1
|
Chang ML, Cheng JS, Chen WT, Shen YJ, Kuo CJ, Chien RN. Mixed cryoglobulinemia decelerates hepatocellular carcinoma development in hepatitis C patients with SVR by downregulating regulatory B cells: a 12-year prospective cohort study. Oncoimmunology 2025; 14:2470128. [PMID: 40008547 PMCID: PMC11866965 DOI: 10.1080/2162402x.2025.2470128] [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: 12/25/2024] [Revised: 02/14/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
How mixed cryoglobulinemia (MC) affects cancer risk in chronic hepatitis C patients with sustained virologic response (SVR) remains unclear. In a 12-year prospective study, post-SVR MC was assessed every 3‒6 months. Among the 891 SVR patients, 265 (29.7%) had baseline (24 weeks after completing anti-HCV therapy) MC, and the 12-year cancer cumulative incidence was 19.7%. Among the 73 patients who developed cancer, 37 (50.7%) had hepatocellular carcinoma (HCC), with the following associated baseline variables: for cancer, male sex, age and alanine aminotransferase (ALT) levels; for HCC, male sex, age, and cirrhosis; and for non-HCC cancer, rheumatoid factor levels. Among patients with post-SVR HCC, the mean time to HCC was longer in those with than in those without baseline MC (1545.4 ± 276.5 vs. 856.9 ± 115.2 days, p = 0.014). Patients with baseline MC had decreased circulating interleukin-10 (IL-10)-positive B cell (CD19+IL-10+cells/CD19+cells) (31.24 ± 16.14 vs. 40.08 ± 15.42%, p = 0.031), regulatory B cell (Breg) (CD19+CD24hi CD27+cells/CD19+cells) (10.45 ± 7.10 vs. 15.76 ± 9.14%, p = 0.035), IL-10-positive Breg (CD19+CD24hiCD27+IL-10+cells/CD19+cells) (5.06 ± 4.68 vs. 8.83 ± 5.46%, p = 0.015) and HCC-infiltrating Breg (18.6 ± 10 vs. 33.51 ± 6.8%, p = 0.022) ratios but comparable circulating and HCC-infiltrating regulatory T cell ratios relative to patients without baseline MC. In conclusion, old male SVR patients with elevated ALT levels or cirrhosis require intensive monitoring for cancer development, especially HCC. Tailored HCC follow-up is needed for SVR patients according to their baseline MC, which might downregulate Bregs to decelerate HCC development for almost 2 years.
Collapse
MESH Headings
- Humans
- Male
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/virology
- Carcinoma, Hepatocellular/etiology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/epidemiology
- Cryoglobulinemia/immunology
- Cryoglobulinemia/complications
- Female
- Liver Neoplasms/immunology
- Liver Neoplasms/virology
- Liver Neoplasms/etiology
- Liver Neoplasms/pathology
- Liver Neoplasms/epidemiology
- Middle Aged
- Prospective Studies
- B-Lymphocytes, Regulatory/immunology
- Hepatitis C, Chronic/drug therapy
- Hepatitis C, Chronic/complications
- Hepatitis C, Chronic/virology
- Hepatitis C, Chronic/immunology
- Sustained Virologic Response
- Aged
- Interleukin-10
- Antiviral Agents/therapeutic use
- Adult
- Hepacivirus
Collapse
Affiliation(s)
- Ming-Ling Chang
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Jur-Shan Cheng
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Clinical Informatics and Medical Statistics Research Center, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Wei-Ting Chen
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yi-Jyun Shen
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Chia-Jung Kuo
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Rong-Nan Chien
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
- Department of Medicine, College of Medicine, Chang Gung University, Taoyuan, Taiwan
| |
Collapse
|
2
|
Gong Y, Liu Y, Jiang F, Wang X. Ocular Immune-Related Adverse Events Associated with PD-1 Inhibitors: From Molecular Mechanisms to Clinical Management. Semin Ophthalmol 2025; 40:288-305. [PMID: 39606920 DOI: 10.1080/08820538.2024.2433636] [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: 08/22/2024] [Revised: 11/12/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
Purpose: To help ophthalmologists and oncologists better understand the ocular irAEs secondary to PD-1 inhibitors , enabling early detection and management of ocular complications.Methods: We reviewed case reports and related literatures on ocular irAEs secondary to PD-1 inhibitors in PubMed, including a total of 70 case reports, summarizing and analyzing the specific conditions of these patients.Results: The most common malignant tumors were melanoma (n = 41; 58.6%) and lung cancer (n = 13; 18.6%). The main PD-1 inhibitors used were pembrolizumab (n = 38; 54.3%) and nivolumab (n = 28; 40%). They may result in various ocular complications, with the most common being uveitis (n = 35; 50%) and myasthenia gravis (n = 13; 18.57%). Adverse events concerning the cornea and the retina were reported in 8 cases each (11.43%). Neuro-ophthalmic adverse events were reported in 6 cases (8.57%). Most of these toxicities responded to topical and systemic steroids. Severe manifestations, however, may require temporary or permanent cessation of PD-1 inhibitors treatment.Conclusions: With the increasing use of PD-1 inhibitors, ophthalmologists need to remain sensitive to the clinical manifestations of adverse events to ensure timely diagnosis and management. To improve their quality of life and reduce mortality, oncologists and ophthalmologists should maintain close cooperation and implement multi-disciplinary treatment.
Collapse
Affiliation(s)
- Yuqi Gong
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | - Yushuai Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| | | | - Xinghua Wang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei Province, China
| |
Collapse
|
3
|
Tan SN, Hao J, Ge J, Yang Y, Liu L, Huang J, Lin M, Zhao X, Wang G, Yang Z, Ni L, Dong C. Regulatory T cells converted from Th1 cells in tumors suppress cancer immunity via CD39. J Exp Med 2025; 222:e20240445. [PMID: 39907686 PMCID: PMC11797014 DOI: 10.1084/jem.20240445] [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: 03/11/2024] [Revised: 10/17/2024] [Accepted: 01/10/2025] [Indexed: 02/06/2025] Open
Abstract
Regulatory T (Treg) cells are known to impede antitumor immunity, yet the regulatory mechanisms and functional roles of these cells remain poorly understood. In this study, through the characterization of multiple cancer models, we identified a substantial presence of peripherally induced Treg cells in the tumor microenvironment (TME). Depletion of these cells triggered antitumor responses and provided potent therapeutic effects by increasing functional CD8+ T cells. Fate-mapping and transfer experiments revealed that IFN-γ-expressing T helper (Th) 1 cells differentiated into Treg cells in response to TGF-β signaling in tumors. Pseudotime trajectory analysis further revealed the terminal differentiation of Th1-like Treg cells from Th1 cells in the TME. Tumor-resident Treg cells highly expressed T-bet, which was essential for their functions in the TME. Additionally, CD39 was highly expressed by T-bet+ Treg cells in both mouse and human tumors, and was necessary for Treg cell-mediated suppression of CD8+ T cell responses. Our study elucidated the developmental pathway of intratumoral Treg cells and highlighted novel strategies for targeting them in cancer patients.
Collapse
Affiliation(s)
- Sang-Nee Tan
- School of Medicine, Westlake University, Hangzhou, China
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Jing Hao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| | - Jing Ge
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| | - Yazheng Yang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Liguo Liu
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Jia Huang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Meng Lin
- School of Medicine, Westlake University, Hangzhou, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Genyu Wang
- School of Pharmaceutical Sciences, Shanghai Jiao Tong University, Shanghai, China
| | - Zhiying Yang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Chen Dong
- School of Medicine, Westlake University, Hangzhou, China
- Shanghai Immune Therapy Institute, Shanghai Jiao Tong University School of Medicine-affiliated Renji Hospital, Shanghai, China
| |
Collapse
|
4
|
Muhammed TM, Jasim SA, Zwamel AH, Rab SO, Ballal S, Singh A, Nanda A, Ray S, Hjazi A, Yasin HA. T lymphocyte-based immune response and therapy in hepatocellular carcinoma: focus on TILs and CAR-T cells. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04035-9. [PMID: 40100377 DOI: 10.1007/s00210-025-04035-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Hepatocellular carcinoma (HCC) is among the leading causes of cancer-related death worldwide. The primary therapies for HCC are liver transplantation, hepatic tumor excision, radiofrequency ablation, and molecular-targeted medicines. An unfavorable prognosis marks HCC and has limited pharmacological response in therapeutic studies. The tumor immune microenvironment (TME) imposes significant selection pressure on HCC, resulting in its evolution and recurrence after various treatments. As the principal cellular constituents of tumor-infiltrating lymphocytes (TILs), T cells have shown both anti-tumor and protumor actions in HCC. T cell-mediated immune responses are pivotal in cancer monitoring and elimination. TILs are recognized for their critical involvement in the progression, prognosis, and immunotherapeutic management of HCC. Foxp3 + , CD8 + , CD3 + , and CD4 + T cells are the extensively researched subtypes of TILs. This article examines the functions and processes of several subtypes of TILs in HCC. Emerging T cell-based therapies, including TILs and chimeric antigen receptor (CAR)-T cell therapy, have shown tumor regression in several clinical and preclinical studies. Herein, it also delves into the existing T cell-based immunotherapies in HCC, with emphasis on TILs and CAR-T cells.
Collapse
Affiliation(s)
- Thikra Majid Muhammed
- Biology Department, College of Education for Pure Sciences, University of Anbar, Anbar, Iraq
| | - Saade Abdalkareem Jasim
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al-Maarif, Anbar, Iraq.
| | - Ahmed Hussein Zwamel
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University, Najaf, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Department of Medical Analysis, Medical Laboratory Technique College, The Islamic University of Babylon, Babylon, Iraq
| | - Safia Obaidur Rab
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Abhayveer Singh
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - Anima Nanda
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to Be University), Bhubaneswar, Odisha, 751003, India
| | - Ahmed Hjazi
- Department of Medical Laboratory, College of Applied Medical Sciences, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia.
| | - Hatif Abdulrazaq Yasin
- Department of Medical Laboratories Technology, Al-Nisour University College, Nisour Seq. Karkh, Baghdad, Iraq
| |
Collapse
|
5
|
Zhou Q, Yang T, Yu X, Li B, Liu J, Mao Y, Guo R, Feng Z, Zhou L, Zeng G, Li N, Liang J, Liu L, Feng P, Shu HB, Chen L. Lanatoside C activates the E3 ligase STUB1 to inhibit FOXP3 transcriptional activity and promote antitumor immunity. EMBO Mol Med 2025; 17:563-588. [PMID: 39979425 PMCID: PMC11904033 DOI: 10.1038/s44321-025-00200-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/29/2024] [Accepted: 02/03/2025] [Indexed: 02/22/2025] Open
Abstract
Regulatory T cells (Tregs) play critical roles in inhibiting antitumor immunity, which is dependent on FOXP3-mediated transcriptional activity. However, no Treg-specific therapeutics has been approved for clinical use. We performed a high-throughput screen of FDA-approved drugs for potential inhibitors of FOXP3 transcriptional activity. These efforts identified Lanatoside C (Lac), which potently inhibits FOXP3 activity by causing degradation of RUNX1, a FOXP3-associated component required for its transcriptional activity. Lac directly binds the E3 ligase STUB1, leading to increased polyubiquitination and proteasomal degradation of RUNX1. Lac inhibits Tregs activity and promotes antitumor immunity in a mouse primary lung cancer model. In addition, Lac synergizes with PD-1 inhibitor to shrink lung cancers driven by mutant KRAS in a mouse model. Our findings suggested that the FDA-approved Lac is a Tregs inhibitor and serves as a candidate drug for cancer patients by its own or in combination with existing therapeutics such as PD-1 inhibitors.
Collapse
Affiliation(s)
- Qian Zhou
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China.
| | - Tong Yang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Xixi Yu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Bo Li
- MOE Key Laboratory of Glucolipid Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, College of Chinese Medicine Research, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Jin Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Yongxin Mao
- MOE Key Laboratory of Glucolipid Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, College of Chinese Medicine Research, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Rongxiang Guo
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Zhuo Feng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Li Zhou
- MOE Key Laboratory of Glucolipid Metabolic Diseases, Guangdong Metabolic Diseases Research Center of Integrated Chinese and Western Medicine, College of Chinese Medicine Research, Guangdong Pharmaceutical University, 510006, Guangzhou, China
| | - Guandi Zeng
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Nan Li
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Jinxia Liang
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Lu Liu
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China
| | - Pengju Feng
- Department of Chemistry, College of Chemistry and Materials Science, Jinan University Guangzhou, 510632, Guangzhou, China
| | - Hong-Bing Shu
- Department of Infectious Diseases, Medical Research Institute, Zhongnan Hospital of Wuhan University, Wuhan University, 430073, Wuhan, China.
| | - Liang Chen
- State Key Laboratory of Bioactive Molecules and Druggability Assessment, Guangdong Basic Research Center of Excellence for Natural Bioactive Molecules and Discovery of Innovative Drugs, College of Life Science and Technology, Jinan University, 510006, Guangzhou, China.
- MOE Key Laboratory of Tumor Molecular Biology and Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, College of Life Science and Technology, Jinan University, 510632, Guangzhou, China.
| |
Collapse
|
6
|
Wang ZB, Zhang X, Fang C, Liu XT, Liao QJ, Wu N, Wang J. Immunotherapy and the ovarian cancer microenvironment: Exploring potential strategies for enhanced treatment efficacy. Immunology 2024; 173:14-32. [PMID: 38618976 DOI: 10.1111/imm.13793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/05/2024] [Indexed: 04/16/2024] Open
Abstract
Despite progress in cancer immunotherapy, ovarian cancer (OC) prognosis continues to be disappointing. Recent studies have shed light on how not just tumour cells, but also the complex tumour microenvironment, contribute to this unfavourable outcome of OC immunotherapy. The complexities of the immune microenvironment categorize OC as a 'cold tumour'. Nonetheless, understanding the precise mechanisms through which the microenvironment influences the effectiveness of OC immunotherapy remains an ongoing scientific endeavour. This review primarily aims to dissect the inherent characteristics and behaviours of diverse cells within the immune microenvironment, along with an exploration into its reprogramming and metabolic changes. It is expected that these insights will elucidate the operational dynamics of the immune microenvironment in OC and lay a theoretical groundwork for improving the efficacy of immunotherapy in OC management.
Collapse
Affiliation(s)
- Zhi-Bin Wang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Xiu Zhang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Chao Fang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hunan Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Xiao-Ting Liu
- The Second People's Hospital of Hunan Province, Changsha, China
| | - Qian-Jin Liao
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Nayiyuan Wu
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| | - Jing Wang
- Hunan Gynecological Tumor Clinical Research Center; Hunan Key Laboratory of Cancer Metabolism; Hunan Cancer Hospital, and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Public Service Platform of Tumor Organoids Technology, Changsha, China
| |
Collapse
|
7
|
Almarii F, Sajin M, Simion G, Dima SO, Herlea V. Analyzing the Spatial Distribution of Immune Cells in Lung Adenocarcinoma. J Pers Med 2024; 14:925. [PMID: 39338178 PMCID: PMC11433064 DOI: 10.3390/jpm14090925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 09/30/2024] Open
Abstract
(1) Background: This study investigates the tumor immune microenvironment, focusing on immune cell distribution in lung adenocarcinoma. (2) Methods: We evaluated fifty cases of lung adenocarcinoma, and suitable areas for further studies were annotated on the histological slides. Two tumor cores per case were obtained, one from the tumor's center and another from its periphery, and introduced into three paraffin receptor blocks for optimized processing efficiency. The 4-micrometer-thick tissue microarray sections were stained for H&E and for CD68, CD163, CD8, CD4, and PD-L1; (3) Results: Our investigation revealed significant correlations between PD-L1 expression in tumor cells and the presence of CD163+ macrophages, between CD4+ cells and CD8+, CD68+, and CD163+ cells, and also between CD8+ T cells and CD163+ cells. Additionally, while we observed some differences in cellular components and densities between the tumor center and periphery, these differences were not statistically significant. However, distinct correlations between PD-L1 and immune cells in these regions were identified, suggesting spatial heterogeneity in the immune landscape. (4) Conclusions: These results emphasize the intricate interactions between immune cells and tumor cells in lung adenocarcinoma. Understanding patient spatial immune profile could improve patient selection for immunotherapy, ensuring that those most likely to benefit are identified.
Collapse
Affiliation(s)
- Florina Almarii
- Department of Pathology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Pathology, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Maria Sajin
- Department of Pathology, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Pathology, Emergency University Hospital, 050098 Bucharest, Romania
| | - George Simion
- Department of Pathology, Emergency University Hospital, 050098 Bucharest, Romania
| | - Simona O Dima
- Department of Pathology, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Surgery, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Histopathology, The Center for Excellence in Translational Medicine, 022328 Bucharest, Romania
| | - Vlad Herlea
- Department of Pathology, Fundeni Clinical Institute, 022328 Bucharest, Romania
- Department of Pathology, "Carol Davila" University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Department of Histopathology, The Center for Excellence in Translational Medicine, 022328 Bucharest, Romania
| |
Collapse
|
8
|
Pu X, Li L, Xu F, Wang Z, Fu Y, Wu H, Ren J, Chen J, Sun B. HER2 amplification subtype intrahepatic cholangiocarcinoma exhibits high mutation burden and T cell exhaustion microenvironment. J Cancer Res Clin Oncol 2024; 150:403. [PMID: 39198311 PMCID: PMC11358322 DOI: 10.1007/s00432-024-05894-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 07/15/2024] [Indexed: 09/01/2024]
Abstract
OBJECTIVE This study aimed to establish a uniform standard for the interpretation of HER2 gene and protein statuses in intrahepatic cholangiocarcinoma (ICC). We also intended to explore the clinical pathological characteristics, molecular features, RNA expression and immune microenvironment of HER2-positive ICC. METHODS We analyzed a cohort of 304 ICCs using immunohistochemistry (IHC) and fluorescence in situ hybridization (FISH) to identify HER2 status. Comprehensive analyses of the clinicopathological, molecular genetic, and RNA expression characterizations of ICCs with varying HER2 statuses were performed using next-generation sequencing. We further investigated the tumor microenvironment of ICCs with different HER2 statuses using IHC and multiplex immunofluorescence staining. RESULTS HER2/CEP17 ratio of ≥ 2.0 and HER2 copy number ≥ 4.0; or HER2 copy number ≥ 6.0 were setup as FISH positive criteria. Based on this criterion, 13 (4.27%, 13/304) samples were classified as having HER2 amplification. The agreement between FISH and IHC results in ICC was poor. HER2-amplified cases demonstrated a higher tumor mutational burden compared to non-amplified cases. No significant differences were observed in immune markers between the two groups. However, an increased density of CD8 + CTLA4 + and CD8 + FOXP3 + cells was identified in HER2 gene-amplified cases. CONCLUSION FISH proves to be more appropriate as the gold standard for HER2 evaluation in ICC. HER2 gene-amplified ICCs exhibit poorer prognosis, higher mutational burden, and T cell exhaustion and immune suppressed microenvironment.
Collapse
Affiliation(s)
- Xiaohong Pu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Lin Li
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Feng Xu
- Department of Medical Imaging, The Affiliated Suqian First People's Hospital of Nanjing Medical University, 223800, Suqian, Jiangsu Province, China
| | - Ziyu Wang
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Yao Fu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Hongyan Wu
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China
| | - Jun Ren
- Department of General Surgery, Northern Jiangsu People's Hospital, Yangzhou University, Yangzhou, 225000, Jiangsu Province, China.
| | - Jun Chen
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China.
| | - Beicheng Sun
- Medical School, Nanjing Drum Tower Hospital, The Affiliated Drum Tower Hospital of Medical School, Nanjing University, Nanjing, 210008, Jiangsu Province, China.
| |
Collapse
|
9
|
Ma J, Shi Y, Lu Q, Huang D. Inflammation-Related Gene ADH1A Regulates the Polarization of Macrophage M1 and Influences the Malignant Progression of Gastric Cancer. J Inflamm Res 2024; 17:4647-4665. [PMID: 39045532 PMCID: PMC11264289 DOI: 10.2147/jir.s452670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 06/15/2024] [Indexed: 07/25/2024] Open
Abstract
Background Gastric cancer (GC) is a malignant tumor originating from the gastric mucosa epithelium, and there is a low survival rate of GC patients after treatment, with a poor prognostic outcome. The inflammatory response within the tumor microenvironment plays an important role in GC progression. Methods We downloaded GC-related datasets and inflammation-related genes from GEO, TCGA and MSigDB databases, performed differential analysis, protein-protein interaction analysis, immunoinfiltration analysis and Lasso analysis to screen inflammation-related hub genes affecting GC progression, and carried out qRT-PCR for validation. In order to explore the role of ADH1A, we constructed overexpressed plasmids, treated GC cells with cGMP/PKG pathway agonist 8-Br-cGMP, and tested cell functions with CCK8, EdU, Transwell, scratch assay and other experiments. On this basis, GC cells were co-cultured with monocyte THP-1 to explore the effect of ADH1A on the polarization of macrophages. Results ADH1A was significantly decreased in GC cells, and its expression trend was consistent with the results of bioinformatics analysis. Therefore, we chose ADH1A for subsequent functional validation. Overexpression of ADH1A in GC cells revealed ADH1A's role in inhibiting the activity, proliferation, migration and invasion of GC cells, promoting apoptosis and secretion of IL-6, IFN-γ, CCL5 and CSF2, and facilitating the transformation of macrophages to a pro-inflammatory M1 phenotype. ssGSEA results demonstrated the potential involvement of ADH1A in the cGMP/PKG signaling pathway, and significant changes in the expression of proteins related to the cGMP/PKG signaling pathway. The use of the cGMP/PKG signaling pathway agonist 8-Br-cGMP in ADH1A-overexpressing GC cells substantiated ADH1A's capacity to inhibit the cGMP/PKG signaling pathway, thereby suppressing the malignant progression of GC and promoting the transformation of macrophages to a pro-inflammatory M1 phenotype. Conclusion ADH1A is able to influence the malignant progression of GC and the transformation of macrophages to the pro-inflammatory M1 phenotype through the cGMP/PKG signaling pathway.
Collapse
Affiliation(s)
- Jun Ma
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Yongkang Shi
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Qiliang Lu
- General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Dongsheng Huang
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| |
Collapse
|
10
|
Qin Y, Sheng Y, Ren M, Hou Z, Xiao L, Chen R. Identification of necroptosis-related gene signatures for predicting the prognosis of ovarian cancer. Sci Rep 2024; 14:11133. [PMID: 38750159 PMCID: PMC11096311 DOI: 10.1038/s41598-024-61849-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 05/10/2024] [Indexed: 05/18/2024] Open
Abstract
Ovarian cancer (OC) is one of the most prevalent and fatal malignant tumors of the female reproductive system. Our research aimed to develop a prognostic model to assist inclinical treatment decision-making.Utilizing data from The Cancer Genome Atlas (TCGA) and copy number variation (CNV) data from the University of California Santa Cruz (UCSC) database, we conducted analyses of differentially expressed genes (DEGs), gene function, and tumor microenvironment (TME) scores in various clusters of OC samples.Next, we classified participants into low-risk and high-risk groups based on the median risk score, thereby dividing both the training group and the entire group accordingly. Overall survival (OS) was significantly reduced in the high-risk group, and two independent prognostic factors were identified: age and risk score. Additionally, three genes-C-X-C Motif Chemokine Ligand 10 (CXCL10), RELB, and Caspase-3 (CASP3)-emerged as potential candidates for an independent prognostic signature with acceptable prognostic value. In Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses, pathways related to immune responses and inflammatory cell chemotaxis were identified. Cellular experiments further validated the reliability and precision of our findings. In conclusion, necroptosis-related genes play critical roles in tumor immunity, and our model introduces a novel strategy for predicting the prognosis of OC patients.
Collapse
Affiliation(s)
- Yuling Qin
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5, Beixiange Road, Xicheng District, Beijing, 100053, China
| | - Yawen Sheng
- Shandong University of Traditional Chinese Medicine, Jinan, 250014, Shandong, China
| | - Mengxue Ren
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5, Beixiange Road, Xicheng District, Beijing, 100053, China
| | - Zitong Hou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5, Beixiange Road, Xicheng District, Beijing, 100053, China
| | - Lu Xiao
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5, Beixiange Road, Xicheng District, Beijing, 100053, China
| | - Ruixue Chen
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, No. 5, Beixiange Road, Xicheng District, Beijing, 100053, China.
| |
Collapse
|
11
|
Schmälter AK, Löhr P, Konrad M, Waidhauser J, Arndt TT, Schiele S, Thoma A, Hackanson B, Rank A. Alterations in Peripheral Lymphocyte Subsets under Immunochemotherapy in Stage IV SCLC Patients: Th17 Cells as Potential Early Predictive Biomarker for Response. Int J Mol Sci 2024; 25:5056. [PMID: 38791096 PMCID: PMC11121216 DOI: 10.3390/ijms25105056] [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: 03/30/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 05/26/2024] Open
Abstract
UICC stage IV small-cell lung cancer (SCLC) is a highly aggressive malignancy without curative treatment options. Several randomized trials have demonstrated improved survival rates through the addition of checkpoint inhibitors to first-line platin-based chemotherapy. Consequently, a combination of chemo- and immunotherapy has become standard palliative treatment. However, no reliable predictive biomarkers for treatment response exist. Neither PD-L1 expression nor tumor mutational burden have proven to be effective predictive biomarkers. In this study, we compared the cellular immune statuses of SCLC patients to a healthy control cohort and investigated changes in peripheral blood B, T, and NK lymphocytes, as well as several of their respective subsets, during treatment with immunochemotherapy (ICT) using flow cytometry. Our findings revealed a significant decrease in B cells, while T cells showed a trend to increase throughout ICT. Notably, high levels of exhausted CD4+ and CD8+ cells, alongside NK subsets, increased significantly during treatment. Furthermore, we correlated decreases/increases in subsets after two cycles of ICT with survival. Specifically, a decrease in Th17 cells indicated a better overall survival. Based on these findings, we suggest conducting further investigation into Th17 cells as a potential early predictive biomarkers for response in patients receiving palliative ICT for stage IV SCLC.
Collapse
Affiliation(s)
- Ann-Kristin Schmälter
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
- Bavarian Cancer Research Center (BZKF), 86156 Augsburg, Germany
| | - Phillip Löhr
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
- Bavarian Cancer Research Center (BZKF), 86156 Augsburg, Germany
| | - Maik Konrad
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
| | - Johanna Waidhauser
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
| | - Tim Tobias Arndt
- Institute of Mathematics, University of Augsburg, 86159 Augsburg, Germany; (T.T.A.); (S.S.)
| | - Stefan Schiele
- Institute of Mathematics, University of Augsburg, 86159 Augsburg, Germany; (T.T.A.); (S.S.)
| | - Alicia Thoma
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
| | - Björn Hackanson
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
- Bavarian Cancer Research Center (BZKF), 86156 Augsburg, Germany
- Department of Medicine I, Medical Center, Faculty of Medicine, University of Freiburg, 79106 Freiburg, Germany
| | - Andreas Rank
- Department of Hematology and Oncology, Augsburg University Hospital and Medical Faculty, Comprehensive Cancer Center Augsburg, 86156 Augsburg, Germany; (P.L.); (M.K.); (J.W.); (A.T.); (B.H.); (A.R.)
| |
Collapse
|
12
|
Wang L, Zhang J, Zhang W, Zheng M, Guo H, Pan X, Li W, Yang B, Ding L. The inhibitory effect of adenosine on tumor adaptive immunity and intervention strategies. Acta Pharm Sin B 2024; 14:1951-1964. [PMID: 38799637 PMCID: PMC11119508 DOI: 10.1016/j.apsb.2023.12.004] [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: 07/05/2023] [Revised: 10/02/2023] [Accepted: 11/14/2023] [Indexed: 05/29/2024] Open
Abstract
Adenosine (Ado) is significantly elevated in the tumor microenvironment (TME) compared to normal tissues. It binds to adenosine receptors (AdoRs), suppressing tumor antigen presentation and immune cell activation, thereby inhibiting tumor adaptive immunity. Ado downregulates major histocompatibility complex II (MHC II) and co-stimulatory factors on dendritic cells (DCs) and macrophages, inhibiting antigen presentation. It suppresses anti-tumor cytokine secretion and T cell activation by disrupting T cell receptor (TCR) binding and signal transduction. Ado also inhibits chemokine secretion and KCa3.1 channel activity, impeding effector T cell trafficking and infiltration into the tumor site. Furthermore, Ado diminishes T cell cytotoxicity against tumor cells by promoting immune-suppressive cytokine secretion, upregulating immune checkpoint proteins, and enhancing immune-suppressive cell activity. Reducing Ado production in the TME can significantly enhance anti-tumor immune responses and improve the efficacy of other immunotherapies. Preclinical and clinical development of inhibitors targeting Ado generation or AdoRs is underway. Therefore, this article will summarize and analyze the inhibitory effects and molecular mechanisms of Ado on tumor adaptive immunity, as well as provide an overview of the latest advancements in targeting Ado pathways in anti-tumor immune responses.
Collapse
Affiliation(s)
- Longsheng Wang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wenxin Zhang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongjie Guo
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaohui Pan
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Wen Li
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Bo Yang
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- The Innovation Institute for Artificial Intelligence in Medicine, Zhejiang University, Hangzhou 310018, China
| | - Ling Ding
- Zhejiang Province Key Laboratory of Anti-Cancer Drug Research, Institute of Pharmacology and Toxicology, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
- Nanhu Brain-Computer Interface Institute, Hangzhou 311100, China
| |
Collapse
|
13
|
Jiang B, Ye X, Wang W, He J, Zhang S, Zhang S, Bao L, Xu X. Comprehensive assessment of regulatory T-cells-related scoring system for predicting the prognosis, immune microenvironment and therapeutic response in hepatocellular carcinoma. Aging (Albany NY) 2024; 16:5288-5310. [PMID: 38461439 PMCID: PMC11006487 DOI: 10.18632/aging.205649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Accepted: 01/23/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Regulatory T cells (Tregs) play important roles in tumor immunosuppression and immune escape. The aim of the present study was to construct a novel Tregs-associated biomarker for the prediction of tumour immune microenvironment (TIME), clinical outcomes, and individualised treatment in hepatocellular carcinoma (HCC). METHODS Single-cell sequencing data were obtained from the three independent cohorts. Cox and LASSO regression were utilised to develop the Tregs Related Scoring System (TRSSys). GSE140520, ICGC-LIRI and CHCC cohorts were used for the validation of TRSSys. Kaplan-Meier, ROC, and Cox regression were utilised for the evaluation of TRSSys. The ESTIMATE, TIMER 2.0, and ssGSEA algorithm were utilised to determine the value of TRSSys in predicting the TIME. GSVA, GO, KEGG, and TMB analyses were used for mechanistic exploration. Finally, the value of TRSSys in predicting drug sensitivity was evaluated based on the oncoPredict algorithm. RESULTS Comprehensive validation showed that TRSSys had good prognostic predictive efficacy and applicability. Additionally, ssGSEA, TIMER and ESTIMATE algorithm suggested that TRSSys could help to distinguish different TIME subtypes and determine the beneficiary population of immunotherapy. Finally, the oncoPredict algorithm suggests that TRSSys provides a basis for individualised treatment. CONCLUSIONS TRSSys constructed in the current study is a novel HCC prognostic prediction biomarker with good predictive efficacy and stability. Additionally, risk stratification based on TRSSys can help to identify the TIME landscape subtypes and provide a basis for individualized treatment options.
Collapse
Affiliation(s)
- Bitao Jiang
- Department of Hematology and Oncology, Beilun District People’s Hospital, Ningbo, China
| | - Xiaojuan Ye
- Radiotherapy Department, The Second People’s Hospital of Wuhu, Wuhu, China
| | - Wenjuan Wang
- Department of Hematology and Oncology, Beilun District People’s Hospital, Ningbo, China
| | - Jiajia He
- Department of Hematology and Oncology, Ningbo Yinzhou No. 2 Hospital, Ningbo, China
| | - Shuyan Zhang
- Pharmacy Department, Beilun District People’s Hospital, Ningbo, China
| | - Song Zhang
- Department of Hematology and Oncology, Beilun District People’s Hospital, Ningbo, China
| | - Lingling Bao
- Department of Hematology and Oncology, Beilun District People’s Hospital, Ningbo, China
| | - Xin Xu
- Department of Hematology and Oncology, Beilun District People’s Hospital, Ningbo, China
| |
Collapse
|
14
|
Zhou H, Yu CY, Wei H. Liposome-based nanomedicine for immune checkpoint blocking therapy and combinatory cancer therapy. Int J Pharm 2024; 652:123818. [PMID: 38253269 DOI: 10.1016/j.ijpharm.2024.123818] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 01/06/2024] [Accepted: 01/16/2024] [Indexed: 01/24/2024]
Abstract
The discovery of immune checkpoint (IC) has led to a wave of leap forward in cancer immunotherapy that represents probably the most promising strategy for cancer therapy. However, the clinical use of immune checkpoint block (ICB) therapy is limited by response rates and side effects. A strategy that addresses the limitations of ICB therapies through combination therapies, using nanocarriers as mediators, has been mentioned in numerous research papers. Liposomes have been probably one of the most extensively used nanocarriers for clinical applications, with broad drug delivery and high safety. A timely review on this hot subject of research, i.e., the application of liposomes for ICB, is thus highly desirable for both fundamental and clinical translatable studies, but remains, to our knowledge, unexplored so far. For this purpose, this review is composed to address the dilemma of ICB therapy and the reasons for this dilemma. We later describe how other cancer treatments have broken this dilemma. Finally, we focus on the role of liposomes in various combinatory cancer therapy. This review is believed to serve as a guidance for the rational design and development of liposome for immunotherapy with enhanced therapeutic efficiency.
Collapse
Affiliation(s)
- Haoyuan Zhou
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China
| | - Cui-Yun Yu
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| | - Hua Wei
- Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, School of Pharmaceutical of Science, Hengyang 421001, China.
| |
Collapse
|
15
|
Whiteside SK, Grant FM, Alvisi G, Clarke J, Tang L, Imianowski CJ, Zhang B, Evans AC, Wesolowski AJ, Conti AG, Yang J, Lauder SN, Clement M, Humphreys IR, Dooley J, Burton O, Liston A, Alloisio M, Voulaz E, Langhorne J, Okkenhaug K, Lugli E, Roychoudhuri R. Acquisition of suppressive function by conventional T cells limits antitumor immunity upon T reg depletion. Sci Immunol 2023; 8:eabo5558. [PMID: 38100544 PMCID: PMC7615475 DOI: 10.1126/sciimmunol.abo5558] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 01/15/2023] [Accepted: 11/10/2023] [Indexed: 12/17/2023]
Abstract
Regulatory T (Treg) cells contribute to immune homeostasis but suppress immune responses to cancer. Strategies to disrupt Treg cell-mediated cancer immunosuppression have been met with limited clinical success, but the underlying mechanisms for treatment failure are poorly understood. By modeling Treg cell-targeted immunotherapy in mice, we find that CD4+ Foxp3- conventional T (Tconv) cells acquire suppressive function upon depletion of Foxp3+ Treg cells, limiting therapeutic efficacy. Foxp3- Tconv cells within tumors adopt a Treg cell-like transcriptional profile upon ablation of Treg cells and acquire the ability to suppress T cell activation and proliferation ex vivo. Suppressive activity is enriched among CD4+ Tconv cells marked by expression of C-C motif receptor 8 (CCR8), which are found in mouse and human tumors. Upon Treg cell depletion, CCR8+ Tconv cells undergo systemic and intratumoral activation and expansion, and mediate IL-10-dependent suppression of antitumor immunity. Consequently, conditional deletion of Il10 within T cells augments antitumor immunity upon Treg cell depletion in mice, and antibody blockade of IL-10 signaling synergizes with Treg cell depletion to overcome treatment resistance. These findings reveal a secondary layer of immunosuppression by Tconv cells released upon therapeutic Treg cell depletion and suggest that broader consideration of suppressive function within the T cell lineage is required for development of effective Treg cell-targeted therapies.
Collapse
Affiliation(s)
- Sarah K Whiteside
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Francis M Grant
- Immunology Programme, Babraham Institute, Babraham Research Campus, Cambridge, Cambridgeshire CB22 3AT, UK
| | - Giorgia Alvisi
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - James Clarke
- La Jolla Institute for Allergy and Immunology, La Jolla, CA, USA
| | - Leqi Tang
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Charlotte J Imianowski
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Baojie Zhang
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Alexander C Evans
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Alexander J Wesolowski
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Alberto G Conti
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Jie Yang
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Sarah N Lauder
- Division of Infection and Immunity/System Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Mathew Clement
- Division of Infection and Immunity/System Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - Ian R Humphreys
- Division of Infection and Immunity/System Immunity University Research Institute, Cardiff University, Cardiff CF14 4XN, UK
| | - James Dooley
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Oliver Burton
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Adrian Liston
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Marco Alloisio
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Emanuele Voulaz
- Department of Biomedical Sciences, Humanitas University, Via Rita Levi Montalcini 4, 20072 Pieve Emanuele, Milan, Italy
- Division of Thoracic Surgery, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Jean Langhorne
- Francis Crick Institute, 1 Midland Road, London NW1 1AT, UK
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Enrico Lugli
- Laboratory of Translational Immunology, IRCCS Humanitas Research Hospital, Via Manzoni 56, 20089 Rozzano, Milan, Italy
| | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| |
Collapse
|
16
|
Zhu Y, Chang S, Liu J, Wang B. Identification of a novel cuproptosis-related gene signature for multiple myeloma diagnosis. Immun Inflamm Dis 2023; 11:e1058. [PMID: 38018590 PMCID: PMC10629272 DOI: 10.1002/iid3.1058] [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: 04/07/2023] [Revised: 08/19/2023] [Accepted: 10/11/2023] [Indexed: 11/30/2023] Open
Abstract
BACKGROUND Multiple myeloma (MM) ranks second among the most prevalent hematological malignancies. Recent studies have unearthed the promise of cuproptosis as a novel therapeutic intervention for cancer. However, no research has unveiled the particular roles of cuproptosis-related genes (CRGs) in the prediction of MM diagnosis. METHODS Microarray data and clinical characteristics of MM patients were obtained from the Gene Expression Omnibus (GEO) database. Differentially expressed gene analysis, least absolute shrinkage and selection operator (LASSO) and support vector machine-recursive feature elimination (SVM-RFE) algorithms were applied to identify potential signature genes for MM diagnosis. Predictive performance was further assessed by receiver operating characteristic (ROC) curves, nomogram analysis, and external data sets. Functional enrichment analysis was performed to elucidate the involved mechanisms. Finally, the expression of the identified genes was validated by quantitative real-time polymerase chain reaction (qRT-PCR) in MM cell samples. RESULTS The optimal gene signature was identified using LASSO and SVM-RFE algorithms based on the differentially expressed CRGs: ATP7A, FDX1, PDHA1, PDHB, MTF1, CDKN2A, and DLST. Our gene signature-based nomogram revealed a high degree of accuracy in predicting MM diagnosis. ROC curves showed the signature had dependable predictive ability across all data sets, with area under the curve values exceeding 0.80. Additionally, functional enrichment analysis suggested significant associations between the signature genes and immune-related pathways. The expression of the genes was validated in MM cells, indicating the robustness of these findings. CONCLUSION We discovered and validated a novel CRG signature with strong predictive capability for diagnosing MM, potentially implicated in MM pathogenesis and progression through immune-related pathways.
Collapse
Affiliation(s)
- Yidong Zhu
- Department of Traditional Chinese Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Shuaikang Chang
- Department of Hematology, Shanghai East HospitalTongji University School of MedicineShanghaiChina
| | - Jun Liu
- Department of Traditional Chinese Medicine, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Bo Wang
- Department of Endocrinology, Yangpu HospitalTongji University School of MedicineShanghaiChina
| |
Collapse
|
17
|
Varisli L, Dancik GM, Tolan V, Vlahopoulos S. Critical Roles of SRC-3 in the Development and Progression of Breast Cancer, Rendering It a Prospective Clinical Target. Cancers (Basel) 2023; 15:5242. [PMID: 37958417 PMCID: PMC10648290 DOI: 10.3390/cancers15215242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 10/27/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Breast cancer (BCa) is the most frequently diagnosed malignant tumor in women and is also one of the leading causes of cancer-related death. Most breast tumors are hormone-dependent and estrogen signaling plays a critical role in promoting the survival and malignant behaviors of these cells. Estrogen signaling involves ligand-activated cytoplasmic estrogen receptors that translocate to the nucleus with various co-regulators, such as steroid receptor co-activator (SRC) family members, and bind to the promoters of target genes and regulate their expression. SRC-3 is a member of this family that interacts with, and enhances, the transcriptional activity of the ligand activated estrogen receptor. Although SRC-3 has important roles in normal homeostasis and developmental processes, it has been shown to be amplified and overexpressed in breast cancer and to promote malignancy. The malignancy-promoting potential of SRC-3 is diverse and involves both promoting malignant behavior of tumor cells and creating a tumor microenvironment that has an immunosuppressive phenotype. SRC-3 also inhibits the recruitment of tumor-infiltrating lymphocytes with effector function and promotes stemness. Furthermore, SRC-3 is also involved in the development of resistance to hormone therapy and immunotherapy during breast cancer treatment. The versatility of SRC-3 in promoting breast cancer malignancy in this way makes it a good target, and methodical targeting of SRC-3 probably will be important for the success of breast cancer treatment.
Collapse
Affiliation(s)
- Lokman Varisli
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Garrett M. Dancik
- Department of Computer Science, Eastern Connecticut State University, Willimantic, CT 06226, USA;
| | - Veysel Tolan
- Department of Molecular Biology and Genetics, Science Faculty, Dicle University, Diyarbakir 21280, Turkey;
| | - Spiros Vlahopoulos
- First Department of Pediatrics, National and Kapodistrian University of Athens, Thivon & Levadeias 8, Goudi, 11527 Athens, Greece
| |
Collapse
|
18
|
Wang L, Zhang W, Zhang J, Zheng M, Pan X, Guo H, Ding L. Inhibitory effect of adenosine on adaptive antitumor immunity and intervention strategies. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:567-577. [PMID: 37916308 PMCID: PMC10630057 DOI: 10.3724/zdxbyxb-2023-0263] [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: 06/02/2023] [Accepted: 09/11/2023] [Indexed: 10/08/2023]
Abstract
Tumors in which the microenvironment is characterized by lack of immune cell infiltration are referred as "cold tumors" and typically exhibit low responsiveness to immune therapy. Targeting the factors contributing to "cold tumors" formation and converting them into "hot tumors" is a novel strategy for improving the efficacy of immunotherapy. Adenosine, a hydrolysis product of ATP, accumulates with a significantly higher concentration in the tumor microenvironments compared with normal tissue and exerts inhibitory effects on tumor-specific adaptive immunity. Tumor cells, dendritic cells, macrophages, and T cells express abundant adenosine receptors on their surfaces. The binding of adenosine to these receptors initiates downstream signaling pathways that suppress tumor antigen presentation and immune cell activation, consequently dampening adaptive immune responses against tumors. Adenosine down-regulates the expression of major histocompatibility complex Ⅱ and co-stimulatory factors on dendritic cells and macrophages, thereby inhibiting antigen presentation to T cells. Adenosine also inhibits ligand-receptor binding and transmembrane signaling on T cells, concomitantly suppressing the secretion of anti-tumor cytokines and impairing T cell activation. Furthermore, adenosine hinders effector T cell trafficking to tumor sites and infiltration by inhibiting chemokine secretion and KCa3.1 channels. Additionally, adenosine promotes the secretion of immunosuppressive cytokines, increases immune checkpoint protein expression, and enhances the activity of immunosuppressive cells, collectively curbing cytotoxic T cell-mediated tumor cell killing. Given the immunosuppressive role of adenosine in adaptive antitumor immunity, several inhibitors targeting adenosine generation or adenosine receptor blockade are currently in preclinical or clinical development with the aim of enhancing the effectiveness of immunotherapies. This review provides an overview of the inhibitory effects of adenosine on adaptive antitumor immunity, elucidate the molecular mechanisms involved, and summarizes the latest advances in application of adenosine inhibition strategies for antitumor immunotherapy.
Collapse
Affiliation(s)
- Longsheng Wang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| | - Wenxin Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Jie Zhang
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Mingming Zheng
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Xiaohui Pan
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Hongjie Guo
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China
| | - Ling Ding
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
19
|
Shan Y, Zhang B, Chen L, Zhang H, Jiang C, You Q, Li Y, Han H, Zhu J. Herpesvirus entry mediator regulates the transduction of Tregs via STAT5/Foxp3 signaling pathway in ovarian cancer cells. Anticancer Drugs 2023; 34:73-80. [PMID: 35946515 DOI: 10.1097/cad.0000000000001336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
The ratio of regulatory T cells (Treg) in peripheral blood of cancer patients has a closely correlation to the occurrence and development of ovarian cancer. In this study, our aim to explore the expression of herpesvirus entry mediator (HVEM) in ovarian cancer and its correlation with Tregs. The expression of HVEM in peripheral blood of ovarian cancer patients was detected by ELISA, and the ratio of CD4+ CD25 + Foxp3 positive Tregs cells was detected by flow cytometry. Ovarian cancer cell lines with high- and low-HVEM expression were constructed. CD4+ cells were co-cultured with ovarian cancer (OC) cells, and the expressions of IL-2 and TGF-β1 in the supernatant of cells were detected by ELISA, and western blot was used to detect the expressions of STAT5, p-STAT5, and Foxp3. The results indicated that the number of Treg cells in the peripheral blood of OC patients increased, and the expression of HVEM increased, the two have a certain correlation. At the same time, the overexpression of HVEM promoted the expression of cytokines IL-2 and TGF- β1, promoted the activation of STAT5 and the expression of Foxp3, leading to an increase in the positive rate of Treg, while the HVEM gene silence group was just the opposite. Our results showed that the expression of HVEM in OC cells has a positive regulation effect on Tregs through the STAT5/Foxp3 signaling pathway. To provide experimental basis and related mechanism for the clinical treatment of ovarian cancer.
Collapse
Affiliation(s)
- Ying Shan
- Department of Obstetrics and Gynecology
| | | | - Li Chen
- Department of Obstetrics and Gynecology
| | - Hu Zhang
- Department of Obstetrics and Gynecology
| | - Cui Jiang
- Department of Obstetrics and Gynecology
| | - Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Yanyi Li
- Department of Obstetrics and Gynecology
- Department of Health Science, Graduate School of Medical, Osaka University, Osaka, Japan
| | | | | |
Collapse
|
20
|
Zhao X, Wang S, Wang S, Xie J, Cui D. mTOR signaling: A pivotal player in Treg cell dysfunction in systemic lupus erythematosus. Clin Immunol 2022; 245:109153. [DOI: 10.1016/j.clim.2022.109153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/30/2022] [Accepted: 10/04/2022] [Indexed: 11/03/2022]
|
21
|
Yang D, Liu J, Liu N, Yin C, Zhang H, Xu J. The prognostic value of tumor mutational burden related 6-gene-based Risk Score in laryngeal cancer patients. BMC Oral Health 2022; 22:510. [DOI: 10.1186/s12903-022-02534-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/25/2022] [Indexed: 11/18/2022] Open
Abstract
Abstract
Background
Laryngeal cancer (LC) is the second frequent malignant head and neck cancer around world, while LC patients’ prognosis is unsatisfactory. This study aims to investigate the prognostic value of tumor mutation burden (TMB)-related genes in LC.
Methods
LC data was downloaded from The Cancer Genome Atlas and Gene Expression Omnibus databases. TMB values of all samples were calculated basing on mutation data. The differentially expressed genes (DEGs) between LC samples with distinct TMB were subjected to univariate and LASSO Cox regression analysis to build Risk Score. Immune cell infiltration analysis was conducted in CIBERSORT.
Results
Between high and low TMB LC samples, we identified 210 DEGs. Of which, six optimal genes were included to construct Risk Score, comprising FOXJ1, EPO, FGF5, SPOCK1, KCNF1 and PSG5. High risk LC patients had significantly poorer overall survival than low risk patients. The nomogram model constructed basing on Risk Score and gender showed good performance in predicting LC patients’ survival probability.
Conclusions
The prognostic Risk Score model, basing on six TMB-related genes (FOXJ1, EPO, FGF5, SPOCK1, KCNF1 and PSG5), was a reliable prognostic model to separate LC patients with different prognoses.
Collapse
|
22
|
Yin J, He X, Qin F, Zheng S, Huang Y, Hu L, Chen Y, Zhong L, Hu W, Li S. m 6A-related lncRNA signature for predicting prognosis and immune response in head and neck squamous cell carcinoma. Am J Transl Res 2022; 14:7653-7669. [PMID: 36505334 PMCID: PMC9730119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Accepted: 09/27/2022] [Indexed: 12/15/2022]
Abstract
OBJECTIVES N6-methyladenosine (m6A) and long non-coding RNAs (lncRNAs) significantly impact the prognosis and the response to immunotherapy in head and neck squamous cell carcinoma (HNSCC). Therefore, this study aimed to develop an m6A-related lncRNA (m6AlncRNA) model for predicting the prognosis and the immunotherapeutic response in HNSCC. METHODS We identified the m6AlncRNAs and constructed a risk assessment signature by using univariable Cox, Least Absolute Shrinkage and Selection Operator (LASSO), and multivariate Cox regression analyses. The Kaplan-Meier analysis, receiver-operating characteristic (ROC) curves, principal component analysis (PCA), decision curve analysis (DCA), consistency index (C-index), and nomogram were applied to assess the risk model. Finally, we investigated the predictability of this model in prognosis and response to immunotherapy and evaluated various novel compounds for the clinical treatment of HNSCC. RESULTS HNSCC patients were assigned to high- and low-risk groups based on the median risk scores, and the high- and low-risk groups had different clinical features, tumor immune infiltration status, tumor immune dysfunction and exclusion (TIDE), tumor mutational burden (TMB), sensitivity to novel potential compounds, and immunotherapeutic response. CONCLUSIONS The model we developed was accurate and efficient in predicting the prognosis of patients with HNSCC. It was also sensitive in stratifying HNSCC patients with good response to immunotherapy. Therefore, our study provided insight into elucidating the processes and mechanisms of m6AlncRNAs.
Collapse
|
23
|
Guizhen Z, Guanchang J, Liwen L, Huifen W, Zhigang R, Ranran S, Zujiang Y. The tumor microenvironment of hepatocellular carcinoma and its targeting strategy by CAR-T cell immunotherapy. Front Endocrinol (Lausanne) 2022; 13:918869. [PMID: 36093115 PMCID: PMC9452721 DOI: 10.3389/fendo.2022.918869] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 08/05/2022] [Indexed: 12/16/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the major subtype of liver cancer, which ranks sixth in cancer incidence and third in mortality. Although great strides have been made in novel therapy for HCC, such as immunotherapy, the prognosis remains less than satisfactory. Increasing evidence demonstrates that the tumor immune microenvironment (TME) exerts a significant role in the evolution of HCC and has a non-negligible impact on the efficacy of HCC treatment. In the past two decades, the success in hematological malignancies made by chimeric antigen receptor-modified T (CAR-T) cell therapy leveraging it holds great promise for cancer treatment. However, in the face of a hostile TME in solid tumors like HCC, the efficacy of CAR-T cells will be greatly compromised. Here, we provide an overview of TME features in HCC, discuss recent advances and challenges of CAR-T immunotherapy in HCC.
Collapse
Affiliation(s)
- Zhang Guizhen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Presion Medicine Cencter Gene Hospital of Henan Province, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ji Guanchang
- Department of Urology People’s Hospital of Puyang, Puyang, China
| | - Liu Liwen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Presion Medicine Cencter Gene Hospital of Henan Province, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Wang Huifen
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Presion Medicine Cencter Gene Hospital of Henan Province, Zhengzhou, China
- Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Ren Zhigang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Sun Ranran
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yu Zujiang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Presion Medicine Cencter Gene Hospital of Henan Province, Zhengzhou, China
| |
Collapse
|
24
|
Alrumaihi F. The Multi-Functional Roles of CCR7 in Human Immunology and as a Promising Therapeutic Target for Cancer Therapeutics. Front Mol Biosci 2022; 9:834149. [PMID: 35874608 PMCID: PMC9298655 DOI: 10.3389/fmolb.2022.834149] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 05/26/2022] [Indexed: 11/13/2022] Open
Abstract
An important hallmark of the human immune system is to provide adaptive immunity against pathogens but tolerance toward self-antigens. The CC-chemokine receptor 7 (CCR7) provides a significant contribution in guiding cells to and within lymphoid organs and is important for acquiring immunity and tolerance. The CCR7 holds great importance in establishing thymic architecture and function and naïve and regulatory T-cell homing in the lymph nodes. Similarly, the receptor is a key regulator in cancer cell migration and the movement of dendritic cells. This makes the CCR7 an important receptor as a drug and prognostic marker. In this review, we discussed several biological roles of the CCR7 and its importance as a drug and prognostic marker.
Collapse
Affiliation(s)
- Faris Alrumaihi
- Department of Medical Laboratories, College of Applied Medical Sciences, Qassim University, Buraydah, Saudi Arabia
| |
Collapse
|
25
|
Fu MS, Pan SX, Cai XQ, Hu YX, Zhang WJ, Pan QC. Analysis of ARHGAP4 Expression With Colorectal Cancer Clinical Characteristics and Prognosis. Front Oncol 2022; 12:899837. [PMID: 35847897 PMCID: PMC9278087 DOI: 10.3389/fonc.2022.899837] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2022] [Accepted: 05/23/2022] [Indexed: 11/21/2022] Open
Abstract
Background This study aims to analyze the correlation between ARHGAP4 in the expression and clinical characteristics of colorectal cancer (CRC), and the influence of ARHGAP4 expression on the prognosis of CRC, and to evaluate whether ARHGAP4 is a potential prognostic oncotarget for CRC. Methods ARHGAP4 was identified using the Gene Expression Omnibus database through weighted gene coexpression network analysis. Using the Gene Expression Profiling Interactive Analysis to perform and analyze the expression and prognosis of ARHGAP4 in CRC. The expression of AGRGAP4 and immune cells was analyzed by the Tumor IMmune Estimation Resource online database. Finally, immunohistochemistry was used to analyze the expression difference and prognosis of ARHGAP4 in CRC and adjacent normal tissues, as well as the relationship between AGRGAP4 expression and clinical features of CRC. Results We identified ARHGAP4 that is related to the recurrence of CRC from GSE97781 data. ARHGAP4 has not been reported in CRC. The high expression of ARHGAP4 in select colon adenocarcinoma indicates a poor prognosis by database analysis. In our clinical data results, ARHGAP4 is highly expressed in CRC and lowly expressed in normal tissues adjacent to cancer. Compared with the low-expression group, the high-expression group has a significantly poorer prognosis. In colon cancer, the B-cell, macrophage, neutrophil, and dendritic-cell levels are downregulated after ARHGAP4 gene knockout; the levels of CD8+ and CD4+ T cells, neutrophils, and dendritic cells are upregulated after the amplification of the ARHGAP4 gene. In addition, ARHGAP4 expression is related to N,M staging and clinical staging. Conclusion ARHGAP4 is highly expressed in CRC, and the high expression of ARHGAP4 has a poor prognosis. The expression of ARHGAP4 in CRC is related to the immune cells such as B cells, CD8+ and CD4+ T cells, macrophages, neutrophils, and dendritic cells. ARHGAP4 is correlated with N,M staging and clinical staging in CRC. ARHGAP4 may be a potential biomarker for the prognosis of CRC.
Collapse
Affiliation(s)
- Ming-sheng Fu
- Department of Gastroenterology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Shu-xian Pan
- Department of Nephrology of Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Xun-quan Cai
- Department of Gastroenterology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Yuan-xin Hu
- Department of Gastroenterology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Wei-jie Zhang
- Department of Gastroenterology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| | - Qin-cong Pan
- Department of Gastroenterology, Shanghai Fifth People’s Hospital, Fudan University, Shanghai, China
| |
Collapse
|
26
|
Li Z, Tuong ZK, Dean I, Willis C, Gaspal F, Fiancette R, Idris S, Kennedy B, Ferdinand JR, Peñalver A, Cabantous M, Murtuza Baker S, Fry JW, Carlesso G, Hammond SA, Dovedi SJ, Hepworth MR, Clatworthy MR, Withers DR. In vivo labeling reveals continuous trafficking of TCF-1+ T cells between tumor and lymphoid tissue. J Exp Med 2022; 219:e20210749. [PMID: 35472220 PMCID: PMC9048291 DOI: 10.1084/jem.20210749] [Citation(s) in RCA: 67] [Impact Index Per Article: 22.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 01/14/2022] [Accepted: 04/06/2022] [Indexed: 12/12/2022] Open
Abstract
Improving the efficacy of immune checkpoint therapies will require a better understanding of how immune cells are recruited and sustained in tumors. Here, we used the photoconversion of the tumor immune cell compartment to identify newly entering lymphocytes, determine how they change over time, and investigate their egress from the tumor. Combining single-cell transcriptomics and flow cytometry, we found that while a diverse mix of CD8 T cell subsets enter the tumor, all CD8 T cells retained within this environment for more than 72 h developed an exhausted phenotype, revealing the rapid establishment of this program. Rather than forming tumor-resident populations, non-effector subsets, which express TCF-1 and include memory and stem-like cells, were continuously recruited into the tumor, but this recruitment was balanced by concurrent egress to the tumor-draining lymph node. Thus, the TCF-1+ CD8 T cell niche in tumors is highly dynamic, with the circulation of cells between the tumor and peripheral lymphoid tissue to bridge systemic and intratumoral responses.
Collapse
Affiliation(s)
- Zhi Li
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Zewen K. Tuong
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cellular Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - Isaac Dean
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Claire Willis
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Fabrina Gaspal
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Rémi Fiancette
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Suaad Idris
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Bethany Kennedy
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - John R. Ferdinand
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Ana Peñalver
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Mia Cabantous
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
| | - Syed Murtuza Baker
- Division of Informatics, Imaging & Data Science, Faculty of Biology, Medicine and Health, the University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Jeremy W. Fry
- ProImmune Ltd., The Magdalen Centre, Oxford Science Park, Oxford, UK
| | | | | | | | - Matthew R. Hepworth
- Lydia Becker Institute of Immunology and Inflammation, Division of Infection, Immunity and Respiratory Medicine, Faculty of Biology, Medicine and Health, the University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Menna R. Clatworthy
- Molecular Immunity Unit, Department of Medicine, University of Cambridge, Cambridge, UK
- Cellular Genetics, Wellcome Trust Sanger Institute, Hinxton, UK
| | - David R. Withers
- Institute of Immunology and Immunotherapy, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| |
Collapse
|
27
|
Yao X, Qi X, Wang Y, Zhang B, He T, Yan T, Zhang L, Wang Y, Zheng H, Zhang G, Guo X. Identification and Validation of an Annexin-Related Prognostic Signature and Therapeutic Targets for Bladder Cancer: Integrative Analysis. BIOLOGY 2022; 11:biology11020259. [PMID: 35205125 PMCID: PMC8869209 DOI: 10.3390/biology11020259] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/03/2022] [Accepted: 02/03/2022] [Indexed: 12/12/2022]
Abstract
Abnormal expression and dysfunction of Annexins (ANXA1-11, 13) have been widely found in several types of cancer. However, the expression pattern and prognostic value of Annexins in bladder cancer (BC) are currently still unknown. In this study, survival analysis by our in-house OSblca web server revealed that high ANXA1/2/3/5/6 expression was significantly associated with poor overall survival (OS) in BC patients, while higher ANXA11 was associated with increased OS. Through Oncomine and GEPIA2 database analysis, we found that ANXA2/3/4/13 were up-regulated, whereas ANXA1/5/6 were down-regulated in BC compared with normal bladder tissues. Further LASSO analysis built an Annexin-Related Prognostic Signature (ARPS, including four members ANXA1/5/6/10) in the TCGA BC cohort and validated it in three independent GEO BC cohorts (GSE31684, GSE32548, GSE48075). Multivariate COX analysis demonstrated that ARPS is an independent prognostic signature for BC. Moreover, GSEA results showed that immune-related pathways, such as epithelial-mesenchymal transition and IL6/JAK/STAT3 signaling were enriched in the high ARPS risk groups, while the low ARPS risk group mainly regulated metabolism-related processes, such as adipogenesis and bile acid metabolism. In conclusion, our study comprehensively analyzed the mRNA expression and prognosis of Annexin family members in BC, constructed an Annexin-related prognostic signature using LASSO and COX regression, and validated it in four independent BC cohorts, which might help to improve clinical outcomes of BC patients, offer insights into the underlying molecular mechanisms of BC development and suggest potential therapeutic targets for BC.
Collapse
Affiliation(s)
- Xitong Yao
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Xinlei Qi
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Yao Wang
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Baokun Zhang
- Beijing Key Laboratory of New Molecular Diagnosis Technologies for Infectious Diseases, Department of Biotechnology, Beijing Institute of Radiation Medicine, Beijing 100850, China;
| | - Tianshuai He
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Taoning Yan
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Lu Zhang
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Yange Wang
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Hong Zheng
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
| | - Guosen Zhang
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
- Correspondence: or (G.Z.); (X.G.); Tel.: +86-18237808750 (G.Z.)
| | - Xiangqian Guo
- Cell Signal Transduction Laboratory, Department of Predictive Medicine, Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Academy for Advanced Interdisciplinary Studies, Henan University, Kaifeng 475004, China; (X.Y.); (X.Q.); (Y.W.); (T.H.); (T.Y.); (L.Z.); (Y.W.); (H.Z.)
- Correspondence: or (G.Z.); (X.G.); Tel.: +86-18237808750 (G.Z.)
| |
Collapse
|
28
|
Chen Y, Zheng X, Wu C. The Role of the Tumor Microenvironment and Treatment Strategies in Colorectal Cancer. Front Immunol 2021; 12:792691. [PMID: 34925375 PMCID: PMC8674693 DOI: 10.3389/fimmu.2021.792691] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 11/15/2021] [Indexed: 12/17/2022] Open
Abstract
Colorectal cancer (CRC) has the second highest mortality rate among all cancers worldwide. Surgery, chemotherapy, radiotherapy, molecular targeting and other treatment methods have significantly prolonged the survival of patients with CRC. Recently, the emergence of tumor immunotherapy represented by immune checkpoint inhibitors (ICIs) has brought new immunotherapy options for the treatment of advanced CRC. As the efficacy of ICIs is closely related to the tumor immune microenvironment (TME), it is necessary to clarify the relationship between the immune microenvironment of CRC and the efficacy of immunotherapy to ensure that the appropriate drugs are selected. We herein review the latest research progress in the immune microenvironment and strategies related to immunotherapy for CRC. We hope that this review helps in the selection of appropriate treatment strategies for CRC patients.
Collapse
Affiliation(s)
- Yaping Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
29
|
Braun C, Weichhart T. mTOR-dependent immunometabolism as Achilles' heel of anticancer therapy. Eur J Immunol 2021; 51:3161-3175. [PMID: 34648202 DOI: 10.1002/eji.202149270] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Revised: 07/07/2021] [Accepted: 10/06/2021] [Indexed: 12/14/2022]
Abstract
Immune cells are important constituents of the tumor microenvironment and essential in eradicating tumor cells during conventional therapies or novel immunotherapies. The mechanistic target of rapamycin (mTOR) signaling pathway senses the intra- and extracellular nutrient status, growth factor supply, and cell stress-related changes to coordinate cellular metabolism and activation dictating effector and memory functions in mainly all hematopoietic immune cells. In addition, the mTOR complex 1 (mTORC1) and mTORC2 are frequently deregulated and become activated in cancer cells to drive cell transformation, survival, neovascularization, and invasion. In this review, we provide an overview of the influence of mTOR complexes on immune and cancer cell function and metabolism. We discuss how mTOR inhibitors aiming to target cancer cells will influence immunometabolic cell functions participating either in antitumor responses or favoring tumor cell progression in individual immune cells. We suggest immunometabolism as the weak spot of anticancer therapy and propose to evaluate patients according to their predominant immune cell subtype in the cancer tissue. Advances in metabolic drug development that hold promise for more effective treatments in different types of cancer will have to consider their effects on the immune system.
Collapse
Affiliation(s)
- Clarissa Braun
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria.,Clinical Division of Endocrinology and Metabolism, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
| | - Thomas Weichhart
- Center of Pathobiochemistry and Genetics, Institute of Medical Genetics, Medical University of Vienna, Vienna, Austria
| |
Collapse
|
30
|
Feng P, Yang Q, Luo L, Sun Y, Lv W, Wan S, Guan Z, Xiao Z, Liu F, Li Z, Dong Z, Yang M. The kinase PDK1 regulates regulatory T cell survival via controlling redox homeostasis. Theranostics 2021; 11:9503-9518. [PMID: 34646383 PMCID: PMC8490516 DOI: 10.7150/thno.63992] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2021] [Accepted: 08/29/2021] [Indexed: 12/17/2022] Open
Abstract
Rationale: Regulatory T cells (Treg cells) play an important role in maintaining peripheral tolerance by suppressing over-activation of effector T cells. The kinase PDK1 plays a pivotal role in conventional T cell development. However, whether PDK1 signaling affects the homeostasis and function of Treg cells remains elusive. Methods: In order to evaluate the role of PDK1 in Treg cells from a genetic perspective, mice carrying the floxed PDK1 allele were crossbred with Foxp3Cre mice to efficiently deleted PDK1 in Foxp3+ Treg cells. Flow cytometry was used to detect the immune cell homeostasis of WT and PDK1fl/flFoxp3Cre mice. RNA-seq was used to assess the differences in transcriptional expression profile of WT and PDK1-deficient Treg cells. The metabolic profiles of WT and PDK1-deficient Treg cells were tested using the Glycolysis Stress Test and Mito Stress Test Kits by the Seahorse XFe96 Analyser. Results: PDK1 was essential for the establishment and maintenance of Treg cell homeostasis and function. Disruption of PDK1 in Treg cells led to a spontaneous fatal systemic autoimmune disorder and multi-tissue inflammatory damage, accompanied by a reduction in the number and function of Treg cells. The deletion of PDK1 in Treg cells destroyed the iron ion balance through regulating MEK-ERK signaling and CD71 expression, resulting in excessive production of intracellular ROS, which did not depend on the down-regulation of mTORC1 signaling. Inhibition of excessive ROS, activated MEK-Erk signaling or overload Fe2+ could partially rescue the survival of PDK1-deficient Treg cells. Conclusion: Our results defined a key finding on the mechanism by which PDK1 regulates Treg cell survival via controlling redox homeostasis.
Collapse
Affiliation(s)
- Peiran Feng
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Quanli Yang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Liang Luo
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Yadong Sun
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Wenkai Lv
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Shuo Wan
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zerong Guan
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zhiqiang Xiao
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Feng Liu
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Zehua Li
- School of Medicine and Institute for Immunology, Tsinghua University, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, 100084, China
| | - Zhongjun Dong
- School of Medicine and Institute for Immunology, Tsinghua University, Beijing Key Lab for Immunological Research on Chronic Diseases, Tsinghua University, Beijing, 100084, China
| | - Meixiang Yang
- Zhuhai Institute of Translational Medicine, Zhuhai People's Hospital (Zhuhai Hospital Affiliated with Jinan University), Jinan University, Zhuhai, Guangdong, 519000, China
- The Biomedical Translational Research Institute, Faculty of Medical Science, Jinan University, Guangzhou, Guangdong, 510632, China
| |
Collapse
|
31
|
Whiteside SK, Grant FM, Gyori DS, Conti AG, Imianowski CJ, Kuo P, Nasrallah R, Sadiyah F, Lira SA, Tacke F, Eil RL, Burton OT, Dooley J, Liston A, Okkenhaug K, Yang J, Roychoudhuri R. CCR8 marks highly suppressive Treg cells within tumours but is dispensable for their accumulation and suppressive function. Immunol Suppl 2021; 163:512-520. [PMID: 33838058 PMCID: PMC8274197 DOI: 10.1111/imm.13337] [Citation(s) in RCA: 52] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 02/02/2023]
Abstract
CD4+ regulatory T (Treg) cells, dependent upon the transcription factor Foxp3, contribute to tumour immunosuppression but are also required for immune homeostasis. There is interest in developing therapies that selectively target the immunosuppressive function of Treg cells within tumours without disrupting their systemic anti-inflammatory function. High levels of expression of chemokine (C-C motif) receptor 8 (CCR8) discriminate Treg cells within tumours from those found in systemic lymphoid tissues. It has recently been proposed that disruption of CCR8 function using blocking anti-CCR8 antibodies results in reduced accumulation of Treg cells within tumours and disruption of their immunosuppressive function. Here, using Ccr8-/- mice, we show that CCR8 function is not required for Treg cell accumulation or immunosuppression in the context of syngeneic MC38 colorectal adenocarcinoma and B16 melanoma tumours. We observed high levels of CCR8 expression on tumour-infiltrating Treg cells which were abolished in Ccr8-/- mice. High levels of CCR8 marked cells with high levels of suppressive function. However, whereas systemic ablation of Treg cells resulted in strikingly diminished tumour burden, growth of subcutaneously implanted tumours was unaffected by systemic CCR8 loss. Consistently, we observed minimal impact of systemic CCR8 ablation on the frequency, phenotype and function of tumour-infiltrating Treg cells and conventional T (Tconv) function. These findings suggest that CCR8 is not required for Treg cell accumulation and immunosuppressive function within tumours and that depletion of CCR8+ Treg cells rather than blockade of CCR8 function is a more promising avenue for selective immunotherapy.
Collapse
Affiliation(s)
- Sarah K. Whiteside
- Department of PathologyUniversity of CambridgeCambridgeUK,Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Francis M. Grant
- Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - David S. Gyori
- Department of PhysiologySemmelweis UniversityBudapestHungary
| | | | - Charlotte J. Imianowski
- Department of PathologyUniversity of CambridgeCambridgeUK,Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Paula Kuo
- Department of PathologyUniversity of CambridgeCambridgeUK,Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Rabab Nasrallah
- Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Firas Sadiyah
- Department of PathologyUniversity of CambridgeCambridgeUK,Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Sergio A. Lira
- Mount Sinai School of MedicineImmunology InstituteNew YorkNYUSA
| | - Frank Tacke
- Department of Hepatology & GastroenterologyCampus Virchow‐Klinikum (CVK) and Campus Charité Mitte (CCM)Charité Universitätsmedizin BerlinBerlinGermany
| | - Robert L. Eil
- Department of SurgeryMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Oliver T. Burton
- Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - James Dooley
- Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Adrian Liston
- Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | | | - Jie Yang
- Department of PathologyUniversity of CambridgeCambridgeUK,Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| | - Rahul Roychoudhuri
- Department of PathologyUniversity of CambridgeCambridgeUK,Immunology ProgrammeBabraham Research CampusBabraham InstituteCambridgeUK
| |
Collapse
|
32
|
Miller MO, Kashyap PC, Becker SL, Thomas RM, Hodin RA, Miller G, Hundeyin M, Pushalkar S, Cohen D, Saxena D, Shogan BD, Morris-Stiff GJ. SSAT State-of-the-Art Conference: Advancements in the Microbiome. J Gastrointest Surg 2021; 25:1885-1895. [PMID: 32989690 DOI: 10.1007/s11605-020-04551-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The microbiome plays a major role in human physiology by influencing obesity, inducing inflammation, and impacting cancer therapies. During the 60th Annual Meeting of the Society of the Alimentary Tract (SSAT) at the State-of-the-Art Conference, experts in the field discussed the influence of the microbiome. This paper is a summary of the influence of the microbiome on obesity, inflammatory bowel disease, pancreatic cancer, cancer therapies, and gastrointestinal optimization. This review shows how the microbiome plays an important role in the development of diseases and surgical complications. Future studies are needed in targeting the gut microbiome to develop individualized therapies.
Collapse
Affiliation(s)
- Miquell O Miller
- Department of General Surgery, Stanford University, 300 Pasteur Dr, Stanford, CA, 94305, USA.
| | - Purna C Kashyap
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Sarah L Becker
- Department of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Ryan M Thomas
- Departments of Surgery, Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, 32610, USA
| | - Richard A Hodin
- Department of Surgery, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - George Miller
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Mautin Hundeyin
- Departments of Surgery and Cell Biology, New York University School of Medicine, New York, NY, 10016, USA
| | - Smruti Pushalkar
- Department of Medicine, New York University School of Medicine, New York, NY, 10016, USA
| | - Deirdre Cohen
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Deepak Saxena
- Department of Basic Science and Craniofacial Biology, New York University College of Dentistry, New York, NY, 10010, USA
| | - Benjamin D Shogan
- Department of Surgery, University of Chicago, Chicago, IL, 60637, USA
| | | |
Collapse
|
33
|
Coexpression of Helios in Foxp3 + Regulatory T Cells and Its Role in Human Disease. DISEASE MARKERS 2021; 2021:5574472. [PMID: 34257746 PMCID: PMC8245237 DOI: 10.1155/2021/5574472] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 06/15/2021] [Indexed: 12/03/2022]
Abstract
Regulatory T cells (Tregs) expressing the Foxp3 transcription factor are indispensable for the maintenance of immune system homeostasis. Tregs may lose Foxp3 expression or be reprogrammed into cells that produce proinflammatory cytokines, for example, Th1-like Tregs, Th2-like Tregs, Th17-like Tregs, and Tfh-like Tregs. Accordingly, selective therapeutic molecules that manipulate Treg lineage stability and/or functional activity might have the potential to improve aberrant immune responses in human disorders. In particular, the transcription factor Helios has emerged as an important marker and modulator of Tregs. Therefore, the current review focuses on recent findings on the expression, function, and mechanisms of Helios, as well as the patterns of Foxp3+ Tregs coexpressing Helios in various human disorders, in order to explore the potential of Helios for the improvement of many immune-related diseases. The studies were selected from PubMed using the library of the Nanjing Medical University in this review. The findings of the included studies indicate that Helios expression stabilizes the phenotype and function of Foxp3+ Tregs in certain inflammatory environments. Further, Tregs coexpressing Helios and Foxp3 were identified as a specific phenotype of stronger suppressor immune cells in both humans and animal models. Importantly, there is ample evidence that Helios-expressing Foxp3+ Tregs are relevant to various human disorders, including connective tissue diseases, infectious diseases, solid organ transplantation-related immunity, and cancer. Thus, Helios+Foxp3+CD4+ Tregs could be a valuable target in human diseases, and their potential should be explored further in the clinical setting.
Collapse
|
34
|
Troulé K, López-Fernández H, García-Martín S, Reboiro-Jato M, Carretero-Puche C, Martorell-Marugán J, Martín-Serrano G, Carmona-Sáez P, Glez-Peña D, Al-Shahrour F, Gómez-López G. DREIMT: a drug repositioning database and prioritization tool for immunomodulation. Bioinformatics 2021; 37:578-579. [PMID: 32818254 DOI: 10.1093/bioinformatics/btaa727] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 07/22/2020] [Accepted: 08/16/2019] [Indexed: 11/13/2022] Open
Abstract
MOTIVATION Drug immunomodulation modifies the response of the immune system and can be therapeutically exploited in pathologies such as cancer and autoimmune diseases. RESULTS DREIMT is a new hypothesis-generation web tool, which performs drug prioritization analysis for immunomodulation. DREIMT provides significant immunomodulatory drugs targeting up to 70 immune cells subtypes through a curated database that integrates 4960 drug profiles and ∼2600 immune gene expression signatures. The tool also suggests potential immunomodulatory drugs targeting user-supplied gene expression signatures. Final output includes drug-signature association scores, FDRs and downloadable plots and results tables. AVAILABILITYAND IMPLEMENTATION http://www.dreimt.org. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Kevin Troulé
- Bioinformatics Unit, Structural Biology Department, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Hugo López-Fernández
- SING Research Group, Biomedical Research Centre (CINBIO), E-36310, Vigo, Spain.,ESEI - Escuela Superior de Ingeniería Informática, Department of Computer Science, Universidad de Vigo, E-32004, Ourense, Spain
| | - Santiago García-Martín
- Bioinformatics Unit, Structural Biology Department, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Miguel Reboiro-Jato
- SING Research Group, Biomedical Research Centre (CINBIO), E-36310, Vigo, Spain.,ESEI - Escuela Superior de Ingeniería Informática, Department of Computer Science, Universidad de Vigo, E-32004, Ourense, Spain
| | - Carlos Carretero-Puche
- Bioinformatics Unit, Structural Biology Department, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Jordi Martorell-Marugán
- Bioinformatics Unit, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
| | - Guillermo Martín-Serrano
- Bioinformatics Unit, Structural Biology Department, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Pedro Carmona-Sáez
- Bioinformatics Unit, GENYO, Centre for Genomics and Oncological Research, Pfizer/University of Granada/Andalusian Regional Government, PTS Granada, E-18016 Granada, Spain
| | - Daniel Glez-Peña
- SING Research Group, Biomedical Research Centre (CINBIO), E-36310, Vigo, Spain.,ESEI - Escuela Superior de Ingeniería Informática, Department of Computer Science, Universidad de Vigo, E-32004, Ourense, Spain
| | - Fátima Al-Shahrour
- Bioinformatics Unit, Structural Biology Department, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| | - Gonzalo Gómez-López
- Bioinformatics Unit, Structural Biology Department, Spanish National Cancer Research Centre (CNIO), Madrid 28029, Spain
| |
Collapse
|
35
|
Immuno-profiling and cellular spatial analysis using five immune oncology multiplex immunofluorescence panels for paraffin tumor tissue. Sci Rep 2021; 11:8511. [PMID: 33875760 PMCID: PMC8055659 DOI: 10.1038/s41598-021-88156-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/05/2021] [Indexed: 02/07/2023] Open
Abstract
Multiplex immunofluorescence (mIF) has arisen as an important tool for immuno-profiling tumor tissues. We updated our manual protocol into an automated protocol that allows the use of up to seven markers in five mIF panels to apply to formalin-fixed paraffin-embedded tumor tissues. Using a tyramide signal amplification system, we optimized five mIF panels that included cytokeratin to characterize malignant cells (MCs), immune checkpoint markers (i.e., PD-L1, B7-H3, B7-H4, IDO-1, VISTA, LAG3, ICOS, TIM3, and OX40), tumor-infiltrating lymphocytic markers (i.e., CD3, CD8, CD45RO, granzyme B, PD-1, and FOXP3), and markers to characterize myeloid-derived suppressor cells (i.e., CD68, CD66b, CD14, CD33, Arg-1, and CD11b). To determine analytical reproducibility and the impact of those panels for immuno-profiling tumor tissues, we performed an exploratory analysis in a set of non–small cell lung cancer (NSCLC) samples. The slides were scanned, and the different cell phenotypes were quantified by simultaneous co-localizations with the markers using image analysis software. Comparison between the time points of staining showed high analytical reproducibility. The analysis of NSCLC cases showed an immunosuppressive microenvironment with PD-L1/PD-1 expression as a predominant axis. Interestingly, high density of MCs expressing B7-H4 was correlated with recurrence. Unexpectedly, MCs expressing OX40 were also detected, and those cells were a closer distance to CD3+T-cells than were MCs expressing other immune checkpoints. Two different cellular patterns of spatial distribution were determined according the CD3 distribution, and the predominant pattern was related with active immunosuppressive interaction with MCs. Our study shows that these five mIF panels can identify multiple targets in a single cell with high reproducibility. The study of different cell populations and their spatial relationship can open new ideas for therapeutic approaches.
Collapse
|
36
|
Wang W, Zou R, Qiu Y, Liu J, Xin Y, He T, Qiu Z. Interaction Networks Converging on Immunosuppressive Roles of Granzyme B: Special Niches Within the Tumor Microenvironment. Front Immunol 2021; 12:670324. [PMID: 33868318 PMCID: PMC8047302 DOI: 10.3389/fimmu.2021.670324] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2021] [Accepted: 03/15/2021] [Indexed: 12/12/2022] Open
Abstract
Granzyme B is a renowned effector molecule primarily utilized by CTLs and NK cells against ill-defined and/or transformed cells during immunosurveillance. The overall expression of granzyme B within tumor microenvironment has been well-established as a prognostic marker indicative of priming immunity for a long time. Until recent years, increasing immunosuppressive effects of granzyme B are unveiled in the setting of different immunological context. The accumulative evidence confounded the roles of granzyme B in immune responses, thereby arousing great interests in characterizing detailed feature of granzyme B-positive niche. In this paper, the granzyme B-related regulatory effects of major suppressor cells as well as the tumor microenvironment that defines such functionalities were longitudinally summarized and discussed. Multiplex networks were built upon the interactions among different transcriptional factors, cytokines, and chemokines that regarded to the initiation and regulation of granzyme B-mediated immunosuppression. The conclusions and prospect may facilitate better interpretations of the clinical significance of granzyme B, guiding the rational development of therapeutic regimen and diagnostic probes for anti-tumor purposes.
Collapse
Affiliation(s)
- Weinan Wang
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Rui Zou
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Ye Qiu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Jishuang Liu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Yu Xin
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Tianzhu He
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China.,School of Basic Medical Sciences, Changchun University of Chinese Medicine, Changchun, China
| | - Zhidong Qiu
- School of Pharmaceutical Sciences, Changchun University of Chinese Medicine, Changchun, China
| |
Collapse
|
37
|
Seed RI, Kobayashi K, Ito S, Takasaka N, Cormier A, Jespersen JM, Publicover J, Trilok S, Combes AJ, Chew NW, Chapman J, Krummel MF, Lou J, Marks J, Cheng Y, Baron JL, Nishimura SL. A tumor-specific mechanism of T reg enrichment mediated by the integrin αvβ8. Sci Immunol 2021; 6:6/57/eabf0558. [PMID: 33771888 DOI: 10.1126/sciimmunol.abf0558] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Revised: 01/06/2021] [Accepted: 02/24/2021] [Indexed: 12/14/2022]
Abstract
Regulatory T cells (Tregs) that promote tumor immune evasion are enriched in certain tumors and correlate with poor prognosis. However, mechanisms for Treg enrichment remain incompletely understood. We described a mechanism for Treg enrichment in mouse and human tumors mediated by the αvβ8 integrin. Tumor cell αvβ8 bound to latent transforming growth factor-β (L-TGF-β) presented on the surface of T cells, resulting in TGF-β activation and immunosuppressive Treg differentiation in vitro. In vivo, tumor cell αvβ8 expression correlated with Treg enrichment, immunosuppressive Treg gene expression, and increased tumor growth, which was reduced in mice by αvβ8 inhibition or Treg depletion. Structural modeling and cell-based studies suggested a highly geometrically constrained complex forming between αvβ8-expressing tumor cells and L-TGF-β-expressing T cells, facilitating TGF-β activation, independent of release and diffusion, and providing limited access to TGF-β inhibitors. These findings suggest a highly localized tumor-specific mechanism for Treg enrichment.
Collapse
Affiliation(s)
- Robert I Seed
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Kenji Kobayashi
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Saburo Ito
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Naoki Takasaka
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Anthony Cormier
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Jillian M Jespersen
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jean Publicover
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Suprita Trilok
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Alexis J Combes
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Nayvin W Chew
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX CoLabs, University of California San Francisco, San Francisco, CA 94143, USA
| | - Jocelyne Chapman
- Department of Gynecology and Oncology, University of California, San Francisco San Francisco, CA 94110, USA
| | - Matthew F Krummel
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jianlong Lou
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - James Marks
- Department of Anesthesia and Perioperative Care, University of California, San Francisco, San Francisco, CA 94110, USA
| | - Yifan Cheng
- Department of Biochemistry and Biophysics, University of California, San Francisco, San Francisco, CA 94158, USA.,Howard Hughes Medical Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Jody L Baron
- Department of Medicine and Liver Center, University of California, San Francisco, San Francisco, CA 94143, USA.,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Stephen L Nishimura
- Department of Pathology, University of California, San Francisco, San Francisco, CA 94110, USA. .,ImmunoX Initiative, University of California, San Francisco, San Francisco, CA 94143, USA
| |
Collapse
|
38
|
Germano G, Lu S, Rospo G, Lamba S, Rousseau B, Fanelli S, Stenech D, Le DT, Hays J, Totaro MG, Amodio V, Chilà R, Mondino A, Diaz LA, Di Nicolantonio F, Bardelli A. CD4 T Cell-Dependent Rejection of Beta-2 Microglobulin Null Mismatch Repair-Deficient Tumors. Cancer Discov 2021; 11:1844-1859. [PMID: 33653693 DOI: 10.1158/2159-8290.cd-20-0987] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Revised: 01/09/2021] [Accepted: 02/25/2021] [Indexed: 11/16/2022]
Abstract
Inactivation of beta-2 microglobulin (B2M) is considered a determinant of resistance to immune checkpoint inhibitors (ICPi) in melanoma and lung cancers. In contrast, B2M loss does not appear to affect response to ICPis in mismatch repair-deficient (MMRd) colorectal tumors where biallelic inactivation of B2M is frequently observed. We inactivated B2m in multiple murine MMRd cancer models. Although MMRd cells would not readily grow in immunocompetent mice, MMRd B2m null cells were tumorigenic and regressed when treated with anti-PD-1 and anti-CTLA4. The efficacy of ICPis against MMRd B2m null tumors did not require CD8+ T cells but relied on the presence of CD4+ T cells. Human tumors expressing low levels of B2M display increased intratumoral CD4+ T cells. We conclude that B2M inactivation does not blunt the efficacy of ICPi in MMRd tumors, and we identify a unique role for CD4+ T cells in tumor rejection. SIGNIFICANCE: B2M alterations, which impair antigen presentation, occur frequently in microsatellite-unstable colorectal cancers. Although in melanoma and lung cancers B2M loss is a mechanism of resistance to immune checkpoint blockade, we show that MMRd tumors respond to ICPis through CD4+ T-cell activation.This article is highlighted in the In This Issue feature, p. 1601.
Collapse
Affiliation(s)
- Giovanni Germano
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Steve Lu
- Ludwig Center and Howard Hughes Medical Institute at Johns Hopkins, Baltimore, Maryland
| | - Giuseppe Rospo
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Simona Lamba
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy
| | - Benoit Rousseau
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Sonia Fanelli
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Denise Stenech
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Dung T Le
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, Maryland
| | - John Hays
- Division of Medical Oncology, Wexner Medical Center and James Cancer Hospital, The Ohio State University, Columbus, Ohio
| | | | - Vito Amodio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Rosaria Chilà
- Department of Oncology, University of Torino, Candiolo, Turin, Italy.,IFOM-the FIRC Institute of Molecular Oncology, Milan, Italy
| | - Anna Mondino
- Division of Immunology, Transplantation and Infectious Diseases, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Luis A Diaz
- Division of Solid Tumor Oncology, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | - Federica Di Nicolantonio
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy.,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| | - Alberto Bardelli
- Candiolo Cancer Institute, FPO-IRCCS, Candiolo, Turin, Italy. .,Department of Oncology, University of Torino, Candiolo, Turin, Italy
| |
Collapse
|
39
|
Churov A, Zhulai G. Targeting adenosine and regulatory T cells in cancer immunotherapy. Hum Immunol 2021; 82:270-278. [PMID: 33610376 DOI: 10.1016/j.humimm.2020.12.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2020] [Revised: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 12/19/2022]
Abstract
Immunosuppressive activity of regulatory T cells (Tregs) is one of the mechanisms promoting carcinogenesis. Intratumoral Tregs have some phenotypic and functional traits that lower the efficiency of antitumor immune response, which makes them a good target for immunotherapy. Several approaches to cancer immunotherapy are being developed along this vector: deletion of tumor-infiltrating Tregs, inhibition of their homing to the tumor microenvironment, and functional downregulation of Tregs. Studies of the past decade have demonstrated the role of Tregs and ectonucleotidases CD39 and CD73 in the generation of immunosuppressive extracellular adenosine. Pharmacological targeting of CD39 and CD73 can restrain the activity of suppressor cells and promote the efficiency of cancer therapy. Here we review the latest data on issues regarding the role of extracellular adenosine and its receptors in antitumor immune response, adenosine generation mechanisms involving Tregs and the membrane proteins CD39 and CD73. Innovative approaches to antitumor immunotherapy and clinical studies of Treg targeting and application of anti-CD39/CD73 antibodies, adenosine receptor antagonists, and small-molecule inhibitors of ectonucleotidase activity are explored.
Collapse
Affiliation(s)
- Alexey Churov
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Russian Federation.
| | - Galina Zhulai
- Institute of Biology, Karelian Research Centre, Russian Academy of Sciences, Petrozavodsk, Russian Federation
| |
Collapse
|
40
|
Farnoosh G, Saeedi-Boroujeni A, Jalali A, Keikhaei B, Mahmoudian-Sani MR. Polymorphisms in genes involved in breast cancer among Iranian patients. Per Med 2021; 18:153-169. [PMID: 33565318 DOI: 10.2217/pme-2020-0003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
This review gives a summary of the important genetic polymorphisms in breast cancer with a focus on people in Iran. Several single nucleotide polymorphisms were considered as breast cancer susceptibility polymorphisms within genes (STK15, ERRs, ESR1, p53, SEP15, AURKA, SHBG, SRC, FAS, VEGF, XRCC1, GST, NFκB1, XPC, XRCC3, sirtuin-3, NKG2D). Cytosine-adenine repeat (IGF-I), rs3877899, G-2548A, GGC (eRF3a/GSPT1), IVS2nt-124A/G have shown an increased risk of breast cancers and a decreased risk has been observed in 4G/5G (PAI-1), rs6505162, tri-nucleotide (GCG TGFBR1). We observed that the signaling pathways and antioxidant related genes are the main molecular processes associated with breast cancer progression. Further studies on types of polymorphisms in breast cancer could validate the prognostic value of biomarkers.
Collapse
Affiliation(s)
- Gholamreza Farnoosh
- Applied Biotechnology Research Center, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Ali Saeedi-Boroujeni
- Department of Immunology, Faculty of Medicine, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.,Immunology Today, Universal Scientific Education & Research Network (USERN), Tehran, Iran
| | - Akram Jalali
- Department of Molecular Medicine & Genetics, School of Medicine Hamadan University of Medical Sciences
| | - Bijan Keikhaei
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Mohammad-Reza Mahmoudian-Sani
- Thalassemia & Hemoglobinopathy Research Center, Health Research Institute, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
41
|
Involvement of CD26 in Differentiation and Functions of Th1 and Th17 Subpopulations of T Lymphocytes. J Immunol Res 2021; 2021:6671410. [PMID: 33542930 PMCID: PMC7843192 DOI: 10.1155/2021/6671410] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Revised: 12/12/2020] [Accepted: 12/18/2020] [Indexed: 12/17/2022] Open
Abstract
CD26, acting as a costimulator of T cell activation, plays an important role in the immune system. However, the role of CD26 in the differentiation of T cell subsets, especially of new paradigms of T cells, such as Th17 and Tregs, is not fully clarified. In the present study, the role of CD26 in T cell differentiation was investigated in vitro. CD26 expression was analyzed in the different subsets of human peripheral blood T lymphocytes after solid-phase immobilized specific anti-CD3 mAb stimulation. Here, the percentage of CD4+ cells significantly increased and most of these cells were coexpressed with CD26, suggesting a close correlation of CD26 expression with the proliferation of CD4+ cells. Subsequently, after immobilized anti-CD3 mAb stimulation, CD26 high-expressing cells (CD26high) were separated from CD26 low-expressing cells (CD26low) by magnetic cell sorting. We found that the percentages of cells secreting Th1 typical cytokines (IL-2, IFN-γ) and Th17 typical cytokines (IL-6, IL-17, and IL-22) or expressing Th17 typical biomarkers (IL-23R, CD161, and CD196) in the CD26high group were markedly higher than in those in the CD26low group. In addition, a coexpression of CD26 with IL-2, IFN-γ, IL-17, IL-22, and IL-23R in lymphocytes was demonstrated by fluorescence microscopy. These results provide direct evidence that the high expression of CD26 is accompanied by the differentiation of T lymphocytes into Th1 and Th17, indicating that CD26 plays a crucial role in regulating the immune response.
Collapse
|
42
|
Kolb HR, Borcherding N, Zhang W. Understanding and Targeting Human Cancer Regulatory T Cells to Improve Therapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1278:229-256. [PMID: 33523451 DOI: 10.1007/978-981-15-6407-9_12] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Regulatory T cells (Tregs) are critical in maintaining immune homeostasis under various pathophysiological conditions. A growing body of evidence demonstrates that Tregs play an important role in cancer progression and that they do so by suppressing cancer-directed immune responses. Tregs have been targeted for destruction by exploiting antibodies against and small-molecule inhibitors of several molecules that are highly expressed in Tregs-including immune checkpoint molecules, chemokine receptors, and metabolites. To date, these strategies have had only limited antitumor efficacy, yet they have also created significant risk of autoimmunity because most of them do not differentiate Tregs in tumors from those in normal tissues. Currently, immune checkpoint inhibitor (ICI)-based cancer immunotherapies have revolutionized cancer treatment, but the resistance to ICI is common and the elevation of Tregs is one of the most important mechanisms. Therapeutic strategies that can selectively eliminate Tregs in the tumor (i.e. therapies that do not run the risk of causing autoimmunity by affecting normal tissue), are urgently needed for the development of cancer immunotherapies. This chapter discusses specific properties of human Tregs under the context of cancer and the various ways to target Treg for cancer immunotherapy.
Collapse
Affiliation(s)
- H Ryan Kolb
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA
| | - Nicholas Borcherding
- Department of Pathology and Immunology, Washington University, St. Louis, MO, USA
| | - Weizhou Zhang
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, Gainesville, FL, USA.
| |
Collapse
|
43
|
Özdemir BC. Androgen Signaling in the Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1270:169-183. [PMID: 33123999 DOI: 10.1007/978-3-030-47189-7_10] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
The key function of mesenchymal/stromal androgen receptor (AR) signaling for prostate development has been well documented by tissue recombination experiments. Some studies have addressed the expression and function of AR in stromal cells in prostate cancer, yet our understanding of the role of stromal AR in other tissues beyond prostate is still insufficient.Genomic analysis has revealed that cellular responses to androgens differ between epithelial and stromal cells. AR in stromal cells seems not to act via classical AR transcription factors such as FOXA1 but rather depends on the JUN/AP1 complex. Stromal AR appears to have tumor-promoting and tumor-protective functions depending on tumor stage. Loss of AR signaling in fibroblasts has been detected already in premalignant lesions in the skin and prostate and has been associated with tumor induction in xenografts of skin cancer and aggressive disease features and poor patient prognosis in prostate cancer. Moreover, AR expression is found on virtually all tissue-infiltrating immune cells and plays critical roles in immune cell function. These findings suggest a potential deleterious impact of current androgen deprivation therapies which inhibit both epithelial and stromal AR, highlighting the need to develop tissue-specific AR inhibitors.
Collapse
Affiliation(s)
- Berna C Özdemir
- Department of Oncology, Lausanne University Hospital, Lausanne, Switzerland. .,International Cancer Prevention Institute, Epalinges, Switzerland.
| |
Collapse
|
44
|
González-Navajas JM, Fan DD, Yang S, Yang FM, Lozano-Ruiz B, Shen L, Lee J. The Impact of Tregs on the Anticancer Immunity and the Efficacy of Immune Checkpoint Inhibitor Therapies. Front Immunol 2021; 12:625783. [PMID: 33717139 PMCID: PMC7952426 DOI: 10.3389/fimmu.2021.625783] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Accepted: 02/02/2021] [Indexed: 12/21/2022] Open
Abstract
Although cancers arise from genetic mutations enabling cells to proliferate uncontrollably, they cannot thrive without failure of the anticancer immunity due in a large part to the tumor environment's influence on effector and regulatory T cells. The field of immune checkpoint inhibitor (ICI) therapy for cancer was born out of the fact that tumor environments paralyze the immune cells that are supposed to clear them by activating the immune checkpoint molecules such as PD-1. While various subsets of effector T cells work collaboratively to eliminate cancers, Tregs enriched in the tumor environment can suppress not only the native anticancer immunity but also diminish the efficacy of ICI therapies. Because of their essential role in suppressing autoimmunity, various attempts to specifically deplete tumor-associated Tregs are currently underway to boost the efficacy of ICI therapies without causing systemic autoimmune responses. A better understanding the roles of Tregs in the anti-cancer immunity and ICI therapies should provide more specific targets to deplete intratumoral Tregs. Here, we review the current understanding on how Tregs inhibit the anti-cancer immunity and ICI therapies as well as the advances in the targeted depletion of intratumoral Tregs.
Collapse
Affiliation(s)
- Jose M. González-Navajas
- Alicante Institute for Health and Biomedical Research (ISABIAL), Hospital General Universitario de Alicante, Alicante, Spain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III, Madrid, Spain
- Department of Pharmacology, Pediatrics and Organic Chemistry, University Miguel Hernández, Elche, Spain
- Institute of Research, Development and Innovation in Healthcare Biotechnology in Elche (IDiBE), University Miguel Hernández, Elche, Spain
- Jose M. González-Navajas
| | - Dengxia Denise Fan
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Shuang Yang
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Fengyuan Mandy Yang
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Beatriz Lozano-Ruiz
- Alicante Institute for Health and Biomedical Research (ISABIAL), Hospital General Universitario de Alicante, Alicante, Spain
- Networked Biomedical Research Center for Hepatic and Digestive Diseases (CIBERehd), Institute of Health Carlos III, Madrid, Spain
| | - Liya Shen
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
| | - Jongdae Lee
- State Key Laboratory of Respiratory Disease, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou, China
- *Correspondence: Jongdae Lee ;
| |
Collapse
|
45
|
Vardaka P, Lozano T, Bot C, Ellery J, Whiteside SK, Imianowski CJ, Farrow S, Walker S, Okkenhaug H, Yang J, Okkenhaug K, Kuo P, Roychoudhuri R. A cell-based bioluminescence assay reveals dose-dependent and contextual repression of AP-1-driven gene expression by BACH2. Sci Rep 2020; 10:18902. [PMID: 33144667 PMCID: PMC7641119 DOI: 10.1038/s41598-020-75732-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2020] [Accepted: 10/12/2020] [Indexed: 02/02/2023] Open
Abstract
Whereas effector CD4+ and CD8+ T cells promote immune activation and can drive clearance of infections and cancer, CD4+ regulatory T (Treg) cells suppress their function, contributing to both immune homeostasis and cancer immunosuppression. The transcription factor BACH2 functions as a pervasive regulator of T cell differentiation, promoting development of CD4+ Treg cells and suppressing the effector functions of multiple effector T cell (Teff) lineages. Here, we report the development of a stable cell-based bioluminescence assay of the transcription factor activity of BACH2. Tetracycline-inducible BACH2 expression resulted in suppression of phorbol 12-myristate 13-acetate (PMA)/ionomycin-driven activation of a luciferase reporter containing BACH2/AP-1 target sequences from the mouse Ifng + 18k enhancer. BACH2 expression repressed the luciferase signal in a dose-dependent manner but this activity was abolished at high levels of AP-1 signalling, suggesting contextual regulation of AP-1 driven gene expression by BACH2. Finally, using the reporter assay developed, we find that the histone deacetylase 3 (HDAC3)-selective inhibitor, RGFP966, inhibits BACH2-mediated repression of signal-driven luciferase expression. In addition to enabling mechanistic studies, this cell-based reporter may enable identification of small molecule agonists or antagonists of BACH2 function for drug development.
Collapse
Affiliation(s)
- Panagiota Vardaka
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Teresa Lozano
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Christopher Bot
- CRUK Therapeutic Discovery Laboratories, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Jonathan Ellery
- CRUK Therapeutic Discovery Laboratories, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Sarah K Whiteside
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Charlotte J Imianowski
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Stuart Farrow
- CRUK Therapeutic Discovery Laboratories, Babraham Research Campus, Cambridge, CB22 3AT, UK
| | - Simon Walker
- Imaging Facility, Babraham Institute, Cambridge, CB22 3AT, UK
| | | | - Jie Yang
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
| | - Paula Kuo
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK
| | - Rahul Roychoudhuri
- Department of Pathology, University of Cambridge, Tennis Court Road, Cambridge, CB2 1QP, UK.
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, CB22 3AT, UK.
| |
Collapse
|
46
|
Grant FM, Yang J, Nasrallah R, Clarke J, Sadiyah F, Whiteside SK, Imianowski CJ, Kuo P, Vardaka P, Todorov T, Zandhuis N, Patrascan I, Tough DF, Kometani K, Eil R, Kurosaki T, Okkenhaug K, Roychoudhuri R. BACH2 drives quiescence and maintenance of resting Treg cells to promote homeostasis and cancer immunosuppression. J Exp Med 2020; 217:e20190711. [PMID: 32515782 PMCID: PMC7478731 DOI: 10.1084/jem.20190711] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 12/19/2019] [Accepted: 05/01/2020] [Indexed: 12/14/2022] Open
Abstract
Regulatory T (Treg) cell populations are composed of functionally quiescent resting Treg (rTreg) cells which differentiate into activated Treg (aTreg) cells upon antigen stimulation. How rTreg cells remain quiescent despite chronic exposure to cognate self- and foreign antigens is unclear. The transcription factor BACH2 is critical for early Treg lineage specification, but its function following lineage commitment is unresolved. Here, we show that BACH2 is repurposed following Treg lineage commitment and promotes the quiescence and long-term maintenance of rTreg cells. Bach2 is highly expressed in rTreg cells but is down-regulated in aTreg cells and during inflammation. In rTreg cells, BACH2 binds to enhancers of genes involved in aTreg differentiation and represses their TCR-driven induction by competing with AP-1 factors for DNA binding. This function promotes rTreg cell quiescence and long-term maintenance and is required for immune homeostasis and durable immunosuppression in cancer. Thus, BACH2 supports a "division of labor" between quiescent rTreg cells and their activated progeny in Treg maintenance and function, respectively.
Collapse
Affiliation(s)
- Francis M. Grant
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Jie Yang
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Rabab Nasrallah
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - James Clarke
- La Jolla Institute for Allergy and Immunology, La Jolla, CA
- Cancer Research UK & National Institute for Health Research Experimental Cancer Sciences Unit, University of Southampton, Southampton, UK
| | - Firas Sadiyah
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Sarah K. Whiteside
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Charlotte J. Imianowski
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Paula Kuo
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Panagiota Vardaka
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Tihomir Todorov
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Nordin Zandhuis
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
| | - Ilinca Patrascan
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - David F. Tough
- Epigenetics DPU, Immunoinflammation Therapeutic Area Unit, GSK Medicines Research Centre, Stevenage, UK
| | - Kohei Kometani
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Robert Eil
- Oregon Health and Science University School of Medicine, Portland, OR
| | - Tomohiro Kurosaki
- Laboratory of Lymphocyte Differentiation, WPI Immunology Frontier Research Center, Osaka University, Osaka, Japan
- Laboratory for Lymphocyte Differentiation, RIKEN Center for Integrative Medical Sciences, Yokohama, Kanagawa, Japan
| | - Klaus Okkenhaug
- Department of Pathology, University of Cambridge, Cambridge, UK
| | - Rahul Roychoudhuri
- Laboratory of Lymphocyte Signalling and Development, Babraham Institute, Cambridge, UK
- Department of Pathology, University of Cambridge, Cambridge, UK
| |
Collapse
|
47
|
Li Z, Lin J, Zhang L, Li J, Zhang Y, Zhao C, Wang H. Comprehensive analysis of multiple parameters associated with tumor immune microenvironment in ARID1A mutant cancers. Future Oncol 2020; 16:2295-2306. [PMID: 32639175 DOI: 10.2217/fon-2020-0243] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Aim: To verify the relationship between ARID1A and tumor immune microenvironment thus immune checkpoint inhibitors (ICIs) response. Material & methods: Several public databases were used to characterize the association between ARID1A gene alteration and tumor immunity. Results: The gene mutation frequency was 8.2% in all cancer types. The ARID1A-mutated cancers have higher scores of mutation count, tumor mutational burden, neoantigen load (p < 0.001) and T cell repertoire, B cell repertoire diversity (p < 0.05). The gene mutation has tight association with multiple-activated immune cells. Survival analysis suggested that patients with ARID1A mutant cancers benefit more from ICIs treatment (p = 0.013). Conclusion: The ARID1A gene mutation was correlated with higher tumor immunogenicity and activated antitumor immune microenvironment, resulting in superior cohort that respond well to ICIs.
Collapse
Affiliation(s)
- Zhenxiang Li
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Jiamao Lin
- Department of Medical Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Lijuan Zhang
- Department of Pediatric Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan 250021, PR China
| | - Jingchao Li
- Department of Radiation Oncology, The People's Hospital of Zhangqiu Area, Jinan 250200, China
| | - Yingyun Zhang
- Department of Radiation Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Chenglong Zhao
- Department of pathology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| | - Haiyong Wang
- Department of Medical Oncology, Shandong Cancer Hospital & Institute, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan 250117, PR China
| |
Collapse
|
48
|
Macciò A, Madeddu C. Blocking inflammation to improve immunotherapy of advanced cancer. Immunology 2020; 159:357-364. [PMID: 31821533 PMCID: PMC7078000 DOI: 10.1111/imm.13164] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/27/2019] [Accepted: 12/02/2019] [Indexed: 12/19/2022] Open
Abstract
The ability to induce functional reprogramming of regulatory T (Treg) cells in the tumor microenvironment is an extremely important therapeutic opportunity. However, when discussing such an approach, the opposing effect that the activation of the Treg cell compartments may have in inducing the immune inflammatory response and its link with the efficacy of immunotherapy should be considered. In fact, Treg reprogramming has a dual effect: immediate, with mechanisms that activate immunosurveillance, and late, mediated by the macrophage activation that yields an inflammatory status that is deleterious for the antineoplastic efficiency of the immune system response. Persistence of the inflammatory response is associated with specific changes of oxidative and glycolytic metabolic pathways that interfere with conventional T-cell activation and function and may be one of the reasons for the failure of immunotherapy in advanced cancer patients. Therefore, in addition to modulating Treg cell action, the combined use of drugs able to block chronic inflammation mediated mainly by macrophages, to counteract the oxidative stress, and to positively regulate the metabolic derangements, could improve the effectiveness of modern immunotherapy. In conclusion, reprogramming of Treg cells may be an appropriate strategy for treating early stages of neoplastic diseases, whereas other immunosuppressive mechanisms should be the target of a combined immunotherapy approach in more advanced phases of cancer.
Collapse
Affiliation(s)
- Antonio Macciò
- Department of Gynecologic OncologyAzienda Ospedaliera BrotzuCagliariItaly
| | - Clelia Madeddu
- Department of Medical Sciences and Public HealthUniversity of CagliariCagliariItaly
| |
Collapse
|
49
|
Bai F, Zhang P, Fu Y, Chen H, Zhang M, Huang Q, Li D, Li B, Wu K. Targeting ANXA1 abrogates Treg-mediated immune suppression in triple-negative breast cancer. J Immunother Cancer 2020; 8:e000169. [PMID: 32300050 PMCID: PMC7204868 DOI: 10.1136/jitc-2019-000169] [Citation(s) in RCA: 78] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2020] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND Regulatory T (Treg) cells play a negative role in anti-tumor immunity against triple-negative breast cancer, so it is of great significance to find the potential therapeutic target of Treg cells. METHODS First, Annexin A1 (ANXA1) expression and survival of patients with breast cancer were analyzed using TCGA data. Then plasma ANXA1 levels in patients with malignant and benign breast tumors were detected by ELISA. Next, the effect of ANXA1 on Treg cells was studied through suppressive assays, and how ANXA1 regulates the function of Treg cells was detected by RNA sequencing. Finally, the in vivo experiment in balb/c mice was conducted to test whether the ANXA1 blocker Boc1 could shrink tumors and affect the function of Treg cells. RESULTS Our data suggest that ANXA1 expression is associated with lower survival and a higher risk of breast malignancy. Suppressive assays show that ANXA1 can enhance the inhibition function of Treg cells. RNA-Sequencing results indicate that Boc1 could reduce the expression of granzyme A mRNA in Treg cells. Animal experiments have been done to show that Boc1 can reduce tumor size and down regulate Treg cell function. CONCLUSIONS ANXA1 can enhance the function of Treg cells and reduce the survival rate of patients with breast cancer. Targeting ANXA1 can reduce Treg cell function and shrink breast tumors.
Collapse
MESH Headings
- Adult
- Aged
- Animals
- Annexin A1/antagonists & inhibitors
- Annexin A1/genetics
- Annexin A1/metabolism
- Apoptosis
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Carcinoma, Ductal, Breast/immunology
- Carcinoma, Ductal, Breast/metabolism
- Carcinoma, Ductal, Breast/pathology
- Carcinoma, Lobular/immunology
- Carcinoma, Lobular/metabolism
- Carcinoma, Lobular/pathology
- Cell Movement
- Cell Proliferation
- Female
- Follow-Up Studies
- Gene Expression Regulation, Neoplastic
- Humans
- Mice
- Mice, Inbred BALB C
- Mice, Nude
- Middle Aged
- Prognosis
- Receptor, ErbB-2/metabolism
- Receptors, Estrogen/metabolism
- Receptors, Progesterone/metabolism
- Survival Rate
- T-Lymphocytes, Regulatory/immunology
- Triple Negative Breast Neoplasms/immunology
- Triple Negative Breast Neoplasms/metabolism
- Triple Negative Breast Neoplasms/pathology
- Tumor Cells, Cultured
- Xenograft Model Antitumor Assays
Collapse
Affiliation(s)
- Fang Bai
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Peng Zhang
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Yipeng Fu
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Hongliang Chen
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Mingdi Zhang
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Qianru Huang
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dan Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Immunology and Microbiology, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kejin Wu
- Breast Surgery, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| |
Collapse
|
50
|
Gallimore A, Quezada SA, Roychoudhuri R. Regulatory T cells in cancer: where are we now? Immunology 2019; 157:187-189. [PMID: 31225653 PMCID: PMC6587319 DOI: 10.1111/imm.13088] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 05/31/2019] [Indexed: 12/17/2022] Open
Abstract
There have been substantial strides forward in our understanding of the contribution of regulatory T (Treg) cells to cancer immunosuppression. In this issue, we present a series of papers highlighting emerging themes on this topic relevant not only to our understanding of the fundamental biology of tumour immunosuppression but also to the design of new immunotherapeutic approaches. The substantially shared biology of CD4+ conventional T (Tconv) and Treg cells necessitates a detailed understanding of the potentially opposing functional consequences that immunotherapies will have on Treg and Tconv cells, a prominent example being the potential for Treg-mediated hyperprogressive disease following anti-PD-1 therapy. Such understanding will aid patient stratification and the rational design of combination therapies. It is also becoming clear, however, that Treg cells within tumours exhibit distinct biological features to both Tconv cells and Treg cells in other tissues. These distinct features provide the opportunity for development of targeted immunotherapies with greater efficacy and reduced potential for inducing systemic toxicity.
Collapse
Affiliation(s)
- Awen Gallimore
- Division of Infection and ImmunityCardiff UniversityCardiffUK
| | - Sergio A. Quezada
- Cancer Immunology UnitUniversity College London (UCL) Cancer InstituteLondonUK
| | - Rahul Roychoudhuri
- Laboratory of Lymphocyte Signalling and DevelopmentBabraham InstituteCambridgeUK
| |
Collapse
|