14
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Blocking Triggering Receptor Expressed on Myeloid Cells‐1‐Positive Tumor‐Associated Macrophages Induced by Hypoxia Reverses Immunosuppression and Anti‐Programmed Cell Death Ligand 1 Resistance in Liver Cancer

      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

          Tumor‐associated macrophages (TAMs) are recognized as antitumor suppressors, but how TAMs behave in the hypoxic environment of hepatocellular carcinoma (HCC) remains unclear. Here, we demonstrated that hypoxia inducible factor 1α induced increased expression of triggering receptor expressed on myeloid cells‐1 (TREM‐1) in TAMs, resulting in immunosuppression. Specifically, TREM‐1‐positive (TREM‐1 +) TAMs abundant at advanced stages of HCC progression indirectly impaired the cytotoxic functions of CD8 + T cells and induced CD8 + T‐cells apoptosis. Biological and functional assays showed that TREM‐1 + TAMs had higher expression of programmed cell death ligand 1 (PD‐L1) under hypoxic environment. However, TREM‐1 + TAMs could abrogate spontaneous and PD‐L1‐blockade‐mediated antitumor effects in vivo, suggesting that TREM‐1 + TAM‐induced immunosuppression was dependent on a pathway separate from PD‐L1/programmed cell death 1 axis. Moreover, TREM‐1 + TAM‐associated regulatory T cells (Tregs) were crucial for HCC resistance to anti‐PD‐L1 therapy. Mechanistically, TREM‐1 + TAMs elevated chemokine (C‐C motif) ligand 20 expression through the extracellular signal‐regulated kinase/NF‐κβ pathway in response to hypoxia and tumor metabolites leading to CCR6 +Foxp3 + Treg accumulation. Blocking the TREM‐1 pathway could significantly inhibit tumor progression, reduce CCR6 +Foxp3 + Treg recruitment, and improve the therapeutic efficacy of PD‐L1 blockade. Thus, these data demonstrated that CCR6 +Foxp3 + Treg recruitment was crucial for TREM‐1 + TAM‐mediated anti‐PD‐L1 resistance and immunosuppression in hypoxic tumor environment. Conclusion: This study highlighted that the hypoxic environment initiated the onset of tumor immunosuppression through TREM‐1 + TAMs attracting CCR6 +Foxp3 + Tregs, and TREM‐1 + TAMs endowed HCC with anti‐PD‐L1 therapy resistance.

          Related collections

          Most cited references49

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

          TIMER: A Web Server for Comprehensive Analysis of Tumor-Infiltrating Immune Cells.

          Recent clinical successes of cancer immunotherapy necessitate the investigation of the interaction between malignant cells and the host immune system. However, elucidation of complex tumor-immune interactions presents major computational and experimental challenges. Here, we present Tumor Immune Estimation Resource (TIMER; cistrome.shinyapps.io/timer) to comprehensively investigate molecular characterization of tumor-immune interactions. Levels of six tumor-infiltrating immune subsets are precalculated for 10,897 tumors from 32 cancer types. TIMER provides 6 major analytic modules that allow users to interactively explore the associations between immune infiltrates and a wide spectrum of factors, including gene expression, clinical outcomes, somatic mutations, and somatic copy number alterations. TIMER provides a user-friendly web interface for dynamic analysis and visualization of these associations, which will be of broad utilities to cancer researchers. Cancer Res; 77(21); e108-10. ©2017 AACR.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            PD-1 blockade induces responses by inhibiting adaptive immune resistance

            Therapies that target the programmed death-1 (PD-1) receptor have shown unprecedented rates of durable clinical responses in patients with various cancer types. 1–5 One mechanism by which cancer tissues limit the host immune response is via upregulation of PD-1 ligand (PD-L1) and its ligation to PD-1 on antigen-specific CD8 T-cells (termed adaptive immune resistance). 6,7 Here we show that pre-existing CD8 T-cells distinctly located at the invasive tumour margin are associated with expression of the PD-1/PD-L1 immune inhibitory axis and may predict response to therapy. We analyzed samples from 46 patients with metastatic melanoma obtained before and during anti-PD1 therapy (pembrolizumab) using quantitative immunohistochemistry, quantitative multiplex immunofluorescence, and next generation sequencing for T-cell receptors (TCR). In serially sampled tumours, responding patients showed proliferation of intratumoural CD8+ T-cells that directly correlated with radiographic reduction in tumour size. Pre-treatment samples obtained from responding patients showed higher numbers of CD8, PD1, and PD-L1 expressing cells at the invasive tumour margin and inside tumours, with close proximity between PD-1 and PD-L1, and a more clonal TCR repertoire. Using multivariate analysis, we established a predictive model based on CD8 expression at the invasive margin and validated the model in an independent cohort of 15 patients. Our findings indicate that tumour regression following therapeutic PD-1 blockade requires pre-existing CD8+ T cells that are negatively regulated by PD-1/PD-L1 mediated adaptive immune resistance.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Tumour-associated macrophages as treatment targets in oncology

              Tumour-associated macrophages (TAMs) are key drivers of tumour-promoting inflammation and cancer progression, and are important determinants of responsiveness to a range of therapies. Herein, the authors summarize the roles of TAMs in cancer, and discuss the potential of TAM-targeted therapeutic strategies to complement and synergize with other anticancer treatments.
                Bookmark

                Author and article information

                Contributors
                zhoulin99@zju.edu.cn
                shusenzheng@zju.edu.cn
                Journal
                Hepatology
                Hepatology
                10.1002/(ISSN)1527-3350
                HEP
                Hepatology (Baltimore, Md.)
                John Wiley and Sons Inc. (Hoboken )
                0270-9139
                1527-3350
                12 April 2019
                July 2019
                : 70
                : 1 ( doiID: 10.1002/hep.v70.1 )
                : 198-214
                Affiliations
                [ 1 ] Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery The First Affiliated Hospital, School of Medicine, Zhejiang University Hangzhou China
                [ 2 ] NHFPC Key Laboratory of Combined Multi‐organ Transplantation Hangzhou China
                [ 3 ] Key Laboratory of the Diagnosis and Treatment of Organ Transplantation CAMS Hangzhou China
                [ 4 ] Key Laboratory of Organ Transplantation Zhejiang Province Hangzhou China
                [ 5 ] Collaborative Innovation Center for Diagnosis Treatment of Infectious Diseases Hangzhou China
                [ 6 ] Department of Hepatobiliary and Pancreatic Surgery Taihe Hospital, Hubei University of Medicine Hubei China
                Author notes
                [*] [* ] Address Correspondence and Reprint Requests to:

                Shusen Zheng, M.D., Ph.D.

                Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University

                No. 79 Qing chun Road

                Hangzhou 310003, China

                E‐mail: shusenzheng@ 123456zju.edu.cn

                Tel.: +1‐86‐571‐87236466

                or

                Lin Zhou, M.D., Ph.D.

                Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, First Affiliated Hospital, School of Medicine, Zhejiang University

                No. 79 Qing chun Road

                Hangzhou 310003, China

                E‐mail: zhoulin99@ 123456zju.edu.cn

                Tel.: +1‐86‐571‐87236466

                [†]

                These authors contributed equally to this work.

                Author information
                https://orcid.org/0000-0003-1459-8261
                Article
                HEP30593
                10.1002/hep.30593
                6618281
                30810243
                f25b36e8-3b29-49a8-895b-0d06a1ee3579
                © 2019 The Authors. Hepatology published by Wiley Periodicals, Inc., on behalf of American Association for the Study of Liver Diseases.

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ 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
                : 11 July 2018
                : 20 February 2019
                Page count
                Figures: 7, Tables: 0, Pages: 2, Words: 17109
                Funding
                Funded by: Science and Technology Project of Health Department of Zhejiang province
                Award ID: No. 2018KY397
                Funded by: National S&T Major Project
                Award ID: No. 2017ZX10203205
                Funded by: Major program of National Natural Science Foundation of China
                Award ID: No. 91542205
                Funded by: National Natural Science Foundation of China
                Award ID: 81572954
                Funded by: Innovative Research Groups of National Natural Science Foundation of China
                Award ID: No. 81721091
                Categories
                Original Article
                Original Articles
                Hepatobiliary Malignancies
                Custom metadata
                2.0
                hep30593
                July 2019
                Converter:WILEY_ML3GV2_TO_NLMPMC version:5.6.5 mode:remove_FC converted:10.07.2019

                Gastroenterology & Hepatology
                Gastroenterology & Hepatology

                Comments

                Comment on this article