1
|
Li L, Pu H, Zhang X, Guo X, Li G, Zhang M. Resistance to PD-1/PD-L1 immune checkpoint blockade in advanced non-small cell lung cancer. Crit Rev Oncol Hematol 2025; 209:104683. [PMID: 40024354 DOI: 10.1016/j.critrevonc.2025.104683] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 03/04/2025] Open
Abstract
Lung cancer is one of the most common malignant tumors, of which non-small cell lung cancer (NSCLC) accounts for about 85 %. Although immune checkpoint inhibitors (ICIs), particularly PD-1/PD-L1 inhibitors, have significantly improved the prognosis of patients with NSCLC. There are still many patients do not benefit from ICIs. Primary resistance remains a major challenge in advanced NSCLC. The cancer-immunity cycle describes the process from antigen release to T cell recognition and killing of the tumor, which provides a framework for understanding anti-tumor immunity. The classical cycle consists of seven steps, and alterations at each stage can result in resistance. This review examines the current status of PD-1/PD-L1 blockade in the treatment of advanced NSCLC and explores potential mechanisms of resistance. We summarize the latest clinical trials of PD-1/PD-L1 inhibitors combined with other therapies and explore potential targets for overcoming primary resistance to PD-1/PD-L1 inhibitors.
Collapse
Affiliation(s)
- Lijun Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Haihong Pu
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaoxin Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Xiaotian Guo
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Guangrui Li
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| | - Minghui Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
2
|
Chuang CH, Guo JC, Kato K, Hsu CH. Exploring novel immunotherapy in advanced esophageal squamous cell carcinoma: Is targeting TIGIT an answer? Esophagus 2025; 22:139-147. [PMID: 39847233 PMCID: PMC11929690 DOI: 10.1007/s10388-024-01105-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/26/2024] [Indexed: 01/24/2025]
Abstract
Esophageal squamous cell carcinoma (ESCC) is a prevalent and highly lethal malignancy in Asia. Recent advancements in immune checkpoint inhibitors (ICIs) have markedly transformed the systemic therapy landscape for ESCC. Anti-PD-1-based combination with chemotherapy or with ipilimumab, an anti-CTLA-4 antibody, have been established as the new standard first-line treatments for patients with advanced ESCC. Moreover, anti-PD-1 monotherapy has demonstrated improved efficacy and survival compared with second-line chemotherapy in previously treated patients with ESCC. Novel ICIs targeting other immune checkpoints also show potential for enhancing anticancer therapy in advanced ESCC.The TIGIT/PVR pathway represents a new immune checkpoint. Preclinical studies have indicated that the dual blockade of TIGIT and PD-1 can enhance antitumor immune responses. Clinical trials have reported that combining anti-TIGIT with anti-PD-1/PD-L1 antibodies elicited clinical responses in patients with advanced ESCC. In the first-line systemic therapy setting, combinations of dual ICIs targeting TIGIT and PD-1/PD-L1 plus platinum-based chemotherapy have demonstrated acceptable toxicity profiles and promising antitumor activity in several phase II trials and one phase III study. However, the role of adding an anti-TIGIT antibody to the current standard of anti-PD-1/PD-L1 plus platinum-based chemotherapy in first-line therapy for advanced ESCC remains to be fully determined, necessitating further clinical trials. Ongoing studies are also investigating the role of anti-TIGIT, with or without anti-PD-1/PD-L1, in locoregional ESCC. Additional research is essential to optimize the potential of anti-TIGIT therapy in ESCC and other malignancies by identifying predictive biomarkers, determining optimal antibody types, and gaining key mechanistic insights.
Collapse
Affiliation(s)
- Chien-Huai Chuang
- Department of Medical Oncology, National Taiwan University Cancer Center, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan
| | - Jhe-Cyuan Guo
- Department of Medical Oncology, National Taiwan University Cancer Center, 7 Chung-Shan South Road, Taipei, 10002, Taiwan
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-Ku, Tokyo, 104-0045, Japan.
- Department of Gastrointestinal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan.
| | - Chih-Hung Hsu
- Department of Medical Oncology, National Taiwan University Cancer Center, 7 Chung-Shan South Road, Taipei, 10002, Taiwan.
- Department of Oncology, National Taiwan University Hospital, Taipei, Taiwan.
- Graduate Institute of Oncology, National Taiwan University College of Medicine, Taipei, Taiwan.
| |
Collapse
|
3
|
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
|
4
|
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
|
5
|
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
|
6
|
Kidacki M, Cho C, Lopez-Giraldez F, Huang B, He J, Gaule P, Chen L, Vesely MD. PD-1H (VISTA) Expression in Cutaneous Squamous Cell Carcinoma Is Correlated with T-Cell Infiltration and Activation. J Invest Dermatol 2025:S0022-202X(25)00116-2. [PMID: 39983979 DOI: 10.1016/j.jid.2025.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/23/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is one of the most common human cancers, with an estimated death rate approaching or exceeding that of melanoma. Immune inhibitory receptor antagonism through the blockade of PD-1 or its ligand, PD-L1, has revolutionized the treatment of cSCC; however, approximately half of patients still fail to respond. The inhibitory receptor PD-1H (also known as VISTA) controls T-cell and myeloid cell functions in preclinical cancer studies. Currently, cancer clinical trials using anti-VISTA-blocking antibodies are ongoing. We sought to determine the extent of VISTA expression in cSCCs and to correlate its expression with PD-L1 expression. Using multiplexed quantitative immunofluorescence of primary cSCC tissues (n = 76), we found that VISTA was expressed in 48% of cSCCs and that PD-L1 was expressed in 58% of cSCCs. We found that high VISTA expression, more than PD-L1 expression, correlated with greater T-cell infiltration and activation, as measured by the proliferation marker Ki-67 and the cytotoxic marker granzyme B. Furthermore, there was no significant correlation between VISTA and PD-L1 coexpression within the same cSCCs, suggesting that individual tumors have distinct immunosuppressive microenvironments. These findings provide a rationale for the targeting of VISTA in cSCC immunotherapy.
Collapse
Affiliation(s)
- Michal Kidacki
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Cho
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Francesc Lopez-Giraldez
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA; Yale Center for Genome Analysis (YCGA), Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Baozhu Huang
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jianwei He
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lieping Chen
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Internal Medicine (Oncology), Yale School of Medicine, New Haven, Connecticut, USA
| | - Matthew D Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
7
|
Cesano A, Augustin R, Barrea L, Bedognetti D, Bruno TC, Carturan A, Hammer C, Ho WS, Kather JN, Kirchhoff T, Lu RO, McQuade J, Najjar YG, Pietrobon V, Ruella M, Shen R, Soldati L, Spencer C, Betof Warner A, Warren S, Ziv E, Marincola FM. Advances in the understanding and therapeutic manipulation of cancer immune responsiveness: a Society for Immunotherapy of Cancer (SITC) review. J Immunother Cancer 2025; 13:e008876. [PMID: 39824527 PMCID: PMC11749597 DOI: 10.1136/jitc-2024-008876] [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: 01/19/2024] [Accepted: 12/12/2024] [Indexed: 01/20/2025] Open
Abstract
Cancer immunotherapy-including immune checkpoint inhibition (ICI) and adoptive cell therapy (ACT)-has become a standard, potentially curative treatment for a subset of advanced solid and liquid tumors. However, most patients with cancer do not benefit from the rapidly evolving improvements in the understanding of principal mechanisms determining cancer immune responsiveness (CIR); including patient-specific genetically determined and acquired factors, as well as intrinsic cancer cell biology. Though CIR is multifactorial, fundamental concepts are emerging that should be considered for the design of novel therapeutic strategies and related clinical studies. Recent advancements as well as novel approaches to address the limitations of current treatments are discussed here, with a specific focus on ICI and ACT.
Collapse
Affiliation(s)
| | - Ryan Augustin
- University of Pittsburgh Department of Medicine, Pittsburgh, Pennsylvania, USA
- Mayo Clinic, Rochester, Minnesota, USA
| | | | | | - Tullia C Bruno
- University of Pittsburgh, UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | | | - Winson S Ho
- University of California San Francisco, San Francisco, California, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Tomas Kirchhoff
- Laura and Isaac Perlmutter Cancer Center, New York University Grossman School of Medicine, New York University Langone Health, New York, NY, USA
| | - Rongze O Lu
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, California, USA
| | - Jennifer McQuade
- University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | - Yana G Najjar
- UPMC Hillman Cancer Center, Pittsburgh, Pennsylvania, USA
| | | | - Marco Ruella
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rhine Shen
- Kite Pharma Inc, Santa Monica, California, USA
| | | | - Christine Spencer
- Parker Institute for Cancer Immunotherapy, San Francisco, California, USA
| | | | | | - Elad Ziv
- University of California San Francisco, San Francisco, California, USA
| | | |
Collapse
|
8
|
Duan Y, Ren X, Guo X, Xie J, Liu Z, Li L. VISTA in hematological malignancies: a review of the literature. Front Immunol 2024; 15:1466839. [PMID: 39742253 PMCID: PMC11685136 DOI: 10.3389/fimmu.2024.1466839] [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: 07/18/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, tumor immunotherapy has become an active research area, with the emergence of immune checkpoint inhibitors (ICIs) revolutionizing immunotherapy. Clinical evidence indicates that programmed cell death protein 1 (PD-1) monoclonal antibodies and other drugs have remarkable therapeutic effects. V-domain Ig suppressor of T-cell activation (VISTA) is a new type of immune checkpoint receptor that is highly expressed in various tumors. It is co-expressed with PD-1, T-cell immunoglobulin domain, mucin domain-3 (Tim-3), T-cell immunoglobulin, and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) and is associated with prognosis, which suggests that it may be a target for immunotherapy. As an immune checkpoint receptor with no mature drugs, VISTA is highly expressed in acute myeloid leukemia (AML), multiple myeloma (MM), and other hematological malignancies; however, its pathogenic mechanism should be defined to better guide treatment.
Collapse
Affiliation(s)
- Yuanjia Duan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Xiaotong Ren
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Xinyu Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Jiayi Xie
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| |
Collapse
|
9
|
Gayen S, Mukherjee S, Dasgupta S, Roy S. Emerging druggable targets for immune checkpoint modulation in cancer immunotherapy: the iceberg lies beneath the surface. Apoptosis 2024; 29:1879-1913. [PMID: 39354213 DOI: 10.1007/s10495-024-02022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
The immune system serves as a fundamental defender against the initiation and progression of cancer. Failure of the immune system augments immunosuppressive action that leading to cancer manifestation. This immunosuppressive effect causes from significant alterations in immune checkpoint expression associated with tumoral progression. The tumor microenvironment promotes immune escape mechanisms that further amplifying immunosuppressive actions. Notably, substantial targeting of immune checkpoints has been pragmatic in the advancement of cancer research. This study highlights a comprehensive review of emerging druggable targets aimed at modulating immune checkpoint co-inhibitory as well as co-stimulatory molecules in response to immune system activation. This modulation has prompted to the development of newer therapeutic insights, eventually inducing immunogenic cell death through immunomodulatory actions. The study emphasizes the role of immune checkpoints in immunogenic regulation of cancer pathogenesis and explores potential therapeutic avenues in cancer immunotherapy.Modulation of Immunosuppressive and Immunostimulatory pathways of immune checkpoints in cancer immunotherapy.
Collapse
Affiliation(s)
- Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sandipan Dasgupta
- Department of Pharmaceutical Technology, Maulana Abul Kalam Azad University of Technology, Kolkata, West Bengal, 741249, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
10
|
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
|
11
|
Ai K, Chen M, Liang Z, Ding X, Gao Y, Zhang H, Wu S, He Y, Li Y. Inhibition of Tumoral VISTA to Overcome TKI Resistance via Downregulation of the AKT/mTOR and JAK2/STAT5 Pathways in Chronic Myeloid Leukemia. Biomol Ther (Seoul) 2024; 32:582-600. [PMID: 39104205 PMCID: PMC11392669 DOI: 10.4062/biomolther.2024.017] [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: 01/22/2024] [Revised: 04/06/2024] [Accepted: 06/10/2024] [Indexed: 08/07/2024] Open
Abstract
Tyrosine kinase inhibitors (TKIs) have revolutionized the treatment landscape for chronic myeloid leukemia (CML). However, TKI resistance poses a significant challenge, leading to treatment failure and disease progression. Resistance mechanisms include both BCR::ABL1-dependent and BCR::ABL1-independent pathways. The mechanisms underlying BCR::ABL1 independence remain incompletely understood, with CML cells potentially activating alternative signaling pathways, including the AKT/mTOR and JAK2/STAT5 pathways, to compensate for the loss of BCR::ABL1 kinase activity. This study explored tumoral VISTA (encoded by VSIR) as a contributing factor to TKI resistance in CML patients and identified elevated tumoral VISTA levels as a marker of resistance and poor survival. Through in vitro and in vivo analyses, we demonstrated that VSIR knockdown and the application of NSC-622608, a novel VISTA inhibitor, significantly impeded CML cell proliferation and induced apoptosis by attenuating the AKT/ mTOR and JAK2/STAT5 pathways, which are crucial for CML cell survival independent of BCR::ABL1 kinase activity. Moreover, VSIR overexpression promoted TKI resistance in CML cells. Importantly, the synergistic effect of NSC-622608 with TKIs offers a potent therapeutic avenue against both imatinib-sensitive and imatinib-resistant CML cells, including those harboring the challenging T315I mutation. Our findings highlight the role of tumoral VISTA in mediating TKI resistance in CML, suggesting that inhibition of VISTA, particularly in combination with TKIs, is an innovative approach to enhancing treatment outcomes in CML patients, irrespective of BCR::ABL1 mutation status. This study not only identified a new pathway contributing to TKI resistance but also revealed the possibility of targeting tumoral VISTA as a means of overcoming this significant clinical challenge.
Collapse
Affiliation(s)
- Kexin Ai
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Mu Chen
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zhao Liang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiangyang Ding
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yang Gao
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Honghao Zhang
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Suwan Wu
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yanjie He
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yuhua Li
- Department of Hematology, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
- Bioland Laboratory (Guangzhou Regenerative Medicine and Health Guangdong Laboratory), Guangzhou 510280, China
| |
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
|
Du G, Dou C, Sun P, Wang S, Liu J, Ma L. Regulatory T cells and immune escape in HCC: understanding the tumor microenvironment and advancing CAR-T cell therapy. Front Immunol 2024; 15:1431211. [PMID: 39136031 PMCID: PMC11317284 DOI: 10.3389/fimmu.2024.1431211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Liver cancer, which most commonly manifests as hepatocellular carcinoma (HCC), is the sixth most common cancer in the world. In HCC, the immune system plays a crucial role in the growth and proliferation of tumor cells. HCC achieve immune escape through the tumor microenvironment, which significantly promotes the development of this cancer. Here, this article introduces and summarizes the functions and effects of regulatory T cells (Tregs) in the tumor microenvironment, highlighting how Tregs inhibit and regulate the functions of immune and tumor cells, cytokines, ligands and receptors, etc, thereby promoting tumor immune escape. In addition, it discusses the mechanism of CAR-T therapy for HCC and elaborate on the relationship between CAR-T and Tregs.
Collapse
Affiliation(s)
- Guangtan Du
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Cunmiao Dou
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Peng Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Leina Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
14
|
Zhan X, Li J, Ding Y, Zhou F, Zeng R, Lei L, Zhang Y, Feng A, Qu Y, Yang Z. Pyroptosis-related long-noncoding RNA signature predicting survival and immunotherapy efficacy in patients with lung squamous cell carcinoma. Clin Exp Med 2024; 24:145. [PMID: 38960987 PMCID: PMC11222204 DOI: 10.1007/s10238-024-01409-w] [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: 06/04/2024] [Accepted: 06/19/2024] [Indexed: 07/05/2024]
Abstract
Pyroptosis-related long-noncoding RNAs (PRlncRNAs) play an important role in cancer progression. However, their role in lung squamous cell carcinoma (LUSC) is unclear. A risk model was constructed using the least absolute shrinkage and selection operator (LASSO) Cox regression analysis based on RNA sequencing data from The Cancer Genome Atlas database. The LUSC cohort was divided into high- and low-risk groups based on the median risk score. For the prognostic value of the model, the Kaplan-Meier analysis, log-rank test, and Cox regression analysis were performed. A nomogram was constructed to predict the prognosis of patients, using a risk score and clinical parameters such as age, sex, clinical stage, and tumor node metastasis classification (TNM) stage. Afterwards, six common algorithms were employed to assess the invasion of immune cells. The Gene Set Enrichment Analysis (GSEA) was conducted to identify differences between patients at high and low risk. Furthermore, the pRRophetic package was employed to forecast the half-maximal inhibitory doses of prevalent chemotherapeutic drugs, while the tumor immune dysfunction and exclusion score was computed to anticipate the response to immunotherapy. The expression levels of the seven PRlncRNAs were examined in both LUSC and normal lung epithelial cell lines using RT-qPCR. Proliferation, migration, and invasion assays were also carried out to investigate the role of MIR193BHG in LUSC cells. Patients in the low-risk group showed prolonged survival in the total cohort or subgroup analysis. The Cox regression analysis showed that the risk model could act as an independent prognostic factor for patients with LUSC. The results of GSEA analysis revealed that the high-risk group showed enrichment of cytokine pathways, Janus tyrosine kinase/signal transducer and activator of the transcription signalling pathway, and Toll-like receptor pathway. Conversely, the low-risk group showed enrichment of several gene repair pathways. Furthermore, the risk score was positively correlated with immune cell infiltration. Moreover, patients in the high-risk category showed reduced responsiveness to conventional chemotherapeutic medications and immunotherapy. The majority of the long noncoding RNAs in the risk model were confirmed to be overexpressed in LUSC cell lines compared to normal lung epithelial cell lines by in vitro tests. Further studies have shown that downregulating the expression of MIR193BHG may inhibit the growth, movement, and infiltration capabilities of LUSC cells, whereas increasing the expression of MIR193BHG could enhance these malignant tendencies. This study found that PRlncRNAs were linked to the prognosis of LUSC patients. The risk model, evaluated across various clinical parameters and treatment modalities, shows potential as a future reference for clinical applications.
Collapse
Affiliation(s)
- Xiang Zhan
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Jixian Li
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Yi Ding
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Fengge Zhou
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Renya Zeng
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Lingli Lei
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Ying Zhang
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Alei Feng
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Yan Qu
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Zhe Yang
- Tumor Research and Therapy Center, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
- Tumor Research and Therapy Center, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
| |
Collapse
|
15
|
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
|
16
|
Konen JM, Wu H, Gibbons DL. Immune checkpoint blockade resistance in lung cancer: emerging mechanisms and therapeutic opportunities. Trends Pharmacol Sci 2024; 45:520-536. [PMID: 38744552 PMCID: PMC11189143 DOI: 10.1016/j.tips.2024.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 04/17/2024] [Accepted: 04/17/2024] [Indexed: 05/16/2024]
Abstract
Immune checkpoint blockade (ICB) therapy works by inhibiting suppressive checkpoints that become upregulated after T cell activation, like PD-1/PD-L1 and CTLA-4. While the initial FDA approvals of ICB have revolutionized cancer therapies and fueled a burgeoning immuno-oncology field, more recent clinical development of new agents has been slow. Here, focusing on lung cancer, we review the latest research uncovering tumor cell intrinsic and extrinsic ICB resistance mechanisms as major hurdles to treatment efficacy and clinical progress. These include genomic and non-genomic tumor cell alterations, along with host and microenvironmental factors like the microbiome, metabolite accumulation, and hypoxia. Together, these factors can cooperate to promote immunosuppression and ICB resistance. Opportunities to prevent resistance are constantly evolving in this rapidly expanding field, with the goal of moving toward personalized immunotherapeutic regimens.
Collapse
Affiliation(s)
- Jessica M Konen
- Department of Hematology and Medical Oncology, Emory University, Atlanta, GA, USA.
| | - Haoyi Wu
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Don L Gibbons
- Department of Thoracic/Head and Neck Medical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA; Department of Molecular and Cellular Oncology, University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
17
|
Pan J, Mahsud I, Ul Haq M, Khan S, Alhomrani M, Alamri AS, Alghamdi SA, ALSuhaymi N, Baothman BK, Almaghrabi S, Ullah S, Jamil M. Comprehensive pan-cancer analysis reveals VSIR as a candidate immunologic, diagnostic, and prognostic biomarker. Am J Transl Res 2024; 16:1630-1642. [PMID: 38883368 PMCID: PMC11170614 DOI: 10.62347/jmbz8836] [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: 11/03/2023] [Accepted: 04/22/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVES Being a checkpoint, the expression level of V-set immunoregulatory receptor (VSIR) serves as an indicator of the extent of immunosuppression. Our objective was to undertake a pan-cancer analysis to examine the expression, genetic alterations, prognosis, and immunologic features associated with VSIR. METHODS The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), GEPIA2, UALCAN, OncoDB, Human Protein Atlas (HPA), STRING, DAVID, cell culture, clinical sample collection, and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used. RESULTS This study comprehensively assessed VSIR across 33 cancers using TCGA and GTEx databases. Differential expression analysis revealed elevated VSIR in several cancers, notably in cholangiocarcinoma, esophageal carcinoma, kidney renal cell carcinoma, and liver hepatocellular carcinoma, while decreased expression was observed in various others. Prognostic analysis highlighted its significant association with reduced overall survival (OS) in ESCA and LIHC. Investigation into cancer stages demonstrated a correlation between VSIR expression and stage in ESCA and LIHC. Promoter methylation analysis indicated decreased VSIR methylation levels in tumors, implicating a role in oncogenesis. Furthermore, subcellular localization predictions, Tumor Mutational Burden (TMB), and Microsatellite Instability (MSI) correlations revealed intriguing insight into VSIR's function. Notably, a positive correlation was identified between VSIR expression and various immune cells in both cancers. Protein-protein interaction (PPI) network construction and gene enrichment analysis elucidated VSIR-associated dysregulated pathways, emphasizing its possible involvement in diverse pathways. Finally, experimental validation using LIHC clinical samples and cell lines confirmed elevated VSIR expression, supporting its oncogenic role. CONCLUSION Collectively, these findings present a comprehensive understanding of VSIR's diverse roles and potential clinical implications in ESCA and LIHC.
Collapse
Affiliation(s)
- Jun Pan
- Department of Thoracic Surgery/Cardiovascular Surgery, The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University Hangzhou 311200, Zhejiang, China
| | | | - Moeen Ul Haq
- Gastroenterology Department, Gomal Medical College/MTI D.I.Khan Pakistan
| | | | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University Taif, Saudi Arabia
- Research Centre for Health Sciences, Taif University Taif, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University Taif, Saudi Arabia
- Research Centre for Health Sciences, Taif University Taif, Saudi Arabia
| | - Saleh A Alghamdi
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University Taif, Saudi Arabia
| | - Naif ALSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences - AlQunfudah, Umm Al-Qura University Mekkah, Saudi Arabia
| | - Bandar K Baothman
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences in Rabigh, King Abdulaziz University Jeddah 21589, Saudi Arabia
| | - Sarah Almaghrabi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University Jeddah 21589, Saudi Arabia
- Center of Innovations in Personalized Medicine (CIPM), King Abdulaziz University 21589 Jeddah, Saudi Arabia
| | | | - Muhammad Jamil
- PARC Arid Zone Research Center Dera Ismail Khan 29050, Pakistan
| |
Collapse
|
18
|
Chen L, Zhao X, Liu X, Ouyang Y, Xu C, Shi Y. Development of small molecule drugs targeting immune checkpoints. Cancer Biol Med 2024; 21:j.issn.2095-3941.2024.0034. [PMID: 38727005 PMCID: PMC11131045 DOI: 10.20892/j.issn.2095-3941.2024.0034] [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: 01/17/2024] [Accepted: 03/28/2024] [Indexed: 05/29/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) are used to relieve and refuel anti-tumor immunity by blocking the interaction, transcription, and translation of co-inhibitory immune checkpoints or degrading co-inhibitory immune checkpoints. Thousands of small molecule drugs or biological materials, especially antibody-based ICIs, are actively being studied and antibodies are currently widely used. Limitations, such as anti-tumor efficacy, poor membrane permeability, and unneglected tolerance issues of antibody-based ICIs, remain evident but are thought to be overcome by small molecule drugs. Recent structural studies have broadened the scope of candidate immune checkpoint molecules, as well as innovative chemical inhibitors. By way of comparison, small molecule drug-based ICIs represent superior oral bioavailability and favorable pharmacokinetic features. Several ongoing clinical trials are exploring the synergetic effect of ICIs and other therapeutic strategies based on multiple ICI functions, including immune regulation, anti-angiogenesis, and cell cycle regulation. In this review we summarized the current progression of small molecule ICIs and the mechanism underlying immune checkpoint proteins, which will lay the foundation for further exploration.
Collapse
Affiliation(s)
- Luoyi Chen
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xinchen Zhao
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Xiaowei Liu
- Institute for Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yujie Ouyang
- Acupuncture and Massage College, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Chuan Xu
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Ying Shi
- Department of Oncology, Sichuan Academy of Medical Sciences, Sichuan Provincial People’s Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| |
Collapse
|
19
|
Zhang T, Yu W, Cheng X, Yeung J, Ahumada V, Norris PC, Pearson MJ, Yang X, van Deursen W, Halcovich C, Nassar A, Vesely MD, Zhang Y, Zhang JP, Ji L, Flies DB, Liu L, Langermann S, LaRochelle WJ, Humphrey R, Zhao D, Zhang Q, Zhang J, Gu R, Schalper KA, Sanmamed MF, Chen L. Up-regulated PLA2G10 in cancer impairs T cell infiltration to dampen immunity. Sci Immunol 2024; 9:eadh2334. [PMID: 38669316 DOI: 10.1126/sciimmunol.adh2334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 10/19/2023] [Accepted: 04/04/2024] [Indexed: 04/28/2024]
Abstract
T cells are often absent from human cancer tissues during both spontaneously induced immunity and therapeutic immunotherapy, even in the presence of a functional T cell-recruiting chemokine system, suggesting the existence of T cell exclusion mechanisms that impair infiltration. Using a genome-wide in vitro screening platform, we identified a role for phospholipase A2 group 10 (PLA2G10) protein in T cell exclusion. PLA2G10 up-regulation is widespread in human cancers and is associated with poor T cell infiltration in tumor tissues. PLA2G10 overexpression in immunogenic mouse tumors excluded T cells from infiltration, resulting in resistance to anti-PD-1 immunotherapy. PLA2G10 can hydrolyze phospholipids into small lipid metabolites, thus inhibiting chemokine-mediated T cell mobility. Ablation of PLA2G10's enzymatic activity enhanced T cell infiltration and sensitized PLA2G10-overexpressing tumors to immunotherapies. Our study implicates a role for PLA2G10 in T cell exclusion from tumors and suggests a potential target for cancer immunotherapy.
Collapse
Affiliation(s)
- Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Weiwei Yu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Xiaoxiao Cheng
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jacky Yeung
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Neurosurgery, Yale University School of Medicine, New Haven, CT, USA
| | - Viviana Ahumada
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | - Xuan Yang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | - Christina Halcovich
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Ala Nassar
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Matthew D. Vesely
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Department of Dermatology, Yale University School of Medicine, New Haven, CT, USA
| | - Yu Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jian-Ping Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Lan Ji
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | | | | | | | | | | | - Dejian Zhao
- Yale Center for Genome Analysis, Yale University, New Haven, CT, USA
| | - Qiuyu Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Jindong Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Runxia Gu
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| | - Kurt A Schalper
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| | - Miguel F Sanmamed
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
- Program of Immunology and Immunotherapy, Center for Applied Medical Research, University of Navarra, Pamplona, Spain
| | - Lieping Chen
- Department of Immunobiology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
20
|
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
|
21
|
Nishizaki D, Kurzrock R, Miyashita H, Adashek JJ, Lee S, Nikanjam M, Eskander RN, Patel H, Botta GP, Nesline MK, Pabla S, Conroy JM, DePietro P, Sicklick JK, Kato S. Viewing the immune checkpoint VISTA: landscape and outcomes across cancers. ESMO Open 2024; 9:102942. [PMID: 38503143 PMCID: PMC10966162 DOI: 10.1016/j.esmoop.2024.102942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Optimizing immune checkpoint inhibitor (ICI) therapy may require identification of co-targetable checkpoint pathways via immune profiling. Herein, we analyzed the transcriptomic expression and clinical correlates of V-domain immunoglobulin suppressor of T-cell activation (VISTA), a promising targetable checkpoint. PATIENTS AND METHODS RNA sequencing was carried out on 514 tissues reflecting diverse advanced/metastatic cancers. Expression of eight immune checkpoint markers [lymphocyte-activation gene 3 (LAG-3), tumor necrosis factor receptor superfamily 14 (TNFRSF14), programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), programmed death-ligand 2 (PD-L2), B- and T-lymphocyte attenuator (BTLA), T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), cytotoxic T-lymphocyte antigen 4 (CTLA-4)], in addition to VISTA, was analyzed, along with clinical outcomes. RESULTS High VISTA RNA expression was observed in 32% of tumors (66/514) and was the most common highly expressed checkpoint among the nine assessed. High VISTA expression was independently correlated with high BTLA, TIM-3, and TNFRSF14, and with a diagnosis of pancreatic, small intestine, and stomach cancer. VISTA transcript levels did not correlate with overall survival (OS) from metastatic/advanced disease in the pan-cancer cohort or with immunotherapy outcome (progression-free survival and OS from the start of ICI) in 217 ICI-treated patients. However, in ICI-treated pancreatic cancer patients (n = 16), median OS was significantly shorter (from immunotherapy initiation) for the high- versus not-high-VISTA groups (0.28 versus 1.21 years) (P = 0.047); in contrast, VISTA levels were not correlated with OS in 36 pancreatic cancer patients who did not receive ICI. CONCLUSION High VISTA expression correlates with high BTLA, TIM-3, and TNFRSF14 checkpoint-related molecules and with poorer post-immunotherapy survival in pancreatic cancer, consistent with prior literature indicating that VISTA is prominently expressed on CD68+ macrophages in pancreatic cancers and requiring validation in larger prospective studies. Immunomic analysis may be important for individualized precision immunotherapy.
Collapse
Affiliation(s)
- D Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla.
| | - R Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, USA; WIN Consortium, Paris, France
| | - H Miyashita
- Dartmouth Cancer Center, Hematology and Medical Oncology, Lebanon
| | - J J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore
| | - S Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - M Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - R N Eskander
- Center for Personalized Cancer Therapy and Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, Moores Cancer Center, La Jolla
| | - H Patel
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - G P Botta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | | | | | | | | | - J K Sicklick
- Division of Surgical Oncology, Department of Surgery, Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, USA
| | - S Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla.
| |
Collapse
|
22
|
Sun C, He Y, Wang G, Zhang G, Zhang Y, Shen H, Hu L, Sun Y, Jiang B, Wang X, Yuan K, Min W, Wang L, Sun H, Xiao Y, Yang P. Design, Synthesis, and Antitumor Activity Evaluation of Novel VISTA Small Molecule Inhibitors. J Med Chem 2024; 67:3590-3605. [PMID: 38412237 DOI: 10.1021/acs.jmedchem.3c02039] [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: 02/29/2024]
Abstract
VISTA (V-domain Ig suppressor of T cell activation) is a novel immune checkpoint protein and represents a promising target for cancer immunotherapy. Here, we report the design, synthesis, and evaluation of a series of methoxy-pyrimidine-based VISTA small molecule inhibitors with potent antitumor activity. By employing molecular docking and microscale thermophoresis (MST) assay, we identified a lead compound A1 that binds to VISTA protein with high affinity and optimized its structure. A4 was then obtained, which exhibited the strongest binding ability to VISTA protein, with a KD value of 0.49 ± 0.20 μM. In vitro, A4 significantly activated peripheral blood mononuclear cells (PBMCs) induced the release of cytokines such as IFN-γ and enhanced the cytotoxicity of PBMCs against tumor cells. In vivo, A4 displayed potent antitumor activity and synergized with PD-L1 antibody to enhance the therapeutic effect against cancer. These results suggest that compound A4 is an effective VISTA small molecule inhibitor, providing a basis for the future development of VISTA-targeted drugs.
Collapse
Affiliation(s)
- Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yuling He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Gefei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Guoyu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hao Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yanze Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Binjian Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Liping Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Haopeng Sun
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
23
|
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
|
24
|
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
|
25
|
Vesely MD, Kidacki M, Gaule P, Gupta S, Chan NNN, Han X, Yeung JT, Chen L. Immune Inhibitory Molecule PD-1 Homolog (VISTA) Colocalizes with CD11b Myeloid Cells in Melanoma and Is Associated with Poor Outcomes. J Invest Dermatol 2024; 144:106-115.e4. [PMID: 37562584 PMCID: PMC11966651 DOI: 10.1016/j.jid.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023]
Abstract
Tumors evade immunity through the overexpression of immune inhibitory molecules in the tumor microenvironment such as PD-L1/B7-H1. An immune inhibitory molecule named PD-1 homolog (also known as V-domain Ig-containing suppressor of T cell activation [VISTA]) functions to control both T cells and myeloid cells. Current clinical trials using anti-VISTA-blocking agents for treatment of cancer are ongoing. We sought to determine the extent of VISTA expression in primary cutaneous melanomas (n = 190), identify the critical cell types expressing VISTA, and correlate its expression with PD-L1 expression using multiplexed quantitative immunofluorescence. Within the tumor subcompartments, VISTA is most highly expressed on CD11b myeloid cells, and PD-L1 is most highly expressed on CD68 myeloid cells in our melanoma cohort. There is little correlation between VISTA and PD-L1 expression intensity, suggesting that individual tumors have distinct immunosuppressive tumor microenvironments. High levels of VISTA expression on CD11b myeloid cells but not PD-L1 expression were associated with greater melanoma recurrence and greater all-cause mortality. Our findings suggest that cell-specific VISTA expression may be a negative prognostic biomarker for melanoma and a future potential therapeutic target.
Collapse
Affiliation(s)
- Matthew D Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.
| | - Michal Kidacki
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Swati Gupta
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nay Nwe Nyein Chan
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xue Han
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jacky T Yeung
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lieping Chen
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Borgeaud M, Sandoval J, Obeid M, Banna G, Michielin O, Addeo A, Friedlaender A. Novel targets for immune-checkpoint inhibition in cancer. Cancer Treat Rev 2023; 120:102614. [PMID: 37603905 DOI: 10.1016/j.ctrv.2023.102614] [Citation(s) in RCA: 45] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/06/2023] [Accepted: 08/09/2023] [Indexed: 08/23/2023]
Abstract
Immune-checkpoint inhibitors have revolutionized cancer therapy, yet many patients either do not derive any benefit from treatment or develop a resistance to checkpoint inhibitors. Intrinsic resistance can result from neoantigen depletion, defective antigen presentation, PD-L1 downregulation, immune-checkpoint ligand upregulation, immunosuppression, and tumor cell phenotypic changes. On the other hand, extrinsic resistance involves acquired upregulation of inhibitory immune-checkpoints, leading to T-cell exhaustion. Current data suggest that PD-1, CTLA-4, and LAG-3 upregulation limits the efficacy of single-agent immune-checkpoint inhibitors. Ongoing clinical trials are investigating novel immune-checkpoint targets to avoid or overcome resistance. This review provides an in-depth analysis of the evolving landscape of potentially targetable immune-checkpoints in cancer. We highlight their biology, emphasizing the current understanding of resistance mechanisms and focusing on promising strategies that are under investigation. We also summarize current results and ongoing clinical trials in this crucial field that could once again revolutionize outcomes for cancer patients.
Collapse
Affiliation(s)
| | | | - Michel Obeid
- Centre Hospitalier Universitaire Vaudois, Switzerland
| | - Giuseppe Banna
- Portsmouth Hospitals University NHS Trust, Portsmouth, UK
| | | | | | - Alex Friedlaender
- Geneva University Hospitals, Switzerland; Clinique Générale Beaulieu, Geneva, Switzerland.
| |
Collapse
|
29
|
Rocco D, Della Gravara L, Ragone A, Sapio L, Naviglio S, Gridelli C. Prognostic Factors in Advanced Non-Small Cell Lung Cancer Patients Treated with Immunotherapy. Cancers (Basel) 2023; 15:4684. [PMID: 37835378 PMCID: PMC10571734 DOI: 10.3390/cancers15194684] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/13/2023] [Accepted: 09/19/2023] [Indexed: 10/15/2023] Open
Abstract
Taking into account the huge epidemiologic impact of lung cancer (in 2020, lung cancer accounted for 2,206,771 of the cases and for 1,796,144 of the cancer-related deaths, representing the second most common cancer in female patients, the most common cancer in male patients, and the second most common cancer in male and female patients) and the current lack of recommendations in terms of prognostic factors for patients selection and management, this article aims to provide an overview of the current landscape in terms of currently available immunotherapy treatments and the most promising assessed prognostic biomarkers, highlighting the current state-of-the-art and hinting at future challenges.
Collapse
Affiliation(s)
- Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, 80131 Naples, Italy;
| | - Luigi Della Gravara
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.D.G.); (L.S.); (S.N.)
| | - Angela Ragone
- Max-Planck Institute of Molecular Physiology, 44227 Dortmund, Germany;
| | - Luigi Sapio
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.D.G.); (L.S.); (S.N.)
| | - Silvio Naviglio
- Department of Precision Medicine, Università degli Studi della Campania “Luigi Vanvitelli”, 80138 Naples, Italy; (L.D.G.); (L.S.); (S.N.)
| | - Cesare Gridelli
- Division of Medical Oncology, “S.G. Moscati” Hospital, Contrada Amoretta, 83100 Avellino, Italy
| |
Collapse
|
30
|
Noelle RJ, Lines JL, Lewis LD, Martell RE, Guillaudeux T, Lee SW, Mahoney KM, Vesely MD, Boyd-Kirkup J, Nambiar DK, Scott AM. Clinical and research updates on the VISTA immune checkpoint: immuno-oncology themes and highlights. Front Oncol 2023; 13:1225081. [PMID: 37795437 PMCID: PMC10547146 DOI: 10.3389/fonc.2023.1225081] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 08/21/2023] [Indexed: 10/06/2023] Open
Abstract
Immune checkpoints limit the activation of the immune system and serve an important homeostatic function but can also restrict immune responses against tumors. Inhibition of specific immune checkpoint proteins such as the B7:CD28 family members programmed cell death protein-1 (PD-1) and cytotoxic T-lymphocyte antigen-4 (CTLA-4) has transformed the treatment of various cancers by promoting the anti-tumor activation of immune cells. In contrast to these effects, the V-domain immunoglobulin suppressor of T-cell activation (VISTA) regulates the steady state of the resting immune system and promotes homeostasis by mechanisms distinct from PD-1 and CTLA-4. The effects of VISTA blockade have been shown to include a decrease in myeloid suppression coupled with proinflammatory changes by mechanisms that are separate and distinct from other immune checkpoint proteins; in some preclinical studies these immune effects appear synergistic. Given the potential benefits of VISTA blockade in the context of cancer therapy, the second Annual VISTA Symposium was convened virtually on September 23, 2022, to review new research from investigators and immuno-oncology experts. Discussions in the meeting extended the knowledge of VISTA biology and the effects of VISTA inhibition, particularly on cells of the myeloid lineage and resting T cells, as three candidate anti-VISTA antibodies are in, or nearing, clinical development.
Collapse
Affiliation(s)
- Randolph J. Noelle
- ImmuNext Inc., Lebanon, NH, United States
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - J. Louise Lines
- Department of Microbiology and Immunology, Dartmouth Cancer Center, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | - Lionel D. Lewis
- Section of Clinical Pharmacology, Department of Medicine, Geisel School of Medicine at Dartmouth and Dartmouth Cancer Center, Hanover, NH, United States
| | - Robert E. Martell
- Curis, Inc., Lexington, MA, United States
- Division of Hematology/Oncology, Tufts Medical Center, Boston, MA, United States
| | | | - Sam W. Lee
- Yale University School of Medicine, New Haven, CT, United States
| | - Kathleen M. Mahoney
- Department of Medical, Division of Medical Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, United States
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, United States
| | - Matthew D. Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, CT, United States
| | | | - Dhanya K. Nambiar
- Department of Radiation Oncology, Stanford School of Medicine, Stanford, CA, United States
| | - Andrew M. Scott
- Olivia Newton-John Cancer Research Institute and School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
- Department of Molecular Imaging and Therapy, Austin Health and Faculty of Medicine, University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
31
|
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
|
32
|
Mo S, Zou L, Hu Y, Chang X, Chen J. Expression of PD-L1 and VISTA in Intraductal Papillary Mucinous Neoplasm With Associated Invasive Carcinoma of the Pancreas. Mod Pathol 2023; 36:100223. [PMID: 37244388 DOI: 10.1016/j.modpat.2023.100223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 05/02/2023] [Accepted: 05/18/2023] [Indexed: 05/29/2023]
Abstract
Early detection and treatment of invasive carcinoma arising in association with intraductal papillary mucinous neoplasm (IPMN), which is biologically and (epi)genetically distinct from conventional pancreatic ductal adenocarcinoma, provide an opportunity to improve the prognosis of this lethal disease. Despite the successful application of programmed death (ligand) 1 (PD-[L]1)-blocking strategies in numerous cancers, the immune microenvironment of IPMN with associated invasive carcinoma remains elusive. Here, we investigated CD8+ T cells, CD68+ macrophages, PD-L1, and V-domain immunoglobulin suppressor of T-cell activation (VISTA) in 60 patients with IPMN with associated invasive carcinoma using immunohistochemistry, explored their correlations with clinicopathologic variables and prognosis, and compared them with those in 76 patients with IPMN without invasive carcinoma (60 low-grade and 16 high-grade lesions). Using antibodies against CD8, CD68, and VISTA, we evaluated tumor-infiltrating immune cells in 5 high-power fields (×400) and calculated the corresponding mean counts. PD-L1 with a combined positive score of ≥1 was regarded as positive, and VISTA expression on tumor cells (TCs) was deemed positive when ≥1% of TCs showed membranous/cytoplasmic staining. A reduction of CD8+ T cells and an increase of macrophages were observed during carcinogenesis. Positive PD-L1 combined positive score and VISTA expression on TCs were 13% and 11% in the intraductal component of IPMN with associated invasive carcinoma, 15% and 12% in the associated invasive carcinoma, and 6% and 4% in IPMN without an invasive carcinoma, respectively. Interestingly, the PD-L1 positivity rate was the highest in a subset of associated invasive carcinomas (predominantly gastric-type-derived) and was associated with higher counts of CD8+ T cells, macrophages, and VISTA+ immune cells. Accumulation of VISTA+ immune cells was observed in the intraductal component of IPMN with associated invasive carcinoma compared with that of low-grade IPMN, whereas in intestinal-type IPMN with associated invasive carcinoma, the number of these cells decreased during the transition from the intraductal component to the associated invasive carcinoma. Survival analysis revealed that a higher number of macrophages predicted poorer prognosis. In conclusion, our results might help in individualized immunotherapeutic strategies for these patients.
Collapse
Affiliation(s)
- Shengwei Mo
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Long Zou
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ya Hu
- Department of General Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoyan Chang
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| | - Jie Chen
- Department of Pathology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
| |
Collapse
|
33
|
Liu X, Zhao Z, Dai W, Liao K, Sun Q, Chen D, Pan X, Feng L, Ding Y, Wei S. The Development of Immunotherapy for the Treatment of Recurrent Glioblastoma. Cancers (Basel) 2023; 15:4308. [PMID: 37686584 PMCID: PMC10486426 DOI: 10.3390/cancers15174308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Revised: 08/04/2023] [Accepted: 08/04/2023] [Indexed: 09/10/2023] Open
Abstract
Recurrent glioblastoma (rGBM) is a highly aggressive form of brain cancer that poses a significant challenge for treatment in neuro-oncology, and the survival status of patients after relapse usually means rapid deterioration, thus becoming the leading cause of death among patients. In recent years, immunotherapy has emerged as a promising strategy for the treatment of recurrent glioblastoma by stimulating the body's immune system to recognize and attack cancer cells, which could be used in combination with other treatments such as surgery, radiation, and chemotherapy to improve outcomes for patients with recurrent glioblastoma. This therapy combines several key methods such as the use of monoclonal antibodies, chimeric antigen receptor T cell (CAR-T) therapy, checkpoint inhibitors, oncolytic viral therapy cancer vaccines, and combination strategies. In this review, we mainly document the latest immunotherapies for the treatment of glioblastoma and especially focus on rGBM.
Collapse
Affiliation(s)
- Xudong Liu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Zihui Zhao
- School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Wufei Dai
- Department of Plastic and Reconstructive Surgery, Shanghai Key Laboratory of Tissue Engineering Research, Shanghai Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China;
| | - Kuo Liao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, China;
| | - Qi Sun
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Dongjiang Chen
- Division of Neuro-Oncology, USC Keck Brain Tumor Center, University of Southern California Keck School of Medicine, Los Angeles, CA 90089, USA;
| | - Xingxin Pan
- Department of Oncology, Livestrong Cancer Institutes, Dell Medical School, The University of Texas at Austin, Austin, TX 78712, USA;
| | - Lishuang Feng
- School of Public Health, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong, China; (Q.S.); (L.F.)
| | - Ying Ding
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China; (X.L.); (Y.D.)
| | - Shiyou Wei
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
34
|
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
|
35
|
Borella R, Paolini A, Aramini B, Gibellini L, Masciale V, Lo Tartaro D, Dominici M, De Biasi S, Cossarizza A. Multiparametric analysis of tumor infiltrating lymphocytes in solid tumors. Methods Cell Biol 2023; 195:39-70. [PMID: 40180454 DOI: 10.1016/bs.mcb.2023.03.006] [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] [Indexed: 04/05/2025]
Abstract
The use of single-cell technologies in characterizing the interactions between immune and cancer cells is in continuous expansion. Indeed, the combination of different single-cell approaches enables the definition of novel phenotypic and functional aspects of the immune cells infiltrating the tumor microenvironment (TME). This approach is promoting the discovery of relevant and reliable predictive biomarkers, along with the development of new promising treatments. In this chapter, we describe the main subsets of tumor-infiltrating lymphocytes from a phenotypical and functional point of view and discuss the use of single-cell technologies used to characterize these cell populations within TME.
Collapse
Affiliation(s)
- Rebecca Borella
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Annamaria Paolini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Beatrice Aramini
- Division of Thoracic Surgery, Department of Medical and Surgical Sciences-DIMEC of the Alma Mater Studiorum-University of Bologna and G.B. Morgagni-L. Pierantoni Hospital, Forlì, Italy
| | - Lara Gibellini
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Valentina Masciale
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Domenico Lo Tartaro
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Massimo Dominici
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy
| | - Sara De Biasi
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy.
| | - Andrea Cossarizza
- Department of Medical and Surgical Sciences for Children & Adults, University of Modena and Reggio Emilia, Modena, Italy; Istituto Nazionale per le Ricerche Cardiovascolari, Bologna, Italy
| |
Collapse
|
36
|
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
|
37
|
Ibrahim R, Saleh K, Chahine C, Khoury R, Khalife N, Cesne AL. LAG-3 Inhibitors: Novel Immune Checkpoint Inhibitors Changing the Landscape of Immunotherapy. Biomedicines 2023; 11:1878. [PMID: 37509517 PMCID: PMC10377063 DOI: 10.3390/biomedicines11071878] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 06/25/2023] [Accepted: 06/28/2023] [Indexed: 07/30/2023] Open
Abstract
One of the most important steps forward in the management of cancer was the discovery of immunotherapy. It has become an essential pillar in the treatment paradigm of cancer patients. Unfortunately, despite the various options presented with immune checkpoint inhibitors (ICIs), the benefit is still limited to select patients and the vast majority of these patients gain either minimal benefit or eventually progress, leaving an unmet need for the development of novel therapeutic agents and strategies. Lymphocyte activation gene-3 (LAG-3), an immune checkpoint receptor protein, is a molecule found on the surface of activated T-cells. It plays a major role in negatively regulating T-cell function thereby providing tumors with an immune escape in the tumor microenvironment (TME). Given its importance in regulating the immune system, LAG-3 has been considered as a promising target in oncology and precision medicine. To date, two LAG-3-directed agents (eftilagimod alpha and relatlimab) have been approved in combination with programmed death-1 (PD-1) inhibitors in the setting of advanced solid tumors. In this review, we discuss the structure of LAG-3, its mechanism of action, and its interaction with its ligands. We also shed light on the emerging treatments targeting LAG-3 for the treatment of solid tumors.
Collapse
Affiliation(s)
- Rebecca Ibrahim
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Khalil Saleh
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Claude Chahine
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Rita Khoury
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Nadine Khalife
- Department of head and neck Oncology, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| | - Axel Le Cesne
- International Department, Gustave Roussy Cancer Campus, 94800 Villejuif, France
| |
Collapse
|
38
|
Bewersdorf JP, Xie Z, Bejar R, Borate U, Boultwood J, Brunner AM, Buckstein R, Carraway HE, Churpek JE, Daver NG, Porta MGD, DeZern AE, Fenaux P, Figueroa ME, Gore SD, Griffiths EA, Halene S, Hasserjian RP, Hourigan CS, Kim TK, Komrokji R, Kuchroo VK, List AF, Loghavi S, Majeti R, Odenike O, Patnaik MM, Platzbecker U, Roboz GJ, Sallman DA, Santini V, Sanz G, Sekeres MA, Stahl M, Starczynowski DT, Steensma DP, Taylor J, Abdel-Wahab O, Xu ML, Savona MR, Wei AH, Zeidan AM. Current landscape of translational and clinical research in myelodysplastic syndromes/neoplasms (MDS): Proceedings from the 1 st International Workshop on MDS (iwMDS) Of the International Consortium for MDS (icMDS). Blood Rev 2023; 60:101072. [PMID: 36934059 DOI: 10.1016/j.blre.2023.101072] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Revised: 03/07/2023] [Accepted: 03/09/2023] [Indexed: 03/18/2023]
Abstract
Biological events that contribute to the pathogenesis of myelodysplastic syndromes/neoplasms (MDS) are becoming increasingly characterized and are being translated into rationally designed therapeutic strategies. Herein, we provide updates from the first International Workshop on MDS (iwMDS) of the International Consortium for MDS (icMDS) detailing recent advances in understanding the genetic landscape of MDS, including germline predisposition, epigenetic and immune dysregulation, the complexities of clonal hematopoiesis progression to MDS, as well as novel animal models of the disease. Connected to this progress is the development of novel therapies targeting specific molecular alterations, the innate immune system, and immune checkpoint inhibitors. While some of these agents have entered clinical trials (e.g., splicing modulators, IRAK1/4 inhibitors, anti-CD47 and anti-TIM3 antibodies, and cellular therapies), none have been approved for MDS. Additional preclinical and clinical work is needed to develop a truly individualized approach to the care of MDS patients.
Collapse
Affiliation(s)
- Jan Philipp Bewersdorf
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Zhuoer Xie
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Rafael Bejar
- Division of Hematology and Oncology, Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | - Uma Borate
- Ohio State University Comprehensive Cancer/ James Cancer Hospital, Ohio State University, Columbus, OH, USA
| | - Jacqueline Boultwood
- Blood Cancer UK Molecular Haematology Unit, Nuffield Division of Clinical Laboratory Sciences, Radcliffe Department of Medicine, University of Oxford, Oxford, UK
| | - Andrew M Brunner
- Leukemia Program, Massachusetts General Hospital Cancer Center, Harvard Medical School, Boston, MA, USA
| | - Rena Buckstein
- Department of Medical Oncology/Hematology, Sunnybrook Health Sciences Centre, Toronto, ON, Canada
| | - Hetty E Carraway
- Leukemia Program, Hematology and Medical Oncology, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jane E Churpek
- Department of Hematology, Oncology, and Palliative Care, Carbone Cancer Center, The University of Wisconsin-Madison, Madison, WI, USA
| | - Naval G Daver
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Matteo Giovanni Della Porta
- IRCCS Humanitas Clinical and Research Center & Humanitas University, Department of Biomedical Sciences, via Manzoni 56, 20089 Rozzano - Milan, Italy
| | - Amy E DeZern
- Division of Oncology, Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins Hospital, Baltimore, MD, USA
| | - Pierre Fenaux
- Hôpital Saint Louis, Assistance Publique Hôpitaux de Paris and Paris Cité University, Paris, France
| | - Maria E Figueroa
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Steven D Gore
- National Cancer Institute, Cancer Therapy Evaluation Program, Bethesda, MD, USA
| | | | - Stephanie Halene
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | | | - Christopher S Hourigan
- Laboratory of Myeloid Malignancies, Hematology Branch, National Heart, Lung, and Blood Institute, and Myeloid Malignancies Program, National Institutes of Health, Bethesda, MD, USA
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rami Komrokji
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Vijay K Kuchroo
- Evergrande Center for Immunologic Diseases, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Alan F List
- Precision BioSciences, Inc., Durham, NC, USA
| | - Sanam Loghavi
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Ravindra Majeti
- Division of Hematology, Department of Medicine, Cancer Institute, and Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Olatoyosi Odenike
- Leukemia Program, University of Chicago Medicine and University of Chicago Comprehensive Cancer Center, Chicago, IL, USA
| | - Mrinal M Patnaik
- Division of Hematology, Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | | | - Gail J Roboz
- Weill Cornell Medical College, New York, NY, USA
| | - David A Sallman
- Department of Malignant Hematology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | | | - Guillermo Sanz
- Health Research Institute La Fe, Valencia, Spain; Hospital Universitario y Politécnico La Fe, Valencia, Spain; CIBERONC, IS Carlos III, Madrid, Spain
| | - Mikkael A Sekeres
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Maximilian Stahl
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Daniel T Starczynowski
- Division of Experimental Hematology and Cancer Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | | | - Justin Taylor
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL, USA
| | - Omar Abdel-Wahab
- Department of Medicine, Leukemia Service, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mina L Xu
- Departments of Pathology & Laboratory Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA
| | - Michael R Savona
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Andrew H Wei
- Department of Haematology, Peter MacCallum Cancer Centre, Royal Melbourne Hospital, Walter and Eliza Hall Institute of Medical Research and University of Melbourne, Victoria, Australia
| | - Amer M Zeidan
- Section of Hematology, Department of Internal Medicine, Yale University School of Medicine and Yale Cancer Center, New Haven, CT, USA.
| |
Collapse
|
39
|
Andhari MD, Antoranz A, De Smet F, Bosisio FM. Recent advancements in tumour microenvironment landscaping for target selection and response prediction in immune checkpoint therapies achieved through spatial protein multiplexing analysis. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 382:207-237. [PMID: 38225104 DOI: 10.1016/bs.ircmb.2023.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2024]
Abstract
Immune checkpoint therapies have significantly advanced cancer treatment. Nevertheless, the high costs and potential adverse effects associated with these therapies highlight the need for better predictive biomarkers to identify patients who are most likely to benefit from treatment. Unfortunately, the existing biomarkers are insufficient to identify such patients. New high-dimensional spatial technologies have emerged as a valuable tool for discovering novel biomarkers by analysing multiple protein markers at a single-cell resolution in tissue samples. These technologies provide a more comprehensive map of tissue composition, cell functionality, and interactions between different cell types in the tumour microenvironment. In this review, we provide an overview of how spatial protein-based multiplexing technologies have fuelled biomarker discovery and advanced the field of immunotherapy. In particular, we will focus on how these technologies contributed to (i) characterise the tumour microenvironment, (ii) understand the role of tumour heterogeneity, (iii) study the interplay of the immune microenvironment and tumour progression, (iv) discover biomarkers for immune checkpoint therapies (v) suggest novel therapeutic strategies.
Collapse
Affiliation(s)
- Madhavi Dipak Andhari
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium; The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Asier Antoranz
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium; The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Frederik De Smet
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium; The Laboratory for Precision Cancer Medicine, Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Francesca Maria Bosisio
- Translational Cell and Tissue Research Unit, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium.
| |
Collapse
|
40
|
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
|
41
|
Monjaras-Avila CU, Lorenzo-Leal AC, Luque-Badillo AC, D'Costa N, Chavez-Muñoz C, Bach H. The Tumor Immune Microenvironment in Clear Cell Renal Cell Carcinoma. Int J Mol Sci 2023; 24:ijms24097946. [PMID: 37175653 PMCID: PMC10178526 DOI: 10.3390/ijms24097946] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 04/15/2023] [Accepted: 04/25/2023] [Indexed: 05/15/2023] Open
Abstract
Clear cell renal cell carcinoma (ccRCC) is a type of kidney cancer that arises from the cells lining the tubes of the kidney. The tumor immune microenvironment (TIME) of ccRCC is a complex interplay of various immune cells, cytokines, and signaling pathways. One of the critical features of the ccRCC TIME is the presence of infiltrating immune cells, including T cells, B cells, natural killer cells, dendritic cells, and myeloid-derived suppressor cells. Among these cells, CD8+ T cells are particularly important in controlling tumor growth by recognizing and killing cancer cells. However, the TIME of ccRCC is also characterized by an immunosuppressive environment that hinders the function of immune cells. Several mechanisms contribute to the immunosuppressive nature of the ccRCC TIME. For instance, ccRCC cells produce cytokines such as interleukin-10 (IL-10) and transforming growth factor-beta (TGF-β), which suppress immune cell activation and promote the differentiation of regulatory T cells (Tregs). Tregs, in turn, dampen the activity of effector T cells and promote tumor growth. In addition, ccRCC cells can express programmed death-ligand 1 (PD-L1), which interacts with the programmed cell death protein 1 (PD-1) receptor on T cells to inhibit their function. In addition, other immune checkpoint proteins, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and lymphocyte activation gene 3 (LAG-3), also contribute to the immunosuppressive milieu of the ccRCC TIME. Finally, the hypoxic and nutrient-poor microenvironment of ccRCC can stimulate the production of immunosuppressive metabolites, such as adenosine and kynurenine, which further impair the function of immune cells. Understanding the complex interplay between tumor cells and the immune system in the ccRCC TIME is crucial for developing effective immunotherapies to treat this disease.
Collapse
Affiliation(s)
- Cesar U Monjaras-Avila
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2635 Laurel Street, Vancouver, BC V6H 3Z6, Canada
| | - Ana C Lorenzo-Leal
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| | - Ana C Luque-Badillo
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2635 Laurel Street, Vancouver, BC V6H 3Z6, Canada
| | - Ninadh D'Costa
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2635 Laurel Street, Vancouver, BC V6H 3Z6, Canada
| | - Claudia Chavez-Muñoz
- Vancouver Prostate Centre, Department of Urologic Sciences, University of British Columbia, 2635 Laurel Street, Vancouver, BC V6H 3Z6, Canada
| | - Horacio Bach
- Division of Infectious Diseases, Department of Medicine, University of British Columbia, 2660 Oak Street, Vancouver, BC V6H 3Z6, Canada
| |
Collapse
|
42
|
Digomann D, Strack J, Heiduk M, Plesca I, Rupp L, Reiche C, Nicolaus S, Beer C, Sommer U, Schmitz M, Distler M, Weitz J, Seifert AM, Seifert L. VISTA Ligation Reduces Antitumor T-Cell Activity in Pancreatic Cancer. Cancers (Basel) 2023; 15:cancers15082326. [PMID: 37190254 DOI: 10.3390/cancers15082326] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2023] [Revised: 04/08/2023] [Accepted: 04/11/2023] [Indexed: 05/17/2023] Open
Abstract
Immunotherapy has shown promising results in multiple solid tumors and hematological malignancies. However, pancreatic ductal adenocarcinoma (PDAC) has been largely refractory to current clinical immunotherapies. The V-domain Ig suppressor of T-cell activation (VISTA) inhibits T-cell effector function and maintains peripheral tolerance. Here, we determine VISTA expression in nontumorous pancreatic (n = 5) and PDAC tissue using immunohistochemistry (n = 76) and multiplex immunofluorescence staining (n = 67). Additionally, VISTA expression on tumor-infiltrating immune cells and matched blood samples (n = 13) was measured with multicolor flow cytometry. Further, the effect of recombinant VISTA on T-cell activation was investigated in vitro, and VISTA blockade was tested in an orthotopic PDAC mouse model in vivo. PDAC showed significantly higher VISTA expression compared to that of a nontumorous pancreas. Patients with a high density of VISTA-expressing tumor cells had reduced overall survival. The VISTA expression of CD4+ and CD8+ T cells was increased after stimulation and particularly after a coculture with tumor cells. We detected a higher level of proinflammatory cytokine (TNFα and IFNγ) expression by CD4+ and CD8+ T cells, which was reversed with the addition of recombinant VISTA. A VISTA blockade reduced tumor weights in vivo. The VISTA expression of tumor cells has clinical relevance, and its blockade may be a promising immunotherapeutic strategy for PDAC.
Collapse
Affiliation(s)
- David Digomann
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Johannes Strack
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Max Heiduk
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
| | - Ioana Plesca
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Luise Rupp
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Charlotte Reiche
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Simone Nicolaus
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Carolin Beer
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Ulrich Sommer
- Institute of Pathology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
| | - Marc Schmitz
- National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
- Institute of Immunology, Faculty of Medicine Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Dresden, 69120 Heidelberg, Germany
| | - Marius Distler
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Dresden, 69120 Heidelberg, Germany
| | - Jürgen Weitz
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Dresden, 69120 Heidelberg, Germany
| | - Adrian M Seifert
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Dresden, 69120 Heidelberg, Germany
| | - Lena Seifert
- Department of Visceral, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- National Center for Tumor Diseases (NCT/UCC), 01307 Dresden, Germany
- German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
- Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, 01307 Dresden, Germany
- Helmholtz-Zentrum Dresden-Rossendorf (HZDR), 01328 Dresden, Germany
- German Cancer Research Center (DKFZ), German Cancer Consortium (DKTK), Partner Site Dresden, 69120 Heidelberg, Germany
- Else Kröner Clinician Scientist Professor for "Translational Tumor Immunological Research", 01307 Dresden, Germany
| |
Collapse
|
43
|
Najjary S, Kros JM, de Koning W, Vadgama D, Lila K, Wolf J, Mustafa DAM. Tumor lineage-specific immune response in brain metastatic disease: opportunities for targeted immunotherapy regimen? Acta Neuropathol Commun 2023; 11:64. [PMID: 37061716 PMCID: PMC10105417 DOI: 10.1186/s40478-023-01542-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Accepted: 03/05/2023] [Indexed: 04/17/2023] Open
Abstract
Metastases in the brain are the most severe and devastating complication of cancer. The incidence of brain metastasis is increasing. Therefore, the need of finding specific druggable targets for brain metastasis is demanding. The aim of this study was to compare the brain (immune) response to brain metastases of the most common tumor lineages, viz., lung adenocarcinoma and breast cancer. Targeted gene expression profiles of 11 brain metastasis of lung adenocarcinoma (BM-LUAD) were compared to 11 brain metastasis of breast cancer (BCBM) using NanoString nCounter PanCancer IO 360™ Panel. The most promising results were validated spatially using the novel GeoMx™ Digital Spatial Profiler (DSP) Technology. Additionally, Immune cell profiles and expression of drug targets were validated by multiplex immunohistochemistry. We found a more active immune response in BM-LUAD as compared to BCBM. In the BM-LUAD, 138 genes were upregulated as compared to BCBM (adj. p ≤ 0.05). Conversely, in BCBM 28 genes were upregulated (adj. p ≤ 0.05). Additionally, genes related to CD45 + cells, T cells, and cytotoxic T cells showed to be expressed higher in BM-LUAD compared to BCBM (adj. p = 0.01, adj. p = 0.023, adj. p = 0.023, respectively). The spatial quantification of the immune cells using the GeoMx DSP technique revealed the significantly higher quantification of CD14 and CD163 in tumor regions of BM-LUAD as compared to BCBM. Importantly, the immune checkpoint VISTA and IDO1 were identified as highly expressed in the BM-LUAD. Multiplex immunohistochemistry confirmed the finding and showed that VISTA is expressed mainly in BM-LUAD tumor cells, CD3 + cells, and to fewer levels in some microglial cells in BM-LUAD. This is the first report on differences in the brain immune response between metastatic tumors of different lineages. We found a far more extensive infiltration of immune cells in BM-LUAD as compared to BCBM. In addition, we found higher expression of VISTA and IDO1 in BM-LUAD. Taken together, targeted immune therapy should be considered to treat patients with BM-LUAD.
Collapse
Affiliation(s)
- Shiva Najjary
- Department of Pathology and Clinical Bioinformatics, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Johan M Kros
- Department of Pathology and Clinical Bioinformatics, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Willem de Koning
- Department of Pathology and Clinical Bioinformatics, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Disha Vadgama
- Department of Pathology and Clinical Bioinformatics, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands
| | - Karishma Lila
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Janina Wolf
- Department of Pathology and Clinical Bioinformatics, Erasmus University Medical Center, Rotterdam, The Netherlands
- Institute of Tissue Medicine and Pathology, University of Bern, Murtenstrasse 31, 3008, Bern, Switzerland
| | - Dana A M Mustafa
- Department of Pathology and Clinical Bioinformatics, The Tumor Immuno-Pathology Laboratory, Erasmus University Medical Center, Dr. Molewaterplein 40, 3015 GD, Rotterdam, The Netherlands.
| |
Collapse
|
44
|
Dussart C, Decaux-Tramoni B, Quesada S, Thomas QD, Benzerouale O, Nicolas E, Fiteni F. [Combination strategies for checkpoint inhibition: Current practices and perspectives]. Bull Cancer 2023:S0007-4551(23)00166-2. [PMID: 37055309 DOI: 10.1016/j.bulcan.2023.03.021] [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: 01/24/2023] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 04/15/2023]
Abstract
T-cell checkpoint blockade therapies have revolutionized treatment protocols and prognosis in patients with cancer. Pointed out by the success of PD-1 (programmed cell death-1) plus CTLA-4 (cytotoxic-T-lymphocyte associated antigen 4) blockade in patients with melanoma, the perspective of new synergistic immunotherapy combinations seems to be an important opportunity to improve outcomes for patients. In this article, we first focus on immunotherapy combinations that have shown their efficiency and that are currently approved in solid tumors. Then, we present a summary of emerging targets with reported pre-clinical efficacy and currently evaluated through ongoing clinical trials and other immunomodulatory molecules in the tumor microenvironment.
Collapse
Affiliation(s)
- Chloé Dussart
- CHU de Nîmes, service d'oncologie médicale, 4, rue du Professeur-Robert-Debré, 30900 Nîmes, France
| | - Baptiste Decaux-Tramoni
- CHU de Nîmes, service d'oncologie médicale, 4, rue du Professeur-Robert-Debré, 30900 Nîmes, France
| | - Stanislas Quesada
- Institut régional du cancer de Montpellier, département d'oncologie médicale, 34298 Montpellier cedex 5, France
| | - Quentin Dominique Thomas
- Institut régional du cancer de Montpellier, département d'oncologie médicale, 34298 Montpellier cedex 5, France
| | - Ouail Benzerouale
- CHU de Nîmes, service d'oncologie médicale, 4, rue du Professeur-Robert-Debré, 30900 Nîmes, France
| | - Emanuel Nicolas
- CHU de Nîmes, service d'oncologie médicale, 4, rue du Professeur-Robert-Debré, 30900 Nîmes, France; Université de Montpellier, Institut Desbrest d'épidémiologie et de santé publique, Inserm UMR 1302, 34090 Montpellier, France
| | - Frédéric Fiteni
- CHU de Nîmes, service d'oncologie médicale, 4, rue du Professeur-Robert-Debré, 30900 Nîmes, France; Université de Montpellier, Institut Desbrest d'épidémiologie et de santé publique, Inserm UMR 1302, 34090 Montpellier, France.
| |
Collapse
|
45
|
Rocco D, Della Gravara L, Battiloro C, Palazzolo G, Gridelli C. Recently approved and emerging monoclonal antibody immune checkpoint inhibitors for the treatment of advanced non-small cell lung cancer. Expert Opin Biol Ther 2023; 23:261-268. [PMID: 36803090 DOI: 10.1080/14712598.2023.2183116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2023]
Abstract
INTRODUCTION CTLA-4/PD-1/PD-L1- directed immune checkpoint inhibitors (ICIs) are one of the standard therapies for the treatment of advanced non-small cell lung cancer (NSCLC). However, some new classes of monoclonal antibodies are emerging as promising therapies for advanced NSCLC. AREAS COVERED Therefore, this paper aims to provide a comprehensive review of the recently approved as well as emerging monoclonal antibody immune checkpoint inhibitors for the treatment of advanced NSCLC. EXPERT OPINION Further and larger studies will be needed to explore the promising emerging data on new ICIs. Future phase III trials could allow us to properly assess the role of each immune checkpoints in the wider context of the tumor microenvironment and thus the best new ICIs to use, the best approach and the most effective subset of patients to select.
Collapse
Affiliation(s)
- Danilo Rocco
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Naples, Italy
| | - Luigi Della Gravara
- Department of Precision Medicine, Università degli studi della Campania "Luigi Vanvitelli", Naples, Italy
| | - Ciro Battiloro
- Department of Pulmonary Oncology, AORN dei Colli Monaldi, Naples, Italy
| | | | - Cesare Gridelli
- Division of Medical Oncology, S.G. Moscati Hospital, Avellino, Italy
| |
Collapse
|
46
|
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
|
47
|
VISTA H-Score Is Significantly Associated with a 5-Year DFS Rate in Oral Squamous Cell Carcinoma. J Clin Med 2023; 12:jcm12041619. [PMID: 36836154 PMCID: PMC9967884 DOI: 10.3390/jcm12041619] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 02/12/2023] [Accepted: 02/14/2023] [Indexed: 02/22/2023] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most common type of oral cancer in the world. Despite its prevalence, it is often recognized in advanced stages (III or IV) when it has already spread to local lymph nodes. In this study, we investigate the V-domain Ig suppressor of T cell activation (VISTA) as a potential prognostic factor in OSCC. Tissue samples were collected from 71 oral squamous cell carcinoma patients to determine protein expression levels (using immunochemistry and the semi-quantitative H-score method). Moreover, RT-qPCR was additionally performed in 35 patients. Clinical factors in our cohort study had no impact on VISTA expression. However, VISTA expression is largely correlated with Il-33 levels in tumor cells and lymphocytes and with PD-L1 in tumor cells. The impact of VISTA expression on overall survival (OS) is rather limited, but in the case of a 5-year survival rate, a significant association has been proven. VISTA seems to be a rather weak clinicopathological marker but needs further evaluation in the context of survival. In addition, the potential of VISTA combination with Il-33 or PD-L1 should be further investigated in OSCC.
Collapse
|
48
|
Zhou K, Li S, Zhao Y, Cheng K. Mechanisms of drug resistance to immune checkpoint inhibitors in non-small cell lung cancer. Front Immunol 2023; 14:1127071. [PMID: 36845142 PMCID: PMC9944349 DOI: 10.3389/fimmu.2023.1127071] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 01/30/2023] [Indexed: 02/10/2023] Open
Abstract
Immune checkpoint inhibitors (ICIs) in the form of anti-CTLA-4 and anti-PD-1/PD-L1 have become the frontier of cancer treatment and successfully prolonged the survival of patients with advanced non-small cell lung cancer (NSCLC). But the efficacy varies among different patient population, and many patients succumb to disease progression after an initial response to ICIs. Current research highlights the heterogeneity of resistance mechanisms and the critical role of tumor microenvironment (TME) in ICIs resistance. In this review, we discussed the mechanisms of ICIs resistance in NSCLC, and proposed strategies to overcome resistance.
Collapse
Affiliation(s)
- Kexun Zhou
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
| | - Shuo Li
- Department of Thoracic Surgery, West China Hospital, Sichuan University, Chengdu, China
- Lung Cancer Center, West China Hospital Sichuan University, Chengdu, China
| | - Yi Zhao
- The First Clinical Medical College of Lanzhou University, Lanzhou University, Lanzhou, China
| | - Ke Cheng
- Abdominal Oncology Ward, Division of Medical Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
- Abdominal Oncology Ward, Division of Radiation Oncology, Cancer Center, State Key Laboratory of Biological Therapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
49
|
Sadeghirad H, Bahrami T, Layeghi SM, Yousefi H, Rezaei M, Hosseini-Fard SR, Radfar P, Warkiani ME, O'Byrne K, Kulasinghe A. Immunotherapeutic targets in non-small cell lung cancer. Immunology 2023; 168:256-272. [PMID: 35933597 DOI: 10.1111/imm.13562] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Accepted: 08/02/2022] [Indexed: 01/17/2023] Open
Abstract
Non-small cell lung cancer (NSCLC) is one of the most common types of cancer in the world and has a 5-year survival rate of ~20%. Immunotherapies have shown promising results leading to durable responses, however, they are only effective for a subset of patients. To determine the best therapeutic approach, a thorough and in-depth profiling of the tumour microenvironment (TME) is required. The TME is a complex network of cell types that form an interconnected network, promoting tumour cell initiation, growth and dissemination. The stroma, immune cells and endothelial cells that comprise the TME generate a plethora of cytotoxic or cytoprotective signalling pathways. In this review, we discuss immunotherapeutic targets in NSCLC tumours and how the TME may influence patients' response to immunotherapy.
Collapse
Affiliation(s)
- Habib Sadeghirad
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| | - Tayyeb Bahrami
- Liver and Digestive Research Center, Research Institute for Health Development, Kurdistan University of Medical Sciences, Sanandaj, Iran
| | - Sepideh M Layeghi
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Hassan Yousefi
- Department of Biochemistry and Molecular Biology, LSUHSC School of Medicine, New Orleans, Louisiana, USA
| | - Meysam Rezaei
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Seyed R Hosseini-Fard
- Department of Medical Genetics, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Payar Radfar
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Majid E Warkiani
- School of Biomedical Engineering, University of Technology Sydney, Sydney, New South Wales, Australia
| | - Ken O'Byrne
- Centre for Genomics and Personalised Health, School of Biomedical Sciences, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Arutha Kulasinghe
- University of Queensland Diamantina Institute, Faculty of Medicine, The University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
50
|
Dutta S, Ganguly A, Chatterjee K, Spada S, Mukherjee S. Targets of Immune Escape Mechanisms in Cancer: Basis for Development and Evolution of Cancer Immune Checkpoint Inhibitors. BIOLOGY 2023; 12:biology12020218. [PMID: 36829496 PMCID: PMC9952779 DOI: 10.3390/biology12020218] [Citation(s) in RCA: 52] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/22/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023]
Abstract
Immune checkpoint blockade (ICB) has emerged as a novel therapeutic tool for cancer therapy in the last decade. Unfortunately, a small number of patients benefit from approved immune checkpoint inhibitors (ICIs). Therefore, multiple studies are being conducted to find new ICIs and combination strategies to improve the current ICIs. In this review, we discuss some approved immune checkpoints, such as PD-L1, PD-1, and CTLA-4, and also highlight newer emerging ICIs. For instance, HLA-E, overexpressed by tumor cells, represents an immune-suppressive feature by binding CD94/NKG2A, on NK and T cells. NKG2A blockade recruits CD8+ T cells and activates NK cells to decrease the tumor burden. NKG2D acts as an NK cell activating receptor that can also be a potential ICI. The adenosine A2A and A2B receptors, CD47-SIRPα, TIM-3, LAG-3, TIGIT, and VISTA are targets that also contribute to cancer immunoresistance and have been considered for clinical trials. Their antitumor immunosuppressive functions can be used to develop blocking antibodies. PARPs, mARTs, and B7-H3 are also other potential targets for immunosuppression. Additionally, miRNA, mRNA, and CRISPR-Cas9-mediated immunotherapeutic approaches are being investigated with great interest. Pre-clinical and clinical studies project these targets as potential immunotherapeutic candidates in different cancer types for their robust antitumor modulation.
Collapse
Affiliation(s)
- Shovan Dutta
- The Center for Immunotherapy & Precision Immuno-Oncology (CITI), Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Anirban Ganguly
- Department of Biochemistry, All India Institute of Medical Sciences, Deoghar 814152, India
| | | | - Sheila Spada
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
- Correspondence: (S.S.); (S.M.)
| | - Sumit Mukherjee
- Department of Cardiothoracic and Vascular Surgery, Albert Einstein College of Medicine, Bronx, NY 10461, USA
- Correspondence: (S.S.); (S.M.)
| |
Collapse
|