11
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Programmed death‐ligand 1 is prognostic factor in esophageal squamous cell carcinoma and is associated with epidermal growth factor receptor

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Programmed death‐ligand 1 ( PD‐L1) expression either indicates immune inhibitory status or concurrent immune response. Although the relationship between PD‐L1 and clinical outcomes has been studied widely in recent years, its role in prognosis of esophageal squamous cell carcinoma ( ESCC) remains unclear. Here, we assessed the significance of PD‐L1 in ESCC and its association with epidermal growth factor receptor ( EGFR) and radiation response. We found that PD‐L1 was present both on the surface of tumor cells and tumor‐infiltrating immune cells. Patients with tumor‐infiltrating immune cell PD‐L1 expression had better survival. PD‐L1 expression on immune cells was an independent prognostic factor for patients with ESCC. PD‐L1 expression either on tumor‐infiltrating immune cells or tumor cells was negatively associated with EGFR expression. EGFR/ PD‐L1 pairs could separate the survival between EGFR low/ PD‐L1 positive and EGFR high/ PD‐L1 negative groups. In ESCC cell lines with EGFR high expression, PD‐L1 expression was induced significantly when EGFR signaling was activated by radiation and was dramatically inhibited by an EGFR tyrosine kinase inhibitor. In conclusion, tumor‐infiltrating immune cell PD‐L1 expression is an independent prognostic factor for ESCC, and the association between EGFR and PD‐L1 is vital to determining survival. It is important to consider radiotherapy‐induced imbalance of pro‐tumor and anti‐tumor immune response. A combination of radiotherapy and PD‐L1‐targeted therapy could be a promising therapeutic strategy for ESCC patients.

          Related collections

          Most cited references18

          • Record: found
          • Abstract: found
          • Article: not found

          Activation of the PD-1 pathway contributes to immune escape in EGFR-driven lung tumors.

          The success in lung cancer therapy with programmed death (PD)-1 blockade suggests that immune escape mechanisms contribute to lung tumor pathogenesis. We identified a correlation between EGF receptor (EGFR) pathway activation and a signature of immunosuppression manifested by upregulation of PD-1, PD-L1, CTL antigen-4 (CTLA-4), and multiple tumor-promoting inflammatory cytokines. We observed decreased CTLs and increased markers of T-cell exhaustion in mouse models of EGFR-driven lung cancer. PD-1 antibody blockade improved the survival of mice with EGFR-driven adenocarcinomas by enhancing effector T-cell function and lowering the levels of tumor-promoting cytokines. Expression of mutant EGFR in bronchial epithelial cells induced PD-L1, and PD-L1 expression was reduced by EGFR inhibitors in non-small cell lung cancer cell lines with activated EGFR. These data suggest that oncogenic EGFR signaling remodels the tumor microenvironment to trigger immune escape and mechanistically link treatment response to PD-1 inhibition. We show that autochthonous EGFR-driven lung tumors inhibit antitumor immunity by activating the PD-1/PD-L1 pathway to suppress T-cell function and increase levels of proinflammatory cytokines. These findings indicate that EGFR functions as an oncogene through non-cell-autonomous mechanisms and raise the possibility that other oncogenes may drive immune escape. ©2013 AACR.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Programmed death ligand-1 expression in non-small cell lung cancer.

            Recent strategies targeting the interaction of the programmed cell death ligand-1 (PD-L1, B7-H1, CD274) with its receptor, PD-1, resulted in promising activity in early phase clinical trials. In this study, we used various antibodies and in situ mRNA hybridization to measure PD-L1 in non-small cell lung cancer (NSCLC) using a quantitative fluorescence (QIF) approach to determine the frequency of expression and prognostic value in two independent populations. A control tissue microarray (TMA) was constructed using PD-L1-transfected cells, normal human placenta and known PD-L1-positive NSCLC cases. Only one of four antibodies against PD-L1 (5H1) validated for specificity on this TMA. In situ PD-L1 mRNA using the RNAscope method was similarly validated. Two cohorts of NSCLC cases in TMAs including 340 cases from hospitals in Greece and 204 cases from Yale University were assessed. Tumors showed PD-L1 protein expression in 36% (Greek) and 25% (Yale) of the cases. PD-L1 expression was significantly associated with tumor-infiltrating lymphocytes in both cohorts. Patients with PD-L1 (both protein and mRNA) expression above the detection threshold showed statistically significant better outcome in both series (log-rank P=0.036 and P=0.027). Multivariate analysis showed that PD-L1 expression was significantly associated with better outcome independent of histology. Measurement of PD-L1 requires specific conditions and some commercial antibodies show lack of specificity. Expression of PD-L1 protein or mRNA is associated with better outcome. Further studies are required to determine the value of this marker in prognosis and prediction of response to treatments targeting this pathway.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              IFN-γ from lymphocytes induces PD-L1 expression and promotes progression of ovarian cancer

              Background: PD-L1 (programmed cell death 1 ligand 1) on tumour cells suppresses host immunity through binding to its receptor PD-1 on lymphocytes, and promotes peritoneal dissemination in mouse models of ovarian cancer. However, how PD-L1 expression is regulated in ovarian cancer microenvironment remains unclear. Methods: The number of CD8-positive lymphocytes and PD-L1 expression in tumour cells was assessed in ovarian cancer clinical samples. PD-L1 expression and tumour progression in mouse models under conditions of altering IFN-γ signals was assessed. Results: The number of CD8-positive cells in cancer stroma was very high in peritoneally disseminated tumours, and was strongly correlated to PD-L1 expression on the tumour cells (P<0.001). In mouse models, depleting IFNGR1 (interferon-γ receptor 1) resulted in lower level of PD-L1 expression in tumour cells, increased the number of tumour-infiltrating CD8-positive lymphocytes, inhibition of peritoneal disseminated tumour growth and longer survival (P=0.02). The injection of IFN-γ into subcutaneous tumours induced PD-L1 expression and promoted tumour growth, and PD-L1 depletion completely abrogated tumour growth caused by IFN-γ injection (P=0.01). Conclusions: Interferon-γ secreted by CD8-positive lymphocytes upregulates PD-L1 on ovarian cancer cells and promotes tumour growth. The lymphocyte infiltration and the IFN-γ status may be the key to effective anti-PD-1 or anti-PD-L1 therapy in ovarian cancer.
                Bookmark

                Author and article information

                Contributors
                wangping@tjmuch.com
                xiaozefen2013@163.com
                Journal
                Cancer Sci
                Cancer Sci
                10.1111/(ISSN)1349-7006
                CAS
                Cancer Science
                John Wiley and Sons Inc. (Hoboken )
                1347-9032
                1349-7006
                25 April 2017
                April 2017
                : 108
                : 4 ( doiID: 10.1111/cas.2017.108.issue-4 )
                : 590-597
                Affiliations
                [ 1 ] Department of Radiation Oncology National Clinical Research Center of Cancer Key Laboratory of Cancer Prevention and TherapyTianjin Medical University Cancer Institute and Hospital TianjinChina
                [ 2 ] Department of Radiation PhysicsUniversity of Texas MD Anderson Cancer Center Houston TexasUSA
                [ 3 ] Departments of Immunology National Clinical Research Center of Cancer Key Laboratory of Cancer Prevention and TherapyTianjin Medical University Cancer Institute and Hospital TianjinChina
                [ 4 ] Department of Radiation Oncology National Cancer Center/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical College Chaoyang District BeijingChina
                [ 5 ] Department of Pathology National Clinical Research Center of Cancer Key Laboratory of Cancer Prevention and TherapyTianjin Medical University Cancer Institute and Hospital TianjinChina
                Author notes
                [*] [* ] Correspondence

                Zefen Xiao, Department of Radiation Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Pan Jia Yuan Nanli number 17, Chaoyang District, Beijing 100021, China.

                Tel: +086‐10‐8778‐8503; Fax: +086‐10‐6770‐6153;

                E‐mail: xiaozefen2013@ 123456163.com

                and

                Ping Wang, Departments of Radiation Oncology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Cancer, Key Laboratory of Cancer Prevention and Therapy, Huan‐Hu‐Xi Road, Ti‐Yuan‐Bei, He XiDistrict, Tianjin 300060, China.

                Tel: +086‐22‐2351‐9953; Fax: +086‐22‐2334‐1405;

                E‐mail: wangping@ 123456tjmuch.com

                Author information
                http://orcid.org/0000-0002-4290-0379
                Article
                CAS13197
                10.1111/cas.13197
                5406530
                28192623
                dc165704-9d4b-4585-bae9-c41ca3ffa23b
                © 2017 The Authors. Cancer Science published by John Wiley & Sons Australia, Ltd on behalf of Japanese Cancer Association.

                This is an open access article under the terms of the Creative Commons Attribution‐NonCommercial License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 18 December 2016
                : 30 January 2017
                : 04 February 2017
                Page count
                Figures: 4, Tables: 2, Pages: 8, Words: 6851
                Funding
                Funded by: Science and Technology Project of Beijing
                Award ID: Z121107001012004
                Funded by: Capital Foundation for Medical Research and Development
                Award ID: 2007–2012
                Funded by: National Natural Science Foundation
                Award ID: 81272512
                Funded by: Chinese Hi‐Tech R&D Program
                Award ID: 2012AA02A503
                Categories
                Original Article
                Original Articles
                Basic and Clinical Immunology
                Custom metadata
                2.0
                cas13197
                April 2017
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.0.9 mode:remove_FC converted:27.04.2017

                Oncology & Radiotherapy
                epidermal growth factor receptor,esophageal squamous cell carcinoma,programmed death‐ligand 1,radiation,tumor‐infiltrating immune cell

                Comments

                Comment on this article