1
|
Dübbel L, Göken-Riebisch A, Koch KW. Intracellular and exosomal localization of the negative checkpoint regulator VISTA in immune cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119966. [PMID: 40262722 DOI: 10.1016/j.bbamcr.2025.119966] [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: 11/18/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
Strategies in immunotherapy often target the immunosuppressive environment of tumor cells. One route of therapeutic interference could involve negative checkpoint regulators of which V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA) has raised more interest recently. The protein is expressed on the surface of tumor cells, T-lymphocytes, and antigen-presenting cells (APCs), but its intracellular expression pattern has not been investigated yet. We examined the intracellular distribution of VISTA and its possible role in translocation processes by immunofluorescence and Western blots. We analyzed the expression and localization of VISTA in murine bone marrow-derived macrophages (BMDMs), human monocyte-derived macrophages, and human T lymphocytes (Jurkat). We obtained different cell fractions and organelles of various cell types and analyzed for the presence of VISTA. Monitoring a VISTA-GFP fusion construct in transfected cell lines HL-60 and THP-1 confirmed VISTA localization in these cell lines. All used cell lines showed the colocalization of VISTA and several vesicle markers together with VISTA staining along microtubule fibers. Additionally, we found VISTA in secreted exosomes and have the first hints for nucleic expression in all tested cell lines. Therefore, the storage of VISTA in vesicles and its potential presence in nuclei resembles two other well-described checkpoint regulators, CTLA-4 and PD-L1, respectively. We conclude that VISTA storage in vesicles enables a fast response to immunogenic stimuli, which needs to be considered for inhibitory experiments. The localization of VISTA in exosomes suggests a signaling function to facilitate cell-cell communication. Furthermore, the VISTA expression in the nucleus proposes a transcriptional role.
Collapse
Affiliation(s)
- Lena Dübbel
- Division of Biochemistry, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany; University Clinic of Gynaecology and Obstetrics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany.
| | - Anna Göken-Riebisch
- University Clinic of Gynaecology and Obstetrics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Division of Biochemistry, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| |
Collapse
|
2
|
Geng Q, Xu J, Du C, Zhang D, Jin Y, Song J, Qu W, Zhang C, Su G, Jiao P. Small molecules targeting immune checkpoint proteins for cancer immunotherapy: a patent and literature review (2020-2024). Expert Opin Ther Pat 2025; 35:409-440. [PMID: 39907457 DOI: 10.1080/13543776.2025.2462849] [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: 08/06/2024] [Revised: 12/20/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025]
Abstract
INTRODUCTION Targeting immune checkpoint proteins (ICPs) via small molecules open a new window for cancer immunotherapy. Herein, we summarize recent advances of small molecules with novel chemical structures targeting ICPs, discusses their anti-tumor efficacies, which are important for the development of novel small molecules for cancer immunotherapy. AREAS COVERED In this review, the latest patents and literature were gathered through the comprehensive searches in the databases of European Patent Office (EPO), Cortellis Drug Discovery Intelligence (CDDI), PubMed and Web of Science using ICPs and compounds as key words. EXPERT OPINION To develop novel weapons to fight against cancer, small molecules targeting ICPs including CTLA-4, LAG-3, PD-L1, Siglec-9, TIM-3, TIGIT, and VISTA have been synthesized and evaluated in succession. Chief among them are the small molecules targeting PD-L1, which have been intensively investigated in recent years. Various in vitro assays such as ALPHA, HTRF binding assay, NFAT assay have been successfully developed to screen novel IPCs inhibitors. However, the in vivo assay, for example, using double-humanized PD-1/PD-L1 (hPD-1/hPD-L1) mouse as evaluation model, are seldom reported. Novel pharmacophores with new working mechanisms such as proteolysis targeting chimeras (PROTACs) and peptides are needed to enhance the therapeutic efficacy.
Collapse
Affiliation(s)
- Qiaohong Geng
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Juanjuan Xu
- Department of Neurology, Changyi People's Hospital, Weifang, Shandong, China
| | - Chunsheng Du
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Deheng Zhang
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Yanrui Jin
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Jiatong Song
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Wenjing Qu
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Changnan Zhang
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| | - Gaoxing Su
- School of Pharmacy, Nantong University, Nantong, Jiangsu, China
| | - Peifu Jiao
- School of Chemistry and Chemical Engineering, Qilu Normal University, Jinan, Shandong, China
| |
Collapse
|
3
|
Sun Y, Puspanathan P, Lim T, Lin D. Advances and challenges in gastric cancer testing: the role of biomarkers. Cancer Biol Med 2025; 22:j.issn.2095-3941.2024.0386. [PMID: 40126094 PMCID: PMC11976707 DOI: 10.20892/j.issn.2095-3941.2024.0386] [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: 12/04/2024] [Accepted: 01/23/2025] [Indexed: 03/25/2025] Open
Abstract
Advances in the identification of molecular biomarkers and the development of targeted therapies have enhanced the prognosis of patients with advanced gastric cancer. Several established biomarkers have been widely integrated into routine clinical diagnostics of gastric cancer to guide personalized treatment. Human epidermal growth factor receptor 2 (HER2) was the first molecular biomarker to be used in gastric cancer with trastuzumab being the first approved targeted therapy for HER2-positive gastric cancer. Programmed death-ligand 1 positivity and microsatellite instability can guide the use of immunotherapies, such as pembrolizumab and nivolumab. More recently, zolbetuximab has been approved for patients with claudin 18.2-positive diseases in some countries. More targeted therapies, including savolitinib for MET-positive patients, are currently under clinical investigation. However, the clinical application of these diagnostic approaches could be hampered by many existing challenges, including invasive and costly sampling methods, variability in immunohistochemistry interpretation, high costs and long turnaround times for next-generation sequencing, the absence of standardized and clinically validated diagnostic cut-off values for some biomarkers, and tumor heterogeneity. Novel testing and analysis techniques, such as artificial intelligence-assisted image analysis and multiplex immunohistochemistry, and emerging therapeutic strategies, including combination therapies that integrate immune checkpoint inhibitors with targeted therapies, offer potential solutions to some of these challenges. This article reviews recent progress in gastric cancer testing, outlines current challenges, and explores future directions for biomarker testing and targeted therapy for gastric cancer.
Collapse
Affiliation(s)
- Yu Sun
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| | | | - Tony Lim
- Division of Pathology, Singapore General Hospital, Singapore 169608, Singapore
| | - Dongmei Lin
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Pathology, Peking University Cancer Hospital & Institute, Beijing 100142, China
| |
Collapse
|
4
|
Ren X, Guo A, Geng J, Chen Y, Wang X, Zhou L, Shi L. Pan-cancer analysis of co-inhibitory molecules revealing their potential prognostic and clinical values in immunotherapy. Front Immunol 2025; 16:1544104. [PMID: 40196117 PMCID: PMC11973099 DOI: 10.3389/fimmu.2025.1544104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The widespread use of immune checkpoint inhibitors (anti-CTLA4 or PD-1) has opened a new chapter in tumor immunotherapy by providing long-term remission for patients. Unfortunately, however, these agents are not universally available and only a minority of patients respond to them. Therefore, there is an urgent need to develop novel therapeutic strategies targeting other co-inhibitory molecules. However, comprehensive information on the expression and prognostic value of co-inhibitory molecules, including co-inhibitory receptors and their ligands, in different cancers is not yet available. Methods We investigated the expression, correlation, and prognostic value of co-inhibitory molecules in different cancer types based on TCGA, UCSC Xena, TIMER, CellMiner datasets. We also examined the associations between the expression of these molecules and the extent of immune cell infiltration. Besides, we conducted a more in-depth study of VISTA. Result The results of differential expression analysis, correlation analysis, and drug sensitivity analysis suggest that CTLA4, PD-1, TIGIT, LAG3, TIM3, NRP1, VISTA, CD80, CD86, PD-L1, PD-L2, PVR, PVRL2, FGL1, LGALS9, HMGB1, SEMA4A, and VEGFA are associated with tumor prognosis and immune cell infiltration. Therefore, we believe that they are hopefully to serve as prognostic biomarkers for certain cancers. In addition, our analysis indicates that VISTA plays a complex role and its expression is related to TMB, MSI, cancer cell stemness, DNA/RNA methylation, and drug sensitivity. Conclusions These co-inhibitory molecules have the potential to serve as prognostic biomarkers and therapeutic targets for a broad spectrum of cancers, given their strong associations with key clinical metrics. Furthermore, the analysis results indicate that VISTA may represent a promising target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Anjie Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiahui Geng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuling Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lian Zhou
- Department of Head&Neck Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
5
|
Luo Y, Yuan Y, Liu D, Peng H, Shen L, Chen Y. Targeting novel immune checkpoints in the B7-H family: advancing cancer immunotherapy from bench to bedside. Trends Cancer 2025:S2405-8033(25)00055-X. [PMID: 40113530 DOI: 10.1016/j.trecan.2025.02.007] [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: 12/04/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
The B7-H family of immune checkpoint molecules is a crucial component of the immune regulatory network for tumors, offering new opportunities to modulate the tumor microenvironment (TME). The B7-H family - which includes B7-H2 (inducible T cell costimulatory ligand, ICOSL), B7-H3, B7-H4, B7-H5 (V-domain immunoglobulin suppressor of T cell activation, VISTA), B7-H6, and B7-H7 (HHLA2) - is known for its diverse roles in regulating innate and adaptive immunity. These molecules can exhibit co-stimulatory or co-inhibitory effects on T cells, influencing processes such as T cell activation, differentiation, and effector functions, and they are involved in the recruitment and polarization of various immune cells. This review explores the structural characteristics, receptor-ligand interactions, and signaling pathways associated with each B7-H family member. We also discuss the family's impact on tumor immunity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Yiming Luo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ye Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Early Drug Development Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing 102200, China.
| |
Collapse
|
6
|
Qi Z, Cheng Y, Wang K, Cai S, Ni X, Wang T, Zhang K, Jiang S, Xiao Y, Zhang X. Discovery, Synthesis, and Activity Evaluation of Novel Small-Molecule Inhibitors Targeting VISTA for Cancer Immunotherapy. J Med Chem 2025; 68:5222-5237. [PMID: 40014385 DOI: 10.1021/acs.jmedchem.4c02031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Immune checkpoint inhibitors (ICIs) have been potent therapeutic options for the treatment of multiple types of cancer. However, not all patients experience benefits from ICIs, and discovering inhibitors targeting novel immune checkpoints is necessary. V-domain Ig suppressor of T-cell activation (VISTA) is a novel immune checkpoint. Blockade of the VISTA pathway enhances antitumor immunity in multiple tumor types. Herein, a series of VISTA inhibitors based on the benzimidazole scaffold were discovered. B3 showed the strongest binding affinity to the VISTA protein with a KD value of 0.452 ± 0.12 μM. In vitro, B3 could effectively activate VISTA-mediated immunosuppression and induce effective VISTA degradation in HepG2 cells. In vivo, B3 improved pharmacokinetics compared to the lead compound 4. Moreover, compound B3 significantly inhibited tumor growth in a CT26 colon cancer model. These results suggest that compound B3 is a promising VISTA small molecule inhibitor and degrader worthy of further development as an antitumor agent.
Collapse
Affiliation(s)
- Zhihao Qi
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yao Cheng
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi Cai
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiang Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- Department of Biomedical Engineering and Diagnostic Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
7
|
Wang J, Wang R, Wang M, Ge J, Wang Y, Li Y, Chen C, He J, Zheng B, Xu M, Jiang X, Liu Y, Chen M, Long J. Cutting-Edge Therapy and Immune Escape Mechanisms in EBV-Associated Tumors. Med Res Rev 2025. [PMID: 40077924 DOI: 10.1002/med.22104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 12/19/2024] [Accepted: 02/21/2025] [Indexed: 03/14/2025]
Abstract
Epstein-Barr virus (EBV), the first identified human tumor virus, significantly influences the immune microenvironment of associated cancers. EBV-induced expression of viral antigens by tumor cells triggers immune recognition and elicits a pro-inflammatory response. While mild inflammation may help eliminate malignant cells, intense inflammation can accelerate tumor progression. Moreover, EBV can establish lifelong latency in human hosts, characterized by low immunogenicity of its proteins and noncoding RNAs. This enables tumor cells to evade immune detection and impair immune cell function, disrupting immune homeostasis. Consequently, EBV-associated malignancies pose a considerable public health challenge globally, often complicating the prognosis of cancer patients under conventional treatment. With deeper research into the oncogenic expressions and mechanisms of EBV, novel targeted therapies against EBV are gaining prominence. This review discusses recent advancements in understanding how EBV helps tumor cells evade immune surveillance and induce immune dysfunction. It also examines the clinical potential of targeting EBV-associated tumors, providing fresh perspectives on the mechanisms and therapeutic strategies for these cancers.
Collapse
Affiliation(s)
- Jie Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Rong Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
- Guangdong Province Key Laboratory of Pharmaceutical Functional Genes, MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Meifeng Wang
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Junshang Ge
- Key Laboratory of Carcinogenesis and Cancer Invasion of the Chinese Ministry of Education, Cancer Research Institute and School of Basic Medicine Sciences, Central South University, Changsha, China
| | - Yian Wang
- The Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University; The Engineering Research Center of Reproduction and Translational Medicine of Hunan Province, Changsha, China
| | - Yanhan Li
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Changan Chen
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Jiale He
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Boshu Zheng
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Meifang Xu
- Department of Pathology and Institute of Oncology, The School of Basic Medical Sciences & Diagnostic Pathology Center, Fujian Medical University, Fuzhou, China
| | - Xianjie Jiang
- Hunan Key Laboratory of Cancer Metabolism, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Yuhang Liu
- Department of Forensic Science, School of Basic Medical Science, Central South University, Changsha, China
| | - Mingfen Chen
- Department of Radiation Oncology, The Second Affiliated Hospital of Fujian Medical University, Fujian Medical University, Quanzhou, China
| | - Jun Long
- Shenzhen Geim Graphene Center, Tsinghua-Berkeley Shenzhen Institute & Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| |
Collapse
|
8
|
Almawash S. Revolutionary Cancer Therapy for Personalization and Improved Efficacy: Strategies to Overcome Resistance to Immune Checkpoint Inhibitor Therapy. Cancers (Basel) 2025; 17:880. [PMID: 40075727 PMCID: PMC11899125 DOI: 10.3390/cancers17050880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/09/2025] [Accepted: 02/12/2025] [Indexed: 03/14/2025] Open
Abstract
Cancer remains a significant public health issue worldwide, standing as a primary contributor to global mortality, accounting for approximately 10 million fatalities in 2020 [...].
Collapse
Affiliation(s)
- Saud Almawash
- Department of Pharmaceutics, College of Pharmacy, Shaqra University, Shaqra 11961, Saudi Arabia
| |
Collapse
|
9
|
Zhang RJ, Kim TK. VISTA-mediated immune evasion in cancer. Exp Mol Med 2024; 56:2348-2356. [PMID: 39482534 PMCID: PMC11612309 DOI: 10.1038/s12276-024-01336-6] [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: 06/06/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 11/03/2024] Open
Abstract
Over the past decade, V-domain immunoglobulin suppressor of T-cell activation (VISTA) has been established as a negative immune checkpoint molecule. Since the role of VISTA in inhibiting T-cell activation was described, studies have demonstrated other diverse regulatory functions in multiple immune cell populations. Furthermore, its relevance has been identified in human cancers. The role of VISTA in cancer immune evasion has been determined, but its mechanisms in the tumor microenvironment remain to be further elucidated. Understanding its contributions to cancer initiation, progression, and resistance to current treatments will be critical to its utility as a target for novel immunotherapies. Here, we summarize the current understanding of VISTA biology in cancer.
Collapse
Affiliation(s)
- Raymond J Zhang
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
| |
Collapse
|
10
|
Wang K, Cai S, Cheng Y, Qi Z, Ni X, Zhang K, Xiao Y, Zhang X, Wang T. Discovery of Benzo[ d]oxazoles as Novel Dual Small-Molecule Inhibitors Targeting PD-1/PD-L1 and VISTA Pathway. J Med Chem 2024; 67:18526-18548. [PMID: 39389791 DOI: 10.1021/acs.jmedchem.4c01899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
The blockers of programmed cell death-1 (PD-1)/programmed cell death-ligand 1 (PD-L1) pathway have achieved great clinical success. However, the limited efficacy and low tumor response rate of anti-PD-1/PD-L1 monotherapy limit the clinical application of PD-1/PD-L1 inhibitors. V-domain immunoglobulin suppressor of T-cell activation (VISTA), a novel checkpoint regulator, exhibits potential synergy with PD-1/PD-L1 in enhancing antitumor immunity. Herein, we report the discovery of benzo[d]oxazole B3 as novel dual small-molecule inhibitors targeting PD-1/PD-L1 and VISTA with high PD-1/PD-L1 inhibitory activity and VISTA binding affinity. B3 rescues the immunosuppression of T-cells mediated by PD-L1 and VISTA and activates antitumor immunity effectively. Moreover, B3 could induce degradation of PD-L1 and VISTA in tumor cell. Furthermore, B3 displays significant in vivo antitumor efficacy in a CT26 mouse model. Our results discover B3 as a promising dual PD-1/PD-L1 and VISTA inhibitor, providing a novel therapeutic strategy to overcome the limitations of current anti-PD-1/PD-L1 therapy.
Collapse
Affiliation(s)
- Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi Cai
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
- Innovation Department of the Research Institute, Nanjing Chia-Tai Tianqing Pharmaceutical Co., Ltd., Nanjing 210046, China
| | - Yao Cheng
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Zhihao Qi
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiang Ni
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
11
|
Vilela T, Valente S, Correia J, Ferreira F. Advances in immunotherapy for breast cancer and feline mammary carcinoma: From molecular basis to novel therapeutic targets. Biochim Biophys Acta Rev Cancer 2024; 1879:189144. [PMID: 38914239 DOI: 10.1016/j.bbcan.2024.189144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
The role of inflammation in cancer is a topic that has been investigated for many years. As established, inflammation emerges as a defining characteristic of cancer, presenting itself as a compelling target for therapeutic interventions in the realm of oncology. Controlling the tumor microenvironment (TME) has gained paramount significance, modifying not only the effectiveness of immunotherapy but also modulating the outcomes and prognoses of standard chemotherapy and other anticancer treatments. Immunotherapy has surfaced as a central focus within the domain of tumor treatments, using immune checkpoint inhibitors as cancer therapy. Immune checkpoints and their influence on the tumor microenvironment dynamic are presently under investigation, aiming to ascertain their viability as therapeutic interventions across several cancer types. Cancer presents a significant challenge in humans and cats, where female breast cancer ranks as the most prevalent malignancy and feline mammary carcinoma stands as the third most frequent. This review seeks to summarize the data about the immune checkpoints cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), lymphocyte activation gene-3 (LAG-3), programmed cell death protein-1 (PD-1), V-domain Ig suppressor of T cell activation (VISTA), and T-cell immunoglobulin and mucin domain 3 (TIM-3) respective ongoing investigations as prospective targets for therapy for human breast cancer, while also outlining findings from studies reported on feline mammary carcinoma (FMC), strengthening the rationale for employing FMC as a representative model in the exploration of human breast cancer.
Collapse
Affiliation(s)
- Tatiana Vilela
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Sofia Valente
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Jorge Correia
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; CIISA-Center of Interdisciplinary Research in Animal Health, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Fernando Ferreira
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; CIISA-Center of Interdisciplinary Research in Animal Health, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal.
| |
Collapse
|
12
|
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
|
13
|
Gao Y, He Y, Tang Y, Chen ZS, Qu M. VISTA: A Novel Checkpoint for Cancer Immunotherapy. Drug Discov Today 2024; 29:104045. [PMID: 38797321 DOI: 10.1016/j.drudis.2024.104045] [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: 02/22/2024] [Revised: 04/20/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a recently identified member of the B7 family of immunoregulatory proteins. It is pivotal for maintaining T cell quiescence and exerts a significant regulatory influence on the immune response to tumors. Accumulating clinical evidence suggests that the influence of VISTA on tumor immunity is more nuanced than initially postulated. Although these revelations add layers of complexity to our understanding of the function of VISTA, they also offer novel avenues for scientific inquiry and potential therapeutic targets. In this review, we scrutinize the current literature pertaining to the expression of VISTA in various of malignancies, aiming to elucidate its intricate roles within the tumor microenvironment and in cancer immunotherapy.
Collapse
Affiliation(s)
- Yu Gao
- Translational Medical Center, Weifang Second People's Hospital, Weifang 261041, Shandong, China
| | - Yanting He
- Department of Pathology, The Affiliated Hospital of Qingdao University, Pingdu 266700, Shandong, China
| | - Yuanyuan Tang
- Translational Medical Center, Weifang Second People's Hospital, Weifang 261041, Shandong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Meihua Qu
- Translational Medical Center, Weifang Second People's Hospital, Weifang 261041, Shandong, China; School of Life Science and Technology, Weifang Medical University, Weifang 261053, Shandon, China.
| |
Collapse
|
14
|
Olbromski M, Mrozowska M, Piotrowska A, Smolarz B, Romanowicz H. The VISTA/VSIG3/PSGL-1 axis: crosstalk between immune effector cells and cancer cells in invasive ductal breast carcinoma. Cancer Immunol Immunother 2024; 73:136. [PMID: 38833004 PMCID: PMC11150347 DOI: 10.1007/s00262-024-03701-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 04/09/2024] [Indexed: 06/06/2024]
Abstract
A checkpoint protein called the V-domain Ig suppressor of T cell activation (VISTA) is important for controlling immune responses. Immune cells that interact with VISTA have molecules, or receptors, known as VISTA receptors. Immune system activity can be modified by the interaction between VISTA and its receptors. Since targeting VISTA or its receptors may be beneficial in certain conditions, VISTA has been studied in relation to immunotherapy for cancer and autoimmune illnesses. The purpose of this study was to examine the expression levels and interactions between VISTA and its receptors, VSIG3 and PSGL-1, in breast cancer tissues. IHC analysis revealed higher levels of proteins within the VISTA/VSIG3/PSGL-1 axis in cancer tissues than in the reference samples (mastopathies). VISTA was found in breast cancer cells and intratumoral immune cells, with membranous and cytoplasmic staining patterns. VISTA was also linked with pathological grade and VSIG3 and PSGL-1 levels. Furthermore, we discovered that the knockdown of one axis member boosted the expression of the other partners. This highlights the significance of VISTA/VSIG3/PSGL-1 in tumor stroma and microenvironment remodeling. Our findings indicate the importance of the VISTA/VSIG3/PSGL-1 axis in the molecular biology of cancer cells and the immune microenvironment.
Collapse
Affiliation(s)
- Mateusz Olbromski
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chalubinskiego 6a, 50-368, Wroclaw, Poland.
| | - Monika Mrozowska
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chalubinskiego 6a, 50-368, Wroclaw, Poland
| | - Aleksandra Piotrowska
- Department of Histology and Embryology, Department of Human Morphology and Embryology, Wroclaw Medical University, Chalubinskiego 6a, 50-368, Wroclaw, Poland
| | - Beata Smolarz
- Department of Pathology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| | - Hanna Romanowicz
- Department of Pathology, Polish Mother's Memorial Hospital Research Institute, 93-338, Lodz, Poland
| |
Collapse
|
15
|
Jlassi A, Rejaibi R, Manai M, Sahraoui G, Guerfali FZ, Charfi L, Mezlini A, Manai M, Mrad K, Doghri R. VISTA/CTLA4/PD1 coexpression on tumor cells confers a favorable immune microenvironment and better prognosis in high-grade serous ovarian carcinoma. Front Oncol 2024; 14:1352053. [PMID: 38634058 PMCID: PMC11022690 DOI: 10.3389/fonc.2024.1352053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 03/18/2024] [Indexed: 04/19/2024] Open
Abstract
Introduction Immunotherapy by blocking immune checkpoints programmed death/ligand (PD1/PDL1) and cytotoxic T-lymphocyte-associated protein 4(CTLA4) has emerged as new therapeutic targets in cancer. However, their efficacy has been limited due to resistance. A new- checkpoint V-domain Ig-containing suppressor of T cell activation (VISTA) has appeared, but the use of its inhibition effect in combination with antibodies targeting PDL1/PD1and CTLA4 has not been reported in ovarian cancer. Methods In this study, we investigated the expressions of VISTA, CTLA4, and PDL1 using immunohistochemistry (IHC)on 135 Formalin-Fixed Paraffin-Embedded (FFPE)tissue samples of High-grade serous carcinoma (HGSOC). VISTA, CTLA4, PDL1, PD1, CD8, CD4, and FOXP3 mRNA extracted from 429 patients with ovarian cancer in the Cancer Genome Atlas (TCGA) database was included as a validation cohort. Correlations between these checkpoints, tumor-infiltrating- lymphocytes (TILs), and survival were analyzed. Results and discussion CTLA4 was detectable in 87.3% of samples, VISTA in 64.7%, PD1 in 56.7%, and PDL1 in 48.1%. PDL1 was the only tested protein associated with an advanced stage (p=0.05). VISTA was associated with PDL1, PD1, and CTLA4 expressions (p=0.005, p=0.001, p=0.008, respectively), consistent with mRNA level analysis from the TCGA database. Univariate analyses showed only VISTA expression (p=0.04) correlated with overall survival (OS). Multivariate analyses showed that VISTA expression (p=0.01) and the coexpression of VISTA+/CTLA4+/PD1+ (p=0.05) were associated with better OS independently of the clinicopathological features. Kaplan-Meier analysis showed that the coexpression of the VISTA+/CTLA4+/PDL1+ and VISTA+/CTLA4+/PD1+ checkpoints on tumor cells (TCs)were associated with OS (p=0.02 and p<0.001; respectively). VISTA+/CTLA4+/PD1+ in TCs and CD4+/CD8+TILswere associated with better 2-yer OS. This correlation may refer to the role of VISTA as a receptor in the TCs and not in the immune cells. Thus, targeting combination therapy blocking VISTA, CTLA4, and PD1 could be a novel and attractive strategy for HGSOC treatment, considering the ambivalent role of VISTA in the HGSOC tumor cells.
Collapse
Affiliation(s)
- Aida Jlassi
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation Salah Azaiz Institute, Tunis, Tunisia
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory, Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Rim Rejaibi
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory, Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Maroua Manai
- Laboratory of Transmission, Control and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ghada Sahraoui
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation Salah Azaiz Institute, Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
| | - Fatma Zahra Guerfali
- Laboratory of Transmission, Control and Immunobiology of Infections, Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Lamia Charfi
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation Salah Azaiz Institute, Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
| | - Amel Mezlini
- Medical Oncology Department, Salah Azaiez Institute, Tunis, Tunisia
| | - Mohamed Manai
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory, Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Karima Mrad
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation Salah Azaiz Institute, Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
| | - Raoudha Doghri
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation Salah Azaiz Institute, Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
| |
Collapse
|
16
|
Nowak KM, Chetty R. Predictive and prognostic biomarkers in gastrointestinal tract tumours. Pathology 2024; 56:205-213. [PMID: 38238239 DOI: 10.1016/j.pathol.2023.12.412] [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: 11/16/2023] [Revised: 12/28/2023] [Accepted: 12/30/2023] [Indexed: 02/18/2024]
Abstract
Tumours of the gastrointestinal tract represent nearly a quarter of all newly diagnosed tumours diagnosed in 2019. Various treatment modalities for gastrointestinal cancers exist, some of which may be guided by biomarkers. Biomarkers act as gauges of either normal or pathogenic processes or responses to an exposure or intervention. They come in many forms. This review explores established and potential molecular/immunohistochemical (IHC) predictive and prognostic biomarkers of the gastrointestinal tract.
Collapse
Affiliation(s)
- Klaudia M Nowak
- Laboratory Medicine Program, Toronto General Hospital, University Health Network, Toronto, Ontario, Canada.
| | | |
Collapse
|
17
|
Cao Y, Yu K, Zhang Z, Gu Y, Gu Y, Li W, Zhang W, Shen Z, Xu J, Qin J. Blockade of V-domain immunoglobulin suppressor of T-cell activation reprograms tumour-associated macrophages and improves efficacy of PD-1 inhibitor in gastric cancer. Clin Transl Med 2024; 14:e1578. [PMID: 38356419 PMCID: PMC10867598 DOI: 10.1002/ctm2.1578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND AND AIMS In gastric cancer, the response rate of programmed cell death protein-1 (PD-1) inhibitor is far from satisfactory, indicating additional nonredundant pathways might hamper antitumour immunity. V-domain immunoglobulin suppressor of T-cell activation (VISTA) has been reported in several malignancies as a novel immune-checkpoint. Nevertheless, the role of VISTA in gastric cancer still remains obscure. Our purpose is to explore the clinical significance and potential mechanism of VISTA in affecting gastric cancer patients' survival and immunotherapeutic responsiveness. METHODS Our study recruited eight independent cohorts with a total of 1403 gastric cancer patients. Immunohistochemistry, multiplex immunofluorescence, flow cytometry or intracellular flow cytometry, quantitative polymerase chain reaction, western blotting, fluorescence-activated cell sorting, magnetic-activated cell sorting, smart-seq2, in vitro cell co-culture and ex vivo tumour inhibition assays were applied to investigate the clinical significance and potential mechanism of VISTA in gastric cancer. RESULTS VISTA was predominantly expressed on tumour-associated macrophages (TAMs), and indicated poor clinical outcomes and inferior immunotherapeutic responsiveness. VISTA+ TAMs showed a mixed phenotype. Co-culture of TAMs and CD8+ T cells indicated that VISTA+ TAMs attenuated effective function of CD8+ T cells. Blockade of VISTA reprogrammed TAMs to a proinflammatory phenotype, reactivated CD8+ T cells and promoted apoptosis of tumour cells. Moreover, blockade of VISTA could also enhance the efficacy of PD-1 inhibitor, suggesting that blockade of VISTA might synergise with PD-1 inhibitor in gastric cancer. CONCLUSIONS Our data revealed that VISTA was an immune-checkpoint associated with immunotherapeutic resistance. Blockade of VISTA reprogrammed TAMs, promoted T-cell-mediated antitumour immunity, and enhanced efficacy of PD-1 inhibitor, which might have implications in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yifan Cao
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Kuan Yu
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Zihao Zhang
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yun Gu
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yichao Gu
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Wandi Li
- Department of ImmunologySchool of Basic Medical Sciences, Fudan UniversityShanghaiChina
| | - Weijuan Zhang
- Department of ImmunologySchool of Basic Medical Sciences, Fudan UniversityShanghaiChina
| | - Zhenbin Shen
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Jiejie Xu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical Sciences, Fudan UniversityShanghaiChina
| | - Jing Qin
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
18
|
Rezagholizadeh F, Tajik F, Talebi M, Taha SR, Shariat Zadeh M, Farhangnia P, Hosseini HS, Nazari A, Mollazadeh Ghomi S, Kamrani Mousavi SM, Haeri Moghaddam N, Khorramdelazad H, Joghataei MT, Safari E. Unraveling the potential of CD8, CD68, and VISTA as diagnostic and prognostic markers in patients with pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1283364. [PMID: 38357542 PMCID: PMC10865497 DOI: 10.3389/fimmu.2024.1283364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Pancreatic cancer is a truculent disease with limited treatment options and a grim prognosis. Immunotherapy has shown promise in treating various types of cancer, but its effectiveness in pancreatic cancer has been lacking. As a result, it is crucial to identify markers associated with immunological pathways in order to improve the treatment outcomes for this deadly cancer. The purpose of this study was to investigate the diagnostic and prognostic significance of three markers, CD8, CD68, and VISTA, in pancreatic ductal adenocarcinoma (PDAC), the most common subtype of pancreatic cancer. Methods We analyzed gene expression data from Gene Expression Omnibus (GEO) database using bioinformatics tools. We also utilized the STRING online tool and Funrich software to study the protein-protein interactions and transcription factors associated with CD8, CD68, and VISTA. In addition, tissue microarray (TMA) and immunohistochemistry (IHC) staining were performed on 228 samples of PDAC tissue and 10 samples of normal pancreatic tissue to assess the expression levels of the markers. We then correlated these expression levels with the clinicopathological characteristics of the patients and evaluated their survival rates. Results The analysis of the GEO data revealed slightly elevated levels of VISTA in PDAC samples compared to normal tissues. However, there was a significant increase in CD68 expression and a notable reduction in CD8A expression in pancreatic cancer. Further investigation identified potential protein-protein interactions and transcription factors associated with these markers. The IHC staining of PDAC tissue samples showed an increased expression of VISTA, CD68, and CD8A in pancreatic cancer tissues. Moreover, we found correlations between the expression levels of these markers and certain clinicopathological features of the patients. Additionally, the survival analysis revealed that high expression of CD8 was associated with better disease-specific survival and progression-free survival in PDAC patients. Conclusion These findings highlight the potential of CD8, CD68, and VISTA as diagnostic and prognostic indicators in PDAC.
Collapse
Affiliation(s)
- Fereshteh Rezagholizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Talebi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), Tehran, Iran
| | - Seyed Reza Taha
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Hamideh Sadat Hosseini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Aram Nazari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Mollazadeh Ghomi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyede Mahtab Kamrani Mousavi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Haeri Moghaddam
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
19
|
Mesquita FP, Lima LB, da Silva EL, Souza PFN, de Moraes MEA, Burbano RMR, Montenegro RC. A Review on Anaplastic Lymphoma Kinase (ALK) Rearrangements and Mutations: Implications for Gastric Carcinogenesis and Target Therapy. Curr Protein Pept Sci 2024; 25:539-552. [PMID: 38424421 DOI: 10.2174/0113892037291318240130103348] [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: 11/04/2023] [Revised: 01/10/2024] [Accepted: 01/17/2024] [Indexed: 03/02/2024]
Abstract
Gastric adenocarcinoma is a complex disease with diverse genetic modifications, including Anaplastic Lymphoma Kinase (ALK) gene changes. The ALK gene is located on chromosome 2p23 and encodes a receptor tyrosine kinase that plays a crucial role in embryonic development and cellular differentiation. ALK alterations can result from gene fusion, mutation, amplification, or overexpression in gastric adenocarcinoma. Fusion occurs when the ALK gene fuses with another gene, resulting in a chimeric protein with constitutive kinase activity and promoting oncogenesis. ALK mutations are less common but can also result in the activation of ALK signaling pathways. Targeted therapies for ALK variations in gastric adenocarcinoma have been developed, including ALK inhibitors that have shown promising results in pre-clinical studies. Future studies are needed to elucidate the ALK role in gastric cancer and to identify predictive biomarkers to improve patient selection for targeted therapy. Overall, ALK alterations are a relevant biomarker for gastric adenocarcinoma treatment and targeted therapies for ALK may improve patients' overall survival.
Collapse
Affiliation(s)
- Felipe Pantoja Mesquita
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | - Luina Benevides Lima
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | - Emerson Lucena da Silva
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | - Pedro Filho Noronha Souza
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
| | | | - Rommel Mario Rodrigues Burbano
- Department of Biological Sciences, Oncology Research Center, Federal University of Pará, Belém, Brazil
- Molecular Biology Laboratory, Ophir Loyola Hospital, Belém, Brazil
| | - Raquel Carvalho Montenegro
- Laboratory, Drug Research and Development Center (NPDM), Federal University of Ceará, Fortaleza, CE, 60430-275, Brazil
- Latinoamericana de Implementación y Validación de guias clinicas Farmacogenomicas (RELIVAF), Brazil
| |
Collapse
|
20
|
Xiao Y, Li ZZ, Zhong NN, Cao LM, Liu B, Bu LL. Charting new frontiers: Co-inhibitory immune checkpoint proteins in therapeutics, biomarkers, and drug delivery systems in cancer care. Transl Oncol 2023; 38:101794. [PMID: 37820473 PMCID: PMC10582482 DOI: 10.1016/j.tranon.2023.101794] [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: 06/23/2023] [Revised: 09/17/2023] [Accepted: 09/29/2023] [Indexed: 10/13/2023] Open
Abstract
Cancer remains a major health concern globally. Immune checkpoint inhibitors (ICIs) target co-inhibitory immune checkpoint molecules and have received approval for treating malignancies like melanoma and non-small cell lung cancer. While CTLA-4 and PD-1/PD-L1 are extensively researched, additional targets such as LAG-3, TIGIT, TIM-3, and VISTA have also demonstrated effective in cancer therapy. Combination treatments, which pair ICIs with interventions such as radiation or chemotherapy, amplify therapeutic outcomes. However, ICIs can lead to diverse side effects, and their varies across patients and cancers. Hence, identifying predictive biomarkers to guide therapy is essential. Notably, expression levels of molecules like PD-1, CTLA-4, and LAG-3 have been linked to tumor progression and ICI therapy responsiveness. Recent advancements in drug delivery systems (DDSs) further enhance ICI therapy efficacy. This review explores predominant DDSs for ICI delivery, such as hydrogel, microparticle, and nanoparticle, which offer improved therapeutic effects and reduced toxicity. In summary, we discuss the future of immune therapy focusing on co-inhibitory checkpoint molecules, pinpoint challenges, and suggest avenues for developing efficient, safer DDSs for ICI transport.
Collapse
Affiliation(s)
- Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Zi-Zhan Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Nian-Nian Zhong
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Lei-Ming Cao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Bing Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China
| | - Lin-Lin Bu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China; Department of Oral & Maxillofacial - Head Neck Oncology, School & Hospital of Stomatology, Wuhan University, Wuhan, 430079, China.
| |
Collapse
|
21
|
Wang B, Ou Z, Zhong W, Huang L, Liao W, Sheng Y, Guo Z, Chen J, Yang W, Chen K, Huang X, Yang T, Lin T, Huang J. Effective Antitumor Immunity Can Be Triggered by Targeting VISTA in Combination with a TLR3-Specific Adjuvant. Cancer Immunol Res 2023; 11:1656-1670. [PMID: 37847894 DOI: 10.1158/2326-6066.cir-23-0117] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/31/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023]
Abstract
Resistance to anti-PD-1/PD-L1 treatment is often associated with accumulation of intratumoral inhibitory macrophages. V-domain immunoglobulin suppressor of T-cell activation (VISTA) is a nonredundant immune checkpoint that can induce both T-cell and myeloid-cell immunosuppression. In this study, we found that high levels of VISTA+ immune cells were associated with advanced stage bladder cancer and predicted poor survival in patients. A combination of high infiltration of VISTA+ immune cells and PD-L1+ immune cells or PD-1+ T cells predicted the worst survival. Flow cytometry and multiplex immunofluorescence analyses confirmed that VISTA expression was higher in macrophages than in T cells or neutrophils, and only VISTA+CD163+ macrophage density predicted poor prognosis in patients with bladder cancer. Toll-like receptor (TLR) agonists are known to trigger the innate immune response in macrophages. We found that the VISTA-specific mAb 13F3 augmented the ability of a TLR3-specific adjuvant to induce macrophage activation in vitro. In the MB49 syngeneic mouse model of bladder cancer, treatment with 13F3 curbed tumor growth and prolonged survival when combined with a TLR3-specific adjuvant. The combination treatment reduced the intratumoral frequency of CD206+ anti-inflammatory macrophages and levels of the immunosuppressive molecule TGFβ1, but it upregulated expression of immunostimulatory molecules (Ifna, Ifnb, and Trail) and increased the CD8+ T cell/regulatory T-cell ratio. These findings indicate that elevated VISTA expression in immune cells, particularly macrophages, is associated with an unfavorable prognosis in patients with bladder cancer and suggest that targeting VISTA in combination with a TLR3-specific adjuvant has translational potential.
Collapse
Affiliation(s)
- Bo Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Ziwei Ou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Wenlong Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Lin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Wenjian Liao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Yiyu Sheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Zhixing Guo
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, P.R. China
| | - Junyu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Wenjuan Yang
- Department of Hematology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Ke Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Xiaodong Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Tenghao Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| |
Collapse
|
22
|
Shekari N, Shanehbandi D, Kazemi T, Zarredar H, Baradaran B, Jalali SA. VISTA and its ligands: the next generation of promising therapeutic targets in immunotherapy. Cancer Cell Int 2023; 23:265. [PMID: 37936192 PMCID: PMC10631023 DOI: 10.1186/s12935-023-03116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel negative checkpoint receptor (NCR) primarily involved in maintaining immune tolerance. It has a role in the pathogenesis of autoimmune disorders and cancer and has shown promising results as a therapeutic target. However, there is still some ambiguity regarding the ligands of VISTA and their interactions with each other. While V-Set and Immunoglobulin domain containing 3 (VSIG-3) and P-selectin glycoprotein ligand-1(PSGL-1) have been extensively studied as ligands for VISTA, the others have received less attention. It seems that investigating VISTA ligands, reviewing their functions and roles, as well as outcomes related to their interactions, may allow an understanding of their full functionality and effects within the cell or the microenvironment. It could also help discover alternative approaches to target the VISTA pathway without causing related side effects. In this regard, we summarize current evidence about VISTA, its related ligands, their interactions and effects, as well as their preclinical and clinical targeting agents.
Collapse
Affiliation(s)
- Najibeh Shekari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Amir Jalali
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
23
|
Luk SJ, Schoppmeyer R, Ijsselsteijn ME, Somarakis A, Acem I, Remst DFG, Cox DT, van Bergen CAM, Briaire-de Bruijn I, Grönloh MLB, van der Meer WJ, Hawinkels LJAC, Koning RI, Bos E, Bovée JVMG, de Miranda NFCC, Szuhai K, van Buul JD, Falkenburg JHF, Heemskerk MHM. VISTA Expression on Cancer-Associated Endothelium Selectively Prevents T-cell Extravasation. Cancer Immunol Res 2023; 11:1480-1492. [PMID: 37695550 DOI: 10.1158/2326-6066.cir-22-0759] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 04/14/2023] [Accepted: 09/01/2023] [Indexed: 09/12/2023]
Abstract
Cancers evade T-cell immunity by several mechanisms such as secretion of anti-inflammatory cytokines, down regulation of antigen presentation machinery, upregulation of immune checkpoint molecules, and exclusion of T cells from tumor tissues. The distribution and function of immune checkpoint molecules on tumor cells and tumor-infiltrating leukocytes is well established, but less is known about their impact on intratumoral endothelial cells. Here, we demonstrated that V-domain Ig suppressor of T-cell activation (VISTA), a PD-L1 homolog, was highly expressed on endothelial cells in synovial sarcoma, subsets of different carcinomas, and immune-privileged tissues. We created an ex vivo model of the human vasculature and demonstrated that expression of VISTA on endothelial cells selectively prevented T-cell transmigration over endothelial layers under physiologic flow conditions, whereas it does not affect migration of other immune cell types. Furthermore, endothelial VISTA correlated with reduced infiltration of T cells and poor prognosis in metastatic synovial sarcoma. In endothelial cells, we detected VISTA on the plasma membrane and in recycling endosomes, and its expression was upregulated by cancer cell-secreted factors in a VEGF-A-dependent manner. Our study reveals that endothelial VISTA is upregulated by cancer-secreted factors and that it regulates T-cell accessibility to cancer and healthy tissues. This newly identified mechanism should be considered when using immunotherapeutic approaches aimed at unleashing T cell-mediated cancer immunity.
Collapse
Affiliation(s)
- Sietse J Luk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Rouven Schoppmeyer
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Leeuwenhoek Centre for Advanced Microscopy, Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Antonios Somarakis
- Department of Radiology, Leiden University Medical Center, Leiden, the Netherlands
| | - Ibtissam Acem
- Department of Orthopedic Surgery, Leiden University Medical Center, Leiden, the Netherlands
- Department of Oncological and Gastrointestinal Surgery, Erasmus MC Cancer Institute, Rotterdam, the Netherlands
| | - Dennis F G Remst
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | - Daan T Cox
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| | | | | | - Max L B Grönloh
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
- Leeuwenhoek Centre for Advanced Microscopy, Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Werner J van der Meer
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
- Leeuwenhoek Centre for Advanced Microscopy, Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Lukas J A C Hawinkels
- Department of Gastroenterology-Hepatology, Leiden University Medical Center, Leiden, the Netherlands
| | - Roman I Koning
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Erik Bos
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Judith V M G Bovée
- Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands
| | | | - Karoly Szuhai
- Department of Cell and Chemical Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Jaap D van Buul
- Molecular Cell Biology Lab, Department of Molecular Hematology, Sanquin Research, Amsterdam, the Netherlands
- Landsteiner Laboratory, Amsterdam UMC, University of Amsterdam, Amsterdam, the Netherlands
- Leeuwenhoek Centre for Advanced Microscopy, Molecular Cytology, Swammerdam Institute for Life Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | | | - Mirjam H M Heemskerk
- Department of Hematology, Leiden University Medical Center, Leiden, The Netherlands
| |
Collapse
|
24
|
Roy D, Gilmour C, Patnaik S, Wang LL. Combinatorial blockade for cancer immunotherapy: targeting emerging immune checkpoint receptors. Front Immunol 2023; 14:1264327. [PMID: 37928556 PMCID: PMC10620683 DOI: 10.3389/fimmu.2023.1264327] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2023] [Accepted: 09/26/2023] [Indexed: 11/07/2023] Open
Abstract
The differentiation, survival, and effector function of tumor-specific CD8+ cytotoxic T cells lie at the center of antitumor immunity. Due to the lack of proper costimulation and the abundant immunosuppressive mechanisms, tumor-specific T cells show a lack of persistence and exhausted and dysfunctional phenotypes. Multiple coinhibitory receptors, such as PD-1, CTLA-4, VISTA, TIGIT, TIM-3, and LAG-3, contribute to dysfunctional CTLs and failed antitumor immunity. These coinhibitory receptors are collectively called immune checkpoint receptors (ICRs). Immune checkpoint inhibitors (ICIs) targeting these ICRs have become the cornerstone for cancer immunotherapy as they have established new clinical paradigms for an expanding range of previously untreatable cancers. Given the nonredundant yet convergent molecular pathways mediated by various ICRs, combinatorial immunotherapies are being tested to bring synergistic benefits to patients. In this review, we summarize the mechanisms of several emerging ICRs, including VISTA, TIGIT, TIM-3, and LAG-3, and the preclinical and clinical data supporting combinatorial strategies to improve existing ICI therapies.
Collapse
Affiliation(s)
- Dia Roy
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Cassandra Gilmour
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| | - Sachin Patnaik
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
| | - Li Lily Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, Cleveland, OH, United States
- Department of Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, United States
| |
Collapse
|
25
|
Wang T, Wang K, Zhang Y, Zhang K, Cai S, Jiang S, Xiao Y, Zhang X. Novel Benzimidazoles as Potent Small-Molecule Inhibitors and Degraders of V-Domain Ig Suppressor of T-Cell Activation (VISTA). J Med Chem 2023; 66:11881-11892. [PMID: 37594853 DOI: 10.1021/acs.jmedchem.3c00484] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/20/2023]
Abstract
The V-domain Ig suppressor of T-cell activation (VISTA) is a promising negative immune checkpoint and plays a critical role in the regulation of the quiescence of naïve T lymphocytes. Most patients however do not experience durable disease control from current immune checkpoint inhibitors and discovery of inhibitors targeting novel immune checkpoints is necessary. Herein, we report our discovery and optimization of benzimidazoles as the bifunctional inhibitors of VISTA. Compound 1 is identified as a bifunctional inhibitor targeting VISTA, which shows good binding affinity to VISTA and induces VISTA degradation in HepG2 cells through an autophagic mechanism. Compound 1 rescues VISTA-mediated immunosuppression effectively and enhances antitumor activity of immune cells. 1 activates the antitumor immunity in vivo and suppresses tumor growth in a CT26 mouse model significantly. Our results show that compound 1 is a promising VISTA inhibitor and degrader and offers novel approach for cancer immunotherapy through VISTA degradation.
Collapse
Affiliation(s)
- Tianyu Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kaizhen Wang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Kuojun Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Shi Cai
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Sheng Jiang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Yibei Xiao
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| | - Xiangyu Zhang
- School of Pharmacy, China Pharmaceutical University, Nanjing 210009, China
| |
Collapse
|
26
|
Issam Salah NEI, Marnissi F, Lakhdar A, Karkouri M, ElBelhadji M, Badou A. The immune checkpoint VISTA is associated with prognosis in patients with malignant uveal melanoma. Front Immunol 2023; 14:1225140. [PMID: 37662962 PMCID: PMC10471992 DOI: 10.3389/fimmu.2023.1225140] [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: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/05/2023] Open
Abstract
Introduction Uveal melanoma (UM) is a rare yet deadly tumor. It is known for its high metastatic potential, which makes it one of the most aggressive and lethal cancers. Recently, immune checkpoints such as Programmed cell Death protein-1 (PD1) and Cytotoxic T-Lymphocyte-Associated significantly increasing patient survival in multiple human cancers, especially cutaneous melanoma. However, patients with UMs were excluded from these studies because of their molecular characteristics, which tend to be widely different from those of cutaneous melanoma. This study aimed to analyze the expression of V domain Ig Suppressor T-cell Activation (VISTA), a novel immune checkpoint, to evaluate its prognosis significance and its correlation with PD1 and CTLA-4. Methods Evaluation of VISTA, CTLA-4, and PD1 expression was performed through TCGA database analysis and immunohistochemistry using two independent cohorts with primary malignant UM. Results and discussion Our results showed that VISTA expression was associated with tumor aggressiveness, T cell exhaustion, and the shortest median overall survival among patients. Surprisingly, PD1 protein expression was negative in all patients, whereas CTLA-4 expression was high in patients with advanced stages. Our findings suggest that VISTA may be a prognostic marker and an attractive treatment strategy for immunotherapy in patients with UM. Exploring its expression profile may predict response to immunotherapy and may lead to the improvement of precision therapy in malignant uveal melanoma patients.
Collapse
Affiliation(s)
- Nour el Imane Issam Salah
- Laboratory of Research on Neurologic, Neurosensorial Diseases and Handicap, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Farida Marnissi
- Department of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd and Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Abdelhakim Lakhdar
- Laboratory of Research on Neurologic, Neurosensorial Diseases and Handicap, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Mehdi Karkouri
- Department of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd and Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Mohamed ElBelhadji
- Department of Adults Ophthalmology, 20 August Hospital 1953, CHU Ibn Rochd, Casablanca, Morocco
| | - Abdallah Badou
- Immuno-Genetics and Human Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research & Innovation, Rabat, Morocco, Mohammed VI University of Sciences and Health, Casablanca, Morocco
| |
Collapse
|
27
|
Li W, Liu Z, Jin K, Shao F, Zeng H, Wang Y, Zhu Y, Xu L, Wang Z, Chang Y, Zhang W. Immune inactivation by VISTA predicts clinical outcome and therapeutic benefit in muscle-invasive bladder cancer. BMC Cancer 2023; 23:661. [PMID: 37452272 PMCID: PMC10347783 DOI: 10.1186/s12885-023-11157-x] [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: 10/24/2022] [Accepted: 07/06/2023] [Indexed: 07/18/2023] Open
Abstract
BACKGROUND V domain Immunoglobulin suppressor of T cell activation (VISTA) has been proved to be a novel immune checkpoint molecule that positively regulates T cell infiltration in several malignancies. However, the clinical impact of VISTA on muscle-invasive bladder cancer (MIBC) patients remains relatively obscure. METHODS This study enrolled 135 MIBC patients from Zhongshan Hospital (ZSHS) and 391 patients from The Cancer Genome Atlas (TCGA) to examine the VISTA expression and immune contexture based on immunohistochemistry (IHC) staining and CIBERSORT algorithm. Additionally, IMvigor210 Cohort included 195 bladder-derived urothelial carcinoma patients to evaluate the efficacy of immunotherapy. Kaplan-Meier curve and Cox regression analyses were conducted to assess clinical outcomes. RESULTS MIBC patients with high VISTA+ immune cells (ICs) possessed poor overall survival and inferior therapeutic responsiveness to adjuvant chemotherapy (ACT), but superior responsiveness to PD-L1 inhibitor. VISTA+ ICs infiltration shaped an immunoevasive context featured by regulatory T cells (Tregs), M2 macrophages, mast cells and exhausted CD8+ T cells infiltration, with increased interleukin 10 (IL-10), transforming growth factor-β (TGF-β) and interferon-γ (IFN-γ), but also elevated T-cell immunoglobulin mucin-3 (TIM-3), lymphocyte activation gene 3 (LAG-3) and T-cell immunoglobulin and ITIM domain (TIGIT), which was also mainly presented in basal-squamous and luminal-infiltrated subtypes of MIBC. CONCLUSION VISTA+ ICs infiltration could be an independent predictor to identify poor prognosis and therapeutic responses (PD-L1 blockade and ACT) in MIBC patients, which was associated with immunoevasive contexture. The novel immune checkpoint VISTA might be utilized as a candidate treatment biomarker in MIBC patients.
Collapse
Affiliation(s)
- Wandi Li
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China
| | - Zhaopei Liu
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Kaifeng Jin
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Fei Shao
- Department of Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han Zeng
- Department of Biochemistry and Molecular Biology, NHC Key Laboratory of Glycoconjugate Research, School of Basic Medical Sciences, Fudan University, Shanghai, China
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yiwei Wang
- Department of Urology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yu Zhu
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Le Xu
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zewei Wang
- Department of Urology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Yuan Chang
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, 200032, China.
| | - Weijuan Zhang
- Department of Immunology, School of Basic Medical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
28
|
Siemsen W, Halske C, Behrens HM, Krüger S, Becker-Pauly C, Röcken C. The putative pleiotropic functions of meprin β in gastric cancer. Gastric Cancer 2023; 26:542-552. [PMID: 36976399 PMCID: PMC10284984 DOI: 10.1007/s10120-023-01385-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Accepted: 03/15/2023] [Indexed: 03/29/2023]
Abstract
BACKGROUND The gastric microbiome and inflammation play a key role in gastric cancer (GC) by regulating the immune response in a complex manner and by inflammatory events supporting carcinogenesis. Meprin β is a zinc endopeptidase and participates in tissue homeostasis, intestinal barrier function and immunological processes. It influences local inflammatory processes, dysbiosis and the microbiome. Here, we tested the hypothesis that meprin β is expressed in GC and of tumor biological significance. PATIENTS AND METHODS Four hundred forty whole mount tissue sections of patients with therapy-naive GC were stained with an anti-meprin β antibody. The histoscore and staining pattern were analyzed for each case. Following dichotomization at the median histoscore into a "low" and "high" group, the expression was correlated with numerous clinicopathological patient characteristics. RESULTS Meprin β was found intracellularly and at the cell membrane of GC. Cytoplasmic expression correlated with the phenotype according to Lauren, microsatellite instability and PD-L1 status. Membranous expression correlated with intestinal phenotype, mucin-1-, E-cadherin-, β-catenin status, mucin typus, microsatellite instability, KRAS mutation and PD-L1-positivity. Patients with cytoplasmic expression of meprin β showed a better overall and tumor-specific survival. CONCLUSIONS Meprin β is differentially expressed in GC and has potential tumor biological relevance. It might function as a tumor suppressor or promotor depending on histoanatomical site and context.
Collapse
Affiliation(s)
- Wiebke Siemsen
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, House U33, 24105, Kiel, Germany
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Christine Halske
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, House U33, 24105, Kiel, Germany
- Institute of Biochemistry, Christian-Albrechts-University, Kiel, Germany
| | - Hans-Michael Behrens
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, House U33, 24105, Kiel, Germany
| | - Sandra Krüger
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, House U33, 24105, Kiel, Germany
| | | | - Christoph Röcken
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, House U33, 24105, Kiel, Germany.
| |
Collapse
|
29
|
Zheng M, Zhang Z, Yu L, Wang Z, Dong Y, Tong A, Yang H. Immune-checkpoint protein VISTA in allergic, autoimmune disease and transplant rejection. Front Immunol 2023; 14:1194421. [PMID: 37435070 PMCID: PMC10330820 DOI: 10.3389/fimmu.2023.1194421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 05/30/2023] [Indexed: 07/13/2023] Open
Abstract
Negative checkpoint regulators (NCRs) reduce the T cell immune response against self-antigens and limit autoimmune disease development. V-domain Ig suppressor of T cell activation (VISTA), a novel immune checkpoint in the B7 family, has recently been identified as one of the NCRs. VISTA maintains T cell quiescence and peripheral tolerance. VISTA targeting has shown promising results in treating immune-related diseases, including cancer and autoimmune disease. In this review, we summarize and discuss the immunomodulatory role of VISTA, its therapeutic potential in allergic, autoimmune disease, and transplant rejection, as well as the current therapeutic antibodies, to present a new method for regulating immune responses and achieving durable tolerance for the treatment of autoimmune disease and transplantation.
Collapse
Affiliation(s)
- Meijun Zheng
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Zongliang Zhang
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Lingyu Yu
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Zeng Wang
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Yijun Dong
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Aiping Tong
- State Key Laboratory of Biotherapy, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| | - Hui Yang
- Department of Otolaryngology, Head and Neck Surgery, West China Hospital, West China Medical School, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
30
|
Chmiel P, Gęca K, Michalski A, Kłosińska M, Kaczyńska A, Polkowski WP, Pelc Z, Skórzewska M. Vista of the Future: Novel Immunotherapy Based on the Human V-Set Immunoregulatory Receptor for Digestive System Tumors. Int J Mol Sci 2023; 24:9945. [PMID: 37373091 PMCID: PMC10297928 DOI: 10.3390/ijms24129945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2023] [Revised: 05/31/2023] [Accepted: 06/07/2023] [Indexed: 06/29/2023] Open
Abstract
While gastrointestinal tumors remain a multifactorial and prevalent group of malignancies commonly treated surgically in combination with chemotherapy and radiotherapy, advancements regarding immunotherapeutic approaches continue to occur. Entering a new era of immunotherapy focused on overcoming resistance to preceding therapies caused the emergence of new therapeutic strategies. A promising solution surfaces with a V-domain Ig suppressor of T-cell activation (VISTA), a negative regulator of a T-cell function expressed in hematopoietic cells. Due to VISTA's ability to act as both a ligand and a receptor, several therapeutic approaches can be potentially developed. A broad expression of VISTA was discovered on various tumor-growth-controlling cells, which proved to increase in specific tumor microenvironment (TME) conditions, thus serving as a rationale behind the development of new VISTA-targeting. Nevertheless, VISTA's ligands and signaling pathways are still not fully understood. The uncertain results of clinical trials suggest the need for future examining inhibitor agents for VISTA and implicating a double immunotherapeutic blockade. However, more research is needed before the breakthrough can be achieved. This review discusses perspectives and novel approaches presented in the current literature. Based on the results of the ongoing studies, VISTA might be considered a potential target in combined therapy, especially for treating gastrointestinal malignancies.
Collapse
|
31
|
Reyes VE. Helicobacter pylori and Its Role in Gastric Cancer. Microorganisms 2023; 11:1312. [PMID: 37317287 PMCID: PMC10220541 DOI: 10.3390/microorganisms11051312] [Citation(s) in RCA: 37] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Revised: 05/08/2023] [Accepted: 05/15/2023] [Indexed: 06/16/2023] Open
Abstract
Gastric cancer is a challenging public health concern worldwide and remains a leading cause of cancer-related mortality. The primary risk factor implicated in gastric cancer development is infection with Helicobacter pylori. H. pylori induces chronic inflammation affecting the gastric epithelium, which can lead to DNA damage and the promotion of precancerous lesions. Disease manifestations associated with H. pylori are attributed to virulence factors with multiple activities, and its capacity to subvert host immunity. One of the most significant H. pylori virulence determinants is the cagPAI gene cluster, which encodes a type IV secretion system and the CagA toxin. This secretion system allows H. pylori to inject the CagA oncoprotein into host cells, causing multiple cellular perturbations. Despite the high prevalence of H. pylori infection, only a small percentage of affected individuals develop significant clinical outcomes, while most remain asymptomatic. Therefore, understanding how H. pylori triggers carcinogenesis and its immune evasion mechanisms is critical in preventing gastric cancer and mitigating the burden of this life-threatening disease. This review aims to provide an overview of our current understanding of H. pylori infection, its association with gastric cancer and other gastric diseases, and how it subverts the host immune system to establish persistent infection.
Collapse
Affiliation(s)
- Victor E Reyes
- Department of Pediatrics and Microbiology & Immunology, University of Texas Medical Branch, 301 University Blvd., Galveston, TX 77555-0372, USA
| |
Collapse
|
32
|
Martin AS, Molloy M, Ugolkov A, von Roemeling RW, Noelle RJ, Lewis LD, Johnson M, Radvanyi L, Martell RE. VISTA expression and patient selection for immune-based anticancer therapy. Front Immunol 2023; 14:1086102. [PMID: 36891296 PMCID: PMC9986543 DOI: 10.3389/fimmu.2023.1086102] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
V-domain Ig suppressor of T-cell activation (VISTA) is a B7 family member that plays key roles in maintaining T cell quiescence and regulation of myeloid cell populations, which together establish it as a novel immunotherapy target for solid tumors. Here we review the growing literature on VISTA expression in relation to various malignancies to better understand the role of VISTA and its interactions with both tumor cells and immune cells expressing other checkpoint molecules within the tumor microenvironment (TME). The biology of VISTA creates several mechanisms to maintain the TME, including supporting the function of myeloid-derived suppressor cells, regulating natural killer cell activation, supporting the survival of regulatory T cells, limiting antigen presentation on antigen-presenting cells and maintaining T cells in a quiescent state. Understanding these mechanisms is an important foundation of rational patient selection for anti-VISTA therapy. We provide a general framework to describe distinct patterns of VISTA expression in correlation with other known predictive immunotherapy biomarkers (programmed cell death ligand 1 and tumor-infiltrating lymphocytes) across solid tumors to facilitate investigation of the most efficacious TMEs for VISTA-targeted treatment as a single agent and/or in combination with anti-programmed death 1/anti-cytotoxic T lymphocyte antigen-4 therapies.
Collapse
Affiliation(s)
- Alexander S. Martin
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA, United States
| | | | | | | | - Randolph J. Noelle
- ImmuNext Inc., Lebanon, NH, United States
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Lionel D. Lewis
- Department of Microbiology and Immunology, Norris Cotton Cancer Center Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Melissa Johnson
- Sarah Cannon at Tennessee Oncology, Nashville, TN, United States
| | | | - Robert E. Martell
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA, United States
- Curis Inc., Lexington, MA, United States
| |
Collapse
|
33
|
Mo S, Zong L, Chen X, Ban X, Li M, Lu Z, Yu S, Chen J. Expression and Prognostic Value of B7 Family Immune Checkpoints in Pancreatic Neuroendocrine Tumors. Arch Pathol Lab Med 2023; 147:193-201. [PMID: 35671167 DOI: 10.5858/arpa.2021-0377-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/09/2021] [Indexed: 02/05/2023]
Abstract
CONTEXT.— Pancreatic neuroendocrine tumors (PanNETs) are rare malignancies with heterogeneous clinical courses requiring novel prognosticators and therapies. B7 family molecules have an important role in various cancers; however, these have not been distinguished in PanNETs. OBJECTIVE.— To investigate the expression and clinical significance of programmed death ligand-1 (PD-L1), programmed death ligand-2 (PD-L2), B7 homolog 3 (B7-H3), B7 homolog 4 (B7-H4), and V-domain immunoglobulin suppressor of T-cell activation (VISTA) in 182 PanNETs (with a high proportion of functioning versus nonfunctioning PanNETs: 51% versus 49%). DESIGN.— Molecules were immunostained by using tissue microarrays from 182 patients with grade 1/2 PanNETs. VISTA-positive microvessel density (VISTA+ MVD) was evaluated in 4 high-power fields (HPFs) (×200) and mean count was calculated; immune cells with 1% or greater VISTA staining were considered positive. PD-L1 tumoral expression was considered positive in samples with 5% or more membranous staining. Tumoral VISTA, stromal PD-L1, PD-L2, B7-H3, and B7-H4 expression were deemed positive if any staining was observed. RESULTS.— VISTA+ MVD was high (≥10.8/HPF) in 45 patients (25%), while VISTA stained positively on immune and tumor cells in 121 (66%) and 0 patients, respectively. Positive PD-L1 tumoral and stromal expression was observed in 23 (13%) and 0 patients, with positive B7-H3 expression in 76 (42%) and 98 (54%) patients, respectively, in these cells; PD-L2 and B7-H4 were not detected. PD-L1 positivity rate was high in functioning PanNETs. Stromal B7-H3 and high VISTA+ MVD correlated with unfavorable clinicopathologic features. Moreover, high VISTA+ MVD was an independent predictor of shorter progression-free survival. CONCLUSIONS.— VISTA may serve as a prognosticator and immunotherapeutic target for patients with pancreatic neuroendocrine tumor (PanNET).
Collapse
Affiliation(s)
- Shengwei Mo
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Mo and Zong contributed equally to this work
| | - Liju Zong
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China.,Mo and Zong contributed equally to this work
| | - Xianlong Chen
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Xinchao Ban
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Mei Li
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Zhaohui Lu
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Shuangni Yu
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Jie Chen
- From the Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| |
Collapse
|
34
|
Röcken C. Predictive biomarkers in gastric cancer. J Cancer Res Clin Oncol 2023; 149:467-481. [PMID: 36260159 PMCID: PMC9889517 DOI: 10.1007/s00432-022-04408-0] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 10/06/2022] [Indexed: 02/04/2023]
Abstract
Predictive biomarkers are the mainstay of precision medicine. This review summarizes the advancements in tissue-based diagnostic biomarkers for gastric cancer, which is considered the leading cause of cancer-related deaths worldwide. A disease seen in the elderly, it is often diagnosed at an advanced stage, thereby limiting therapeutic options. In Western countries, neoadjuvant/perioperative (radio-)chemotherapy is administered, and adjuvant chemotherapy is administered in the East. The morpho-molecular classification of gastric cancer has opened novel avenues identifying Epstein-Barr-Virus (EBV)-positive, microsatellite instable, genomically stable and chromosomal instable gastric cancers. In chromosomal instable tumors, receptor tyrosine kinases (RKTs) (e.g., EGFR, FGFR2, HER2, and MET) are frequently overexpressed. Gastric cancers such as microsatellite instable and EBV-positive types often express immune checkpoint molecules, such as PD-L1 and VISTA. Genomically stable tumors show alterations in claudin 18.2. Next-generation sequencing is increasingly being used to search for druggable targets in advanced palliative settings. However, most tissue-based biomarkers of gastric cancer carry the risk of a sampling error due to intratumoral heterogeneity, and adequate tissue sampling is of paramount importance.
Collapse
Affiliation(s)
- C. Röcken
- Department of Pathology, Christian-Albrechts-University, Arnold-Heller-Str. 3, Haus 14, Haus U33, 24105 Kiel, Germany
| |
Collapse
|
35
|
Jlassi A, Manai M, Morjen M, Sahraoui G, Elasmi Allal M, ELBini-Dhouib I, Naija L, Charfi L, Rejaibi R, Ben Ahmed M, Marrakchi N, Srairi-Abid N, Mezlini A, Manai M, Mrad K, Doghri R. VISTA+/CD8+ status correlates with favorable prognosis in Epithelial ovarian cancer. PLoS One 2023; 18:e0278849. [PMID: 36952478 PMCID: PMC10035885 DOI: 10.1371/journal.pone.0278849] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 11/23/2022] [Indexed: 03/25/2023] Open
Abstract
Immunotherapy by blocking immune checkpoint regulators has emerged as a new targeted therapy for some cancers. Among them V-domain Ig suppressor of Tcell activation (VISTA) which is identified as a novel checkpoint regulator in ovarian cancer. This study aimed to investigate the VISTA role in Epithelial ovarian cancer (EOC), and its relationship with tumor-infiltrating lymphocytes (TILs) markers and its prognostic value. The expression of VISTA, CD3, CD8, CD4, FOXP3, and CD56 was assessed in 168 EOC tissue microarrays (TMA) by immunohistochemistry (IHC). In addition, associations between VISTA, TILs, clinicopathological variables, and overall survival (OS) were analyzed. VISTA expression in IGRov1 cells, as well as in PBMC of EOC patient, was evaluated by western blot. VISTA expression was detected in 64,28% of tissues, among which 42.3% were positive for tumor cells (TCs), and 47,9% were positive for immune cells (ICs). In univariate analysis, VISTA expression was significantly associated with a high density of TILs:CD3+ (p = 0,001), CD4+ (p = 0,002) and CD8+ (p≤0,001), in ICs but not in TCs. In terms of OS, multivariate analysis showed a significant association between the high density of CD8+ TILs and VISTA positive staining in ICs (p = 0,044), but not in TCs (p = 0,108). Kaplan-Meier curves demonstrated no correlation between VISTA expression and prolonged OS in both ICs (p = 0,841) and TCs (p = 0,090). Classification of EOC tumor microenvironment based on VISTA and CD8+TILs expression, demonstrated four immune subtypes: VISTA+/CD8+, VISTA+/CD8-, VISTA-/CD8+ and VISTA-/CD8-. The dual positive VISTA+/CD8+ subtype was significantly associated with prolonged OS in both TCs and ICs (p = 0,012 and p≤0,01, respectively), whereas patients with VISTA+/CD8- had the worst OS. Our results showed that VISTA is highly expressed in the IGRov1 cell line and LT-CD8 from a patient with EOC. Our results highlighted the association of VISTA expression and CD8+ TILs in EOC, with prolonged OS in patients with VISTA+/CD8+ and proposed VISTA as a potential immunotherapeutic target in EOC.
Collapse
Affiliation(s)
- Aida Jlassi
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
| | - Maroua Manai
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
- Human Genetics Laboratory (LR99ES10), Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Department of Medicine, Division of Hematology-oncology, New York, New York, United States of America
| | - Maram Morjen
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Ghada Sahraoui
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | | | - Ines ELBini-Dhouib
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Lamia Naija
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Department of Surgical Oncology, Salah Aziz Institute, Tunis, Tunisia
| | - Lamia Charfi
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Rim Rejaibi
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
| | - Melika Ben Ahmed
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Laboratory of Transmission, Control and Immunobiology of Infections - LR16IPT02, Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Naziha Marrakchi
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Najet Srairi-Abid
- Laboratory of Biomolecules, Venoms and Theranostic Applications (LR20IPT01), Pasteur Institute of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Amel Mezlini
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
- Department of Medical Oncology, Salah Aziz Institute, Tunis, Tunisia
| | - Mohamed Manai
- Department of Biology, Mycology, Pathologies and Biomarkers Laboratory (LR16ES05), Faculty of Sciences of Tunis, University of Tunis El Manar, Ariana, Tunisia
| | - Karima Mrad
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| | - Raoudha Doghri
- Research Laboratory of Precision Medicine/Personalized Medicine and Oncology Investigation (LR21SP01), Tunis, Tunisia
- Department of Pathology, Salah Azaiez Institute, Tunis, Tunisia
- Faculty of Medicine of Tunis, University of Tunis, El Manar, Tunis, Tunisia
| |
Collapse
|
36
|
Rezouki I, Zohair B, Ssi SA, Karkouri M, Razzouki I, Elkarroumi M, Badou A. High VISTA expression is linked to a potent epithelial-mesenchymal transition and is positively correlated with PD1 in breast cancer. Front Oncol 2023; 13:1154631. [PMID: 37152039 PMCID: PMC10157209 DOI: 10.3389/fonc.2023.1154631] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 04/04/2023] [Indexed: 05/09/2023] Open
Abstract
Breast cancer is the most common type of tumor in women worldwide. Immune checkpoint inhibitors, particularly anti-PDL1, have shown promise as a therapeutic approach for managing this disease. However, this type of immunotherapy still fails to work for some patients, leading researchers to explore alternative immune checkpoint targets. The Ig suppressor of T cell activation domain V (VISTA) has emerged as a novel immune checkpoint that delivers inhibitory signals to T cells and has demonstrated encouraging results in various cancers. Our study investigated the association of VISTA expression with clinicopathological parameters in breast cancer patients, its involvement in the Epithelial-Mesenchymal-Transition (EMT) process, and its correlation with PD1 expression. Transcriptomic analysis revealed that VISTA was associated with lobular and metaplastic histological type, tumor size, lymph node status, ER and PR negative status, and the TNBC molecular subtype. Furthermore, VISTA expression was strongly associated with an immunosuppressive tumor microenvironment. Immunohistochemistry analysis corroborated the transcriptomic results, indicating that VISTA was expressed in most immune cells (94%) and was significantly expressed in breast cancer tumor cells compared to matched adjacent tissues. Our study also showed for the first time that VISTA overexpression in breast cancer cells could be associated with the EMT process. Additionally, we identified a positive correlation between VISTA and PD-1 expression. Together, these results highlight the immunosuppressive effect of VISTA in breast cancer patients and suggest that bi-specific targeting of VISTA and PD-1 in combination therapy could be beneficial for these patients.
Collapse
Affiliation(s)
- Ibtissam Rezouki
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Basma Zohair
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Saadia Ait Ssi
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Mehdi Karkouri
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Ibtissam Razzouki
- Laboratory of Pathological Anatomy, University Hospital Center (CHU) Ibn Rochd, Hassan II University, Casablanca, Morocco
| | - Mohamed Elkarroumi
- Department of Obstetrics and Gynecology, University Hospital Center (CHU) Ibn Rochd, Casablanca, Morocco
| | - Abdallah Badou
- Laboratory of Immunogenetics and Human Pathologies, Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
- Mohammed VI Center for Research and Innovation, Rabat, Morocco, and Mohammed VI University of Sciences and Health, Casablanca, Morocco
- *Correspondence: Abdallah Badou, ; ;
| |
Collapse
|
37
|
Catalano M, Shabani S, Venturini J, Ottanelli C, Voltolini L, Roviello G. Lung Cancer Immunotherapy: Beyond Common Immune Checkpoints Inhibitors. Cancers (Basel) 2022; 14:6145. [PMID: 36551630 PMCID: PMC9777293 DOI: 10.3390/cancers14246145] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2022] [Revised: 11/27/2022] [Accepted: 11/27/2022] [Indexed: 12/15/2022] Open
Abstract
Immunotherapy is an ever-expanding field in lung cancer treatment research. Over the past two decades, there has been significant progress in identifying immunotherapy targets and creating specific therapeutic agents, leading to a major paradigm shift in lung cancer treatment. However, despite the great success achieved with programmed death protein 1/ligand 1 (PD-1/PD-L1) monoclonal antibodies and with anti-PD-1/PD-L1 plus anti-cytotoxic T-lymphocyte antigen 4 (CTLA-4), only a minority of lung cancer patients respond to treatment, and of these many subsequently experience disease progression. In addition, immune-related adverse events sometimes can be life-threatening, especially when anti-CTLA-4 and anti-PD-1 are used in combination. All of this prompted researchers to identify novel immune checkpoints targets to overcome these limitations. Lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin (Ig) and Immunoreceptor Tyrosine-Based Inhibitory Motif (ITIM) domain (TIGIT), T cell immunoglobulin and mucin-domain containing-3 (TIM-3) are promising molecules now under investigation. This review aims to outline the current role of immunotherapy in lung cancer and to examine efficacy and future applications of the new immune regulating molecules.
Collapse
Affiliation(s)
- Martina Catalano
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Sonia Shabani
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Jacopo Venturini
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Carlotta Ottanelli
- School of Human Health Sciences, University of Florence, 50134 Florence, Italy
| | - Luca Voltolini
- Thoraco-Pulmonary Surgery Unit, Careggi University Hospital, 50134 Florence, Italy
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| | - Giandomenico Roviello
- Department of Experimental and Clinical Medicine, University of Florence, 50134 Florence, Italy
| |
Collapse
|
38
|
Mortezaee K, Majidpoor J, Najafi S. VISTA immune regulatory effects in bypassing cancer immunotherapy: Updated. Life Sci 2022; 310:121083. [DOI: 10.1016/j.lfs.2022.121083] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/04/2022] [Accepted: 10/12/2022] [Indexed: 11/07/2022]
|
39
|
Ma S, Chen F. Common strategies for effective immunotherapy of gastroesophageal cancers using immune checkpoint inhibitors. Pathol Res Pract 2022; 238:154110. [PMID: 36155325 DOI: 10.1016/j.prp.2022.154110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 08/25/2022] [Accepted: 08/31/2022] [Indexed: 11/21/2022]
Abstract
Gastroesophageal cancers (GECs) are very prevalent around the world and rank as the second cause of all cancer-related deaths in men and women and demonstrate a very poor prognosis. Currently, the treatment options for these malignancies are very limited and the response rates are also very low. Recently, immune checkpoint inhibitors (ICIs) have been proposed for immunotherapy of GECs; although preliminary results obtained from the clinical trials of ICIs in GECs were promising, they have shown to be effective only in a few subsets of patients who had a previous immune response to the tumor. In order to maximize the efficacy of ICIs in GECs, as well as identify the patients who will likely benefit from ICIs, several predictive biomarkers, such as Programmed death-ligand 1 (PD-L1) have been developed and evaluated. Since the single ICI therapies resulted in poor treatment response, several clinical studies began to explore various combinations of one or two ICIs with other anti-cancer treatment approaches, including chemotherapy, radiotherapy, and anti-angiogenesis therapy. These combinations demonstrated a more effective response among the ICIs-responsive patients and even in some instances sensitized the non-responsive individuals. This review is aimed to summarize the efforts made so far for improving the effectiveness of ICIs in the treatment of patients with GECs. Furthermore, multiple aspects of translational medicine such as available biomarkers and interactions between tumor and the immune system, as well as clinical aspects regarding the combination therapies and results of clinical trials will be discussed.
Collapse
Affiliation(s)
- Shuang Ma
- Cancer Center, Department of Pathology, Zhejiang Provincial People's Hospital (Affiliated People's Hospital, Hangzhou Medical College), Hangzhou 310014, China.
| | - Fei Chen
- Department of Gastroenterology, Tiantai People's Hospital of Zhejiang Province (Tiantai Branch of Zhejiang People's Hospital), Taizhou 317200, China.
| |
Collapse
|
40
|
Therapeutic targets and biomarkers of tumor immunotherapy: response versus non-response. Signal Transduct Target Ther 2022; 7:331. [PMID: 36123348 PMCID: PMC9485144 DOI: 10.1038/s41392-022-01136-2] [Citation(s) in RCA: 227] [Impact Index Per Article: 75.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2022] [Revised: 06/25/2022] [Accepted: 07/25/2022] [Indexed: 02/05/2023] Open
Abstract
Cancers are highly complex diseases that are characterized by not only the overgrowth of malignant cells but also an altered immune response. The inhibition and reprogramming of the immune system play critical roles in tumor initiation and progression. Immunotherapy aims to reactivate antitumor immune cells and overcome the immune escape mechanisms of tumors. Represented by immune checkpoint blockade and adoptive cell transfer, tumor immunotherapy has seen tremendous success in the clinic, with the capability to induce long-term regression of some tumors that are refractory to all other treatments. Among them, immune checkpoint blocking therapy, represented by PD-1/PD-L1 inhibitors (nivolumab) and CTLA-4 inhibitors (ipilimumab), has shown encouraging therapeutic effects in the treatment of various malignant tumors, such as non-small cell lung cancer (NSCLC) and melanoma. In addition, with the advent of CAR-T, CAR-M and other novel immunotherapy methods, immunotherapy has entered a new era. At present, evidence indicates that the combination of multiple immunotherapy methods may be one way to improve the therapeutic effect. However, the overall clinical response rate of tumor immunotherapy still needs improvement, which warrants the development of novel therapeutic designs as well as the discovery of biomarkers that can guide the prescription of these agents. Learning from the past success and failure of both clinical and basic research is critical for the rational design of studies in the future. In this article, we describe the efforts to manipulate the immune system against cancer and discuss different targets and cell types that can be exploited to promote the antitumor immune response.
Collapse
|
41
|
Kuzevanova A, Apanovich N, Mansorunov D, Korotaeva A, Karpukhin A. The Features of Checkpoint Receptor—Ligand Interaction in Cancer and the Therapeutic Effectiveness of Their Inhibition. Biomedicines 2022; 10:biomedicines10092081. [PMID: 36140182 PMCID: PMC9495440 DOI: 10.3390/biomedicines10092081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 08/14/2022] [Accepted: 08/22/2022] [Indexed: 12/12/2022] Open
Abstract
To date, certain problems have been identified in cancer immunotherapy using the inhibition of immune checkpoints (ICs). Despite the excellent effect of cancer therapy in some cases when blocking the PD-L1 (programmed death-ligand 1) ligand and the immune cell receptors PD-1 (programmed cell death protein 1) and CTLA4 (cytotoxic T-lymphocyte-associated protein 4) with antibodies, the proportion of patients responding to such therapy is still far from desirable. This situation has stimulated the exploration of additional receptors and ligands as targets for immunotherapy. In our article, based on the analysis of the available data, the TIM-3 (T-cell immunoglobulin and mucin domain-3), LAG-3 (lymphocyte-activation gene 3), TIGIT (T-cell immunoreceptor with Ig and immunoreceptor tyrosine-based inhibitory motif (ITIM) domains), VISTA (V-domain Ig suppressor of T-cell activation), and BTLA (B- and T-lymphocyte attenuator) receptors and their ligands are comprehensively considered. Data on the relationship between receptor expression and the clinical characteristics of tumors are presented and are analyzed together with the results of preclinical and clinical studies on the therapeutic efficacy of their blocking. Such a comprehensive analysis makes it possible to assess the prospects of receptors of this series as targets for anticancer therapy. The expression of the LAG-3 receptor shows the most unambiguous relationship with the clinical characteristics of cancer. Its inhibition is the most effective of the analyzed series in terms of the antitumor response. The expression of TIGIT and BTLA correlates well with clinical characteristics and demonstrates antitumor efficacy in preclinical and clinical studies, which indicates their high promise as targets for anticancer therapy. At the same time, the relationship of VISTA and TIM-3 expression with the clinical characteristics of the tumor is contradictory, and the results on the antitumor effectiveness of their inhibition are inconsistent.
Collapse
|
42
|
Establishment of a Monoclonal Antibody-Based Enzyme-Linked Immunosorbent Assay to Measure Soluble B7-H5 in Patients with Cancer. J Immunol Res 2022; 2022:3013185. [PMID: 35966819 PMCID: PMC9371821 DOI: 10.1155/2022/3013185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/18/2022] [Indexed: 11/28/2022] Open
Abstract
B7-H5, an immune checkpoint molecule, is markedly upregulated in multiple cancers and plays an important role in tumor progression and immune escape. However, the expression and significance of soluble B7-H5 (sB7-H5) in cancer remain unclear. Herein, we generated two novel mouse anti-human B7-H5 monoclonal antibodies (mAbs) 2E5 and 7B10, which had different epitopes. Based on the two mAbs, a sandwich enzyme-linked immunosorbent assay (ELISA) system was developed. Using this ELISA, we found that compared with healthy controls (HCs), sB7-H5 levels were significantly increased in the serum of patients with gastric cancer (GC), colorectal cancer (CRC), and lung cancer (LC) and were associated with TNM stage and metastasis. Receiver operating characteristic (ROC) curve analysis showed that sB7-H5 has diagnostic value for GC, CRC, and LC. Collectively, our findings delineate that sB7-H5 may be used as a predictor for diagnosis of cancer and a potential therapeutic target for cancer treatment.
Collapse
|
43
|
Tang XY, Xiong YL, Shi XG, Zhao YB, Shi AP, Zheng KF, Liu YJ, Jiang T, Ma N, Zhao JB. IGSF11 and VISTA: a pair of promising immune checkpoints in tumor immunotherapy. Biomark Res 2022; 10:49. [PMID: 35831836 PMCID: PMC9277907 DOI: 10.1186/s40364-022-00394-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Accepted: 06/22/2022] [Indexed: 11/20/2022] Open
Abstract
Immunotherapy has become the major treatment for tumors in clinical practice, but some intractable problems such as the low response rate and high rates of immune-related adverse events still hinder the progress of tumor immunotherapy. Hence, it is essential to explore additional immunotherapy treatment targets. In this review, we focus on the structure, expression and expression-related mechanisms, interactions, biological functions and the progress in preclinical/clinical research of IGSF11 and VISTA in tumors. We cover the progress in recent research with this pair of immune checkpoints in tumor immune regulation, proliferation, immune resistance and predictive prognosis. Both IGSF11 and VISTA are highly expressed in tumors and are modulated by various factors. They co-participate in the functional regulation of immune cells and the inhibition of cytokine production. Besides, in the downregulation of IGSF11 and VISTA, both inhibit the growth of some tumors. Preclinical and clinical trials all emphasize the predictive role of IGSF11 and VISTA in the prognosis of tumors, and that the predictive role of the same gene varies from tumor to tumor. At present, further research is proving the enormous potential of IGSF11 and VISTA in tumors, and especially the role of VISTA in tumor immune resistance. This may prove to be a breakthrough to solve the current clinical immune resistance, and most importantly, since research has focused on VISTA but less on IGSF11, IGSF11 may be the next candidate for tumor immunotherapy.
Collapse
Affiliation(s)
- Xi-Yang Tang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Yan-Lu Xiong
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Xian-Gui Shi
- College of Basic Medicine, Air Force Medical University, Xi'an, 710032, Shaanxi, China
| | - Ya-Bo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - An-Ping Shi
- Department of Radiology & Functional and Molecular Imaging Key Lab of Shaanxi Province, Tangdu Hospital, Fourth Military Medical University (Air Force Medical University), 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Kai-Fu Zheng
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Yu-Jian Liu
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China
| | - Tao Jiang
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| | - Nan Ma
- Department of Ophthalmology, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, 710038, Xi'an, China.
| | - Jin-Bo Zhao
- Department of Thoracic Surgery, Tangdu Hospital, Air Force Medical University, 569 Xinsi Road, Xi'an, 710038, Shaanxi, China.
| |
Collapse
|
44
|
Gupta M, Chandan K, Sarwat M. Natural Products and their Derivatives as Immune Check Point Inhibitors: Targeting Cytokine/Chemokine Signalling in Cancer. Semin Cancer Biol 2022; 86:214-232. [PMID: 35772610 DOI: 10.1016/j.semcancer.2022.06.009] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Revised: 06/15/2022] [Accepted: 06/24/2022] [Indexed: 11/29/2022]
Abstract
Cancer immunotherapy is the new generation and widely accepted form of tumour treatment. It is, however, associated with exclusive challenges which include organ-specific inflammation, and single-target strategies. Therefore, approaches that can enhance the efficiency of existing immunotherapies and expand their indications are required for the further development of immunotherapy. Natural products and medicines are stated to have this desired effect on cancer immunotherapy (adoptive immune-cells therapy, cancer vaccines, and immune-check point inhibitors). They refurbish the immunosuppressed tumour microenvironment, which is the primary location of interaction of tumour cells with the host immune system. Various immune cell subsets, via interaction with cytokine/chemokine receptors, are recruited into this microenvironment, and these subsets have roles in tumour progression and treatment responsiveness. This review summarises cytokine/chemokine signalling, types of cancer immunotherapy and the herbal medicine-derived natural products targeting cytokine/chemokines and immune checkpoints. These natural compounds possess immunomodulatory activities and exert their anti-tumour effect by either blocking the interaction or modulating the expression of the proteins linked with immune checkpoint signaling pathways. Some compounds also show a synergistic effect in combination with existing monoclonal antibody drugs to reverse the tumour microenvironment. Additionally, we have also reported some studies about the derivatives and formulations used to overcome the limitations of natural forms. This review can provide important insights for directing future research.
Collapse
Affiliation(s)
- Meenakshi Gupta
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India
| | - Kumari Chandan
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India
| | - Maryam Sarwat
- Amity Institute of Pharmacy, Amity University, Noida-201313, Uttar Pradesh, India.
| |
Collapse
|
45
|
Zhang Z, Bu L, Luo J, Guo J. Targeting protein kinases benefits cancer immunotherapy. Biochim Biophys Acta Rev Cancer 2022; 1877:188738. [PMID: 35660645 DOI: 10.1016/j.bbcan.2022.188738] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2022] [Revised: 05/16/2022] [Accepted: 05/28/2022] [Indexed: 02/07/2023]
Abstract
Small-molecule kinase inhibitors have been well established and successfully developed in the last decades for cancer target therapies. However, intrinsic or acquired drug resistance is becoming the major barrier for their clinical application. With the development of immunotherapies, in particular the discovery of immune checkpoint inhibitors (ICIs), the combination of ICIs with other therapies have recently been extensively explored, among which combination of ICIs with kinase inhibitors achieves promising clinical outcome in a plethora of cancer types. Here we comprehensively summarize the potent roles of protein kinases in modulating immune checkpoints both in tumor and immune cells, and reshaping tumor immune microenvironments by evoking innate immune response and neoantigen generation or presentation. Moreover, the clinical trial and approval of combined administration of kinase inhibitors with ICIs are collected, highlighting the precise strategies to benefit cancer immune therapies.
Collapse
Affiliation(s)
- Zhengkun Zhang
- Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China; Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Lang Bu
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China
| | - Junhang Luo
- Department of Urology, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| | - Jianping Guo
- Institute of Precision Medicine, the First Affiliated Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510080, China.
| |
Collapse
|
46
|
Immune Checkpoint Inhibitors in Cancer Therapy. Curr Oncol 2022; 29:3044-3060. [PMID: 35621637 PMCID: PMC9139602 DOI: 10.3390/curroncol29050247] [Citation(s) in RCA: 552] [Impact Index Per Article: 184.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Revised: 04/20/2022] [Accepted: 04/20/2022] [Indexed: 11/17/2022] Open
Abstract
The discovery of immune checkpoint proteins such as PD-1/PDL-1 and CTLA-4 represents a significant breakthrough in the field of cancer immunotherapy. Therefore, humanized monoclonal antibodies, targeting these immune checkpoint proteins have been utilized successfully in patients with metastatic melanoma, renal cell carcinoma, head and neck cancers and non-small lung cancer. The US FDA has successfully approved three different categories of immune checkpoint inhibitors (ICIs) such as PD-1 inhibitors (Nivolumab, Pembrolizumab, and Cemiplimab), PDL-1 inhibitors (Atezolimumab, Durvalumab and Avelumab), and CTLA-4 inhibitor (Ipilimumab). Unfortunately, not all patients respond favourably to these drugs, highlighting the role of biomarkers such as Tumour mutation burden (TMB), PDL-1 expression, microbiome, hypoxia, interferon-γ, and ECM in predicting responses to ICIs-based immunotherapy. The current study aims to review the literature and updates on ICIs in cancer therapy.
Collapse
|
47
|
Chen W, Qie C, Hu X, Wang L, Jiang J, Liu W, Liu J. A small molecule inhibitor of VSIG-8 prevents its binding to VISTA. Invest New Drugs 2022; 40:690-699. [DOI: 10.1007/s10637-022-01244-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Accepted: 03/31/2022] [Indexed: 12/19/2022]
|
48
|
Hong S, Yuan Q, Xia H, Dou Y, Sun T, Xie T, Zhang Z, He W, Dong C, Lu J, Guo L, Ni L. Establishment of an Ex Vivo Tissue Culture Model for Evaluation of Antitumor Efficacy in Clear Cell Renal Cell Carcinoma. Front Oncol 2022; 12:851191. [PMID: 35463322 PMCID: PMC9019348 DOI: 10.3389/fonc.2022.851191] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 03/03/2022] [Indexed: 12/03/2022] Open
Abstract
There are many potential immunotherapeutic targets for cancer immunotherapy, which should be assessed for efficacy before they enter clinical trials. Here we established an ex vivo cultured patient-derived tumor tissue model to evaluate antitumor effectiveness of one VISTA inhibitor, given that our previous study showed that VISTA was selectively highly expressed in human clear cell renal cell carcinoma (ccRCC) tumors. We observed that all the tested patients responded to the anti-VISTA monoclonal antibody as manifested by TNF-α production, but only a small fraction were responders to the anti-PD-1 antibody. Co-blockade of VISTA and PD-1 resulted in a synergistic effect in 20% of RCC patients. Taken together, these findings indicate that this ex vivo tumor slice culture model represents a viable tool to evaluate antitumor efficacies for the inhibitors of immune checkpoints and further supports that VISTA could serve as a promising target for immunotherapy in ccRCC.
Collapse
Affiliation(s)
- Shanjuan Hong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Qing Yuan
- Department of Urology, The Third Medical Center of Chinese Peoples Liberation Army (PLA) General Hospital, Beijing, China
| | - Haizhui Xia
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Yuan Dou
- R&D Center, Suzhou Kanova Biopharmaceutical Co., Ltd., Suzhou, China
| | - Tiantian Sun
- R&D Center, Suzhou Kanova Biopharmaceutical Co., Ltd., Suzhou, China
| | - Tian Xie
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
| | - Zhiyin Zhang
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Wei He
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Center for Human Disease Immuno-Monitoring, Beijing Friendship Hospital, Beijing, China
| | - Jian Lu
- Department of Urology, Peking University Third Hospital, Beijing, China
| | - Li Guo
- R&D Center, Suzhou Kanova Biopharmaceutical Co., Ltd., Suzhou, China
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing, China
- Center for Human Disease Immuno-Monitoring, Beijing Friendship Hospital, Beijing, China
| |
Collapse
|
49
|
Biggi AFB, Elgui de Oliveira D. The Epstein-Barr Virus Hacks Immune Checkpoints: Evidence and Consequences for Lymphoproliferative Disorders and Cancers. Biomolecules 2022; 12:397. [PMID: 35327589 PMCID: PMC8946074 DOI: 10.3390/biom12030397] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/25/2022] [Accepted: 03/02/2022] [Indexed: 12/12/2022] Open
Abstract
The Epstein-Barr Virus (EBV) is a gammaherpesvirus involved in the etiopathogenesis of a variety of human cancers, mostly of lymphoid and epithelial origin. The EBV infection participates in both cell transformation and tumor progression, also playing an important role in subverting immune responses against cancers. The homeostasis of the immune system is tightly regulated by inhibitory mechanisms affecting key immune effectors, such as T lymphocytes and NK cells. Collectively known as immune checkpoints, these mechanisms rely on a set of cellular receptors and ligands. These molecules may be candidate targets for immune checkpoints blockade-an emergent and promising modality of immunotherapy already proven to be valuable for a variety of human cancers. The EBV was lately suspected to interfere with the expression of immune checkpoint molecules, notably PD-1 and its ligands, found to be overexpressed in cases of Hodgkin lymphoma, nasopharyngeal, and gastric adenocarcinomas associated with the viral infection. Even though there is compelling evidence showing that the EBV interferes with other immune checkpoint regulators (e.g., CTLA-4, LAG-3, TIM-3, and VISTA), the published data are still scarce. Herein, we discuss the current state of the knowledge on how the EBV interferes with the activity of immune checkpoints regulators, as well as its implications considering the immune checkpoints blockade for clinical management of the EBV-associated malignancies, notably lymphomas.
Collapse
Affiliation(s)
| | - Deilson Elgui de Oliveira
- Department of Pathology, Medical School, São Paulo State University (UNESP), Botucatu 18618-687, SP, Brazil
- ViriCan, Institute for Biotechnology (IBTEC), São Paulo State University (UNESP), Botucatu 18607-440, SP, Brazil
| |
Collapse
|
50
|
Schrumpf T, Behrens HM, Haag J, Krüger S, Röcken C. FGFR2 overexpression and compromised survival in diffuse-type gastric cancer in a large central European cohort. PLoS One 2022; 17:e0264011. [PMID: 35167603 PMCID: PMC8846517 DOI: 10.1371/journal.pone.0264011] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Accepted: 02/01/2022] [Indexed: 12/24/2022] Open
Abstract
The significance of fibroblast growth factor receptor 2 (FGFR2) in gastric cancer (GC) has been studied predominantly in Asian patient cohorts. Data on White patients are scarce. Here, we aimed to independently validate the expression and putative tumor biological significance of FGFR2 in a large non-Asian GC cohort. Immunohistochemistry (IHC) was performed on large-area tissue sections from 493 patients with GC and evaluated using the HScore. GCs with moderate and strong FGFR2 expression were studied for Fgfr2 amplification using chromogenic in situ hybridization (CISH). Median overall survival was determined using the Kaplan–Meier method. The majority [240 (99.1%)] of FGFR2-positive GCs showed a variable combination of staining intensities with marked intratumoral heterogeneity, including weak [198 (40.2%) cases], moderate [145 (29.4%)], and strong [108 (21.9%)] staining in diverse combinations. 250 (50.9%) GCs expressed no FGFR2. Fgfr2 gene amplification was found in 40% of selected cases with high protein expression and was also heterogeneous at the cell level. FGFR2 protein expression did not correlate with patient survival in the entire cohort However, using different cutoff values, a negative correlation between FGFR2-expression and patient outcome was found for diffuse-type GC. FGFR2 expression was associated with a lower tumor grade and intestinal phenotype (p≤0.0001). FGFR2–positive diffuse-type GCs classify a small subset of patients with a poor tumor specific survival (5.29±1.3 vs. 14.67±1.9 months; p = 0.004).
Collapse
Affiliation(s)
- Thorben Schrumpf
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Hans-Michael Behrens
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Jochen Haag
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Sandra Krüger
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
| | - Christoph Röcken
- Dept. of Pathology, Christian-Albrechts-University, University Hospital Schleswig-Holstein, Kiel, Germany
- * E-mail:
| |
Collapse
|