151
|
Abstract
PURPOSE OF REVIEW Cancer cells evade immune surveillance partly due to the immunosuppressive features of the tumor microenvironment (TME). Currently approved immuno-oncology drugs for the treatment of lung cancer are aimed to inhibit immune checkpoints, such as programmed death protein-1 (PD-1), PD ligand-1 (PD-L1) and cytotoxic T lymphocyte-associated antigen 4. Despite these, researchers are currently racing to create the optimal cancer immunotherapy treatments. RECENT FINDINGS Novel immunotherapeutic drugs mainly act on activated immune cells and exert their therapeutic effects by enhancing antitumor responses. In this article, we review new therapies for the treatment of lung cancer that enhance T cell priming, remove coinhibitory signals, supply costimulatory signals and condition the TME. SUMMARY As more immunotherapeutic targets are in studies, designing multimodal strategies to provide greater efficacy with lower toxicity will be necessary.
Collapse
Affiliation(s)
- Ling Peng
- Department of Respiratory Disease, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang Province
| | - Zibing Wang
- Department of Immunotherapy, Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan Province, China
| | - Justin Stebbing
- Division of Cancer, Department of Surgery and Cancer, Imperial College London, London, UK
| | - Zhentao Yu
- Department of Thoracic Surgery, National Cancer Center, National Clinical Research Center for Cancer, Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
152
|
Chen Y, Feng R, He B, Wang J, Xian N, Huang G, Zhang Q. PD-1H Expression Associated With CD68 Macrophage Marker Confers an Immune-Activated Microenvironment and Favorable Overall Survival in Human Esophageal Squamous Cell Carcinoma. Front Mol Biosci 2021; 8:777370. [PMID: 34950702 PMCID: PMC8688962 DOI: 10.3389/fmolb.2021.777370] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 11/04/2021] [Indexed: 12/19/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is the most common type of esophageal carcinoma (EC) in China. Although the PD-1 inhibitor pembrolizumab has been approved to treat patients with EC, its therapeutic efficacy is limited. Thus, additional immunotherapeutic targets for EC treatment are needed. Programmed Death-1 Homolog (PD-1H) is a negative checkpoint regulator that inhibits antitumor immune responses. Here, PD-1H expression in 114 patients with ESCC was evaluated by immunohistochemistry. Next, 12 randomly selected tumor tissue sections were used to assess the colocalization of PD-1H protein and multiple immune markers by multiplex immunohistochemistry. Our results demonstrated that PD-1H was expressed at high frequency in ESCC tumor tissues (85.1%). PD-1H protein was predominantly expressed in CD68+ tumor-associated macrophages and expressed at low levels in CD4+ T cells and CD8+ T cells in ESCC tumor tissues. Furthermore, based on ESCC data in The Cancer Genome Atlas (TCGA), the gene expression levels of PD-1H were positively associated with the infiltration levels of immune-activated cells especially CD8+ cytotoxic T cells. In contrast, the gene expression levels of PD-1H were negatively correlated with myeloid-derived suppressor cells (MDSCs). Importantly, PD-1H expression in tumor sites was significantly correlated with favorable overall survival in patients with ESCC. Collectively, our findings first provided direct information on the PD-1H expression pattern and distribution in ESCC, and positive correlation of PD-1H expression with overall survival suggested PD-1H expression levels could be a significant prognostic indicator for patients with ESCC. Future studies need to explore the immunoregulatory of PD-1H in the tumor microenvironment of ESCC.
Collapse
Affiliation(s)
- Yuangui Chen
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Rui Feng
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China.,Department of Oncology, Fujian Medical University Union Hospital, Fuzhou, China
| | - Bailin He
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Jun Wang
- Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, China
| | - Na Xian
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Gangxiong Huang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Qiuyu Zhang
- Department of Immunology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China.,Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| |
Collapse
|
153
|
Hu L, Chen L, Xiao Z, Zheng X, Chen Y, Xian N, Cho C, Luo L, Huang G, Chen L. Ablation of T cell-associated PD-1H enhances functionality and promotes adoptive immunotherapy. JCI Insight 2021; 7:148247. [PMID: 34905507 PMCID: PMC8855794 DOI: 10.1172/jci.insight.148247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 12/08/2021] [Indexed: 11/17/2022] Open
Abstract
Programmed death-1 homolog (PD-1H) is a co-inhibitory molecule that negatively regulates T cell-mediated immune responses. In this study, we determined whether ablation of T cell-associated PD-1H could enhance adoptive T cell therapy in experimental tumor models. The expression of PD-1H is up-regulated in activated and tumor-infiltrating CD8+ T cells. Activated CD8+ T cells from PD-1H deficient (KO) mice exhibited increased cell proliferation, cytokine production and anti-tumor activity in vitro. Adoptive transfer of PD-1H-KO CD8+ T cells resulted in the regression of established syngeneic mouse tumors. Similar results were obtained when PD-1H was ablated in T cells by CRISPR/Cas9-mediated gene silencing. Furthermore, ablation of PD-1H in CAR-T cells significantly improved their anti-tumor activity against human xenografts in vivo. Our results indicate that T cell-associated PD-1H could suppress immunity in the tumor microenvironment and targeting PD-1H may improve T cell adoptive immunotherapy.
Collapse
Affiliation(s)
- Li Hu
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Ling Chen
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Zexiu Xiao
- Laboratory of Immunotherapy, Sun Yat-sen University, Guangzhou, China
| | - Xu Zheng
- Laboratory of Immunotherapy, Sun Yat-sen University, Guangzhou, China
| | - Yuangui Chen
- Department of Radiation Oncology, Fujian Medical University, Fuzhou, China
| | - Na Xian
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Christina Cho
- Department of Immunobiology, Yale University, New Haven, United States of America
| | - Liqun Luo
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Gangxiong Huang
- Institute of Immunotherapy, Fujian Medical University, Fuzhou, China
| | - Lieping Chen
- Department of Immunobiology, Yale University, New Haven, United States of America
| |
Collapse
|
154
|
Li L, Shi W, Zhou J. Effect of CMNa combined with radiotherapy on the tumor immune microenvironment of mouse cervical cancer cell transplantation tumor model. Bioengineered 2021; 12:1066-1077. [PMID: 33784955 PMCID: PMC8806344 DOI: 10.1080/21655979.2021.1899532] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2021] [Accepted: 03/02/2021] [Indexed: 01/30/2023] Open
Abstract
In this study, we construct a subcutaneous tumor mice model of U14 cells, observe the tumor growth, and detect the expression of Foxp3 and VISTA in cervical cancer tissues and adjacent tissues during CMNa-enhancing radiotherapy.From the 15th day, compared with the control group, the tumor volume changes in each treatment group were significant (P < 0.01). CMNa combined with radiotherapy had an interactive effect and a positive effect in inhibiting tumor volume growth. There was no significant difference in the expression of Foxp3 and VISTA in mouse cervical cancer tissues and adjacent tissues in each group. The Foxp3 level in the RT group was the highest, and the CMNa group was the lowest. The VISTA level of the CMNa+RT group was the highest, the RT group is followed by, and the Control group is the lowest. The Foxp3 level of the CMNa group did not change much at each different point. The Foxp3 level in RT and CMNa+RT group gradually decreased after a transient increase, and the VISTA level in the CMNa+RT group increased more.Our results show that CMNa can enhance the efficacy of radiotherapy, and at the same time can reduce the compensatory increase in regulatory T cell Foxp3 levels caused by radiotherapy, and reduce the radiotherapy response. However, in the course of the treatment of the two, there may be a substantial increase in the level of VISTA, and the combined application of VISTA inhibitors may increase the anti-tumor response.
Collapse
Affiliation(s)
- Li Li
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Weiqiang Shi
- Department of Pathology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juying Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
155
|
Abstract
The V-domain Ig Suppressor of T-cell Activation (VISTA) is an immune checkpoint regulator that suppresses immune responses and is readily expressed on human and murine myeloid cells and T cells. This immunosuppressive pathway can be activated using VISTA agonists. Here, we report the development of murine anti-human VISTA (anti-hVISTA) monoclonal antibodies (mAbs), anti-hVISTA nanobodies (Nbs), and cross-reactive rat anti-murine/human VISTA (anti-hmVISTA) mAbs. All mAbs and Nbs generated bound to VISTA (human and/or murine) with dissociation constants in the sub-nanomolar or low nanomolar range. Competition analysis revealed that the selected Nbs bound the same or a nearby epitope(s) as the human VISTA-specific mAbs. However, the cross-reactive mAbs only partially competed with Nbs for binding to hVISTA. All mAbs and one Nb (hVISTANb7) were able to strongly detect VISTA expression on primary human monocytes. Importantly, the murine anti-hVISTA mAbs 7E12 and 7G5 displayed strong agonistic activity in human peripheral blood mononuclear cell cultures, while Nb7 and rat anti-hmVISTA mAbs 3C3, 7C6, 7C7, and 7G1 also behaved as hVISTA agonists, albeit to a lesser extent. Cross-reactive mAbs 7C7 and 7G1 further displayed agonistic potential in murine splenocyte assays. Importantly, mAb 7G1 significantly reduced inflammation associated with the murine model of imiquimod-induced psoriasis. These agonistic VISTA mAbs may represent therapeutic leads to treat inflammatory disorders.
Collapse
Affiliation(s)
| | - Amanda Sparkes
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada
| | - Ema Romão
- Cellular and Molecular Immunology Lab, Vrije Universiteit Brussel, Ixelles, Belgium
| | - Shrayasee Saha
- Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada
| | - Jean Gariépy
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada.,Department of Pharmaceutical Sciences, University of Toronto, Toronto, Canada.,Department of Medical Biophysics, University of Toronto, Toronto, Canada
| |
Collapse
|
156
|
Gameiro A, Nascimento C, Correia J, Ferreira F. VISTA Is a Diagnostic Biomarker and Immunotherapy Target of Aggressive Feline Mammary Carcinoma Subtypes. Cancers (Basel) 2021; 13:cancers13215559. [PMID: 34771722 PMCID: PMC8583306 DOI: 10.3390/cancers13215559] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 10/31/2021] [Accepted: 11/04/2021] [Indexed: 01/03/2023] Open
Abstract
Simple Summary Mammary tumors are common in cats, showing aggressive behavior and few therapeutic options. Recently, feline mammary carcinomas have become a reliable cancer model for human breast cancer studies, due to the similarities between the two species. Thus, the identification of new tumor biomarkers and therapeutic targets to improve cat’s prognosis is needed. VISTA is an important immune checkpoint protein that has gained importance over the past few years in women’s cancers. In this study, the serum VISTA levels and tumor expression were analyzed in cats with mammary tumors, being correlated with other immune checkpoints. In the diseased animals, VISTA is overexpressed in more aggressive tumor subtypes (HER2-positive and triple-negative), showing a positive correlation with the expression of VISTA in tumor-infiltrating lymphocytes, and is associated with an immunosuppressive status, suggesting that VISTA could be a promising non-invasive prognostic biomarker and therapeutic target in cats with mammary carcinomas, as reported in humans. Abstract Feline mammary carcinoma (FMC) is a common neoplasia, showing aggressive clinicopathological features, without viable therapeutic options. The study of tumor microenvironment has gained importance, due to the ability to control tumor progression by regulating the immune response. Considering the lack of knowledge, feline serum VISTA levels from cats with mammary carcinoma were compared with healthy controls, and with serum levels of PD-1/PD-L1, CTLA-4, LAG-3, IL-6, and TNF-α. In parallel, VISTA tumor expression was evaluated in FMC samples. The obtained data revealed that serum VISTA levels were significantly higher in cats presenting HER2-positive (p = 0.0025) or triple-negative subtypes (p = 0.0019), with higher serum levels in luminal A (p = 0.0025) correlated to the presence of metastasis (p = 0.0471). Furthermore, in HER2-positive or triple-negative tumors, correlations were obtained between serum VISTA levels and the serum levels of the above-mentioned molecules. In tumors, VISTA expression revealed a stronger intensity in cancer cells, when compared to TILs (p < 0.0001). Stratifying the samples by subtypes, a higher number of VISTA-positive TILs was observed in the HER2-positive subtype, compared with triple-negative tumors (p = 0.0138). In conclusion, results support the development of therapeutic strategies for HER2-positive and triple-negative FMC subtypes, reinforcing the use of cats as a human oncology model.
Collapse
|
157
|
Ghouzlani A, Lakhdar A, Rafii S, Karkouri M, Badou A. The immune checkpoint VISTA exhibits high expression levels in human gliomas and associates with a poor prognosis. Sci Rep 2021; 11:21504. [PMID: 34728682 PMCID: PMC8563991 DOI: 10.1038/s41598-021-00835-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Accepted: 10/06/2021] [Indexed: 12/14/2022] Open
Abstract
In human gliomas, anti-tumor T cell responses are inhibited through induction of local and systemic immunosuppression. Immune checkpoint blockade is proving to be a success in several types of cancers. However, many studies reported that the treatment of glioblastoma patients with anti-CTLA-4 or anti-PD-1 has no survival benefit compared to standard chemotherapy. This study aimed to investigate the expression and role of VISTA, a newly described immune checkpoint regulator, in human gliomas. mRNA expression was assessed in a total of 87 samples from glioma patients. 57 glioma tissues were taken at different grades. 20 peripheral blood mononuclear cells (PBMC) samples were taken before surgery and ten after surgery, all from the same set of patients. As for the control, ten specimens of PBMC were taken from healthy donors. Protein expression using immunohistochemistry was performed for 30 patients. The Cancer Genome Atlas (TCGA) data set, was also used to investigate VISTA expression through analysis of RNA-seq data of 667 glioma patients. In the Moroccan cohort, VISTA gene expression was significantly upregulated in glioma tissues related to PBMC of healthy donors. This high expression was specific to patient tissues since VISTA expression in PBMC was low when assessed either before or after surgery. Besides, VISTA exhibited higher expression levels in grade III/IV relative to grade I/II glioma patients. Interestingly, VISTA correlated positively with PD-1 expression. PD-1 also showed elevated expressions in higher glioma grades. The TCGA cohort corroborated these observations. Indeed, VISTA was also found to be strongly expressed in high grades. It was positively correlated with other critical immune checkpoints. Finally, increased VISTA transcript levels were associated with weak overall survival of glioma patients. Our study highlighted a correlation between high levels of VISTA expression and poor prognosis in glioma patients. VISTA might be involved in glioma progression and could be considered as a possible new therapeutic target, especially in advanced gliomas.
Collapse
Affiliation(s)
- Amina Ghouzlani
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Abdelhakim Lakhdar
- Department of Neurosurgery, UHC Ibn Rochd, Casablanca, Morocco.,Laboratory of Research on Neurologic, Neurosensorial Diseases and Handicap, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Soumaya Rafii
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco
| | - Mehdi Karkouri
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco.,Department of Pathology, CHU Ibn Rochd, Casablanca, Morocco
| | - Abdallah Badou
- Cellular and Molecular Pathology Laboratory, Faculty of Medicine and Pharmacy, Hassan II University, Casablanca, Morocco.
| |
Collapse
|
158
|
Lee SW. VISTA Stimulation of VSIG4-Positive Macrophages Strongly Suppresses T Cell Proliferation via Excessive Nitric Oxide Production in Sepsis. Biol Pharm Bull 2021; 44:1645-1652. [PMID: 34433706 DOI: 10.1248/bpb.b21-00616] [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: 11/22/2022]
Abstract
Organ damage and immune deficiency are important problems in sepsis caused by an excessive immune response. There is controversy about the cause of immune suppression. In this study, we investigated the roles of macrophages that exhibit excessive activity on T cell immunity. Peritoneal macrophages from mice with cecal ligation and puncture (CLP)-induced sepsis migrated to different organs. In particular, V-set immunoglobulin (Ig)-domain-containing 4 (VSIG4) positive macrophages appeared in the spleen 48 h after CLP induction. When cocultured with splenic T cells, VSIG4(+) cells inhibited the proliferation of activated T cells through the release of nitric oxide (NO) compared to VSIG4(-) cells. Stimulation of VSIG4(+) cells with V-domain Ig suppressor of T cell activation (VISTA) antibody increased the expression of several cytokine genes and the release of NO, but not phagocytosis, compared to those of hamster IgG-stimulated VSIG4(+) cells. When cocultured with splenic T cells, VISTA-stimulated VSIG4(+) cells induced excessive T cell suppression via more NO secretion compared to hamster IgG-stimulated VSIG4(+) cells. Taken together, the current study demonstrates that VSIG4(+) peritoneal macrophages play important roles in inducing immunosuppression and that VISTA acts as a costimulatory receptor in these cells. These data suggest that blocking the migration of VSIG4(+) cells might alleviate excessive immune activity and that blocking VISTA on VSIG4(+) macrophages might play a crucial role in the development of new therapies to prevent T cell suppression in sepsis.
Collapse
Affiliation(s)
- Soo-Woong Lee
- Innovative Therapeutics Research Institute, College of Medicine, Inje University
- Department of Convergence Biomedical Science, College of Medicine, Inje University
| |
Collapse
|
159
|
Yang F, Zhang Y, Chen Z, Zhang L. VISTA Blockade Aggravates Bone Loss in Experimental Murine Apical Periodontitis. Front Immunol 2021; 12:738586. [PMID: 34691045 PMCID: PMC8529274 DOI: 10.3389/fimmu.2021.738586] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Accepted: 09/24/2021] [Indexed: 11/19/2022] Open
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a novel coinhibitory immune checkpoint molecule that maintains immune homeostasis. The present study explored the role of VISTA in human and murine inflammatory tissues of apical periodontitis (AP). VISTA was upregulated in inflammatory tissues of human AP. In mice, the expression of VISTA gradually increased with the development of mouse experimental apical periodontitis (MAP), the CD3+ T cells, CD11b+ myeloid cells, and FOXP3+ regulatory T cells also gradually accumulated. Moreover, a blockade of VISTA using a mouse in vivo anti-VISTA antibody aggravated periapical bone loss and enhanced the infiltration of immune cells in an experimental mouse periapical periodontitis model. The collective results suggest that VISTA serves as a negative regulator of the development and bone loss of apical periodontitis.
Collapse
Affiliation(s)
- Fuhua Yang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Yifei Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Zhi Chen
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| | - Lu Zhang
- The State Key Laboratory Breeding Base of Basic Science of Stomatology (Hubei-MOST) and Key Laboratory for Oral Biomedicine of Ministry of Education (KLOBM), School and Hospital of Stomatology, Wuhan University, Wuhan, China
- Department of Endodontics, School and Hospital of Stomatology, Wuhan University, Wuhan, China
| |
Collapse
|
160
|
Li K, Shi H, Zhang B, Ou X, Ma Q, Chen Y, Shu P, Li D, Wang Y. Myeloid-derived suppressor cells as immunosuppressive regulators and therapeutic targets in cancer. Signal Transduct Target Ther 2021; 6:362. [PMID: 34620838 PMCID: PMC8497485 DOI: 10.1038/s41392-021-00670-9] [Citation(s) in RCA: 426] [Impact Index Per Article: 106.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 02/21/2021] [Accepted: 06/01/2021] [Indexed: 02/05/2023] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogenic population of immature myeloid cells with immunosuppressive effects, which undergo massive expansion during tumor progression. These cells not only support immune escape directly but also promote tumor invasion via various non-immunological activities. Besides, this group of cells are proved to impair the efficiency of current antitumor strategies such as chemotherapy, radiotherapy, and immunotherapy. Therefore, MDSCs are considered as potential therapeutic targets for cancer therapy. Treatment strategies targeting MDSCs have shown promising outcomes in both preclinical studies and clinical trials when administrated alone, or in combination with other anticancer therapies. In this review, we shed new light on recent advances in the biological characteristics and immunosuppressive functions of MDSCs. We also hope to propose an overview of current MDSCs-targeting therapies so as to provide new ideas for cancer treatment.
Collapse
Affiliation(s)
- Kai Li
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China
| | - Houhui Shi
- Department of Gynecology and Obstetrics, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second Hospital, Sichuan University, 610041, Chengdu, China
| | - Benxia Zhang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China
| | - Xuejin Ou
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China
| | - Qizhi Ma
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China
| | - Yue Chen
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China
| | - Pei Shu
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China
| | - Dan Li
- Institute of Respiratory Health, Frontiers Science Center for Disease-related Molecular Network, and Precision Medicine Center, Precision Medicine Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, 610041, Chengdu, China.
| | - Yongsheng Wang
- Department of Thoracic Oncology, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center, 610041, Chengdu, China. .,Clinical Trial Center, West China Hospital, Sichuan University, 610041, Chengdu, China.
| |
Collapse
|
161
|
Bolandi N, Derakhshani A, Hemmat N, Baghbanzadeh A, Asadzadeh Z, Afrashteh Nour M, Brunetti O, Bernardini R, Silvestris N, Baradaran B. The Positive and Negative Immunoregulatory Role of B7 Family: Promising Novel Targets in Gastric Cancer Treatment. Int J Mol Sci 2021; 22:ijms221910719. [PMID: 34639059 PMCID: PMC8509619 DOI: 10.3390/ijms221910719] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 09/28/2021] [Accepted: 10/01/2021] [Indexed: 12/30/2022] Open
Abstract
Gastric cancer (GC), with a heterogeneous nature, is the third leading cause of death worldwide. Over the past few decades, stable reductions in the incidence of GC have been observed. However, due to the poor response to common treatments and late diagnosis, this cancer is still considered one of the lethal cancers. Emerging methods such as immunotherapy with immune checkpoint inhibitors (ICIs) have transformed the landscape of treatment for GC patients. There are presently eleven known members of the B7 family as immune checkpoint molecules: B7-1 (CD80), B7-2 (CD86), B7-H1 (PD-L1, CD274), B7-DC (PDCD1LG2, PD-L2, CD273), B7-H2 (B7RP1, ICOS-L, CD275), B7-H3 (CD276), B7-H4 (B7x, B7S1, Vtcn1), B7-H5 (VISTA, Gi24, DD1α, Dies1 SISP1), B7-H6 (NCR3LG1), B7-H7 (HHLA2), and Ig-like domain-containing receptor 2 (ILDR2). Interaction of the B7 family of immune-regulatory ligands with the corresponding receptors resulted in the induction and inhibition of T cell responses by sending co-stimulatory and co-inhibitory signals, respectively. Manipulation of the signals provided by the B7 family has significant potential in the management of GC.
Collapse
Affiliation(s)
- Nadia Bolandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Laboratory of Experimental Pharmacology, IRCCS Istituto Tumori Giovanni Paolo II, 70124 Bari, Italy
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Zahra Asadzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
| | - Mina Afrashteh Nour
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Biochemistry, Faculty of Medicine, Urmia University of Medical Sciences, Urmia 571478334, Iran
| | - Oronzo Brunetti
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, University of Catania, Via S. Sofia 97, 95100 Catania, Italy;
| | - Nicola Silvestris
- Medical Oncology Unit—IRCCS Istituto Tumori “Giovanni Paolo II” of Bari, 70124 Bari, Italy;
- Department of Biomedical Sciences and Human Oncology (DIMO), University of Bari, 70124 Bari, Italy
- Correspondence: (N.S.); (B.B.); Tel.: +98-413-3371440 (B.B.); Fax: +98-413-3371311 (B.B.)
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran; (N.B.); (A.D.); (N.H.); (A.B.); (Z.A.); (M.A.N.)
- Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Pharmaceutical Analysis Research Center, Tabriz University of Medical Sciences, Tabriz 516615731, Iran
- Correspondence: (N.S.); (B.B.); Tel.: +98-413-3371440 (B.B.); Fax: +98-413-3371311 (B.B.)
| |
Collapse
|
162
|
Hatzioannou A, Boumpas A, Papadopoulou M, Papafragkos I, Varveri A, Alissafi T, Verginis P. Regulatory T Cells in Autoimmunity and Cancer: A Duplicitous Lifestyle. Front Immunol 2021; 12:731947. [PMID: 34539668 PMCID: PMC8446642 DOI: 10.3389/fimmu.2021.731947] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/18/2021] [Indexed: 01/08/2023] Open
Abstract
Regulatory T (Treg) cells, possess a strategic role in the maintenance of immune homeostasis, and their function has been closely linked to development of diverse pathologies including autoimmunity and cancer. Comprehensive studies in various disease contexts revealed an increased plasticity as a characteristic of Treg cells. Although Treg cell plasticity comes in various flavors, the major categories enclose the loss of Foxp3 expression, which is the master regulator of Treg cell lineage, giving rise to “ex-Treg” cells and the “fragile” Treg cells in which FOXP3 expression is retained but accompanied by the engagement of an inflammatory program and attenuation of the suppressive activity. Treg cell plasticity possess a tremendous therapeutic potential either by inducing Treg cell de-stabilization to promote anti-tumor immunity, or re-enforcing Treg cell stability to attenuate chronic inflammation. Herein, we review the literature on the Treg cell plasticity with lessons learned in autoimmunity and cancer and discuss challenges and open questions with potential therapeutic implications.
Collapse
Affiliation(s)
- Aikaterini Hatzioannou
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany
| | - Athina Boumpas
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Miranta Papadopoulou
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Iosif Papafragkos
- Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece.,Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| | - Athina Varveri
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Themis Alissafi
- Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece
| | - Panayotis Verginis
- Institute for Clinical Chemistry and Laboratory Medicine, Faculty of Medicine, Technische Universität Dresden, Dresden, Germany.,Center of Clinical, Experimental Surgery & Translational Research, Biomedical Research Foundation of the Academy of Athens, Athens, Greece.,Institute of Molecular Biology and Biotechnology (IMBB), Foundation for Research and Technology - Hellas (FORTH), Heraklion, Greece.,Laboratory of Immune Regulation and Tolerance, Division of Basic Sciences, Medical School, University of Crete, Heraklion, Greece
| |
Collapse
|
163
|
Hosseinzadeh R, Feizisani F, Shomali N, Abdelbasset WK, Hemmatzadeh M, Gholizadeh Navashenaq J, Jadidi-Niaragh F, Bokov DO, Janebifam M, Mohammadi H. PD-1/PD-L1 blockade: Prospectives for immunotherapy in cancer and autoimmunity. IUBMB Life 2021; 73:1293-1306. [PMID: 34538007 DOI: 10.1002/iub.2558] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 08/10/2021] [Accepted: 09/06/2021] [Indexed: 12/14/2022]
Abstract
Immune checkpoint blockade therapy (ICBT) has become a successful cancer treatment approach in the field of cancer immunotherapy. Blockade of programmed death 1 (PD-1) and programmed death-ligand 1 (PD-L1) with monoclonal antibodies have been known as successful examples of cancer immunotherapy in recent years. Although ICBT has been shown to be beneficial in cancers, such benefits have only been seen in a portion of cancer patients. In this regard, enhancing the therapeutic effects of inhibiting PD-1 and PD-L1 and reducing the side effects of this approach can be considered as a potential approach in a successful ICBT. In this review, we have highlighted new viewpoints regarding improving the therapeutic effect of PD-1 and PD-L1 blockades in cancer therapy. Besides, their expression levels as a biomarker with prognostic value, their role in intestinal microbiota modulation, combination therapy, and immune-related side effects (irAEs) have been discussed.
Collapse
Affiliation(s)
- Ramin Hosseinzadeh
- Department of Medical Immunology, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Fahimeh Feizisani
- Student Research Committee, Sarab Faculty of Medical Sciences, Sarab, Iran
| | - Navid Shomali
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Walid Kamal Abdelbasset
- Department of Health and Rehabilitation Sciences, College of Applied Medical Sciences, Prince Sattam bin Abdulaziz University, Al Kharj, Saudi Arabia.,Department of Physical Therapy, Kasr Al-Aini Hospital, Cairo University, Giza, Egypt
| | - Maryam Hemmatzadeh
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Farhad Jadidi-Niaragh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Faculty of Medicine, Department of Immunology, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dmitry O Bokov
- Institute of Pharmacy, Sechenov First Moscow State Medical University, Moscow, Russian Federation.,Laboratory of Food Chemistry, Federal Research Center of Nutrition, Biotechnology and Food Safety, Moscow, Russian Federation
| | - Morteza Janebifam
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamed Mohammadi
- Department of Immunology, School of Medicine, Alborz University of Medical Sciences, Karaj, Iran.,Noncommunicable Diseases Research Center, Alborz University of Medical Sciences, Karaj, Iran
| |
Collapse
|
164
|
Wang J, Wu H, Chen Y, Zhu J, Sun L, Li J, Yao Z, Chen Y, Zhang X, Xia S, Chen W, Shi T. B7-H5 blockade enhances CD8 + T-cell-mediated antitumor immunity in colorectal cancer. Cell Death Discov 2021; 7:248. [PMID: 34537815 PMCID: PMC8449782 DOI: 10.1038/s41420-021-00628-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 08/15/2021] [Accepted: 08/26/2021] [Indexed: 12/22/2022] Open
Abstract
Negative immune checkpoint blockade immunotherapy has shown potential for multiple malignancies including colorectal cancer (CRC). B7-H5, a novel negative immune checkpoint regulator, is highly expressed in tumor tissues and promotes tumor immune escape. However, the clinical significance of B7-H5 expression in CRC and the role of B7-H5 in the tumor microenvironment (TME) has not been fully clarified. In this study, we observed that high B7-H5 expression in CRC tissues was significantly correlated with the lymph node involvement, AJCC stage, and survival of CRC patients. A significant inverse correlation was also observed between B7-H5 expression and CD8+ T-cell infiltration in CRC tissues. Kaplan-Meier analysis showed that patients with high B7-H5 expression and low CD8+ T-cell infiltration had the worst prognosis in our cohort of CRC patients. Remarkably, both high B7-H5 expression and low CD8+ T infiltration were risk factors for overall survival. Additionally, B7-H5 blockade using a B7-H5 monoclonal antibody (B7-H5 mAb) effectively suppressed the growth of MC38 colon cancer tumors by enhancing the infiltration and Granzyme B production of CD8+ T cells. Importantly, the depletion of CD8+ T cells obviously abolished the antitumor effect of B7-H5 blockade in the MC38 tumors. In sum, our findings suggest that B7-H5 may be a valuably prognostic marker for CRC and a potential target for CRC immunotherapy.
Collapse
Affiliation(s)
- Jiayu Wang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Hongya Wu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yanjun Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Jinghan Zhu
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Linqing Sun
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Juntao Li
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Zhendong Yao
- Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuqi Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Xueguang Zhang
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.,Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China.,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Suhua Xia
- Department of Oncology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Weichang Chen
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China. .,Department of Gastroenterology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Tongguo Shi
- Jiangsu Institute of Clinical Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China. .,Suzhou Key Laboratory for Tumor Immunology of Digestive Tract, The First Affiliated Hospital of Soochow University, Suzhou, China. .,Jiangsu Key Laboratory of Clinical Immunology, Soochow University, Suzhou, China. .,Jiangsu Key Laboratory of Gastrointestinal Tumor Immunology, The First Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
165
|
Li L, Xu XT, Wang LL, Qin SB, Zhou JY. Expression and clinicopathological significance of Foxp3 and VISTA in cervical cancer. Am J Transl Res 2021; 13:10428-10438. [PMID: 34650712 PMCID: PMC8507058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 01/27/2021] [Indexed: 06/13/2023]
Abstract
OBJECTIVE To detect the expression differences of Foxp3 and VISTA in chronic cervical inflammation, cervical intraepithelial neoplasia, and cervical cancer, and to explore the role of Foxp3 and VISTA in the development of cervical cancer and the effect of Foxp3 and VISTA on the prognosis of cervical cancer, to provide a theoretical basis for clinical immunotherapy of cervical cancer. METHODS We collected 130 paraffin specimens of cervical tissue, which included 70 cases of cervical cancer tissue, 40 cases of cervical intraepithelial neoplasia tissues and 20 cases of chronic cervicitis. The expression of Foxp3 and VISTA in each group was detected, and the study was conducted based on the clinicopathological characteristics of the patients. The patients were followed up and the prognosis was statistically analyzed. RESULT 1. The expression of Foxp3 and VISTA was statistically different between the cervical cancer group and other groups. 2. Expressions of Foxp3 and VISTA were significantly correlated. 3. In 70 cases of cervical cancer, the expression of Foxp3 and VISTA was related to the clinical stage. 4. The 3-year survival rate of 70 patients with cervical cancer was 72.9%, and there were no factors affecting 3-year OS found. The expression of Foxp3 and VISTA was significantly correlated with the prognosis of cervical cancer. Foxp3 and VISTA double positive expression group had the worst prognosis. CONCLUSION 1. In cervical cancer, the expression of Foxp3 and VISTA was significantly higher than that of cervical intraepithelial neoplasia and chronic cervicitis, which suggested that they were closely related to the occurrence and growth of cervical cancer. 2. The expression of Foxp3 and VISTA was significantly related. 3. The positive expression of Foxp3 and VISTA could be used as independent prognostic factors for cervical cancer prognosis providing a strong basis for cervical cancer immunotherapy.
Collapse
Affiliation(s)
- Li Li
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Xiao-Ting Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Li-Li Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Song-Bing Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| | - Ju-Ying Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Soochow University Suzhou 215006, China
| |
Collapse
|
166
|
Pisibon C, Ouertani A, Bertolotto C, Ballotti R, Cheli Y. Immune Checkpoints in Cancers: From Signaling to the Clinic. Cancers (Basel) 2021; 13:cancers13184573. [PMID: 34572799 PMCID: PMC8468441 DOI: 10.3390/cancers13184573] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/19/2022] Open
Abstract
The immune system is known to help fight cancers. Ten years ago, the first immune checkpoint inhibitor targeting CTLA4 was approved by the FDA to treat patients with metastatic melanoma. Since then, immune checkpoint therapies have revolutionized the field of oncology and the treatment of cancer patients. Numerous immune checkpoint inhibitors have been developed and tested, alone or in combination with other treatments, in melanoma and other cancers, with overall clear benefits to patient outcomes. However, many patients fail to respond or develop resistance to these treatments. It is therefore essential to decipher the mechanisms of action of immune checkpoints and to understand how immune cells are affected by signaling to be able to understand and overcome resistance. In this review, we discuss the signaling and effects of each immune checkpoint on different immune cells and their biological and clinical relevance. Restoring the functionality of T cells and their coordination with other immune cells is necessary to overcome resistance and help design new clinical immunotherapy strategies. In this respect, NK cells have recently been implicated in the resistance to anti-PD1 evoked by a protein secreted by melanoma, ITGBL1. The complexity of this network will have to be considered to improve the efficiency of future immunotherapies and may lead to the discovery of new immune checkpoints.
Collapse
Affiliation(s)
- Céline Pisibon
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Amira Ouertani
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Corine Bertolotto
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Robert Ballotti
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
| | - Yann Cheli
- Université Côte d’Azur, 06103 Nice, France; (C.P.); (A.O.); (C.B.); (R.B.)
- INSERM, Centre Méditerranéen de Médecine Moléculaire, Biology and Pathologies of Melanocytes, Team1, 06200 Nice, France
- Correspondence:
| |
Collapse
|
167
|
Kelly WJ, Giles AJ, Gilbert M. T lymphocyte-targeted immune checkpoint modulation in glioma. J Immunother Cancer 2021; 8:jitc-2019-000379. [PMID: 32051289 PMCID: PMC7057419 DOI: 10.1136/jitc-2019-000379] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2020] [Indexed: 02/07/2023] Open
Abstract
Immunomodulatory therapies targeting inhibitory checkpoint molecules have revolutionized the treatment of solid tumor malignancies. Concerns about whether systemic administration of an immune checkpoint inhibitor could impact primary brain tumors were answered with the observation of definitive responses in pediatric patients harboring hypermutated gliomas. Although initial clinical results in patients with glioblastoma (GBM) were disappointing, recently published results have demonstrated a potential survival benefit in patients with recurrent GBM treated with neoadjuvant programmed cell death protein 1 blockade. While these findings necessitate verification in subsequent studies, they support the possibility of achieving clinical meaningful immune responses in malignant primary brain tumors including GBM, a disease in dire need of additional therapeutic options. There are several challenges involved in treating glioma with immune checkpoint modulators including the immunosuppressive nature of GBM itself with high inhibitory checkpoint expression, the immunoselective blood brain barrier impairing the ability for peripheral lymphocytes to traffic to the tumor microenvironment and the high prevalence of corticosteroid use which suppress lymphocyte activation. However, by simultaneously targeting multiple costimulatory and inhibitory pathways, it may be possible to achieve an effective antitumoral immune response. To this end, there are now several novel agents targeting more recently uncovered “second generation” checkpoint molecules. Given the multiplicity of drugs being considered for combination regimens, an increased understanding of the mechanisms of action and resistance combined with more robust preclinical and early clinical testing will be needed to be able to adequately test these agents. This review summarizes our current understanding of T lymphocyte-modulating checkpoint molecules as it pertains to glioma with the hope for a renewed focus on the most promising therapeutic strategies.
Collapse
Affiliation(s)
| | - Amber Jin Giles
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| | - Mark Gilbert
- Neuro-Oncology Branch, National Cancer Institute, Bethesda, Maryland, USA
| |
Collapse
|
168
|
Im E, Sim DY, Lee HJ, Park JE, Park WY, Ko S, Kim B, Shim BS, Kim SH. Immune functions as a ligand or a receptor, cancer prognosis potential, clinical implication of VISTA in cancer immunotherapy. Semin Cancer Biol 2021; 86:1066-1075. [PMID: 34428551 DOI: 10.1016/j.semcancer.2021.08.008] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2021] [Revised: 07/28/2021] [Accepted: 08/17/2021] [Indexed: 01/15/2023]
Abstract
Since cancer immunotherapy with immune checkpoint inhibitors of PD/PDL-1 and CTLA-4 limited efficacy to the patients due to resistance during the current decade, novel target is required for customized treatment due to tumor heterogeneity. V-domain Ig-containing suppressor of T cell activation (VISTA), a programmed death protein-1(PD-1) homolog expressed on T cells and on antigen presenting cells(APC), has emerged as a new target in several cancers. Though VISTA inhibitors including CA-170 are considered attractive in cancer immunotherapy to date, the information on VISTA as a potent biomarker of cancer prognosis and its combination therapy is still lacking to date. Thus, in this review, we discussed extracellular domain, ligands, expression, immune functions and clinical implications of VISTA and finally suggested conclusion and perspectives.
Collapse
Affiliation(s)
- Eunji Im
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Deok Yong Sim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Hyo-Jung Lee
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Ji Eon Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Woon Yi Park
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - SeongGyu Ko
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bonglee Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea
| | - Bum Sang Shim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| | - Sung-Hoon Kim
- Cancer Molecular Target Herbal Research Lab, College of Korean Medicine, Kyung Hee University, Seoul 02447, Republic of Korea.
| |
Collapse
|
169
|
High-Affinity Anti-VISTA Antibody Protects against Sepsis by Inhibition of T Lymphocyte Apoptosis and Suppression of the Inflammatory Response. Mediators Inflamm 2021; 2021:6650329. [PMID: 34366711 PMCID: PMC8339895 DOI: 10.1155/2021/6650329] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 06/08/2021] [Accepted: 07/06/2021] [Indexed: 12/29/2022] Open
Abstract
Background B7 family members and ligands have been identified as critical checkpoints in orchestrating the immune response during sepsis. V-domain Ig suppressor of T cell activation (VISTA) is a new inhibitory immune checkpoint involved in restraining T cell response. Previous studies demonstrated that VISTA engagement on T cells and myeloid cells could transmit inhibitory signals, resulting in reduced activation and function. The current study was designed to determine the potential therapeutic effects of a high-affinity anti-VISTA antibody (clone MH5A) in a murine model of sepsis. Methods Polymicrobial sepsis was induced in male C57BL/6 mice via cecal ligation and puncture. Expression profiles of VISTA on T lymphocytes and macrophage were examined at 24 and 72 h postsurgery. The effects of anti-VISTA mAb on the 7-day survival, lymphocyte apoptosis, cytokine expression, bacterial burden, and vital organ damage were determined. Furthermore, the effects of anti-VISTA mAb on CD3+ T cell apoptosis and macrophage activation were determined in vitro. Results VISTA was substantially expressed on T cells and macrophages in sham-operated mice; septic peritonitis did not induce significant changes in the expression profiles. Treatment with MH5A improved the survival of septic mice, accompanied by reduced lymphocyte apoptosis, decreased cytokine expression, and enhanced bacterial clearance. Engagement of VISTA receptor with MH5A mitigated CD3+ T cell apoptosis cultured from CLP mice and suppressed LPS-induced cytokine production by macrophage in vitro. Conclusion The present study identified VISTA as a novel immune checkpoint in the regulation of T cell and macrophage response during sepsis. Modulation of the VISTA pathway might offer a promising opportunity in the immunotherapy for sepsis.
Collapse
|
170
|
Mattox AK, Roelands J, Saal TM, Cheng Y, Rinchai D, Hendrickx W, Young GD, Diefenbach TJ, Berger AE, Westra WH, Bishop JA, Faquin WC, Marincola FM, Pittet MJ, Bedognetti D, Pai SI. Myeloid Cells Are Enriched in Tonsillar Crypts, Providing Insight into the Host Tropism of Human Papillomavirus. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1774-1786. [PMID: 34303699 DOI: 10.1016/j.ajpath.2021.06.012] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 05/14/2021] [Accepted: 06/14/2021] [Indexed: 10/20/2022]
Abstract
Viruses are the second leading cause of cancer worldwide, and human papillomavirus (HPV)-associated head and neck cancers are increasing in incidence in the United States. HPV preferentially infects the crypts of the tonsils rather than the surface epithelium. The present study sought to characterize the unique microenvironment within the crypts to better understand the host tropism of HPV to a lymphoid-rich organ. Laser-capture microdissection of distinct anatomic areas (crypts, surface epithelium, and germinal centers) of the tonsil, coupled with transcriptional analysis and multiparameter immunofluorescence staining, was performed and demonstrated that the tonsillar crypts are enriched with myeloid populations that co-express multiple canonical and noncanonical immune checkpoints, including PD-L1, CTLA-4, HAVCR2 (TIM-3), ADORA2A, IDO1, BTLA, LGALS3, CDH1, CEACAM1, PVR, and C10orf54 (VISTA). The resident monocytes may foster a permissive microenvironment that facilitates HPV infection and persistence. Furthermore, the myeloid populations within HPV-associated tonsil cancers co-express the same immune checkpoints, providing insight into potential novel immunotherapeutic targets for HPV-associated head and neck cancers.
Collapse
Affiliation(s)
- Austin K Mattox
- Department of Oncology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Jessica Roelands
- Cancer Program, Research Branch, Sidra Medicine, Doha, Qatar; Department of Surgery, Leiden University Medical Center, Leiden, the Netherlands
| | - Talia M Saal
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Yang Cheng
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts
| | - Darawan Rinchai
- Cancer Program, Research Branch, Sidra Medicine, Doha, Qatar
| | | | - Geoffrey D Young
- Miami Cancer Institute and Department of Surgery, Florida International University, Miami, Florida
| | | | - Alan E Berger
- Division of Allergy and Clinical Immunology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - William H Westra
- Department of Pathology, Icahn School of Medicine at the Mount Sinai Hospital, New York, New York
| | - Justin A Bishop
- Department of Pathology, UT Southwestern Medical Center, Dallas, Texas
| | - William C Faquin
- Department of Pathology, Massachusetts General Hospital, Boston, Massachusetts
| | | | - Mikael J Pittet
- Center for Systems Biology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | | | - Sara I Pai
- Division of Surgical Oncology, Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts; Department of Otolaryngology-Head and Neck Surgery, Massachusetts Eye and Ear, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
171
|
Tagliamento M, Bironzo P, Novello S. New emerging targets in cancer immunotherapy: the role of VISTA. ESMO Open 2021; 4:e000683. [PMID: 32554470 PMCID: PMC7305420 DOI: 10.1136/esmoopen-2020-000683] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 03/24/2020] [Accepted: 03/26/2020] [Indexed: 01/19/2023] Open
Abstract
The immune surveillance system is complex and regulated by different actors. Programmed death protein-ligand 1 (PD-L1), the only approved biomarker in clinical practice, has proven to be imperfect in selecting patients to immune checkpoint inhibitors treatment. Therefore, new biomarkers, and new therapeutic targets, are needed to maximise the efficacy of immunotherapy. V-domain Ig Suppressor of T-cell Activation (VISTA) is a programmed death protein-1 (PD-1) homolog expressed on T cells and on antigen-presenting cells, which regulates processes of activation and repression of the immune system with not yet completely clarified mechanisms. Its blockage has demonstrated in vitro and in vivo antitumour activity. The clinical research of VISTA antagonists is ongoing. Particularly, CA-170, an orally delivered dual inhibitor of VISTA and PD-L1, has shown to have clinical efficacy in phase I and II clinical trials in different advanced solid tumour types. Further data are needed to define whether this drug class can become a new therapeutic option for patients with VISTA expressing cancers.
Collapse
Affiliation(s)
- Marco Tagliamento
- Department of Medical Oncology, Medical Oncology 2, University of Genova & IRCCS Ospedale Policlinico San Martino, Genova, Italy.
| | - Paolo Bironzo
- Oncology Department, Thoracic Oncology, University of Turin & San Luigi Hospital, Orbassano (Turin), Italy
| | - Silvia Novello
- Oncology Department, Thoracic Oncology, University of Turin & San Luigi Hospital, Orbassano (Turin), Italy
| |
Collapse
|
172
|
Wright Q, Gonzalez Cruz JL, Wells JW, Leggatt GR. PD-1 and beyond to Activate T Cells in Cutaneous Squamous Cell Cancers: The Case for 4-1BB and VISTA Antibodies in Combination Therapy. Cancers (Basel) 2021; 13:3310. [PMID: 34282763 PMCID: PMC8269268 DOI: 10.3390/cancers13133310] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 06/24/2021] [Accepted: 06/27/2021] [Indexed: 01/02/2023] Open
Abstract
Non-melanoma skin cancers (NMSC) have a higher incidence than all other cancers combined with cutaneous squamous cell carcinoma (cSCC), capable of metastasis, representing approximately 20% of NMSCs. Given the accessibility of the skin, surgery is frequently employed to treat localized disease, although certain localities, the delineation of clear margins, frequency and recurrence of tumors can make these cancers inoperable in a subset of patients. Other treatment modalities, including cryotherapy, are commonly used for individual lesions, with varying success. Immunotherapy, particularly with checkpoint antibodies, is increasingly a promising therapeutic approach in many cancers, offering the potential advantage of immune memory for protection against lesion recurrence. This review addresses a role for PD-1, 4-1BB and VISTA checkpoint antibodies as monotherapies, or in combination as a therapeutic treatment for both early and late-stage cSCC.
Collapse
Affiliation(s)
| | | | | | - Graham R. Leggatt
- The University of Queensland Diamantina Institute, The University of Queensland, Woolloongabba, QLD 4102, Australia; (Q.W.); (J.L.G.C.); (J.W.W.)
| |
Collapse
|
173
|
Peng X, Zhao L, Liu J, Guo X, Ding Y. Comparative transcriptome analyses of the liver between Xenocypris microlepis and Xenocypris davidi under low copper exposure. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2021; 236:105850. [PMID: 34022695 DOI: 10.1016/j.aquatox.2021.105850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 06/12/2023]
Abstract
Copper is one of the most ubiquitous environmental pollutants worldwide. Previous studies have focused on the toxicology of high copper exposure, while there has been comparatively less research on the biological effects of low copper exposure. Low concentrations of copper often exist in freshwater ecosystems, and its impact on the fish is unclear. Both Xenocypris microlepis and Xenocypris davidi are bottom-feeding fishes widely distributed in freshwater ecosystems of China, and they are more likely to be contaminated by low concentrations of copper. Low copper exposure may have effects on molecular regulation at the level of gene expression in the two Xenocypris species. To investigate gene expression differences involved in the response to low copper concentrations between X. microlepis and X. davidi, we established the responses to low copper exposure of 0.01 mg/L for 14 days at the transcriptional level, and RNA-Seq was used to perform a comparative transcriptomic analysis of the liver. A total of 74,135 and 60,894 unigenes from X. microlepis and X. davidi were assembled by transcriptome profiling, respectively. Among these, 84 genes of X. microlepis and 165 genes of X. davidi were identified as differentially expressed genes (DEGs). There were 60 and 135 up-regulated, 24 and 30 down-regulated genes in the two species, respectively. Comparative transcriptome analyses identified five differentially co-expressed genes (DCGs) related to low copper exposure from the DEGs of the two Xenocypris species. The five DCGs were related to the fishes' growth, antioxidant system, immune system and heavy metal tolerance. The results could help us to understand the molecular mechanisms of the response to low copper exposure, and the data should provide a valuable transcriptomic resource for the genus Xenocypris.
Collapse
Affiliation(s)
- Xinliang Peng
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China; College of Fisheries, Xinyang Agriculture and Forestry University, Xinyang 464000, China
| | - Liangjie Zhao
- College of Fisheries, Xinyang Agriculture and Forestry University, Xinyang 464000, China; Fishery Biological Engineering Technology Research Center of Henan Province, Xinyang 464000, China
| | - Jun Liu
- College of Fisheries, Xinyang Agriculture and Forestry University, Xinyang 464000, China; Fishery Biological Engineering Technology Research Center of Henan Province, Xinyang 464000, China
| | - Xusheng Guo
- College of Fisheries, Xinyang Agriculture and Forestry University, Xinyang 464000, China; Fishery Biological Engineering Technology Research Center of Henan Province, Xinyang 464000, China.
| | - Yu Ding
- Fisheries College, Guangdong Ocean University, Zhanjiang 524088, China.
| |
Collapse
|
174
|
Tu VY, Ayari A, O’Connor RS. Beyond the Lactate Paradox: How Lactate and Acidity Impact T Cell Therapies against Cancer. Antibodies (Basel) 2021; 10:25. [PMID: 34203136 PMCID: PMC8293081 DOI: 10.3390/antib10030025] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 06/14/2021] [Accepted: 06/22/2021] [Indexed: 11/16/2022] Open
Abstract
T cell therapies, including CAR T cells, have proven more effective in hematologic malignancies than solid tumors, where the local metabolic environment is distinctly immunosuppressive. In particular, the acidic and hypoxic features of the tumor microenvironment (TME) present a unique challenge for T cells. Local metabolism is an important consideration for activated T cells as they undergo bursts of migration, proliferation and differentiation in hostile soil. Tumor cells and activated T cells both produce lactic acid at high rates. The role of lactic acid in T cell biology is complex, as lactate is an often-neglected carbon source that can fuel TCA anaplerosis. Circulating lactate is also an important means to regulate redox balance. In hypoxic tumors, lactate is immune-suppressive. Here, we discuss how intrinsic- (T cells) as well as extrinsic (tumor cells and micro-environmental)-derived metabolic factors, including lactate, suppress the ability of antigen-specific T cells to eradicate tumors. Finally, we introduce recent discoveries that target the TME in order to potentiate T cell-based therapies against cancer.
Collapse
Affiliation(s)
- Violet Y. Tu
- Center for Cellular Immunotherapies, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Biological Physics, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Asma Ayari
- Nucleus Biologics, LLC., San Diego, CA 92127, USA;
| | - Roddy S. O’Connor
- Center for Cellular Immunotherapies, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA;
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine of the University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
175
|
Rogers BM, Smith L, Dezso Z, Shi X, DiGiammarino E, Nguyen D, Sethuraman S, Zheng P, Choi D, Zhang D, Nguyen A, McGuire K, Liu W, Chung N, Chao DT, Ye S, Starbeck-Miller GR. VISTA is an activating receptor in human monocytes. THE JOURNAL OF EXPERIMENTAL MEDICINE 2021; 218:212264. [PMID: 34106206 PMCID: PMC8193568 DOI: 10.1084/jem.20201601] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/28/2020] [Revised: 03/01/2021] [Accepted: 05/10/2021] [Indexed: 11/26/2022]
Abstract
As indicated by its name, V-domain Ig suppressor of T cell activation (VISTA) is thought to serve primarily as an inhibitory protein that limits immune responses. VISTA antibodies can dampen the effects of several concomitantly elicited activation signals, including TCR and TLR activation, but it is currently unclear if VISTA agonism could singly affect immune cell biology. In this study, we discovered two novel VISTA antibodies and characterized their effects on human peripheral blood mononuclear cells by scRNA/CITE-seq. Both antibodies appeared to agonize VISTA in an Fc-functional manner to elicit transcriptional and functional changes in monocytes consistent with activation. We also used pentameric VISTA to identify Syndecan-2 and several heparan sulfate proteoglycan synthesis genes as novel regulators of VISTA interactions with monocytic cells, adding further evidence of bidirectional signaling. Together, our study highlights several novel aspects of VISTA biology that have yet to be uncovered in myeloid cells and serves as a foundation for future research.
Collapse
Affiliation(s)
| | | | | | - Xu Shi
- AbbVie Biotherapeutics Inc., Redwood City, CA
| | | | | | | | | | | | - Dong Zhang
- AbbVie Biotherapeutics Inc., Redwood City, CA
| | | | | | - Wei Liu
- AbbVie Biotherapeutics Inc., Redwood City, CA
| | | | | | - Shiming Ye
- AbbVie Biotherapeutics Inc., Redwood City, CA
| | | |
Collapse
|
176
|
Mei Y, Bi WL, Agolia J, Hu C, Giantini Larsen AM, Meredith DM, Al Abdulmohsen S, Bale T, Dunn GP, Abedalthagafi M, Dunn IF. Immune profiling of pituitary tumors reveals variations in immune infiltration and checkpoint molecule expression. Pituitary 2021; 24:359-373. [PMID: 33492612 DOI: 10.1007/s11102-020-01114-3] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/27/2020] [Indexed: 12/30/2022]
Abstract
PURPOSE Pituitary tumors are the second most common primary brain tumors. Functional tumors demonstrate increased PD-L1 expression, but expression of other checkpoint regulators has not been characterized. We sought to characterize the immune microenvironment of human pituitary tumors to identify new treatment opportunities. METHODS 72 pituitary tumors were evaluated for expression of the immune regulatory markers programmed death ligand 1 (PD-L1), programmed death ligand 2 (PD-L2), V-domain Ig suppressor of T cell activation (VISTA), lymphocyte activation gene 3 (LAG3) and tumor necrosis factor receptor superfamily member 4 (OX40) by immunohistochemistry (IHC). Lymphocyte infiltration, macrophage infiltration, and angiogenesis were analyzed using IHC. Expression of pituitary tumor initiating cell marker CD15 and mismatch repair proteins MutS protein homolog 2 (MSH2) and MutS protein homolog 6 (MSH6) was also assessed. RESULTS Pituitary tumors were infiltrated by macrophages and T cells, and they expressed varying levels of PD-L1, PD-L2, VISTA, LAG3, and OX40. Functional tumors and tumors with high expression of tumor stem cell markers had higher immune cell infiltration and greater expression of immunosuppressive checkpoint regulators. Increased PD-L1 and LAG3 and reduced VISTA were observed in primary tumors compared to recurrent tumors. CONCLUSION Immune cell infiltration and checkpoint regulator expression vary depending on functional status and presence of pituitary tumor initiating cells. Functional tumors may have a particularly immunosuppressive microenvironment. Further studies of immune checkpoint blockade of pituitary tumors, particularly functional tumors, are warranted, though combination therapy may be required.
Collapse
Affiliation(s)
- Yu Mei
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Wenya Linda Bi
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA.
- Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA.
| | - James Agolia
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
| | - Changchen Hu
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
- Department of Neurosurgery, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan, China
| | | | - David M Meredith
- Department of Pathology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, USA
| | - Sally Al Abdulmohsen
- Center for Skull Base and Pituitary Surgery, Department of Neurosurgery, Harvard Medical School, Brigham and Women's Hospital, 60 Fenwood Road, Boston, MA, 02115, USA
- King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Tejus Bale
- Department of Neuropathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Gavin P Dunn
- Department of Neurosurgery, Washington University School of Medicine, St. Louis, MO, USA
- Andrew M. and Jane M. Bursky Center for Human Immunology and Immunotherapy Programs, Washington University School of Medicine, St. Louis, MO, USA
| | - Malak Abedalthagafi
- King Fahad Medical City and King Abdulaziz City for Science and Technology, Riyadh, Saudi Arabia
| | - Ian F Dunn
- Department of Neurosurgery, University of Oklahoma Health Sciences Center, HHDC Suite 4000, 1000 N. Lincoln Blvd, Oklahoma City, OK, 73104, USA.
| |
Collapse
|
177
|
Hosseinkhani N, Derakhshani A, Shadbad MA, Argentiero A, Racanelli V, Kazemi T, Mokhtarzadeh A, Brunetti O, Silvestris N, Baradaran B. The Role of V-Domain Ig Suppressor of T Cell Activation (VISTA) in Cancer Therapy: Lessons Learned and the Road Ahead. Front Immunol 2021; 12:676181. [PMID: 34093577 PMCID: PMC8172140 DOI: 10.3389/fimmu.2021.676181] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Accepted: 04/29/2021] [Indexed: 12/12/2022] Open
Abstract
Immune checkpoints (ICs) have pivotal roles in regulating immune responses. The inhibitory ICs in the tumor microenvironment (TME) have been implicated in the immune evasion of tumoral cells. Therefore, identifying and targeting these inhibitory ICs might be critical for eliminating tumoral cells. V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel inhibitory IC that is expressed on myeloid cells, lymphoid cells, and tumoral cells; therefore, VISTA can substantially regulate innate and adaptive anti-tumoral immune responses. Besides, growing evidence indicates that VISTA blockade can enhance the sensitivity of tumoral cells to conventional IC-based immunotherapy, e.g., cytotoxic T lymphocyte antigen 4 (CTLA-4) inhibitors. In this regard, the current study aimed to review the current evidence about the structure and expression pattern of VISTA, its role in TME, the clinicopathological significance of VISTA, and its prognostic values in various cancers. Besides, this review intended to collect the lessons from the recent pre-clinical and clinical studies and propose a strategy to overcome tumor immune-resistance states.
Collapse
Affiliation(s)
- Negar Hosseinkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Afshin Derakhshani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Mahdi Abdoli Shadbad
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Antonella Argentiero
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Vito Racanelli
- Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Oronzo Brunetti
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy
| | - Nicola Silvestris
- Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Istituto Tumori "Giovanni Paolo II" of Bari, Bari, Italy.,Department of Biomedical Sciences and Human Oncology, Aldo Moro University of Bari, Bari, Italy
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.,Department of Immunology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.,Research Center for Pharmaceutical Nanotechnology, Tabriz University of Medical Sciences, Tabriz, Iran
| |
Collapse
|
178
|
Jiang M, Wu C, Zhang L, Sun C, Wang H, Xu Y, Sun H, Zhu J, Zhao W, Fang Q, Yu J, Chen P, Wu S, Zheng Z, He Y, Zhou C. FOXP3-based immune risk model for recurrence prediction in small-cell lung cancer at stages I-III. J Immunother Cancer 2021; 9:jitc-2021-002339. [PMID: 34006632 PMCID: PMC8137193 DOI: 10.1136/jitc-2021-002339] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/20/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Immunotherapies may prolong the survival of patients with small-cell lung cancer (SCLC) to some extent. The role of forkhead box protein P3 (FOXP3) in tumor microenvironment (TME) remains controversial. We aimed to examine FOXP3-related expression characteristics and prognostic values and to develop a clinically relevant predictive system for SCLC. METHODS We enrolled 102 patients with histologically confirmed SCLC at stages I-III. Through immunohistochemistry, we determined the expression pattern of FOXP3 and its association with other immune biomarkers. By machine learning and statistical analysis, we constructed effective immune risk score models. Furthermore, we examined FOXP3-related enrichment pathways and TME traits in distinct cohorts. RESULTS In SCLC, FOXP3 level was significantly associated with status of programmed death-ligand 1 (PD-L1), programmed cell death protein 1 (PD-1), CD4, CD8, and CD3 (p=0.002, p=0.001, p=0.002, p=0.030, and p<0.001). High FOXP3 expression showed longer relapse-free survival (RFS) than the low-level group (41.200 months, 95% CI 26.937 to 55.463, vs 14.000 months, 95% CI 8.133 to 19.867; p=0.008). For tumor-infiltrating lymphocytes (TILs), subgroup analysis demonstrated FOXP3 and PD-1, PD-L1, lymphocyte activation gene-3, CD3, CD4, or CD8 double positive were significantly correlated with longer RFS. We further performed importance evaluation for immune biomarkers, constructed an immune risk score incorporating the top three important biomarkers, FOXP3, TIL PD-L1, and CD8, and found their independently prognostic role to predict SCLC relapse. Better predictive performance was achieved in this immune risk model compared with single-indicator-based or two-indicator-based prediction systems (area under the curve 0.715 vs 0.312-0.711). Then, relapse prediction system integrating clinical staging and immune risk score was established, which performed well in different cohorts. High FOXP3-related genes were enriched in several immune-related pathways, and the close relationships of interleukin-2, CD28, basic excision repair genes MUTYH, POLD1, POLD2, and oxidative phosphorylation related gene cytochrome c oxidase subunit 8A with FOXP3 expression were revealed. Moreover, we found low-immune risk score group had statistically higher activated CD4+ memory T cells (p=0.014) and plasma cells (p=0.049) than the high-risk group. The heterogeneity of tumor-infiltrating immune cells might represent a promising feature for risk prediction in SCLC. CONCLUSION FOXP3 interacts closely with immune biomarkers on tumor-infiltrating cells in TME. This study highlighted the crucial prognostic value and promising clinical applications of FOXP3 in SCLC.
Collapse
Affiliation(s)
- Minlin Jiang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Chunyan Wu
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Liping Zhang
- Department of Pathology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Chenglong Sun
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Hao Wang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Yi Xu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Hui Sun
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Jun Zhu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Wencheng Zhao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Qiyu Fang
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Jia Yu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Peixin Chen
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Shengyu Wu
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Zixuan Zheng
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China.,Tongji University, No 1239 Siping Road, Shanghai 200433, China
| | - Yayi He
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| | - Caicun Zhou
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University Medical School Cancer Institute, Tongji University School of Medicine, No 507 Zhengmin Road, Shanghai 200433, China
| |
Collapse
|
179
|
Mehdizadeh S, Bayatipoor H, Pashangzadeh S, Jafarpour R, Shojaei Z, Motallebnezhad M. Immune checkpoints and cancer development: Therapeutic implications and future directions. Pathol Res Pract 2021; 223:153485. [PMID: 34022684 DOI: 10.1016/j.prp.2021.153485] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/10/2021] [Accepted: 05/13/2021] [Indexed: 02/08/2023]
Abstract
Over the past few decades, different inhibitory receptors have been identified, which have played prominent roles in reducing anti-tumor immune responses. The role of immune checkpoint inhibitors in cancer was revealed by critical blockade of the cytotoxic T lymphocyte-associated antigen-4 (CTLA-4) and programmed cell death protein-1 (PD-1) checkpoints. Immune checkpoint inhibitors, including anti-PD-1 (nivolumab and pembrolizumab), anti-PD-L1 (Atezolizumab, avelumab, and duravulumab), and anti-CTLA-4 (ipilimumab, tremelimumab), are currently FDA-approved treatment options for a broad range of cancer types. However, regarding immunotherapy advances in recent years, most studies have been focused on finding the antibodies against other inhibitory immune checkpoints in the tumor microenvironment such as lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin, and mucin domain 3 (TIM-3), B7-homolog 3 (B7-H3), V-domain immunoglobulin-containing suppressor of T-cell activation (VISTA), diacylglycerol kinase-α (DGK-α), T cell immunoglobulin and ITIM domain (TIGIT), and B and T lymphocyte attenuator (BTLA). This immune checkpoint exerts differential inhibitory impacts on various types of lymphocytes. The suppression of immune responses demonstrates a surprising synergy with PD-1. Therefore, most antibodies against these immune checkpoints are undertaking clinical trials for cancer immunotherapy of advanced solid tumors and hematologic malignancies. In this review, we will summarize recent findings of immune checkpoint and the role of monoclonal antibodies in cancer immunotherapy targeting these receptors.
Collapse
Affiliation(s)
- Saber Mehdizadeh
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Hashem Bayatipoor
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Salar Pashangzadeh
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Roghayeh Jafarpour
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Zeinab Shojaei
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Motallebnezhad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran; Immunology Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
180
|
V-Domain Ig Suppressor of T Cell Activation (VISTA) Expression Is an Independent Prognostic Factor in Multiple Myeloma. Cancers (Basel) 2021; 13:cancers13092219. [PMID: 34066382 PMCID: PMC8124446 DOI: 10.3390/cancers13092219] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 12/27/2022] Open
Abstract
Simple Summary Multiple myeloma (MM) is characterized by loss of anti-tumor T-cell immunity. The precise mechanisms by which malignant plasma cells escape T-cell immunity are unknown, although upregulation of checkpoint molecules is seen in progressive disease. The aim of our study was to investigate mechanisms of escape from T-cell immunity. We observed that the expression of V-domain Ig suppressor of T cell activation (VISTA) in the tumor microenvironment is an independent prognostic factor for survival in MM and its major cellular source is tumor infiltrating CD11B+ cells. The combination of high VISTA expression in the tumor combined with low infiltration of CD8+ cells compared to the surrounding stromal tissue is significantly associated with poor survival. These finding have identified VISTA as an interesting target for inhibition to circumvent escape of T-cell immunity. Abstract Multiple myeloma (MM) is characterized by loss of anti-tumor T cell immunity. Despite moderate success of treatment with anti-PD1 antibodies, effective treatment is still challenged by poor T cell-mediated control of MM. To better enable identification of shortcomings in T-cell immunity that relate to overall survival (OS), we interrogated transcriptomic data of bone marrow samples from eight clinical trials (n = 1654) and one trial-independent patient cohort (n = 718) for multivariate analysis. Gene expression of V-domain Ig suppressor of T cell activation (VISTA) was observed to correlate to OS [hazard ratio (HR): 0.72; 95% CI: 0.61–0.83; p = 0.005]. Upon imaging the immune contexture of MM bone marrow tissues (n = 22) via multiplex in situ stainings, we demonstrated that VISTA was expressed predominantly by CD11b+ myeloid cells. The combination of abundance of VISTA+, CD11b+ cells in the tumor but not stromal tissue together with low presence of CD8+ T cells in the same tissue compartment, termed a high VISTA-associated T cell exclusion score, was significantly associated with short OS [HR: 16.6; 95% CI: 4.54–62.50; p < 0.0001]. Taken together, the prognostic value of a combined score of VISTA+, CD11b+ and CD8+ cells in the tumor compartment could potentially be utilized to guide stratification of MM patients for immune therapies.
Collapse
|
181
|
Lee JB, Ha SJ, Kim HR. Clinical Insights Into Novel Immune Checkpoint Inhibitors. Front Pharmacol 2021; 12:681320. [PMID: 34025438 PMCID: PMC8139127 DOI: 10.3389/fphar.2021.681320] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Accepted: 04/22/2021] [Indexed: 12/17/2022] Open
Abstract
The success of immune checkpoint inhibitors (ICIs), notably anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4) as well as inhibitors of CTLA-4, programmed death 1 (PD-1), and programmed death ligand-1 (PD-L1), has revolutionized treatment options for solid tumors. However, the lack of response to treatment, in terms of de novo or acquired resistance, and immune related adverse events (IRAE) remain as hurdles. One mechanisms to overcome the limitations of ICIs is to target other immune checkpoints associated with tumor microenvironment. Immune checkpoints such as lymphocyte activation gene-3 (LAG-3), T cell immunoglobulin and ITIM domain (TIGIT), T cell immunoglobulin and mucin-domain containing-3 (TIM-3), V-domain immunoglobulin suppressor of T cell activation (VISTA), B7 homolog 3 protein (B7-H3), inducible T cell costimulatory (ICOS), and B and T lymphocyte attenuator (BTLA) are feasible and promising options for treating solid tumors, and clinical trials are currently under active investigation. This review aims to summarize the clinical aspects of the immune checkpoints and introduce novel agents targeting these checkpoints.
Collapse
Affiliation(s)
- Jii Bum Lee
- Division of Hemato-oncology, Wonju Severance Christian Hospital, Yonsei University Wonju College of Medicine, Wonju, South Korea.,Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang-Jun Ha
- Department of Biochemistry, College of Life Science & Biotechnology, Yonsei University, Seoul, South Korea
| | - Hye Ryun Kim
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
182
|
Wei G, Zhang H, Zhao H, Wang J, Wu N, Li L, Wu J, Zhang D. Emerging immune checkpoints in the tumor microenvironment: Implications for cancer immunotherapy. Cancer Lett 2021; 511:68-76. [PMID: 33957184 DOI: 10.1016/j.canlet.2021.04.021] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 12/14/2022]
Abstract
Immune checkpoints within the tumor microenvironment (TME) play important roles in modulating host antitumor immunity. Checkpoint-based immunotherapies (e.g. immune checkpoint inhibitors) have revolutionized cancer therapy. However, there are still many drawbacks with current checkpoint immunotherapies in clinical practice, such as unresponsiveness, resistance, tumor hyperprogression, autoimmune-related adverse events, and limited efficacy with some solid malignances. These drawbacks highlight the need to further investigate the mechanisms underlying the therapeutic effects, as well as the need to identify new targets for cancer immunotherapy. With the discovery of emerging immune checkpoints in the TME, the development of strategies targeting the pivotal immunomodulators for cancer treatment has been significantly advanced in the past decade. In this review, we summarize and classify the novel emerging immune checkpoints beyond the extensively studied ones (e.g. PD-1, PD-L1, CTLA-4, LAG-3 and TIM-3) in the TME, and provide an update on the clinical trials targeting these key immune molecules.
Collapse
Affiliation(s)
- Gaigai Wei
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Huiling Zhang
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Haiping Zhao
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jing Wang
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Nana Wu
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Leying Li
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Jiaying Wu
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Duanwu Zhang
- Children's Hospital of Fudan University, National Children's Medical Center, And Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
183
|
Abstract
Renal cell carcinoma (RCC) is increasing in incidence and one third of newly diagnosed cases already are metastatic. The metastatic spread of solid tumors renders RCC incurable by surgical resection and consequently more difficult to treat. New molecular-targeted therapies have played a pivotal role in RCC treatment. Unfortunately, tumors frequently develop resistance to these targeted therapies by activating bypass pathways in which alternative signaling or biochemical pathways are activated in response to targeted inhibition of a signaling pathway, allowing cancer cells to continue to survive. Although the advent of immunotherapy with checkpoint inhibitors has led to significant changes in the treatment landscape for advanced RCC, many issues remain to be resolved. For these reasons, there is an urgent need to develop novel therapies and new treatment paradigms for patients with RCC. Much research has been performed thus far in identifying novel targets and treatment strategies in RCC and many of these currently are under investigation and/or in clinical trials. In this article, we discuss therapeutic options in the management of RCC with a focus on the new therapeutic approaches currently investigated in research and for use in the clinic. We divide these potential novel therapies into five groups: nonbiologics, small-molecule drugs, biologics, immunomodulatory therapies, and peptide drugs. We also present some therapeutics and treatment paradigms.
Collapse
Affiliation(s)
- David C Yang
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA
| | - Ching-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, University of California Davis, Davis, CA.
| |
Collapse
|
184
|
Yum JEI, Hong YK. Terminating Cancer by Blocking VISTA as a Novel Immunotherapy: Hasta la vista, baby. Front Oncol 2021; 11:658488. [PMID: 33937071 PMCID: PMC8085549 DOI: 10.3389/fonc.2021.658488] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 03/30/2021] [Indexed: 01/06/2023] Open
Abstract
VISTA is an up-and-coming immune checkpoint molecule that can become the target of new cancer immunotherapy treatments. Immune cells in the tumor microenvironment can largely influence the progression of cancer through inhibitory and stimulatory pathways. Indeed, VISTA is expressed on many immune cells, including T cells, myeloid-derived suppressor cells, tumor-associated macrophages, and dendritic cells. VISTA has predominantly been shown to act in an immune-suppressing manner that enables cancer progression. This review will delve into results from preclinical murine studies of anti-VISTA monoclonal antibody treatments, bring together recent studies that detect VISTA expression on immune cells from patient tumors of various cancers, and discuss ongoing clinical trials involving VISTA.
Collapse
Affiliation(s)
- Ji-Eun Irene Yum
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| | - Young-Kwon Hong
- Department of Surgery, Keck School of Medicine, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
185
|
Seo WI, Lee CH, Jung SJ, Lee DS, Park HY, Jeong DH, Kim W, Chung JI, Choi I. Expression of VISTA on tumor-infiltrating immune cells correlated with short intravesical recurrence in non-muscle-invasive bladder cancer. Cancer Immunol Immunother 2021; 70:3113-3122. [PMID: 33770210 DOI: 10.1007/s00262-021-02906-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Accepted: 02/28/2021] [Indexed: 12/24/2022]
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA) is an immune checkpoint molecule expressed in hematopoietic cells, granulocytes, macrophages, and monocytes. However, few studies to date have investigated VISTA expression, especially its clinical utility, in bladder cancer. The present retrospective study aimed to examine VISTA, programmed death ligand-1 (PD-L1), and CD45 expression by immunohistochemical and immunofluorescence staining of archived pathological tissue samples from 159 patients with primary bladder cancer. The correlation between VISTA expression in immune cells (ICs) and clinicopathologic variables including PD-L1 expression in ICs was examined. Briefly, the rates of VISTA-positive ICs and VISTA-positive tumor cells were 67.9% (108/159) and 30.8% (49/159), respectively. The VISTA expression in ICs of patients with bladder cancer, including those with non-muscle-invasive bladder cancer (NMIBC), was positively correlated with tumor stage, grade, size, and multiplicity. The VISTA expression in ICs was stronger in bladder cancer cases with PD-L1-positive ICs than in those with PD-L1-negative ICs (p < 0.001). The mean intravesical recurrence-free survival was shorter in NMIBC cases with VISTA-positive ICs than in those with VISTA-negative ICs (34.0 vs 39.9 months, p = 0.03, log-rank test). In this first study to investigate VISTA expression in bladder cancer, these results implicate VISTA as a potential immunotherapeutic target and immunologic biomarker in bladder cancer.
Collapse
Affiliation(s)
- Won Ik Seo
- Department of Urology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Chan Ho Lee
- Department of Urology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Soo Jin Jung
- Department of Pathology, Busan Paik Hospital, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan, 47392, Republic of Korea.
| | - Dae Sim Lee
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.,Paik Institute of Clinical Research, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Ha Young Park
- Department of Pathology, Busan Paik Hospital, Inje University College of Medicine, 75 Bokji-ro, Busanjin-gu, Busan, 47392, Republic of Korea
| | - Dae Hoon Jeong
- Department of Obstetrics and Gynecology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea.,Paik Institute of Clinical Research, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Wansuk Kim
- Department of Urology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Jae Il Chung
- Department of Urology, Busan Paik Hospital, Inje University College of Medicine, Busan, Republic of Korea
| | - Inhak Choi
- Department of Microbiology and Immunology, Innovative Therapeutics Research Institute, Inje University College of Medicine, Busan, Korea
| |
Collapse
|
186
|
Xie X, Chen C, Chen W, Jiang J, Wang L, Li T, Sun H, Liu J. Structural Basis of VSIG3: The Ligand for VISTA. Front Immunol 2021; 12:625808. [PMID: 33841409 PMCID: PMC8027081 DOI: 10.3389/fimmu.2021.625808] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Accepted: 03/11/2021] [Indexed: 12/30/2022] Open
Abstract
B7 family members and their receptors play key roles in regulating T cell responses, and constitute very attractive targets for developing immunotherapeutic drugs. V-Set and Immunoglobulin domain containing 3 (VSIG3), a ligand for the novel B7 family immune checkpoint V-domain immunoglobulin suppressor of T cell activation (VISTA), can significantly inhibit T cell functions. Inhibitors targeting the VISTA/VSIG3 pathway are of great significance in tumor immunology. Here, we show the crystal structure of the extracellular domain (ECD) of the human VSIG3 protein at 2.64 angstrom resolution, and we produce recombinant human VSIG-3 ECD in both CHO cells and E. coli. Furthermore, we demonstrated the interaction of VISTA and VSIG3 by coimmunoprecipitation (Co-IP). Based on protein-protein docking for VISTA and VSIG3, we report a small molecule inhibitor of VSIG3 K284-3046 and evaluate its biological activities in vitro. This study was the first to reveal the crystal structure of VSIG3, and provides the structural basis for designing antibodies or compounds for the unique VSIG3/VISTA coinhibitory pathway in the treatment of cancers, autoimmune diseases and may be beneficial of designing vaccines.
Collapse
Affiliation(s)
- Xiaoxue Xie
- New Drug Screening Center, China Pharmaceutical University, Nanjing, China
| | - Caiping Chen
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Wenting Chen
- New Drug Screening Center, China Pharmaceutical University, Nanjing, China
| | - Jingwei Jiang
- New Drug Screening Center, China Pharmaceutical University, Nanjing, China
| | - Lanlan Wang
- New Drug Screening Center, China Pharmaceutical University, Nanjing, China
| | - Tingting Li
- New Drug Screening Center, China Pharmaceutical University, Nanjing, China
| | - Hongbin Sun
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- New Drug Screening Center, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
187
|
Rohatgi A, Kirkwood JM. Beyond PD-1: The Next Frontier for Immunotherapy in Melanoma. Front Oncol 2021; 11:640314. [PMID: 33732652 PMCID: PMC7958874 DOI: 10.3389/fonc.2021.640314] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 01/07/2021] [Indexed: 12/14/2022] Open
Abstract
The advent of first and second-generation immune checkpoint blockade (ICI) has resulted in improved survival of patients with metastatic melanoma over the past decade. However, the majority of patients ultimately progress despite these treatments, which has served as an impetus to consider a range of subsequent therapies. Many of the next generation of immunotherapeutic agents focus on modifying the immune system to overcome resistance to checkpoint blockade. ICI resistance can be understood as primary, or acquired-where the latter is the most common scenario. While there are several postulated mechanisms by which resistance, particularly acquired resistance, occurs, the predominant escape mechanisms include T cell exhaustion, upregulation of alternative inhibitory checkpoint receptors, and alteration of the tumor microenvironment (TME) into a more suppressive, anti-inflammatory state. Therapeutic agents in development are designed to work by combating one or more of these resistance mechanisms. These strategies face the added challenge of minimizing immune-related toxicities, while improving antitumor efficacy. This review focuses upon the following categories of novel therapeutics: 1) alternative inhibitory receptor pathways; 2) damage- or pathogen-associated molecular patterns (DAMPs/PAMPs); and 3) immune cell signaling mediators. We present the current state of these therapies, including preclinical and clinical data available for these targets under development.
Collapse
Affiliation(s)
| | - John M. Kirkwood
- Hillman Cancer Center, University of Pittsburgh Medical Center, Pittsburgh, PA, United States
| |
Collapse
|
188
|
Advanced Molecular Characterization Using Digital Spatial Profiling Technology on Immunooncology Targets in Methylated Compared with Unmethylated IDH-Wildtype Glioblastoma. JOURNAL OF ONCOLOGY 2021; 2021:8819702. [PMID: 33995529 PMCID: PMC8096575 DOI: 10.1155/2021/8819702] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/09/2020] [Revised: 01/24/2021] [Accepted: 02/04/2021] [Indexed: 01/05/2023]
Abstract
Introduction Glioblastoma (GBM) is the most common primary adult brain tumour with a median overall survival (OS) of 12-15 months. Molecular characterization of multiple immunooncology targets in GBM may help target novel immunotherapeutic strategies. We used NanoString GeoMx® Digital Spatial Profiling (DSP) to assess multiple immunooncology protein targets in methylated versus unmethylated IDH-wild-type glioblastoma. Methods NanoString GeoMx® DSP technology uses multiple primary antibodies conjugated to indexing DNA oligos with a UV photocleavable linker. Tissue regions of interest (ROIs) are selected with bound fluorescent antibodies; oligos are released via a UV-mediated linker and quantitated. We used DSP multiplex analysis of 31 immunooncology proteins and controls (CD4, CD14, CD68, CD8A, B7-H3, PD-L1, CD19, FOXP3, CD44, STAT3 (phospho Y705), CD45, Pan Cytokeratin, MS4A1/CD20, CD45RO, PD1, CD3, beta-2 microglobulin, VISTA, Bcl2, GZMB, PTEN, beta-catenin, CD56, Ki-67, STAT3, AKT, p-Akt, S6, Histone H3, IgG Rabbit control, and Mouse IgG control) from ROIs in a cohort of 10 IDH-wild-type glioblastomas (5 methylated and 5 unmethylated). An nCounter platform allowed quantitative comparisons of antibodies between ROIs in MGMT methylated and unmethylated tumours. Mean protein expression counts between methylated and unmethylated GBM were compared using technical and biological replicates. Results The analysis showed 10/27 immunooncology target proteins were significantly increased in methylated versus unmethylated IDH-wild-type glioblastoma tumour core (false discovery rate (FDR) <0.1 by Benjamini-Hochberg procedure). Conclusions NanoString GeoMx® DSP was used to analyse multiple immunooncology protein target expression in methylated versus unmethylated IDH-wild-type glioblastoma. In this small study, there was a statistical increase in CD4, CD14, CD68, CD8A, B7-H3, PDL-1, CD19, FOXP3, CD44, and STAT3 protein expression in methylated versus unmethylated GBM tumour core; however, this requires larger cohort validation. Advanced multiplex immunooncological biomarker analysis may be useful in identifying biomarkers for novel immunotherapeutic agents in GBMs.
Collapse
|
189
|
Ait Boujmia OK. V-domain Ig suppressor of T cell activation (VISTA) inhibition is a new approach to cancer therapy: a Bibliometric study. Naunyn Schmiedebergs Arch Pharmacol 2021; 394:1057-1065. [PMID: 33616679 DOI: 10.1007/s00210-021-02068-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 02/15/2021] [Indexed: 12/24/2022]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a transmembrane protein that plays a crucial role in the regulation of antitumor immunity. Therapeutic intervention that inhibits the VISTA pathway constitutes a new approach in the treatment of cancer. The aim of the present study was to provide a bibliometric literature review of VISTA research in the field of cancer. Published articles on the topic were retrieved from the Web of Science Core Collection Database of Hassan II University from the beginning of the database to November 14, 2019. The articles were analyzed and a lot of information was available on the Web of Science, such as the number of citations, the names of the authors, country, publication year, Web of Science categories, and journal. A total of 76 papers (research and review articles) were retrieved from the Web of Science Core Collection Database to introduce VISTA research in cancer topic. All of the articles were published in English during the period between 2011 and 2019; the annual publications number has increased from 1 in 2011 to 22 in 2019. Cancer immunology immunotherapy journal, Cancer immunology research journal, and Cancer research journal, each one has published 3 articles (3.9% of the total publications), the impact factors of the journals ranged from 2.34 to 10.19. The author who has published high number of articles was Noelle RJ with 11 articles; according to the keyword co-occurrence, VISTA was the most frequent keyword with a frequency of 42.1%, followed by immune (36.8%). This is the first work that treats the application of bibliometric methods in VISTA research in the oncology field and represents an important bibliographic source for future studies on the role of VISTA in cancer and immunotherapy of cancer.
Collapse
Affiliation(s)
- Oum Kaltoum Ait Boujmia
- Laboratory of Cellular and Molecular Pathology, Faculty of Medicine and Pharmacy of Casablanca, University Hassan II, Casablanca, Morocco. .,Medical School of Casablanca, 19 Rue Tarik Ibnou Ziad, BP. 9154, Casablanca, Morocco.
| |
Collapse
|
190
|
Rosenbaum SR, Knecht M, Mollaee M, Zhong Z, Erkes DA, McCue PA, Chervoneva I, Berger AC, Lo JA, Fisher DE, Gershenwald JE, Davies MA, Purwin TJ, Aplin AE. FOXD3 Regulates VISTA Expression in Melanoma. Cell Rep 2021; 30:510-524.e6. [PMID: 31940493 PMCID: PMC6995351 DOI: 10.1016/j.celrep.2019.12.036] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2019] [Revised: 11/01/2019] [Accepted: 12/12/2019] [Indexed: 12/22/2022] Open
Abstract
Immune checkpoint inhibitors have improved patient survival in melanoma, but the innate resistance of many patients necessitates the investigation of alternative immune targets. Many immune checkpoint proteins lack proper characterization, including V-domain Ig suppressor of T cell activation (VISTA). VISTA expression on immune cells can suppress T cell activity; however, few studies have investigated its expression and regulation in cancer cells. In this study, we observe that VISTA is expressed in melanoma patient samples and cell lines. Tumor cell-specific expression of VISTA promotes tumor onset in vivo, associated with increased intratumoral T regulatory cells, and enhanced PDL-1 expression on tumor-infiltrating macrophages. VISTA transcript levels are regulated by the stemness factor Forkhead box D3 (FOXD3). BRAF inhibition upregulates FOXD3 and reduces VISTA expression. Overall, this study demonstrates melanoma cell expression of VISTA and its regulation by FOXD3, contributing to the rationale for therapeutic strategies that combine targeted inhibitors with immune checkpoint blockade. VISTA is an understudied immune checkpoint protein. Through the analysis of patient samples and studies in mouse models, Rosenbaum et al. investigate the functional consequences of VISTA expression on melanoma cells. Furthermore, they demonstrate that the BRAF-regulated transcription factor FOXD3 negatively regulates VISTA expression.
Collapse
Affiliation(s)
- Sheera R Rosenbaum
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Meghan Knecht
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Mehri Mollaee
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Zhijiu Zhong
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Dan A Erkes
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Peter A McCue
- Department of Pathology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Inna Chervoneva
- Division of Biostatistics in the Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Adam C Berger
- Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA; Department of Surgery, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Jennifer A Lo
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Michael A Davies
- Department of Melanoma Medical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Timothy J Purwin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - Andrew E Aplin
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA; Sidney Kimmel Cancer Center, Thomas Jefferson University, Philadelphia, PA 19107, USA.
| |
Collapse
|
191
|
ElTanbouly MA, Zhao Y, Schaafsma E, Burns CM, Mabaera R, Cheng C, Noelle RJ. VISTA: A Target to Manage the Innate Cytokine Storm. Front Immunol 2021; 11:595950. [PMID: 33643285 PMCID: PMC7905033 DOI: 10.3389/fimmu.2020.595950] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/11/2020] [Indexed: 12/12/2022] Open
Abstract
In recent years, the success of immunotherapy targeting immunoregulatory receptors (immune checkpoints) in cancer have generated enthusiastic support to target these receptors in a wide range of other immune related diseases. While the overwhelming focus has been on blockade of these inhibitory pathways to augment immunity, agonistic triggering via these receptors offers the promise of dampening pathogenic inflammatory responses. V-domain Ig suppressor of T cell activation (VISTA) has emerged as an immunoregulatory receptor with constitutive expression on both the T cell and myeloid compartments, and whose agonistic targeting has proven a unique avenue relative to other checkpoint pathways to suppress pathologies mediated by the innate arm of the immune system. VISTA agonistic targeting profoundly changes the phenotype of human monocytes towards an anti-inflammatory cell state, as highlighted by striking suppression of the canonical markers CD14 and Fcγr3a (CD16), and the almost complete suppression of both the interferon I (IFN-I) and antigen presentation pathways. The insights from these very recent studies highlight the impact of VISTA agonistic targeting of myeloid cells, and its potential therapeutic implications in the settings of hyperinflammatory responses such as cytokine storms, driven by dysregulated immune responses to viral infections (with a focus on COVID-19) and autoimmune diseases. Collectively, these findings suggest that the VISTA pathway plays a conserved, non-redundant role in myeloid cell function.
Collapse
Affiliation(s)
- Mohamed A. ElTanbouly
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| | - Yanding Zhao
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
| | - Evelien Schaafsma
- Department of Molecular and Systems Biology, Geisel School of Medicine at Dartmouth, Hanover, NH, United States
| | | | - Rodwell Mabaera
- Department of Medicine, Norris Cotton Cancer Center, Lebanon, NH, United States
| | - Chao Cheng
- Department of Medicine, Baylor College of Medicine, Houston, TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
| | - Randolph J. Noelle
- Department of Microbiology and Immunology, Norris Cotton Cancer Center, Geisel School of Medicine at Dartmouth, Lebanon, NH, United States
| |
Collapse
|
192
|
Recent Advancements in the Mechanisms Underlying Resistance to PD-1/PD-L1 Blockade Immunotherapy. Cancers (Basel) 2021; 13:cancers13040663. [PMID: 33562324 PMCID: PMC7915065 DOI: 10.3390/cancers13040663] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Accepted: 02/03/2021] [Indexed: 02/08/2023] Open
Abstract
Simple Summary Immune checkpoint blockade targeting PD-1/PD-L1 has a promising therapeutic efficacy in different tumors, but a significant percentage of patients cannot benefit from this therapy due to primary and acquired resistance during treatment. This review summarizes the recent findings of PD-L1 role in resistance to therapies through the PD-1/PD-L1 pathway and other correlating signaling pathways. A special focus will be given to the key mechanisms underlying resistance to the PD-1/PD-L1 blockade in cancer immunotherapy. Furthermore, we also discuss the promising combination of therapeutic strategies for patients resistant to the PD-1/PD-L1 blockade in order to enhance the efficacy of immune checkpoint inhibitors. Abstract Release of immunoreactive negative regulatory factors such as immune checkpoint limits antitumor responses. PD-L1 as a significant immunosuppressive factor has been involved in resistance to therapies such as chemotherapy and target therapy in various cancers. Via interacting with PD-1, PD-L1 can regulate other factors or lead to immune evasion of cancer cells. Besides, immune checkpoint blockade targeting PD-1/PD-L1 has promising therapeutic efficacy in the different tumors, but a significant percentage of patients cannot benefit from this therapy due to primary and acquired resistance during treatment. In this review, we described the utility of PD-L1 expression levels for predicting poor prognosis in some tumors and present evidence for a role of PD-L1 in resistance to therapies through PD-1/PD-L1 pathway and other correlating signaling pathways. Afterwards, we elaborate the key mechanisms underlying resistance to PD-1/PD-L1 blockade in cancer immunotherapy. Furthermore, promising combination of therapeutic strategies for patients resistant to PD-1/PD-L1 blockade therapy or other therapies associated with PD-L1 expression was also summarized.
Collapse
|
193
|
Gao X, Sui H, Zhao S, Gao X, Su Y, Qu P. Immunotherapy Targeting Myeloid-Derived Suppressor Cells (MDSCs) in Tumor Microenvironment. Front Immunol 2021; 11:585214. [PMID: 33613512 PMCID: PMC7889583 DOI: 10.3389/fimmu.2020.585214] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2020] [Accepted: 12/17/2020] [Indexed: 12/11/2022] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) are a heterogeneous population of immature myeloid cells that accumulate in tumor-bearing hosts to reduce T cells activity and promote tumor immune escape in the tumor microenvironment (TME). The immune system in the TME can be stimulated to elicit an anti-tumor immune response through immunotherapy. The main theory of immunotherapy resides on the plasticity of the immune system and its capacity to be re-educated into a potent anti-tumor response. Thus, MDSCs within the TME became one of the major targets to improve the efficacy of tumor immunotherapy, and therapeutic strategies for tumor MDSCs were developed in the last few years. In the article, we analyzed the function of tumor MDSCs and the regulatory mechanisms of agents targeting MDSCs in tumor immunotherapy, and reviewed their therapeutic effects in MDSCs within the TME. Those data focused on discussing how to promote the differentiation and maturation of MDSCs, reduce the accumulation and expansion of MDSCs, and inhibit the function, migration and recruitment of MDSCs, further preventing the growth, invasion and metastasis of tumor. Those investigations may provide new directions for cancer therapy.
Collapse
Affiliation(s)
- Xidan Gao
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Hongshu Sui
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Shang Zhao
- Department of Pathophysiology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Xingmei Gao
- Department of Neurology, People's Hospital of Binzhou, Binzhou, China
| | - Yanping Su
- Department of Histology and Embryology, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, China
| | - Peng Qu
- Center for Cancer Research, National Cancer Institute, Frederick, MD, United States
| |
Collapse
|
194
|
Hou Z, Pan Y, Fei Q, Lin Y, Zhou Y, Liu Y, Guan H, Yu X, Lin X, Lu F, Huang H. Prognostic significance and therapeutic potential of the immune checkpoint VISTA in pancreatic cancer. J Cancer Res Clin Oncol 2021; 147:517-531. [PMID: 33237432 PMCID: PMC7817580 DOI: 10.1007/s00432-020-03463-9] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 11/09/2020] [Indexed: 12/11/2022]
Abstract
OBJECTIVE V-domain Ig suppressor of T cell activation (VISTA) is a novel immune checkpoint protein that belongs to the B7 family. The aim of this study was to investigate the prognostic significance and therapeutic potential of VISTA in patients with pancreatic cancer. METHODS Using immunohistochemistry (IHC), we examined the expression of VISTA and demonstrated the associations between the VISTA and overall survival in 223 PDAC patients from 2 different unrelated retrospective cohorts. The multiplex immunofluorescence was performed to illuminate the relationship between VISTA expression and tumor-infiltrating immune cell subclusters of PDAC. We also verified the findings in The Cancer Genome Atlas (TCGA) dataset. The anti-tumor effect of anti-VISTA therapy was studied by the mouse model with liver metastases of PDAC. RESULTS The VISTA protein was highly expressed in 25.6% of tumor cells (TCs), 38.1% of immune cells, and 26.0% of endothelial cells in 223 PDAC tumor tissues. VISTA expression in TCs was significantly associated with prolonged overall survival. Multiplex immunofluorescence analysis revealed that VISTA level was positively correlated with CD68+ macrophages, CD3+ T cells, and CD19+ B cells in PDAC. However, a higher expression level of VISTA was detected in tumor-infiltrating CD68+ macrophages than in CD3+ T and CD19+ B cells. Furthermore, anti-VISTA antibody treatment significantly reduced the number of metastatic nodules in livers of mouse models of PDAC with liver metastases. CONCLUSION VISTA expressed in TCs is associated with a favorable prognosis in PDAC. Moreover, immunotherapy with anti-VISTA antibodies may potentially be an effective treatment strategy against PDAC.
Collapse
Affiliation(s)
- Zelin Hou
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Yu Pan
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Qinglin Fei
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
- The Cancer Center, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Yali Lin
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Yuanyuan Zhou
- Department of Geriatrics, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Ying Liu
- Department of Endocrinology, Quanzhou Hospital of Traditional Chinese Medicine, Quanzhou, 362000 China
| | - Hongdan Guan
- Department of Radiation Oncology, Fujian Medical University Union Hospital, Fuzhou, 350001 China
| | - Xunbin Yu
- Department of Pathology, Fujian Provincialial Hospital, Fuzhou, 350001 China
| | - Xianchao Lin
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Fengchun Lu
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| | - Heguang Huang
- Department of General Surgery, Fujian Medical University Union Hospital, No. 29 Xinquan Road, Fuzhou, 350001 People’s Republic of China
| |
Collapse
|
195
|
VISTA: A Mediator of Quiescence and a Promising Target in Cancer Immunotherapy. Trends Immunol 2021; 42:209-227. [PMID: 33495077 PMCID: PMC8088836 DOI: 10.1016/j.it.2020.12.008] [Citation(s) in RCA: 135] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 12/13/2020] [Accepted: 12/24/2020] [Indexed: 12/18/2022]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a B7 family member that maintains T cell and myeloid quiescence and is a promising target for combination cancer immunotherapy. During inflammatory challenges, VISTA activity reprograms macrophages towards reduced production of proinflammatory cytokines and increased production of interleukin (IL)-10 and other anti-inflammatory mediators. The interaction of VISTA with its ligands is regulated by pH, and the acidic pH ~6.0 in the tumor microenvironment (TME) facilitates VISTA binding to P-selectin glycoprotein ligand 1 (PSGL-1). Targeting intratumoral pH might be a way to reduce the immunoinhibitory activity of the VISTA pathway and enhance antitumor immune responses. We review differences among VISTA therapeutics under development as candidate immunotherapies, focusing on VISTA binding partners and the unique structural features of this interaction.
Collapse
|
196
|
Kulasinghe A, Taheri T, O'Byrne K, Hughes BGM, Kenny L, Punyadeera C. Highly Multiplexed Digital Spatial Profiling of the Tumor Microenvironment of Head and Neck Squamous Cell Carcinoma Patients. Front Oncol 2021; 10:607349. [PMID: 33542903 PMCID: PMC7851078 DOI: 10.3389/fonc.2020.607349] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2020] [Accepted: 12/03/2020] [Indexed: 01/10/2023] Open
Abstract
Background Immune checkpoint inhibitors (ICI) have shown durable and long-term benefits in a subset of head and neck squamous cell carcinoma (HNSCC) patients. To identify patient-responders from non-responders, biomarkers are needed which are predictive of outcome to ICI therapy. Cues in the tumor microenvironment (TME) have been informative in understanding the tumor-immune contexture. Methods In this preliminary study, the NanoString GeoMx™ Digital Spatial Profiling (DSP) technology was used to determine the immune marker and compartment specific measurements in a cohort of HNSCC tumors from patients receiving ICI therapy. Results Our data revealed that markers involved with immune cell infiltration (CD8 T-cells) were not predictive of outcome to ICI therapy. Rather, a number of immune cell types and protein markers (CD4, CD68, CD45, CD44, CD66b) were found to correlate with progressive disease. Cross platform comparison with the Opal Vectra (Perkin Elmer) for a number of markers across similar regions of interest demonstrated concordance for pan-cytokeratin, CD8, and PD-L1. Conclusion This study, to our knowledge, represents the first digital spatial analysis of HNSCC tumors. A larger cohort of HNSCC will be required to orthogonally validate the findings.
Collapse
Affiliation(s)
- Arutha Kulasinghe
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| | - Touraj Taheri
- Department of Pathology, Royal Brisbane and Women's Hospital, Brisbane, QLD, Australia.,Royal Brisbane and Women's Hospital, Herston, QLD, Australia
| | - Ken O'Byrne
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia.,Princess Alexandra Hospital, Woolloongabba, QLD, Australia
| | - Brett G M Hughes
- Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Liz Kenny
- Royal Brisbane and Women's Hospital, Herston, QLD, Australia.,School of Medicine, University of Queensland, Brisbane, QLD, Australia
| | - Chamindie Punyadeera
- The School of Biomedical Sciences, Institute of Health and Biomedical Innovation, Queensland University of Technology, Kelvin Grove, QLD, Australia.,Translational Research Institute, Brisbane, QLD, Australia
| |
Collapse
|
197
|
Cao X, Ren X, Zhou Y, Mao F, Lin Y, Wu H, Sun Q. VISTA Expression on Immune Cells Correlates With Favorable Prognosis in Patients With Triple-Negative Breast Cancer. Front Oncol 2021; 10:583966. [PMID: 33505908 PMCID: PMC7829913 DOI: 10.3389/fonc.2020.583966] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Accepted: 11/26/2020] [Indexed: 01/26/2023] Open
Abstract
V-domain Ig suppressor of T-cell activation (VISTA), a newly discovered negative immune checkpoint, is thought to be related to immunotherapy resistance and may become a new immune therapeutic target. Here, we evaluated the expression of VISTA in a cohort containing 254 patients with untreated triple-negative breast cancer. The relevance of VISTA expression, clinicopathologic parameters, expression of other immune markers, and prognosis were investigated in the whole cohort. Genomic analysis of 139 triple-negative breast cancer (TNBC) patients from the cancer genome atlas (TCGA) was also performed. VISTA was expressed in the immune cells (ICs) and in the tumor cells (TCs) in 87.8% (223/254) and 18.5% (47/254) of the cohort, respectively. VISTA-positive ICs were associated with no lymph node metastasis (p < 0.001), American Joint Committee on Cancer (AJCC) stage I and II (p = 0.001) and basal-like subtype (p < 0.001). VISTA expression in ICs positively correlated with some tumor-infiltrating lymphocytes (TILs) types, particularly with the CD4+TILs, which was consistent with mRNA level analysis from the TCGA database. Survival analysis showed that patients with VISTA-positive ICs had prolonged relapse-free and overall survival compared with the negative ones, especially among T1-2N0 stage patients. Multivariate analysis showed that it independently predicted the prognosis. These data confirmed the regulatory role of VISTA in anti-tumor immunity, changed our perception of VISTA as a negative immune checkpoint, and suggested VISTA as a potential therapeutic target for TNBC.
Collapse
Affiliation(s)
- Xi Cao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of MedicalSciences, Beijing, China
| | - Xinyu Ren
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yidong Zhou
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of MedicalSciences, Beijing, China
| | - Feng Mao
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of MedicalSciences, Beijing, China
| | - Yan Lin
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of MedicalSciences, Beijing, China
| | - Huanwen Wu
- Department of Pathology, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Qiang Sun
- Department of Breast Surgery, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of MedicalSciences, Beijing, China
| |
Collapse
|
198
|
The Functional Crosstalk between Myeloid-Derived Suppressor Cells and Regulatory T Cells within the Immunosuppressive Tumor Microenvironment. Cancers (Basel) 2021; 13:cancers13020210. [PMID: 33430105 PMCID: PMC7827203 DOI: 10.3390/cancers13020210] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/1970] [Revised: 12/13/2020] [Accepted: 01/06/2021] [Indexed: 12/14/2022] Open
Abstract
Simple Summary Immunotherapy improved the therapeutic landscape for patients with advanced cancer diseases. However, many patients do not benefit from immunotherapy. The bidirectional crosstalk between myeloid-derived suppressor cells (MDSC) and regulatory T cells (Treg) contributes to immune evasion, limiting the success of immunotherapy by checkpoint inhibitors. This review aims to outline the current knowledge of the role and the immunosuppressive properties of MDSC and Treg within the tumor microenvironment (TME). Furthermore, we will discuss the importance of the functional crosstalk between MDSC and Treg for immunosuppression, issuing particularly the role of cell adhesion molecules. Lastly, we will depict the impact of this interaction for cancer research and discuss several strategies aimed to target these pathways for tumor therapy. Abstract Immune checkpoint inhibitors (ICI) have led to profound and durable tumor regression in some patients with metastatic cancer diseases. However, many patients still do not derive benefit from immunotherapy. Here, the accumulation of immunosuppressive cell populations within the tumor microenvironment (TME), such as myeloid-derived suppressor cells (MDSC), tumor-associated macrophages (TAM), and regulatory T cells (Treg), contributes to the development of immune resistance. MDSC and Treg expand systematically in tumor patients and inhibit T cell activation and T effector cell function. Numerous studies have shown that the immunosuppressive mechanisms exerted by those inhibitory cell populations comprise soluble immunomodulatory mediators and receptor interactions. The latter are also required for the crosstalk of MDSC and Treg, raising questions about the relevance of cell–cell contacts for the establishment of their inhibitory properties. This review aims to outline the current knowledge on the crosstalk between these two cell populations, issuing particularly the potential role of cell adhesion molecules. In this regard, we further discuss the relevance of β2 integrins, which are essential for the differentiation and function of leukocytes as well as for MDSC–Treg interaction. Lastly, we aim to describe the impact of such bidirectional crosstalk for basic and applied cancer research and discuss how the targeting of these pathways might pave the way for future approaches in immunotherapy.
Collapse
|
199
|
Drakes ML, Stiff PJ. Ovarian Cancer: Therapeutic Strategies to Overcome Immune Suppression. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1330:33-54. [PMID: 34339029 DOI: 10.1007/978-3-030-73359-9_3] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Ovarian cancer generally escapes diagnosis until the advanced stages. High-grade serous ovarian cancer (HGSOC) is the most frequently occurring form of this malaise and is a disease which has the highest mortality rate of gynecologic cancers. Over recent years it has been revealed that the course of such cancers can be significantly influenced by the nature of immune cells in tumors at the time of diagnosis and by immune cells induced by therapy. Numerous investigators have since focused on disease biology to identify biomarkers or therapeutic targets. Yet, while over the past decade there have been significant improvements in state-of-the-art surgery for ovarian cancer as frontline therapy, there have been limited advancements in the development of novel curative or management drugs for this disease. This chapter discusses the major elements of immune suppression in HGSOC from a biological viewpoint, mechanisms of overcoming resistance to therapies, and recent therapy aimed at improving patient care and survival.
Collapse
Affiliation(s)
- Maureen L Drakes
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA.
| | - Patrick J Stiff
- Department of Medicine, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, IL, USA
| |
Collapse
|
200
|
Wuerdemann N, Pütz K, Eckel H, Jain R, Wittekindt C, Huebbers CU, Sharma SJ, Langer C, Gattenlöhner S, Büttner R, Speel EJ, Suchan M, Wagner S, Quaas A, Klussmann JP. LAG-3, TIM-3 and VISTA Expression on Tumor-Infiltrating Lymphocytes in Oropharyngeal Squamous Cell Carcinoma-Potential Biomarkers for Targeted Therapy Concepts. Int J Mol Sci 2020; 22:E379. [PMID: 33396515 PMCID: PMC7796181 DOI: 10.3390/ijms22010379] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 12/24/2020] [Accepted: 12/25/2020] [Indexed: 12/12/2022] Open
Abstract
Tumor growth and survival requires a particularly effective immunosuppressant tumor microenvironment (TME) to escape destruction by the immune system. While immunosuppressive checkpoint markers like programmed cell death 1 ligand (PD-L1) are already being targeted in clinical practice, lymphocyte-activation-protein 3 (LAG-3), T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) and V-domain Ig suppressor of T cell activation (VISTA) inhibitors are currently under investigation in clinical trials. Reliable findings on the expression status of those immune checkpoint inhibitors on tumor-infiltrating lymphocytes (TILs) in the TME of oropharyngeal squamous cell carcinoma (OPSCC) are lacking. This work aims to describe the expression of LAG-3, TIM-3, and VISTA expression in the TME of OPSCC. We created a tissue microarray of paraffin-embedded tumor tissue of 241 OPSCC. Expression of the immune checkpoint protein LAG-3, TIM-3, and VISTA in OPSCC was evaluated using immunohistochemistry and results were correlated with CD8+ T-cell inflammation and human papillomavirus (HPV)-status. 73 OPSCC stained positive for LAG-3 (31%; HPV+:44%; HPV-:26%, p = 0.006), 122 OPSCC stained positive for TIM-3 (51%; HPV+:70%; HPV-:44%, p < 0.001) and 168 OPSCC (70%; HPV+:75%; HPV-:68%, p = 0.313) for VISTA. CD8+ T-cells were significantly associated with LAG-3, TIM-3 and VISTA expression (p < 0.001, p < 0.001, p = 0.007). Immune checkpoint therapy targeting LAG-3, TIM-3, and/or VISTA could be a promising treatment strategy especially in HPV-related OPSCC. Future clinical trials investigating the efficacy of a checkpoint blockade in consideration of LAG-3, TIM-3, and VISTA expression are required.
Collapse
Affiliation(s)
- Nora Wuerdemann
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, 35392 Giessen, Germany; (C.W.); (S.J.S.); (C.L.); (S.W.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| | - Katharina Pütz
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (K.P.); (R.B.); (A.Q.)
| | - Hans Eckel
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| | - Rishabh Jain
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
| | - Claus Wittekindt
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, 35392 Giessen, Germany; (C.W.); (S.J.S.); (C.L.); (S.W.)
| | - Christian U. Huebbers
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
- Jean-Uhrmacher-Institute for Otorhinolaryngological Research, University of Cologne, Geibelstrasse 29–31, 50931 Cologne, Germany
| | - Shachi J. Sharma
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, 35392 Giessen, Germany; (C.W.); (S.J.S.); (C.L.); (S.W.)
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| | - Christine Langer
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, 35392 Giessen, Germany; (C.W.); (S.J.S.); (C.L.); (S.W.)
| | - Stefan Gattenlöhner
- Institute of Pathology, University of Giessen, Langhansstrasse 10, 35392 Giessen, Germany;
| | - Reinhard Büttner
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (K.P.); (R.B.); (A.Q.)
| | - Ernst-Jan Speel
- Department of Pathology, GROW-School for Oncology and Developmental Biology, Maastricht University Medical Center, Universiteitssingel 40, 6229 ER Maastricht, The Netherlands;
| | - Malte Suchan
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| | - Steffen Wagner
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Giessen, Klinikstrasse 33, 35392 Giessen, Germany; (C.W.); (S.J.S.); (C.L.); (S.W.)
| | - Alexander Quaas
- Institute of Pathology, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (K.P.); (R.B.); (A.Q.)
| | - Jens P. Klussmann
- Department of Otorhinolaryngology, Head and Neck Surgery, Medical Faculty, University of Cologne, Kerpener Strasse 62, 50937 Cologne, Germany; (H.E.); (R.J.); (C.U.H.); (M.S.); (J.P.K.)
- Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Robert-Koch-Str. 21, 50931 Cologne, Germany
| |
Collapse
|