1
|
Tang XS, Xu CL, Li N, Zhang JQ, Tang Y. Landscape of four different stages of human gastric cancer revealed by single-cell sequencing. World J Gastrointest Oncol 2025; 17:97125. [PMID: 39958562 PMCID: PMC11756019 DOI: 10.4251/wjgo.v17.i2.97125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 10/12/2024] [Accepted: 11/08/2024] [Indexed: 01/18/2025] Open
Abstract
BACKGROUND Gastric cancer (GC) poses a substantial risk to human health due to its high prevalence and mortality rates. Nevertheless, current therapeutic strategies remain insufficient. Single-cell RNA sequencing (scRNA-seq) offers the potential to provide comprehensive insights into GC pathogenesis. AIM To explore the distribution and dynamic changes of cell populations in the GC tumor microenvironment using scRNA-seq techniques. METHODS Cancerous tissues and paracancerous tissues were obtained from patients diagnosed with GC at various stages (I, II, III, and IV). Single-cell suspensions were prepared and analyzed using scRNA-seq to examine transcriptome profiles and cell-cell interactions. Additionally, quantitative real-time polymerase chain reaction (qRT-PCR) and flow cytometry were applied for measuring the expression of cluster of differentiation (CD) 2, CD3D, CD3E, cytokeratin 19, cytokeratin 8, and epithelial cell adhesion molecules. RESULTS Transcriptome data from 73645 single cells across eight tissues of four patients were categorized into 25 distinct cell clusters, representing 10 different cell types. Variations were observed in these cell type distribution. The adjacent epithelial cells in stages II and III exhibited a degenerative trend. Additionally, the quantity of CD4 T cells and CD8 T cells were evidently elevated in cancerous tissues. Interaction analysis displayed a remarkable increase in interaction between B cells and other mast cells in stages II, III, and IV of GC. These findings were further validated through qRT-PCR and flow cytometry, demonstrating elevated T cells and declined epithelial cells within the cancerous tissues. CONCLUSION This study provides a comprehensive analysis of cell dynamics across GC stages, highlighting key interactions within the tumor microenvironment. These findings offer valuable insights for developing novel therapeutic strategies.
Collapse
Affiliation(s)
- Xu-Shan Tang
- Department of Gastroenterology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uighur Autonomous Region, China
| | - Chun-Lei Xu
- Department of Gastroenterology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uighur Autonomous Region, China
| | - Na Li
- Department of Gastroenterology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uighur Autonomous Region, China
| | - Jian-Qing Zhang
- Department of Outpatient, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830011, Xinjiang Uighur Autonomous Region, China
| | - Yong Tang
- Department of Gastroenterology, Tumor Hospital Affiliated to Xinjiang Medical University, Urumqi 830011, Xinjiang Uighur Autonomous Region, China
| |
Collapse
|
2
|
Tang T, Wang W, Gan L, Bai J, Tan D, Jiang Y, Zheng P, Zhang W, He Y, Zuo Q, Zhang L. TIGIT expression in extrahepatic cholangiocarcinoma and its impact on CD8 + T cell exhaustion: implications for immunotherapy. Cell Death Dis 2025; 16:90. [PMID: 39939322 PMCID: PMC11822069 DOI: 10.1038/s41419-025-07388-4] [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: 09/23/2024] [Revised: 01/13/2025] [Accepted: 01/22/2025] [Indexed: 02/14/2025]
Abstract
Extrahepatic cholangiocarcinoma (ECCA) is a malignant tumor. The precise role of T-cell immunoreceptor with Ig and ITIM domains (TIGIT), an emerging immunosuppressive receptor, in ECCA, and its impact on CD8+ T cell exhaustion (Tex) remains unclear. We performed single-cell RNA sequencing (scRNA-seq) to characterize tumor-infiltrating lymphocytes (TILs) isolated from ECCA. We found that TIGIT was significantly overexpressed in TOX+CD8 T cells. Tissue microarray and immunohistochemistry staining demonstrated that increased TIGIT expression was associated with poorer patient survival. Flow cytometry analysis revealed that TIGIT+CD8+ T cells exhibited decreased TNF-α, IFN-γ, and TCF-1 expression, accompanied by elevated PD-1 and TIM-3 expression compared to TIGIT-CD8+ T cells. In the patient-derived xenograft (PDX) model, the anti-TIGIT treatment group demonstrated reduced tumor weight, enhanced CD8 frequency, and an increased IFN-γ proportion compared to the PBS treatment group. The TIGIT antibody-treated group exhibited a notably higher fraction of GRZB, and anti-TIGIT treatment led to elevated TCF-1 protein levels and decreased protein levels of TOX1 and NR4A1. Moreover, TIGIT+CD8 T cells from TILs appear to be in a state of exhaustion with low potential killing capacity in ECCA, as shown by scRNA-seq. Taken together, the present study underscores the significant role of TIGIT in ECCA, contributing to T cell exhaustion and a compromised CD8+ T cell immune response. Targeting TIGIT presents a promising therapeutic avenue to enhance the CD8+ T-cell response, thereby potentially improving ECCA therapeutic benefits.
Collapse
Affiliation(s)
- Tengqian Tang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China
| | - Wenhao Wang
- College of Pharmacy, Henan University, Kaifeng, 475001, PR China
| | - Lang Gan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China
| | - Jie Bai
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China
| | - Dehong Tan
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China
| | - Yan Jiang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China
| | - Ping Zheng
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China
| | - Weijun Zhang
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China
| | - Yu He
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China.
| | - Qianfei Zuo
- Department of Microbiology and Biochemical Pharmacy, College of Pharmacy, Army Medical University, Chongqing, 400038, PR China.
| | - Leida Zhang
- Department of Hepatobiliary Surgery, First Affiliated Hospital, Army Medical University, Chongqing, 400038, PR China.
| |
Collapse
|
3
|
Liang R, Liu L, Ding D, Li Y, Ren J, Wei B. CD155 promotes the progression of colorectal cancer by restraining CD8 + T cells via the PI3K/AKT/NF-κB pathway. Cancer Immunol Immunother 2025; 74:94. [PMID: 39891722 PMCID: PMC11787123 DOI: 10.1007/s00262-025-03947-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: 06/29/2024] [Accepted: 01/15/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND CD155 is a crucial factor in the regulation of T cell function and contributes to immune escape. CD155 upregulation has been found in several types of cancer. However, the mechanism by which CD155 regulates CD8+ T cell function in colorectal cancer remains unclear. Here we investigated the role and mechanism of CD155 in the regulation of CD8+ T cell function. METHODS We studied the expression of CD155 in colorectal cancer tissues through western blot, immunohistochemistry, and the TCGA database. We verified the effects of CD155 on the functions of colorectal cancer cells and CD8+ T cells through in vitro experiments. We demonstrated that CD155 affects CD8+ T cell migration and thus promotes tumor growth in a mouse subcutaneous tumor model. We then tested the changes in the PI3K/AKT/NF-κB pathway in CD8+ T cells by flow cytometry. RESULTS We demonstrated that stable CD155 expression was negatively correlated with prognosis in colorectal cancer patients. In vitro experiments confirmed that CD155 does not affect tumor cell proliferation, migration, or invasion. We also revealed that CD155 downregulated the function and migration of CD8+ T cells in vivo and in vitro. Furthermore, CD155 might regulate CD8+ T cells function via the PI3K/AKT/NF-κB pathway. CONCLUSION This study revealed that CD155 can promote the progression of colorectal cancer by regulating the PI3K / AKT-NF-κB pathway to promote the depletion of CD8+ T cells and reduce their migration to the tumor microenvironment. CD155 may become an important prognostic biomarker and an effective target for colorectal cancer immunotherapy.
Collapse
Affiliation(s)
- Rongpu Liang
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Liting Liu
- Department of Pediatrics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Dongbing Ding
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China
| | - Yiquan Li
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Jiannan Ren
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China
| | - Bo Wei
- Department of Gastrointestinal Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, 510630, People's Republic of China.
- Department of Gastrointestinal Surgery, Lingnan Hospital, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong, People's Republic of China.
| |
Collapse
|
4
|
Ke J, Huang S, He Z, Lei S, Lin S, Duan M. TIGIT Regulates T Cell Inflammation in Airway Inflammatory Diseases. Inflammation 2025; 48:15-24. [PMID: 38780694 DOI: 10.1007/s10753-024-02045-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: 03/16/2024] [Revised: 05/06/2024] [Accepted: 05/06/2024] [Indexed: 05/25/2024]
Abstract
TIGIT, a co-inhibitory receptor found on T cells and NK cells, transmits inhibitory signals upon binding to its ligand. This interaction suppresses the activation of various signaling pathways, leading to functional exhaustion of cells, ultimately dampening excessive inflammatory responses or facilitating immune evasion in tumors. Dysregulated TIGIT expression has been noted in T cells across different inflammatory conditions, exhibiting varying effects based on T cell subsets. TIGIT predominantly restrains the effector function of pro-inflammatory T cells, upholds the suppressive function of regulatory T cells, and influences Tfh maturation. Mechanistically, the IL27-induced transcription factors c-Maf and Blimp-1 are believed to be key regulators of TIGIT expression in T cells. Notably, TIGIT expression in T cells is implicated in lung diseases, particularly airway inflammatory conditions such as lung cancer, obstructive pulmonary disease, interstitial lung disease, sarcoidosis, and COVID-19. This review emphasizes the significance of TIGIT in the context of T cell immunity and airway inflammatory diseases.
Collapse
Affiliation(s)
- Junyi Ke
- Guangxi Medical University, Nanning, China
- The Second Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Shu Huang
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | | | - Siyu Lei
- Wuming Hospital of Guangxi Medical University, Nanning, China
| | - Shiya Lin
- Guangxi Medical University, Nanning, China
| | - Minchao Duan
- Wuming Hospital of Guangxi Medical University, Nanning, China.
- Department of Respiratory Medicine, Wuming Hospital of Guangxi Medical University, No.26 Yongning Road, Wuming District, Nanning, 530100, China.
| |
Collapse
|
5
|
Li G, Wen Z, Xiong S. Microenvironmental β-TrCP negates amino acid transport to trigger CD8 + T cell exhaustion in human non-small cell lung cancer. Cell Rep 2025; 44:115128. [PMID: 39754718 DOI: 10.1016/j.celrep.2024.115128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/05/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
CD8+ T cell exhaustion (Tex) has been widely acknowledged in human cancer, while the underlying mechanisms remain unclear. Here, we demonstrate that reduced amino acid (aa) metabolism and mTOR inactivation are accountable for Tex in human non-small cell lung cancer (NSCLC). NSCLC cells impede the T cell-intrinsic transcription of SLC7A5 and SLC38A1, disrupting aa transport and consequently leading to mTOR inactivation. Further, the ubiquitination of YAP1 protein is the basis for NSCLC-mediated transcriptional inhibition of aa transporters. Mechanistically, NSCLC cells transfer β-TrCP-containing exosomes into T cells, inducing YAP1 ubiquitination and Tex. Consequently, inhibiting cancer-associated β-TrCP effectively restores the anti-tumor immune response of CD8+ T cells and curtails tumor growth in NSCLC patient-derived organoids. Together, our findings highlight a β-TrCP-dependent mechanism in steering intrinsic metabolic adaptation and CD8+ Tex, emphasizing microenvironmental β-TrCP as an immune checkpoint for therapeutic exploration against human NSCLC.
Collapse
Affiliation(s)
- Ge Li
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China
| | - Zhenke Wen
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| | - Sidong Xiong
- The Fourth Affiliated Hospital of Soochow University, Institutes of Biology and Medical Sciences, Suzhou Medical College of Soochow University, Soochow University, Suzhou, China; Jiangsu Key Laboratory of Infection and Immunity, Soochow University, Suzhou, China.
| |
Collapse
|
6
|
Korpan M, Puhr HC, Berger JM, Friedrich A, Prager GW, Preusser M, Ilhan-Mutlu A. Current Landscape of Molecular Biomarkers in Gastroesophageal Tumors and Potential Strategies for Co-Expression Patterns. Cancers (Basel) 2025; 17:340. [PMID: 39941712 PMCID: PMC11816248 DOI: 10.3390/cancers17030340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/14/2025] [Accepted: 01/18/2025] [Indexed: 02/16/2025] Open
Abstract
The treatment of metastasized gastroesophageal adenocarcinoma largely depends on molecular profiling based on immunohistochemical procedures. Therefore, the examination of HER2, PD-L1, and dMMR/MSI is recommended by the majority of clinical practice guidelines, as positive expression leads to different treatment approaches. Data from large phase-III trials and consequent approvals in various countries enable physicians to offer their patients several therapy options including immunotherapy, targeted therapy, or both combined with chemotherapy. The introduction of novel therapeutic targets such as CLDN18.2 leads to a more complex decision-making process as a significant number of patients show positive results for the co-expression of other biomarkers besides CLDN18.2. The aim of this review is to summarize the current biomarker landscape of patients with metastatic gastroesophageal tumors, its direct clinical impact on daily decision-making, and to evaluate current findings on biomarker co-expression. Furthermore, possible treatment strategies with multiple biomarker expression are discussed.
Collapse
Affiliation(s)
- Martin Korpan
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Hannah Christina Puhr
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Julia M. Berger
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Alexander Friedrich
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Gerald W. Prager
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Matthias Preusser
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
- Christian Doppler Laboratory for Personalized Immunotherapy, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| | - Aysegül Ilhan-Mutlu
- Division of Oncology, Department of Medicine I, Medical University of Vienna, Waehringer Guertel 18-20, 1090 Vienna, Austria
| |
Collapse
|
7
|
Sun J, Tian Y, Yang C. Target therapy of TIGIT; a novel approach of immunotherapy for the treatment of colorectal cancer. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:231-241. [PMID: 39158733 DOI: 10.1007/s00210-024-03346-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 07/30/2024] [Indexed: 08/20/2024]
Abstract
The T cell immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT), a newly discovered checkpoint, is characterized by its elevated expression on CD4 + T cells, CD8 + T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). Research to date has been shown that TIGIT has been linked to exhaustion of NK cell both and T cells in numerous cancers. CD155, being the specific ligand of TIGIT in humans, emerges as a key target for immunotherapy owing to its crucial interaction with TIGIT. Furthermore, numerous studies have demonstrated that the combination of TIGIT with other immune checkpoint inhibitors (ICIs) and/or traditional treatments elicits a potent antitumor response in colorectal cancer (CRC). This review provides an overview of the structure, function, and signaling pathways associated with TIGIT across multiple immune system cell types. Additionally, focusing on the role of TIGIT in the progression of CRC, this study reviewed various studies exploring TIGIT-based immunotherapy in CRC.
Collapse
Affiliation(s)
- Jing Sun
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Yan Tian
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China
| | - Changqing Yang
- Department of Gastroenterology, Heping Hospital Affiliated to Changzhi Medical College, Changzhi, 046000, China.
| |
Collapse
|
8
|
Wu Y, Jiang X, Yu Z, Xing Z, Ma Y, Qing H. Mechanisms of Anti-PD Therapy Resistance in Digestive System Neoplasms. Recent Pat Anticancer Drug Discov 2025; 20:1-25. [PMID: 38305306 DOI: 10.2174/0115748928269276231120103256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 09/25/2023] [Accepted: 10/03/2023] [Indexed: 02/03/2024]
Abstract
Digestive system neoplasms are highly heterogeneous and exhibit complex resistance mechanisms that render anti-programmed cell death protein (PD) therapies poorly effective. The tumor microenvironment (TME) plays a pivotal role in tumor development, apart from supplying energy for tumor proliferation and impeding the body's anti-tumor immune response, the TME actively facilitates tumor progression and immune escape via diverse pathways, which include the modulation of heritable gene expression alterations and the intricate interplay with the gut microbiota. In this review, we aim to elucidate the mechanisms underlying drug resistance in digestive tumors, focusing on immune-mediated resistance, microbial crosstalk, metabolism, and epigenetics. We will highlight the unique characteristics of each digestive tumor and emphasize the significance of the tumor immune microenvironment (TIME). Furthermore, we will discuss the current therapeutic strategies that hold promise for combination with cancer immune normalization therapies. This review aims to provide a thorough understanding of the resistance mechanisms in digestive tumors and offer insights into potential therapeutic interventions.
Collapse
Affiliation(s)
- Yuxia Wu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xiangyan Jiang
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zeyuan Yu
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Zongrui Xing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yong Ma
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Huiguo Qing
- Department of General Surgery, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| |
Collapse
|
9
|
Choudhury A, Chatterjee S, Dalui S, Ghosh P, Daptary AH, Mollah GK, Bhattacharyya A. Breast cancer cell derived exosomes reduces glycolysis of activated CD8 + T cells in a AKT-mTOR dependent manner. Cell Biol Int 2025; 49:45-54. [PMID: 39285531 DOI: 10.1002/cbin.12241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 08/21/2024] [Accepted: 09/03/2024] [Indexed: 12/18/2024]
Abstract
Cytotoxic CD8+ T cells plays a pivotal role in the adaptive immune system to protect the organism against infections and cancer. During activation and response, T cells undergo a metabolic reprogramming that involves various metabolic pathways, with a predominant reliance on glycolysis to meet their increased energy demands and enhanced effector response. Recently, extracellular vesicles (EVs) known as exosomes have been recognized as crucial signaling mediators in regulating the tumor microenvironment (TME). Recent reports indicates that exosomes may transfer biologically functional molecules to the recipient cells, thereby facilitate cancer progression, angiogenesis, metastasis, drug resistance, and immunosuppression by reprogramming the metabolism of cancer cells. This study sought to enlighten possible involvement of cancer-derived exosomes in CD8 + T cell glucose metabolism and discover a regulated signalome as a mechanism of action. We observed reduction in glucose metabolism due to downregulation of AKT/mTOR signalome in activated CD8 + T cells after cancer derived exosome exposure. In-vivo murine breast tumor studies showed better tumor control and antitumor CD8 + T cell glycolysis and effector response after abrogation of exosome release from breast cancer cells. Summarizing, the present study establishes an immune evasion mechanism of breast cancer cell secreted exosomes that will act as a foundation for future precision cancer therapeutics.
Collapse
Affiliation(s)
- Abhishek Choudhury
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| | - Soumya Chatterjee
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| | - Shauryabrota Dalui
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| | - Pronabesh Ghosh
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| | | | | | - Arindam Bhattacharyya
- Immunology Laboratory, Department of Zoology, University of Calcutta, Kolkata, India
| |
Collapse
|
10
|
Yin K, Guo Y, Wang J, Guo S, Zhang C, Dai Y, Guo Y, Dai C. Identification of a novel immune checkpoint-related gene signature predicts prognosis and immunotherapy in breast cancer and experiment verification. Sci Rep 2024; 14:31065. [PMID: 39730892 DOI: 10.1038/s41598-024-82266-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2024] [Accepted: 12/04/2024] [Indexed: 12/29/2024] Open
Abstract
Breast cancer (BRCA) is one of the pivotal causes of female death worldwide. And the morbidity and mortality of breast cancer have increased rapidly. Immune checkpoints are important to maintain immune tolerance and are regarded as important therapeutic targets. However, research for BRCA were limited to single immune checkpoint-related gene (ICG) and few studies have systematically explored expression profile of Immune checkpoint-related genes or attempted to construct a prognostic gene risk model based on immune checkpoint-related genes. We identified immune checkpoint-related differentially expressed genes (DEGs) in BRCA and normal breast tissues from TCGA database. A 7-gene signature was created by utilizing the univariate Cox regression model with least absolute shrinkage and selection operator (LASSO) Cox regression method. In addition, we conducted a nomogram to predict the prognostic significance. This tool enables quantitative prediction of patient prognosis, serving as a valuable reference for clinical decision-making, thereby improving patient outcomes. Relationships between our risk model and clinical indicators, TME (Tumor Microenvironment), immune cell infiltration, immune response and drug susceptibility were investigated. A set of in vitro cell assays was conducted to decipher the relationship between MAP2K6 and proliferation, invasion, migration, colony formation and apoptosis rate of breast cancer cells. As a result, we established a prognostic model composed of seven ICGs in BRCA. Based on the median risk score, BRCA patients were equally assigned into two groups of high- and low-risk. High-risk BRCA patients have poorer OS (overall survival) than low-risk patients. In addition, there were remarkable differences between these two groups in clinicopathological features, TME, immune cell infiltration, immune response and drug susceptibility. The results of GO and KEGG analyses indicated that DEGs between the high- and low-risk groups were involved in immune-related biological processes and pathways. GSEA analysis also showed that a number of immune-related pathways were notably enriched in the low-risk group. Finally, results of cell-based assays indicated that MAP2K6 may play a pivotal role in the initiation and progression of breast cancer as a tumor suppressor gene. In conclusion, we created a novel ICG signature that has the potential to predict the survival and drug sensitivity of BRCA patients. Furthermore, this study indicated that MAP2K6 may serve as a novel target for BRCA therapy.
Collapse
Affiliation(s)
- Ke Yin
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yangyang Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Jinqiu Wang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Shenchao Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Chunxu Zhang
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China
| | - Yongping Dai
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Yu Guo
- Department of Thyroid and Breast Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Chen Dai
- Digestive Diseases Center, The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, Guangdong, China.
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-Sen University, No. 628 Zhenyuan Road, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
11
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
12
|
Hosseininejad-Chafi M, Eftekhari Z, Oghalaie A, Behdani M, Sotoudeh N, Kazemi-Lomedasht F. Nanobodies as innovative immune checkpoint modulators: advancing cancer immunotherapy. Med Oncol 2024; 42:36. [PMID: 39719469 DOI: 10.1007/s12032-024-02588-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 12/14/2024] [Indexed: 12/26/2024]
Abstract
The immune system relies on a delicate balance between attacking harmful pathogens and preserving the body's own tissues, a balance maintained by immune checkpoints. These checkpoints play a critical role in preventing autoimmune diseases by restraining excessive immune responses while allowing the immune system to recognize and destroy abnormal cells, such as tumors. In recent years, immune checkpoint inhibitors (ICIs) have become central to cancer therapy, enabling the immune system to target and eliminate cancer cells that evade detection. Traditional antibodies, such as IgGs, have been widely used in immune therapies but are limited by their size and complexity. Nanobodies (Nbs), derived from camelid heavy-chain-only antibodies, offer a promising alternative. These small, stable antibody fragments retain the antigen-binding specificity of traditional antibodies but have enhanced solubility and the ability to target otherwise inaccessible epitopes. This review explores the use of Nbs as ICIs, emphasizing their potential in cancer immunotherapy and other immune-related treatments. Their unique structural properties and small size make Nbs highly effective tools for modulating immune responses, representing a novel approach in the evolving landscape of checkpoint inhibitor therapies.
Collapse
Affiliation(s)
- Mohammad Hosseininejad-Chafi
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Zohre Eftekhari
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Akbar Oghalaie
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Mahdi Behdani
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Nazli Sotoudeh
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran
| | - Fatemeh Kazemi-Lomedasht
- Venom and Biotherapeutics Molecules Laboratory, Biotechnology Department, Biotechnology Research Center, Pasteur Institute of Iran, Tehran, 1316943551, Iran.
| |
Collapse
|
13
|
Wang Y, Li N, Chen X, Zhao Y, Qu L, Cai D. Mechanistic insights into sevoflurane-induced hippocampal neuronal damage and cognitive dysfunction through the NEAT1/Nrf2 signaling axis in aged rats. Cell Biol Toxicol 2024; 41:13. [PMID: 39707048 PMCID: PMC11662051 DOI: 10.1007/s10565-024-09964-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Accepted: 11/29/2024] [Indexed: 12/23/2024]
Abstract
The use of anesthetics during surgery can cause severe neurological damage and cognitive dysfunction in elderly patients. However, this health issue currently lacks corresponding therapeutic strategies. This research involved the utilization of single-cell RNA sequencing (scRNA-seq) and transcriptomic assessment to pinpoint crucial cell classifications and molecular pathways, as well as the lncRNA expression profiles, that undergo substantial alterations in aged rats experiencing sevoflurane-induced cognitive impairment. The results of our investigation pointed towards the enrichment of differentially expressed genes in neurons within the Nrf2/ARE signaling pathway, alongside an elevated expression of lncRNA NEAT1. Subsequently, by constructing a rat model to induce neuronal dysfunction with sevoflurane and performing experiments both in vivo and in vitro (including TUNEL staining, H&E staining, immunohistochemistry, immunofluorescence, and flow cytometry to assess apoptosis levels), we confirmed that NEAT1 inhibits the Nrf2/ARE/HO-1 pathway-related factors. Sevoflurane promotes oxidative stress and apoptosis in primary hippocampal neurons through the NEAT1/Nrf2/ARE/HO-1 axis. This study elucidates the molecular mechanism by which sevoflurane induces hippocampal neuronal damage and cognitive decline in elderly rats via the regulation of the lncRNA NEAT1/Nrf2 signaling axis. We discovered that upregulation of NEAT1 suppresses the Nrf2 signaling pathway, further inducing neuronal damage and cognitive dysfunction, furnishing an essential citation to grasp the molecular pathways involved in neuronal harm and devising corresponding treatment methodologies.
Collapse
Affiliation(s)
- Yiliang Wang
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, 110122, Liaoning, People's Republic of China
| | - Nu Li
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China
| | - Xiaoyu Chen
- Department of Anesthesiology, General Hospital of Northern Theater Command, Shenyang, 110001, Liaoning, People's Republic of China
| | - Yue Zhao
- Department of Anesthesiology, Second Affiliated Hospital of Liaoning University of Traditional Chinese Medicine, Shenyang, 110001, Liaoning, People's Republic of China
| | - Letian Qu
- Department of Geriatrics, The First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| | - Dasheng Cai
- Department of Anesthesiology, the First Hospital of China Medical University, Shenyang, 110001, Liaoning, People's Republic of China.
| |
Collapse
|
14
|
Tang Q, Leng S, Tan Y, Cheng H, Liu Q, Wang Z, Xu Y, Zhu L, Wang C. Chitosan/dextran-based organohydrogel delivers EZH2 inhibitor to epigenetically reprogram chemo/immuno-resistance in unresectable metastatic melanoma. Carbohydr Polym 2024; 346:122645. [PMID: 39245506 DOI: 10.1016/j.carbpol.2024.122645] [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: 03/06/2024] [Revised: 08/16/2024] [Accepted: 08/20/2024] [Indexed: 09/10/2024]
Abstract
Melanoma either intrinsically possesses resistance or rapidly acquires resistance to anti-tumor therapy, which often leads to local recurrence or distant metastasis after resection. In this study, we found histone 3 lysine 27 (H3K27) demethylated by an inhibitor of histone methyltransferase EZH2 could epigenetically reverse the resistance to chemo-drug paclitaxel (PTX), or enhance the efficacy of immune checkpoint inhibitor anti-TIGIT via downregulating TIGIT ligand CD155. Next, to address the complexity in the combination of multiple bioactive molecules with distinct therapeutic properties, we developed a polysaccharides-based organohydrogel (OHG) configured with a heterogenous network. Therein, hydroxypropyl chitosan (HPC)-stabilized emulsions for hydrophobic drug entrapment were crosslinked with oxidized dextran (Odex) to form a hydrophilic gel matrix to facilitate antibody accommodation, which demonstrated a tunable sustained release profile by optimizing emulsion/gel volume ratios. As results, local injection of OHG loaded with EZH2 inhibitor UNC1999, PTX and anti-TIGIT did not only synergistically enhance the cytotoxicity of PTX, but also reprogrammed the immune resistance via bi-directionally blocking TIGIT/CD155 axis, leading to the recruitment of cytotoxic effector cells into tumor and conferring a systemic immune memory to prevent lung metastasis. Hence, this polysaccharides-based OHG represents a potential in-situ epigenetic-, chemo- and immunotherapy platform to treat unresectable metastatic melanoma.
Collapse
Affiliation(s)
- Qi Tang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China
| | - Shaolong Leng
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, PR China
| | - Yinqiu Tan
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan 430022, PR China
| | - Huan Cheng
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, PR China
| | - Qi Liu
- The First Dongguan Affiliated Hospital Guangdong Medical University No. 42, Jiaoping Road Dongguan, Guangdong 523710, PR China
| | - Zhongjuan Wang
- Department of Pharmacy, Yan'an Hospital Affiliated to Kunming Medical University, No.245, People East Road, Kunming 650051, PR China
| | - Yunsheng Xu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, PR China.
| | - Linyu Zhu
- Department of Dermatovenereology, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, PR China.
| | - Cuifeng Wang
- School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, PR China; Department of neurosurgery, JiuJiang Hospital of Traditional Chinese Medicine, Jiujiang, PR China.
| |
Collapse
|
15
|
Gibbs RJ, Chambers AC, Hill DJ. The emerging role of Fusobacteria in carcinogenesis. Eur J Clin Invest 2024; 54 Suppl 2:e14353. [PMID: 39674881 DOI: 10.1111/eci.14353] [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: 09/27/2024] [Accepted: 11/04/2024] [Indexed: 12/17/2024]
Abstract
The Fusobacterium genus comprises Gram-negative, obligate anaerobic bacteria that typically reside in the periodontium of the oral cavity, gastrointestinal tract, and female genital tract. The association of Fusobacterial spp. with colorectal tumours is widely accepted, with further evidence that this pathogen may also be implicated in the development of other malignancies. Fusobacterial spp. influence malignant cell behaviours and the tumour microenvironment in various ways, which can be related to the multiple surface adhesins expressed. These adhesins include Fap2 (fibroblast-activated protein 2), CpbF (CEACAM binding protein of Fusobacteria), FadA (Fusobacterium adhesin A) and FomA (Fusobacterial outer membrane protein A). This review outlines the influence of Fusobacteria in promoting cancer initiation and progression, impacts of therapeutic outcomes and discusses potential therapeutic interventions where appropriate.
Collapse
|
16
|
Yamada Y, Miyoshi H, Takeuchi M, Nakashima K, Yamada K, Kato T, Tanaka K, Kohno K, Imaizumi Y, Miyazaki Y, Ohshima K. TIGIT expression on neoplastic cells is a poor prognostic factor for adult T-cell leukaemia/lymphoma. Pathology 2024; 56:993-999. [PMID: 39266421 DOI: 10.1016/j.pathol.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 05/02/2024] [Accepted: 06/02/2024] [Indexed: 09/14/2024]
Abstract
Adult T-cell leukaemia/lymphoma (ATLL) is an aggressive peripheral T-cell neoplasm with a poor prognosis. T-cell immunoreceptor with immunoglobulin and immunoreceptor tyrosine-based inhibitory motif domains (TIGIT) is an immune checkpoint receptor expressed on T and natural killer cells. Although increased TIGIT expression in the tumour microenvironment is associated with poor prognosis in various neoplasms, its relevance in ATLL remains unknown. Herein, we investigated the clinicopathological impact of TIGIT expression on ATLL using immunohistochemistry. TIGIT expression was detected in 21 of 84 patients (25%). A partial association between the clinical features and immune checkpoint molecules and the expression of TIGIT was found including sIL-2R, CD86 and GITR. TIGIT-positive patients [median survival time (MST) 8.9 months, 95% confidence interval (CI) 7.7-15.6] had inferior overall survival compared with TIGIT-negative patients (MST 18.7 months, 95% CI 12.0-36.4) (p=0.0124]. TIGIT expression maintained its prognostic value for overall survival in both univariate and multivariate analyses [hazard ratio (HR) 1.909; 95% CI 1.044-3.488; p=0.0356]. Further studies are required to clarify the clinical and biological significance of TIGIT expression in patients with ATLL.
Collapse
Affiliation(s)
- Yuichi Yamada
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan; Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan
| | - Hiroaki Miyoshi
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan.
| | - Mai Takeuchi
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kazutaka Nakashima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Kyohei Yamada
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Takeharu Kato
- Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan
| | - Ken Tanaka
- Department of Hematology/Oncology, Wakayama Medical University, Wakayama, Japan
| | - Kei Kohno
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| | - Yoshitaka Imaizumi
- Department of Hematology, National Hospital Organization Nagasaki Medical Center, Omura, Japan
| | - Yasushi Miyazaki
- Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan; Department of Hematology, Nagasaki University Hospital, Nagasaki, Japan; Department of Hematology, Atomic Bomb Disease and Hibakusha Medicine Unit, Atomic Bomb Disease Institute, Nagasaki University, Nagasaki, Japan
| | - Koichi Ohshima
- Department of Pathology, Kurume University School of Medicine, Kurume, Japan
| |
Collapse
|
17
|
Huang F, Wang F, Hu Q, Li Y, Jiang D. PTGR1-mediated immune evasion mechanisms in late-stage triple-negative breast cancer: mechanisms of M2 macrophage infiltration and CD8 + T cell suppression. Apoptosis 2024; 29:2002-2024. [PMID: 39068625 DOI: 10.1007/s10495-024-01991-0] [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] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
Triple-negative breast cancer (TNBC) is a heterogeneous disease characterized by metabolic dysregulation. Tumor cell immune escape plays an indispensable role in the development of TNBC tumors. Furthermore, in the abstract, we explicitly mention the techniques used and enhance the clarity and impact of our findings. "Based on bioinformatics analysis results, we utilized CRISPR/Cas9 technology to knockout the target gene and established a mouse model of breast cancer. Through experiments such as CCK8, scratch assay, and Transwell assay, we further investigated the impact of target gene knockout on the malignant behavior of tumor cells. Subsequently, we conducted immunohistochemistry and Western Blot experiments to study the expression of macrophage polarization and infiltration-related markers and evaluate the effect of the target gene on macrophage polarization. Next, through co-culture experiments, we simulated the tumor microenvironment and used immunohistochemistry staining to observe and analyze the distribution and activation status of M2 macrophages and CD8+ T cells in the co-culture system. We validated in vivo experiments the molecular mechanism by which the target gene regulates immune cell impact on TNBC progression.
Collapse
Affiliation(s)
- Fang Huang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China
| | - Fuhe Wang
- Department of General surgery, Hebei Yiling Hospital, Shijiazhuang, 050000, P. R. China
| | - Qilu Hu
- Department of Radiotherapy, Heze Traditional Chinese Medicine Hospital, Heze, 274008, P. R. China
| | - Ying Li
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China
| | - Da Jiang
- Department of Medical Oncology, The Fourth Hospital of Hebei Medical University, East Campus, No.169 Tianshan Street, Shijiazhuang, 050000, Hebei Province, P. R. China.
| |
Collapse
|
18
|
Jin K, Zhao D, Zhou J, Zhang X, Wang Y, Wu Z. Pulsed electromagnetic fields inhibit IL-37 to alleviate CD8 + T cell dysfunction and suppress cervical cancer progression. Apoptosis 2024; 29:2108-2127. [PMID: 39404933 DOI: 10.1007/s10495-024-02006-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/12/2024] [Indexed: 11/10/2024]
Abstract
Pulsed electromagnetic field (PEMF) therapy is a potential non-invasive treatment to modulate immune responses and inhibit tumor growth. Cervical cancer (CC) is influenced by IL-37-mediated immune regulation, making PEMF therapy a potential strategy to impede CC progression. This study aimed to elucidate the effects of PEMF on IL-37 regulation and its molecular mechanisms in CC. CC cell-xenografted mouse models, including IL-37 transgenic (IL-37tg) mice, were used to assess tumor growth through in vivo fluorescence imaging and analyze CC cell apoptosis via flow cytometry. TCGA-CESC transcriptome and clinical data were analyzed to identify key inflammation and immune-related genes. CD8+ T cell models were stimulated with PEMF, and apoptosis, oxidative stress, and inflammatory factor expression were analyzed through RT-qPCR, Western blot, and flow cytometry. PEMF treatment significantly inhibited IL-37 expression (p < 0.05), promoted inflammatory factor release (TNF-α and IL-6), and activated oxidative stress, leading to increased CC cell apoptosis (p < 0.05). IL-37 interaction with SMAD3 impacted the p38/NF-κB signaling pathway, modulating CD8+ T cell activity and cytotoxicity. Co-culture of Hela cells with CD8+ T cells under PEMF treatment showed reduced proliferation (by 40%), migration, and invasion (p < 0.05). In vivo experiments with CC-bearing mice demonstrated that PEMF treatment downregulated IL-37 expression (p < 0.05), enhanced CD8+ T cell function, and inhibited tumor growth (p < 0.05). These molecular mechanisms were validated through RT-qPCR, Western blot, and immunohistochemistry. Thus, PEMF therapy inhibits CC progression by downregulating IL-37 and improving CD8+ T cell function via the SMAD3/p38/NF-κB signaling pathway.
Collapse
Affiliation(s)
- Ke Jin
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Dan Zhao
- Department of Oncology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Jun Zhou
- School of Electronic Science and Engineering, University of Electronic Science and Technology of China, Chengdu, 611731, China
| | - Xun Zhang
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West 2nd Section, First Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China
| | - Yujue Wang
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West 2nd Section, First Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China.
| | - Zhao Wu
- Department of Obstetrics and Gynecology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32 West 2nd Section, First Ring Road, Qingyang District, Chengdu, 610072, Sichuan Province, China.
| |
Collapse
|
19
|
Yang MQ, Zhang SL, Sun L, Huang LT, Yu J, Zhang JH, Tian Y, Han CB, Ma JT. Targeting mitochondria: restoring the antitumor efficacy of exhausted T cells. Mol Cancer 2024; 23:260. [PMID: 39563438 PMCID: PMC11575104 DOI: 10.1186/s12943-024-02175-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 11/10/2024] [Indexed: 11/21/2024] Open
Abstract
Immune checkpoint blockade therapy has revolutionized cancer treatment, but resistance remains prevalent, often due to dysfunctional tumor-infiltrating lymphocytes. A key contributor to this dysfunction is mitochondrial dysfunction, characterized by defective oxidative phosphorylation, impaired adaptation, and depolarization, which promotes T cell exhaustion and severely compromises antitumor efficacy. This review summarizes recent advances in restoring the function of exhausted T cells through mitochondria-targeted strategies, such as metabolic remodeling, enhanced biogenesis, and regulation of antioxidant and reactive oxygen species, with the aim of reversing the state of T cell exhaustion and improving the response to immunotherapy. A deeper understanding of the role of mitochondria in T cell exhaustion lays the foundation for the development of novel mitochondria-targeted therapies and opens a new chapter in cancer immunotherapy.
Collapse
Affiliation(s)
- Mei-Qi Yang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Shu-Ling Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Li Sun
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Le-Tian Huang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jing Yu
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Jie-Hui Zhang
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Yuan Tian
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Cheng-Bo Han
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| | - Jie-Tao Ma
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
- Department of Oncology, Innovative Cancer Drug Research and Development Engineering Center of Liaoning Province, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
20
|
Matsuoka T, Yashiro M. Molecular Mechanism for Malignant Progression of Gastric Cancer Within the Tumor Microenvironment. Int J Mol Sci 2024; 25:11735. [PMID: 39519285 PMCID: PMC11546171 DOI: 10.3390/ijms252111735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/23/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Gastric cancer (GC) is one of the most common cancers worldwide. Most patients are diagnosed at the progressive stage of GC, and progress in the development of effective anti-GC drugs has been insufficient. The tumor microenvironment (TME) regulates various functions of tumor cells, and interactions between the cellular and molecular components of the TME-e.g., inflammatory cells, fibroblasts, vasculature cells, and innate and adaptive immune cells-promote the aggressiveness of cancer cells and dissemination to distant organs. This review summarizes the roles of various TME cells and molecules in regulating the malignant progression and metastasis of GC. We also address the important roles of signaling pathways in mediating the interaction between cancer cells and the different components of the GC TME. Finally, we discuss the implications of these molecular mechanisms for developing novel and effective therapies targeting molecular and cellular components of the GC TME to control the malignant progression of GC.
Collapse
Affiliation(s)
- Tasuku Matsuoka
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| | - Masakazu Yashiro
- Department of Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan;
- Institute of Medical Genetics, Osaka Metropolitan University, 1-4-3 Asahi-machi, Abeno-ku, Osaka 5458585, Japan
| |
Collapse
|
21
|
Thomas-Jardin S, Suresh S, Arce A, Novaresi N, Stein E, Thomas L, Lewis C, Ahn C, Evers BM, Salvatierra ME, Lui W, Khan K, Solis Soto LM, Wistuba I, Minna JD, O'Donnell KA. Coordinated translational control of multiple immune checkpoints by the integrated stress response pathway in lung cancer. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.23.619897. [PMID: 39554171 PMCID: PMC11565990 DOI: 10.1101/2024.10.23.619897] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
The integrated stress response (ISR) is an adaptive pathway hijacked by cancer cells to survive cellular stresses in the tumor microenvironment. ISR activation potently induces Programmed Death Ligand 1 (PD-L1), leading to suppression of anti-tumor immunity. Here we sought to uncover additional immune checkpoint proteins regulated by the ISR to elucidate mechanisms of tumor immune escape. We show that CD155 and PD-L1 are coordinately induced by the ISR, enhancing translation of both immune checkpoint proteins through bypass of inhibitory upstream open reading frames (uORFs) in their 5' UTRs. Analysis of primary human lung tumors identifies a significant correlation between PD-L1 and CD155 expression. ISR activation accelerates tumorigenesis and inhibits T cell function, effects that can be overcome by combining PD-1 blockade with the ISR inhibitor ISRIB. These studies uncover a novel mechanism by which two immune checkpoint proteins are coordinately regulated and suggest a new therapeutic strategy for lung cancer patients. Statement of Significance This study uncovers a novel mechanism for the coordinated translational regulation of the PD-L1/PD1 and CD155/TIGIT immune checkpoint pathways and highlights the ISR as a therapeutic vulnerability for lung cancer. Inhibition of the ISR pathway bolsters PD-1 blockade, potentially unveiling a new therapeutic strategy for lung cancer patients.
Collapse
Affiliation(s)
- Shayna Thomas-Jardin
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Shruthy Suresh
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
- CSIR-Institute of Genomics & Integrative Biology, New Delhi 110025, India
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Ariana Arce
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Nicole Novaresi
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Emily Stein
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Lisa Thomas
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
| | - Chul Ahn
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
- O'Donnell School of Public Health, UT Southwestern Medical Center, Dallas, TX, USA
| | - Bret M Evers
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Maria E Salvatierra
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center
| | - Wei Lui
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center
| | - Khaja Khan
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center
| | | | - Ignacio Wistuba
- Department of Translational Molecular Pathology, UT MD Anderson Cancer Center
| | - John D Minna
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, UT Southwestern Medical Center, Dallast, TX, USA
| | - Kathryn A O'Donnell
- Department of Molecular Biology, UT Southwestern Medical Center, Dallas, TX, USA
- Harold C. Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX
- Hamon Center for Regenerative Science and Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
22
|
Hu T, Liu CH, Lei M, Zeng Q, Li L, Tang H, Zhang N. Metabolic regulation of the immune system in health and diseases: mechanisms and interventions. Signal Transduct Target Ther 2024; 9:268. [PMID: 39379377 PMCID: PMC11461632 DOI: 10.1038/s41392-024-01954-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/18/2024] [Accepted: 08/11/2024] [Indexed: 10/10/2024] Open
Abstract
Metabolism, including glycolysis, oxidative phosphorylation, fatty acid oxidation, and other metabolic pathways, impacts the phenotypes and functions of immune cells. The metabolic regulation of the immune system is important in the pathogenesis and progression of numerous diseases, such as cancers, autoimmune diseases and metabolic diseases. The concept of immunometabolism was introduced over a decade ago to elucidate the intricate interplay between metabolism and immunity. The definition of immunometabolism has expanded from chronic low-grade inflammation in metabolic diseases to metabolic reprogramming of immune cells in various diseases. With immunometabolism being proposed and developed, the metabolic regulation of the immune system can be gradually summarized and becomes more and more clearer. In the context of many diseases including cancer, autoimmune diseases, metabolic diseases, and many other disease, metabolic reprogramming occurs in immune cells inducing proinflammatory or anti-inflammatory effects. The phenotypic and functional changes of immune cells caused by metabolic regulation further affect and development of diseases. Based on experimental results, targeting cellular metabolism of immune cells becomes a promising therapy. In this review, we focus on immune cells to introduce their metabolic pathways and metabolic reprogramming, and summarize how these metabolic pathways affect immune effects in the context of diseases. We thoroughly explore targets and treatments based on immunometabolism in existing studies. The challenges of translating experimental results into clinical applications in the field of immunometabolism are also summarized. We believe that a better understanding of immune regulation in health and diseases will improve the management of most diseases.
Collapse
Affiliation(s)
- Tengyue Hu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Chang-Hai Liu
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Min Lei
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Qingmin Zeng
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China
| | - Li Li
- Division of Renal and endocrinology, Qin Huang Hospital, Xi'an, China
| | - Hong Tang
- Center of Infectious Diseases, West China Hospital of Sichuan University, Chengdu, China.
- Laboratory of Infectious and Liver Diseases, Institution of Infectious Diseases, West China Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| | - Nannan Zhang
- West China School of clinical medical, West China Second University Hospital, Sichuan University, Chengdu, China.
- National Center for Birth Defect Monitoring, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, China.
- State Key Laboratory of Biotherapy/Collaborative Innovation Center of Biotherapy, West China Second University Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
23
|
Nader NE, Frederico SC, Miller T, Huq S, Zhang X, Kohanbash G, Hadjipanayis CG. Barriers to T Cell Functionality in the Glioblastoma Microenvironment. Cancers (Basel) 2024; 16:3273. [PMID: 39409893 PMCID: PMC11476085 DOI: 10.3390/cancers16193273] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 09/17/2024] [Accepted: 09/19/2024] [Indexed: 10/20/2024] Open
Abstract
Glioblastoma (GBM) is an aggressive primary brain tumor depicted by a cold tumor microenvironment, low immunogenicity, and limited effective therapeutic interventions. Its location in the brain, a highly immune-selective organ, acts as a barrier, limiting immune access and promoting GBM dissemination, despite therapeutic interventions. Currently, chemotherapy and radiation combined with surgical resection are the standard of care for GBM treatment. Although immune checkpoint blockade has revolutionized the treatment of solid tumors, its observed success in extracranial tumors has not translated into a significant survival benefit for GBM patients. To develop effective immunotherapies for GBM, it is vital to tailor treatments to overcome the numerous immunosuppressive barriers that inhibit T cell responses to these tumors. In this review, we address the unique physical and immunological barriers that make GBM challenging to treat. Additionally, we explore potential therapeutic mechanisms, studied in central nervous system (CNS) and non-CNS cancers, that may overcome these barriers. Furthermore, we examine current and promising immunotherapy clinical trials and immunotherapeutic interventions for GBM. By highlighting the array of challenges T cell-based therapies face in GBM, we hope this review can guide investigators as they develop future immunotherapies for this highly aggressive malignancy.
Collapse
Affiliation(s)
- Noor E. Nader
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (N.E.N.); (S.C.F.); (T.M.)
| | - Stephen C. Frederico
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (N.E.N.); (S.C.F.); (T.M.)
- Harvard Medical School, Boston, MA 02115, USA
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Tracy Miller
- School of Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA; (N.E.N.); (S.C.F.); (T.M.)
| | - Sakibul Huq
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA 15213, USA;
| | - Xiaoran Zhang
- Sloan Kettering Memorial Cancer Center, New York, NY 10065, USA;
| | - Gary Kohanbash
- Sloan Kettering Memorial Cancer Center, New York, NY 10065, USA;
| | | |
Collapse
|
24
|
Li W, Wei J, Cheng M, Liu M. Unveiling promising targets in gastric cancer therapy: A comprehensive review. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200857. [PMID: 39280587 PMCID: PMC11396074 DOI: 10.1016/j.omton.2024.200857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 09/18/2024]
Abstract
Gastric cancer (GC) poses a significant global health challenge, ranking fifth in incidence and third in mortality among all malignancies worldwide. Its insidious onset, aggressive growth, proclivity for metastasis, and limited treatment options have contributed to its high fatality rate. Traditional approaches for GC treatment primarily involve surgery and chemotherapy. However, there is growing interest in targeted therapies and immunotherapies. This comprehensive review highlights recent advancements in GC targeted therapy and immunotherapy. It delves into the mechanisms of various strategies, underscoring their potential in GC treatment. Additionally, the review evaluates the efficacy and safety of relevant clinical trials. Despite the benefits observed in numerous advanced GC patients with targeted therapies and immunotherapies, challenges persist. We discuss pertinent strategies to overcome these challenges, thereby providing a solid foundation for enhancing the clinical effectiveness of targeted therapies and immunotherapies.
Collapse
Affiliation(s)
- Wenke Li
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Jing Wei
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Mo Cheng
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| | - Ming Liu
- Gastric Cancer Center/Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan Province 610041, China
| |
Collapse
|
25
|
Xia RJ, Du XY, Shen LW, Ma JG, Xu SM, Fan RF, Qin JW, Yan L. Roles of the tumor microenvironment in the resistance to programmed cell death protein 1 inhibitors in patients with gastric cancer. World J Gastrointest Oncol 2024; 16:3820-3831. [PMID: 39350980 PMCID: PMC11438768 DOI: 10.4251/wjgo.v16.i9.3820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/21/2024] [Revised: 05/27/2024] [Accepted: 08/09/2024] [Indexed: 09/09/2024] Open
Abstract
Despite the continuous developments and advancements in the treatment of gastric cancer (GC), which is one of the most prevalent types of cancer in China, the overall survival is still poor for most patients with advanced GC. In recent years, with the progress in tumor immunology research, attention has shifted toward immunotherapy as a therapeutic approach for GC. Programmed cell death protein 1 (PD-1) inhibitors, as novel immunosuppressive medications, have been widely utilized in the treatment of GC. However, many patients are still resistant to PD-1 inhibitors and experience recurrence in the advanced stages of PD-1 immunotherapy. To reduce the occurrence of drug resistance and recurrence in GC patients receiving PD-1 immunotherapy, to maximize the clinical activity of immunosuppressive drugs, and to elicit a lasting immune response, it is essential to research the tumor microenvironment mechanisms leading to PD-1 inhibitor resistance in GC patients. This article reviews the progress in studying the factors influencing the resistance to PD-1 inhibitors in the GC tumor microenvironment, aiming to provide insights and a basis for reducing resistance to PD-1 inhibitors for GC patients in the future.
Collapse
Affiliation(s)
- Ren-Jie Xia
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Xiao-Yu Du
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
- Department of Medicine, Northwest Minzu University, Lanzhou 730050, Gansu Province, China
| | - Li-Wen Shen
- Department of Medical Support Center, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Guo Ma
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Shu-Mei Xu
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Rui-Fang Fan
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Jian-Wei Qin
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| | - Long Yan
- Department of General Surgery, The 940th Hospital of Joint Logistic Support Force of Chinese People’s Liberation Army, Lanzhou 730050, Gansu Province, China
| |
Collapse
|
26
|
Heidari-Foroozan M, Rezalotfi A, Rezaei N. The molecular landscape of T cell exhaustion in the tumor microenvironment and reinvigoration strategies. Int Rev Immunol 2024; 43:419-440. [PMID: 39257319 DOI: 10.1080/08830185.2024.2401352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 08/31/2023] [Accepted: 09/02/2024] [Indexed: 09/12/2024]
Abstract
Immunotherapy has emerged as a promising therapeutic approach for cancer treatment by harnessing the immune system to target cancer cells. However, the efficacy of immunotherapy is hindered by the tumor microenvironment (TME), comprising regulatory T cells (Tregs), macrophages, myeloid-derived suppressor cells (MDSCs), neutrophils, soluble factors (TGF-β, IL-35, IL-10), and hypoxia. These components interact with inhibitory receptors (IRs) on T cells, leading to alterations in T cell transcriptomes, epigenomes, and metabolism, ultimately resulting in T cell exhaustion and compromising the effectiveness of immunotherapy. T cell exhaustion occurs in two phases: pre-exhaustion and exhaustion. Pre-exhausted T cells exhibit reversibility and distinct molecular properties compared to terminally exhausted T cells. Understanding these differences is crucial for designing effective interventions. This comprehensive review summarizes the characteristics of pre-exhausted and exhausted T cells and elucidates the influence of TME components on T cell activity, transcriptomes, epigenomes, and metabolism, ultimately driving T cell exhaustion in cancer. Additionally, potential intervention strategies for reversing exhaustion are discussed. By gaining insights into the mechanisms underlying T cell exhaustion and the impact of the TME, this review aims to inform the development of innovative approaches for combating T cell exhaustion and enhancing the efficacy of immunotherapy in cancer treatment.
Collapse
Affiliation(s)
- Mahsa Heidari-Foroozan
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Cancer Immunology Project (CIP), Universal Scientific Education and Research Network (USERN), Tehran, Iran
| | - Alaleh Rezalotfi
- Institute of Immunology, Hannover Medical School, Hannover, Germany
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Nima Rezaei
- Department of Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center Hospital, Dr. Qarib St, Keshavarz Blvd, Tehran, Iran
- Research Center for Immunodeficiencies, Children's Medical Center, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
27
|
Tiersma JF, Evers B, Bakker BM, Reijngoud DJ, de Bruyn M, de Jong S, Jalving M. Targeting tumour metabolism in melanoma to enhance response to immune checkpoint inhibition: A balancing act. Cancer Treat Rev 2024; 129:102802. [PMID: 39029155 DOI: 10.1016/j.ctrv.2024.102802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Revised: 07/08/2024] [Accepted: 07/10/2024] [Indexed: 07/21/2024]
Abstract
Immune checkpoint inhibition has transformed the treatment landscape of advanced melanoma and long-term survival of patients is now possible. However, at least half of the patients do not benefit sufficiently. Metabolic reprogramming is a hallmark of cancer cells and may contribute to both tumour growth and immune evasion by the tumour. Preclinical studies have indeed demonstrated that modulating tumour metabolism can reduce tumour growth while improving the functionality of immune cells. Since metabolic pathways are commonly shared between immune and tumour cells, it is essential to understand how modulating tumour metabolism in patients influences the intricate balance of pro-and anti-tumour immune effects in the tumour microenvironment. The key question is whether modulating tumour metabolism can inhibit tumour cell growth as well as facilitate an anti-tumour immune response. Here, we review current knowledge on the effect of tumour metabolism on the immune response in melanoma. We summarise metabolic pathways in melanoma and non-cancerous cells in the tumour microenvironment and discuss models and techniques available to study the metabolic-immune interaction. Finally, we discuss clinical use of these techniques to improve our understanding of how metabolic interventions can tip the balance towards a favourable, immune permissive microenvironment in melanoma patients.
Collapse
Affiliation(s)
- J F Tiersma
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - B Evers
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, and Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - B M Bakker
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, and Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - D J Reijngoud
- Laboratory of Pediatrics, Section Systems Medicine of Metabolism and Signalling, and Center for Liver, Digestive and Metabolic Diseases, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - M de Bruyn
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - S de Jong
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - M Jalving
- Department of Medical Oncology, University Medical Center Groningen, University of Groningen, Groningen, The Netherlands.
| |
Collapse
|
28
|
Chong X, Madeti Y, Cai J, Li W, Cong L, Lu J, Mo L, Liu H, He S, Yu C, Zhou Z, Wang B, Cao Y, Wang Z, Shen L, Wang Y, Zhang X. Recent developments in immunotherapy for gastrointestinal tract cancers. J Hematol Oncol 2024; 17:65. [PMID: 39123202 PMCID: PMC11316403 DOI: 10.1186/s13045-024-01578-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
The past few decades have witnessed the rise of immunotherapy for Gastrointestinal (GI) tract cancers. The role of immune checkpoint inhibitors (ICIs), particularly programmed death protein 1 (PD-1) and PD ligand-1 antibodies, has become increasingly pivotal in the treatment of advanced and perioperative GI tract cancers. Currently, anti-PD-1 plus chemotherapy is considered as first-line regimen for unselected advanced gastric/gastroesophageal junction adenocarcinoma (G/GEJC), mismatch repair deficient (dMMR)/microsatellite instability-high (MSI-H) colorectal cancer (CRC), and advanced esophageal cancer (EC). In addition, the encouraging performance of claudin18.2-redirected chimeric antigen receptor T-cell (CAR-T) therapy in later-line GI tract cancers brings new hope for cell therapy in solid tumour treatment. Nevertheless, immunotherapy for GI tumour remains yet precise, and researchers are dedicated to further maximising and optimising the efficacy. This review summarises the important research, latest progress, and future directions of immunotherapy for GI tract cancers including EC, G/GEJC, and CRC.
Collapse
Affiliation(s)
- Xiaoyi Chong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yelizhati Madeti
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jieyuan Cai
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Wenfei Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Cong
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Jialin Lu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Liyang Mo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Huizhen Liu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Siyi He
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Chao Yu
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhiruo Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Boya Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Yanshuo Cao
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Zhenghang Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China
| | - Yakun Wang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| | - Xiaotian Zhang
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital and Institute, 52 Fucheng Road, Hai-Dian District, Beijing, 100142, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Beijing Key Laboratory of Carcinogenesis and Translational Research, Department of Gastrointestinal Oncology, Peking University Cancer Hospital and Institute, Beijing, 100142, China.
| |
Collapse
|
29
|
Murakami K, Ganguly S. The Nectin family ligands, PVRL2 and PVR, in cancer immunology and immunotherapy. Front Immunol 2024; 15:1441730. [PMID: 39156900 PMCID: PMC11327090 DOI: 10.3389/fimmu.2024.1441730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 07/22/2024] [Indexed: 08/20/2024] Open
Abstract
In recent years, immunotherapy has emerged as a crucial component of cancer treatment. However, its efficacy remains limited across various cancer types, highlighting unmet needs. Poliovirus receptor-related 2 (PVRL2) and Poliovirus receptor (PVR) are members of the Nectin and Nectin-like Molecules family, known for their role as cell-cell adhesion molecules. With the development of immunotherapy, their involvement in tumor immune mechanisms as immune checkpoint factors has garnered significant attention. PVRL2 and PVR are predominantly expressed on tumor cells and antigen-presenting cells, binding to PVRIG and TIGIT, respectively, which are primarily found on T and NK cells, thereby suppressing antitumor immunity. Notably, gynecological cancers such as ovarian and endometrial cancers exhibit high expression levels of PVRL2 and PVR, with similar trends observed in various other solid and hematologic tumors. Targeting these immune checkpoint pathways offers a promising therapeutic avenue, potentially in combination with existing treatments. However, the immunomodulatory mechanism involving these bindings, known as the DNAM-1 axis, is complex, underscoring the importance of understanding it for developing novel therapies. This article comprehensively reviews the immunomodulatory mechanisms centered on PVRL2 and PVR, elucidating their implications for various cancer types.
Collapse
Affiliation(s)
| | - Sudipto Ganguly
- The Bloomberg~Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| |
Collapse
|
30
|
Zhan T, Zou Y, Han Z, Tian X, Chen M, Liu J, Yang X, Zhu Q, Liu M, Chen W, Chen M, Huang X, Tan J, Liu W, Tian X. Single-cell sequencing combined with spatial transcriptomics reveals that the IRF7 gene in M1 macrophages inhibits the occurrence of pancreatic cancer by regulating lipid metabolism-related mechanisms. Clin Transl Med 2024; 14:e1799. [PMID: 39118300 PMCID: PMC11310283 DOI: 10.1002/ctm2.1799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 08/10/2024] Open
Abstract
AIM The main focus of this study is to explore the molecular mechanism of IRF7 regulation on RPS18 transcription in M1-type macrophages in pancreatic adenocarcinoma (PAAD) tissue, as well as the transfer of RPS18 by IRF7 via exosomes to PAAD cells and the regulation of ILF3 expression. METHODS By utilising single-cell RNA sequencing (scRNA-seq) data and spatial transcriptomics (ST) data from the Gene Expression Omnibus database, we identified distinct cell types with significant expression differences in PAAD tissue. Among these cell types, we identified those closely associated with lipid metabolism. The differentially expressed genes within these cell types were analysed, and target genes relevant to prognosis were identified. Flow cytometry was employed to assess the expression levels of target genes in M1 and M2 macrophages. Cell lines with target gene knockout were constructed using CRISPR/Cas9 editing technology, and cell lines with target gene knockdown and overexpression were established using lentiviral vectors. Additionally, a co-culture model of exosomes derived from M1 macrophages with PAAD cells was developed. The impact of M1 macrophage-derived exosomes on the lipid metabolism of PAAD cells in the model was evaluated through metabolomics analysis. The effects of M1 macrophage-derived exosomes on the viability, proliferation, division, migration and apoptosis of PAAD cells were assessed using MTT assay, flow cytometry, EdU assay, wound healing assay, Transwell assay and TUNEL staining. Furthermore, a mouse PAAD orthotopic implantation model was established, and bioluminescence imaging was utilised to assess the influence of M1 macrophage-derived exosomes on the intratumoural formation capacity of PAAD cells, as well as measuring tumour weight and volume. The expression of proliferation-associated proteins in tumour tissues was examined using immunohistochemistry. RESULTS Through combined analysis of scRNA-seq and ST technologies, we discovered a close association between M1 macrophages in PAAD samples and lipid metabolism signals, as well as a negative correlation between M1 macrophages and cancer cells. The construction of a prognostic risk score model identified RPS18 and IRF7 as two prognostically relevant genes in M1 macrophages, exhibiting negative and positive correlations, respectively. Mechanistically, it was found that IRF7 in M1 macrophages can inhibit the transcription of RPS18, reducing the transfer of RPS18 to PAAD cells via exosomes, consequently affecting the expression of ILF3 in PAAD cells. IRF7/RPS18 in M1 macrophages can also suppress lipid metabolism, cell viability, proliferation, migration, invasion and intratumoural formation capacity of PAAD cells, while promoting cell apoptosis. CONCLUSION Overexpression of IRF7 in M1 macrophages may inhibit RPS18 transcription, reduce the transfer of RPS18 from M1 macrophage-derived exosomes to PAAD cells, thereby suppressing ILF3 expression in PAAD cells, inhibiting the lipid metabolism pathway, and curtailing the viability, proliferation, migration, invasion of PAAD cells, as well as enhancing cell apoptosis, ultimately inhibiting tumour formation in PAAD cells in vivo. Targeting IRF7/RPS18 in M1 macrophages could represent a promising immunotherapeutic approach for PAAD in the future.
Collapse
Affiliation(s)
- Ting Zhan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Yanli Zou
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Zheng Han
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - XiaoRong Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Mengge Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jiaxi Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiulin Yang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Qingxi Zhu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Meng Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Wei Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Mingtao Chen
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xiaodong Huang
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
- Department of GastroenterologyZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Jie Tan
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Weijie Liu
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| | - Xia Tian
- Department of GastroenterologyWuHan Third Hospital (Tongren Hospital of WuHan University)WuhanChina
| |
Collapse
|
31
|
Chen H, Molberg K, Carrick K, Niu S, Rivera Colon G, Gwin K, Lewis C, Lea J, Panwar V, Zheng W, Castrillon DH, Lucas E. Expression and Prognostic Significance of LAG-3, TIGIT, VISTA, and IDO1 in Endometrial Serous Carcinoma. Mod Pathol 2024; 37:100532. [PMID: 38848896 DOI: 10.1016/j.modpat.2024.100532] [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: 01/31/2024] [Revised: 04/27/2024] [Accepted: 05/29/2024] [Indexed: 06/09/2024]
Abstract
Endometrial serous carcinoma (ESC) is an uncommon, aggressive type of endometrial cancer. While immune checkpoint blockade has emerged as a promising treatment option for endometrial carcinomas, research on the expression of immune checkpoints that could serve as prospective immunotherapy targets in ESC is limited. We examined the prevalence and prognostic value of lymphocyte-activation gene 3 (LAG-3), T-cell immunoglobulin and ITIM domain (TIGIT), V-domain immunoglobulin (Ig) suppressor of T-cell activation (VISTA), and indoleamine 2,3-dioxygenase 1 (IOD1) in 94 cases of ESC and correlated their expression with CD8+ and FOXP3+ tumor-infiltrating lymphocytes (TILs). We observed a positive correlation among LAG-3, TIGIT, and VISTA expressed on immune cells, and among these markers and CD8+ and FOXP3+ TIL densities. In Kaplan-Meier survival analysis, tumors with high levels of LAG-3 and TIGIT expression had better progression-free survival (PFS) and overall survival (OS) than those with lower levels of expression (LAG-3: PFS, P = .03, OS, P = .04; TIGIT: PFS, P = .01, OS, P = .009). In multivariate analysis, only high TIGIT expression was of independent prognostic value for better OS. VISTA expression in immune or tumor cells, and IDO1 expression in tumor cells, did not show a significant association with survival. Our data indicate that LAG-3, TIGIT, and VISTA immune checkpoints have roles in the microenvironment of ESC, and their expression patterns highlight the complex interactions among the different components of this system. High levels of these markers, together with high CD8+ TIL, suggest the potential immunogenicity of a subset of these tumors. Further studies are needed to elucidate the roles of various immune components in the ESC microenvironment and their association with intrinsic tumor properties.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Female
- Humans
- Middle Aged
- Antigens, CD/metabolism
- B7 Antigens/metabolism
- Biomarkers, Tumor/analysis
- Cystadenocarcinoma, Serous/pathology
- Cystadenocarcinoma, Serous/mortality
- Cystadenocarcinoma, Serous/immunology
- Endometrial Neoplasms/pathology
- Endometrial Neoplasms/immunology
- Endometrial Neoplasms/mortality
- Endometrial Neoplasms/genetics
- Indoleamine-Pyrrole 2,3,-Dioxygenase/biosynthesis
- Indoleamine-Pyrrole 2,3,-Dioxygenase/metabolism
- Indoleamine-Pyrrole 2,3,-Dioxygenase/analysis
- Lymphocyte Activation Gene 3 Protein
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Prognosis
- Receptors, Immunologic/metabolism
- Tumor Microenvironment/immunology
Collapse
Affiliation(s)
- Hao Chen
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Kyle Molberg
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Kelley Carrick
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Shuang Niu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Glorimar Rivera Colon
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Katja Gwin
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Cheryl Lewis
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jayanthi Lea
- Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Vandana Panwar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas
| | - Wenxin Zheng
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Diego H Castrillon
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas; Harold C. Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Obstetrics and Gynecology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Elena Lucas
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Pathology, Parkland Hospital, Dallas, Texas.
| |
Collapse
|
32
|
Szereday L, Nagy DU, Vastag F, Mezosi L, Meggyes M. Immunological Profiling of CD8 + and CD8 - NK Cell Subpopulations and Immune Checkpoint Alterations in Early-Onset Preeclampsia and Healthy Pregnancy. Int J Mol Sci 2024; 25:8378. [PMID: 39125946 PMCID: PMC11313567 DOI: 10.3390/ijms25158378] [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: 06/17/2024] [Revised: 07/25/2024] [Accepted: 07/30/2024] [Indexed: 08/12/2024] Open
Abstract
Despite the numerous studies on the clinical aspects of early-onset preeclampsia, our understanding of the immunological consequences of inadequate placenta development remains incomplete. The Th1-predominance characteristic of early-onset preeclampsia significantly impacts maternal immunotolerance, and the role of immune checkpoint molecules in these mechanisms is yet to be fully elucidated. Our study aims to fill these crucial knowledge gaps. A total of 34 pregnant women diagnosed with early-onset preeclampsia and 34 healthy pregnant women were enrolled in this study. A mononuclear cell fragment from the venous blood was separated and frozen. The CD8+ and CD8- NK cell subpopulations were identified and compared to their immune checkpoint molecule expressions using multicolor flow cytometry. The serum CD226 levels were measured by ELISA. Based on our measures, the frequency of the CD8- subpopulation was significantly higher than that of the CD8+ counterpart in both the NKdim and NKbright subsets. Significantly lower CD226 surface expressions were detected in the preeclamptic group compared to healthy women in all the investigated subpopulations. However, while no difference was observed in the level of the soluble CD226 molecule between the two groups, the CD112 and CD155 surface expressions were significantly different. Our study's findings underscore the significant role of the CD8+ and CD8- NK subpopulations in the Th1-dominated immune environment. This deepens our understanding of early-onset preeclampsia and suggests that each subpopulation could contribute to the compensation mechanisms and the restoration of the immunological balance in this condition, a crucial step toward developing effective interventions.
Collapse
Affiliation(s)
- Laszlo Szereday
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, 12 Szigeti Street, 7624 Pécs, Hungary; (L.S.); (L.M.)
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pécs, Hungary
| | - David U. Nagy
- Institute of Geobotany/Plant Ecology, Martin-Luther-University, Große Steinstraße 79/80, D-06108 Halle (Saale), Germany;
| | - Fanni Vastag
- Department of Obstetrics and Gynaecology, Medical School, University of Pécs, 7624 Pécs, Hungary;
| | - Livia Mezosi
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, 12 Szigeti Street, 7624 Pécs, Hungary; (L.S.); (L.M.)
| | - Matyas Meggyes
- Department of Medical Microbiology and Immunology, Medical School, University of Pécs, 12 Szigeti Street, 7624 Pécs, Hungary; (L.S.); (L.M.)
- Janos Szentagothai Research Centre, 20 Ifjusag Street, 7624 Pécs, Hungary
| |
Collapse
|
33
|
Yasuda T, Wang YA. Gastric cancer immunosuppressive microenvironment heterogeneity: implications for therapy development. Trends Cancer 2024; 10:627-642. [PMID: 38600020 PMCID: PMC11292672 DOI: 10.1016/j.trecan.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 03/15/2024] [Accepted: 03/18/2024] [Indexed: 04/12/2024]
Abstract
Although immunotherapy has revolutionized solid tumor treatment, durable responses in gastric cancer (GC) remain limited. The heterogeneous tumor microenvironment (TME) facilitates immune evasion, contributing to resistance to conventional and immune therapies. Recent studies have highlighted how specific TME components in GC acquire immune escape capabilities through cancer-specific factors. Understanding the underlying molecular mechanisms and targeting the immunosuppressive TME will enhance immunotherapy efficacy and patient outcomes. This review summarizes recent advances in GC TME research and explores the role of the immune-suppressive system as a context-specific determinant. We also provide insights into potential treatments beyond checkpoint inhibition.
Collapse
Affiliation(s)
- Tadahito Yasuda
- Brown Center for Immunotherapy, Department of Medicine, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Y Alan Wang
- Brown Center for Immunotherapy, Department of Medicine, Melvin and Bren Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| |
Collapse
|
34
|
Shi H, Chen S, Chi H. Immunometabolism of CD8 + T cell differentiation in cancer. Trends Cancer 2024; 10:610-626. [PMID: 38693002 PMCID: PMC11342304 DOI: 10.1016/j.trecan.2024.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/19/2024] [Accepted: 03/21/2024] [Indexed: 05/03/2024]
Abstract
CD8+ cytotoxic T lymphocytes (CTLs) are central mediators of tumor immunity and immunotherapies. Upon tumor antigen recognition, CTLs differentiate from naive/memory-like toward terminally exhausted populations with more limited function against tumors. Such differentiation is regulated by both immune signals, including T cell receptors (TCRs), co-stimulation, and cytokines, and metabolism-associated processes. These immune signals shape the metabolic landscape via signaling, transcriptional and post-transcriptional mechanisms, while metabolic processes in turn exert spatiotemporal effects to modulate the strength and duration of immune signaling. Here, we review the bidirectional regulation between immune signals and metabolic processes, including nutrient uptake and intracellular metabolic pathways, in shaping CTL differentiation and exhaustion. We also discuss the mechanisms underlying how specific nutrient sources and metabolite-mediated signaling events orchestrate CTL biology. Understanding how metabolic programs and their interplay with immune signals instruct CTL differentiation and exhaustion is crucial to uncover tumor-immune interactions and design novel immunotherapies.
Collapse
Affiliation(s)
- Hao Shi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA
| | - Sidi Chen
- Department of Genetics, Yale University School of Medicine, New Haven, CT, USA; System Biology Institute, Integrated Science & Technology Center, West Haven, CT, USA.
| | - Hongbo Chi
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, USA.
| |
Collapse
|
35
|
Yan ZX, Dong Y, Qiao N, Zhang YL, Wu W, Zhu Y, Wang L, Cheng S, Xu PP, Zhou ZS, Sheng LS, Zhao WL. Cholesterol efflux from C1QB-expressing macrophages is associated with resistance to chimeric antigen receptor T cell therapy in primary refractory diffuse large B cell lymphoma. Nat Commun 2024; 15:5183. [PMID: 38890370 PMCID: PMC11189439 DOI: 10.1038/s41467-024-49495-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Accepted: 06/03/2024] [Indexed: 06/20/2024] Open
Abstract
Chimeric antigen receptor T (CAR-T) cell therapy has demonstrated promising efficacy in early trials for relapsed/refractory diffuse large B cell lymphoma (DLBCL). However, its efficacy in treating primary refractory DLBCL has not been comprehensively investigated, and the underlying resistance mechanisms remain unclear. Here, we report the outcomes of a phase I, open-label, single-arm clinical trial of relmacabtagene autoleucel (relma-cel), a CD19-targeted CAR-T cell product, with safety and efficacy as primary endpoints. Among the 12 enrolled patients, 8 experienced grade 4 hematologic toxicity of treatment-emergent adverse event. No grade ≥3 cytokine release syndrome or neurotoxicity occurred. Single-cell RNA sequencing revealed an increase proportion of C1QB-expressing macrophages in patients with progressive disease before CAR-T cell therapy. Cholesterol efflux from M2 macrophages was found to inhibit CAR-T cells cytotoxicity by inducing an immunosuppressive state in CD8+ T cells, leading to their exhaustion. Possible interactions between macrophages and CD8+ T cells, mediating lipid metabolism (AFR1-FAS), immune checkpoint activation, and T cell exhaustion (LGALS9-HAVCR2, CD86-CTLA4, and NECTIN2-TIGIT) were enhanced during disease progression. These findings suggest that cholesterol efflux from macrophages may trigger CD8+ T cell exhaustion, providing a rationale for metabolic reprogramming to counteract CAR-T treatment failure. Chinadrugtrials.org.cn identifier: CTR20200376.
Collapse
MESH Headings
- Humans
- Lymphoma, Large B-Cell, Diffuse/therapy
- Lymphoma, Large B-Cell, Diffuse/immunology
- Lymphoma, Large B-Cell, Diffuse/metabolism
- Lymphoma, Large B-Cell, Diffuse/genetics
- Macrophages/metabolism
- Macrophages/immunology
- Immunotherapy, Adoptive/methods
- Middle Aged
- Female
- Male
- Cholesterol/metabolism
- Receptors, Chimeric Antigen/metabolism
- Receptors, Chimeric Antigen/immunology
- Receptors, Chimeric Antigen/genetics
- Aged
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Adult
- Drug Resistance, Neoplasm
Collapse
Affiliation(s)
- Zi-Xun Yan
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yan Dong
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Niu Qiao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yi-Lun Zhang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Wen Wu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yue Zhu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Li Wang
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shu Cheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Peng-Peng Xu
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zi-Song Zhou
- JW Therapeutics (Shanghai) Co. Ltd, Shanghai, 200025, China
| | - Ling-Shuang Sheng
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wei-Li Zhao
- Shanghai Institute of Hematology, State Key Laboratory of Medical Genomics, National Research Center for Translational Medicine at Shanghai, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
- Pôle de Recherches Sino-Français en Science du Vivant et Génomique, Laboratory of Molecular Pathology, Shanghai, 200025, China.
| |
Collapse
|
36
|
Yin N, Li X, Zhang X, Xue S, Cao Y, Niedermann G, Lu Y, Xue J. Development of pharmacological immunoregulatory anti-cancer therapeutics: current mechanistic studies and clinical opportunities. Signal Transduct Target Ther 2024; 9:126. [PMID: 38773064 PMCID: PMC11109181 DOI: 10.1038/s41392-024-01826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
Immunotherapy represented by anti-PD-(L)1 and anti-CTLA-4 inhibitors has revolutionized cancer treatment, but challenges related to resistance and toxicity still remain. Due to the advancement of immuno-oncology, an increasing number of novel immunoregulatory targets and mechanisms are being revealed, with relevant therapies promising to improve clinical immunotherapy in the foreseeable future. Therefore, comprehending the larger picture is important. In this review, we analyze and summarize the current landscape of preclinical and translational mechanistic research, drug development, and clinical trials that brought about next-generation pharmacological immunoregulatory anti-cancer agents and drug candidates beyond classical immune checkpoint inhibitors. Along with further clarification of cancer immunobiology and advances in antibody engineering, agents targeting additional inhibitory immune checkpoints, including LAG-3, TIM-3, TIGIT, CD47, and B7 family members are becoming an important part of cancer immunotherapy research and discovery, as are structurally and functionally optimized novel anti-PD-(L)1 and anti-CTLA-4 agents and agonists of co-stimulatory molecules of T cells. Exemplified by bispecific T cell engagers, newly emerging bi-specific and multi-specific antibodies targeting immunoregulatory molecules can provide considerable clinical benefits. Next-generation agents also include immune epigenetic drugs and cytokine-based therapeutics. Cell therapies, cancer vaccines, and oncolytic viruses are not covered in this review. This comprehensive review might aid in further development and the fastest possible clinical adoption of effective immuno-oncology modalities for the benefit of patients.
Collapse
Affiliation(s)
- Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xintong Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Shaolong Xue
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, PR China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
- Institute of Disaster Medicine & Institute of Emergency Medicine, Sichuan University, No. 17, Gaopeng Avenue, Chengdu, 610041, Sichuan, PR China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site DKTK-Freiburg, Robert-Koch-Strasse 3, 79106, Freiburg, Germany.
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
37
|
Wu JW, Liu Y, Dai XJ, Liu HM, Zheng YC, Liu HM. CD155 as an emerging target in tumor immunotherapy. Int Immunopharmacol 2024; 131:111896. [PMID: 38518596 DOI: 10.1016/j.intimp.2024.111896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/08/2024] [Accepted: 03/16/2024] [Indexed: 03/24/2024]
Abstract
CD155 is an immunoglobulin-like protein overexpressed in almost all the tumor cells, which not only promotes proliferation, adhesion, invasion, and migration of tumor cells, but also regulates immune responses by interacting with TIGIT, CD226 or CD96 receptors expressed on several immune cells, thereby modulating the functionality of these cellular subsets. As a novel immune checkpoint, the inhibition of CD155/TIGIT, either as a standalone treatment or in conjunction with other immune checkpoint inhibitors, has demonstrated efficacy in managing advanced solid malignancies. In this review, we summarize the intricate relationship between on tumor surface CD155 and its receptors, with further discussion on how they regulate the occurrence of tumor immune escape. In addition, novel therapeutic strategies and clinical trials targeting CD155 and its receptors are summarized, providing a strong rationale and way forward for the development of next-generation immunotherapies.
Collapse
Affiliation(s)
- Jiang-Wan Wu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Ying Liu
- Henan Engineering Research Center for Application & Translation of Precision Clinical Pharmacy, Department of Pharmacy, The First Affiliated Hospital of Zhengzhou University, 1 Jianshe East Road, Zhengzhou 450052, China
| | - Xing-Jie Dai
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Hong-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China
| | - Yi-Chao Zheng
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| | - Hui-Min Liu
- State Key Laboratory of Esophageal Cancer Prevention & Treatment, Key Laboratory of Advanced Drug Preparation Technologies, Ministry of Education of China, Key Laboratory of Henan Province for Drug Quality and Evaluation, XNA Platform, School of Pharmaceutical Sciences, Zhengzhou University, 100 Kexue Avenue, Zhengzhou, Henan 450001, China.
| |
Collapse
|
38
|
Jiang S, Wang W, Yang Y. TIGIT: A potential immunotherapy target for gynecological cancers. Pathol Res Pract 2024; 255:155202. [PMID: 38367600 DOI: 10.1016/j.prp.2024.155202] [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: 10/23/2023] [Revised: 01/23/2024] [Accepted: 02/05/2024] [Indexed: 02/19/2024]
Abstract
Gynecological cancer represents a significant global health challenge, and conventional treatment modalities have demonstrated limited efficacy. However, recent investigations into immune checkpoint pathways have unveiled promising opportunities for enhancing the prognosis of patients with cancer. Among these pathways, TIGIT has surfaced as a compelling candidate owing to its capacity to augment the immune function of NK and T cells through blockade, thereby yielding improved anti-tumor effects and prolonged patient survival. Global clinical trials exploring TIGIT blockade therapy have yielded promising preliminary findings. Nevertheless, further research is imperative to comprehensively grasp the potential of TIGIT-based immunotherapy in optimizing therapeutic outcomes for gynecological cancers. This review primarily delineates the regulatory network and immunosuppressive mechanism of TIGIT, expounds upon its expression and therapeutic potential in three major gynecological cancers, and synthesizes the clinical trials of TIGIT-based cancer immunotherapy. Such insights aim to furnish novel perspectives and serve as reference points for subsequent research and clinical application targeting TIGIT in gynecological cancers.
Collapse
Affiliation(s)
- Siyue Jiang
- The third People's Hospital of Suining, Suining, Sichuan, China
| | - Wenhua Wang
- First Clinical Medical College of Lanzhou University, Lanzhou, Gansu, China
| | - Yongxiu Yang
- Department of Obstetrics and Gynecology, First Hospital of Lanzhou University, Key Laboratory of Gynecologic Oncology Gansu Province, Lanzhou, Gansu, China.
| |
Collapse
|
39
|
Xie W, Yu S, Hou NY, Yan L, Cao QX, Dan ZJ, Yuan XM, Lu HJ, Liu J, Pang MH. Relationship between expression of CD155 and TIGIT and clinicopathological features in gastrointestinal neuroendocrine tumors. WORLD CHINESE JOURNAL OF DIGESTOLOGY 2024; 32:123-133. [DOI: 10.11569/wcjd.v32.i2.123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/26/2024]
|
40
|
Li C, Liu H, Duan Z. Expression of the immune checkpoint molecules CD226 and TIGIT in preeclampsia patients. BMC Immunol 2024; 25:12. [PMID: 38326745 PMCID: PMC10848455 DOI: 10.1186/s12865-024-00603-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 01/30/2024] [Indexed: 02/09/2024] Open
Abstract
BACKGROUND Imbalanced immune responses are involved in developing preeclampsia (PE). We wish to explore the expression and potential changes of immune checkpoint molecules TIGIT, CD226 and CD155 in PE patients. METHODS The expression of the immune checkpoint molecules TIGIT, CD226 and CD155 in different lymphocyte subpopulations was determined by flow cytometry in 24 patients with PE and compared to 24 healthy pregnant women of the same gestational age as the controls.Serum CD155 was detected by ELISA in the patients with PE compared to controls. RESULTS The percentages of CD4+ and CD8+ T lymphocytes in the peripheral blood of PE patients were not significantly different from those of the controls, whereas the regulatory T cells (Tregs) in PE patients were significantly lower than those in controls (6.43 ± 1.77% vs. 7.48 ± 1.71%, P = 0.0420). The expression of TIGIT and CD226 showed different percentages on CD4+ T cells, CD8+ T cells and Treg cells. However, the difference in the percentages of TIGIT, CD226 on these T cells between the two groups was not statistically significant. The level of CD155 in peripheral serum of PE patients was 6.64 ± 1.79 ng/ml, which was not significantly different from that in the control group 5.61 ± 1.77 ng/ml, P = 0.0505. The present results demonstrate that TIGIT, CD226 and CD155 are not present at altered immune conditions in the peripheral blood of patients with PE, compared with normal pregnant women. CONCLUSION The immune checkpoint molecules TIGIT, CD226 and CD155 are not abnormally expressed in PE patients.
Collapse
Affiliation(s)
- Cui Li
- Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, China
| | - Haiyan Liu
- Obstetrics Department, Obstetrics and Gynecology Hospital of Fudan University, Shanghai, China
| | - Zhongliang Duan
- Clinical Laboratory, Obstetrics and Gynecology Hospital of Fudan University, 419 Fangxie Road, Shanghai, 200011, China.
| |
Collapse
|
41
|
Zhang Q, Yang C, Gao X, Dong J, Zhong C. Phytochemicals in regulating PD-1/PD-L1 and immune checkpoint blockade therapy. Phytother Res 2024; 38:776-796. [PMID: 38050789 DOI: 10.1002/ptr.8082] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Revised: 10/27/2023] [Accepted: 11/12/2023] [Indexed: 12/06/2023]
Abstract
Clinical treatment and preclinical studies have highlighted the role of immune checkpoint blockade in cancer treatment. Research has been devoted to developing immune checkpoint inhibitors in combination with other drugs to achieve better efficacy or reduce adverse effects. Phytochemicals sourced from vegetables and fruits have demonstrated antiproliferative, proapoptotic, anti-migratory, and antiangiogenic effects against several cancers. Phytochemicals also modulate the tumor microenvironment such as T cells, regulatory T cells, and cytokines. Recently, several phytochemicals have been reported to modulate immune checkpoint proteins in in vivo or in vitro models. Phytochemicals decreased programmed cell death ligand-1 expression and synergized programmed cell death receptor 1 (PD-1) monoclonal antibody to suppress tumor growth. Combined administration of phytochemicals and PD-1 monoclonal antibody enhanced the tumor growth inhibition as well as CD4+ /CD8+ T-cell infiltration. In this review, we discuss immune checkpoint molecules as potential therapeutic targets of cancers. We further assess the impact of phytochemicals including carotenoids, polyphenols, saponins, and organosulfur compounds on cancer PD-1/programmed cell death ligand-1 immune checkpoint molecules and document their combination effects with immune checkpoint inhibitors on various malignancies.
Collapse
Affiliation(s)
- Qi Zhang
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenying Yang
- Yinzhou Center for Disease Control and Prevention, Ningbo, China
| | - Xingsu Gao
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ju Dong
- Department of Public Health, School of Medicine & Holistic Integrative Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Caiyun Zhong
- Department of Nutrition and Food Safety, School of Public Health, Nanjing Medical University, Nanjing, China
| |
Collapse
|
42
|
Hoffman-Censits J, Grivas P, Powles T, Hawley J, Tyroller K, Seeberger S, Guenther S, Jacob N, Mehr KT, Hahn NM. The JAVELIN Bladder Medley trial: avelumab-based combinations as first-line maintenance in advanced urothelial carcinoma. Future Oncol 2024; 20:179-190. [PMID: 37671748 DOI: 10.2217/fon-2023-0492] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/07/2023] Open
Abstract
Results from JAVELIN Bladder 100 established avelumab (anti-PD-L1) first-line maintenance as the standard-of-care treatment for patients with advanced urothelial carcinoma (UC) that has not progressed with first-line platinum-based chemotherapy. We describe the design of JAVELIN Bladder Medley (NCT05327530), an ongoing phase II, multicenter, randomized, open-label, parallel-arm, umbrella trial. Overall, 252 patients with advanced UC who are progression-free following first-line platinum-based chemotherapy will be randomized 1:2:2:2 to receive maintenance therapy with avelumab alone (control group) or combined with sacituzumab govitecan (anti-Trop-2/topoisomerase inhibitor conjugate), M6223 (anti-TIGIT) or NKTR-255 (recombinant human IL-15). Primary end points are progression-free survival per investigator and safety/tolerability of the combination regimens. Secondary end points include overall survival, objective response and duration of response per investigator, and pharmacokinetics.
Collapse
Affiliation(s)
- Jean Hoffman-Censits
- Departments of Medical Oncology & Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| | - Petros Grivas
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Thomas Powles
- Department of Genitourinary Oncology, Barts Cancer Institute, Experimental Cancer Medicine Centre, Queen Mary University of London, St Bartholomew's Hospital, London, UK
| | - Jessica Hawley
- University of Washington, Fred Hutchinson Cancer Center, Seattle, WA, USA
| | - Karin Tyroller
- EMD Serono Research & Development Institute, Inc., Billerica, MA, USA, an affiliate of Merck KGaA
| | | | | | | | | | - Noah M Hahn
- Departments of Medical Oncology & Urology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Johns Hopkins Medical Institutions, Baltimore, MD, USA
| |
Collapse
|
43
|
Zhang P, Liu X, Gu Z, Jiang Z, Zhao S, Song Y, Yu J. Targeting TIGIT for cancer immunotherapy: recent advances and future directions. Biomark Res 2024; 12:7. [PMID: 38229100 PMCID: PMC10790541 DOI: 10.1186/s40364-023-00543-z] [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: 09/28/2023] [Accepted: 11/08/2023] [Indexed: 01/18/2024] Open
Abstract
As a newly identified checkpoint, T cell immunoreceptor with immunoglobulin and tyrosine-based inhibitory motif (ITIM) domain (TIGIT) is highly expressed on CD4+ T cells, CD8+ T cells, natural killer (NK) cells, regulatory T cells (Tregs), and tumor-infiltrating lymphocytes (TILs). TIGIT has been associated with NK cell exhaustion in vivo and in individuals with various cancers. It not only modulates NK cell survival but also mediates T cell exhaustion. As the primary ligand of TIGIT in humans, CD155 may be the main target for immunotherapy due to its interaction with TIGIT. It has been found that the anti-programmed cell death protein 1 (PD-1) treatment response in cancer immunotherapy is correlated with CD155 but not TIGIT. Anti-TIGIT alone and in combination with anti-PD-1 agents have been tested for cancer immunotherapy. Although two clinical studies on advanced lung cancer had positive results, the TIGIT-targeted antibody, tiragolumab, recently failed in two new trials. In this review, we highlight the current developments on TIGIT for cancer immunotherapy and discuss the characteristics and functions of TIGIT.
Collapse
Affiliation(s)
- Peng Zhang
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Medical Key Laboratory of Thoracic Oncology, Zhengzhou, 450052, Henan, China
| | - Xinyuan Liu
- Institute of Biomedical Informatics, Bioinformatics Center, Henan Provincial Engineering Center for Tumor Molecular Medicine, School of Basic Medical Sciences, Henan University, Kaifeng, 475004, Henan, China
| | - Zhuoyu Gu
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
- Henan Medical Key Laboratory of Thoracic Oncology, Zhengzhou, 450052, Henan, China
| | - Zhongxing Jiang
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Song Zhao
- Department of Thoracic Surgery, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Yongping Song
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
| | - Jifeng Yu
- Department of Hematology, the First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China.
- Henan International Joint Laboratory of Nuclear Protein Gene Regulation, Henan University College of Medicine, Kaifeng, 475004, Henan, China.
| |
Collapse
|
44
|
Rong Y, Liu SH, Tang MZ, Wu ZH, Ma GR, Li XF, Cai H. Analysis of the potential biological value of pyruvate dehydrogenase E1 subunit β in human cancer. World J Gastrointest Oncol 2024; 16:144-181. [PMID: 38292838 PMCID: PMC10824119 DOI: 10.4251/wjgo.v16.i1.144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/28/2023] [Accepted: 12/01/2023] [Indexed: 01/11/2024] Open
Abstract
BACKGROUND The pyruvate dehydrogenase E1 subunit β (PDHB) gene which regulates energy metabolism is located in mitochondria. However, few studies have elucidated the role and mechanism of PDHB in different cancers. AIM To comprehensive pan-cancer analysis of PDHB was performed based on bioinformatics approaches to explore its tumor diagnostic and prognostic value and tumor immune relevance in cancer. In vitro experiments were performed to examine the biological regulation of PDHB in liver cancer. METHODS Pan-cancer data related to PDHB were obtained from the Cancer Genome Atlas (TCGA) database. Analysis of the gene expression profiles of PDHB was based on TCGA and Genotype Tissue Expression Dataset databases. Cox regression analysis and Kaplan-Meier methods were used to assess the correlation between PDHB expression and survival prognosis in cancer patients. The correlation between PDHB and receiver operating characteristic diagnostic curve, clinicopathological staging, somatic mutation, tumor mutation burden (TMB), microsatellite instability (MSI), DNA methylation, and drug susceptibility in pan-cancer was also analyzed. Various algorithms were used to analyze the correlation between PDHB and immune cell infiltration and tumor chemotaxis environment, as well as the co-expression analysis of PDHB and immune checkpoint (ICP) genes. The expression and functional phenotype of PDHB in single tumor cells were studied by single-cell sequencing, and the functional enrichment analysis of PDHB-related genes was performed. The study also validated the level of mRNA or protein expression of PDHB in several cancers. Finally, in vitro experiments verified the regulatory effect of PDHB on the proliferation, migration, and invasion of liver cancer. RESULTS PDHB was significantly and differently expressed in most cancers. PDHB was significantly associated with prognosis in patients with a wide range of cancers, including kidney renal clear cell carcinoma, kidney renal papillary cell carcinoma, breast invasive carcinoma, and brain lower grade glioma. In some cancers, PDHB expression was clearly associated with gene mutations, clinicopathological stages, and expression of TMB, MSI, and ICP genes. The expression of PDHB was closely related to the infiltration of multiple immune cells in the immune microenvironment and the regulation of tumor chemotaxis environment. In addition, single-cell sequencing results showed that PDHB correlated with different biological phenotypes of multiple cancer single cells. This study further demonstrated that down-regulation of PDHB expression inhibited the proliferation, migration, and invasion functions of hepatoma cells. CONCLUSION As a member of pan-cancer, PDHB may be a novel cancer marker with potential value in diagnosing cancer, predicting prognosis, and in targeted therapy.
Collapse
Affiliation(s)
- Yao Rong
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Song-Hua Liu
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Ming-Zheng Tang
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| | - Zhi-Hang Wu
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Guo-Rong Ma
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Xiao-Feng Li
- First Clinical Medical College, Gansu University of Chinese Medicine, Lanzhou 730000, Gansu Province, China
| | - Hui Cai
- General Surgery Clinical Medical Center, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- Key Laboratory of Molecular Diagnostics and Precision Medicine for Surgical Oncology in Gansu Province, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
- NHC Key Laboratory of Diagnosis and Therapy of Gastrointestinal Tumor, Gansu Provincial Hospital, Lanzhou 730000, Gansu Province, China
| |
Collapse
|
45
|
Huang M, Yu X, Wang Q, Jiang Z, Li X, Chen W, Song C. The immune checkpoint TIGIT/CD155 promotes the exhaustion of CD8 + T cells in TNBC through glucose metabolic reprogramming mediated by PI3K/AKT/mTOR signaling. Cell Commun Signal 2024; 22:35. [PMID: 38216949 PMCID: PMC10785424 DOI: 10.1186/s12964-023-01455-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 12/21/2023] [Indexed: 01/14/2024] Open
Abstract
OBJECTIVE The CD155/TIGIT axis has attracted considerable interest as an emerging immune checkpoint with potential applications in cancer immunotherapy. Our research focused on investigating the role of CD155/TIGIT checkpoints in the progression of triple-negative breast cancer (TNBC). METHODS We evaluated CD155 and TIGIT expression in TNBC tissues using both immunohistochemistry (IHC) and gene expression profiling. Our experiments, both in vivo and in vitro, provided evidence that inhibiting the CD155/TIGIT pathway reinstates the ability of CD8 + T cells to generate cytokines. To assess the impact of CD155/TIGIT signaling blockade, we utilized Glucose Assay Kits and Lactate Assay Kits to measure alterations in glucose and lactate levels within CD8 + T cells. We employed western blotting (WB) to investigate alterations in glycolytic-related proteins within the PI3K/AKT/mTOR pathways following the inhibition of CD155/TIGIT signaling. RESULTS CD155 exhibits heightened expression within TNBC tissues and exhibits a negative correlation with the extent of infiltrating CD8 + T cells. Furthermore, patients with TNBC demonstrate elevated levels of TIGIT expression. Our findings indicate that the interaction between CD155 and TIGIT disrupts the glucose metabolism of CD8 + T cells by suppressing the activation of the PI3K/AKT/mTOR signaling pathway, ultimately leading to the reduced production of cytokines by CD8 + T cells. Both in vivo and in vitro experiments have conclusively demonstrated that the inhibition of CD155/TIGIT interaction reinstates the capacity of CD8 + T cells to generate cytokines. Moreover, in vivo administration of the blocking antibody against TIGIT not only inhibits tumor growth but also augments the functionality of CD8 + T lymphocytes. CONCLUSIONS Our research findings strongly suggest that CD155/TIGIT represents a promising therapeutic target for treating TNBC.
Collapse
Affiliation(s)
- Mingyao Huang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350011, China
| | - Xiaoqin Yu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China
| | - Qing Wang
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350011, China
| | - Zirong Jiang
- Department of Thyroid and Breast Surgery, Ningde Municipal Hospital of Ningde Normal University, Ningde, 352100, China
| | - Xiaofen Li
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350011, China
| | - Wei Chen
- Department of Oncology Surgery, Fujian Provincial Hospital, Fuzhou, 350001, China
| | - Chuangui Song
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350011, China.
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, 350001, China.
| |
Collapse
|
46
|
Park D, Jeon WJ, Yang C, Castillo DR. Advancing Esophageal Cancer Treatment: Immunotherapy in Neoadjuvant and Adjuvant Settings. Cancers (Basel) 2024; 16:318. [PMID: 38254805 PMCID: PMC10813716 DOI: 10.3390/cancers16020318] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 01/03/2024] [Accepted: 01/08/2024] [Indexed: 01/24/2024] Open
Abstract
Locally advanced esophageal cancer (LAEC) poses a significant and persistent challenge in terms of effective treatment. Traditionally, the primary strategy for managing LAEC has involved concurrent neoadjuvant chemoradiation followed by surgery. However, achieving a pathologic complete response (pCR) has proven to be inconsistent, and despite treatment, roughly half of patients experience locoregional recurrence or metastasis. Consequently, there has been a paradigm shift towards exploring the potential of immunotherapy in reshaping the landscape of LAEC management. Recent research has particularly focused on immune checkpoint inhibitors, investigating their application in both neoadjuvant and adjuvant settings. These inhibitors, designed to block specific proteins in immune cells, are meant to enhance the immune system's ability to target and combat cancer cells. Emerging evidence from these studies suggests the possibility of a mortality benefit, indicating that immunotherapy may contribute to improved overall survival rates for individuals grappling with esophageal cancer. This manuscript aims to meticulously review the existing literature surrounding neoadjuvant and adjuvant immunotherapy in the context of LAEC management. The intention is to thoroughly examine the methodologies and findings of relevant studies, providing a comprehensive synthesis of the current understanding of the impact of immunotherapy on esophageal cancer.
Collapse
Affiliation(s)
- Daniel Park
- University of California, San Francisco-Fresno Branch Campus, Fresno, CA 93701, USA;
| | - Won Jin Jeon
- Loma Linda University Medical Center, Loma Linda, CA 92354, USA;
| | - Chieh Yang
- Department of Internal Medicine for UCSF, University of California, and UC Riverside, Riverside, CA 92521, USA;
| | - Dani Ran Castillo
- City of Hope-Duarte, Department of Hematology & Oncology, Duarte, CA 91010, USA
| |
Collapse
|
47
|
Chen R, Feng C, Chen L, Zheng X, Fang W, Wu S, Gao X, Chen C, Yang J, Wu Y, Chen Y, Zheng P, Hu N, Yuan M, Fu Y, Ying H, Zhou J, Jiang J. Single-cell RNA sequencing indicates cordycepin remodels the tumor immune microenvironment to enhance TIGIT blockade's anti-tumor effect in colon cancer. Int Immunopharmacol 2024; 126:111268. [PMID: 37992442 DOI: 10.1016/j.intimp.2023.111268] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2023] [Revised: 11/16/2023] [Accepted: 11/16/2023] [Indexed: 11/24/2023]
Abstract
Both preclinical and clinical studies have extensively proven the effectiveness of TIGIT inhibitors in tumor immunotherapy. However, it has been discovered that the presence of CD226 on tumor-infiltrating lymphocytes is crucial for the effectiveness of both anti-TIGIT therapy alone and when combined with anti-PD-1 therapy for tumors. In our investigation, we observed that cordycepin therapy significantly augmented the expression of the Cd226 gene. As a result, it was hypothesized that cordycepin therapy could enhance the effectiveness of anti-TIGIT therapy. By employing single-cell RNA sequencing analysis of immune cells in the MC38 tumor model, we discovered that cordycepin combined with anti-TIGIT therapy led to a significant increase in the proportion of NK cells within the tumor immune microenvironment. This increased NK cell activity and decreased the expression of inhibitory receptors and exhaustion marker genes. In the combination therapy group, CD8+ T cells had lower exhaustion state scores and increased cytotoxicity, indicating a better immune response. The combination therapy group increased DCs in the tumor immune microenvironment and promoted cellular interaction with CD4+ T cell and CD8+ T cell populations while decreasing Treg cell interactions. In conclusion, cordycepin with anti-TIGIT therapy in colon cancer could reshape the tumor immune microenvironment and have notable anticancer effects.
Collapse
Affiliation(s)
- Rongzhang Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Chen Feng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Weiwei Fang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Xinran Gao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Can Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Jiayi Yang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Yue Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Yuanyuan Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Panpan Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Nan Hu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Maoling Yuan
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Yuanyuan Fu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Department of Gynecology, Changzhou Traditional Chinese Medicine Hospital, Changzhou, China.
| | - Hanjie Ying
- College of Biotechnology and Pharmaceutical Engineering, Nanjing Tech University, Nanjing, Jiangsu 211816, China.
| | - Jun Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China; Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.
| |
Collapse
|
48
|
Zhang S, Zhang X, Yang H, Liang T, Bai X. Hurdle or thruster: Glucose metabolism of T cells in anti-tumour immunity. Biochim Biophys Acta Rev Cancer 2024; 1879:189022. [PMID: 37993001 DOI: 10.1016/j.bbcan.2023.189022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 09/11/2023] [Accepted: 10/08/2023] [Indexed: 11/24/2023]
Abstract
Glucose metabolism is essential for the activation, differentiation and function of T cells and proper glucose metabolism is required to maintain effective T cell immunity. Dysregulation of glucose metabolism is a hallmark of cancer, and the tumour microenvironment (TME2) can create metabolic barriers in T cells that inhibit their anti-tumour immune function. Targeting glucose metabolism is a promising approach to improve the capacity of T cells in the TME. The efficacy of common immunotherapies, such as immune checkpoint inhibitors (ICIs3) and adoptive cell transfer (ACT4), can be limited by T-cell function, and the treatment itself can affect T-cell metabolism. Therefore, understanding the relationship between immunotherapy and T cell glucose metabolism helps to achieve more effective anti-tumour therapy. In this review, we provide an overview of T cell glucose metabolism and how T cell metabolic reprogramming in the TME regulates anti-tumour responses, briefly describe the metabolic patterns of T cells during ICI and ACT therapies, which suggest possible synergistic strategies.
Collapse
Affiliation(s)
- Sirui Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Xiaozhen Zhang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Hanshen Yang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| | - Xueli Bai
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China; Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, Zhejiang, China; Cancer Center, Zhejiang University, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
49
|
Shang Z, Ma Z, Wu E, Chen X, Tuo B, Li T, Liu X. Effect of metabolic reprogramming on the immune microenvironment in gastric cancer. Biomed Pharmacother 2024; 170:116030. [PMID: 38128177 DOI: 10.1016/j.biopha.2023.116030] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/03/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023] Open
Abstract
Gastric cancer (GC) is a malignant tumor of the gastrointestinal tract with a high mortality rate worldwide, a low early detection rate and a poor prognosis. The rise of metabolomics has facilitated the early detection and treatment of GC. Metabolism in the GC tumor microenvironment (TME) mainly includes glucose metabolism, lipid metabolism and amino acid metabolism, which provide energy and nutrients for GC cell proliferation and migration. Abnormal tumor metabolism can influence tumor progression by regulating the functions of immune cells and immune molecules in the TME, thereby contributing to tumor immune escape. Thus, in this review, we summarize the impact of metabolism on the TME during GC progression. We also propose novel strategies to modulate antitumor immune responses by targeting metabolism.
Collapse
Affiliation(s)
- Zhengye Shang
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Zhiyuan Ma
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Enqin Wu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Xingzhao Chen
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Biguang Tuo
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China
| | - Taolang Li
- Department of General Surgery, Affiliated Hospital of Zunyi Medical University, Dalian Road 149, Zunyi 563000, China.
| | - Xuemei Liu
- Department of Gastroenterology, Digestive Disease Hospital, Affiliated Hospital of Zunyi Medical University, Zunyi, Guizhou Province, China.
| |
Collapse
|
50
|
Spehner L, Orillard E, Falcoz A, Lepiller Q, Bouard A, Almotlak H, Kim S, Curtit E, Meynard G, Jary M, Nardin C, Asgarov K, Abdeljaoued S, Chartral U, Mougey V, Ben Khelil M, Lopez M, Loyon R, Vernerey D, Adotevi O, Borg C, Mansi L, Kroemer M. Predictive biomarkers and specific immune responses of COVID-19 mRNA vaccine in patients with cancer: prospective results from the CACOV-VAC trial. BMJ ONCOLOGY 2023; 2:e000054. [PMID: 39886486 PMCID: PMC11235023 DOI: 10.1136/bmjonc-2023-000054] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 10/17/2023] [Indexed: 02/01/2025]
Abstract
Objective Vaccinated patients with cancer in follow-up studies showed a high seropositivity rate but impaired antibody titres and T cell responses following mRNA vaccine against COVID-19. Besides clinical characteristics and the type of anticancer treatment before vaccination, the identification of patients susceptible to non-response following vaccination using immunological markers is worth to be investigated. Methods and analysis All patients (n=138, solid cancers) were included in the CACOV-VAC Study comprising three cohorts ((neo)-adjuvant, metastatic and surveillance). Immune responses were assessed using, respectively, anti-receptor-binding domain (RBD) SARS-CoV-S-IgG assay and interferon-γ ELISpot assay 3 months following the prime vaccination dose. Immunophenotyping of T cells and immunosuppressive cells from peripheral blood was performed before the prime dose. The serological threshold 3563 AU/mL was used to discriminate non-responders or suboptimal responders versus responders. Results Most patients achieved seroconversion after receiving the two doses of vaccine (97.6%). The median serological level of anti-RBD SARS-CoV-S-IgG was equal to 3029 for patients at the metastatic stage. The patient's age was the main demographic characteristic that influenced vaccine efficacy. Among the immunological parameters measured at baseline, lower TIGIT (T cell immunoreceptor with Ig and ITIM domains) expression on CD8 T cells was associated with a better vaccine immunogenicity both in terms of humoral and cellular immune responses. Conclusion Despite a high seroconversion rate, median serological levels of patients with cancer, particularly elderly patients, were below the threshold equal to 3563 AU/mL considered as a humoral correlate of protection against SARS-CoV-2. Our findings suggest that the inhibitory receptor TIGIT might be an interesting predictive biomarker of COVID-19 vaccine immunogenicity and beyond in an anticancer vaccine context. Trial registration number ClinicalTrials.gov Registry (NCT04836793).
Collapse
Affiliation(s)
- Laurie Spehner
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Emeline Orillard
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Antoine Falcoz
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Methodology and Quality of Life Unit in Oncology, CHU Besançon, Besançon, France
| | | | - Adeline Bouard
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
| | - Hamadi Almotlak
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Stefano Kim
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Elsa Curtit
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | | | - Marine Jary
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Medical Oncology, Hôpital Jean Minjoz, Besançon, France
| | - Charlee Nardin
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Dermatology, CHU Besançon, Besançon, France
| | - Kamal Asgarov
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
| | - Syrine Abdeljaoued
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Ugo Chartral
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Virginie Mougey
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Myriam Ben Khelil
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Morgane Lopez
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Romain Loyon
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
| | - Dewi Vernerey
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Methodology and Quality of Life Unit in Oncology, CHU Besançon, Besançon, France
| | - Olivier Adotevi
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Christophe Borg
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
| | - Laura Mansi
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- Service d'oncologie médicale, CHU Besançon, Besançon, France
| | - Marie Kroemer
- Université Franche-Comté, INSERM, EFS BFC, UMR1098 RIGHT Interactions Greffon-Hôte-Tumeur/Ingénierie Cellulaire et Génique, Besançon, France
- ITAC Platform, University of Franche-Comté, Besançon, France
- Department of Pharmacy, University Hospital Centre Besançon, Besançon, France
| |
Collapse
|