1
|
Dübbel L, Göken-Riebisch A, Koch KW. Intracellular and exosomal localization of the negative checkpoint regulator VISTA in immune cells. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119966. [PMID: 40262722 DOI: 10.1016/j.bbamcr.2025.119966] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 04/10/2025] [Accepted: 04/17/2025] [Indexed: 04/24/2025]
Abstract
Strategies in immunotherapy often target the immunosuppressive environment of tumor cells. One route of therapeutic interference could involve negative checkpoint regulators of which V-domain immunoglobulin (Ig)-containing suppressor of T-cell activation (VISTA) has raised more interest recently. The protein is expressed on the surface of tumor cells, T-lymphocytes, and antigen-presenting cells (APCs), but its intracellular expression pattern has not been investigated yet. We examined the intracellular distribution of VISTA and its possible role in translocation processes by immunofluorescence and Western blots. We analyzed the expression and localization of VISTA in murine bone marrow-derived macrophages (BMDMs), human monocyte-derived macrophages, and human T lymphocytes (Jurkat). We obtained different cell fractions and organelles of various cell types and analyzed for the presence of VISTA. Monitoring a VISTA-GFP fusion construct in transfected cell lines HL-60 and THP-1 confirmed VISTA localization in these cell lines. All used cell lines showed the colocalization of VISTA and several vesicle markers together with VISTA staining along microtubule fibers. Additionally, we found VISTA in secreted exosomes and have the first hints for nucleic expression in all tested cell lines. Therefore, the storage of VISTA in vesicles and its potential presence in nuclei resembles two other well-described checkpoint regulators, CTLA-4 and PD-L1, respectively. We conclude that VISTA storage in vesicles enables a fast response to immunogenic stimuli, which needs to be considered for inhibitory experiments. The localization of VISTA in exosomes suggests a signaling function to facilitate cell-cell communication. Furthermore, the VISTA expression in the nucleus proposes a transcriptional role.
Collapse
Affiliation(s)
- Lena Dübbel
- Division of Biochemistry, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany; University Clinic of Gynaecology and Obstetrics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany.
| | - Anna Göken-Riebisch
- University Clinic of Gynaecology and Obstetrics, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| | - Karl-Wilhelm Koch
- Division of Biochemistry, Carl von Ossietzky Universität Oldenburg, Ammerländer Heerstraße 114-118, 26129 Oldenburg, Germany
| |
Collapse
|
2
|
Ibrahim A, Mohamady Farouk Abdalsalam N, Liang Z, Kashaf Tariq H, Li R, O Afolabi L, Rabiu L, Chen X, Xu S, Xu Z, Wan X, Yan D. MDSC checkpoint blockade therapy: a new breakthrough point overcoming immunosuppression in cancer immunotherapy. Cancer Gene Ther 2025; 32:371-392. [PMID: 40140724 PMCID: PMC11976280 DOI: 10.1038/s41417-025-00886-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 02/25/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025]
Abstract
Despite the success of cancer immunotherapy in treating hematologic malignancies, their efficacy in solid tumors remains limited due to the immunosuppressive tumor microenvironment (TME), which is mainly formed by myeloid-derived suppressor cells (MDSCs). MDSCs not only exert potent immunosuppressive effects that hinder the success of immune checkpoint inhibitors (ICIs) and adaptive cellular therapies, but they also promote tumor advancement through non-immunological pathways, including promoting angiogenesis, driving epithelial-mesenchymal transition (EMT), and contributing to the establishment of pre-metastatic environments. While targeting MDSCs alone or in combination with conventional therapies has shown limited success, emerging evidence suggests that MDSC checkpoint blockade in combination with other immunotherapies holds great promise in overcoming both immunological and non-immunological barriers. In this review, we discussed the dual roles of MDSCs, with a particular emphasis on their underexplored checkpoints blockade strategies. We discussed the rationale behind combination strategies, their potential advantages in overcoming MDSC-mediated immunosuppression, and the challenges associated with their development. Additionally, we highlight future research directions aimed at optimizing combination immunotherapies to enhance cancer therapeutic effectiveness, particularly in solid tumor therapies where MDSCs are highly prevalent.
Collapse
Affiliation(s)
- Abdulrahman Ibrahim
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Nada Mohamady Farouk Abdalsalam
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Zihao Liang
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Hafiza Kashaf Tariq
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Rong Li
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
| | - Lukman O Afolabi
- Department of Pediatrics, Indiana University School of Medicine, 1234 Notre Dame Ave, South Bend, IN, 46617, USA
| | - Lawan Rabiu
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China
- University of Chinese Academy of Sciences, 100864, Beijing, China
| | - Xuechen Chen
- College of Pharmacy, Jinan University, 511436, Guangzhou, China.
| | - Shu Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, 518106, Shenzhen, China
| | - Zhiming Xu
- Cancer Center, Shenzhen Guangming District People's Hospital, 518106, Shenzhen, China.
| | - Xiaochun Wan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| | - Dehong Yan
- Guangdong Immune Cell Therapy Engineering and Technology Research Center, Center for Protein and Cell-Based Drugs, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, 518055, Shenzhen, China.
- University of Chinese Academy of Sciences, 100864, Beijing, China.
| |
Collapse
|
3
|
Ren X, Guo A, Geng J, Chen Y, Wang X, Zhou L, Shi L. Pan-cancer analysis of co-inhibitory molecules revealing their potential prognostic and clinical values in immunotherapy. Front Immunol 2025; 16:1544104. [PMID: 40196117 PMCID: PMC11973099 DOI: 10.3389/fimmu.2025.1544104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 03/03/2025] [Indexed: 04/09/2025] Open
Abstract
Background The widespread use of immune checkpoint inhibitors (anti-CTLA4 or PD-1) has opened a new chapter in tumor immunotherapy by providing long-term remission for patients. Unfortunately, however, these agents are not universally available and only a minority of patients respond to them. Therefore, there is an urgent need to develop novel therapeutic strategies targeting other co-inhibitory molecules. However, comprehensive information on the expression and prognostic value of co-inhibitory molecules, including co-inhibitory receptors and their ligands, in different cancers is not yet available. Methods We investigated the expression, correlation, and prognostic value of co-inhibitory molecules in different cancer types based on TCGA, UCSC Xena, TIMER, CellMiner datasets. We also examined the associations between the expression of these molecules and the extent of immune cell infiltration. Besides, we conducted a more in-depth study of VISTA. Result The results of differential expression analysis, correlation analysis, and drug sensitivity analysis suggest that CTLA4, PD-1, TIGIT, LAG3, TIM3, NRP1, VISTA, CD80, CD86, PD-L1, PD-L2, PVR, PVRL2, FGL1, LGALS9, HMGB1, SEMA4A, and VEGFA are associated with tumor prognosis and immune cell infiltration. Therefore, we believe that they are hopefully to serve as prognostic biomarkers for certain cancers. In addition, our analysis indicates that VISTA plays a complex role and its expression is related to TMB, MSI, cancer cell stemness, DNA/RNA methylation, and drug sensitivity. Conclusions These co-inhibitory molecules have the potential to serve as prognostic biomarkers and therapeutic targets for a broad spectrum of cancers, given their strong associations with key clinical metrics. Furthermore, the analysis results indicate that VISTA may represent a promising target for cancer therapy.
Collapse
Affiliation(s)
- Xiaoyu Ren
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Anjie Guo
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Jiahui Geng
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Yuling Chen
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Xue Wang
- School of Life Sciences, Chongqing University, Chongqing, China
| | - Lian Zhou
- Department of Head&Neck Cancer Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Lei Shi
- School of Life Sciences, Chongqing University, Chongqing, China
| |
Collapse
|
4
|
Luo Y, Yuan Y, Liu D, Peng H, Shen L, Chen Y. Targeting novel immune checkpoints in the B7-H family: advancing cancer immunotherapy from bench to bedside. Trends Cancer 2025:S2405-8033(25)00055-X. [PMID: 40113530 DOI: 10.1016/j.trecan.2025.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025]
Abstract
The B7-H family of immune checkpoint molecules is a crucial component of the immune regulatory network for tumors, offering new opportunities to modulate the tumor microenvironment (TME). The B7-H family - which includes B7-H2 (inducible T cell costimulatory ligand, ICOSL), B7-H3, B7-H4, B7-H5 (V-domain immunoglobulin suppressor of T cell activation, VISTA), B7-H6, and B7-H7 (HHLA2) - is known for its diverse roles in regulating innate and adaptive immunity. These molecules can exhibit co-stimulatory or co-inhibitory effects on T cells, influencing processes such as T cell activation, differentiation, and effector functions, and they are involved in the recruitment and polarization of various immune cells. This review explores the structural characteristics, receptor-ligand interactions, and signaling pathways associated with each B7-H family member. We also discuss the family's impact on tumor immunity and potential therapeutic strategies.
Collapse
Affiliation(s)
- Yiming Luo
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Ye Yuan
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Dan Liu
- Early Drug Development Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Haoxin Peng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing 102200, China.
| |
Collapse
|
5
|
Mi Y, Jiang P, Luan J, Feng L, Zhang D, Gao X. Peptide‑based therapeutic strategies for glioma: Current state and prospects. Peptides 2025; 185:171354. [PMID: 39922284 DOI: 10.1016/j.peptides.2025.171354] [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: 11/23/2024] [Revised: 01/21/2025] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
Glioma is a prevalent form of primary malignant central nervous system tumor, characterized by its cellular invasiveness, rapid growth, and the presence of the blood-brain barrier (BBB)/blood-brain tumor barrier (BBTB). Current therapeutic approaches, such as chemotherapy and radiotherapy, have shown limited efficacy in achieving significant antitumor effects. Therefore, there is an urgent demand for new treatments. Therapeutic peptides represent an innovative class of pharmaceutical agents with lower immunogenicity and toxicity. They are easily modifiable via chemical means and possess deep tissue penetration capabilities which reduce side effects and drug resistance. These unique pharmacokinetic characteristics make peptides a rapidly growing class of new therapeutics that have demonstrated significant progress in glioma treatment. This review outlines the efforts and accomplishments in peptide-based therapeutic strategies for glioma. These therapeutic peptides can be classified into four types based on their anti-tumor function: tumor-homing peptides, inhibitor/antagonist peptides targeting cell surface receptors, interference peptides, and peptide vaccines. Furthermore, we briefly summarize the results from clinical trials of therapeutic peptides in glioma, which shows that peptide-based therapeutic strategies exhibit great potential as multifunctional players in glioma therapy.
Collapse
Affiliation(s)
- Yajing Mi
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Pengtao Jiang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Jing Luan
- Institute of Basic and Translational Medicine, Xi'an Medical University, Xi'an, Shaanxi, China
| | - Lin Feng
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Dian Zhang
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China
| | - Xingchun Gao
- Institute of Basic Medical Sciences, School of Basic Medical Science, Xi'an Medical University, Xi'an, China; Shaanxi Key Laboratory of Brain Disorders, School of Basic Medical Science, Xi'an Medical University, Xi'an, China.
| |
Collapse
|
6
|
Jin S, Li T, Liu L, Gao T, Zhang T, Yuan D, Di J, Guo Z, Luo Z, Yuan H, Liu J. V-domain immunoglobulin suppressor of T-cell activation and programmed death receptor 1 dual checkpoint blockade enhances antitumour immunity and survival in glioblastoma. Br J Pharmacol 2025; 182:1306-1323. [PMID: 39626657 DOI: 10.1111/bph.17404] [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: 04/23/2024] [Revised: 09/30/2024] [Accepted: 10/19/2024] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND AND PURPOSE The current therapy cannot meet the needs of glioblastoma (GBM). V-domain immunoglobulin suppressor of T-cell activation (VISTA) is significantly up-regulated in GBM patients; however, its therapeutic potential in GBM is still unclear. EXPERIMENTAL APPROACH Flow cytometry was used to detect the expression of VISTA and the co-expression pattern of VISTA and programmed death receptor 1 (PD-1) on brain infiltrating lymphocytes of GBM mice. Monoclonal antibody therapy was used to evaluate the therapeutic effect of α-VISTA monotherapy and α-VISTA combined with α-PD-1 on GBM mice. Transcriptome analysis, flow cytometry, and immunofluorescence were used to detect changes of immune microenvironment in mouse brain tumours. Immunofluorescence and TCGA data analysis were used to further validate the combined treatment strategy on patient data. KEY RESULTS Compared with normal mice, the frequency of VISTA expression and co-expression of VISTA and PD-1 on tumour-infiltrating lymphocytes (TILs) in tumour-bearing mice was increased. Anti-VISTA monotherapy significantly up-regulated multiple immune stimulation-related pathways and moderately prolonged mouse survival time. Blocking the immune checkpoint VISTA and PD-1 significantly prolonged the survival time of mice and cured about 80% of the mice; CD8+ T cells played an important role in this process. In addition, we found that the expression of VISTA and PD-1 was significantly up-regulated in GBM patients by immunofluorescence, and patients with high expression of VISTA and PD-1 were associated with poor overall survival. This combination of blocking the immune checkpoint VISTA and PD-1 may achieve clinical transformation in GBM.
Collapse
Affiliation(s)
- Shasha Jin
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tao Li
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Liu Liu
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Ting Gao
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Tingting Zhang
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Dingyi Yuan
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jianwen Di
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhanying Guo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Zhijie Luo
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Haoliang Yuan
- Jiangsu Key Laboratory of Drug Discovery for Metabolic Disease, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| | - Jun Liu
- New Drug Screening and Pharmacodynamics Evaluation Center, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, China
| |
Collapse
|
7
|
Kidacki M, Cho C, Lopez-Giraldez F, Huang B, He J, Gaule P, Chen L, Vesely MD. PD-1H (VISTA) Expression in Cutaneous Squamous Cell Carcinoma Is Correlated with T-Cell Infiltration and Activation. J Invest Dermatol 2025:S0022-202X(25)00116-2. [PMID: 39983979 DOI: 10.1016/j.jid.2025.01.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Revised: 01/14/2025] [Accepted: 01/20/2025] [Indexed: 02/23/2025]
Abstract
Cutaneous squamous cell carcinoma (cSCC) is one of the most common human cancers, with an estimated death rate approaching or exceeding that of melanoma. Immune inhibitory receptor antagonism through the blockade of PD-1 or its ligand, PD-L1, has revolutionized the treatment of cSCC; however, approximately half of patients still fail to respond. The inhibitory receptor PD-1H (also known as VISTA) controls T-cell and myeloid cell functions in preclinical cancer studies. Currently, cancer clinical trials using anti-VISTA-blocking antibodies are ongoing. We sought to determine the extent of VISTA expression in cSCCs and to correlate its expression with PD-L1 expression. Using multiplexed quantitative immunofluorescence of primary cSCC tissues (n = 76), we found that VISTA was expressed in 48% of cSCCs and that PD-L1 was expressed in 58% of cSCCs. We found that high VISTA expression, more than PD-L1 expression, correlated with greater T-cell infiltration and activation, as measured by the proliferation marker Ki-67 and the cytotoxic marker granzyme B. Furthermore, there was no significant correlation between VISTA and PD-L1 coexpression within the same cSCCs, suggesting that individual tumors have distinct immunosuppressive microenvironments. These findings provide a rationale for the targeting of VISTA in cSCC immunotherapy.
Collapse
Affiliation(s)
- Michal Kidacki
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Christina Cho
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Francesc Lopez-Giraldez
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA; Yale Center for Genome Analysis (YCGA), Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Baozhu Huang
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Jianwei He
- Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Patricia Gaule
- Department of Pathology, Yale School of Medicine, New Haven, Connecticut, USA
| | - Lieping Chen
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Immunobiology, Yale School of Medicine, New Haven, Connecticut, USA; Department of Internal Medicine (Oncology), Yale School of Medicine, New Haven, Connecticut, USA
| | - Matthew D Vesely
- Department of Dermatology, Yale School of Medicine, New Haven, Connecticut, USA.
| |
Collapse
|
8
|
Struckmeier AK, Gosau M, Smeets R. Immunotherapeutic strategies beyond the PD-1/PD-L1 pathway in head and neck squamous cell carcinoma - A scoping review on current developments in agents targeting TIM-3, TIGIT, LAG-3, and VISTA. Oral Oncol 2025; 161:107145. [PMID: 39705929 DOI: 10.1016/j.oraloncology.2024.107145] [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: 10/10/2024] [Revised: 11/24/2024] [Accepted: 12/11/2024] [Indexed: 12/23/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) poses a considerable challenge due to its high incidence and mortality rates. Immunotherapy targeting PD-(L)1 emerges as a promising approach for HNSCC, as it has the potential to trigger a broad and long-lasting anti-tumor response. Nevertheless, the effectiveness of immunotherapy encounters hurdles, and only a small proportion of patients benefit, with many eventually experiencing relapse. Consequently, there is a pursuit of strategies to enhance overall treatment outcomes. Understanding the mechanisms driving resistance to PD-(L)1 inhibition and devising strategies to overcome these challenges are vital for advancing more effective treatments. Furthermore, gaining insights into the mechanisms of action and safety profiles of novel combination therapies is critical for their successful adoption in clinical practice. As a result, current research is dedicated to investigating various immunotherapeutic agents beyond the PD-1/PD-L1 axis. This review offers a comprehensive overview of the existing immunotherapy strategies in HNSCC with a focus on TIM-3, TIGIT, LAG-3, and VISTA. The aim is to lay a strong foundation for the continual advancement of therapies for HNSCC.
Collapse
Affiliation(s)
- Ann-Kristin Struckmeier
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany.
| | - Martin Gosau
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany
| | - Ralf Smeets
- Department of Oral and Maxillofacial Surgery, University Medical Center Hamburg-Eppendorf, Martinistraße 52, 20246 Hamburg, Germany; Department of Oral and Maxillofacial Surgery, Division of Regenerative Orofacial Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| |
Collapse
|
9
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
10
|
Zhao Y, Andoh T, Charles F, Reddy P, Paul K, Goar H, Durdana I, Golder C, Hardy A, Juntilla MM, Yang SR, Lin CY, Sagiv-Barfi I, Geller BS, Moore S, Thakkar D, Boyd-Kirkup JD, Peng Y, Ford JM, Telli ML, Zhang S, Kurian AW, West RB, Yue T, Lipchik AM, Snyder MP, Gruber JJ. VISTA-induced tumor suppression by a four amino acid intracellular motif. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.05.631401. [PMID: 39803490 PMCID: PMC11722267 DOI: 10.1101/2025.01.05.631401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/24/2025]
Abstract
VISTA is a key immune checkpoint receptor under investigation for cancer immunotherapy; however, its signaling mechanisms remain unclear. Here we identify a conserved four amino acid (NPGF) intracellular motif in VISTA that suppresses cell proliferation by constraining cell-intrinsic growth receptor signaling. The NPGF motif binds to the adapter protein NUMB and recruits Rab11 endosomal recycling machinery. We identify and characterize a class of triple-negative breast cancers with high VISTA expression and low proliferative index. In tumor cells with high VISTA levels, the NPGF motif sequesters NUMB at endosomes, which interferes with epidermal growth factor receptor (EGFR) trafficking and signaling to suppress tumor growth. These effects do not require canonical VISTA ligands, nor a functioning immune system. As a consequence of VISTA expression, EGFR receptor remains abnormally phosphorylated and cannot propagate ligand-induced signaling. Mutation of the VISTA NPGF domain reverts VISTA-induced growth suppression in multiple breast cancer mouse models. These results define a mechanism by which VISTA represses NUMB to control malignant epithelial cell growth and signaling. They also define distinct intracellular residues that are critical for VISTA-induced cell-intrinsic signaling that could be exploited to improve immunotherapy.
Collapse
Affiliation(s)
- Yan Zhao
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Tina Andoh
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Fatima Charles
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Priyanka Reddy
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Kristina Paul
- Departments of Genetics, Stanford University, Palo Alto, CA, 94305
| | - Harsh Goar
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Ishrat Durdana
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Caiden Golder
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Ashley Hardy
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | | | - Soo-Ryum Yang
- Department Pathology, Stanford University, Palo Alto, CA, 94305
| | - Chien-Yu Lin
- Department Pathology, Stanford University, Palo Alto, CA, 94305
| | | | | | - Stephen Moore
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| | - Dipti Thakkar
- Hummingbird Bioscience, 61 Science Park Road, #06-15/24, Singapore 117525
| | | | - Yan Peng
- Department of Pathology, UT Southwestern Medical Center, Dallas, TX, 75235
| | - James M. Ford
- Department Medicine, Stanford University, Palo Alto, CA, 94305
| | | | - Song Zhang
- Peter O’Donnell Jr. School of Public Health, UT Southwestern Medical Center, Dallas, TX, 75235
| | | | - Robert B. West
- Department Pathology, Stanford University, Palo Alto, CA, 94305
| | - Tao Yue
- Departments of Surgery and Immunology, Center for Organogenesis Research and Trauma, UT Southwestern Medical Center, Dallas, TX 75235
| | - Andrew M. Lipchik
- Eugene Applebaum College of Pharmacy and Health Science, Wayne State University, Detroit, MI, 48201
| | | | - Joshua J. Gruber
- Departments of Medicine and Molecular Biology, Cecil H. and Ida Green Center for Reproductive Sciences, UT Southwestern Medical Center, Dallas, TX, 75235
| |
Collapse
|
11
|
Timis T, Buruiana S, Dima D, Nistor M, Muresan XM, Cenariu D, Tigu AB, Tomuleasa C. Advances in Cell and Immune Therapies for Melanoma. Biomedicines 2025; 13:98. [PMID: 39857682 PMCID: PMC11761552 DOI: 10.3390/biomedicines13010098] [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: 11/28/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/27/2025] Open
Abstract
The incidence rate of cutaneous melanoma is on the rise worldwide, due to increased exposure to UV radiation, aging populations, and exposure to teratogen agents. However, diagnosis is more precise, and the increased number of new cases is related to the improved diagnosis tools. Despite better early diagnosis and better therapies, melanoma has remained a significant public health challenge because of its aggressive behavior and high potential for metastasis. In 2020, cutaneous melanoma constituted approximately 1.3% of all cancer deaths that occurred within the European Union, thereby highlighting the necessity for effective prevention, timely diagnosis, and sustainable treatment measures, especially as a growing number of cases occur among younger patients. Melanoma is regarded as one of the most inflamed cancers due to its high immune cell presence and strong response to immunotherapy, fueling the need for development of immune-driven innovative treatments. Approved therapies, including immune checkpoint inhibitors (e.g., anti-PD-1 and anti-CTLA-4), have notably improved survival rates in melanoma. However, the limitations of the PD-1/PD-L1 and CTLA-4 axes inhibitors, such as low response rates, treatment resistance, and toxicity, have driven the need for continued research and advancements in treatment strategies. Current clinical trials are exploring various combinations of immune checkpoint inhibitors with costimulatory receptor agonists, chemotherapy, targeted therapies, and other immunotherapies, with the goal of improving outcomes and reducing side effects for melanoma patients. Emerging approaches, including adoptive cell therapy with tumor-infiltrating lymphocytes (TILs) and oncolytic virotherapy, are showing promise. While CAR-T cell therapy has been less successful in melanoma compared to blood cancers, ongoing research is addressing challenges like the tumor microenvironment and antigen specificity. This review provides an overview of the requirement for advances in these medications, to mark a significant step forward in melanoma management, set to bring a fresh breath of hope for patients.
Collapse
Affiliation(s)
- Tanase Timis
- Department of Hematology, Iuliu Hațieganu University of Medicine and Pharmacy, 400347 Cluj-Napoca, Romania
- Department of Oncology, Bistrița Emergency Hospital, 420094 Bistrița, Romania
| | - Sanda Buruiana
- Department of Hematology, Nicolae Testemițanu University of Medicine and Pharmacy, MD-2004 Chisinau, Moldova
| | - Delia Dima
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
| | - Madalina Nistor
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ximena Maria Muresan
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Diana Cenariu
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Adrian-Bogdan Tigu
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| | - Ciprian Tomuleasa
- Department of Hematology, Ion Chiricuta Oncology Institute, 400015 Cluj-Napoca, Romania
- Department of Personalized Medicine and Rare Diseases, MEDFUTURE—Institute for Biomedical Research, “Iuliu Hațieganu” University of Medicine and Pharmacy, 400349 Cluj-Napoca, Romania
| |
Collapse
|
12
|
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
|
13
|
Arafat Hossain M. A comprehensive review of immune checkpoint inhibitors for cancer treatment. Int Immunopharmacol 2024; 143:113365. [PMID: 39447408 DOI: 10.1016/j.intimp.2024.113365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2024] [Revised: 09/28/2024] [Accepted: 10/05/2024] [Indexed: 10/26/2024]
Abstract
Immunology-based therapies are emerging as an effective cancer treatment, using the body's immune system to target tumors. Immune checkpoints, which regulate immune responses to prevent tissue damage and autoimmunity, are often exploited by cancer cells to avoid destruction. The discovery of checkpoint proteins like PD-1/PD-L1 and CTLA-4 was pivotal in developing cancer immunotherapy. Immune checkpoint inhibitors (ICIs) have shown great success, with FDA-approved drugs like PD-1 inhibitors (Nivolumab, Pembrolizumab, Cemiplimab), PD-L1 inhibitors (Atezolizumab, Durvalumab, Avelumab), and CTLA-4 inhibitors (Ipilimumab, Tremelimumab), alongside LAG-3 inhibitor Relatlimab. Research continues on new checkpoints like TIM-3, VISTA, B7-H3, BTLA, and TIGIT. Biomarkers like PDL-1 expression, tumor mutation burden, interferon-γ presence, microbiome composition, and extracellular matrix characteristics play a crucial role in predicting responses to immunotherapy with checkpoint inhibitors. Despite their effectiveness, not all patients experience the same level of benefit, and organ-specific immune-related adverse events (irAEs) such as rash or itching, colitis, diarrhea, hyperthyroidism, and hypothyroidism may occur. Given the rapid advancements in this field and the variability in patient outcomes, there is an urgent need for a comprehensive review that consolidates the latest findings on immune checkpoint inhibitors, covering their clinical status, biomarkers, resistance mechanisms, strategies to overcome resistance, and associated adverse effects. This review aims to fill this gap by providing an analysis of the current clinical status of ICIs, emerging biomarkers, mechanisms of resistance, strategies to enhance therapeutic efficacy, and assessment of adverse effects. This review is crucial to furthering our understanding of ICIs and optimizing their application in cancer therapy.
Collapse
Affiliation(s)
- Md Arafat Hossain
- Department of Pharmacy, Bangabandhu Sheikh Mujibur Rahman Science and Technology University, Gopalganj 8100, Bangladesh.
| |
Collapse
|
14
|
Duan Y, Ren X, Guo X, Xie J, Liu Z, Li L. VISTA in hematological malignancies: a review of the literature. Front Immunol 2024; 15:1466839. [PMID: 39742253 PMCID: PMC11685136 DOI: 10.3389/fimmu.2024.1466839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 11/29/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, tumor immunotherapy has become an active research area, with the emergence of immune checkpoint inhibitors (ICIs) revolutionizing immunotherapy. Clinical evidence indicates that programmed cell death protein 1 (PD-1) monoclonal antibodies and other drugs have remarkable therapeutic effects. V-domain Ig suppressor of T-cell activation (VISTA) is a new type of immune checkpoint receptor that is highly expressed in various tumors. It is co-expressed with PD-1, T-cell immunoglobulin domain, mucin domain-3 (Tim-3), T-cell immunoglobulin, and immunoreceptor tyrosine-based inhibitory motif domain (TIGIT) and is associated with prognosis, which suggests that it may be a target for immunotherapy. As an immune checkpoint receptor with no mature drugs, VISTA is highly expressed in acute myeloid leukemia (AML), multiple myeloma (MM), and other hematological malignancies; however, its pathogenic mechanism should be defined to better guide treatment.
Collapse
Affiliation(s)
- Yuanjia Duan
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Xiaotong Ren
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Xinyu Guo
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Jiayi Xie
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Zhaoyun Liu
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| | - Lijuan Li
- Department of Hematology, Tianjin Medical University General Hospital, Tianjin, China
- Tianjin Key Laboratory of Bone Marrow Failure and Malignant Hemopoietic Clone Control, Tianjin, China
- Tianjin Institute of Hematology, Tianjin, China
| |
Collapse
|
15
|
Yu X, Zhong M, Wang C, Shi Y, Xing C, Yu K, Lin Y, Ou R, Yang J, Zhu L. Effect of peripheral blood absolute monocyte count at admission on relapse-free survival in patients with idiopathic thrombotic thrombocytopenic purpura in remission. Front Med (Lausanne) 2024; 11:1463086. [PMID: 39736971 PMCID: PMC11683109 DOI: 10.3389/fmed.2024.1463086] [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: 07/11/2024] [Accepted: 11/29/2024] [Indexed: 01/01/2025] Open
Abstract
Introduction Peripheral monocytes have been reported to correlate with clinical outcomes in various types of malignancies. Previous reports have also shown that acute-phase thrombotic thrombocytopenic purpura (TTP) plasma could induce the activation of monocytes. However, the significance of peripheral blood absolute monocyte count (AMC) in idiopathic TTP remains an unanswered question. The primary aim of this retrospective study was to evaluate the prognostic value of AMC at admission in idiopathic TTP patients in remission. Methods A total of 37 patients with idiopathic TTP who survived their first episode of the disease and achieved remission following treatment were eligible for inclusion in the study. SPSS and Stata were used to analysis. Results There were 1 patient (2.7%) with low AMC (< 0.12 × 109/L), 27 patients (73.0%) with normal AMC (0.12-0.80 × 109/L), and 9 patients (24.3%) with high AMC (> 0.80 × 109/L) at admission. Ten (27.0%) of 37 patients in our cohort subsequently relapsed (1 in the low AMC group and 9 in the normal AMC group). Survival analysis showed that there was a trend of higher relapse-free survival (RFS) rate in patients having increased A MC (log-rank test, p = 0.026). Univariate analysis revealed that increased AMC at admission was significantly associated with higher RFS (hazard ratio = 0.12, 95% confidence interval: 0.02-0.62, p = 0.011). Discussion Our results suggest that increased AMC at admission could represent a predictor of higher RFS in TTP patients having survived their first episode of the disease and achieved remission following treatment.
Collapse
Affiliation(s)
- Xiaomin Yu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Mingzhu Zhong
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chen Wang
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yifen Shi
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chongyun Xing
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Kang Yu
- Department of Hematology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Ying Lin
- Department of Hematology, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Rongying Ou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Junjun Yang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Liqing Zhu
- Department of Clinical Laboratory, the First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
16
|
Wu L, Bai R, Zhang Y, Chen H, Wu J, Chen Z, Wang H, Zhao L. METTL3-VISTA axis-based combination immunotherapy for APC truncation colorectal cancer. J Immunother Cancer 2024; 12:e009865. [PMID: 39653551 PMCID: PMC11629097 DOI: 10.1136/jitc-2024-009865] [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: 06/13/2024] [Accepted: 11/17/2024] [Indexed: 12/12/2024] Open
Abstract
OBJECTIVE Although immune checkpoint blockade (ICB) therapy represents a bright spot in antitumor immunotherapy, its clinical benefits in colorectal cancer (CRC) are limited. Therefore, a new target for mediating CRC immunosuppression is urgently needed. Adenomatous polyposis coli (APC) mutations have been reported as early-stage characteristic events in CRC, but the role of truncated APC in the CRC immune microenvironment remains unclear and its clinical significance has yet to be explored. DESIGN Adenocarcinoma formation in the colon of the APCMin/+ mouse model, which displays features associated with the translation of truncated APC proteins, was induced by azoxymethane/dextran sodium sulfate. Multiplexed immunohistochemical consecutive staining on single slides and flow cytometry were used to explore the activation of immune cells and the expression of the immune checkpoint V-domain immunoglobulin suppressor of T-cell activation (VISTA) in the CRC tissues of APCWT and APCMin/+ mice. The construction of truncated APC vectors and an initial subserosal graft tumor mouse model was employed to mimic the tumor microenvironment (TME) during APC mutation. Methylated RNA immunoprecipitation-quantitative PCR assays were performed to investigate the N6-methyladenosine (m6A)-dependent transcriptional regulation of hypoxia-inducible factor-1 alpha (HIF1α) by methyltransferase-like protein 3 (METTL3). Mettl3fl/fl vil1-cre+/- mice were used to demonstrate that targeting METTL3 is a mediator that mitigates the deleterious effects of the APC978∆-HIF1α axis on antitumor immunity. A chimeric VISTA humanized mouse model was used to evaluate the drug efficacy of the VISTA-targeted compound onvatilimab. RESULTS We showed that APC978∆, a truncated APC protein, mediated overexpression of METTL3, resulting in m6A methylation of HIF1α messenger RNA and high expression of HIF1α. Furthermore, HIF1α promotes the migration of myeloid-derived suppressor cells to the TME by binding to the promoters of MCP-1 and MIF. In addition, HIF1α enhances the expression of the immune checkpoint VISTA on CRC cells, weakening tumor immune monitoring. CONCLUSIONS We elucidate that an underappreciated function of truncated APC in CRC is its ability to drive an immunosuppressive program that boosts tumor progression. Our work could provide a new perspective for the clinical application of immunotherapy in patients with CRC resistant to ICB therapy.
Collapse
Affiliation(s)
- Ling Wu
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Rui Bai
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Yujie Zhang
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Hao Chen
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Jianghua Wu
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Zetao Chen
- Department of Neurosurgery, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Hui Wang
- Department of Medical Oncology, Guangzhou Medical University Affiliated Cancer Hospital, Guangzhou, Guangdong, China
| | - Liang Zhao
- Department of Pathology, Shunde Hospital of Southern Medical University, Foshan, Guangdong, China
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Peng M, Lu X, Guo J, Yin X, Zhang J, Li X, Zou Y. A pair of promising immune checkpoints PSGL-1 and VISTA from immunotolerance to immunotherapy. Biomark Res 2024; 12:151. [PMID: 39617949 PMCID: PMC11610313 DOI: 10.1186/s40364-024-00693-8] [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: 09/19/2024] [Accepted: 11/17/2024] [Indexed: 01/25/2025] Open
Abstract
Immune checkpoints are crucial for regulating immune responses and maintaining self-tolerance, as they play a pivotal role in preventing autoimmunity and facilitating tumor immune evasion. This review concentrates on the immune checkpoint molecules PSGL-1 and VISTA. Both molecules are highly expressed in hematopoietic cells, including T cells and myeloid cells. VISTA functions both as a ligand on myeloid cells, where it regulates cytokine production, chemotaxis, and phagocytosis while promoting their differentiation into a tolerogenic phenotype and as a receptor on T cells, where it contributes to T cell quiescence. PSGL-1, which acts as a binding partner for VISTA, further inhibits T-cell activation and fosters tolerance within the acidic tumor microenvironment. Our review provides a comprehensive analysis of the structure, expression, and biological functions of PSGL-1 and VISTA and emphasizes their therapeutic potential in cancer treatment, autoimmune diseases, and transplantation. The dual role of these checkpoints in immune regulation presents novel opportunities for advancing cancer immunotherapy and developing new strategies for managing autoimmune conditions.
Collapse
Affiliation(s)
- Manqing Peng
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China
| | - Xiaofang Lu
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China
| | - Junshuang Guo
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China
| | - Xiangli Yin
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China
| | - Jing Zhang
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China
| | - Xin Li
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China
| | - Yizhou Zou
- Department of Immunology, School of Basic Medicine, Central South University, Changsha, Hunan, 410000, China.
| |
Collapse
|
18
|
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
|
19
|
Zhang RJ, Kim TK. VISTA-mediated immune evasion in cancer. Exp Mol Med 2024; 56:2348-2356. [PMID: 39482534 PMCID: PMC11612309 DOI: 10.1038/s12276-024-01336-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 08/04/2024] [Accepted: 08/11/2024] [Indexed: 11/03/2024] Open
Abstract
Over the past decade, V-domain immunoglobulin suppressor of T-cell activation (VISTA) has been established as a negative immune checkpoint molecule. Since the role of VISTA in inhibiting T-cell activation was described, studies have demonstrated other diverse regulatory functions in multiple immune cell populations. Furthermore, its relevance has been identified in human cancers. The role of VISTA in cancer immune evasion has been determined, but its mechanisms in the tumor microenvironment remain to be further elucidated. Understanding its contributions to cancer initiation, progression, and resistance to current treatments will be critical to its utility as a target for novel immunotherapies. Here, we summarize the current understanding of VISTA biology in cancer.
Collapse
Affiliation(s)
- Raymond J Zhang
- Medical Scientist Training Program, Vanderbilt University School of Medicine, Nashville, TN, USA
- Program in Cancer Biology, Vanderbilt University, Nashville, TN, USA
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Tae Kon Kim
- Division of Hematology/Oncology, Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA.
- Vanderbilt Ingram Cancer Center, Nashville, TN, 37232, USA.
| |
Collapse
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
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
|
24
|
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, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
- Committee on Clinical Pharmacology and Pharmacogenomics, University of Chicago; Chicago, USA
| | - Liangliang Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Xianbin Yu
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, University of Chicago; Chicago, USA
- Howard Hughes Medical Institute, University of Chicago; Chicago, USA
| | - Xuezhi Cao
- Guangzhou National Laboratory, Bio-Island; Guangzhou, China
| | - Jiaai Wang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Xiaona Huang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Kaiting Yang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Andras Piffko
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
- Department of Neurosurgery, University Medical Center Hamburg-Eppendorf; Hamburg, Germany
| | - Katherine Chen
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Stephen Y. Luo
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Emile Naccasha
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - 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, USA
| | - Chuan He
- Department of Chemistry, Department of Biochemistry and Molecular Biology, and Institute for Biophysical Dynamics, University of Chicago; Chicago, USA
- Howard Hughes Medical Institute, University of Chicago; Chicago, USA
| | - Yang-xin Fu
- Department of Basic Medical Science, Tsinghua University; Beijing, China
| | - Hua Laura Liang
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| | - Ralph R. Weichselbaum
- Department of Radiation and Cellular Oncology, University of Chicago; Chicago, USA
- Ludwig Center for Metastasis Research, University of Chicago; Chicago, USA
| |
Collapse
|
25
|
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
|
26
|
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
|
27
|
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
|
28
|
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
|
29
|
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
|
30
|
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
|
31
|
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
|
32
|
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
|
33
|
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
|
34
|
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
|
35
|
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
|
36
|
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
|
37
|
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
|
38
|
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
|
39
|
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
|
40
|
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
|
41
|
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
|
42
|
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
|
43
|
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
|
44
|
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
|
45
|
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
|
46
|
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
|
47
|
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: 13] [Impact Index Per Article: 13.0] [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
|
48
|
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
|
49
|
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 PMCID: PMC10930979 DOI: 10.3390/cells13050413] [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: 12/19/2023] [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; (C.-W.W.); (A.I.)
- 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; (C.-W.W.); (A.I.)
| | - 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; (C.-W.W.); (A.I.)
| |
Collapse
|
50
|
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
|