1
|
Palecki J, Bhasin A, Bernstein A, Mille PJ, Tester WJ, Kelly WK, Zarrabi KK. T-Cell redirecting bispecific antibodies: a review of a novel class of immuno-oncology for advanced prostate cancer. Cancer Biol Ther 2024; 25:2356820. [PMID: 38801069 PMCID: PMC11135853 DOI: 10.1080/15384047.2024.2356820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Accepted: 05/14/2024] [Indexed: 05/29/2024] Open
Abstract
Novel T-cell immunotherapies such as bispecific T-cell engagers (BiTEs) are emerging as promising therapeutic strategies for prostate cancer. BiTEs are engineered bispecific antibodies containing two distinct binding domains that allow for concurrent binding to tumor-associated antigens (TAAs) as well as immune effector cells, thus promoting an immune response against cancer cells. Prostate cancer is rich in tumor associated antigens such as, but not limited to, PSMA, PSCA, hK2, and STEAP1 and there is strong biologic rationale for employment of T-cell redirecting BiTEs within the prostate cancer disease space. Early generation BiTE constructs employed in clinical study have demonstrated meaningful antitumor activity, but challenges related to drug delivery, immunogenicity, and treatment-associated adverse effects limited their success. The ongoing development of novel BiTE constructs continues to address these barriers and to yield promising results in terms of efficacy and safety. This review will highlight some of most recent developments of BiTE therapies for patients with advanced prostate cancer and the evolving data surrounding BiTE constructs undergoing clinical evaluation.
Collapse
Affiliation(s)
- Julia Palecki
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Amman Bhasin
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Andrew Bernstein
- Department of Internal Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Patrick J. Mille
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - William J. Tester
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Wm. Kevin Kelly
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Kevin K. Zarrabi
- Department of Medical Oncology, Sidney Kimmel Cancer Center, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
2
|
Gayen S, Mukherjee S, Dasgupta S, Roy S. Emerging druggable targets for immune checkpoint modulation in cancer immunotherapy: the iceberg lies beneath the surface. Apoptosis 2024; 29:1879-1913. [PMID: 39354213 DOI: 10.1007/s10495-024-02022-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/03/2024]
Abstract
The immune system serves as a fundamental defender against the initiation and progression of cancer. Failure of the immune system augments immunosuppressive action that leading to cancer manifestation. This immunosuppressive effect causes from significant alterations in immune checkpoint expression associated with tumoral progression. The tumor microenvironment promotes immune escape mechanisms that further amplifying immunosuppressive actions. Notably, substantial targeting of immune checkpoints has been pragmatic in the advancement of cancer research. This study highlights a comprehensive review of emerging druggable targets aimed at modulating immune checkpoint co-inhibitory as well as co-stimulatory molecules in response to immune system activation. This modulation has prompted to the development of newer therapeutic insights, eventually inducing immunogenic cell death through immunomodulatory actions. The study emphasizes the role of immune checkpoints in immunogenic regulation of cancer pathogenesis and explores potential therapeutic avenues in cancer immunotherapy.Modulation of Immunosuppressive and Immunostimulatory pathways of immune checkpoints in cancer immunotherapy.
Collapse
Affiliation(s)
- Sakuntala Gayen
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Swarupananda Mukherjee
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India
| | - Sandipan Dasgupta
- Department of Pharmaceutical Technology, Maulana Abul Kalam Azad University of Technology, Kolkata, West Bengal, 741249, India
| | - Souvik Roy
- Department of Pharmaceutical Technology, NSHM Knowledge Campus, Kolkata-Group of Institutions, 124, B. L. Saha Road, Tara Park, Behala, Kolkata, West Bengal, 700053, India.
| |
Collapse
|
3
|
Zhang RJ, Kim TK. VISTA-mediated immune evasion in cancer. Exp Mol Med 2024:10.1038/s12276-024-01336-6. [PMID: 39482534 DOI: 10.1038/s12276-024-01336-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 11/03/2024] Open
Abstract
Over the past decade, V-domain immunoglobulin suppressor of T-cell activation (VISTA) has been established as a negative immune checkpoint molecule. Since the role of VISTA in inhibiting T-cell activation was described, studies have demonstrated other diverse regulatory functions in multiple immune cell populations. Furthermore, its relevance has been identified in human cancers. The role of VISTA in cancer immune evasion has been determined, but its mechanisms in the tumor microenvironment remain to be further elucidated. Understanding its contributions to cancer initiation, progression, and resistance to current treatments will be critical to its utility as a target for novel immunotherapies. Here, we summarize the current understanding of VISTA biology in cancer.
Collapse
Affiliation(s)
- Raymond J Zhang
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
| |
Collapse
|
4
|
Burvenich IJG, Wichmann CW, McDonald AF, Guo N, Rigopoulos A, Huynh N, Vail M, Allen S, O'Keefe GJ, Scott FE, Soikes R, Angelides S, Roemeling RV, Scott AM. Targeting of immune checkpoint regulator V-domain Ig suppressor of T-cell activation (VISTA) with 89Zr-labelled CI-8993. Eur J Nucl Med Mol Imaging 2024; 51:3863-3873. [PMID: 39060374 PMCID: PMC11527895 DOI: 10.1007/s00259-024-06854-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Accepted: 07/17/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND CI-8993 is a fully human IgG1κ monoclonal antibody (mAb) that binds specifically to immune checkpoint molecule VISTA (V-domain Ig suppressor of T-cell activation). Phase I safety has been established in patients with advanced cancer (NCT02671955). To determine the pharmacokinetics and biodistribution of CI-8993 in patients, we aimed to develop 89Zr-labelled CI-8993 and validate PET imaging and quantitation in preclinical models prior to a planned human bioimaging trial. METHODS CI-8993 and human isotype IgG1 control were conjugated to the metal ion chelator p-isothiocyanatobenzyl-desferrioxamine (Df). Quality of conjugates were assessed by SE-HPLC, SDS-PAGE, and FACS. After radiolabelling with zirconium-89 (89Zr), radioconjugates were assessed for radiochemical purity, immunoreactivity, antigen binding affinity, and serum stability in vitro. [89Zr]Zr-Df-CI-8993 alone (1 mg/kg, 4.6 MBq) or in combination with 30 mg/kg unlabelled CI-8993, as well as isotype control [89Zr]Zr-Df-IgG1 (1 mg/kg, 4.6 MBq) were assessed in human VISTA knock-in female (C57BL/6 N-Vsirtm1.1(VSIR)Geno, huVISTA KI) or control C57BL/6 mice bearing syngeneic MB49 bladder cancer tumours; and in BALB/c nu/nu mice bearing pancreatic Capan-2 tumours. RESULTS Stable constructs with an average chelator-to-antibody ratio of 1.81 were achieved. SDS-PAGE and SE-HPLC showed integrity of CI-8993 was maintained after conjugation; and ELISA indicated no impact of conjugation and radiolabelling on binding to human VISTA. PET imaging and biodistribution in MB49 tumour-bearing huVISTA KI female mice showed specific localisation of [89Zr]Zr-Df-CI-8993 to VISTA in spleen and tumour tissues expressing human VISTA. Specific tumour uptake was also demonstrated in Capan-2 xenografted BALB/c nu/nu mice. CONCLUSIONS We radiolabelled and validated [89Zr]Zr-Df-CI-8993 for specific binding to huVISTA in vivo. Our results demonstrate that 89Zr-labelled CI-8993 is now suitable for targeting and imaging VISTA expression in human trials.
Collapse
Affiliation(s)
- Ingrid Julienne Georgette Burvenich
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Christian Werner Wichmann
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Alexander Franklin McDonald
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
| | - Nancy Guo
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
| | - Angela Rigopoulos
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
| | - Nhi Huynh
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
| | - Mary Vail
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Stacey Allen
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | - Graeme Joseph O'Keefe
- Department of Molecular Imaging and Therapy, Austin Health, Melbourne, VIC, Australia
| | - Fiona Elizabeth Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia
| | | | | | | | - Andrew Mark Scott
- Tumour Targeting Laboratory, Olivia Newton-John Cancer Research Institute, Level 5 ONJ Centre, 145 Studley Road, Heidelberg, VIC, 3084, Australia.
- School of Cancer Medicine, La Trobe University, Melbourne, VIC, Australia.
- Department of Medicine, University of Melbourne, Melbourne, VIC, Australia.
| |
Collapse
|
5
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
6
|
Liu J, Liang Y, Yang H, Wang X, Zeng X, Zhuang R, Du J, Zhang X, Guo Z. Small-Molecule Radiotracers for Visualization of V-Domain Immunoglobulin Suppressor of T Cell Activation. J Med Chem 2024; 67:17690-17700. [PMID: 39305257 DOI: 10.1021/acs.jmedchem.4c01690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA) plays a critical role in regulating innate and adaptive immune responses within the tumor immune microenvironment. Quantifying VISTA expression is necessary to determine whether patients respond to a related combination immunotherapy. This study developed two 68Ga-labeled small-molecule probes ([68Ga]Ga-DCA and [68Ga]Ga-DNCA) for visualizing and differentiating VISTA expression. These probes exhibited excellent targeting capabilities for multiple tumor types (including B16-F10, 4T1, MC38, and CT26 tumors), consistent with the levels of VISTA expression determined by immunoblotting. Co-injection of inhibitor CA-170 led to decreased tumor uptake of both [68Ga]Ga-DCA and [68Ga]Ga-DNCA. [68Ga]Ga-DCA was used to verify the feasibility of monitoring VISTA expression in lung metastasis models. In summary, this study describes the use of 68Ga-labeled CA-170 analogues as small-molecule probes for imaging VISTA. This could provide a visual method and enable personalized immunotherapy in patients.
Collapse
Affiliation(s)
- Jia Liu
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
- Department of Nuclear Technology and Application, China Institute of Atomic Energy, P.O. Box 275(12), Beijing 102413, China
| | - Yuanyuan Liang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Hongzhang Yang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Xueqi Wang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Xinying Zeng
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Rongqiang Zhuang
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
| | - Jin Du
- Department of Nuclear Technology and Application, China Institute of Atomic Energy, P.O. Box 275(12), Beijing 102413, China
- China Isotope & Radiation Corporation, No. 66 Changwa Zhongjie, Haidian, Beijing 100089, China
- CAEA Center of Excellence on Nuclear Technology Application for Engineering and Industrialization of Radiopharmaceuticals, No. 1 Sanqiang Road, Xinzhen, Fangshan District, Beijing 102413, China
| | - Xianzhong Zhang
- Theranostics and Translational Research Center, Institute of Clinical Medicine, Department of Nuclear Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, No. 1 Shuaifuyuan, Dongcheng, Beijing 100730, China
| | - Zhide Guo
- State Key Laboratory of Vaccines for Infectious Diseases, Center for Molecular Imaging and Translational Medicine, Xiang An Biomedicine Laboratory, School of Public Health, Xiamen University, 4221-116 Xiang'An South Road, Xiamen 361102, China
| |
Collapse
|
7
|
Kamali AN, Hamedifar H, Eisenhut M, Bautista JM. Multiple myeloma and the potential of new checkpoint inhibitors for immunotherapy. Ther Adv Vaccines Immunother 2024; 12:25151355241288453. [PMID: 39399301 PMCID: PMC11467827 DOI: 10.1177/25151355241288453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/15/2024] Open
Abstract
Multiple myeloma (MM), a cancer of the bone marrow, is categorized as the second most common hematological malignancy of adults in the Western world. Despite dramatic improvements in immunotherapies in the field of cancers, MM immunotherapy has not been promising until now. Recent clinical studies of immune checkpoint inhibitor therapy, either alone or in combination with anticancer drugs, showed excessive side effects or low efficacy, particularly in advanced MM patients. In this context, lymphocyte levels of exhaustion markers play a pivotal role in the MM tumor microenvironment (TME). Hence in the present review, the mechanisms relevant to MM of five inhibitory molecules including T-cell immunoreceptor with Ig and ITIM domains (TIGIT), T-cell immunoglobulin, and mucin domain 3 (Tim-3), lymphocyte activation gene-3 (LAG-3), V-domain Ig Suppressor of T-cell activation and killer immunoglobulin-like receptors along with bispecific T-cell antibodies (BsAbs) will be discussed. Further, we summarized the underlying biology of these checkpoints in cancer and their rapidly emerging role in pathways in MM along with presenting recent clinical trials in context.
Collapse
Affiliation(s)
- Ali N. Kamali
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Haleh Hamedifar
- CinnaGen Medical Biotechnology Research Center, Alborz University of Medical Sciences, Karaj, Iran
- CinnaGen Research and Production Co., Alborz, Iran
| | - Michael Eisenhut
- Department of Pediatrics, Luton & Dunstable University Hospital, Luton, UK
| | - Jose M. Bautista
- Department of Biochemistry and Molecular Biology, Complutense University of Madrid, Faculty of Veterinary Sciences, Madrid, Spain
- Research Institute Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
8
|
Mashhouri S, Rahmati A, Azimi A, Fava RA, Ismail IH, Walker J, Elahi S. Targeting Dectin-1 and or VISTA enhances anti-tumor immunity in melanoma but not colorectal cancer model. Cell Oncol (Dordr) 2024; 47:1735-1756. [PMID: 38668817 PMCID: PMC11467025 DOI: 10.1007/s13402-024-00950-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/11/2024] [Indexed: 06/27/2024] Open
Abstract
PURPOSE Acquired resistance to immune checkpoint blockers (ICBs) is a major barrier in cancer treatment, emphasizing the need for innovative strategies. Dectin-1 (gene Clec7a) is a C-type lectin receptor best known for its ability to recognize β-glucan-rich structures in fungal cell walls. While Dectin-1 is expressed in myeloid cells and tumor cells, its significance in cancer remains the subject of controversy. METHODS Using Celc7a-/- mice and curdlan administration to stimulate Dectin-1 signaling, we explored its impact. VISTA KO mice were employed to assess VISTA's role, and bulk RNAseq analyzed curdlan effects on neutrophils. RESULTS Our findings reveal myeloid cells as primary Dectin-1 expressing cells in the tumor microenvironment (TME), displaying an activated phenotype. Strong Dectin-1 co-expression/co-localization with VISTA and PD-L1 in TME myeloid cells was observed. While Dectin-1 deletion lacked protective effects, curdlan stimulation significantly curtailed B16-F10 tumor progression. RNAseq and pathway analyses supported curdlan's role in triggering a cascade of events leading to increased production of pro-inflammatory mediators, potentially resulting in the recruitment and activation of immune cells. Moreover, we identified a heterogeneous subset of Dectin-1+ effector T cells in the TME. Similar to mice, human myeloid cells are the prominent cells expressing Dectin-1 in cancer patients. CONCLUSION Our study proposes Dectin-1 as a potential adjunctive target with ICBs, orchestrating a comprehensive engagement of innate and adaptive immune responses in melanoma. This innovative approach holds promise for overcoming acquired resistance to ICBs in cancer treatment, offering avenues for further exploration and development.
Collapse
Affiliation(s)
- Siavash Mashhouri
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Amirhossein Rahmati
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Ako Azimi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Roy A Fava
- Department of Veterans Affairs Medical Center, Research Service, White River Junction, VT, USA
- Department of Medicine, Geisel School of Medicine at Dartmouth, Dartmouth Hitchcock Medical Center, Lebanon, NH, USA
| | - Ismail Hassan Ismail
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
- Biophysics Department, Faculty of Science, Cairo University, Giza, 12613, Egypt
| | - John Walker
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada
| | - Shokrollah Elahi
- Department of Dentistry, Division of Foundational Sciences, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Department of Oncology, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Canada.
- Li Ka Shing Institute of Virology, University of Alberta, Edmonton, Canada.
| |
Collapse
|
9
|
Bugno J, Wang L, Yu X, Cao X, Wang J, Huang X, Yang K, Piffko A, Chen K, Luo SY, Naccasha E, Hou Y, Fu S, He C, Fu YX, Liang HL, Weichselbaum RR. Targeting the Dendritic Cell-Secreted Immunoregulatory Cytokine CCL22 Alleviates Radioresistance. Clin Cancer Res 2024; 30:4450-4463. [PMID: 38691100 PMCID: PMC11444901 DOI: 10.1158/1078-0432.ccr-23-3616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/20/2024] [Accepted: 04/29/2024] [Indexed: 05/03/2024]
Abstract
PURPOSE Radiation-mediated immune suppression limits efficacy and is a barrier in cancer therapy. Radiation induces negative regulators of tumor immunity including regulatory T cells (Treg). Mechanisms underlying Treg infiltration after radiotherapy (RT) are poorly defined. Given that conventional dendritic cells (cDC) maintain Treg, we sought to identify and target cDC signaling to block Treg infiltration after radiation. EXPERIMENTAL DESIGN Transcriptomics and high dimensional flow cytometry revealed changes in murine tumor cDC that not only mediate Treg infiltration after RT but also associate with worse survival in human cancer datasets. Antibodies perturbing a cDC-CCL22-Treg axis were tested in syngeneic murine tumors. A prototype interferon-anti-epidermal growth factor receptor fusion protein (αEGFR-IFNα) was examined to block Treg infiltration and promote a CD8+ T cell response after RT. RESULTS Radiation expands a population of mature cDC1 enriched in immunoregulatory markers that mediates Treg infiltration via the Treg-recruiting chemokine CCL22. Blocking CCL22 or Treg depletion both enhanced RT efficacy. αEGFR-IFNα blocked cDC1 CCL22 production while simultaneously inducing an antitumor CD8+ T cell response to enhance RT efficacy in multiple EGFR-expressing murine tumor models, including following systemic administration. CONCLUSIONS We identify a previously unappreciated cDC mechanism mediating Treg tumor infiltration after RT. Our findings suggest blocking the cDC1-CCL22-Treg axis augments RT efficacy. αEGFR-IFNα added to RT provided robust antitumor responses better than systemic free interferon administration and may overcome clinical limitations to interferon therapy. Our findings highlight the complex behavior of cDC after RT and provide novel therapeutic strategies for overcoming RT-driven immunosuppression to improve RT efficacy. See related commentary by Kalinski et al., p. 4260.
Collapse
Affiliation(s)
- Jason Bugno
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago, Chicago, Illinois
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Xianbin Yu
- Department of Chemistry, University of Chicago, Chicago, Illinois
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois
- Howard Hughes Medical Institute, University of Chicago, Chicago, Illinois
| | - Xuezhi Cao
- Guangzhou National Laboratory, Guangzhou, China
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Katherine Chen
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Stephen Y Luo
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Emile Naccasha
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Yuzhu Hou
- Department of Pathogenic Microbiology and Immunology, School of Basic Medical Sciences, Xi'an Jiaotong University, Xi'an, China
| | - Sherry Fu
- UT Southwestern Medical School, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chuan He
- Department of Chemistry, University of Chicago, Chicago, Illinois
- Department of Biochemistry and Molecular Biology, University of Chicago, Chicago, Illinois
- Institute for Biophysical Dynamics, University of Chicago, Chicago, Illinois
- Howard Hughes Medical Institute, University of Chicago, Chicago, Illinois
| | - Yang-Xin Fu
- Department of Basic Medical Science, Tsinghua University, Beijing, China
| | - Hua L Liang
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| | - Ralph R Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago, Chicago, Illinois
- Ludwig Center for Metastasis Research, University of Chicago, Chicago, Illinois
| |
Collapse
|
10
|
Deng Y, Shi M, Yi L, Naveed Khan M, Xia Z, Li X. Eliminating a barrier: Aiming at VISTA, reversing MDSC-mediated T cell suppression in the tumor microenvironment. Heliyon 2024; 10:e37060. [PMID: 39286218 PMCID: PMC11402941 DOI: 10.1016/j.heliyon.2024.e37060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 08/10/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Immune checkpoint inhibitors (ICIs) have revolutionized cancer treatment by producing remarkable clinical outcomes for patients with various cancer types. However, only a subset of patients benefits from immunotherapeutic interventions due to the primary and acquired resistance to ICIs. Myeloid-derived suppressor cells (MDSCs) play a crucial role in creating an immunosuppressive tumor microenvironment (TME) and contribute to resistance to immunotherapy. V-domain Ig suppressor of T cell activation (VISTA), a negative immune checkpoint protein highly expressed on MDSCs, presents a promising target for overcoming resistance to current ICIs. This article provides an overview of the evidence supporting VISTA's role in regulating MDSCs in shaping the TME, thus offering insights into how to overcome immunotherapy resistance.
Collapse
Affiliation(s)
- Yayuan Deng
- The First College of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Mengjia Shi
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Lin Yi
- The First College of Clinical Medicine, Chongqing Medical University, Chongqing, China
| | - Muhammad Naveed Khan
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhijia Xia
- Department of General, Visceral, and Transplant Surgery, Ludwig-Maximilians-University Munich, Munich, 81377, Germany
| | - Xiaosong Li
- Clinical Molecular Medicine Testing Center, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
- Western(Chongqing) Collaborative Innovation Center for Intelligent Diagnostics and Digital Medicine, Chongqing National Biomedicine Industry Park, No. 28 Gaoxin Avenue, High-tech Zone, Chongqing, 401329, China
| |
Collapse
|
11
|
Liu S, Ji F, Ding Y, Ding B, Feng S, Brennick C, Lin H, Zhang T, Shen Y. VISTA: A promising target for overcoming immune evasion in gynecologic cancers. Int Immunopharmacol 2024; 138:112655. [PMID: 38986302 DOI: 10.1016/j.intimp.2024.112655] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/28/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Immune checkpoint blockade (ICB) therapy has revolutionized cancer treatment but has shown limited efficacy in gynecologic cancers. VISTA (V-domain Ig suppressor of T-cell activation), a member of the B7 family, is emerging as another checkpoint that regulates the anti-tumor immune responses within the tumor microenvironment. This paper reviews the structure, expression, and mechanism of action of VISTA. Furthermore, it highlights recent advances in VISTA-blocking therapies and their potential in improving outcomes for patients with gynecologic cancers. By understanding the role of VISTA in mediating the immune evasion of gynecologic tumors, we can develop more effective combinatory treatment strategies that could overcome resistance to current ICB therapies.
Collapse
Affiliation(s)
- Sicong Liu
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210003, China
| | - Feng Ji
- Department of Clinical Science and Research, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yue Ding
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210003, China
| | - Bo Ding
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210003, China
| | - Songwei Feng
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210003, China
| | - Cory Brennick
- Department of Immunobiology, Yale University, New Haven, CT 06511, USA
| | - Hao Lin
- Department of Clinical Science and Research, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China.
| | - Tianxiang Zhang
- Department of Immunobiology, Yale University, New Haven, CT 06511, USA.
| | - Yang Shen
- Department of Obstetrics and Gynecology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210003, China.
| |
Collapse
|
12
|
Yin J, Chen J, Wang T, Sun H, Yan Y, Zhu C, Huang L, Chen Z. Coinhibitory Molecule VISTA Play an Important Negative Regulatory Role in the Immunopathology of Bronchial Asthma. J Asthma Allergy 2024; 17:813-832. [PMID: 39246611 PMCID: PMC11378793 DOI: 10.2147/jaa.s449867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/01/2024] [Indexed: 09/10/2024] Open
Abstract
Objective To investigate the significance of VISTA in bronchial asthma and its impact on the disease. Methods Human peripheral blood of asthma children was gathered. The expression concentrations of VISTA, IL-4, IL-6, CD25, CD40L, and PD-L2 in peripheral blood plasma were detected by ELISA. We established the mouse model of asthma and intervened with agonistic anti-VISTA mAb (4C11) and VISTA fusion protein. ELISA, flow cytometry, and Western blotting were performed to detect the expression levels of Th1, Th2, and Th17 cell subsets and related characteristic cytokines, as well as the protein levels of MAPKs, NF-κB, and TRAF6 in lung tissues. In addition, the infiltration of eosinophils and inflammatory cells, airway mucus secretion, and VISTA protein expression in lung histopathological sections of different groups of mice were analyzed. Results The concentration of VISTA in human asthma group decreased significantly (p < 0.05); A positive correlation was observed between VISTA and CD40L. The intervention of 4C11 mAb and fusion protein respectively during the induction period increase the differentiation of Th1 cells and the secretion of IFN-γ, and inhibit the differentiation of Th2 and Th17 cells, as well as the secretion of IL-4, IL-5, IL-13 and IL-17, partially reduce the pathological changes of asthma in mouse lungs and correct the progress of asthma. The MAPK, NF-κB, and TRAF6 protein levels were the middle range in the 4C11 mAb and fusion protein groups (p < 0.05). Conclusion The findings suggest VISTA may play a negative regulatory role in the occurrence and development of bronchial asthma.
Collapse
Affiliation(s)
- Jianqun Yin
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Jiawei Chen
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Ting Wang
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Huiming Sun
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Yongdong Yan
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Canhong Zhu
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Li Huang
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| | - Zhengrong Chen
- Respiratory Department, Children's Hospital of Soochow University, Suzhou, People's Republic of China
| |
Collapse
|
13
|
Zhao H, Zhang W, Lu Y, Dong Y, He Z, Zhen H, Li Q. Inosine enhances the efficacy of immune-checkpoint inhibitors in advanced solid tumors: A randomized, controlled, Phase 2 study. Cancer Med 2024; 13:e70143. [PMID: 39267574 PMCID: PMC11393481 DOI: 10.1002/cam4.70143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 08/05/2024] [Accepted: 08/12/2024] [Indexed: 09/17/2024] Open
Abstract
BACKGROUND This study aimed to evaluate whether inosine enhances the efficacy of immune-checkpoint inhibitors in human malignant solid tumors. METHODS This single-center, prospective, randomized, open-label study was conducted, from January 2021 to December 2022, in Beijing Friendship Hospital, Capital Medical University, and participants were randomly assigned (1:1) to either the inosine (trial) or non-inosine (control) group that received inosine (dosage: 0.2 g, three times/day) + PD-1/PD-L1 inhibitor or only PD-1/PD-L1 inhibitor ± targeted ± chemotherapy, respectively. Efficacy was assessed every 6 weeks (i.e., after every two-three treatment cycles). The primary endpoint was the objective response rate (ORR); the secondary endpoints were disease control rate, overall survival (OS), and progression-free survival (PFS). The trial was registered at ClinicalTrials.gov (NCT05809336). RESULTS Among the 172 participants with advanced malignant solid tumors, 86 each were assigned to the inosine and non-inosine groups, wherein the median PFS (95% CI) was 7.00 (5.31-8.69) and 4.40 (3.10-5.70) months, respectively (hazard ratio [HR] 0.63; 95% CI 0.44-0.90, p = 0.011), and the ORR was 26.7% and 15.1%, respectively (p = 0.061). In the inosine and non-inosine groups, the median OS was not reached and was 29.67 (95% CI 17.40-41.94) months, respectively (HR 1.05 [95% CI 0.59-1.84], p = 0.874). Compared with the non-inosine group, the median PFS and ORR of the inosine group were significantly prolonged and improved in the multiple exploratory subgroup analyses. The safety analysis showed that Grades 3 and 4 adverse reactions occurred in 25 (29%) and 31 (36%) patients in the inosine and non-inosine groups, respectively, and tended to decrease in the inosine group compared with the non-inosine group. CONCLUSION Inosine had a tendency to enhance the efficacy of immune-checkpoint inhibitors and reduced immunotherapy-related adverse reactions.
Collapse
Affiliation(s)
- Haiqing Zhao
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
- Internal Medicine DepartmentPeople's Hospital of Shen chi CountyShanxiPeople's Republic of China
| | - Wei Zhang
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Yuting Lu
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Yin Dong
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Zhihao He
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Hongchao Zhen
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| | - Qin Li
- Department of Oncology, Beijing Friendship HospitalCapital Medical UniversityBeijingPeople's Republic of China
| |
Collapse
|
14
|
Vilela T, Valente S, Correia J, Ferreira F. Advances in immunotherapy for breast cancer and feline mammary carcinoma: From molecular basis to novel therapeutic targets. Biochim Biophys Acta Rev Cancer 2024; 1879:189144. [PMID: 38914239 DOI: 10.1016/j.bbcan.2024.189144] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 05/29/2024] [Accepted: 06/19/2024] [Indexed: 06/26/2024]
Abstract
The role of inflammation in cancer is a topic that has been investigated for many years. As established, inflammation emerges as a defining characteristic of cancer, presenting itself as a compelling target for therapeutic interventions in the realm of oncology. Controlling the tumor microenvironment (TME) has gained paramount significance, modifying not only the effectiveness of immunotherapy but also modulating the outcomes and prognoses of standard chemotherapy and other anticancer treatments. Immunotherapy has surfaced as a central focus within the domain of tumor treatments, using immune checkpoint inhibitors as cancer therapy. Immune checkpoints and their influence on the tumor microenvironment dynamic are presently under investigation, aiming to ascertain their viability as therapeutic interventions across several cancer types. Cancer presents a significant challenge in humans and cats, where female breast cancer ranks as the most prevalent malignancy and feline mammary carcinoma stands as the third most frequent. This review seeks to summarize the data about the immune checkpoints cytotoxic T-lymphocyte-associated antigen 4 (CTLA-4), lymphocyte activation gene-3 (LAG-3), programmed cell death protein-1 (PD-1), V-domain Ig suppressor of T cell activation (VISTA), and T-cell immunoglobulin and mucin domain 3 (TIM-3) respective ongoing investigations as prospective targets for therapy for human breast cancer, while also outlining findings from studies reported on feline mammary carcinoma (FMC), strengthening the rationale for employing FMC as a representative model in the exploration of human breast cancer.
Collapse
Affiliation(s)
- Tatiana Vilela
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Sofia Valente
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal
| | - Jorge Correia
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; CIISA-Center of Interdisciplinary Research in Animal Health, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal
| | - Fernando Ferreira
- Faculty of Veterinary Medicine, University of Lisbon, Avenida da Universidade Técnica, 1300-477 Lisbon, Portugal; CIISA-Center of Interdisciplinary Research in Animal Health, 1300-477 Lisbon, Portugal; Associate Laboratory for Animal and Veterinary Sciences (AL4AnimalS), 1300-477 Lisbon, Portugal.
| |
Collapse
|
15
|
Moon TJ, Ta HM, Bhalotia A, Paulsen KE, Hutchinson DW, Arkema GM, Choi AS, Haynie MG, Ogunnaike L, Dever M, Wang LL, Karathanasis E. Nanoparticles targeting immune checkpoint protein VISTA induce potent antitumor immunity. J Immunother Cancer 2024; 12:e008977. [PMID: 39209454 PMCID: PMC11367342 DOI: 10.1136/jitc-2024-008977] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND Immune checkpoint protein V-domain immunoglobulin suppressor of T cell activation (VISTA) controls antitumor immunity and is a valuable target for cancer immunotherapy. Previous mechanistic studies have indicated that VISTA impairs the toll-like receptor (TLR)-mediated activation of myeloid antigen-presenting cells, promoting the expansion of myeloid-derived suppressor cells, and suppressing tumor-reactive cytotoxic T cell function. METHODS The aim of this study was to develop a dual-action lipid nanoparticle (dual-LNP) coloaded with VISTA-specific siRNA and TLR9 agonist CpG oligonucleotide. We used three murine preclinical tumor models, melanoma YUMM1.7, melanoma B16F10, and colon carcinoma MC38 to assess the functional synergy of the two cargoes of the dual LNP and therapeutic efficacy. RESULTS The dual-LNP synergistically augmented antitumor immune responses and rejected large established tumors whereas LNPs containing VISTA siRNA or CpG alone were ineffective. In comparison with therapies using the soluble CpG and a VISTA-specific monoclonal antibody, the dual-LNP demonstrated superior therapeutic efficacy yet with reduced systemic inflammatory cytokine production. In three murine models, the dual-LNP treatment achieved a high cure rate. Tumor rejection was associated with influx of immune cells to tumor tissues, augmented dendritic cell activation, production of proinflammatory cytokines, and improved function of cytotoxic T cells. CONCLUSIONS Our studies show the dual-LNP ensured codelivery of its synergistic cargoes to tumor-infiltrating myeloid cells, leading to simultaneous silencing of VISTA and stimulation of TLR9. As a result, the dual-LNP drove a highly potent antitumor immune response that rejected large aggressive tumors, thus may be a promising therapeutic platform for treating immune-cold tumors.
Collapse
Affiliation(s)
- Taylor J Moon
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Hieu Minh Ta
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
| | - Anubhuti Bhalotia
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Kai E Paulsen
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Diarmuid W Hutchinson
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Gabrielle M Arkema
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Andrew S Choi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Michiko G Haynie
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Laolu Ogunnaike
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Margee Dever
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
| | - Li Lily Wang
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Department of Translational Hematology and Oncology Research, Cleveland Clinic, Cleveland, Ohio, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Efstathios Karathanasis
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, Ohio, USA
- Case Comprehensive Cancer Center, Cleveland, Ohio, USA
| |
Collapse
|
16
|
Fu Y, Guo X, Sun L, Cui T, Wu C, Wang J, Liu Y, Liu L. Exploring the role of the immune microenvironment in hepatocellular carcinoma: Implications for immunotherapy and drug resistance. eLife 2024; 13:e95009. [PMID: 39146202 PMCID: PMC11326777 DOI: 10.7554/elife.95009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Accepted: 08/04/2024] [Indexed: 08/17/2024] Open
Abstract
Hepatocellular carcinoma (HCC), the most common type of liver tumor, is a leading cause of cancer-related deaths, and the incidence of liver cancer is still increasing worldwide. Curative hepatectomy or liver transplantation is only indicated for a small population of patients with early-stage HCC. However, most patients with HCC are not candidates for radical resection due to disease progression, leading to the choice of the conventional tyrosine kinase inhibitor drug sorafenib as first-line treatment. In the past few years, immunotherapy, mainly immune checkpoint inhibitors (ICIs), has revolutionized the clinical strategy for HCC. Combination therapy with ICIs has proven more effective than sorafenib, and clinical trials have been conducted to apply these therapies to patients. Despite significant progress in immunotherapy, the molecular mechanisms behind it remain unclear, and immune resistance is often challenging to overcome. Several studies have pointed out that the complex intercellular communication network in the immune microenvironment of HCC regulates tumor escape and drug resistance to immune response. This underscores the urgent need to analyze the immune microenvironment of HCC. This review describes the immunosuppressive cell populations in the immune microenvironment of HCC, as well as the related clinical trials, aiming to provide insights for the next generation of precision immunotherapy.
Collapse
Affiliation(s)
- Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Xinyu Guo
- Department of General Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Chenghui Wu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, China
| |
Collapse
|
17
|
Jin S, Liu W, He X, Zhang Y, Chen W, Wu Y, Liu J. VISTA deficiency exerts anti-tumor effects in breast cancer through regulating macrophage polarization. Int Immunopharmacol 2024; 136:112365. [PMID: 38820964 DOI: 10.1016/j.intimp.2024.112365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 05/20/2024] [Accepted: 05/27/2024] [Indexed: 06/02/2024]
Abstract
Growing evidence had showed that tumor-associated macrophages (TAMs) have a tumor-promoting M2 phenotype which could drive pathological phenomena. In breast cancer, TAMs are abundantly present and may play an important role in the development of breast cancer. V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel inhibitory checkpoint and immunotherapy target for tumor through regulating immune response. However, its effects on macrophages have not been investigated, which was also the focus of this study. Here, the scRNA-seq data further revealed that VISTA was highly expressed in multiple macrophage subclusters. In vitro experiments showed that the absence of VISTA enhanced the M1 polarization of macrophages, inhibited the M2 polarization of macrophages and the proliferation and phagocytosis of 4 T1 cells induced by M2-CM. VISTA regulated the activation of STAT1 and STAT6 signaling pathways in the process of macrophage polarization. In vivo experiments demonstrated that VISTA deficient mice exhibited reduced tumor growth, possibly due to the increase of M1 macrophages and the decrease of M2 macrophages. In summary, our study is the first to reveal the effect of VISTA on macrophages in breast cancer, which showed that VISTA affects tumor growth by critically regulating the macrophage polarization through the STAT pathway.
Collapse
Affiliation(s)
- Shasha Jin
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China
| | - Wanmei Liu
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China
| | - Xiaoyu He
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China
| | - Yuxin Zhang
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China
| | - Wenting Chen
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China
| | - Yinhao Wu
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China
| | - Jun Liu
- New Drug Screening Center, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
18
|
Du G, Dou C, Sun P, Wang S, Liu J, Ma L. Regulatory T cells and immune escape in HCC: understanding the tumor microenvironment and advancing CAR-T cell therapy. Front Immunol 2024; 15:1431211. [PMID: 39136031 PMCID: PMC11317284 DOI: 10.3389/fimmu.2024.1431211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Accepted: 07/12/2024] [Indexed: 08/15/2024] Open
Abstract
Liver cancer, which most commonly manifests as hepatocellular carcinoma (HCC), is the sixth most common cancer in the world. In HCC, the immune system plays a crucial role in the growth and proliferation of tumor cells. HCC achieve immune escape through the tumor microenvironment, which significantly promotes the development of this cancer. Here, this article introduces and summarizes the functions and effects of regulatory T cells (Tregs) in the tumor microenvironment, highlighting how Tregs inhibit and regulate the functions of immune and tumor cells, cytokines, ligands and receptors, etc, thereby promoting tumor immune escape. In addition, it discusses the mechanism of CAR-T therapy for HCC and elaborate on the relationship between CAR-T and Tregs.
Collapse
Affiliation(s)
- Guangtan Du
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Cunmiao Dou
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Medical Department of Qingdao University, Qingdao, China
| | - Peng Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Shasha Wang
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jia Liu
- Department of Pharmacology, School of Pharmacy, Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| | - Leina Ma
- Department of Oncology, The Affiliated Hospital of Qingdao University, Qingdao, China
- Qingdao Cancer Institute, Qingdao, China
| |
Collapse
|
19
|
Muñoz Perez N, Pensabene JM, Galbo PM, Sadeghipour N, Xiu J, Moziak K, Yazejian RM, Welch RL, Bell WR, Sengupta S, Aulakh S, Eberhart CG, Loeb DM, Eskandar E, Zheng D, Zang X, Martin AM. VISTA Emerges as a Promising Target against Immune Evasion Mechanisms in Medulloblastoma. Cancers (Basel) 2024; 16:2629. [PMID: 39123357 PMCID: PMC11312086 DOI: 10.3390/cancers16152629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/12/2024] [Accepted: 07/18/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND Relapsed medulloblastoma (MB) poses a significant therapeutic challenge due to its highly immunosuppressive tumor microenvironment. Immune checkpoint inhibitors (ICIs) have struggled to mitigate this challenge, largely due to low T-cell infiltration and minimal PD-L1 expression. Identifying the mechanisms driving low T-cell infiltration is crucial for developing more effective immunotherapies. METHODS We utilize a syngeneic mouse model to investigate the tumor immune microenvironment of MB and compare our findings to transcriptomic and proteomic data from human MB. RESULTS Flow cytometry reveals a notable presence of CD45hi/CD11bhi macrophage-like and CD45int/CD11bint microglia-like tumor-associated macrophages (TAMs), alongside regulatory T-cells (Tregs), expressing high levels of the inhibitory checkpoint molecule VISTA. Compared to sham control mice, the CD45hi/CD11bhi compartment significantly expands in tumor-bearing mice and exhibits a myeloid-specific signature composed of VISTA, CD80, PD-L1, CTLA-4, MHCII, CD40, and CD68. These findings are corroborated by proteomic and transcriptomic analyses of human MB samples. Immunohistochemistry highlights an abundance of VISTA-expressing myeloid cells clustering at the tumor-cerebellar border, while T-cells are scarce and express FOXP3. Additionally, tumor cells exhibit immunosuppressive properties, inhibiting CD4 T-cell proliferation in vitro. Identification of VISTA's binding partner, VSIG8, on tumor cells, and its correlation with increased VISTA expression in human transcriptomic analyses suggests a potential therapeutic target. CONCLUSIONS This study underscores the multifaceted mechanisms of immune evasion in MB and highlights the therapeutic potential of targeting the VISTA-VSIG axis to enhance anti-tumor responses.
Collapse
Affiliation(s)
- Natalia Muñoz Perez
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Juliana M. Pensabene
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Phillip M. Galbo
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | | | - Joanne Xiu
- Caris Life Sciences, Phoenix, AZ 85040, USA; (N.S.); (J.X.)
| | - Kirsten Moziak
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Rita M. Yazejian
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Rachel L. Welch
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - W. Robert Bell
- Department of Clinical Pathology & Laboratory Medicine, School of Medicine, Indiana University, 340 West 10th Street Fairbanks Hall, Indianapolis, IN 46202, USA;
| | - Soma Sengupta
- Department of Neurology & Neurosurgery, University of North Carolina at Chapel Hill, 170 Manning Drive, Chapel Hill, NC 27599, USA;
| | - Sonikpreet Aulakh
- Department of Internal Medicine, West Virginia University, 64 Medical Center Drive, Morgantown, WV 26506, USA;
| | - Charles G. Eberhart
- Department of Pathology, Johns Hopkins School of Medicine, 600 N Wolfe St., Baltimore, MD 21287, USA;
| | - David M. Loeb
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Emad Eskandar
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Deyou Zheng
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Xingxing Zang
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| | - Allison M. Martin
- Department of Pediatrics, Albert Einstein College of Medicine, 1300 Morris Park Avenue, Bronx, NY 10461, USA; (J.M.P.); (P.M.G.J.); (K.M.); (R.M.Y.); (R.L.W.); (D.M.L.); (E.E.); (D.Z.); (X.Z.)
| |
Collapse
|
20
|
Gao Y, He Y, Tang Y, Chen ZS, Qu M. VISTA: A Novel Checkpoint for Cancer Immunotherapy. Drug Discov Today 2024; 29:104045. [PMID: 38797321 DOI: 10.1016/j.drudis.2024.104045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Revised: 04/20/2024] [Accepted: 05/21/2024] [Indexed: 05/29/2024]
Abstract
V-domain Ig suppressor of T cell activation (VISTA) is a recently identified member of the B7 family of immunoregulatory proteins. It is pivotal for maintaining T cell quiescence and exerts a significant regulatory influence on the immune response to tumors. Accumulating clinical evidence suggests that the influence of VISTA on tumor immunity is more nuanced than initially postulated. Although these revelations add layers of complexity to our understanding of the function of VISTA, they also offer novel avenues for scientific inquiry and potential therapeutic targets. In this review, we scrutinize the current literature pertaining to the expression of VISTA in various of malignancies, aiming to elucidate its intricate roles within the tumor microenvironment and in cancer immunotherapy.
Collapse
Affiliation(s)
- Yu Gao
- Translational Medical Center, Weifang Second People's Hospital, Weifang 261041, Shandong, China
| | - Yanting He
- Department of Pathology, The Affiliated Hospital of Qingdao University, Pingdu 266700, Shandong, China
| | - Yuanyuan Tang
- Translational Medical Center, Weifang Second People's Hospital, Weifang 261041, Shandong, China
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, NY 11439, USA.
| | - Meihua Qu
- Translational Medical Center, Weifang Second People's Hospital, Weifang 261041, Shandong, China; School of Life Science and Technology, Weifang Medical University, Weifang 261053, Shandon, China.
| |
Collapse
|
21
|
Wu Y, Chen D, Gao Y, Xu Q, Zhou Y, Ni Z, Na M. Immunosuppressive regulatory cells in cancer immunotherapy: restrain or modulate? Hum Cell 2024; 37:931-943. [PMID: 38814516 DOI: 10.1007/s13577-024-01083-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 05/22/2024] [Indexed: 05/31/2024]
Abstract
Immunosuppressive regulatory cells (IRCs) play important roles in negatively regulating immune response, and are mainly divided into myeloid-derived suppressor cells (MDSCs) and regulatory T cells (Tregs). Large numbers of preclinical and clinical studies have shown that inhibition or reduction of IRCs could effectively elevate antitumor immune responses. However, several studies also reported that excessive inhibition of IRCs function is one of the main reasons causing the side effects of cancer immunotherapy. Therefore, the reasonable regulation of IRCs is crucial for improving the safety and efficiency of cancer immunotherapy. In this review, we summarised the recent research advances in the cancer immunotherapy by regulating the proportion of IRCs, and discussed the roles of IRCs in regulating tumour immune evasion and drug resistance to immunotherapies. Furthermore, we also discussed how to balance the potential opportunities and challenges of using IRCs to improve the safety of cancer immunotherapies.
Collapse
Affiliation(s)
- Yan Wu
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, 210031, Jiangsu, People's Republic of China
- School of Medicine, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Dongfeng Chen
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yang Gao
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau, 999078, China
| | - Qinggang Xu
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Yang Zhou
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Zhong Ni
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China
| | - Manli Na
- Department of Burn and Plastic Surgery, The Affiliated Hospital of Jiangsu University, Zhenjiang, 210031, Jiangsu, People's Republic of China.
- School of Life Sciences, Jiangsu University, Zhenjiang, 212013, Jiangsu, People's Republic of China.
- International Genome Center, Jiangsu University, 301 Xuefu Road, Zhenjiang, 212013, Jiangsu, People's Republic of China.
| |
Collapse
|
22
|
Tang D, Zhao L, Yan F, Ren C, Xu K, Zhao K. Expression of VISTA regulated via IFN-γ governs endogenous T-cell function and exhibits correlation with the efficacy of CD19 CAR-T cell treated B-malignant mice. J Immunother Cancer 2024; 12:e008364. [PMID: 38925679 PMCID: PMC11202651 DOI: 10.1136/jitc-2023-008364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/30/2024] [Indexed: 06/28/2024] Open
Abstract
BACKGROUND Despite continuous improvements in the new target and construction of chimeric antigen receptor (CAR)-T, relapse remains a significant challenge following CAR-T therapy. Tumor microenvironment (TME) strongly correlates with the efficacy of CAR-T therapy. V-domain Ig suppressor of T-cell activation (VISTA), which exerts a multifaceted and controversial role in regulating the TME, acts not only as a ligand on antigen-presenting cells but also functions as a receptor on T cells. However, the characteristics and underlying mechanisms governing endogenous T-cell activation by VISTA, which are pivotal for reshaping the TME, remain incompletely elucidated. METHODS The immunocompetent B acute lymphoblastic leukemia (B-ALL), lymphoma, and melanoma murine models were employed to investigate the characteristics of endogenous T cells within the TME following CD19 and hCAIX CAR-T cell therapy, respectively. Furthermore, we examined the role of VISTA controlled by interferon (IFN)-γ signaling in regulating endogenous T-cell activation and functionality in B-ALL mice. RESULTS We demonstrated that the administration of CD19 CAR-T or hCAIX CAR-T cell therapy elicited augmented immune responses of endogenous T cells within the TME of B-ALL, lymphoma, and melanoma mice, thereby substantiating the efficacy of CAR-T cell efficacy. However, in the TME lacking IFN-γ signaling, VISTA levels remained elevated, resulting in attenuated cytotoxicity of endogenous T cells and reduced B-ALL recipient survival. Mice treated with CD19 CAR-T cells exhibited increased proportions of endogenous memory T cells during prolonged remission, which possessed the tumor-responsive capabilities to protect against B-ALL re-challenge. Compared with wild-type (WT) CAR-T treated mice, the administration of IFN-γ-/- CAR-T to both WT and IFN-γ-/- recipients resulted in a reduction in the numbers of endogenous CD4+ and CD8+ effectors, while exhibiting increased populations of naïve-like CD4+ T and memory CD8+ T cells. VISTA expression consistently remained elevated in resting or memory CD4+ T cells, with distinct localization from programmed cell death protein-1 (PD-1) expressing T subsets. Blocking the VISTA signal enhanced dendritic cell-induced proliferation and cytokine production by syngeneic T cells. CONCLUSION Our findings confirm that endogenous T-cell activation and functionality are regulated by VISTA, which is associated with the therapeutic efficiency of CAR-T and provides a promising therapeutic strategy for relapse cases in CAR-T therapy.
Collapse
Affiliation(s)
- Donghai Tang
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Li Zhao
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Fen Yan
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Chunxiao Ren
- Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kailin Xu
- Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| | - Kai Zhao
- Xuzhou Medical University, Xuzhou, Jiangsu, China
- Department of Hematology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, Jiangsu, China
| |
Collapse
|
23
|
Liu J, Wang C, Jiang Y, Zhou Y, Chen L, Qian Z, Liu L, Wu D, Zhang Y. Comprehensive characterization of B7 family members in breast cancer: B7-H5 switch reverses breast cancer from "immuno-cold" into "immuno-hot" status. Cancer Cell Int 2024; 24:205. [PMID: 38858701 PMCID: PMC11165836 DOI: 10.1186/s12935-024-03392-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 06/02/2024] [Indexed: 06/12/2024] Open
Abstract
The members of the classic B7 family regulate the immune microenvironment of several malignant tumors. However, the potential relationship between the B7 family and the breast cancer (BrCa) tumor immune microenvironment has remained elusive. In the present study, we provide a comprehensive explanation of the expression, clinical significance, mutation, and immune cell infiltration of B7 family molecules in BrCa. First, we recruited 10 patients with BrCa surgery from the Wuxi Maternal and Child Health Hospital and performed single-cell RNA sequencing (scRNA-seq) analysis to investigate the distribution of B7 family members in multiple immune cell subsets. We focused on B7-2, B7-H3, and B7-H5 molecules of the B7 family and constructed tumor microarrays by self-recruiting patients to perform multiple immunohistochemical (mIHC) analyses and study tumor expression of B7-2, B7-H3, B7-H5 and CD8+ immune cell infiltration. B7-H5 displayed a strong correlation with CD8+ immune cell infiltration. In summary, B7-H5 provides a new perspective for the identification of immunothermal subtypes of BrCa and could function as a switch to reverse BrCa from an "immunologically cold" state to an "immunologically hot" state.
Collapse
Affiliation(s)
- Jiayu Liu
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, No.48 Huaishu Road, Wuxi, Jiangsu, 214002, China
| | - Cenzhu Wang
- Wuxi People's Hospital, Wuxi Medical Center, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Nanjing Medical University, Jiangsu, 214000, China
- Department of Oncology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Jiangsu, 214023, China
| | - Ying Jiang
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, No.48 Huaishu Road, Wuxi, Jiangsu, 214002, China
| | - Yunxu Zhou
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, No.48 Huaishu Road, Wuxi, Jiangsu, 214002, China
| | - Lingyan Chen
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Jiangsu, 214000, China
| | - Zhiwen Qian
- Wuxi Maternal and Child Health Hospital, Nanjing Medical University, Jiangsu, 214000, China
| | - Lu Liu
- Wuxi Maternal and Child Health Hospital, Jiangnan University, Jiangsu, 214002, China
| | - Danping Wu
- Wuxi Maternal and Child Health Hospital, Jiangnan University, Jiangsu, 214002, China
| | - Yan Zhang
- Department of Oncology, Wuxi Maternal and Child Health Care Hospital, Wuxi School of Medicine, Jiangnan University, No.48 Huaishu Road, Wuxi, Jiangsu, 214002, China.
- Wuxi Maternal and Child Health Hospital, Jiangnan University, Jiangsu, 214002, China.
| |
Collapse
|
24
|
Yin N, Li X, Zhang X, Xue S, Cao Y, Niedermann G, Lu Y, Xue J. Development of pharmacological immunoregulatory anti-cancer therapeutics: current mechanistic studies and clinical opportunities. Signal Transduct Target Ther 2024; 9:126. [PMID: 38773064 PMCID: PMC11109181 DOI: 10.1038/s41392-024-01826-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 03/25/2024] [Accepted: 03/28/2024] [Indexed: 05/23/2024] Open
Abstract
Immunotherapy represented by anti-PD-(L)1 and anti-CTLA-4 inhibitors has revolutionized cancer treatment, but challenges related to resistance and toxicity still remain. Due to the advancement of immuno-oncology, an increasing number of novel immunoregulatory targets and mechanisms are being revealed, with relevant therapies promising to improve clinical immunotherapy in the foreseeable future. Therefore, comprehending the larger picture is important. In this review, we analyze and summarize the current landscape of preclinical and translational mechanistic research, drug development, and clinical trials that brought about next-generation pharmacological immunoregulatory anti-cancer agents and drug candidates beyond classical immune checkpoint inhibitors. Along with further clarification of cancer immunobiology and advances in antibody engineering, agents targeting additional inhibitory immune checkpoints, including LAG-3, TIM-3, TIGIT, CD47, and B7 family members are becoming an important part of cancer immunotherapy research and discovery, as are structurally and functionally optimized novel anti-PD-(L)1 and anti-CTLA-4 agents and agonists of co-stimulatory molecules of T cells. Exemplified by bispecific T cell engagers, newly emerging bi-specific and multi-specific antibodies targeting immunoregulatory molecules can provide considerable clinical benefits. Next-generation agents also include immune epigenetic drugs and cytokine-based therapeutics. Cell therapies, cancer vaccines, and oncolytic viruses are not covered in this review. This comprehensive review might aid in further development and the fastest possible clinical adoption of effective immuno-oncology modalities for the benefit of patients.
Collapse
Affiliation(s)
- Nanhao Yin
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xintong Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Xuanwei Zhang
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
| | - Shaolong Xue
- Department of Gynecology and Obstetrics, West China Second University Hospital, Sichuan University, No. 20, Section 3, South Renmin Road, Chengdu, 610041, Sichuan, PR China
| | - Yu Cao
- Department of Emergency Medicine, Laboratory of Emergency Medicine, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China
- Institute of Disaster Medicine & Institute of Emergency Medicine, Sichuan University, No. 17, Gaopeng Avenue, Chengdu, 610041, Sichuan, PR China
| | - Gabriele Niedermann
- Department of Radiation Oncology, Medical Center-University of Freiburg, Faculty of Medicine, University of Freiburg, German Cancer Consortium (DKTK) Partner Site DKTK-Freiburg, Robert-Koch-Strasse 3, 79106, Freiburg, Germany.
| | - You Lu
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| | - Jianxin Xue
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center & State Key Laboratory of Biotherapy, and The National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, No. 37, Guoxue Lane, Chengdu, 610041, Sichuan, PR China.
- Laboratory of Clinical Cell Therapy, West China Hospital, Sichuan University, No. 2222, Xinchuan Road, Chengdu, 610041, Sichuan, PR China.
| |
Collapse
|
25
|
Ta HM, Roy D, Zhang K, Alban T, Juric I, Dong J, Parthasarathy PB, Patnaik S, Delaney E, Gilmour C, Zakeri A, Shukla N, Rupani A, Phoon YP, Liu C, Avril S, Gastman B, Chan T, Wang LL. LRIG1 engages ligand VISTA and impairs tumor-specific CD8 + T cell responses. Sci Immunol 2024; 9:eadi7418. [PMID: 38758807 PMCID: PMC11334715 DOI: 10.1126/sciimmunol.adi7418] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 04/25/2024] [Indexed: 05/19/2024]
Abstract
Immune checkpoint blockade is a promising approach to activate antitumor immunity and improve the survival of patients with cancer. V-domain immunoglobulin suppressor of T cell activation (VISTA) is an immune checkpoint target; however, the downstream signaling mechanisms are elusive. Here, we identify leucine-rich repeats and immunoglobulin-like domains 1 (LRIG1) as a VISTA binding partner, which acts as an inhibitory receptor by engaging VISTA and suppressing T cell receptor signaling pathways. Mice with T cell-specific LRIG1 deletion developed superior antitumor responses because of expansion of tumor-specific cytotoxic T lymphocytes (CTLs) with increased effector function and survival. Sustained tumor control was associated with a reduction of quiescent CTLs (TCF1+ CD62Lhi PD-1low) and a reciprocal increase in progenitor and memory-like CTLs (TCF1+ PD-1+). In patients with melanoma, elevated LRIG1 expression on tumor-infiltrating CD8+ CTLs correlated with resistance to immunotherapies. These results delineate the role of LRIG1 as an inhibitory immune checkpoint receptor and propose a rationale for targeting the VISTA/LRIG1 axis for cancer immunotherapy.
Collapse
Affiliation(s)
- Hieu Minh Ta
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Dia Roy
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Keman Zhang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Tyler Alban
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Ivan Juric
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Juan Dong
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Prerana B. Parthasarathy
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Sachin Patnaik
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Elizabeth Delaney
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Cassandra Gilmour
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
| | - Amin Zakeri
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Nidhi Shukla
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Amit Rupani
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Yee Peng Phoon
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Caini Liu
- Department of Inflammation and Immunology, Cleveland Clinic Lerner College of Medicine, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Stefanie Avril
- Department of Pathology, University Hospitals Cleveland Medical Center, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Brian Gastman
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
| | - Timothy Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Lerner Research Institute, Cleveland Clinic Foundation, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Li Lily Wang
- Department of Translational Hematology and Oncology Research, Taussig Cancer Institute, Cleveland Clinic, Cleveland, OH, USA
- Department of Molecular Medicine, Cleveland Clinic Lerner College of Medicine, Case Western Reserve University, Cleveland, OH, USA
- Case Comprehensive Cancer Center, Cleveland, OH, USA
| |
Collapse
|
26
|
Pan J, Mahsud I, Ul Haq M, Khan S, Alhomrani M, Alamri AS, Alghamdi SA, ALSuhaymi N, Baothman BK, Almaghrabi S, Ullah S, Jamil M. Comprehensive pan-cancer analysis reveals VSIR as a candidate immunologic, diagnostic, and prognostic biomarker. Am J Transl Res 2024; 16:1630-1642. [PMID: 38883368 PMCID: PMC11170614 DOI: 10.62347/jmbz8836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 04/22/2024] [Indexed: 06/18/2024]
Abstract
OBJECTIVES Being a checkpoint, the expression level of V-set immunoregulatory receptor (VSIR) serves as an indicator of the extent of immunosuppression. Our objective was to undertake a pan-cancer analysis to examine the expression, genetic alterations, prognosis, and immunologic features associated with VSIR. METHODS The Cancer Genome Atlas (TCGA), Genotype-Tissue Expression (GTEx), GEPIA2, UALCAN, OncoDB, Human Protein Atlas (HPA), STRING, DAVID, cell culture, clinical sample collection, and reverse transcription quantitative polymerase chain reaction (RT-qPCR) were used. RESULTS This study comprehensively assessed VSIR across 33 cancers using TCGA and GTEx databases. Differential expression analysis revealed elevated VSIR in several cancers, notably in cholangiocarcinoma, esophageal carcinoma, kidney renal cell carcinoma, and liver hepatocellular carcinoma, while decreased expression was observed in various others. Prognostic analysis highlighted its significant association with reduced overall survival (OS) in ESCA and LIHC. Investigation into cancer stages demonstrated a correlation between VSIR expression and stage in ESCA and LIHC. Promoter methylation analysis indicated decreased VSIR methylation levels in tumors, implicating a role in oncogenesis. Furthermore, subcellular localization predictions, Tumor Mutational Burden (TMB), and Microsatellite Instability (MSI) correlations revealed intriguing insight into VSIR's function. Notably, a positive correlation was identified between VSIR expression and various immune cells in both cancers. Protein-protein interaction (PPI) network construction and gene enrichment analysis elucidated VSIR-associated dysregulated pathways, emphasizing its possible involvement in diverse pathways. Finally, experimental validation using LIHC clinical samples and cell lines confirmed elevated VSIR expression, supporting its oncogenic role. CONCLUSION Collectively, these findings present a comprehensive understanding of VSIR's diverse roles and potential clinical implications in ESCA and LIHC.
Collapse
Affiliation(s)
- Jun Pan
- Department of Thoracic Surgery/Cardiovascular Surgery, The First People's Hospital of Xiaoshan District, Xiaoshan Affiliated Hospital of Wenzhou Medical University Hangzhou 311200, Zhejiang, China
| | | | - Moeen Ul Haq
- Gastroenterology Department, Gomal Medical College/MTI D.I.Khan Pakistan
| | | | - Majid Alhomrani
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University Taif, Saudi Arabia
- Research Centre for Health Sciences, Taif University Taif, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University Taif, Saudi Arabia
- Research Centre for Health Sciences, Taif University Taif, Saudi Arabia
| | - Saleh A Alghamdi
- Department of Clinical Laboratories Sciences, The Faculty of Applied Medical Sciences, Taif University Taif, Saudi Arabia
| | - Naif ALSuhaymi
- Department of Emergency Medical Services, Faculty of Health Sciences - AlQunfudah, Umm Al-Qura University Mekkah, Saudi Arabia
| | - Bandar K Baothman
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences in Rabigh, King Abdulaziz University Jeddah 21589, Saudi Arabia
| | - Sarah Almaghrabi
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University Jeddah 21589, Saudi Arabia
- Center of Innovations in Personalized Medicine (CIPM), King Abdulaziz University 21589 Jeddah, Saudi Arabia
| | | | - Muhammad Jamil
- PARC Arid Zone Research Center Dera Ismail Khan 29050, Pakistan
| |
Collapse
|
27
|
Ferencz B, Török K, Pipek O, Fillinger J, Csende K, Lantos A, Černeková R, Mitták M, Škarda J, Delongová P, Megyesfalvi E, Schelch K, Lang C, Solta A, Boettiger K, Brcic L, Lindenmann J, Rényi-Vámos F, Aigner C, Berta J, Megyesfalvi Z, Döme B. Expression patterns of novel immunotherapy targets in intermediate- and high-grade lung neuroendocrine neoplasms. Cancer Immunol Immunother 2024; 73:114. [PMID: 38693435 PMCID: PMC11063022 DOI: 10.1007/s00262-024-03704-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 04/14/2024] [Indexed: 05/03/2024]
Abstract
BACKGROUND Advancements in immunotherapeutic approaches only had a modest impact on the therapy of lung neuroendocrine neoplasms (LNENs). Our multicenter study aimed to investigate the expression patterns of novel immunotherapy targets in intermediate- and high-grade LNENs. METHODS The expressions of V-domain Ig suppressor of T cell activation (VISTA), OX40L, Glucocorticoid-induced TNF receptor (GITR), and T cell immunoglobulin and mucin domain 3 (TIM3) proteins were measured by immunohistochemistry in surgically resected tumor samples of 26 atypical carcinoid (AC), 49 large cell neuroendocrine lung cancer (LCNEC), and 66 small cell lung cancer (SCLC) patients. Tumor and immune cells were separately scored. RESULTS Tumor cell TIM3 expression was the highest in ACs (p < 0.001), whereas elevated tumor cell GITR levels were characteristic for both ACs and SCLCs (p < 0.001 and p = 0.011, respectively). OX40L expression of tumor cells was considerably lower in ACs (vs. SCLCs; p < 0.001). Tumor cell VISTA expression was consistently low in LNENs, with no significant differences across histological subtypes. ACs were the least immunogenic tumors concerning immune cell abundance (p < 0.001). Immune cell VISTA and GITR expressions were also significantly lower in these intermediate-grade malignancies than in SCLCs or in LCNECs. Immune cell TIM3 and GITR expressions were associated with borderline prognostic significance in our multivariate model (p = 0.057 and p = 0.071, respectively). CONCLUSIONS LNEN subtypes have characteristic and widely divergent VISTA, OX40L, GITR, and TIM3 protein expressions. By shedding light on the different expression patterns of these immunotherapy targets, the current multicenter study provides support for the future implementation of novel immunotherapeutic approaches.
Collapse
Affiliation(s)
- Bence Ferencz
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Klára Török
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Orsolya Pipek
- Department of Physics of Complex Systems, Eotvos Lorand University, Budapest, Hungary
| | - János Fillinger
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Kristóf Csende
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
| | - András Lantos
- National Koranyi Institute of Pulmonology, Budapest, Hungary
| | - Radoslava Černeková
- Department of Pulmonary Diseases and Tuberculosis, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Marcel Mitták
- Surgical Clinic, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Jozef Škarda
- Medical Faculty, Institute of Clinical and Molecular Pathology, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Pathology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Patricie Delongová
- Department of Pathology, University Hospital Ostrava and Faculty of Medicine, University of Ostrava, Ostrava, Czech Republic
| | - Evelyn Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- Department of Clinical Pharmacology, National Institute of Oncology, Chest and Abdominal Tumors Chemotherapy "B", Budapest, Hungary
| | - Karin Schelch
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Center for Cancer Research, Medical University of Vienna, Vienna, Austria
| | - Christian Lang
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
- Division of Pulmonology, Department of Medicine II, Medical University of Vienna, Vienna, Austria
| | - Anna Solta
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Kristiina Boettiger
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Luka Brcic
- Diagnostic and Research Institute of Pathology, Medical University of Graz, Graz, Austria
| | - Jörg Lindenmann
- Division of Thoracic and Hyperbaric Surgery, Department of Surgery, Medical University of Graz, Graz, Austria
| | - Ferenc Rényi-Vámos
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- National Institute of Oncology and National Tumor Biology Laboratory, Budapest, Hungary
| | - Clemens Aigner
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Judit Berta
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
| | - Zsolt Megyesfalvi
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary
- National Koranyi Institute of Pulmonology, Budapest, Hungary
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria
| | - Balázs Döme
- Department of Thoracic Surgery, Semmelweis University and National Institute of Oncology, Budapest, Hungary.
- National Koranyi Institute of Pulmonology, Budapest, Hungary.
- Department of Thoracic Surgery, Comprehensive Cancer Center Vienna, Medical University of Vienna, Waehringer Guertel 18-20, 1090, Vienna, Austria.
- Department of Translational Medicine, Lund University, Lund, Sweden.
| |
Collapse
|
28
|
Nishizaki D, Kurzrock R, Miyashita H, Adashek JJ, Lee S, Nikanjam M, Eskander RN, Patel H, Botta GP, Nesline MK, Pabla S, Conroy JM, DePietro P, Sicklick JK, Kato S. Viewing the immune checkpoint VISTA: landscape and outcomes across cancers. ESMO Open 2024; 9:102942. [PMID: 38503143 PMCID: PMC10966162 DOI: 10.1016/j.esmoop.2024.102942] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Revised: 12/18/2023] [Accepted: 02/16/2024] [Indexed: 03/21/2024] Open
Abstract
BACKGROUND Optimizing immune checkpoint inhibitor (ICI) therapy may require identification of co-targetable checkpoint pathways via immune profiling. Herein, we analyzed the transcriptomic expression and clinical correlates of V-domain immunoglobulin suppressor of T-cell activation (VISTA), a promising targetable checkpoint. PATIENTS AND METHODS RNA sequencing was carried out on 514 tissues reflecting diverse advanced/metastatic cancers. Expression of eight immune checkpoint markers [lymphocyte-activation gene 3 (LAG-3), tumor necrosis factor receptor superfamily 14 (TNFRSF14), programmed cell death protein 1 (PD-1), programmed death-ligand 1 (PD-L1), programmed death-ligand 2 (PD-L2), B- and T-lymphocyte attenuator (BTLA), T-cell immunoglobulin and mucin domain-containing protein 3 (TIM-3), cytotoxic T-lymphocyte antigen 4 (CTLA-4)], in addition to VISTA, was analyzed, along with clinical outcomes. RESULTS High VISTA RNA expression was observed in 32% of tumors (66/514) and was the most common highly expressed checkpoint among the nine assessed. High VISTA expression was independently correlated with high BTLA, TIM-3, and TNFRSF14, and with a diagnosis of pancreatic, small intestine, and stomach cancer. VISTA transcript levels did not correlate with overall survival (OS) from metastatic/advanced disease in the pan-cancer cohort or with immunotherapy outcome (progression-free survival and OS from the start of ICI) in 217 ICI-treated patients. However, in ICI-treated pancreatic cancer patients (n = 16), median OS was significantly shorter (from immunotherapy initiation) for the high- versus not-high-VISTA groups (0.28 versus 1.21 years) (P = 0.047); in contrast, VISTA levels were not correlated with OS in 36 pancreatic cancer patients who did not receive ICI. CONCLUSION High VISTA expression correlates with high BTLA, TIM-3, and TNFRSF14 checkpoint-related molecules and with poorer post-immunotherapy survival in pancreatic cancer, consistent with prior literature indicating that VISTA is prominently expressed on CD68+ macrophages in pancreatic cancers and requiring validation in larger prospective studies. Immunomic analysis may be important for individualized precision immunotherapy.
Collapse
Affiliation(s)
- D Nishizaki
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla.
| | - R Kurzrock
- MCW Cancer Center and Genomic Sciences and Precision Medicine Center, Medical College of Wisconsin, Milwaukee, USA; WIN Consortium, Paris, France
| | - H Miyashita
- Dartmouth Cancer Center, Hematology and Medical Oncology, Lebanon
| | - J J Adashek
- Department of Oncology, The Sidney Kimmel Comprehensive Cancer Center, The Johns Hopkins Hospital, Baltimore
| | - S Lee
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - M Nikanjam
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - R N Eskander
- Center for Personalized Cancer Therapy and Division of Gynecologic Oncology, Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, Moores Cancer Center, La Jolla
| | - H Patel
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | - G P Botta
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla
| | | | | | | | | | - J K Sicklick
- Division of Surgical Oncology, Department of Surgery, Center for Personalized Cancer Therapy, University of California San Diego, La Jolla, USA
| | - S Kato
- Center for Personalized Cancer Therapy and Division of Hematology and Oncology, Department of Medicine, University of California San Diego, Moores Cancer Center, La Jolla.
| |
Collapse
|
29
|
Li S, Wang G, Ren Y, Liu X, Wang Y, Li J, Liu H, Yang J, Xing J, Zhang Y, He C, Xu S, Hou X, Li N. Expression and function of VISTA on myeloid cells. Biochem Pharmacol 2024; 222:116100. [PMID: 38428824 DOI: 10.1016/j.bcp.2024.116100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 02/04/2024] [Accepted: 02/27/2024] [Indexed: 03/03/2024]
Abstract
V-domain containing Ig Suppressor of T cell Activation (VISTA) is predominantly expressed on myeloid cells and functions as a ligand/receptor/soluble molecule. In inflammatory responses and immune responses, VISTA regulates multiple functions of myeloid cells, such as chemotaxis, phagocytosis, T cell activation. Since inflammation and immune responses are critical in many diseases, VISTA is a promising therapeutic target. In this review, we will describe the expression and function of VISTA on different myeloid cells, including neutrophils, monocytes, macrophages, dendritic cells (DCs), myeloid-derived suppressor cells (MDSCs). In addition, we will discuss whether the functions of VISTA on these cells impact the disease processing.
Collapse
Affiliation(s)
- Siyu Li
- Health Science Center, Ningbo University, Ningbo, China.
| | - Geng Wang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yan Ren
- Health Science Center, Ningbo University, Ningbo, China.
| | - Xinyue Liu
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yixuan Wang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Jianing Li
- Health Science Center, Ningbo University, Ningbo, China.
| | - Hua Liu
- Health Science Center, Ningbo University, Ningbo, China.
| | - Jiaqiang Yang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Jingjun Xing
- Health Science Center, Ningbo University, Ningbo, China.
| | - Yanru Zhang
- Health Science Center, Ningbo University, Ningbo, China.
| | - Canxia He
- Health Science Center, Ningbo University, Ningbo, China.
| | - Suling Xu
- Department of Dermatology, the First Affiliated Hospital of Ningbo University, Ningbo, China.
| | - Xin Hou
- Health Science Center, Ningbo University, Ningbo, China.
| | - Na Li
- Health Science Center, Ningbo University, Ningbo, China; Department of Dermatology, the First Affiliated Hospital of Ningbo University, Ningbo, China.
| |
Collapse
|
30
|
Lin Y, Choukrani G, Dubbel L, Rockstein L, Freile JA, Qi Y, Wiersma V, Zhang H, Koch KW, Ammatuna E, Schuringa JJ, van Meerten T, Huls G, Bremer E. VISTA drives macrophages towards a pro-tumoral phenotype that promotes cancer cell phagocytosis yet down-regulates T cell responses. Exp Hematol Oncol 2024; 13:35. [PMID: 38553748 PMCID: PMC10979580 DOI: 10.1186/s40164-024-00501-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 03/13/2024] [Indexed: 04/02/2024] Open
Abstract
BACKGROUND VISTA is a well-known immune checkpoint in T cell biology, but its role in innate immunity is less established. Here, we investigated the role of VISTA on anticancer macrophage immunity, with a focus on phagocytosis, macrophage polarization and concomitant T cell activation. METHODS Macrophages, differentiated from VISTA overexpressed THP-1 cells and cord blood CD34+ cell-derived monocytes, were used in phagocytosis assay using B lymphoma target cells opsonized with Rituximab. PBMC-derived macrophages were used to assess the correlation between phagocytosis and VISTA expression. qRT-PCR, flow cytometry, and enzyme-linked immunosorbent assay were performed to analyze the impact of VISTA on other checkpoints and M1/M2-like macrophage biology. Additionally, flow cytometry was used to assess the frequency of CD14+ monocytes expressing VISTA in PBMCs from 65 lymphoma patients and 37 healthy donors. RESULTS Ectopic expression of VISTA in the monocytic model cell line THP-1 or in primary monocytes triggered differentiation towards the macrophage lineage, with a marked increase in M2-like macrophage-related gene expression and decrease in M1-like macrophage-related gene expression. VISTA expression in THP-1 and monocyte-derived macrophages strongly downregulated expression of SIRPα, a prominent 'don't eat me' signal, and augmented phagocytic activity of macrophages against cancer cells. Intriguingly, expression of VISTA's extracellular domain alone sufficed to trigger phagocytosis in ∼ 50% of cell lines, with those cell lines also directly binding to recombinant human VISTA, indicating ligand-dependent and -independent mechanisms. Endogenous VISTA expression was predominantly higher in M2-like macrophages compared to M0- or M1-like macrophages, with a positive correlation observed between VISTA expression in M2c macrophages and their phagocytic activity. VISTA-expressing macrophages demonstrated a unique cytokine profile, characterized by reduced IL-1β and elevated IL-10 secretion. Furthermore, VISTA interacted with MHC-I and downregulated its surface expression, leading to diminished T cell activation. Notably, VISTA surface expression was identified in monocytes from all lymphoma patients but was less prevalent in healthy donors. CONCLUSIONS Collectively, VISTA expression associates with and drives M2-like activation of macrophages with a high phagocytic capacity yet a decrease in antigen presentation capability to T cells. Therefore, VISTA is a negative immune checkpoint regulator in macrophage-mediated immune suppression.
Collapse
Affiliation(s)
- Yusheng Lin
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Ghizlane Choukrani
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Lena Dubbel
- Faculty VI, School of Medicine and Health Sciences, Department for human Medicine, Carl von Ossietzky Universität Oldenburg, University Clinic for Gynecology, Oldenburg, Germany
| | - Lena Rockstein
- Faculty VI, School of Medicine and Health Sciences, Department for human Medicine, Carl von Ossietzky Universität Oldenburg, University Clinic for Gynecology, Oldenburg, Germany
| | - Jimena Alvarez Freile
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Yuzhu Qi
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Valerie Wiersma
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Hao Zhang
- Institute of Precision Cancer Medicine and Pathology, Jinan University Medical College, Guangzhou, Guangdong, China
| | - Karl-Wilhelm Koch
- Faculty VI, School of Medicine and Health Sciences, Dept. of Neuroscience, Carl von Ossietzky Universität Oldenburg, Oldenburg, Germany
| | - Emanuele Ammatuna
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Jan Jacob Schuringa
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Tom van Meerten
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Gerwin Huls
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands
| | - Edwin Bremer
- Department of Hematology, University Medical Center Groningen (UMCG), University of Groningen, Groningen, 9713 EZ, The Netherlands.
| |
Collapse
|
31
|
Sun C, He Y, Wang G, Zhang G, Zhang Y, Shen H, Hu L, Sun Y, Jiang B, Wang X, Yuan K, Min W, Wang L, Sun H, Xiao Y, Yang P. Design, Synthesis, and Antitumor Activity Evaluation of Novel VISTA Small Molecule Inhibitors. J Med Chem 2024; 67:3590-3605. [PMID: 38412237 DOI: 10.1021/acs.jmedchem.3c02039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/29/2024]
Abstract
VISTA (V-domain Ig suppressor of T cell activation) is a novel immune checkpoint protein and represents a promising target for cancer immunotherapy. Here, we report the design, synthesis, and evaluation of a series of methoxy-pyrimidine-based VISTA small molecule inhibitors with potent antitumor activity. By employing molecular docking and microscale thermophoresis (MST) assay, we identified a lead compound A1 that binds to VISTA protein with high affinity and optimized its structure. A4 was then obtained, which exhibited the strongest binding ability to VISTA protein, with a KD value of 0.49 ± 0.20 μM. In vitro, A4 significantly activated peripheral blood mononuclear cells (PBMCs) induced the release of cytokines such as IFN-γ and enhanced the cytotoxicity of PBMCs against tumor cells. In vivo, A4 displayed potent antitumor activity and synergized with PD-L1 antibody to enhance the therapeutic effect against cancer. These results suggest that compound A4 is an effective VISTA small molecule inhibitor, providing a basis for the future development of VISTA-targeted drugs.
Collapse
Affiliation(s)
- Chengliang Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yuling He
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Gefei Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Guoyu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yu Zhang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Hao Shen
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Lingrong Hu
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Yanze Sun
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Binjian Jiang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Xiao Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Kai Yuan
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Wenjian Min
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Liping Wang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| | - Haopeng Sun
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
| | - Yibei Xiao
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Pharmacology, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing 401135, China
| | - Peng Yang
- State Key Laboratory of Natural Medicines and Jiangsu Key Laboratory of Drug Design and Optimization, China Pharmaceutical University, Nanjing 210009, China
- Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing 211198, China
- Institute of Innovative Drug Discovery and Development, China Pharmaceutical University, Nanjing 211198, China
| |
Collapse
|
32
|
Wang F, Fu K, Wang Y, Pan C, Wang X, Liu Z, Yang C, Zheng Y, Li X, Lu Y, To KKW, Xia C, Zhang J, Shi Z, Hu Z, Huang M, Fu L. Small-molecule agents for cancer immunotherapy. Acta Pharm Sin B 2024; 14:905-952. [PMID: 38486980 PMCID: PMC10935485 DOI: 10.1016/j.apsb.2023.12.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 11/28/2023] [Accepted: 12/06/2023] [Indexed: 03/17/2024] Open
Abstract
Cancer immunotherapy, exemplified by the remarkable clinical benefits of the immune checkpoint blockade and chimeric antigen receptor T-cell therapy, is revolutionizing cancer therapy. They induce long-term tumor regression and overall survival benefit in many types of cancer. With the advances in our knowledge about the tumor immune microenvironment, remarkable progress has been made in the development of small-molecule drugs for immunotherapy. Small molecules targeting PRR-associated pathways, immune checkpoints, oncogenic signaling, metabolic pathways, cytokine/chemokine signaling, and immune-related kinases have been extensively investigated. Monotherapy of small-molecule immunotherapeutic drugs and their combinations with other antitumor modalities are under active clinical investigations to overcome immune tolerance and circumvent immune checkpoint inhibitor resistance. Here, we review the latest development of small-molecule agents for cancer immunotherapy by targeting defined pathways and highlighting their progress in recent clinical investigations.
Collapse
Affiliation(s)
- Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Yujue Wang
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Zeyu Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| | - Ying Zheng
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Xiaopeng Li
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Yu Lu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong 999077, China
| | - Chenglai Xia
- Affiliated Foshan Maternity and Child Healthcare Hospital, Southern Medical University, Foshan 528000, China
| | - Jianye Zhang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target & Clinical Pharmacology, NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| | - Zhi Shi
- Department of Cell Biology & Institute of Biomedicine, National Engineering Research Center of Genetic Medicine, MOE Key Laboratory of Tumor Molecular Biology, Guangdong Provincial Key Laboratory of Bioengineering Medicine, College of Life Science and Technology, Jinan University, Guangzhou 510632, China
| | - Zeping Hu
- School of Pharmaceutical Sciences, Tsinghua-Peking Center for Life Sciences, Beijing Frontier Research Center for Biological Structure, Tsinghua University, Beijing 100084, China
| | - Min Huang
- State Key Laboratory of Drug Research, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou 510060, China
| |
Collapse
|
33
|
Shah V, Panchal V, Shah A, Vyas B, Agrawal S, Bharadwaj S. Immune checkpoint inhibitors in metastatic melanoma therapy (Review). MEDICINE INTERNATIONAL 2024; 4:13. [PMID: 38410760 PMCID: PMC10895472 DOI: 10.3892/mi.2024.137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Accepted: 01/26/2024] [Indexed: 02/28/2024]
Abstract
An increase in the incidence of melanoma has been observed in recent decades, which poses a significant challenge due to its poor prognosis in the advanced and metastatic stages. Previously, chemotherapy and high doses of interleukin-2 were available treatments for melanoma; however, they offered limited survival benefits and were associated with severe toxicities. The treatment of metastatic melanoma has been transformed by new developments in immunotherapy. Immune checkpoint inhibitors (ICIs), monoclonal antibodies that target cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4), programmed cell death protein 1 (PD-1) and its ligand, PDL-1, have emerged as promising therapeutic options. Commonly used ICIs, such as ipilimumab, nivolumab and pembrolizumab, have been found to be associated with an improved median overall survival, recurrence-free survival and response rates compared to traditional chemotherapies. Combination therapies involving different types of ICIs, such as anti-PD1 with anti-CTLA-4, have further enhanced the overall survival and response rates by targeting various phases of T-cell activation. Additionally, the development of novel biomarkers has facilitated the assessment of responses to ICI therapy, with tissue and serum-based prognostic and predictive biomarkers now available. The increased response observed with ICIs also provides potential for immune-related adverse effects on various organ systems. Further research is required to evaluate the efficacy and safety of various combinations of ICIs, while ongoing clinical trials explore the potential of newer ICIs. Concerns regarding the development of resistance to ICIs also warrant attention. The present review summarizes and discusses the advent of ICIs with a marked significant breakthrough in the treatment of metastatic melanoma, providing improved outcomes compared to traditional therapies.
Collapse
Affiliation(s)
- Vedant Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Viraj Panchal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Abhi Shah
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Bhavya Vyas
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Siddharth Agrawal
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| | - Sanket Bharadwaj
- Department of Medicine, Smt. N.H.L. Municipal Medical College and Sardar Vallabhbhai Patel Institute of Medical Sciences and Research (SVPISMR), Ahmedabad, Gujarat 380058, India
| |
Collapse
|
34
|
Wang CW, Biswas PK, Islam A, Chen MK, Chueh PJ. The Use of Immune Regulation in Treating Head and Neck Squamous Cell Carcinoma (HNSCC). Cells 2024; 13:413. [PMID: 38474377 DOI: 10.3390/cells13050413] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/20/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Immunotherapy has emerged as a promising new treatment modality for head and neck cancer, offering the potential for targeted and effective cancer management. Squamous cell carcinomas pose significant challenges due to their aggressive nature and limited treatment options. Conventional therapies such as surgery, radiation, and chemotherapy often have limited success rates and can have significant side effects. Immunotherapy harnesses the power of the immune system to recognize and eliminate cancer cells, and thus represents a novel approach with the potential to improve patient outcomes. In the management of head and neck squamous cell carcinoma (HNSCC), important contributions are made by immunotherapies, including adaptive cell therapy (ACT) and immune checkpoint inhibitor therapy. In this review, we are focusing on the latter. Immune checkpoint inhibitors target proteins such as programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) to enhance the immune response against cancer cells. The CTLA-4 inhibitors, such as ipilimumab and tremelimumab, have been approved for early-stage clinical trials and have shown promising outcomes in terms of tumor regression and durable responses in patients with advanced HNSCC. Thus, immune checkpoint inhibitor therapy holds promise in overcoming the limitations of conventional therapies. However, further research is needed to optimize treatment regimens, identify predictive biomarkers, and overcome potential resistance mechanisms. With ongoing advancements in immunotherapy, the future holds great potential for transforming the landscape of oral tumor treatment and providing new hope for patients.
Collapse
Affiliation(s)
- Che-Wei Wang
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Pulak Kumar Biswas
- Institute of Molecular Medicine, National Cheng Kung University, Tainan 70101, Taiwan
| | - Atikul Islam
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| | - Mu-Kuan Chen
- Department of Otorhinolaryngology-Head and Neck Surgery, Changhua Christian Hospital, Changhua 50006, Taiwan
| | - Pin Ju Chueh
- Institute of Biomedical Sciences, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
35
|
Hosseinkhani N, Hemmat N, Baghbani E, Baghbanzadeh A, Kazemi T, Mokhtarzadeh A, Jafarlou M, Amin Doustvandi M, Baradaran B. Dual silencing of tumor-intrinsic VISTA and CTLA-4 stimulates T-cell mediated immune responses and inhibits MCF7 breast cancer development. Gene 2024; 896:148043. [PMID: 38042220 DOI: 10.1016/j.gene.2023.148043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2023] [Revised: 11/16/2023] [Accepted: 11/28/2023] [Indexed: 12/04/2023]
Abstract
BACKGROUND As inhibitory immune checkpoint molecules, cytotoxic T-lymphocyte-associated antigen-4 (CTLA-4) and V-domain Ig suppressor of T-cell activation (VISTA) can be expressed in tumoral cells and facilitate immune evasion of tumoral cells. Herein, we studied the significance of tumor-intrinsic CTLA-4 and VISTA silencing in tumor development and inflammatory factors expression in a co-culture system with MCF7 and T-cells. METHODS MCF7 cells were transfected with 60 pmol of CTLA-siRNA, VISTA-siRNA, and dual VISTA-/CTLA-4-siRNA. The MTT assay was performed to study the effect of CTLA-4 and VISTA knockdown on the viability of MCF7 cells. Colony formation and wound-healing assays were performed to investigate the effect of CTLA-4 and VISTA silencing on the clonogenicity and migration of MCF7 cells. Flow cytometry was used to study the significance of CTLA-4 and VISTA knockdown on the apoptosis and cell cycle of MCF7 cells. Also, a co-culture system with MCF7 and T-cells was developed to study the expression levels of IL-2, IFN-γ, TNF-α, TGF-β, and IL-10 following CTLA-4 and VISTA knockdown. The expression levels of caspase3, Bax, Bcl2, and MMP-9 were also investigated using quantitative real-time PCR. Finally, the TCGA Breast Cancer and GSE45827 datasets were analyzed to study the potential prognostic values of VISTA and CTLA-4, their expression difference in luminal A breast cancer and non-tumoral tissues, and their correlation in luminal A breast cancer tissues. RESULTS Combined knockdown of tumor-intrinsic VISTA and CTLA-4 is superior in upregulating IL-2, IFN-γ, and TNF-α, downregulating TGF-β and IL-10 in T lymphocytes. Also, the combined silencing arrests the cell cycle at the sub-G1 phase, decreases migration, inhibits clonogenicity, and reduces cell viability of MCF7 cells. This combined treatment upregulates caspase 9 and BAX and downregulates MMP-9 in MCF7 cells. Our in-silico results have demonstrated a significant positive correlation between CTLA-4 and VISTA in luminal A breast cancer. CONCLUSION The additive effect of the combined knockdown of tumor-intrinsic VISTA and CTLA-4 can substantially upregulate pro-inflammatory factors, downregulate anti-inflammatory factors, and inhibit tumor development in MCF7 cells. The significant positive correlation between VISTA and CTLA-4 in luminal A breast cancer might support the idea that a network of inhibitory immune checkpoint molecules regulates anti-tumoral immune responses; thus, combinational immune checkpoint molecules blockade can be suggested.
Collapse
Affiliation(s)
- Negar Hosseinkhani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nima Hemmat
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Elham Baghbani
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Amir Baghbanzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mahdi Jafarlou
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - 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.
| |
Collapse
|
36
|
Alaluf E, Shalamov MM, Sonnenblick A. Update on current and new potential immunotherapies in breast cancer, from bench to bedside. Front Immunol 2024; 15:1287824. [PMID: 38433837 PMCID: PMC10905744 DOI: 10.3389/fimmu.2024.1287824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2023] [Accepted: 01/12/2024] [Indexed: 03/05/2024] Open
Abstract
Impressive advances have been seen in cancer immunotherapy during the last years. Although breast cancer (BC) has been long considered as non-immunogenic, immunotherapy for the treatment of BC is now emerging as a new promising therapeutic approach with considerable potential. This is supported by a plethora of completed and ongoing preclinical and clinical studies in various types of immunotherapies. However, a significant gap between clinical oncology and basic cancer research impairs the understanding of cancer immunology and immunotherapy, hampering cancer therapy research and development. To exploit the accumulating available data in an optimal way, both fundamental mechanisms at play in BC immunotherapy and its clinical pitfalls must be integrated. Then, clinical trials must be critically designed with appropriate combinations of conventional and immunotherapeutic strategies. While there is room for major improvement, this updated review details the immunotherapeutic tools available to date, from bench to bedside, in the hope that this will lead to rethinking and optimizing standards of care for BC patients.
Collapse
Affiliation(s)
- Emmanuelle Alaluf
- Medical Oncology Clinic, Institut Jules Bordet, Université Libre de Bruxelles, Brussels, Belgium
- Department of Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Amir Sonnenblick
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
- Oncology Division, Tel Aviv Sourasky Medical Center, Tel Aviv, Israel
| |
Collapse
|
37
|
Burke KP, Chaudhri A, Freeman GJ, Sharpe AH. The B7:CD28 family and friends: Unraveling coinhibitory interactions. Immunity 2024; 57:223-244. [PMID: 38354702 PMCID: PMC10889489 DOI: 10.1016/j.immuni.2024.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Revised: 01/17/2024] [Accepted: 01/17/2024] [Indexed: 02/16/2024]
Abstract
Immune responses must be tightly regulated to ensure both optimal protective immunity and tolerance. Costimulatory pathways within the B7:CD28 family provide essential signals for optimal T cell activation and clonal expansion. They provide crucial inhibitory signals that maintain immune homeostasis, control resolution of inflammation, regulate host defense, and promote tolerance to prevent autoimmunity. Tumors and chronic pathogens can exploit these pathways to evade eradication by the immune system. Advances in understanding B7:CD28 pathways have ushered in a new era of immunotherapy with effective drugs to treat cancer, autoimmune diseases, infectious diseases, and transplant rejection. Here, we discuss current understanding of the mechanisms underlying the coinhibitory functions of CTLA-4, PD-1, PD-L1:B7-1 and PD-L2:RGMb interactions and less studied B7 family members, including HHLA2, VISTA, BTNL2, and BTN3A1, as well as their overlapping and unique roles in regulating immune responses, and the therapeutic potential of these insights.
Collapse
Affiliation(s)
- Kelly P Burke
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA; Department of Immunology, Harvard Medical School, Boston, MA 02115, USA
| | - Apoorvi Chaudhri
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Gordon J Freeman
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA 02115, USA
| | - Arlene H Sharpe
- Department of Immunology, Harvard Medical School, Boston, MA 02115, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA; Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Di JW, Wang YX, Ma RX, Luo ZJ, Chen WT, Liu WM, Yuan DY, Zhang YY, Wu YH, Chen CP, Liu J. Repositioning baloxavir marboxil as VISTA agonist that ameliorates experimental asthma. Cell Biol Toxicol 2024; 40:12. [PMID: 38340268 PMCID: PMC10858940 DOI: 10.1007/s10565-024-09852-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
V-type immunoglobulin domain-containing suppressor of T-cell activation (VISTA), a novel negative checkpoint regulator, plays an essential role in allergic pulmonary inflammation in mice. Treatment with a VISTA agonistic antibody could significantly improve asthma symptoms. Thus, for allergic asthma treatment, VISTA targeting may be a compelling approach. In this study, we examined the functional mechanism of VISTA in allergic pulmonary inflammation and screened the FDA-approved drugs for VISTA agonists. By using mass cytometry (CyTOF), we found that VISTA deficiency primarily increased lung macrophage infiltration in the OVA-induced asthma model, accompanied by an increased proportion of M1 macrophages (CD11b+F4/80+CD86+) and a decreased proportion of M2 macrophages (CD11b+F4/80+CD206+). Further in vitro studies showed that VISTA deficiency promoted M1 polarization and inhibited M2 polarization of bone marrow-derived macrophages (BMDMs). Importantly, we discovered baloxavir marboxil (BXM) as a VISTA agonist by virtual screening of FDA-approved drugs. The surface plasmon resonance (SPR) assays revealed that BXM (KD = 1.07 µM) as well as its active form, baloxavir acid (BXA) (KD = 0.21 µM), could directly bind to VISTA with high affinity. Notably, treatment with BXM significantly ameliorated asthma symptoms, including less lung inflammation, mucus secretion, and the generation of Th2 cytokines (IL-5, IL-13, and IL-4), which were dramatically attenuated by anti-VISTA monoclonal antibody treatment. BXM administration also reduced the pulmonary infiltration of M1 macrophages and raised M2 macrophages. Collectively, our study indicates that VISTA regulates pulmonary inflammation in allergic asthma by regulating macrophage polarization and baloxavir marboxil, and an old drug might be a new treatment for allergic asthma through targeting VISTA.
Collapse
Affiliation(s)
- Jian-Wen Di
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Yi-Xin Wang
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Rui-Xue Ma
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Zhi-Jie Luo
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Wen-Ting Chen
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Wan-Mei Liu
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Ding-Yi Yuan
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Yu-Ying Zhang
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Yin-Hao Wu
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China
| | - Cai-Ping Chen
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.
- Chongqing Innovation Institute of China Pharmaceutical University, Chongqing, 401135, China.
| | - Jun Liu
- New Drug Screening Center, China Pharmaceutical University, Nanjing, 210009, China.
| |
Collapse
|
39
|
Kim TK, Han X, Hu Q, Vandsemb EN, Fielder CM, Hong J, Kim KW, Mason EF, Plowman RS, Wang J, Wang Q, Zhang JP, Badri T, Sanmamed MF, Zheng L, Zhang T, Alawa J, Lee SW, Zeidan AM, Halene S, Pillai MM, Chandhok NS, Lu J, Xu ML, Gore SD, Chen L. PD-1H/VISTA mediates immune evasion in acute myeloid leukemia. J Clin Invest 2024; 134:e164325. [PMID: 38060328 PMCID: PMC10836799 DOI: 10.1172/jci164325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 12/06/2023] [Indexed: 02/02/2024] Open
Abstract
Acute myeloid leukemia (AML) presents a pressing medical need in that it is largely resistant to standard chemotherapy as well as modern therapeutics, such as targeted therapy and immunotherapy, including anti-programmed cell death protein (anti-PD) therapy. We demonstrate that programmed death-1 homolog (PD-1H), an immune coinhibitory molecule, is highly expressed in blasts from the bone marrow of AML patients, while normal myeloid cell subsets and T cells express PD-1H. In studies employing syngeneic and humanized AML mouse models, overexpression of PD-1H promoted the growth of AML cells, mainly by evading T cell-mediated immune responses. Importantly, ablation of AML cell-surface PD-1H by antibody blockade or genetic knockout significantly inhibited AML progression by promoting T cell activity. In addition, the genetic deletion of PD-1H from host normal myeloid cells inhibited AML progression, and the combination of PD-1H blockade with anti-PD therapy conferred a synergistic antileukemia effect. Our findings provide the basis for PD-1H as a potential therapeutic target for treating human AML.
Collapse
Affiliation(s)
- Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine
- Vanderbilt Center for Immunobiology, and
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
- Vanderbilt Ingram Cancer Center, Nashville, Tennessee, USA
- Section of Medical Oncology
- Section of Hematology, Department of Medicine, and
| | - Xue Han
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Pelotonia Institute for Immuno-Oncology, OSUCCC–James Cancer Hospital
- Department of Microbial Infection and Immunity, The Ohio State University, Columbus, Ohio, USA
| | - Qianni Hu
- Division of Hematology/Oncology, Department of Medicine
| | - Esten N. Vandsemb
- Department of Acute Medicine, Oslo University Hospital, Oslo, Norway
| | | | - Junshik Hong
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | | | - Emily F. Mason
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - R. Skipper Plowman
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center
| | - Jun Wang
- Department of Pathology, New York University Grossman School of Medicine, New York, New York, USA
| | - Qi Wang
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China
| | - Jian-Ping Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Ti Badri
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Miguel F. Sanmamed
- Division of Immunology and Immunotherapy, CIMA, Universidad de Navarra, Pamplona, Spain
| | - Linghua Zheng
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
- Pelotonia Institute for Immuno-Oncology, OSUCCC–James Cancer Hospital
| | - Tianxiang Zhang
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Jude Alawa
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Sang Won Lee
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| | | | | | | | - Namrata S. Chandhok
- Sylvester Comprehensive Cancer Center, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Jun Lu
- Department of Genetics and
| | - Mina L. Xu
- Department of Pathology, Yale University School of Medicine, New Haven, Connecticut, USA
| | - Steven D. Gore
- Section of Hematology, Department of Medicine, and
- National Cancer Institute, Cancer Therapy Evaluation Program, Investigational Drug Branch, Bethesda, Maryland, USA
| | - Lieping Chen
- Section of Medical Oncology
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
40
|
Cao Y, Yu K, Zhang Z, Gu Y, Gu Y, Li W, Zhang W, Shen Z, Xu J, Qin J. Blockade of V-domain immunoglobulin suppressor of T-cell activation reprograms tumour-associated macrophages and improves efficacy of PD-1 inhibitor in gastric cancer. Clin Transl Med 2024; 14:e1578. [PMID: 38356419 PMCID: PMC10867598 DOI: 10.1002/ctm2.1578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2023] [Revised: 01/21/2024] [Accepted: 01/24/2024] [Indexed: 02/16/2024] Open
Abstract
BACKGROUND AND AIMS In gastric cancer, the response rate of programmed cell death protein-1 (PD-1) inhibitor is far from satisfactory, indicating additional nonredundant pathways might hamper antitumour immunity. V-domain immunoglobulin suppressor of T-cell activation (VISTA) has been reported in several malignancies as a novel immune-checkpoint. Nevertheless, the role of VISTA in gastric cancer still remains obscure. Our purpose is to explore the clinical significance and potential mechanism of VISTA in affecting gastric cancer patients' survival and immunotherapeutic responsiveness. METHODS Our study recruited eight independent cohorts with a total of 1403 gastric cancer patients. Immunohistochemistry, multiplex immunofluorescence, flow cytometry or intracellular flow cytometry, quantitative polymerase chain reaction, western blotting, fluorescence-activated cell sorting, magnetic-activated cell sorting, smart-seq2, in vitro cell co-culture and ex vivo tumour inhibition assays were applied to investigate the clinical significance and potential mechanism of VISTA in gastric cancer. RESULTS VISTA was predominantly expressed on tumour-associated macrophages (TAMs), and indicated poor clinical outcomes and inferior immunotherapeutic responsiveness. VISTA+ TAMs showed a mixed phenotype. Co-culture of TAMs and CD8+ T cells indicated that VISTA+ TAMs attenuated effective function of CD8+ T cells. Blockade of VISTA reprogrammed TAMs to a proinflammatory phenotype, reactivated CD8+ T cells and promoted apoptosis of tumour cells. Moreover, blockade of VISTA could also enhance the efficacy of PD-1 inhibitor, suggesting that blockade of VISTA might synergise with PD-1 inhibitor in gastric cancer. CONCLUSIONS Our data revealed that VISTA was an immune-checkpoint associated with immunotherapeutic resistance. Blockade of VISTA reprogrammed TAMs, promoted T-cell-mediated antitumour immunity, and enhanced efficacy of PD-1 inhibitor, which might have implications in the treatment of gastric cancer.
Collapse
Affiliation(s)
- Yifan Cao
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Kuan Yu
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Zihao Zhang
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yun Gu
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Yichao Gu
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Wandi Li
- Department of ImmunologySchool of Basic Medical Sciences, Fudan UniversityShanghaiChina
| | - Weijuan Zhang
- Department of ImmunologySchool of Basic Medical Sciences, Fudan UniversityShanghaiChina
| | - Zhenbin Shen
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| | - Jiejie Xu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical Sciences, Fudan UniversityShanghaiChina
| | - Jing Qin
- Department of General SurgeryZhongshan Hospital, Fudan UniversityShanghaiChina
| |
Collapse
|
41
|
Wright QG, Sinha D, Wells JW, Frazer IH, Gonzalez Cruz JL, Leggatt GR. Peritumoral administration of immunomodulatory antibodies as a triple combination suppresses skin tumor growth without systemic toxicity. J Immunother Cancer 2024; 12:e007960. [PMID: 38296598 PMCID: PMC10831460 DOI: 10.1136/jitc-2023-007960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
BACKGROUND Skin cancers, particularly keratinocyte cancers, are the most commonly diagnosed tumors. Although surgery is often effective in early-stage disease, skin tumors are not always easily accessible, can reoccur and have the ability to metastasize. More recently, immunotherapies, including intravenously administered checkpoint inhibitors, have been shown to control some skin cancers, but with off-target toxicities when used in combination. Our study investigated whether peritumoral administration of an antibody combination targeting PD-1, 4-1BB (CD137) and VISTA might control skin tumors and lead to circulating antitumor immunity without off-target toxicity. METHODS The efficacy of combination immunotherapy administered peritumorally or intravenously was tested using transplantable tumor models injected into mouse ears (primary tumors) or subcutaneously in flank skin (secondary tumors). Changes to the tumor microenvironment were tracked using flow cytometry while tumor-specific, CD8 T cells were identified through enzyme-linked immunospot (ELISPOT) assays. Off-target toxicity of the combination immunotherapy was assessed via serum alanine aminotransferase ELISA and histological analysis of liver sections. RESULTS The data showed that local administration of antibody therapy eliminated syngeneic murine tumors transplanted in the ear skin at a lower dose than required intravenously, and without measured hepatic toxicity. Tumor elimination was dependent on CD8 T cells and was associated with an increased percentage of CD8 T cells expressing granzyme B, KLRG1 and Eomes, and a decreased population of CD4 T cells including CD4+FoxP3+ cells in the treated tumor microenvironment. Importantly, untreated, distal tumors regressed following antibody treatment of a primary tumor, and immune memory prevented growth of subcutaneous flank tumors administered 50 days after regression of a primary tumor. CONCLUSIONS Together, these data suggest that peritumoral immunotherapy for skin tumors offers advantages over conventional intravenous delivery, allowing antibody dose sparing, improved safety and inducing long-term systemic memory. Future clinical trials of immunotherapy for primary skin cancer should focus on peritumoral delivery of combinations of immune checkpoint antibodies.
Collapse
Affiliation(s)
- Quentin G Wright
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Debottam Sinha
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - James W Wells
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Ian H Frazer
- Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
42
|
Rezagholizadeh F, Tajik F, Talebi M, Taha SR, Shariat Zadeh M, Farhangnia P, Hosseini HS, Nazari A, Mollazadeh Ghomi S, Kamrani Mousavi SM, Haeri Moghaddam N, Khorramdelazad H, Joghataei MT, Safari E. Unraveling the potential of CD8, CD68, and VISTA as diagnostic and prognostic markers in patients with pancreatic ductal adenocarcinoma. Front Immunol 2024; 15:1283364. [PMID: 38357542 PMCID: PMC10865497 DOI: 10.3389/fimmu.2024.1283364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Accepted: 01/15/2024] [Indexed: 02/16/2024] Open
Abstract
Introduction Pancreatic cancer is a truculent disease with limited treatment options and a grim prognosis. Immunotherapy has shown promise in treating various types of cancer, but its effectiveness in pancreatic cancer has been lacking. As a result, it is crucial to identify markers associated with immunological pathways in order to improve the treatment outcomes for this deadly cancer. The purpose of this study was to investigate the diagnostic and prognostic significance of three markers, CD8, CD68, and VISTA, in pancreatic ductal adenocarcinoma (PDAC), the most common subtype of pancreatic cancer. Methods We analyzed gene expression data from Gene Expression Omnibus (GEO) database using bioinformatics tools. We also utilized the STRING online tool and Funrich software to study the protein-protein interactions and transcription factors associated with CD8, CD68, and VISTA. In addition, tissue microarray (TMA) and immunohistochemistry (IHC) staining were performed on 228 samples of PDAC tissue and 10 samples of normal pancreatic tissue to assess the expression levels of the markers. We then correlated these expression levels with the clinicopathological characteristics of the patients and evaluated their survival rates. Results The analysis of the GEO data revealed slightly elevated levels of VISTA in PDAC samples compared to normal tissues. However, there was a significant increase in CD68 expression and a notable reduction in CD8A expression in pancreatic cancer. Further investigation identified potential protein-protein interactions and transcription factors associated with these markers. The IHC staining of PDAC tissue samples showed an increased expression of VISTA, CD68, and CD8A in pancreatic cancer tissues. Moreover, we found correlations between the expression levels of these markers and certain clinicopathological features of the patients. Additionally, the survival analysis revealed that high expression of CD8 was associated with better disease-specific survival and progression-free survival in PDAC patients. Conclusion These findings highlight the potential of CD8, CD68, and VISTA as diagnostic and prognostic indicators in PDAC.
Collapse
Affiliation(s)
- Fereshteh Rezagholizadeh
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Fatemeh Tajik
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Morteza Talebi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Genetics and Molecular Biology, Faculty of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Human and Animal Cell Bank, Iranian Biological Resource Center (IBRC), Tehran, Iran
| | - Seyed Reza Taha
- Oncopathology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | | | - Pooya Farhangnia
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Hamideh Sadat Hosseini
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Aram Nazari
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Shabnam Mollazadeh Ghomi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Immunology Board for Transplantation and Cell-Based Therapeutics (ImmunoTACT), Universal Scientific Education and Research Network (USERN), Tehran, Iran
- Department of Pathology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Seyede Mahtab Kamrani Mousavi
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Niloofar Haeri Moghaddam
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Medical Nanotechnology, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Hossein Khorramdelazad
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Mohammad Taghi Joghataei
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Elahe Safari
- Department of Immunology, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
43
|
Zhang K, Zakeri A, Alban T, Dong J, Ta HM, Zalavadia AH, Branicky A, Zhao H, Juric I, Husich H, Parthasarathy PB, Rupani A, Drazba JA, Chakraborty AA, Ching-Cheng Huang S, Chan T, Avril S, Wang LL. VISTA promotes the metabolism and differentiation of myeloid-derived suppressor cells by STAT3 and polyamine-dependent mechanisms. Cell Rep 2024; 43:113661. [PMID: 38175754 PMCID: PMC10851928 DOI: 10.1016/j.celrep.2023.113661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2023] [Revised: 10/20/2023] [Accepted: 12/20/2023] [Indexed: 01/06/2024] Open
Abstract
Myeloid-derived suppressor cells (MDSCs) impair antitumor immune responses. Identifying regulatory circuits during MDSC development may bring new opportunities for therapeutic interventions. We report that the V-domain suppressor of T cell activation (VISTA) functions as a key enabler of MDSC differentiation. VISTA deficiency reduced STAT3 activation and STAT3-dependent production of polyamines, which causally impaired mitochondrial respiration and MDSC expansion. In both mixed bone marrow (BM) chimera mice and myeloid-specific VISTA conditional knockout mice, VISTA deficiency significantly reduced tumor-associated MDSCs but expanded monocyte-derived dendritic cells (DCs) and enhanced T cell-mediated tumor control. Correlated expression of VISTA and arginase-1 (ARG1), a key enzyme supporting polyamine biosynthesis, was observed in multiple human cancer types. In human endometrial cancer, co-expression of VISTA and ARG1 on tumor-associated myeloid cells is associated with poor survival. Taken together, these findings unveil the VISTA/polyamine axis as a central regulator of MDSC differentiation and warrant therapeutically targeting this axis for cancer immunotherapy.
Collapse
Affiliation(s)
- Keman Zhang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Amin Zakeri
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Tyler Alban
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Juan Dong
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Hieu M Ta
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Ajay H Zalavadia
- Imaging Core Facility, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Andrelie Branicky
- Imaging Core Facility, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Haoxin Zhao
- Imaging Core Facility, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Ivan Juric
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Hanna Husich
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Prerana B Parthasarathy
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Amit Rupani
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Judy A Drazba
- Imaging Core Facility, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Abhishek A Chakraborty
- Department of Cancer Biology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Stanley Ching-Cheng Huang
- Department of Pathology, University Hospitals Cleveland Medical Center, and Case Western Reserve University School of Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Timothy Chan
- Center for Immunotherapy and Precision Immuno-Oncology, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA
| | - Stefanie Avril
- Department of Pathology, University Hospitals Cleveland Medical Center, and Case Western Reserve University School of Medicine, Cleveland, OH, USA; Case Comprehensive Cancer Center, Cleveland, OH, USA
| | - Li Lily Wang
- Department of Translational Hematology and Oncology Research, Cleveland Clinic Foundation, 9500 Euclid Avenue, Cleveland, OH, USA.
| |
Collapse
|
44
|
Wang J, Zhao Y, Liao P, Huang S, Huang Y, Chen S, Li Y, Zhong L. Immune checkpoint expression patterns on T cell subsets in light-chain amyloidosis: VISTA, PD-1, and TIGIT as potential therapeutic targets. BLOOD SCIENCE 2024; 6:e00181. [PMID: 38226018 PMCID: PMC10789457 DOI: 10.1097/bs9.0000000000000181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 12/21/2023] [Indexed: 01/17/2024] Open
Abstract
Amyloid light chain (AL) amyloidosis is a rare plasma cell dyscrasia with dismal prognosis. This study aims to investigate the T-cell immune checkpoint expression patterns in systemic AL amyloidosis and its relationship with clinicobiological traits. We examined the frequencies of V-domain immunoglobulin suppressor of T cell activation+ (VISTA+), programmed cell death 1+ (PD-1+), T cell immunoglobulin and mucin-domain-containing-3+ (Tim-3+), T cell immunoreceptor with Ig and ITIM domains+ (TIGIT+) T cells in peripheral blood (PB) and bone marrow (BM) from 19 patients with newly diagnosed AL amyloidosis. Patients with AL amyloidosis had significantly higher percentages of VISTA+ and PD-1+ T cells in PB than healthy individuals (HIs), with no statistical differences in BM. The percentages of some double-positive T cells in PB were also considerably higher in AL amyloidosis than those in HIs. Additionally, the patients with renal involvement had more PD-1+ and TIGIT+ T cells than the patients without, and PD-1+CD3+%, PD-1+CD4+%, PD-1+Treg% were positively correlated with 24-hour proteinuria levels. Furthermore, the AL amyloidosis patients had higher counts of PD-1+ Treg in PB than multiple myeloma (MM) patients, while the MM patients had higher counts of TIGIT+ T cells than AL amyloidosis patients. Collectively, this is the first report of elevated proportions of VISTA+ and PD-1+ T cells in PB of AL amyloidosis patients, indicating an immunosuppressive milieu, and the increased PD-1+ and TIGIT+ T cells were associated with renal damage. VISTA, PD-1, and TIGIT may be potential targets for reversing T-cell exhaustion in AL amyloidosis.
Collapse
Affiliation(s)
- Jinghua Wang
- Department of Hematology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Yujie Zhao
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Pengjun Liao
- Department of Hematology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shuxin Huang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Youxue Huang
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Shaohua Chen
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Yangqiu Li
- Key Laboratory for Regenerative Medicine of Ministry of Education, Institute of Hematology, School of Medicine, Jinan University, Guangzhou, China
| | - Liye Zhong
- Department of Hematology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
45
|
Vesely MD, Kidacki M, Gaule P, Gupta S, Chan NNN, Han X, Yeung JT, Chen L. Immune Inhibitory Molecule PD-1 Homolog (VISTA) Colocalizes with CD11b Myeloid Cells in Melanoma and Is Associated with Poor Outcomes. J Invest Dermatol 2024; 144:106-115.e4. [PMID: 37562584 DOI: 10.1016/j.jid.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 08/12/2023]
Abstract
Tumors evade immunity through the overexpression of immune inhibitory molecules in the tumor microenvironment such as PD-L1/B7-H1. An immune inhibitory molecule named PD-1 homolog (also known as V-domain Ig-containing suppressor of T cell activation [VISTA]) functions to control both T cells and myeloid cells. Current clinical trials using anti-VISTA-blocking agents for treatment of cancer are ongoing. We sought to determine the extent of VISTA expression in primary cutaneous melanomas (n = 190), identify the critical cell types expressing VISTA, and correlate its expression with PD-L1 expression using multiplexed quantitative immunofluorescence. Within the tumor subcompartments, VISTA is most highly expressed on CD11b myeloid cells, and PD-L1 is most highly expressed on CD68 myeloid cells in our melanoma cohort. There is little correlation between VISTA and PD-L1 expression intensity, suggesting that individual tumors have distinct immunosuppressive tumor microenvironments. High levels of VISTA expression on CD11b myeloid cells but not PD-L1 expression were associated with greater melanoma recurrence and greater all-cause mortality. Our findings suggest that cell-specific VISTA expression may be a negative prognostic biomarker for melanoma and a future potential therapeutic target.
Collapse
Affiliation(s)
- Matthew D Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.
| | - Michal Kidacki
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Swati Gupta
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Nay Nwe Nyein Chan
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Xue Han
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Microbial Infection and Immunity, The Ohio State University College of Medicine, Columbus, Ohio, USA; Pelotonia Institute for Immuno-Oncology, The Ohio State University College of Medicine, Columbus, Ohio, USA
| | - Jacky T Yeung
- Department of Neurosurgery, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lieping Chen
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Medicine (Medical Oncology), Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
46
|
Iadonato S, Ovechkina Y, Lustig K, Cross J, Eyde N, Frazier E, Kabi N, Katz C, Lance R, Peckham D, Sridhar S, Talbaux C, Tihista I, Xu M, Guillaudeux T. A highly potent anti-VISTA antibody KVA12123 - a new immune checkpoint inhibitor and a promising therapy against poorly immunogenic tumors. Front Immunol 2023; 14:1311658. [PMID: 38152397 PMCID: PMC10751915 DOI: 10.3389/fimmu.2023.1311658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Accepted: 11/20/2023] [Indexed: 12/29/2023] Open
Abstract
Background Immune checkpoint therapies have led to significant breakthroughs in cancer patient treatment in recent years. However, their efficiency is variable, and resistance to immunotherapies is common. VISTA is an immune-suppressive checkpoint inhibitor of T cell response belonging to the B7 family and a promising novel therapeutic target. VISTA is expressed in the immuno-suppressive tumor microenvironment, primarily by myeloid lineage cells, and its genetic knockout or antibody blockade restores an efficient antitumor immune response. Methods Fully human monoclonal antibodies directed against VISTA were produced after immunizing humanized Trianni mice and single B cell sequencing. Anti-VISTA antibodies were evaluated for specificity, cross-reactivity, monocyte and T cell activation, Fc-effector functions, and antitumor efficacy using in vitro and in vivo models to select the KVA12123 antibody lead candidate. The pharmacokinetics and safety profiles of KVA12123 were evaluated in cynomolgus monkeys. Results Here, we report the development of a clinical candidate anti-VISTA monoclonal antibody, KVA12123. KVA12123 showed high affinity binding to VISTA through a unique epitope distinct from other clinical-stage anti-VISTA monoclonal antibodies. This clinical candidate demonstrated high specificity against VISTA with no cross-reactivity detected against other members of the B7 family. KVA12123 blocked VISTA binding to its binding partners. KVA12123 induced T cell activation and demonstrated NK-mediated monocyte activation. KVA12123 treatment mediated strong single-agent antitumor activity in several syngeneic tumor models and showed enhanced efficacy in combination with anti-PD-1 treatment. This clinical candidate was engineered to improve its pharmacokinetic characteristics and reduce Fc-effector functions. It was well-tolerated in preclinical toxicology studies in cynomolgus monkeys, where hematology, clinical chemistry evaluations, and clinical observations revealed no indicators of toxicity. No cytokines associated with cytokine release syndrome were elevated. Conclusion These results establish that KVA12123 is a promising drug candidate with a distinct but complementary mechanism of action of the first generation of immune checkpoint inhibitors. This antibody is currently evaluated alone and in combination with pembrolizumab in a Phase 1/2 open-label clinical trial in patients with advanced solid tumors.
Collapse
|
47
|
Sabaie H, Tamimi P, Gharesouran J, Salkhordeh Z, Asadi MR, Sharifi-Bonab M, Shirvani-Farsani Z, Taheri M, Sayad A, Rezazadeh M. Expression analysis of inhibitory B7 family members in Alzheimer's disease. Metab Brain Dis 2023; 38:2563-2572. [PMID: 37665469 DOI: 10.1007/s11011-023-01274-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 08/04/2023] [Indexed: 09/05/2023]
Abstract
Alzheimer's disease (AD) is a global health problem due to its complexity, which frequently makes the development of treatment methods extremely difficult. Therefore, new methodologies are necessary to investigate the pathophysiology of AD and to treat AD. The interaction of immune modulation and neurodegeneration has added new dimensions in current knowledge of AD etiology and offers an attractive opportunity for the discovery of novel biomarkers and therapies. Using quantitative polymerase chain reaction, we compared the expression levels of inhibitory B7 family members (B7-1, B7-2, B7-H1, B7-DC, B7-H3, B7-H4, B7-H5, B7-H7, and ILDR2), as immune regulators, in the peripheral blood of late-onset AD (LOAD) patients (n = 50) and healthy individuals (n = 50). The levels of B7-2, B7-H4, ILDR2, and B7-DC expression were significantly higher in-patient blood samples than in control blood samples. Furthermore, we discovered a substantial positive correlation between all gene expression levels. In addition, the current study indicated that ILDR2, B7-H4, B7-2, and B7-DC might serve as diagnostic biomarkers to identify LOAD patients from healthy persons. The present work provides additional evidence for the significance of inhibitory B7 family members to the etiology of LOAD.
Collapse
Affiliation(s)
- Hani Sabaie
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parham Tamimi
- School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Jalal Gharesouran
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zoha Salkhordeh
- Department of Medical Genetics, Faculty of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Mohammad Reza Asadi
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mirmohsen Sharifi-Bonab
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Zeinab Shirvani-Farsani
- Department of Cell and Molecular Biology, Faculty of Life Sciences and Technology, Shahid Beheshti University, Tehran, Iran
| | - Mohammad Taheri
- Institute of Human Genetics, Jena University Hospital, Jena, Germany.
- Urology and Nephrology Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Arezou Sayad
- Department of Medical Genetics, Loghman Hakim Hospital, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Maryam Rezazadeh
- Clinical Research Development Unit of Tabriz Valiasr Hospital, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Medical Genetics, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
48
|
Liu J, Lin WP, Xiao Y, Yang QC, Bushabu Fidele N, Yu HJ, Sun ZJ. VISTA blockade alleviates immunosuppression of MDSCs in oral squamous cell carcinoma. Int Immunopharmacol 2023; 125:111128. [PMID: 37907049 DOI: 10.1016/j.intimp.2023.111128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/17/2023] [Accepted: 10/22/2023] [Indexed: 11/02/2023]
Abstract
V-domain Ig suppressor of T-cell activation (VISTA) is a novel immune checkpoint regulator that can inhibit T cell-mediated antitumor immunity. Although the use of anti-VISTA monoclonal antibody has demonstrated encouraging outcomes in the therapy of various malignancies, its specific impact and underlying mechanisms in oral squamous cell carcinoma (OSCC) remain to be explored. In this work, we analyzed human OSCC tissue microarrays, human peripheral blood mononuclear cells, and immunocompetent transgenic mouse models to investigate the relationship between high VISTA expression and markers of myeloid-derived immunosuppressive cells (MDSCs; CD11b, CD33, Arginase-1), tumor-associated macrophages (CD68, CD163, CD206), and T cell function (CD8, PD-L1, Granzyme B). In OSCC, we discovered that VISTA was highly expressed and stably expressed in MDSCs. Furthermore, we established a mouse OSCC orthotopic xenograft tumor model to investigate the impact of VISTA blockade on the tumor microenvironment. We found that VISTA blockade reduces the immunosuppressive microenvironment and delays tumor growth. This is achieved by suppressing the quantity and function of MDSCs while boosting the function of tumor-infiltrating T cells. Our research indicated that VISTA expressed by MDSCs has a crucial function in the progression of OSCC and that VISTA blockade therapy is a promising immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Jie Liu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China
| | - Wen-Ping Lin
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China
| | - Yao Xiao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China
| | - Qi-Chao Yang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China
| | - Nyimi Bushabu Fidele
- The National Key Laboratory of Basic Science of Stomatology of Kinshasa University, School of Dental Medicine, University of Kinshasa, Kinshasa B.P. 834 KIN XI, Democratic Republic of Congo
| | - Hai-Jun Yu
- Department of Radiation and Medical Oncology, Hubei Province Cancer Clinical Study Center, Hubei Key Laboratory of Tumor Biological Behaviors, Zhongnan Hospital of Wuhan University, Wuhan 430071, PR China.
| | - Zhi-Jun Sun
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, Key Laboratory of Oral Biomedicine Ministry of Education, Hubei Key Laboratory of Stomatology, School & Hospital of Stomatology, Frontier Science Center for Immunology and Metabolism, Wuhan University, Wuhan 430079, PR China; Department of Oral Maxillofacial-Head Neck Oncology, School and Hospital of Stomatology, Wuhan University, Wuhan 430079, PR China.
| |
Collapse
|
49
|
Wang B, Ou Z, Zhong W, Huang L, Liao W, Sheng Y, Guo Z, Chen J, Yang W, Chen K, Huang X, Yang T, Lin T, Huang J. Effective Antitumor Immunity Can Be Triggered by Targeting VISTA in Combination with a TLR3-Specific Adjuvant. Cancer Immunol Res 2023; 11:1656-1670. [PMID: 37847894 DOI: 10.1158/2326-6066.cir-23-0117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2023] [Revised: 07/31/2023] [Accepted: 10/16/2023] [Indexed: 10/19/2023]
Abstract
Resistance to anti-PD-1/PD-L1 treatment is often associated with accumulation of intratumoral inhibitory macrophages. V-domain immunoglobulin suppressor of T-cell activation (VISTA) is a nonredundant immune checkpoint that can induce both T-cell and myeloid-cell immunosuppression. In this study, we found that high levels of VISTA+ immune cells were associated with advanced stage bladder cancer and predicted poor survival in patients. A combination of high infiltration of VISTA+ immune cells and PD-L1+ immune cells or PD-1+ T cells predicted the worst survival. Flow cytometry and multiplex immunofluorescence analyses confirmed that VISTA expression was higher in macrophages than in T cells or neutrophils, and only VISTA+CD163+ macrophage density predicted poor prognosis in patients with bladder cancer. Toll-like receptor (TLR) agonists are known to trigger the innate immune response in macrophages. We found that the VISTA-specific mAb 13F3 augmented the ability of a TLR3-specific adjuvant to induce macrophage activation in vitro. In the MB49 syngeneic mouse model of bladder cancer, treatment with 13F3 curbed tumor growth and prolonged survival when combined with a TLR3-specific adjuvant. The combination treatment reduced the intratumoral frequency of CD206+ anti-inflammatory macrophages and levels of the immunosuppressive molecule TGFβ1, but it upregulated expression of immunostimulatory molecules (Ifna, Ifnb, and Trail) and increased the CD8+ T cell/regulatory T-cell ratio. These findings indicate that elevated VISTA expression in immune cells, particularly macrophages, is associated with an unfavorable prognosis in patients with bladder cancer and suggest that targeting VISTA in combination with a TLR3-specific adjuvant has translational potential.
Collapse
Affiliation(s)
- Bo Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Ziwei Ou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Wenlong Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Lin Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Wenjian Liao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Yiyu Sheng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Zhixing Guo
- Department of Ultrasound, Sun Yat-Sen University Cancer Center, State Key Laboratory of Oncology in South China, Guangzhou, P.R. China
| | - Junyu Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Wenjuan Yang
- Department of Hematology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Ke Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Xiaodong Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Tenghao Yang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| | - Jian Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Guangdong-Hong Kong Joint Laboratory for RNA Medicine, Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-sen (Zhongshan) University, Guangzhou, P.R. China
| |
Collapse
|
50
|
Shekari N, Shanehbandi D, Kazemi T, Zarredar H, Baradaran B, Jalali SA. VISTA and its ligands: the next generation of promising therapeutic targets in immunotherapy. Cancer Cell Int 2023; 23:265. [PMID: 37936192 PMCID: PMC10631023 DOI: 10.1186/s12935-023-03116-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 10/27/2023] [Indexed: 11/09/2023] Open
Abstract
V-domain immunoglobulin suppressor of T cell activation (VISTA) is a novel negative checkpoint receptor (NCR) primarily involved in maintaining immune tolerance. It has a role in the pathogenesis of autoimmune disorders and cancer and has shown promising results as a therapeutic target. However, there is still some ambiguity regarding the ligands of VISTA and their interactions with each other. While V-Set and Immunoglobulin domain containing 3 (VSIG-3) and P-selectin glycoprotein ligand-1(PSGL-1) have been extensively studied as ligands for VISTA, the others have received less attention. It seems that investigating VISTA ligands, reviewing their functions and roles, as well as outcomes related to their interactions, may allow an understanding of their full functionality and effects within the cell or the microenvironment. It could also help discover alternative approaches to target the VISTA pathway without causing related side effects. In this regard, we summarize current evidence about VISTA, its related ligands, their interactions and effects, as well as their preclinical and clinical targeting agents.
Collapse
Affiliation(s)
- Najibeh Shekari
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Dariush Shanehbandi
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Tohid Kazemi
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Habib Zarredar
- Tuberculosis and Lung Disease Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Immunology, School of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Seyed Amir Jalali
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|