022] Open
Abstract
Regulatory T cells (Tregs) suppress the host immune response and maintain immune homeostasis. Tregs also promote cancer progression and are involved in resistance to immune checkpoint inhibitor treatments. Recent studies identified selective CCR8 expression on tumor-infiltrating Tregs; CCR8+ Tregs have been indicated as a possible new target of cancer immunotherapy. Here, we investigated the features of CCR8+ Tregs in lung cancer patients. CCR8+ Tregs were highly activated and infiltration of CCR8+ Tregs in tumors was associated with poor prognosis in lung cancer patients. We also investigated their immune suppressive function, especially the influence on cytotoxic T lymphocyte cell function. The Cancer Genome Atlas analysis revealed that CD8 T cell activities were suppressed in high CCR8-expressing tumors. Additionally, depletion of CCR8+ cells enhanced CD8 T cell function in an ex vivo culture of lung tumor-infiltrating cells. Moreover, CCR8+ Tregs, but not CCR8− Tregs, induced from human PBMCs markedly suppressed CD8 T cell cytotoxicity. Finally, we demonstrated the therapeutic effect of targeting CCR8 in a murine model of lung cancer. These findings reveal the significance of CCR8+ Tregs for immunosuppression in lung cancer, especially via cytotoxic T lymphocyte cell suppression, and suggest the potential value of CCR8-targeted therapy for cancer treatment.
Collapse
|
31
|
CCR8-targeted specific depletion of clonally expanded Treg cells in tumor tissues evokes potent tumor immunity with long-lasting memory. Proc Natl Acad Sci U S A 2022; 119:2114282119. [PMID: 35140181 PMCID: PMC8851483 DOI: 10.1073/pnas.2114282119] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/03/2022] [Indexed: 12/15/2022] Open
Abstract
Immunosuppressive Foxp3-expressing regulatory T cells (Tregs) in tumor tissues are assumed to be clonally expanding via recognizing tumor-associated antigens. By single-cell RNA sequencing, we have searched for the molecules that are specifically expressed by such multiclonal tumor Tregs, but not by tumor-infiltrating effector T cells or natural Tregs in other tissues. The search revealed the chemokine receptor CCR8 as a candidate. Treatment of tumor-bearing mice with cell-depleting anti-CCR8 antibody indeed selectively removed multiclonal tumor Tregs without affecting effector T cells or tissue Tregs, eradicating established tumors with induction of potent tumor-specific effector/memory T cells and without activating autoimmune T cells. Thus, specific depletion of clonally expanding tumor Tregs is clinically instrumental for evoking effective tumor immunity without autoimmune adverse effects. Foxp3-expressing CD25+CD4+ regulatory T cells (Tregs) are abundant in tumor tissues. Here, hypothesizing that tumor Tregs would clonally expand after they are activated by tumor-associated antigens to suppress antitumor immune responses, we performed single-cell analysis on tumor Tregs to characterize them by T cell receptor clonotype and gene-expression profiles. We found that multiclonal Tregs present in tumor tissues predominantly expressed the chemokine receptor CCR8. In mice and humans, CCR8+ Tregs constituted 30 to 80% of tumor Tregs in various cancers and less than 10% of Tregs in other tissues, whereas most tumor-infiltrating conventional T cells (Tconvs) were CCR8–. CCR8+ tumor Tregs were highly differentiated and functionally stable. Administration of cell-depleting anti-CCR8 monoclonal antibodies (mAbs) indeed selectively eliminated multiclonal tumor Tregs, leading to cure of established tumors in mice. The treatment resulted in the expansion of CD8+ effector Tconvs, including tumor antigen-specific ones, that were more activated and less exhausted than those induced by PD-1 immune checkpoint blockade. Anti-CCR8 mAb treatment also evoked strong secondary immune responses against the same tumor cell line inoculated several months after tumor eradication, indicating that elimination of tumor-reactive multiclonal Tregs was sufficient to induce memory-type tumor-specific effector Tconvs. Despite induction of such potent tumor immunity, anti-CCR8 mAb treatment elicited minimal autoimmunity in mice, contrasting with systemic Treg depletion, which eradicated tumors but induced severe autoimmune disease. Thus, specific removal of clonally expanding Tregs in tumor tissues for a limited period by cell-depleting anti-CCR8 mAb treatment can generate potent tumor immunity with long-lasting memory and without deleterious autoimmunity.
Collapse
|
32
|
Moser B. Chemokine Receptor-Targeted Therapies: Special Case for CCR8. Cancers (Basel) 2022; 14:511. [PMID: 35158783 PMCID: PMC8833710 DOI: 10.3390/cancers14030511] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Revised: 01/09/2022] [Accepted: 01/17/2022] [Indexed: 11/16/2022] Open
Abstract
Immune checkpoint blockade inhibitors (CBIs) targeting cytotoxic T lymphocyte associated protein-4 (CTLA-4) and program death receptor-1 (PD-1) or its ligand-1 (PD-L1) have transformed the outlook of many patients with cancer. This remarkable progress has highlighted, from the translational point of view, the importance of immune cells in the control of tumor progression. There is still room for improvement, since current CBI therapies benefit a minority of patients. Moreover, interference with immune checkpoint receptors frequently causes immune related adverse events (irAEs) with life-threatening consequences in some of the patients. Immunosuppressive cells in the tumor microenvironment (TME), including intratumoral regulatory T (Treg) cells, tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs), contribute to tumor progression and correlate with a negative disease outlook. Recent reports revealed the selective expression of the chemokine receptor CCR8 on tumor Treg cells, making CCR8 a promising target in translational research. In this review, I summarize our current knowledge about the cellular distribution and function of CCR8 in physiological and pathophysiological processes. The discussion includes an assessment of how the removal of CCR8-expressing cells might affect both anti-tumor immunity as well as immune homeostasis at remote sites. Based on these considerations, CCR8 appears to be a promising novel target to be considered in future translational research.
Collapse
Affiliation(s)
- Bernhard Moser
- Division of Infection & Immunity, Henry Wellcome Building, School of Medicine, Cardiff University, Cardiff CF14 4XN, UK
| |
Collapse
|
33
|
Chen BJ, Zhao JW, Zhang DH, Zheng AH, Wu GQ. Immunotherapy of Cancer by Targeting Regulatory T cells. Int Immunopharmacol 2022; 104:108469. [PMID: 35008005 DOI: 10.1016/j.intimp.2021.108469] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2021] [Revised: 12/05/2021] [Accepted: 12/14/2021] [Indexed: 01/23/2023]
Abstract
Regulatory T (Treg) cells maintain immune homeostasis by inhibiting abnormal/overactive immune responses to both autogenic and nonautogenic antigens. Treg cells play an important role in immune tolerance, autoimmune diseases, infectious diseases, organ transplantation, and tumor diseases. Treg cells have two functional characteristics: T cell anergy and immunosuppression. Treg cells remain immune unresponsive to high concentrations of interleukin-2 and anti-CD3 monoclonal antibodies. In addition, the activation of Treg cells after TCR-mediated signal stimulation inhibits the activation and proliferation of effector T cells. In the process of tumor development, Treg cells accumulate locally in the tumor and lead to tumor escape by inducing anergy and immunosuppression. It is believed that targeted elimination of Treg cells can activate tumor-specific effector T cells and improve the efficiency of cancer immunotherapy. Therefore, inhibition/clearance of Treg cells is a promising strategy for enhancing antitumor immunity. Here, we review studies of cancer immunotherapies targeting Treg cells.
Collapse
Affiliation(s)
- Bo-Jin Chen
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Jing-Wen Zhao
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Da-Hong Zhang
- Department of Urology Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Ai-Hong Zheng
- Department of Oncology Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| | - Guo-Qing Wu
- Department of Oncology Center, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, China.
| |
Collapse
|
34
|
Karin N. Chemokines in the Landscape of Cancer Immunotherapy: How They and Their Receptors Can Be Used to Turn Cold Tumors into Hot Ones? Cancers (Basel) 2021; 13:6317. [PMID: 34944943 PMCID: PMC8699256 DOI: 10.3390/cancers13246317] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/13/2021] [Accepted: 12/14/2021] [Indexed: 02/07/2023] Open
Abstract
Over the last decade, monoclonal antibodies to immune checkpoint inhibitors (ICI), also known as immune checkpoint blockers (ICB), have been the most successful approach for cancer therapy. Starting with mAb to cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors in metastatic melanoma and continuing with blockers of the interactions between program cell death 1 (PD-1) and its ligand program cell death ligand 1 (PDL-1) or program cell death ligand 2 (PDL-2), that have been approved for about 20 different indications. Yet for many cancers, ICI shows limited success. Several lines of evidence imply that the limited success in cancer immunotherapy is associated with attempts to treat patients with "cold tumors" that either lack effector T cells, or in which these cells are markedly suppressed by regulatory T cells (Tregs). Chemokines are a well-defined group of proteins that were so named due to their chemotactic properties. The current review focuses on key chemokines that not only attract leukocytes but also shape their biological properties. CXCR3 is a chemokine receptor with 3 ligands. We suggest using Ig-based fusion proteins of two of them: CXL9 and CXCL10, to enhance anti-tumor immunity and perhaps transform cold tumors into hot tumors. Potential differences between CXCL9 and CXCL10 regarding ICI are discussed. We also discuss the possibility of targeting the function or deleting a key subset of Tregs that are CCR8+ by monoclonal antibodies to CCR8. These cells are preferentially abundant in several tumors and are likely to be the key drivers in suppressing anti-cancer immune reactivity.
Collapse
Affiliation(s)
- Nathan Karin
- Department of Immunology, Faculty of Medicine, Technion, P.O. Box 9697, Haifa 31096, Israel
| |
Collapse
|
35
|
Fattahi S, Karimi M, Ghatreh-Samani M, Taheri F, Shirzad H, Mohammad Alibeigi F, Anjomshoa M, Bagheri N. Correlation between aryl hydrocarbon receptor and IL-17 + and Foxp3 + T-cell infiltration in bladder cancer. Int J Exp Pathol 2021; 102:249-259. [PMID: 34762773 DOI: 10.1111/iep.12392] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Revised: 01/08/2021] [Accepted: 01/21/2021] [Indexed: 01/02/2023] Open
Abstract
Bladder cancer (BC) is one of the most prevalent cancers around the world and, if not treated well, has high morbidity and mortality. Many studies have indicated that there may be various roles for the aryl hydrocarbon receptor (AHR) in the immune system. The aim of this study was to determine the frequency of Foxp3+ regulatory T (Treg) and T helper 17 cells (Th17) in BC tissue in comparison with controls and determine the relationship between AHR, Foxp3+ Treg and Th17 cells in BC. A total of 40 patients with BC were enrolled in this study. The control group was selected from non-tumoural parts of bladder tissues from the patients who have undergone cystoscopy. The percentage of regulatory T cells (Foxp3+ /CD4+ ) and Th17 (IL-17+ /CD4+ ), as well as AHR+ cells in BC tissues and controls, were determined by immunohistochemistry. The results of this study showed that the number of Foxp3+ Treg and Th17 is significantly higher in bladder tumour tissues in comparison with non-tumoural tissues. Also, the percentage of AHR+ lymphocytes and AHR+ cells was increased significantly in bladder tumour tissues rather than non-tumoural tissues. This study also found a relation between AHR and Foxp3+ /CD4+ T lymphocytes ratio cells in BC. The percentage of Foxp3+ Tregs and AHR+ cells were significantly correlated with the grade and stage of BC. An increase in the percentage of Foxp3+ Treg and Th17 cells may play an important role in tumour immunity; and determining the relationship between AHR and differentiation of Th17/Foxp3+ Treg in BC can lead to a potential cancer therapeutic possibility.
Collapse
Affiliation(s)
- Soheila Fattahi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Monireh Karimi
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Mahdi Ghatreh-Samani
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Fatemeh Taheri
- Department of Pathology, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Hedayatollah Shirzad
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | | | - Maryam Anjomshoa
- Department of Anatomical Sciences, Faculty of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Nader Bagheri
- Cellular and Molecular Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
36
|
Glasner A, Plitas G. Tumor resident regulatory T cells. Semin Immunol 2021; 52:101476. [PMID: 33906820 DOI: 10.1016/j.smim.2021.101476] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/16/2020] [Revised: 04/08/2021] [Accepted: 04/13/2021] [Indexed: 02/08/2023]
Abstract
The immune system mediates powerful effector mechanisms to protect against a diversity of pathogens and equally as important regulatory functions, to limit collateral damage of inflammation, prevent misguided immune responses to "self", and promote tissue repair. Inadequate regulatory control can lead to a variety of inflammatory disorders including autoimmunity, metabolic syndrome, allergies, and progression of malignancies. Cancers evolve complex mechanisms to thwart immune eradication including coopting normal host regulatory processes. This is most evident in the analysis of tumor infiltrating lymphocytes (TILs), where a preponderance of immunosuppressive immune cells, such as regulatory T (Treg) cells are found. Treg cells express the X-chromosome linked transcription factor Foxp3 and play a crucial role in maintaining immune homeostasis by suppressing inflammatory responses in diverse biological settings. Treg cells in the tumor microenvironment promote tumor development and progression by dampening anti-tumor immune responses, directly supporting the survival of transformed cells through elaboration of growth factors, and interacting with accessory cells in tumors such as fibroblasts and endothelial cells. Current insights into the phenotype and function of tumor associated Treg cells have opened up opportunities for their selective targeting in cancer with the goal of alleviating their suppression of anti-tumor immune responses while maintaining overall immune homeostasis. Here, we review Treg cell biology in the context of the tumor microenvironment (TME), and the important role they play in cancer immunotherapy.
Collapse
Affiliation(s)
- Ariella Glasner
- Immunology Program and Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA
| | - George Plitas
- Immunology Program and Ludwig Center for Cancer Immunotherapy, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA; Breast Service, Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY, 10065, USA.
| |
Collapse
|
37
|
Recruitment and Expansion of Tregs Cells in the Tumor Environment-How to Target Them? Cancers (Basel) 2021; 13:cancers13081850. [PMID: 33924428 PMCID: PMC8069615 DOI: 10.3390/cancers13081850] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/04/2021] [Accepted: 04/08/2021] [Indexed: 12/22/2022] Open
Abstract
Simple Summary The immune response against cancer is generated by effector T cells, among them cytotoxic CD8+ T cells that destroy cancer cells and helper CD4+ T cells that mediate and support the immune response. This antitumor function of T cells is tightly regulated by a particular subset of CD4+ T cells, named regulatory T cells (Tregs), through different mechanisms. Even if the complete inhibition of Tregs would be extremely harmful due to their tolerogenic role in impeding autoimmune diseases in the periphery, the targeted blockade of their accumulation at tumor sites or their targeted depletion represent a major therapeutic challenge. This review focuses on the mechanisms favoring Treg recruitment, expansion and stabilization in the tumor microenvironment and the therapeutic strategies developed to block these mechanisms. Abstract Regulatory T cells (Tregs) are present in a large majority of solid tumors and are mainly associated with a poor prognosis, as their major function is to inhibit the antitumor immune response contributing to immunosuppression. In this review, we will investigate the mechanisms involved in the recruitment, amplification and stability of Tregs in the tumor microenvironment (TME). We will also review the strategies currently developed to inhibit Tregs’ deleterious impact in the TME by either inhibiting their recruitment, blocking their expansion, favoring their plastic transformation into other CD4+ T-cell subsets, blocking their suppressive function or depleting them specifically in the TME to avoid severe deleterious effects associated with Treg neutralization/depletion in the periphery and normal tissues.
Collapse
|
38
|
Liu F, Wu H. CC Chemokine Receptors in Lung Adenocarcinoma: The Inflammation-Related Prognostic Biomarkers and Immunotherapeutic Targets. J Inflamm Res 2021; 14:267-285. [PMID: 33574689 PMCID: PMC7872903 DOI: 10.2147/jir.s278395] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Accepted: 11/26/2020] [Indexed: 12/11/2022] Open
Abstract
Background Lung adenocarcinoma (LUAD) is the most common type of lung cancer with a high incidence and increased mortality. CC chemokine receptors were participating in the modulation of the tumor microenvironment and involved in carcinogenesis and tumor development. However, the potential mechanistic values of CC chemokine receptors as clinical biomarkers and therapeutic targets in LUAD have not been fully clarified. Methodology ONCOMINE, UALCAN, GEPIA, Kaplan-Meier Plotter, SurvExpress, MethSurv, SurvivalMeth, cBioPortal, String, GeneMANIA, DAVID, Metascape, TRRUST, LinkedOmics, and Timer were applied in this work. Results The transcriptional levels of CCR1/10 in LUAD tissues were significantly reduced while the transcriptional levels of CCR3/6/7/8 were significantly elevated, and the expression of CCR1 was the highest in LUAD among these CC chemokine receptors. A significant correlation was found between the expression of CCR2/4/6/7 and the pathological stage of LUAD patients. There were significant associations between CCR2/3/4/5/6/10 expression levels and OS in LUAD, and LUAD patients with high transcriptional levels of CCR3/4 had inferior first-progression survival. In addition, the prognostic values of CC chemokine receptors signature in LUAD were explored in three independent cohorts, the high-risk group displayed unfavorable OS compared with the low-risk group, and the LUAD cases in the high-risk group also suffered inferior RFS than that in the low-risk group. And for the prognostic value of the DNA methylation of CC chemokine receptors, we found 1 CpG of CCR2, 2 CpGs of CCR3, 1 CpG of CCR4, 3 CpGs of CCR6, 3 CpGs of CCR7, 1 CpG of CCR8, and 3 CpGs of CCR9 were significantly associated with prognosis in LUAD patients. However, the DNA methylation signature analysis showed there was no statistically significant association between the high- and low-risk group. For potential mechanism, the neighbor gene networks, interaction analyses, functional enrichment analyses of CC chemokine receptors in LUAD were performed, the transcription factor targets, kinase targets, and miRNA targets of CC chemokine receptors were also identified in LUAD. We also found significant correlations among CC chemokine receptors expression and the infiltration of immune cells, the tumor infiltration levels among LUAD with different somatic copy number alterations of these chemokine receptors were also assessed. Moreover, the Cox proportional hazard model showed that CCR1/2/10, B_cell, CD4_Tcell were significantly related to the clinical outcome of LUAD patients. Conclusion CC chemokine receptors might serve as immunotherapeutic targets and prognostic biomarkers in LUAD.
Collapse
Affiliation(s)
- Fangteng Liu
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang, Jiangxi, 330009, People's Republic of China.,Faculty of Medicine, University of Munich, Munich, 80336, Germany
| | - Hengyu Wu
- Department of Breast Surgery, The Third Hospital of Nanchang, Nanchang, Jiangxi, 330009, People's Republic of China
| |
Collapse
|
39
|
Bayati F, Mohammadi M, Valadi M, Jamshidi S, Foma AM, Sharif-Paghaleh E. The Therapeutic Potential of Regulatory T Cells: Challenges and Opportunities. Front Immunol 2021; 11:585819. [PMID: 33519807 PMCID: PMC7844143 DOI: 10.3389/fimmu.2020.585819] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Accepted: 11/27/2020] [Indexed: 12/22/2022] Open
Abstract
Regulatory T cells (Tregs) are an immunosuppressive subgroup of CD4+ T cells which are identified by the expression of forkhead box protein P3 (Foxp3). The modulation capacity of these immune cells holds an important role in both transplantation and the development of autoimmune diseases. These cells are the main mediators of self-tolerance and are essential for avoiding excessive immune reactions. Tregs play a key role in the induction of peripheral tolerance that can prevent autoimmunity, by protecting self-reactive lymphocytes from the immune reaction. In contrast to autoimmune responses, tumor cells exploit Tregs in order to prevent immune cell recognition and anti-tumor immune response during the carcinogenesis process. Recently, numerous studies have focused on unraveling the biological functions and principles of Tregs and their primary suppressive mechanisms. Due to the promising and outstanding results, Tregs have been widely investigated as an alternative tool in preventing graft rejection and treating autoimmune diseases. On the other hand, targeting Tregs for the purpose of improving cancer immunotherapy is being intensively evaluated as a desirable and effective method. The purpose of this review is to point out the characteristic function and therapeutic potential of Tregs in regulatory immune mechanisms in transplantation tolerance, autoimmune diseases, cancer therapy, and also to discuss that how the manipulation of these mechanisms may increase the therapeutic options.
Collapse
Affiliation(s)
- Fatemeh Bayati
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Mahsa Mohammadi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran, Iran
| | - Maryam Valadi
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Saeid Jamshidi
- Research & Development Department, Aryogen Pharmed, Karaj, Iran
| | - Arron Munggela Foma
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Ehsan Sharif-Paghaleh
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Department of Imaging Chemistry and Biology, School of Biomedical Engineering and Imaging Sciences, Faculty of Life Sciences and Medicine, King’s College London, London, United Kingdom
| |
Collapse
|
40
|
Li F, Teng H, Liu M, Liu B, Zhang D, Xu Z, Wang Y, Zhou H. Prognostic Value of Immune-Related Genes in the Tumor Microenvironment of Bladder Cancer. Front Oncol 2020; 10:1302. [PMID: 32850407 PMCID: PMC7399341 DOI: 10.3389/fonc.2020.01302] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2020] [Accepted: 06/23/2020] [Indexed: 12/12/2022] Open
Abstract
The tumor microenvironment (TME) is a complex system that plays an important role in tumor development and progression, but the current knowledge about its effect on bladder cancer (BC) is scarce. In this study, we performed a comprehensive analysis of the relationship between the TME and gene expression profiles to identify prognostic biomarkers for BC. The ESTIMATE algorithm was used to calculate immune and stromal scores of BC patients who were obtained from the Gene Expression Omnibus database. We found that the immune and stromal scores were associated with clinical characteristics and the prognosis of BC patients. Based on these scores, 104 immune-related differentially expressed genes were identified. Further, functional enrichment analysis revealed that these genes were mainly involved in the immune-related biological processes and signaling pathways. Three prognostic genes were then identified and used to establish a risk prediction model using Cox regression analyses. Kaplan–Meier survival analysis showed that the expression levels of COL1A1, COMP, and SERPINE2 significantly correlated with cancer-specific survival and overall survival of BC patients. Additionally, we validated the prognostic values of these genes using two independent cohorts from The Cancer Genome Atlas and Gene Expression Omnibus databases. Finally, the relationships between the three prognostic genes and several immune cells were evaluated using Tumor Immune Estimation Resource, indicating that the expression levels of COL1A1, COMP, and SERPINE2 correlated positively with the tumor infiltration levels of CD4+ T cells and macrophages. In conclusion, the current study comprehensively analyzed the TME and presented immune-related prognostic genes for BC, providing new insights into immunotherapeutic strategies for BC patients.
Collapse
Affiliation(s)
- Faping Li
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Haolin Teng
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Mingdi Liu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Bin Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Difei Zhang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Zhixiang Xu
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Yishu Wang
- Key Laboratory of Pathobiology, Ministry of Education, Jilin University, Changchun, China
| | - Honglan Zhou
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|