1
|
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
|
2
|
Hu L, Sun C, Yuan K, Yang P. Expression, regulation, and function of PD-L1 on non-tumor cells in the tumor microenvironment. Drug Discov Today 2024; 29:104181. [PMID: 39278561 DOI: 10.1016/j.drudis.2024.104181] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2024] [Revised: 09/03/2024] [Accepted: 09/11/2024] [Indexed: 09/18/2024]
Abstract
Antiprogrammed death ligand 1 (PD-L1) therapy is a leading immunotherapy, but only some patients with solid cancers benefit. Overwhelming evidence has revealed that PD-L1 is expressed on various immune cells in the tumor microenvironment (TME), including macrophages, dendritic cells, and regulatory T cells, modulating tumor immunity and influencing tumor progression. PD-L1 can also be located on tumor cell membranes as well as in exosomes and cytoplasm. Accordingly, the dynamic expression and various forms of PD-L1 might explain the therapy's limited efficacy and resistance. Herein a systematic summary of the expression of PD-L1 on different immune cells and their regulatory mechanisms is provided to offer a solid foundation for future studies.
Collapse
Affiliation(s)
- 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
| | - 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
| | - 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.
| | - 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
|
3
|
Zhao G, Li P, Suo Y, Li C, Yang S, Zhang Z, Wu Z, Shen C, Hu H. An integrated pan-cancer assessment of prognosis, immune infiltration, and immunotherapy response for B7 family using multi-omics data. Life Sci 2024; 353:122919. [PMID: 39034028 DOI: 10.1016/j.lfs.2024.122919] [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: 04/27/2024] [Revised: 07/04/2024] [Accepted: 07/16/2024] [Indexed: 07/23/2024]
Abstract
AIMS B7 molecules (B7s) are crucial synergistic signals for effective immune surveillance against tumor cells. While previous studies have explored the association between the B7 family and cancer, most have been limited to specific genes or cancer subtypes. MAIN METHODS Our study utilized multi-omics data to investigate potential correlations between B7s expression (B7s exp.) and prognosis, clinicopathological features, somatic mutations (SMs), copy number variations (CNVs), immune characteristics, tumor microenvironment (TME), microsatellite instability, tumor mutation burden, immune checkpoint gene (ICG), and drug responsiveness in TCGA tumors. Furthermore, the connection between B7s exp. and immunotherapy (IT) performance assessed in various validated datasets. Following this, immune infiltration analysis (IIA) was conducted based on B7s exp., CNVs, or SMs in bladder cancer (BLCA), complemented by real-time PCR (RT-PCR) and protein confirmation of B7-H3. KEY FINDINGS Across most cancer types, B7s exp. was related to prognosis, clinicopathological characteristics, mutations, CNVs, ICG, TMB, TME. The examination of sensitivity to anticancer drugs unveiled correlations between B7 molecules and different drug sensitivities. Specific B7s exp. patterns were linked to the clinical effectiveness of IT. Using GSEA, several enriched immune-related functions and pathways were identified. Particularly in BLCA, IIA revealed significant connections between B7 CNVs, mutation status, and various immune cell infiltrates. RT-PCR confirmed elevated B7-H3 gene levels in BLCA tumor tissues. SIGNIFICANCE This study confirmed the significance of B7s exp. and genomic changes in predicting outcomes and treatment across different cancer types. Moreover, they indicate a critical function of B7s in BLCA and their potential as IT biomarkers.
Collapse
Affiliation(s)
- Gangjian Zhao
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Peng Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Yong Suo
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chenyun Li
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Shaobo Yang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhe Zhang
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Zhouliang Wu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China
| | - Chong Shen
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| | - Hailong Hu
- Department of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China; Tianjin Key Laboratory of Urology, Tianjin Institute of Urology, The Second Hospital of Tianjin Medical University, Tianjin, China.
| |
Collapse
|
4
|
Dawidowicz M, Kot A, Mielcarska S, Psykała K, Kula A, Waniczek D, Świętochowska E. B7H4 Role in Solid Cancers: A Review of the Literature. Cancers (Basel) 2024; 16:2519. [PMID: 39061159 PMCID: PMC11275172 DOI: 10.3390/cancers16142519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 06/27/2024] [Accepted: 07/09/2024] [Indexed: 07/28/2024] Open
Abstract
Anti-cancer immunotherapies entirely changed the therapeutic approach to oncological patients. However, despite the undeniable success of anti-PD-1, PD-L1, and CTLA-4 antibody treatments, their effectiveness is limited either by certain types of malignancies or by the arising problem of cancer resistance. B7H4 (aliases B7x, B7H4, B7S1, VTCN1) is a member of a B7 immune checkpoint family with a distinct expression pattern from classical immune checkpoint pathways. The growing amount of research results seem to support the thesis that B7H4 might be a very potent therapeutic target. B7H4 was demonstrated to promote tumour progression in immune "cold" tumours by promoting migration, proliferation of tumour cells, and cancer stem cell persistence. B7H4 suppresses T cell effector functions, including inflammatory cytokine production, cytolytic activity, proliferation of T cells, and promoting the polarisation of naïve CD4 T cells into induced Tregs. This review aimed to summarise the available information about B7H4, focusing in particular on clinical implications, immunological mechanisms, potential strategies for malignancy treatment, and ongoing clinical trials.
Collapse
Affiliation(s)
- Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland
| | - Anna Kot
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| | - Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| | - Katarzyna Psykała
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 19 Jordana, 41-800 Zabrze, Poland
| |
Collapse
|
5
|
Liu M, Wu S, Wu H, Zhou Y, Zhang X, Zhu D, Jiang J. Ferredoxin 1: a gatekeeper in halting lung adenocarcinoma progression through activation of the GPRIN2 signaling pathway. J Transl Med 2024; 22:510. [PMID: 38802900 PMCID: PMC11131317 DOI: 10.1186/s12967-024-05277-6] [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: 03/05/2024] [Accepted: 05/04/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Lung adenocarcinoma (LUAD) is a highly lethal form of lung cancer. Despite advancements in treatments, managing LUAD is still challenging due to its aggressive behavior. Recent studies indicate that various molecular pathways, including the dysregulation of ferredoxin 1 (FDX1), play roles in LUAD progression. FDX1, a crucial protein in cellular redox reactions and energy metabolism, has been linked to several cancers. However, its exact role in the development of LUAD is not yet fully understood. METHODS We investigated the role of ferredoxin 1 (FDX1) in LUAD progression through analysis of its expression in LUAD tissues and its impact on patient survival. Functional assays were performed to assess the effects of FDX1 overexpression on LUAD cell proliferation, migration, and invasion. A xenograft model was employed to evaluate the tumorigenesis potential of LUAD cells with FDX1 overexpression. Mechanistic insights into FDX1 regulation were gained through depletion experiments targeting the G protein-regulated inducer of neurite outgrowth 2 (GPRIN2)/PI3K signaling pathway. RESULTS FDX1 expression was down-regulated in LUAD tissues, correlating with shorter patient survival. Overexpression of FDX1 suppressed LUAD cell proliferation, migration, and invasion in vitro, and inhibited tumorigenesis in vivo. Mechanistically, the GPRIN2/PI3K signaling pathway was implicated in FDX1 regulation, as depletion of GPRIN2 reversed the effects of FDX1 overexpression on cellular functions. CONCLUSIONS Our findings highlight FDX1 as a potential tumor suppressor in LUAD, acting through modulation of the GPRIN2/PI3K signaling pathway. These results suggest FDX1 as a promising therapeutic target for LUAD treatment, warranting further investigation into its clinical relevance.
Collapse
Affiliation(s)
- Ming Liu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
| | - Shaoxian Wu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Haoyu Wu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - You Zhou
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China
| | - Xinyu Zhang
- Department of Respiratory and Critical Care Medicine, The Affiliated Changzhou No.2 People's Hospital of Nanjing Medical University, Changzhou, China
- Changzhou Medical Center, Nanjing Medical University, Changzhou, China
- Dalian Medical University, Dalian, China
| | - Dawei Zhu
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| | - Jingting Jiang
- Department of Tumor Biological Diagnosis and Treatment Center, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
6
|
Dawidowicz M, Kula A, Mielcarska S, Świętochowska E, Waniczek D. Prognostic Value of B7H4 Expression in Patients with Solid Cancers: A Systematic Review and Meta-Analysis. Int J Mol Sci 2024; 25:5045. [PMID: 38732263 PMCID: PMC11084412 DOI: 10.3390/ijms25095045] [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: 04/08/2024] [Revised: 04/27/2024] [Accepted: 05/02/2024] [Indexed: 05/13/2024] Open
Abstract
V-set domain-containing T-cell activation inhibitor 1 (aliases VTCN1, B7H4) participates in tumour immune escape by delivering inhibitory signals to T cells. The purpose of this article was to assess the B7H4 prognostic value in solid cancers. Three databases were searched for relevant articles. The main endpoints were overall survival (OS), disease-specific survival (DSS), progression-free survival (PFS), recurrence-free survival (RFS), and disease-free survival (DFS). Appropriate hazard ratios (HRs) were pooled. The R studio software (version 4.0.3) was used for data analysis. Thirty-one studies met the inclusion criteria. High expression of B7H4 was associated with worse OS (HR = 1.52, 95% CI: 1.37-1.68) but not with DSS (HR = 1.14, 95% CI: 0.49-2.63), RFS (HR = 1.77, 95% CI: 0.75-4.18), DFS (HR = 1.29, 95% CI: 0.8-2.09), or PFS (HR = 1.71, 95% CI: 0.91-3.2) in patients with solid cancers. High expression of B7H4 is associated with a poorer prognosis in patients with solid cancers. B7H4 is a promising prognostic biomarker and immunotherapeutic target for various solid cancers because of its activity in cancer immunity and tumourigenesis.
Collapse
Affiliation(s)
- Miriam Dawidowicz
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland;
| | - Agnieszka Kula
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland;
| | - Sylwia Mielcarska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland; (S.M.); (E.Ś.)
| | - Elżbieta Świętochowska
- Department of Medical and Molecular Biology, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-800 Zabrze, Poland; (S.M.); (E.Ś.)
| | - Dariusz Waniczek
- Department of Oncological Surgery, Faculty of Medical Sciences in Zabrze, Medical University of Silesia, 41-808 Katowice, Poland;
| |
Collapse
|
7
|
Huang H, Ge J, Fang Z, Wu S, Jiang H, Lang Y, Chen J, Xiao W, Xu B, Liu Y, Chen L, Zheng X, Jiang J. Precursor exhausted CD8 +T cells in colorectal cancer tissues associated with patient's survival and immunotherapy responsiveness. Front Immunol 2024; 15:1362140. [PMID: 38510246 PMCID: PMC10950923 DOI: 10.3389/fimmu.2024.1362140] [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: 12/27/2023] [Accepted: 02/26/2024] [Indexed: 03/22/2024] Open
Abstract
Exhausted CD8+T cells represent a distinct cellular lineage that emerges during both chronic infections and cancers. Recent studies have shown that persistent antigen exposure can drive the differentiation of precursor exhausted CD8+T cells, termed Tpex cells, which are characterized as TCF-1+PD-1+CD8+T cells. Elevated Tpex cell frequencies in the tumor microenvironment (TME) are associated with improved overall survival (OS) in cancer patients and heightened responsiveness to anti-PD-1 therapy. In our present study, we utilized multi-color immunohistochemistry (mIHC) to determine the localization and clinical implications of tumor-infiltrating Tpex cells within the TME of human colorectal cancer (CRC) tissues. We also conducted a multi-omics integrative analysis using single-cell RNA sequencing (scRNA-seq) data derived from both the murine MC38 tumor model and human CRC tissues. This analysis helped delineate the transcriptional and functional attributes of Tpex cells within the CRC TME. Furthermore, we employed spatial transcriptome sequencing data from CRC patients to investigate the interactions between Tpex cells and other immune cell subsets within the TME. In conclusion, our study not only established a method for Tpex cell detection using mIHC technology but also confirmed that assessing Tpex cells within the CRC TME could be indicative of patients' survival. We further uncovered the transcriptional and functional characteristics of Tpex cells in the TME and ascertained their pivotal role in the efficacy of immunotherapy against CRC.
Collapse
Affiliation(s)
- Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Junwei Ge
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Zhang Fang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Hongwei Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Yanyan Lang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Wenlu Xiao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Yingting Liu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Jiangsu, Changzhou, China
| |
Collapse
|
8
|
Ge J, Chen J, Shen Q, Zheng X, Chen X, Shi L, Chen L, Xu B. Comprehensive Analysis of the Immunosuppressive Function of Regulatory T Cells in Human Hepatocellular Carcinoma Tissues. Cancer Control 2024; 31:10732748241251580. [PMID: 38712609 DOI: 10.1177/10732748241251580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Immune-based therapies are commonly employed to combat hepatocellular carcinoma (HCC). However, the presence of immune-regulating elements, especially regulatory T cells (Tregs), can dramatically impact the treatment efficacy. A deeper examination of the immune-regulation mechanisms linked to these inhibitory factors and their impact on HCC patient outcomes is warranted. METHODS We employed multicolor fluorescence immunohistochemistry (mIHC) to stain Foxp3, cytokeratin, and nuclei on an HCC tissue microarray (TMA). Leveraging liver cancer transcriptome data from TCGA, we built a prognostic model focused on Treg-associated gene sets and represented it with a nomogram. We then sourced liver cancer single-cell RNA sequencing data (GSE140228) from the GEO database, selectively focusing on Treg subsets, and conducted further analyses, including cell-to-cell communication and pseudo-time trajectory examination. RESULTS Our mIHC results revealed a more substantial presence of Foxp3+Tregs in HCC samples than in adjacent normal tissue samples (P < .001). An increased presence of Foxp3+Tregs in HCC samples correlated with unfavorable patient outcomes (HR = 1.722, 95% CI:1.023-2.899, P = .041). The multi-factorial prognosis model we built from TCGA liver cancer data highlighted Tregs as a standalone risk determinant for predicting outcomes (HR = 3.84, 95% CI:2.52-5.83, P < .001). Re-analyzing the scRNA-seq dataset (GSE140228) showcased distinctive gene expression patterns in Tregs from varying tissues. Interactions between Tregs and other CD4+T cell types were predominantly governed by the CXCL13/CXCR3 signaling pathway. Communication pathways between Tregs and macrophages primarily involved MIF-CD74/CXCR4, LGALS9/CD45, and PTPRC/MRC1. Additionally, macrophages could influence Tregs via HLA-class II and CD4 interactions. CONCLUSION An elevated presence of Tregs in HCC samples correlated with negative patient outcomes. Elucidating the interplay between Tregs and other immune cells in HCC could provide insights into the modulatory role of Tregs within HCC tissues.
Collapse
Affiliation(s)
- Junwei Ge
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Qiong Shen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xuemin Chen
- Radiological Intervention Center, Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liangrong Shi
- Radiological Intervention Center, Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
9
|
Liu B, Liu Z, Gao C. Relationship Between CD8+ T Cells and Prognosis of Esophageal Cancer Patients: A Systematic Review and Meta-analysis. Mol Biotechnol 2024; 66:138-150. [PMID: 37060513 DOI: 10.1007/s12033-023-00733-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Accepted: 03/22/2023] [Indexed: 04/16/2023]
Abstract
Tumor infiltrating lymphocytes (TILs), especially CD8+ T cells, play an important role in the process of anti-tumor immune response and are significantly correlated with the prognosis of esophageal cancer (EC), but there are also inconsistent conclusions. This study aimed to comprehensively evaluate the relationship between invasive CD8+ T cells and the prognosis in patients with EC through meta-analysis, and to provide a basis for prognosis and immunotherapy for EC. Articles related to CD8+ T cells and EC prognosis in PubMed, Cochrane Library, Embase, and CNKI were searched. Cancer specific survival (CSS), overall survival (OS) and disease-free survival (DFS) served as endpoint events. Besides, Stata15.0 was adopted for meta-analysis, and hazard ratio (HR) and 95% confidence interval (95%CI) for calculation of combined effect sizes. Total 547 articles were retrieved and 27 articles were finally enrolled, including 3988 cases of EC patients. Meta-analysis showed that high CD8 expression levels in tumor tissues, especially those in cancer nests, were associated with longer OS (HR = 0.74, 95% CI 0.67-0.81) and DFS (HR = 0.90, 95% CI 0.85-0.95) in EC patients (P < 0.05). CD8+ T cells play an important role in the prognosis of EC patients and are indispensable components for the immune score of EC.
Collapse
Affiliation(s)
- Ben Liu
- Department of Medical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China
| | - Zeyuan Liu
- Department of Radiation Oncology, Nanjing Jiangning Hospital (The Affiliated Jiangning Hospital of Nanjing Medical University), Nanjing, 211000, Jiangsu, China
| | - Chao Gao
- Department of Medical Oncology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221000, Jiangsu, China.
| |
Collapse
|
10
|
Mortezaee K. B7x in cancer immunity and immunotherapy. Int Immunopharmacol 2023; 118:110133. [PMID: 37028278 DOI: 10.1016/j.intimp.2023.110133] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/29/2023] [Accepted: 03/30/2023] [Indexed: 04/08/2023]
Abstract
B7x (also called B7-H4) is a co-inhibitory molecule of B7 family that is highly expressed in non-inflamed or cold cancers, and its aberrant expression is contributed to cancer progression and poor outcomes. B7x preferentially expresses on antigen-presenting cells (APCs) and in tumor cells, and it acts as an alternative anti-inflammatory immune checkpoint for hampering peripheral immune responses. Augmented infiltration of immunosuppressive cells, reduced proliferation and effector function of CD4+ and CD8+ T cells, and increased generation of regulatory T cells (Tregs) are outcomes of increased B7x activity in cancer. Evaluation of B7x in sera can be exploited as an effective biomarker of response in cancer patients. B7x overexpression generally occurs in programmed death-ligand 1 (PD-L1)- cancers and is involved in tumor resistance to anti-programmed death-1 (PD-1), anti-PD-L1 or anti-cytotoxic T lymphocyte associated antigen-4 (CTLA-4) therapy. Co-expression of B7x receptor with PD-1 on CD8+ T cells has made the anti-B7x a fruitful approach for reinvigoration of the functionality of exhausted T cells and is served as a complementary regimen in patients who are irresponsive to the common immune checkpoint inhibitor (ICI) therapy. An advance in the field is the development of bispecific antibodies against B7x with other regulatory molecules within tumor microenvironment (TME).
Collapse
|
11
|
Zhang L, Yu B, Liu Z, Wei J, Pan J, Jiang C, Li Z. Analysis of the Clinicopathological Characteristics, Prognosis, and Lymphocyte Infiltration of Esophageal Neuroendocrine Neoplasms: A Surgery-Based Cohort and Propensity-Score Matching Study. Cancers (Basel) 2023; 15:cancers15061732. [PMID: 36980618 PMCID: PMC10046526 DOI: 10.3390/cancers15061732] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/31/2023] [Accepted: 02/22/2023] [Indexed: 03/14/2023] Open
Abstract
Background: Esophageal neuroendocrine neoplasms (E-NENs) are a rare and poorly reported subtype of esophageal carcinoma. We analyzed the differences in clinicopathological features, prognosis, and tumor-infiltrating lymphocytes (TILs) between E-NENs and esophageal squamous cell carcinoma (ESCC). Methods: A total of 3620 patients who underwent esophagectomy were enrolled retrospectively. The study cohort was divided into two groups (E-NENs and ESCC) through propensity-score matching, and the prognosis and TILs were compared between the two groups. The TILs were assessed using tumor specimens (including six cases of ESCC, six cases of neuroendocrine carcinomas [NECs], and six cases of mixed neuroendocrine–non-neuroendocrine neoplasms [MiNENs]). Results: E-NENs accounted for 3.0% (107/3620) of cases, among which there were just 3 neuroendocrine tumor cases, 51 NEC cases, and 53 MiNENs cases. After matching, esophageal neuroendocrine carcinomas (E-NECs) showed both poorer 5-year overall survival (OS; 35.4% vs. 54.8%, p = 0.0019) and recurrence-free survival (RFS; 29.3% vs. 48.9%, p < 0.001) compared with ESCC. However, the differences were not prominent in the subgroup with stage I. No significant survival benefit was observed for E-NECs with multimodal therapy. Multivariate analysis demonstrated that E-NECs are an independent risk factor for OS and RFS. In the exploratory analysis, E-NECs were associated with less infiltration of immune cells compared with ESCC. Conclusion: E-NECs are significantly associated with a poorer prognosis than ESCC except for early-stage disease. The fewer TILs within the tumor microenvironment of E-NECs compared with ESCC results in weaker anti-tumor immunity and may lead to a poorer prognosis.
Collapse
Affiliation(s)
- Long Zhang
- Department of Thoracic Surgery, Section of Esophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Boyao Yu
- Department of Thoracic Surgery, Section of Esophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhichao Liu
- Department of Thoracic Surgery, Section of Esophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jinzhi Wei
- Department of Pathology, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Jie Pan
- Department of Thoracic Surgery, Section of Esophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Chao Jiang
- Department of Thoracic Surgery, Section of Esophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
| | - Zhigang Li
- Department of Thoracic Surgery, Section of Esophageal Surgery, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200030, China
- Correspondence:
| |
Collapse
|
12
|
Hwang C, Lee HJ, Na JY, Kim KH, Song YJ, Kim JY, Kim K, Shin DH, Park JY, Kim SY, Lee JH, Choi KU. The stromal tumor-infiltrating lymphocytes, cancer stemness, epithelial-mesenchymal transition, and B7-H4 expression in ovarian serous carcinoma. J Ovarian Res 2023; 16:3. [PMID: 36609273 PMCID: PMC9825048 DOI: 10.1186/s13048-022-01076-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 12/12/2022] [Indexed: 01/09/2023] Open
Abstract
BACKGROUND B7-H4 is expressed in various types of cancers and its expression inversely correlates with the degree of tumor-infiltrating lymphocytes (TILs). Studies have shown the relationship between B7-H4, cancer stem cell (CSC) properties, and epithelial-mesenchymal transition (EMT) in various cancers. However, very few studies have investigated the relationship between B7-H4, TILs, cancer stemness, and EMT in epithelial ovarian cancer (EOC). The present study aimed to elucidate whether B7-H4 is involved in immune evasion and examine whether B7-H4 is associated with cancer stemness or EMT in ovarian serous carcinoma, the most common type of EOC. The clinical significance of B7-H4 was also investigated to evaluate its potential as a therapeutic target. METHODS A total of 145 patients included in this study. The degree of stromal TILs was evaluated using hematoxylin and eosin (H&E)-stained slides. Immunohistochemical analysis of B7-H4, CSC-related biomarkers (CD24, CD44s, CD133, and ALDH1), and EMT-related biomarkers (E-cadherin, N-cadherin, and vimentin) was performed using tissue microarray. qRT-PCR for VTCN1, CD24, CD44, PROM1, ALDH1, CDH1, CDH2, and VIM genes was performed on 38 frozen tissue samples. The mRNA expression levels were analyzed using Gene Expression Profiling Interactive Analysis (GEPIA) online analysis tool. RESULTS B7-H4 protein expression positively correlated with the degree of stromal TILs. CD24, CD44s, and CD133 expression showed a positive correlation with B7-H4 expression at both the protein and mRNA levels, but ALDH1 correlated only at the protein level. E-cadherin expression was positively correlated with B7-H4 expression at both the protein and mRNA levels. N-cadherin and vimentin expression was inversely related to B7-H4 expression only at the mRNA level. B7-H4 positive patients were associated with higher tumor grade and lower overall survival rate than B7-H4 negative patients, especially in ovarian serous carcinoma with low stromal TILs. CONCLUSIONS The present study demonstrates that B7-H4 may not be involved in the immune evasion mechanism, but is involved in cancer stemness and mesenchymal-epithelial transition. In addition, B7-H4 may be a therapeutic target for the treatment of ovarian serous carcinoma, especially with low stromal TILs.
Collapse
Affiliation(s)
- Chungsu Hwang
- Research Institute for Convergence of Biomedical Science and Technology, Pusan National Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Hyun Jung Lee
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Ju-Young Na
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Ki Hyung Kim
- grid.412588.20000 0000 8611 7824Department of Obstetrics and Gynecology, Pusan National University Hospital, 179 Gudeok-Ro, Seo-Gu, Busan, 49241 South Korea
| | - Yong Jung Song
- grid.412591.a0000 0004 0442 9883Department of Obstetrics and Gynecology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Jee Yeon Kim
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Kyungbin Kim
- grid.262229.f0000 0001 0719 8572Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, 179 Gudeok-Ro, Seo-Gu, Busan, 49241 South Korea
| | - Dong Hoon Shin
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Joon Young Park
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - So Young Kim
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Jung Hee Lee
- grid.412591.a0000 0004 0442 9883Department of Pathology, Pusan National University Yangsan Hospital, 20, Geumo-Ro, Mulguem-Eup, Yangsan-Si, Gyeongsangnam-Do South Korea
| | - Kyung Un Choi
- grid.262229.f0000 0001 0719 8572Department of Pathology, Pusan National University Hospital and Pusan National University School of Medicine, 179 Gudeok-Ro, Seo-Gu, Busan, 49241 South Korea
| |
Collapse
|
13
|
Xiao L, Guan X, Xiang M, Wang Q, Long Q, Yue C, Chen L, Liu J, Liao C. B7 family protein glycosylation: Promising novel targets in tumor treatment. Front Immunol 2022; 13:1088560. [PMID: 36561746 PMCID: PMC9763287 DOI: 10.3389/fimmu.2022.1088560] [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: 11/03/2022] [Accepted: 11/22/2022] [Indexed: 12/12/2022] Open
Abstract
Cancer immunotherapy, including the inhibition of immune checkpoints, improves the tumor immune microenvironment and is an effective tool for cancer therapy. More effective and alternative inhibitory targets are critical for successful immune checkpoint blockade therapy. The interaction of the immunomodulatory ligand B7 family with corresponding receptors induces or inhibits T cell responses by sending co-stimulatory and co-inhibitory signals respectively. Blocking the glycosylation of the B7 family members PD-L1, PD-L2, B7-H3, and B7-H4 inhibited the self-stability and receptor binding of these immune checkpoint proteins, leading to immunosuppression and rapid tumor progression. Therefore, regulation of glycosylation may be the "golden key" to relieve tumor immunosuppression. The exploration of a more precise glycosylation regulation mechanism and glycan structure of B7 family proteins is conducive to the discovery and clinical application of antibodies and small molecule inhibitors.
Collapse
Affiliation(s)
- Linlin Xiao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China,Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Xiaoyan Guan
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China,Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Mingli Xiang
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China,Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Wang
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Qian Long
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China,Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Chaoyi Yue
- School of Medicine and Technology, Zunyi Medical University, Zunyi, China
| | - Lulu Chen
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China,Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China
| | - Jianguo Liu
- Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China,*Correspondence: Chengcheng Liao, ; Jianguo Liu,
| | - Chengcheng Liao
- Department of Orthodontics II, Affiliated Stomatological Hospital of Zunyi Medical University, Zunyi, China,Oral Disease Research Key Laboratory of Guizhou Tertiary Institution, School of Stomatology, Zunyi Medical University, Zunyi, China,*Correspondence: Chengcheng Liao, ; Jianguo Liu,
| |
Collapse
|
14
|
Nagata Y, Yamamoto S, Kato K. Immune checkpoint inhibitors in esophageal cancer: Clinical development and perspectives. Hum Vaccin Immunother 2022; 18:2143177. [PMID: 36375821 PMCID: PMC9746438 DOI: 10.1080/21645515.2022.2143177] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/11/2022] [Accepted: 10/31/2022] [Indexed: 11/16/2022] Open
Abstract
Esophageal cancer is the sixth most common cause of cancer-related mortality worldwide. The standard treatment for unresectable esophageal cancer is systemic chemotherapy. However, the survival benefit is limited, with a median overall survival of less than 10 months. The advent of immune checkpoint inhibitors (ICIs), including programmed cell death-1 antibodies, has revolutionized the treatment paradigm for esophageal cancer. Since demonstrating promising efficacy with manageable safety in several clinical trials, ICIs has finally reached the point where they can be used in various tumor stages in the clinical setting. ICIs are most promising treatments that can be expected to improve the prognosis in patients with esophageal cancer now and in the future. This review outlines the mechanisms, results of clinical trials, and prospects for future studies of ICIs in esophageal cancer. It also discusses clinical questions and challenges in the therapeutic development of ICIs.
Collapse
Affiliation(s)
- Yusuke Nagata
- Department of Gastroenterology, Nagano Municipal Hospital, Nagano, Japan
| | - Shun Yamamoto
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Head and Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
15
|
Li Y, Huang H, Zhu Y, Xu B, Chen J, Liu Y, Zheng X, Chen L. Increased expression of METTL3 in pancreatic cancer tissues associates with poor survival of the patients. World J Surg Oncol 2022; 20:283. [PMID: 36058919 PMCID: PMC9442951 DOI: 10.1186/s12957-022-02743-7] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 07/16/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Methyltransferase-like 3 (METTL3) expression could be found in various normal and cancerous tissues. As of now, the clinical significance of METTL3 expression in human pancreatic cancer (PC) tissues still remains to be understood. Our present study aims to investigate the prognostic value and clinical implications of METTL3 expression in PC tissues. METHODS The TCGA, GTEx, and GEO public databases were used to study the mRNA expression level of the m6A family members and its relationship among PC tissues and normal pancreatic tissue. The immunohistochemistry was used to analyze the difference of METTL3 expression between cancer tissues and adjacent normal tissues. The prognostic value was evaluated by using the Log-rank survival analysis and Cox model analysis. PAAD samples from TCGA and GEO databases were used to perform the immune infiltration analysis and gene set enrichment analysis based on the genes that were highly correlated with METTL3. RESULTS Based on the analysis of TCGA, GTEx, and GEO public database, we found that the m6A family members showed a higher correlation in PC tissues compared to normal pancreatic tissues, and the mRNA expression level of the m6A family members showed a significant difference between PC tissues and adjacent normal tissues. Moreover, scRNA-seq data indicated that METTL3 showed a higher expression level in malignant epithelial cells. Our immunohistochemistry results also confirmed that the intensity of METTL3 immunostaining in PC tissues was significantly higher than that in adjacent normal tissues (P = 0.015). The overall survival (OS) of PC patients with high expression of METTL3 protein were significantly poorer than those with low expression of METTL3 protein (HR = 1.788, 95% CI 1.071-2.984, P = 0.026). Further analysis of PC data from the database showed that METTL3 expression was associated with a variety of tumor-infiltrating immune cells and was involved in m6A modification and metabolism in PC tissues. CONCLUSION Increased METTL3 expression at the protein level could be found in PC tissues, suggesting that the METTL3 expression was involved in the progression of PC and could serve as an important marker for prognostic prediction of this malignancy.
Collapse
Affiliation(s)
- Yuan Li
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Yulan Zhu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Junjun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Yingting Liu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China. .,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China. .,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003, People's Republic of China.
| |
Collapse
|
16
|
Zhu D, Liu Y, Chen J, Wang Q, Li Y, Zhu Y, Feng J, Jiang J. The methyltransferase METTL3 promotes tumorigenesis via mediating HHLA2 mRNA m6A modification in human renal cell carcinoma. Lab Invest 2022; 20:298. [PMID: 35794583 PMCID: PMC9258210 DOI: 10.1186/s12967-022-03496-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Accepted: 06/22/2022] [Indexed: 01/22/2023]
Abstract
Abstract
Background
As an important N6-methyladenosine (m6A) regulator, abnormal expression of methyltransferase-like protein 3 (METTL3) has been reported in certain human cancers. Although some data have shown that METTL3 plays an essential role in the progression of clear-cell renal cell carcinoma RCC (ccRCC), the detailed mechanism still remains largely undetermined.
Methods
Immunohistochemistry (IHC) assay was used to examine the expression of METTL3 and its clinical implications in human ccRCC by using tissue-microarray (TMA). The cellular models based on ccRCC cell lines such as 786-O and ACHN, were established by operating METTL3 and HHLA2 via knockdown or overexpression, followed by in vitro cellular function studies and in vivo subcutaneous transplantation tumor model.
Results
We found that METTL3 expression in ccRCC tissues was significantly higher compared with adjacent normal tissues. We also found the overall survival (OS) of the patients with low METTL3 expression was significantly better compared with the patients with high METTL3 expression. Furthermore, HHLA2highMETTL3high could serve as a better prognostic predictor for ccRCC patients. Depletion of METTL3 could significantly inhibit the cell viability, migration, and invasion abilities in ccRCC cell lines. Cellular studies further revealed that METTL3 could regulate HHLA2 expression via m6A modification of HHLA2 mRNA. In vitro studies revealed that HHLA2 overexpression could reverse the inhibition of cellular functions mediated by METTL3 depletion. The subcutaneous transplantation tumor model confirmed that HHLA2 overexpression could reverse the inhibition of tumor growth mediated by METTL3 depletion.
Conclusion
Our study indicated that METTL3 served as an important prognostic predictor for ccRCC patients, and we demonstrated a novel regulatory mechanism of HHLA2 by mRNA epigenetic modification via METTL3. Moreover, we found that the METTL3/HHLA2 axis could promote tumorigenesis of ccRCC. Collectively, our current findings provided new insights into the therapeutic strategy against this malignancy targeting METTL3.
Collapse
|
17
|
Harada K, Yamamoto S, Kato K. Pembrolizumab for the treatment of advanced esophageal cancer. Future Oncol 2022; 18:2311-2319. [PMID: 35418242 DOI: 10.2217/fon-2022-0108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The prognosis of patients with advanced esophageal cancer (EC) remains poor and there are limited effective therapeutic agents for EC. Pembrolizumab monotherapy exerts clinically meaningful benefits for advanced esophageal squamous cell carcinoma patients with a combined positive score of ≥10. Additionally, pembrolizumab plus doublet chemotherapy results in a significant survival benefit for patients with advanced EC as first-line treatment compared with chemotherapy alone. We provide an overview of immune checkpoint inhibitors and present important clinical data related to treatment for EC patients. In our opinion, pembrolizumab plus chemotherapy should be the standard first-line treatment for patients with advanced EC, regardless of histology and combined positive score. Biomarker studies to identify patient populations in which immune checkpoint inhibitors are expected to show efficacy are needed.
Collapse
Affiliation(s)
- Kentaro Harada
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Shun Yamamoto
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Ken Kato
- Department of Head & Neck, Esophageal Medical Oncology, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
18
|
Liu B, Xiao X, Lin Z, Lou Y, Zhao L. PDGFRB is a potential prognostic biomarker and correlated with immune infiltrates in gastric cancer. Cancer Biomark 2021; 34:251-264. [PMID: 34958001 DOI: 10.3233/cbm-210335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Gastric cancer (GC) is a common cancer with high mortality and morbidity rates worldwide. Although medical and surgical treatments have improved, the mechanisms of the progression of GC remain unclear. Platelet-derived growth factor receptor-β (PDGFRB) plays a pivotal role in angiogenesis and tumor cell proliferation and has been suggested as a prognostic marker of cancer. This study aimed to explore the relationship of PDGFRB expression with clinicopathologic characteristics, immune cell infiltration status, and prognosis in GC. In this study, we visualized the expression and prognostic values of PDGFRB in GC using the Oncomine, UALCAN, GEPIA, and Kaplan-Meier Plotter databases. And then we explored the potential relationships between PDGFRB expression and the levels of immune cell infiltration using the TIMER, GEPIA databases and CIBERSORT algorithm. Furthermore, LinkedOmics analysis was performed to explore the functions for PDGFRB. The results showed close correlations between PDGFRB and immune cell infiltration especially M2 Macrophage infiltration in GC. High PDGFRB expression was related to poor outcomes in GC. High PDGFRB expression can negatively affect GC prognosis by promoting angiogenesis and modulating the tumor immune microenvironment. These results strongly suggest that PDGFRB can be used as a prognostic biomarker of GC and provide novel insights into possible immunotherapeutic targets.
Collapse
Affiliation(s)
- Baohong Liu
- State Key Laboratory of Veterinary Etiological Biology; Key Laboratory of Veterinary Parasitology of Gansu Province; Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Lanzhou, Gansu, China
| | - Xingxing Xiao
- Wenzhou Key Laboratory of Sanitary Microbiology; Key Laboratory of Laboratory Medicine, Ministry of Education, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Ziqin Lin
- Wenzhou Key Laboratory of Sanitary Microbiology; Key Laboratory of Laboratory Medicine, Ministry of Education, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yongliang Lou
- Wenzhou Key Laboratory of Sanitary Microbiology; Key Laboratory of Laboratory Medicine, Ministry of Education, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingling Zhao
- Wenzhou Key Laboratory of Sanitary Microbiology; Key Laboratory of Laboratory Medicine, Ministry of Education, China; School of Laboratory Medicine and Life Science, Wenzhou Medical University, Wenzhou, Zhejiang, China
| |
Collapse
|
19
|
Niu H, Song F, Wei H, Li Y, Huang H, Wu C. Inhibition of BRD4 Suppresses the Growth of Esophageal Squamous Cell Carcinoma. Cancer Invest 2021; 39:826-841. [PMID: 34519605 DOI: 10.1080/07357907.2021.1975736] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Bromodomain-containing protein 4 (BRD4) binds acetylated lysine residues on histones to facilitate the epigenetic regulation of many genes, and it plays a key role in many cancer types. Despite many prior reports that have explored the importance of BRD4 in oncogenesis and the regulation of epigenetic memory, its role in esophageal squamous cell carcinoma (ESCC) progression is poorly understood. Here, we investigated BRD4 expression in human ESCC tissues to understand how it regulates the biology of these tumor cells. METHODS BRD4 expression in ESCC tissues was measured via immunohistochemical staining. BRD4 inhibition in the Eca-109 and KYSE-150 ESCC cell lines was conducted to explore its functional role in these tumor cells. RESULTS BRD4 overexpression was observed in ESCC tissues and cells, and inhibiting the function of the gene impaired the proliferative, invasive, and migratory activity of these cells while promoting their apoptosis. Cyclin D1 and c-Myc expression were also suppressed by BRD4 inhibition, and the expression of key epithelial-mesenchymal transition markers including E-cadherin and Vimentin was markedly altered by such inhibition. CONCLUSIONS BRD4 plays key functional roles in the biology of ESCC, proposing that it could be a viable therapeutic target for treating this cancer type.
Collapse
Affiliation(s)
- Haiyu Niu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Feixue Song
- Department of Oncology, Lanzhou University Second Hospital, Lanzhou, China
| | - Hanwen Wei
- Department of Cardiology, The First People's Hospital of Lanzhou, Lanzhou, China
| | - Yuan Li
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Hao Huang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
20
|
Granulocyte-Macrophage Colony-Stimulating Factor-Activated Neutrophils Express B7-H4 That Correlates with Gastric Cancer Progression and Poor Patient Survival. J Immunol Res 2021; 2021:6613247. [PMID: 33763491 PMCID: PMC7962878 DOI: 10.1155/2021/6613247] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 02/07/2021] [Accepted: 02/13/2021] [Indexed: 02/07/2023] Open
Abstract
Neutrophils are prominent components of gastric cancer (GC) tumors and exhibit distinct phenotypes in GC environment. However, the phenotype, regulation, and clinical relevance of neutrophils in human GC are presently unknown. Here, immunohistochemistry, real-time PCR, and flow cytometry analyses were performed to examine levels and phenotype of neutrophils in samples from 41 patients with GC, and also isolated, stimulated, and/or cultured neutrophils for in vitro regulation assays. Finally, we performed Kaplan-Meier plots for overall survival by using the log-rank test to evaluate the clinical relevance of neutrophils and their subsets. In our study, neutrophils in tumor tissues were significantly higher than those in nontumor tissues and were positively associated with tumor progression but negatively correlated with GC patient survival. Most intratumoral neutrophils showed an activated CD54+ phenotype and expressed high-level immunosuppressive molecule B7-H4. Tumor tissue culture supernatants from GC patients induced neutrophils to express CD54 and B7-H4 in both time-dependent and dose-dependent manners. Locally enriched CD54+ neutrophils and B7-H4+ neutrophils positively correlated with increased granulocyte-macrophage colony-stimulating factor (GM-CSF) detection ex vivo, and in vitro GM-CSF induced the expression of CD54 and B7-H4 on neutrophils in a time-dependent and dose-dependent manner. Moreover, GC tumor-derived GM-CSF activated neutrophils and induced neutrophil B7-H4 expression via Janus kinase (JAK)-signal transducer and activator of transcription 3 (STAT3) signaling pathway activation. Furthermore, higher intratumoral B7-H4+ neutrophil percentage/number was found in GC patients with advanced tumor node metastasis stage and reduced overall survival following surgery. Our results illuminate a novel regulating mechanism of B7-H4 expression on tumor-activated neutrophils in GC, suggesting that functional inhibition of these novel GM-CSF-B7-H4 pathways may be a suitable therapeutic strategy to treat the immune tolerance feature of GC.
Collapse
|
21
|
Song X, Zhou Z, Li H, Xue Y, Lu X, Bahar I, Kepp O, Hung MC, Kroemer G, Wan Y. Pharmacologic Suppression of B7-H4 Glycosylation Restores Antitumor Immunity in Immune-Cold Breast Cancers. Cancer Discov 2020; 10:1872-1893. [PMID: 32938586 DOI: 10.1158/2159-8290.cd-20-0402] [Citation(s) in RCA: 90] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Revised: 07/15/2020] [Accepted: 09/11/2020] [Indexed: 11/16/2022]
Abstract
Despite widespread utilization of immunotherapy, treating immune-cold tumors has proved to be a challenge. Here, we report that expression of the immune checkpoint molecule B7-H4 is prevalent among immune-cold triple-negative breast cancers (TNBC), where its expression inversely correlates with that of PD-L1. Glycosylation of B7-H4 interferes with its interaction/ubiquitination by AMFR, resulting in B7-H4 stabilization. B7-H4 expression inhibits doxorubicin-induced cell death through the suppression of eIF2α phosphorylation required for calreticulin exposure vis-à-vis the cancer cells. NGI-1, which inhibits B7-H4 glycosylation causing its ubiquitination and subsequent degradation, improves the immunogenic properties of cancer cells treated with doxorubicin, enhancing their phagocytosis by dendritic cells and their capacity to elicit CD8+ IFNγ-producing T-cell responses. In preclinical models of TNBC, a triple combination of NGI-1, camsirubicin (a noncardiotoxic doxorubicin analogue) and PD-L1 blockade was effective in reducing tumor growth. Collectively, our findings uncover a strategy for targeting the immunosuppressive molecule B7-H4. SIGNIFICANCE: This work unravels the regulation of B7-H4 stability by ubiquitination and glycosylation, which affects tumor immunogenicity, particularly regarding immune-cold breast cancers. The inhibition of B7-H4 glycosylation can be favorably combined with immunogenic chemotherapy and PD-L1 blockade to achieve superior immuno-infiltration of cold tumors, as well as improved tumor growth control.See related commentary by Pearce and Läubli, p. 1789.This article is highlighted in the In This Issue feature, p. 1775.
Collapse
Affiliation(s)
- Xinxin Song
- Department of Obstetrics and Gynecology, Department of Pharmacology, the Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Process Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Zhuan Zhou
- Department of Obstetrics and Gynecology, Department of Pharmacology, the Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Process Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois
| | - Hongchun Li
- Research Center for Computer-Aided Drug Discovery, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Beijing, China.,Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yifan Xue
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Xinghua Lu
- Department of Biomedical Informatics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Ivet Bahar
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Oliver Kepp
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Paris, France
| | - Mien-Chie Hung
- Graduate Institute of Biomedical Sciences, Research Center for Cancer Biology and Center for Molecular Medicine, China Medical University, Taiwan
| | - Guido Kroemer
- Equipe labellisée par la Ligue contre le Cancer, Université de Paris, Sorbonne Université, Metabolomics and Cell Biology Platforms, Institut Gustave Roussy, Paris, France.,Pôle de Biologie, Hôpital Européen Georges Pompidou, France.,Suzhou Institute for Systems Medicine, Chinese Academy of Medical Sciences, Beijing, China.,Department of Women's and Children's Health, Karolinska Institute, Karolinska University Hospital, Stockholm, Sweden
| | - Yong Wan
- Department of Obstetrics and Gynecology, Department of Pharmacology, the Robert H. Lurie Comprehensive Cancer Center, Chemistry of Life Process Institute, Northwestern University Feinberg School of Medicine, Chicago, Illinois.
| |
Collapse
|
22
|
Li Q, Zhang D, He W, Chen T, Yan Z, Gao X, Chen L, Zheng X, Xu B, Lu B, Jiang J. CD8 + T cells located in tertiary lymphoid structures are associated with improved prognosis in patients with gastric cancer. Oncol Lett 2020; 20:2655-2664. [PMID: 32782582 PMCID: PMC7400769 DOI: 10.3892/ol.2020.11828] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 02/11/2020] [Indexed: 01/01/2023] Open
Abstract
The presence of tumor infiltrating lymphocytes (TILs) and tertiary lymphoid structures (TLSs) in tumor tissues are of great prognostic significance in several types of human cancer. The present study investigated the density of TILs and TLSs in gastric cancer (GC) tissues and their association with pathological parameters. Moreover, the clinical significance of follicular CD8+ cytotoxic T cells present within the germinal centers of the tumor-associated TLSs was investigated. Immunohistochemistry and H&E staining were used to examine the infiltration and distribution patterns of TILs, TLSs and germinal center (gc) CD8+ TILs in tumor tissues obtained from 63 patients with GC. The number of TILs, TLSs, combination of TILs and TLSs (TILs-TLSs) and gcCD8+ TILs were used to define tumoral immune parameters, and the prognostic value of these parameters was assessed. The analysis revealed that patients with GC with increased levels of TILs, TLSs, or gcCD8+ TILs exhibited improved overall survival. In addition, gcCD8+ TILs levels were significantly associated with patient age, histological grade and pTN stage. Increased levels of TILs-TLSs were positively associated with nerve invasion, tumor thrombus, nodal metastasis and histological grade. Multivariate Cox regression analysis revealed that TILs-TLSs and gcCD8+ TILs were independent prognostic factors. The data obtained in the present study demonstrated that high levels of tumoral immune parameters are important independent prognostic predictors for human GC. The results also suggested a possible role of gcCD8+ TILs in tumor immune surveillance.
Collapse
Affiliation(s)
- Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Dachuan Zhang
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China.,Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Wenting He
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Tongbing Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Zhantao Yan
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Xie Gao
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China.,Department of Tumor Biological Treatment, Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, P.R. China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China.,Department of Tumor Biological Treatment, Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, P.R. China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China.,Department of Tumor Biological Treatment, Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, P.R. China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Binfeng Lu
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China.,Department of Tumor Biological Treatment, Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, P.R. China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, P.R. China
| |
Collapse
|
23
|
He W, Zhang D, Liu H, Chen T, Xie J, Peng L, Zheng X, Xu B, Li Q, Jiang J. The High Level of Tertiary Lymphoid Structure Is Correlated With Superior Survival in Patients With Advanced Gastric Cancer. Front Oncol 2020; 10:980. [PMID: 32733793 PMCID: PMC7358602 DOI: 10.3389/fonc.2020.00980] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2019] [Accepted: 05/18/2020] [Indexed: 01/05/2023] Open
Abstract
Background: A tertiary lymphoid structure (TLS) is a crucial component of the tumor microenvironment, which reflects the anti-tumor immune response in the host. The aim of the present study was to carry out a histopathological evaluation for TLS and assess its prognostic value in gastric cancer (GC). Methods: A total of 1,033 cases that have received a gastrectomy were reviewed, including 914 in the primary cohort and 119 in the validation cohort. TLS was assessed by optical microscopy and verified by immunohistochemistry. A total of five histopathological evaluation methods were compared in the primary cohort and validated in the validation cohort. In addition, MECA-79 and CD21 were used to verify the accuracy of the histopathological scoring system for TLS. The association among TLS, clinicopathological parameters, and patient prognosis was analyzed. Results: TLS as assessed by morphology and immunohistochemistry were significantly correlated and consistent. The morphological evaluation of TLS was accurate. Typically, the high level of TLS was significantly correlated with tumor size (P = 0.047), histological grade (P = 0.039), pTN stage (P = 0.044), and WHO subtype (P < 0.001). In addition, TLShi was a positive indicator of overall survival, as determined by Kaplan–Meier survival (P = 0.038) and multivariate Cox regression analyses (hazard ratio = 0.794, 95% CI: 0.668–0.942, P = 0.008). According to the results, TLShi had a positive effect on the primary cohort patients with pTN stages II and III (P = 0.027, P = 0.042). Conclusions: The histopathological evaluation of TLS was accurate. Diagnosis based solely on hematoxylin and eosin staining of the sections did not easily distinguish tumor-associated TLS. The density of TLS in the center of the tumor was found to be more indicative of patient prognosis than TLS in the invasive margin, with the levels of total TLS shown to best correlate with overall survival in patients with advanced-stage GC.
Collapse
Affiliation(s)
- Wenting He
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Dachuan Zhang
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Hong Liu
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Tongbing Chen
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jun Xie
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Lei Peng
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| | - Qing Li
- Department of Pathology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, China.,Institute of Cell Therapy, Soochow University, Changzhou, China
| |
Collapse
|
24
|
Gao Y, Guo W, Geng X, Zhang Y, Zhang G, Qiu B, Tan F, Xue Q, Gao S, He J. Prognostic value of tumor-infiltrating lymphocytes in esophageal cancer: an updated meta-analysis of 30 studies with 5,122 patients. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:822. [PMID: 32793667 PMCID: PMC7396260 DOI: 10.21037/atm-20-151] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Background The prognostic role of tumor-infiltrating lymphocytes (TILs) in esophageal cancer (EC) patients is controversial; therefore, we performed a meta-analysis to obtain a consensus. Methods The PubMed, PubMed Central, Embase, Cochrane Library, and Web of Science databases were searched. The pooled hazard ratios (HRs) with 95% confidence intervals (CIs) were calculated using fixed effect or random effect models depending on the heterogeneity. Results A total of 30 articles comprising 5,122 patients were included in this meta-analysis. High levels of generalized TIL infiltration were associated with better overall survival (OS) (HR =0.67, 95% CI: 0.47–0.95, P=0.02) in EC patients. High CD8+ T-cell infiltration and high CD4+ T-cell infiltration were associated with better OS (HR =0.68, 95% CI: 0.60–0.78, P<0.001; HR =0.70, 95% CI: 0.57–0.85, P<0.001, respectively). However, the pooled results showed that neither CD3+ nor FOXP3+ T-cell infiltration were associated with patient survival (P>0.05). Moreover, for esophageal squamous cell carcinoma (ESCC), high CD8+ T lymphocyte infiltration in the TN (Tumor nest) or TS (Tumor stroma) significantly predicted better OS (pooled HR =0.70, 95% CI: 0.57–0.85; P=0.001; pooled HR =0.77, 95% CI: 0.65–0.91; P=0.003). Conclusions High levels of generalized TILs, high CD8+ T-cell infiltration and high CD4+ T-cell infiltration have the potential to serve as prognostic markers in EC patients. Moreover, high CD8+ TIL in TNs or TS can predict better OS in ESCC patients.
Collapse
Affiliation(s)
- Yibo Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Guo
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiao Geng
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yidong Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.,Big Data Institute, Li Ka Shing Center for Health Information and Discovery, University of Oxford, Oxford, United Kingdom
| | - Guochao Zhang
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Bin Qiu
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fengwei Tan
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qi Xue
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shugeng Gao
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jie He
- Department of Thoracic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
25
|
Hao J, Li M, Zhang T, Yu H, Liu Y, Xue Y, An R, Wang S. Prognostic Value of Tumor-Infiltrating Lymphocytes Differs Depending on Lymphocyte Subsets in Esophageal Squamous Cell Carcinoma: An Updated Meta-Analysis. Front Oncol 2020; 10:614. [PMID: 32411602 PMCID: PMC7198736 DOI: 10.3389/fonc.2020.00614] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2020] [Accepted: 04/03/2020] [Indexed: 12/15/2022] Open
Abstract
Background: Tumor-infiltrating lymphocytes (TILs) play a role in the anti-tumor immune response, and are often found in esophageal squamous cell carcinoma (ESCC). Methods: We performed a systematic review and meta-analysis, aiming to establish pooled estimates for survival outcomes of TILs based on their abundance and infiltrating location. A literature search of PubMed/Medline, Embase, Web of Science and the Cochrane Library was conducted. Studies that investigated the prognostic significance of generalized, CD8+, CD4+, FoxP3+, CD3+, and CD45O+ TILs in ESCC patients were included. Results: In pooled analysis, generalized TILs infiltrating the entire tumor mass were positively associated with disease-free survival (DFS), with a univariate-related hazard ratio (HR) of 0.630 [95% confidence interval (CI) 0.415-0.955], and also positively associated with overall survival (OS), with a univariate-related HR of 0.586 (0.447-0.770) and a multivariate-related HR of 0.621 (0.439-0.878). The pan-tumor, intra-tumor and peri-tumor CD8+ TILs had a favorable effect on OS, with univariate-related HRs of 0.733 (0.555-0.968), 0.797 (0.660-0.962), and 0.776 (0.635-0.948), respectively. Similar results were observed in CD8+ TILs that infiltrated the whole tumor mass, with a multivariate-related HR of 0.705 (0.524-0.947). CD4+, FoxP3+, CD3+, and CD45O+ TILs were not linked to DFS or OS. Subtypes and spatial locations of TILs seemed to influence study outcomes. Conclusions: Experimental and analytical methods of future studies should be carefully designed to avoid overestimating the effect of TILs on prognosis. Our meta-analysis confirms the prognostic efficacy of generalized TILs and CD8+ TILs in esophageal squamous cell carcinoma (ESCC) patients.
Collapse
Affiliation(s)
- Jiatao Hao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,General Department, Zhongshan Hospital of Fudan University, Shanghai, China
| | - Meng Li
- Neurology Department, Baoji Central Hospital, Baoji, China
| | - Taohong Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Hui Yu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Yan Xue
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ruifang An
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shuai Wang
- Department of Thoracic Surgery, Zhongshan Hospital of Fudan University, Shanghai, China
| |
Collapse
|
26
|
A self-designed CpG ODN enhanced the anti-melanoma effect of pimozide. Int Immunopharmacol 2020; 83:106397. [PMID: 32220805 DOI: 10.1016/j.intimp.2020.106397] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2019] [Revised: 03/07/2020] [Accepted: 03/09/2020] [Indexed: 12/20/2022]
Abstract
Melanomas represent the deadliest form of skin cancers. Due to the intricacy of tumorigenesis, it is emergent to find effective therapies for melanomas. Researches have proved that pimozide inhibits the growth of melanoma, but the limited curing effect needs to be further improved. Nowadays, tumor immunotherapy has been widely recognized as the sole therapy that can eradicate cancers. Cytosine-phosphate-guanine oligodeoxynucleotide (CpG ODN), TLR9 receptor agonist, can significantly enhance anti-tumor immune responses. This study explored the therapeutic effect of pimozide combined with CpG ODN on melanoma-bearing mice. The results showed that pimozide combined with CpG ODN effectively inhibited the growth of melanoma and prolonged the survival of melanoma-bearing mice, inhibited the expression of MMP2 and p-Stat5, increased the infiltration of CD4+ and CD8+ T cells in tumor, raised the ratios of CD4+, CD8+ T cells and NK cells. These all indicated that the combination treatment improved the anti-tumor effect of pimozide on mice. The anti-tumor mechanism might be attributed to cell apoptosis induction, invasion inhibition, and immune regulation. A more effective combination treatment concerning with pimozide is being under investigation.
Collapse
|
27
|
The importance of exosomal PDL1 in tumour immune evasion. Nat Rev Immunol 2020; 20:209-215. [DOI: 10.1038/s41577-019-0264-y] [Citation(s) in RCA: 213] [Impact Index Per Article: 42.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/06/2019] [Indexed: 01/01/2023]
|
28
|
Xu X, Xiao Y, Hong B, Hao B, Qian Y. Combined detection of CA19-9 and B7-H4 in the diagnosis and prognosis of pancreatic cancer. Cancer Biomark 2020; 25:251-257. [PMID: 31282407 DOI: 10.3233/cbm-190067] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
BACKGROUND There were no specific indicators for the early detection of pancreatic cancer. OBJECTIVE To analyze the diagnostic and prognostic value of CA19-9 plus B7-H4 detection in preoperative serum or surgical tissues of patients with pancreatic cancer. METHODS One hundred and eighty-eight patients with pancreatic cancer and 25 controls were recruited. Their preoperative serum CA19-9 level was detected chemiluminescently, and B7-H4 expression in pancreatic cancer tissues was assessed immunohistochemically. The diagnostic and prognostic utility of detecting CA19-9, B7-H4 and their combination was evaluated. RESULTS CA19-9 and B7-H4 levels were significantly upregulated in patients with pancreatic cancer compared with those in controls. The diagnostic value of combined CA19-9 plus B7-H4 detection was markedly better than that achieved from their separate detection analyzed with receiver operating characteristic curve. Sensitivity and specificity of the combined detection in the pancreatic cancer group was significantly increased compared with single detection. B7-H4 detection showed better prognostic value than detection of CA19-9. However, CA19-9 had high sensitivity and low specificity, while B7-H4 showed the opposite. The sensitivity of the combined prognosis was not significantly different to B7-H4 alone. CONCLUSION The combined detection of CA19-9 and B7-H4 could become a new method for the clinical diagnosis and prognosis of pancreatic cancer.
Collapse
Affiliation(s)
- Xiangming Xu
- Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Colorectal Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yufei Xiao
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bo Hong
- Department of Pathology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Bing Hao
- Key Laboratory of Combined Multi-Organ Transplantation, Ministry of Public Health, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yun Qian
- Department of Clinical Laboratory, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| |
Collapse
|
29
|
Zhou H, Dong J, Guo L, Wang X, Wang K, Cai X, Yang S. The prognostic value of B7-H6 in esophageal squamous cell carcinoma. Sci Rep 2019; 9:18122. [PMID: 31792298 PMCID: PMC6889130 DOI: 10.1038/s41598-019-54731-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2019] [Accepted: 11/18/2019] [Indexed: 02/07/2023] Open
Abstract
B7-H6, a member of the B7 family molecules, participates in the clearance of tumor cells by binding to NKp30 on NK cells. B7-H6 expression level in esophageal squamous cell carcinoma (ESCC) and the clinical value remain unknown. The goal of this study was to determine the expression of B7-H6 in ESCC and further explore its clinical significance. We retrospectively collected the clinical data of 145 patients diagnosed with ESCC between January 2007 and December 2008. The expression of B7-H6 of the pathological tissue samples was detected by immunohistochemistry. The chi-square (χ2) test was used to analyse the relationships of B7-H6 and clinicopathological characteristics. Survival and hazard functions were estimated using the Kaplan-Meier method, and survival between groups was compared using the two-sided log-rank test. The Cox proportional hazards regression model was used to adjust for the risk factors related to overall survival (OS). 133/145 (91.72%) of the ESCC tissue samples exhibited B7-H6 expression. The expression level of B7-H6 was correlated with T stage (P = 0.036) and lymphatic metastasis status (P = 0.044). High B7-H6 expression (P = 0.003) was associated with a significantly worse OS than low B7-H6 expression. Multivariate Cox proportional hazards regression analysis demonstrated that tumour size (P = 0.021), B7-H6 expression (P = 0.025) and lymphatic metastasis status (P = 0.049) were independent prognostic factors of OS for ESCC. Collectively, our findings suggest that B7-H6 is widely expressed in ESCC samples. And B7-H6 may represent a predictor of poor prognosis for ESCC.
Collapse
Affiliation(s)
- Huan Zhou
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Jun Dong
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China
| | - Liyi Guo
- Department of Radiotherapy, Huiyang Hospital Affiliated Southern Medical University, Huizhou, Guangdong, China
| | - Xicheng Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Kailin Wang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China
| | - Xiuyu Cai
- Department of VIP Region, Sun Yat-Sen University Cancer Center, Guangzhou, Guangdong, China.
| | - Shu Yang
- Department of Oncology, The First Affiliated Hospital of Guangdong Pharmaceutical University, Guangzhou, Guangdong, China.
| |
Collapse
|
30
|
Wang JY, Wang WP. B7-H4, a promising target for immunotherapy. Cell Immunol 2019; 347:104008. [PMID: 31733822 DOI: 10.1016/j.cellimm.2019.104008] [Citation(s) in RCA: 60] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 10/23/2019] [Accepted: 11/02/2019] [Indexed: 02/07/2023]
Abstract
The coinhibitory molecule B7-H4, an important member of the B7 family, is abnormally expressed in tumors, inflammation and autoimmune diseases. B7-H4 negatively regulates T cell immune response and promotes immune escape by inhibiting the proliferation, cytokine secretion, and cell cycle of T cells. Moreover, B7-H4 plays an extremely important role in tumorigenesis and tumor development including cell proliferation, invasion, metastasis, anti-apoptosis, etc. In addition, B7-H4 has the other biological functions, such as protection against type 1 diabetes (T1D) and islet cell transplantation. Therefore, B7-H4 has been identified as a novel marker or a therapeutic target for the treatment of tumors, inflammation, autoimmune diseases, and organ transplantation. Here, we summarized the expression profiles, physiological and pathological functions, and regulatory mechanisms of B7-H4, the signaling pathways involved, as well as B7-H4-based immunotherapy.
Collapse
Affiliation(s)
- Jia-Yu Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China
| | - Wei-Peng Wang
- Center for Drug Metabolism and Pharmacokinetics, College of Pharmaceutical Sciences, Soochow University, Suzhou 215123, China.
| |
Collapse
|
31
|
Jiang Y, Lin J, Zhang J, Lu S, Wang C, Tong Y. Expression of co-inhibitory molecules B7-H4 and B7-H1 in Epstein-Barr virus positive diffuse large B-cell lymphoma and their roles in tumor invasion. Pathol Res Pract 2019; 215:152684. [PMID: 31679792 DOI: 10.1016/j.prp.2019.152684] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/26/2019] [Accepted: 10/06/2019] [Indexed: 11/25/2022]
Abstract
To investigate the relationship between immunoregulatory molecules B7-H4 and B7-H1 in Epstein-Barr positive diffuse large B-cell lymphoma (EBV+DLBCL). Immunohistochemistry was used to detect the expression of B7-H4 and B7-H1 in tumor tissues of 13 patients with EBV+DLBCL. The expression levels of B7-H4 and B7-H1 in four diffuse large B-cell lymphoma cell lines (SU-DHL-4, SU-DHL-10, SU-DHL-6, Pfeiffer) were analyzed by flow cytometry. Transwell invasion assays were conducted to observe the invasive ability of cell lines. B7-H4 and B7-H1 were expressed in 84.62% and 100% tumor specimens of EBV+DLBCL. The overexpression of B7-H4 and B7-H1 was found in 46.15% and 23.08% tumor samples of EBV+DLBCL. There was a medium negative correlation between the expression levels of B7-H4 and B7-H1 (r = -0.667, P = 0.013, spearman rank correlation). The expression levels of B7-H1 in four diffuse large B-cell lymphoma cell lines were positively correlated with their invasive ability, whereas the expression levels of B7-H4 were not. Here, we provide evidence for the negative relationship between B7-H4 and B7-H1 in EBV+DLBCL. The expression of B7-H1 in EBV+DLBCL appears to be the dominant factor which affects tumor aggressiveness. When B7-H1 expression weakens, the molecule B7-H4 may become the dominant factor of prognosis in patients with EBV+DLBCL.
Collapse
Affiliation(s)
- Ying Jiang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Jun Lin
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China
| | - Jing Zhang
- Department of Integrated Therapy, Fudan University Shanghai Cancer Center, 270 Dongan Road, Shanghai, 200032, China; Department of Oncology, Shanghai Medical College, Fudan University, 138 Yi xue yuan Road, Shanghai, 200032, China
| | - Shasha Lu
- Department of Hematology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai 200127, China
| | - Chun Wang
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| | - Yin Tong
- Department of Hematology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
32
|
Su W, Xiao W, Chen L, Zhou Q, Zheng X, Ju J, Jiang J, Wang Z. Decreased IFIT2 Expression In Human Non-Small-Cell Lung Cancer Tissues Is Associated With Cancer Progression And Poor Survival Of The Patients. Onco Targets Ther 2019; 12:8139-8149. [PMID: 31632065 PMCID: PMC6781603 DOI: 10.2147/ott.s220698] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2019] [Accepted: 09/18/2019] [Indexed: 01/05/2023] Open
Abstract
Background IFIT2 (interferon-induced proteins with tetratricopeptide repeats 2), also known as ISG54, is an important interferon-stimulated gene family protein, which has been confirmed to play a crucial role in anti-cancer as well as anti-virus process. In the present study, we aimed to investigate the IFIT2 expression in human non-small-cell cancer (NSCLC) tissues and its clinical implications. Methods The immunohistochemistry assay was used to identify the clinical significance and prognostic value of IFIT2 expression in NSCLC tissues. The depletion of IFIT2 was achieved using RNAi approach to assess the role of IFIT2 in the regulation of biological behaviors in human lung cancer cell lines. Results Decreased IFIT2 expression was found in human NSCLC tissues (both in lung adenocarcinoma and lung squamous cell carcinoma) in contrast to the adjacent normal tissues (both P<0.0001, respectively). We did not find any significant correlations between the IFIT2 expression and patient’s clinicopathological features. The survival analysis showed that the overall survival (OS) of patients in IFIT2 low expression group was significantly poorer than that in IFIT2 high expression group (in lung adenocarcinoma: P=0.027; and in lung squamous cell carcinoma: P=0.029). The Cox model analysis also indicated that the distant metastasis (P=0.043) could be used as an independent prognostic factor for lung adenocarcinoma patients, and the lymph node metastasis (P=0.045) and IFIT2 low expression (P=0.020) could be used as independent prognostic factors for lung squamous cell carcinoma patients. Moreover, the depletion of IFIT2 in human lung cancer cell lines A549, H1975 and SK-MES-1 significantly increased the cellular abilities, such as viability, migration and invasion. Conclusion Decreased IFIT2 was involved in the initiation and the progression of human NSCLC, and its underlying mechanisms still needs further investigation.
Collapse
Affiliation(s)
- Wenya Su
- Department of Respiration, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, People's Republic of China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Wenlu Xiao
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, People's Republic of China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, People's Republic of China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Qi Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, People's Republic of China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, People's Republic of China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Jingfang Ju
- Stony Brook University, Stony Brook, NY 11794, USA
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China.,Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003, People's Republic of China.,Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| | - Zhigang Wang
- Department of Respiration, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, People's Republic of China
| |
Collapse
|
33
|
Prognostic significance of resident CD103 +CD8 +T cells in human colorectal cancer tissues. Acta Histochem 2019; 121:657-663. [PMID: 31153587 DOI: 10.1016/j.acthis.2019.05.009] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 05/07/2019] [Accepted: 05/22/2019] [Indexed: 02/07/2023]
Abstract
The prognostic significance and clinical implications of resident CD103+CD8+T cells in human colorectal cancer tissues still remains largely unexplored. In our present study, we aimed to characterize the resident CD8+T cells in human colorectal cancer tissues by using double staining of CD103 and CD8, and further evaluated the prognostic significance of resident CD8+T cells in colorectal cancer. We found that the OS rate of the colorectal cancer patients with higher infiltration of CD8+T cells, or with higher numbers of resident CD103+CD8+T cells, or with higher ratio of CD103+CD8+T cells over total CD8+T cells in cancer tissues was significantly better than that of the patients with lower infiltration of CD8+T cells, or with lower numbers of resident CD103+CD8+T cells, or with higher ratio of CD103+CD8+T cells over total CD8+T cells in cancer tissues, respectively. Moreover, higher infiltration of CD8+T cells in colorectal cancer tissues was significantly and inversely correlated with advanced TNM stage. Higher numbers of resident CD103+CD8+T cells in colorectal cancer tissues were significantly and inversely correlated with distant metastasis status. Higher ratio of CD103+CD8+T cells over total CD8+T cells in colorectal cancer tissues was significantly and inversely correlated with age status. The COX model analysis demonstrated that higher infiltration of CD8+T cells, higher numbers of resident CD103+CD8+T cells, or higher ratio of CD103+CD8+T cells over total CD8+T cells in colorectal cancer tissues, could serve as independent prognostic predictors for colorectal cancer patients. Taken together, our present study demonstrated the density of tumor infiltrating CD8+T cells or the numbers of resident CD103+CD8+T cells in colorectal tissues could be used as an important prognostic predictor for this malignancy.
Collapse
|
34
|
Zheng X, Xu K, Chen L, Zhou Y, Jiang J. Prognostic value of TIM-1 expression in human non-small-cell lung cancer. J Transl Med 2019; 17:178. [PMID: 31138322 PMCID: PMC6537328 DOI: 10.1186/s12967-019-1931-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2019] [Accepted: 05/20/2019] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND T-cell immunoglobulin and mucin domain 1 (TIM-1) is an important co-stimulatory molecule which serves as a surface marker for T cell activation, especially for Th2 cells. Recently, many studies have also shown that TIM-1 can be abnormally expressed in human cancers and may have a potential role in promoting cancer progression. METHODS The immunohistochemistry was used to examine the TIM-1 expression in human non-small-cell lung carcinoma (NSCLC) tissues. The cellular studies were performed to investigate the role of TIM-1 in the regulation of biological functions of human lung cancer cell lines. RESULTS We found that the TIM-1 expression was increased in human NSCLC tissues compared with the adjacent normal tissues, and the OS rate of NSCLC patients with higher TIM-1 expression was significantly lower compared with the ones with lower TIM-1 expression. The COX model showed that higher TIM-1 expression in lung cancer tissues could be used as an independent prognostic predictor for the patients. Furthermore, we depleted TIM-1 in NSCLC cell lines A549 and SK-MES-1, and the cellular functional studies also revealed that depletion of TIM-1 could significantly inhibit the cell viability as well as the abilities of migration and invasion. In addition, our microarray data showed that certain signaling pathways were altered and enriched after depletion of TIM-1. We subsequently verified that PI3K/Akt signaling pathway was involved in the TIM-1-mediated regulation of cellular functions in NSCLC cells. CONCLUSION Our findings supported the notion that TIM-1 could serve as a potential therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003 People’s Republic of China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
| | - Kai Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003 People’s Republic of China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003 People’s Republic of China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
| | - You Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003 People’s Republic of China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, Changzhou, Jiangsu 213003 People’s Republic of China
- Institute of Cell Therapy, Soochow University, Changzhou, Jiangsu 213003 People’s Republic of China
| |
Collapse
|
35
|
Deng H, Shi H, Chen L, Zhou Y, Jiang J. Over-expression of Nectin-4 promotes progression of esophageal cancer and correlates with poor prognosis of the patients. Cancer Cell Int 2019; 19:106. [PMID: 31043861 PMCID: PMC6480822 DOI: 10.1186/s12935-019-0824-z] [Citation(s) in RCA: 52] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 04/09/2019] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Nectin-4, also known as PVRL4 (poliovirus-receptor-like 4), is specifically expressed in the embryo and placenta. Recent studies have reported that the Nectin-4 is over-expressed in multiple human cancers, and such abnormal expression is associated with cancer progression and poor prognosis of the patients. In the present study, we aimed to characterize the expression pattern of Nectin-4 in human esophageal cancer (EC) tissues, and to investigate its clinical implications, prognostic value and regulatory effects on cellular functions of EC cells. METHODS In the present study, we first examined Nectin-4 expression in human EC tissues by using immunohistochemistry (IHC) assay and analyzed the clinical associations. Then the cellular studies in vitro and the nude mice tumor model in vivo were used to examine the regulatory role of Nectin-4 in the progression of EC. RESULTS Our results demonstrated that over-expression of Nectin-4 in human EC tissues was significantly associated with tumor size, depth of tumor invasion, and poor prognosis of the patients. The intervention of Nectin-4 expression in EC cell lines showed that the increased Nectin-4 expression could significantly promote the cell viability, migration, invasion and tumor formation. CONCLUSIONS Our present data unveiled that Nectin-4 played an important role in tumor biology and could serve as a useful prognostic predictor of human EC.
Collapse
Affiliation(s)
- Haifeng Deng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Research Center for Cancer Immunotherapy of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Hongbing Shi
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Research Center for Cancer Immunotherapy of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Research Center for Cancer Immunotherapy of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - You Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Research Center for Cancer Immunotherapy of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Research Center for Cancer Immunotherapy of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| |
Collapse
|
36
|
Chen L, Zhu D, Feng J, Zhou Y, Wang Q, Feng H, Zhang J, Jiang J. Overexpression of HHLA2 in human clear cell renal cell carcinoma is significantly associated with poor survival of the patients. Cancer Cell Int 2019; 19:101. [PMID: 31015801 PMCID: PMC6469208 DOI: 10.1186/s12935-019-0813-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2019] [Accepted: 04/03/2019] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND It is well known that human clear cell renal cell carcinoma (ccRCC) is a highly immunogenic and chemo-resistant tumor. Recently, emerging data suggest that the immune checkpoint blockade therapy is an important breakthrough in the treatment against ccRCC. HHLA2, a recently reported member of B7 family, is uniquely expressed in humans but not in mice, and it plays an important role in the functional inhibition of CD4 and CD8 T cells. Herein, we aimed to study the clinical implications of HHLA2 expression in human ccRCC and its potential regulatory role in the biological functions of the cancer cells. METHODS In the present study, we examined HHLA2 expression in human ccRCC tissues and analyzed the clinical implications as well as prognostic value. The intervention of HHLA2 in human ccRCC cell lines ACHN and 786-O was performed and its effect on the cellular function of the cells was also analyzed. We also identified the differentially expressed genes upon HHLA2 knockdown in ccRCC cell lines by using gene microarray analysis. RESULTS We found that higher HHLA2 mRNA expression level in human ccRCC tissues compared with that in adjacent normal tissues based on TCGA data, and the HHLA2 expression at mRNA level was positively and significantly correlated with PD-L1, PD-L2, B7-H6, but negatively and significantly correlated with B7-H3. Moreover, our immunohistochemistry study showed that the staining intensity of HHLA2 in human ccRCC tissues was significantly higher than that in the adjacent normal tissues, and the overall survival rate of ccRCC patients with higher HHLA2 expression was significantly poorer than that of the patients with lower HHLA2 expression. Higher expression of HHLA2 in ccRCC tissues was positively and significantly associated with larger tumor size and advanced TNM stage. The COX model revealed that the parameters including patient's age, TNM stage and HHLA2 expression level could be used as the independent risk factors respectively for the prognostic prediction of the patients. Our cellular study showed that upon knockdown of HHLA2 expression in human ccRCC cell lines, the cell viability, the migration and the invasion ability were significantly inhibited, while the cell cycle arrest at G1 phase was induced and the expressions of Cyclin D1, c-Myc and Cyclin E1 were decreased. In addition, according to the microarray data, the expressions of epithelia-to-mesenchymal transition markers, such as E-cadherin, N-cadherin and Vimentin, were significantly changed after knockdown of HHLA2 expression. CONCLUSIONS Our findings indicated that HHLA2 was involved in the progression of human ccRCC and could be used as an important prognostic predictor for this malignancy.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003 Jiangsu China
| | - Dawei Zhu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003 Jiangsu China
| | - Jun Feng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003 Jiangsu China
| | - You Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003 Jiangsu China
| | - Qi Wang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003 Jiangsu China
| | - Huijing Feng
- Department of Oncology, Shanxi Academy of Medical Sciences, Shanxi Dayi Hospital, Taiyuan, 030032 Shanxi China
| | - Junping Zhang
- Department of Oncology, Shanxi Academy of Medical Sciences, Shanxi Dayi Hospital, Taiyuan, 030032 Shanxi China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, Soochow University, Changzhou, 213003 Jiangsu China
| |
Collapse
|
37
|
Jiang C, Zhu Y, Tang S, Zhang G, Lin Q, Xu Y, Shang J. High PD-L1 expression is associated with a favorable prognosis in patients with esophageal squamous cell carcinoma undergoing postoperative adjuvant radiotherapy. Oncol Lett 2018; 17:1626-1634. [PMID: 30675222 PMCID: PMC6341902 DOI: 10.3892/ol.2018.9747] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2017] [Accepted: 03/07/2018] [Indexed: 02/07/2023] Open
Abstract
Previous results on the prognostic value of programmed death-ligand (PD-L)1 expression in patients with esophageal squamous cell carcinoma (ESCC) remain limited and conflicting. The present study aimed to determine whether PD-L1 expression status predicts prognosis in patients with ESCC, particularly in those undergoing different postoperative treatments. Immunohistochemical staining for PD-L1 was performed on surgical specimens that were obtained from 246 patients with ESCC, who underwent surgical treatment but did not undergo preoperative chemotherapy, radiotherapy, targeted therapy or immune therapy. The association of PD-L1 expression with the clinicopathological factors and the association of PD-L1 expression with survival of patients with ESCC, including subgroups of patients undergoing different postoperative treatments (surgery alone, surgery with adjuvant chemotherapy, surgery with adjuvant radiotherapy and surgery with adjuvant chemo-radiotherapy groups), were statistically analyzed. Positive PD-L1 expression was significantly associated with advanced tumor-node metastasis stage (P=0.022). Median overall survival (OS) time was compared between patients with positive PD-L1 expression and those with negative PD-L1 expression in the overall patient population. In patients who were treated with postoperative adjuvant radiotherapy, the prognosis was significantly improved in patients who were PD-L1-positive compared with those who were PD-L1-negative (P=0.046). In patients treated with adjuvant chemotherapy, median OS was poorer in patients with positive PD-L1 expression compared with those with negative PD-L1 expression. However, the difference was not significant. Multivariate Cox regression analysis demonstrated that PD-L1 expression status was not an independent prognostic factor in patients with ESCC. High PD-L1 expression was associated with a favorable prognosis in patients with ESCC undergoing postoperative adjuvant radiotherapy, and it was concluded that patients with positive PD-L1 expression might benefit from postoperative adjuvant radiotherapy.
Collapse
Affiliation(s)
- Chenxue Jiang
- First Clinical Medical School, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Yaoyao Zhu
- First Clinical Medical School, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| | - Shuiqin Tang
- Department of Medical Ethics Committee, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Gu Zhang
- Department of Medical Ethics Committee, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Qingren Lin
- Department of Thoracic Oncology Radiation, Zhejiang Cancer Hospital, Hangzhou, Zhejiang 310022, P.R. China
| | - Yaping Xu
- Department of Radiation Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai 200433, P.R. China
| | - Jinbiao Shang
- First Clinical Medical School, Wenzhou Medical University, Wenzhou, Zhejiang 325035, P.R. China
| |
Collapse
|
38
|
De la Cruz-Rosas A, Martínez-Tovar A, Ramos-Peñafiel C, Collazo-Jaloma J, Olarte-Carrillo I. Pattern of differential expression of costimulatory molecules in myeloma cell line MM1.R. REVISTA MÉDICA DEL HOSPITAL GENERAL DE MÉXICO 2018. [DOI: 10.1016/j.hgmx.2016.08.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
|
39
|
Chen L, Feng J, Wu S, Xu B, Zhou Y, Wu C, Jiang J. Decreased RIG-I expression is associated with poor prognosis and promotes cell invasion in human gastric cancer. Cancer Cell Int 2018; 18:144. [PMID: 30250402 PMCID: PMC6146491 DOI: 10.1186/s12935-018-0639-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Accepted: 09/10/2018] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND Retinoic acid-induced protein I (RIG-I), known as a cytoplastic pattern recognition receptor, can recognize exogenous viral RNAs, and then initiate immune response. Recently, numerous studies also showed that RIG-I play an important role in oncogenesis and cancer progression as well. As of now, the expression pattern and the role of RIG-I in gastric cancer still remain largely unexplored. In this study, we investigated the clinical associations of RIG-I expression in human gastric cancer tissues and further explore its important contribution in the regulation of malignant phenotype of gastric cancer cells. METHODS Immunohistochemistry was performed to study the correlation between patients' clinical parameters and RIG-I expression in gastric cancer tissues. Knockdown of RIG-I was achieved by RNAi technology to examine the contribution of RIG-I in the regulation of biological functions in the cell lines of human gastric cancer. The Affymetrix GeneChip was performed to figure out the differential gene expression profile between RIG-I wild type and RIG-I knockdown cell lines of gastric cancer. RESULTS Immunohistochemistry result demonstrated that the expression of RIG-I in gastric cancer tissues significantly correlated with pathological stage and patients' prognoses. Furthermore, decreased RIG-I expression in human gastric cancer cell lines could significantly increase the cell migration, cell viability, and the ratio of cells in G2/M phase. Our microarray analysis also revealed that the differentially expressed gene profiles were enriched in related signal pathways or biological processes in KEGG or GO analysis respectively. CONCLUSIONS Our present findings showed that the decreased RIG-I expression significantly correlated with patients' prognoses, and such down-regulation could promote the cell invasion in this malignancy.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jun Feng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Shaoxian Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - You Zhou
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| |
Collapse
|
40
|
Chen L, Feng J, Xu B, Zhou Y, Zheng X, Wu C, Jiang J. B7-H6 expression in human hepatocellular carcinoma and its clinical significance [corrected]. Cancer Cell Int 2018; 18:126. [PMID: 30186042 PMCID: PMC6122564 DOI: 10.1186/s12935-018-0627-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2018] [Accepted: 08/29/2018] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Recent studies have suggested that B7-H6, a new member of the B7 family of ligands, not only is a crucial regulator of NK cell-mediated immune responses but also has important clinical implications due to its abnormal expression in many human cancers. We have previously reported that higher B7-H6 expression levels in ovarian cancer tissues are positively correlated with tumor metastasis and cancer progression. To date, the expression of B7-H6 in human hepatocellular carcinoma (HCC) and the clinical significance of B7-H6 expression still remain elusive. METHODS In the present study, the expression level of B7-H6 was examined in both HCC tissues and HCC cell lines (HepG2 and SMMC-7721). And the clinical significance of B7-H6 was analyzed as well. RESULTS Our results revealed that B7-H6 was expressed abnormally in HCC tissues, which was greatly related to tumor size. The TCGA data also showed that the B7-H6 mRNA expression level was significantly negatively correlated with the survival of HCC patients. Next, to investigate the functions of B7-H6 in HCC, we successfully constructed B7-H6 knockdown expression human HCC cell lines using the RNA interference technology. Our studies showed that reduced expression of B7-H6 in HepG2 and SMMC-7721 cells significantly attenuated cell proliferation as well as cell migration and invasion. Besides, depletion of B7-H6 greatly induced cell cycle arrest at G1 phase. And also B7-H6 knockdown in HCC cell lines dramatically decreased the C-myc, C-fos and Cyclin-D1 expression. CONCLUSIONS Our present findings suggested that B7-H6 played an important role in oncogenesis of HCC on cellular level, and B7-H6 could be employed to develop immunotherapeutic approaches targeting this malignancy.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jun Feng
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Bin Xu
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - You Zhou
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Xiao Zheng
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Changping Wu
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| | - Jingting Jiang
- Department of Tumor Biological Treatment and Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Jiangsu Engineering Research Center for Tumor Immunotherapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
- Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, 213003 Jiangsu China
| |
Collapse
|
41
|
Overexpression of low density lipoprotein receptor-related protein 1 (LRP1) is associated with worsened prognosis and decreased cancer immunity in clear-cell renal cell carcinoma. Biochem Biophys Res Commun 2018; 503:1537-1543. [PMID: 30033103 DOI: 10.1016/j.bbrc.2018.07.076] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Accepted: 07/16/2018] [Indexed: 12/12/2022]
Abstract
AIM Clear-cell renal cell carcinoma (ccRCC) is characterized with underlying genetic disorders and the role of low density lipoprotein receptor-related protein 1 (LRP1) in ccRCC is unknown. METHOD An in silico exploratory analysis using multiple public genetic datasets was used to establish association between LRP1 expression and clinicopathological parameters. Associations of interest were validated using 155 ccRCC samples using immunohistochemistry. RESULTS LRP1 was overexpressed in tumor compared with normal kidney tissue. Increased LRP1 expression in ccRCC was associated with advanced stage, grade and worsened overall survival and progression-free survival. Functional annotation indicated an immune-modulatory role of LRP1 in ccRCC. LRP1 expression was significantly correlated with expressions of PBRM1, SETD2, and KDM5C. Positive correlations between LRP1 and pro-angiogenic factors ERAP1, SCG2, STAB1, and RUNX1 were observed. LRP1 expression was positively correlated with PD-L2 level. Negative correlations between LRP1 and anti-angiogenic factors EMCN and IL18 were observed. LRP1 expression was not associated with microvessel density (MVD) yet was negatively correlated with tumor-infiltrating lymphocytes (TIL). CONCLUSION LRP1 is associated with worsened prognosis in ccRCC and is related to cancer immune modulation. LRP1-targeted therapy can be of therapeutic potential.
Collapse
|
42
|
Li J, Lee Y, Li Y, Jiang Y, Lu H, Zang W, Zhao X, Liu L, Chen Y, Tan H, Yang Z, Zhang MQ, Mak TW, Ni L, Dong C. Co-inhibitory Molecule B7 Superfamily Member 1 Expressed by Tumor-Infiltrating Myeloid Cells Induces Dysfunction of Anti-tumor CD8 + T Cells. Immunity 2018; 48:773-786.e5. [PMID: 29625896 DOI: 10.1016/j.immuni.2018.03.018] [Citation(s) in RCA: 155] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 12/20/2017] [Accepted: 03/12/2018] [Indexed: 11/30/2022]
Abstract
The molecular mechanisms whereby CD8+ T cells become "exhausted" in the tumor microenvironment remain unclear. Programmed death ligand-1 (PD-L1) is upregulated on tumor cells and PD-1-PD-L1 blockade has significant efficacy in human tumors; however, most patients do not respond, suggesting additional mechanisms underlying T cell exhaustion. B7 superfamily member 1 (B7S1), also called B7-H4, B7x, or VTCN1, negatively regulates T cell activation. Here we show increased B7S1 expression on myeloid cells from human hepatocellular carcinoma correlated with CD8+ T cell dysfunction. B7S1 inhibition suppressed development of murine tumors. Putative B7S1 receptor was co-expressed with PD-1 but not T cell immunoglobulin and mucin-domain containing-3 (Tim-3) at an activated state of early tumor-infiltrating CD8+ T cells, and B7S1 promoted T cell exhaustion, possibly through Eomes overexpression. Combinatorial blockade of B7S1 and PD-1 synergistically enhanced anti-tumor immune responses. Collectively, B7S1 initiates dysfunction of tumor-infiltrating CD8+ T cells and may be targeted for cancer immunotherapy.
Collapse
Affiliation(s)
- Jing Li
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Younghee Lee
- Departments of Immunology, MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Yanjian Li
- MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic & Systems Biology, TNLIST; School of Medicine, Tsinghua University, Beijing 100084, China
| | - Yu Jiang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Huiping Lu
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Wenjuan Zang
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Xiaohong Zhao
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Liguo Liu
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Yang Chen
- MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic & Systems Biology, TNLIST; School of Medicine, Tsinghua University, Beijing 100084, China
| | - Haidong Tan
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Zhiying Yang
- Department of Hepatobiliary Surgery, China-Japan Friendship Hospital, Beijing 100029, China
| | - Michael Q Zhang
- MOE Key Laboratory of Bioinformatics; Bioinformatics Division and Center for Synthetic & Systems Biology, TNLIST; School of Medicine, Tsinghua University, Beijing 100084, China; Department of Biological Sciences, Center for Systems Biology, The University of Texas, Dallas, TX 75080, USA
| | - Tak W Mak
- The Campbell Family Cancer Research Institute and University Health Network, Toronto, ON M5G 2C1, Canada
| | - Ling Ni
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China
| | - Chen Dong
- Institute for Immunology and School of Medicine, Tsinghua University, Beijing 100084, China; Beijing Key Lab for Immunological Research on Chronic Diseases, Beijing 100084, China.
| |
Collapse
|
43
|
Jiang T, Wu W, Zhang H, Zhang X, Zhang D, Wang Q, Huang L, Wang Y, Hang C. High expression of B7-H6 in human glioma tissues promotes tumor progression. Oncotarget 2018; 8:37435-37447. [PMID: 28415577 PMCID: PMC5514920 DOI: 10.18632/oncotarget.16391] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2016] [Accepted: 03/01/2017] [Indexed: 02/07/2023] Open
Abstract
B7-H6, a new member of B7-family ligand, also known as NCR3LG1, plays an important role in NK cells mediated immune responses. Many studies have shown that it is highly expressed in various human cancers, and its expression levels are significantly associated with cancer patients’ clinicopathological parameters and postoperative prognoses. But, still the exact role of B7-H6 expression in human glioma remains elusive. In the present study, we have characterized the B7-H6 expression in the human glioma tissues as well as glioma cell lines, U87 and U251. We observed that B7-H6 was highly expressed in the human glioma tissues, and its expression was significantly associated with cancer progression. By using the RNA interference technology, we successfully ablated B7-H6 expression in human glioma cell lines to further study its contribution towards various biological features of this malignancy. Our study identified that the B7-H6 knockdown in U87 and U251 glioma cells significantly suppressed cell proliferation, migration, invasion, and enhanced apoptosis along with induction of cell cycle arrest. It thus suggested that B7-H6 play an important role in the regulation of the biological behavior of these glioma cells. However, the detailed mechanism of B7-H6 mediated regulation of glioma cancer cell transformation and its prognostic value merits further investigation.
Collapse
Affiliation(s)
- Tianwei Jiang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Wei Wu
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Huasheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Xiangsheng Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Dingding Zhang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Qiang Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Lei Huang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Ye Wang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| | - Chunhua Hang
- Department of Neurosurgery, Jinling Hospital, School of Medicine, Nanjing University, Nanjing, Jiangsu Province, China
| |
Collapse
|
44
|
Xu B, Chen L, Li J, Zheng X, Shi L, Wu C, Jiang J. Prognostic value of tumor infiltrating NK cells and macrophages in stage II+III esophageal cancer patients. Oncotarget 2018; 7:74904-74916. [PMID: 27736796 PMCID: PMC5342711 DOI: 10.18632/oncotarget.12484] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2016] [Accepted: 09/17/2016] [Indexed: 12/18/2022] Open
Abstract
The detailed understanding of the immunobiology of tumor microenvironment has recently translated into new therapeutic approach against human cancers. Besides the role of immune cells mediating adaptive immune responses, the tumor infiltrating components of the innate immune system including, neutrophils, mast cells, NK cells, and macrophages, also role importantly in anti-tumor immunity. In our present study, we retrospectively analyzed the prognostic value of the densities of tumor infiltrating NK cells and macrophages in esophageal cancer tissues derived from stage II+III patients. Our results showed that the density of the infiltrating NK cells in tumor stroma was significantly associated with nodal status. In addition, the densities of the infiltrating NK cells in tumor nest, and the infiltrating macrophages in tumor nest as well as in tumor stroma, were significantly associated with patients' postoperative prognoses. Furthermore, the combination of infiltrating NK cells in tumor nest and stroma, or the combination of infiltrating macrophages in tumor nest and stroma, could also be used as important prognostic tool in predicting the survival of the stage II+III esophageal cancer patients.
Collapse
Affiliation(s)
- Bin Xu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| | - Jing Li
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| | - Xiao Zheng
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| | - Liangrong Shi
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Jiangsu Changzhou 213003, China
| |
Collapse
|
45
|
Song X, Shao Y, Gu W, Xu C, Mao H, Pei H, Jiang J. Prognostic role of high B7-H4 expression in patients with solid tumors: a meta-analysis. Oncotarget 2018; 7:76523-76533. [PMID: 27058425 PMCID: PMC5363528 DOI: 10.18632/oncotarget.8598] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2015] [Accepted: 03/28/2016] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND Recently, many studies have shown that B7-H4 exhibits altered expression in various cancers. We performed a meta-analysis to evaluate the prognostic role of B7-H4 expression in solid tumors. RESULTS Data from 18 observational studies and 2467 patients were summarized. An elevated baseline B7-H4 was significantly associated with worse OS (pooled HR = 1.79; 95% CI = 1.56-2.06). Differences across subgroups of tumor type, patients' ethnicity, analysis type, HR obtain method and cut-off value were not significant (PD = 0.313, PD = 0.716, PD = 0.896, PD = 0.290 and PD = 0.153, respectively). Furthermore, patients with high B7-H4 had a significantly shorter DFS (pooled HR = 2.12; 95%CI = 1.45-3.09). MATERIALS AND METHODS We searched PubMed, Embase and the Cochrane Library (last update by November 26, 2015) to identify studies assessing the effect of B7-H4 on survival of cancer patients. Pooled hazard ratios (HRs) for overall survival (OS) and disease-free survival (DFS) were estimated using fixed-effects models and random-effects models respectively. CONCLUSIONS This meta-analysis clarified that high B7-H4 expression in tissue was significantly associated with poor survival in patients with solid tumors. Future clinical studies are warranted to determine whether B7-H4 blockade has a favorable effect on disease recurrence and mortality.
Collapse
Affiliation(s)
- Xing Song
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| | - Yingjie Shao
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| | - Wendong Gu
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| | - Chao Xu
- Department of Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| | - Huihui Mao
- Department of Laboratory, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| | - Honglei Pei
- Department of Radiation Oncology, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou 213003, People's Republic of China
| |
Collapse
|
46
|
PD-1 and its ligands are important immune checkpoints in cancer. Oncotarget 2018; 8:2171-2186. [PMID: 27974689 PMCID: PMC5356790 DOI: 10.18632/oncotarget.13895] [Citation(s) in RCA: 229] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 11/21/2016] [Indexed: 12/31/2022] Open
Abstract
Checkpoint programmed death-1 (PD-1)/programmed cell death ligands (PD-Ls) have been identified as negative immunoregulatory molecules that promote immune evasion of tumor cells. The interaction of PD-1 and PD-Ls inhibits the function of T cells and tumor-infiltrating lymphocytes (TIL) while increasing the function of immunosuppressive regulatory T cells (Tregs). This condition causes the tumor cells to evade immune response. Thus, the blockade of PD-1/PD-L1 enhances anti-tumor immunity by reducing the number and/or the suppressive activity of Tregs and by restoring the activity of effector T cells. Furthermore, some monoclonal antibodies blockading PD-1/PD-Ls axis have achieved good effect and received Food and Drug Administration approval. The role of PD-1/PD-Ls in tumors has been well studied, but little is known on the mechanism by which PD-1 blocks T-cell activation. In this study, we provide a brief overview on the discovery and regulatory mechanism of PD-1 and PD-L1 dysregulation in tumors, as well as the function and signaling pathway of PD-1 and its ligands; their roles in tumor evasion and clinical treatment were also studied.
Collapse
|
47
|
Chen L, Xie Q, Wang Z, Shi L, Wu C, Jiang J. Assessment of combined expression of B7-H3 and B7-H4 as prognostic marker in esophageal cancer patients. Oncotarget 2018; 7:77237-77243. [PMID: 27764786 PMCID: PMC5363583 DOI: 10.18632/oncotarget.12628] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Accepted: 09/14/2016] [Indexed: 02/06/2023] Open
Abstract
The co-stimulatory ligands of B7-family have been confirmed to play an important role in negatively regulating the T-cell mediated anti-tumor immunity. In addition, these inhibitory molecules are also aberrantly expressed on various human cancers tissues, and significantly associated with cancer progression and patients' poor prognoses. We have previously reported that B7-H3 and B7-H4 ligands are highly expressed in human esophageal cancer tissues. Herein, we tried to further analyze the value of their combined expression on prognostic prediction for esophageal cancer patients. We found that the combined expression of both B7-H3 and B7-H4 could be used as a valuable risk factor for predicting the prognosis of esophageal cancer patients (P=0.003). Moreover the status of these patients with high expression of both B7-H3 and B7-H4, was positively and significantly associated with the tumor invasion depth (P=0.0414) and TNM stage (P=0.0414). The Cox multivariate proportional hazards regression analysis revealed that the tumor size (P=0.007), the TNM stage (P=0.024) and the status of both B7-H3 and B7-H4 high expression (P=0.011), could be used as an independent risk factor for predicting patients' postoperative prognosis, respectively. In conclusion, our data indicated that the combined application of B7-H3 and B7-H4 expression can be effectively used as a prognostic marker in esophageal cancer patients.
Collapse
Affiliation(s)
- Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Quanqin Xie
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Zhigang Wang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Department of Respiratory Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Liangrong Shi
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Changping Wu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| | - Jingting Jiang
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Research Center for Cancer Immunotherapy Technology of Jiangsu Province, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China.,Institute of Cell Therapy, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, China
| |
Collapse
|
48
|
Zhang X, Cai L, Zhang G, Shen Y, Huang J. B7-H4 promotes tumor growth and metastatic progression in lung cancer by impacting cell proliferation and survival. Oncotarget 2017; 8:18861-18871. [PMID: 28061481 PMCID: PMC5386653 DOI: 10.18632/oncotarget.14475] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 12/16/2016] [Indexed: 12/21/2022] Open
Abstract
Aberrant expression of B7-H4 occurs across a broad spectrum of human cancers. The aim of this study was to investigate the key role of B7-H4 during tumorigenesis and metastasis of human lung cancer. Our data showed that the shRNA-mediated disruption of B7-H4 markedly inhibited tumor cell proliferation, invasion and migration, increased cell apoptosis and arrested cell cycle at G0/G1. These changes were accompanied by a marked increase in Bax and caspase-3/caspase-8, but a decrease in Bcl-2, cyclinD1 and activation of AKT. In addition, our shRNA-mediated disruption of B7-H4 led to a marked decrease in tumor growth in the immune-compromised mice. Importantly, B7-H4 was expressed in 53.33% of lung carcinomas from our patient cohort (n = 90), but not in any of adjacent non-cancerous tissues, according to our IHC analyses. In particular, B7-H4 expression appeared to be associated with lymph node metastasis (P = 0.008) and TNM stage (P = 0.012). Taken together, our study demonstrates a strong promoting role of B7-H4 in lung tumor growth, progression and metastasis, and supports its potential as a therapeutic target for the treatment of the disease.
Collapse
Affiliation(s)
- Xiuqin Zhang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Department of Respiratory Medicine, The Affiliated Hospital of Jiangnan University (WuXi No.4 People's Hospital), Wuxi 214000, China
| | - Liming Cai
- Department of Respiratory Medicine, The Affiliated Hospital of Jiangnan University (WuXi No.4 People's Hospital), Wuxi 214000, China
| | - Guangbo Zhang
- Clinical Immunology Laboratory of Jiangsu Province, Suzhou 215006, China
| | - Yu Shen
- Clinical Immunology Laboratory of Jiangsu Province, Suzhou 215006, China
| | - Jianan Huang
- Department of Respiratory Medicine, The First Affiliated Hospital of Soochow University, Suzhou 215006, China.,Clinical Immunology Laboratory of Jiangsu Province, Suzhou 215006, China
| |
Collapse
|
49
|
Mao Y, Chen L, Wang F, Zhu D, Ge X, Hua D, Sun J. Cancer cell-expressed B7-H3 regulates the differentiation of tumor-associated macrophages in human colorectal carcinoma. Oncol Lett 2017; 14:6177-6183. [PMID: 29113264 DOI: 10.3892/ol.2017.6935] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 06/15/2017] [Indexed: 12/15/2022] Open
Abstract
Co-stimulatory molecule B7 homolog 3 protein (B7-H3) has been described as an important tumor antigen in various human tumors. The exact role of B7-H3 in tumor progression and its receptor are still ambiguous. The phenotype and the function of tumor-associated macrophages (TAMs) in human solid tumors are complicated and could contribute to the shaping of the tumor microenvironment. In the present study, B7-H3 expression and lymphocyte infiltration were investigated by immunohistochemistry in 117 colorectal carcinoma (CRC) patients. B7-H3 expression was positively associated with the infiltrating density of macrophage in CRC tissues, and B7-H3 expression and the infiltrating density of macrophages were negatively associated with the overall survival rate of patients. The putative B7-H3 receptor was found on activated monocytes and macrophages, indicating the direct function of B7-H3 signal on macrophages. Additional results revealed that during the differentiation of TAMs, B7-H3 promoted the polarization of type 2 macrophages (M2s) and switch of the M1 phenotype to the M2 phenotype. Thus, B7-H3 signaling promotes M2 differentiation via the putative receptor on monocytes and macrophages. Targeting the manipulation of TAMs through the B7-H3 pathway may be valuable for the development of novel immunotherapeutic strategies against human CRC.
Collapse
Affiliation(s)
- Yong Mao
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China.,Department of Immunology, The Fourth Hospital of Wuxi, Wuxi, Jiangsu 214062, P.R. China
| | - Lujun Chen
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Fengming Wang
- Testing Center, Center for Disease Prevention and Control, Changzhou, Jiangsu 213000, P.R. China
| | - Dawei Zhu
- Department of Tumor Biological Treatment, The Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu 213003, P.R. China
| | - Xiaosong Ge
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China
| | - Dong Hua
- Department of Oncology, The Affiliated Hospital of Jiangnan University, Wuxi, Jiangsu 214062, P.R. China.,Department of Immunology, The Fourth Hospital of Wuxi, Wuxi, Jiangsu 214062, P.R. China
| | - Jing Sun
- Department of Immunology, Medical College of Soochow University, Suzhou, Jiangsu 215123, P.R. China
| |
Collapse
|
50
|
Clinical significance of the B7-H4 as a novel prognostic marker in breast cancer. Gene 2017; 623:24-28. [DOI: 10.1016/j.gene.2017.04.003] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Revised: 03/29/2017] [Accepted: 04/04/2017] [Indexed: 01/20/2023]
|