3
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Advances in immunology and immunotherapy for mesenchymal gastrointestinal cancers

      review-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

          Mesenchymal gastrointestinal cancers are represented by the gastrointestinal stromal tumors (GISTs) which occur throughout the whole gastrointestinal tract, and affect human health and economy globally. Curative surgical resections and tyrosine kinase inhibitors (TKIs) are the main managements for localized GISTs and recurrent/metastatic GISTs, respectively. Despite multi-lines of TKIs treatments prolonged the survival time of recurrent/metastatic GISTs by delaying the relapse and metastasis of the tumor, drug resistance developed quickly and inevitably, and became the huge obstacle for stopping disease progression. Immunotherapy, which is typically represented by immune checkpoint inhibitors (ICIs), has achieved great success in several solid tumors by reactivating the host immune system, and been proposed as an alternative choice for GIST treatment. Substantial efforts have been devoted to the research of immunology and immunotherapy for GIST, and great achievements have been made. Generally, the intratumoral immune cell level and the immune-related gene expressions are influenced by metastasis status, anatomical locations, driver gene mutations of the tumor, and modulated by imatinib therapy. Systemic inflammatory biomarkers are regarded as prognostic indicators of GIST and closely associated with its clinicopathological features. The efficacy of immunotherapy strategies for GIST has been widely explored in pre-clinical cell and mouse models and clinical experiments in human, and some patients did benefit from ICIs. This review comprehensively summarizes the up-to-date advancements of immunology, immunotherapy and research models for GIST, and provides new insights and perspectives for future studies.

          Related collections

          Most cited references281

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

          Hallmarks of Cancer: New Dimensions

          The hallmarks of cancer conceptualization is a heuristic tool for distilling the vast complexity of cancer phenotypes and genotypes into a provisional set of underlying principles. As knowledge of cancer mechanisms has progressed, other facets of the disease have emerged as potential refinements. Herein, the prospect is raised that phenotypic plasticity and disrupted differentiation is a discrete hallmark capability, and that nonmutational epigenetic reprogramming and polymorphic microbiomes both constitute distinctive enabling characteristics that facilitate the acquisition of hallmark capabilities. Additionally, senescent cells, of varying origins, may be added to the roster of functionally important cell types in the tumor microenvironment. SIGNIFICANCE: Cancer is daunting in the breadth and scope of its diversity, spanning genetics, cell and tissue biology, pathology, and response to therapy. Ever more powerful experimental and computational tools and technologies are providing an avalanche of "big data" about the myriad manifestations of the diseases that cancer encompasses. The integrative concept embodied in the hallmarks of cancer is helping to distill this complexity into an increasingly logical science, and the provisional new dimensions presented in this perspective may add value to that endeavor, to more fully understand mechanisms of cancer development and malignant progression, and apply that knowledge to cancer medicine.
            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

              IFN-γ–related mRNA profile predicts clinical response to PD-1 blockade

              Programmed death-1–directed (PD-1–directed) immune checkpoint blockade results in durable antitumor activity in many advanced malignancies. Recent studies suggest that IFN-γ is a critical driver of programmed death ligand-1 (PD-L1) expression in cancer and host cells, and baseline intratumoral T cell infiltration may improve response likelihood to anti–PD-1 therapies, including pembrolizumab. However, whether quantifying T cell–inflamed microenvironment is a useful pan-tumor determinant of PD-1–directed therapy response has not been rigorously evaluated. Here, we analyzed gene expression profiles (GEPs) using RNA from baseline tumor samples of pembrolizumab-treated patients. We identified immune-related signatures correlating with clinical benefit using a learn-and-confirm paradigm based on data from different clinical studies of pembrolizumab, starting with a small pilot of 19 melanoma patients and eventually defining a pan-tumor T cell–inflamed GEP in 220 patients with 9 cancers. Predictive value was independently confirmed and compared with that of PD-L1 immunohistochemistry in 96 patients with head and neck squamous cell carcinoma. The T cell–inflamed GEP contained IFN-γ–responsive genes related to antigen presentation, chemokine expression, cytotoxic activity, and adaptive immune resistance, and these features were necessary, but not always sufficient, for clinical benefit. The T cell–inflamed GEP has been developed into a clinical-grade assay that is currently being evaluated in ongoing pembrolizumab trials.
                Bookmark

                Author and article information

                Contributors
                cxdeng@um.edu.mo
                zenglli6@mail.sysu.edu.cn
                heyulong@mail.sysu.edu.cn
                zhchangh@mail.sysu.edu.cn
                Journal
                Mol Cancer
                Mol Cancer
                Molecular Cancer
                BioMed Central (London )
                1476-4598
                18 April 2023
                18 April 2023
                2023
                : 22
                : 71
                Affiliations
                [1 ]GRID grid.511083.e, ISNI 0000 0004 7671 2506, Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, , The Seventh Affiliated Hospital of Sun Yat-Sen University, ; Shenzhen, 518107 Guangdong China
                [2 ]GRID grid.511083.e, ISNI 0000 0004 7671 2506, Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research, Scientific Research Center, , The Seventh Affiliated Hospital of Sun Yat-Sen University, ; Shenzhen, 518107 Guangdong China
                [3 ]GRID grid.511083.e, ISNI 0000 0004 7671 2506, Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research, Digestive Diseases Center, , The Seventh Affiliated Hospital of Sun Yat-Sen University, ; Shenzhen, 518107 Guangdong China
                [4 ]GRID grid.440601.7, ISNI 0000 0004 1798 0578, Shenzhen Key Laboratory for Drug Addiction and Medication Safety, Department of Ultrasound, , Peking University Shenzhen Hospital, ; Shenzhen, 518036 China
                [5 ]GRID grid.437123.0, ISNI 0000 0004 1794 8068, MOE Frontiers Science Center for Precision Oncology, Faculty of Health Sciences, , Institute of Translational Medicine, Cancer Center, University of Macau, ; Macau SAR, 999078 China
                [6 ]GRID grid.412615.5, ISNI 0000 0004 1803 6239, Department of Gastrointestinal Surgery, , The First Affiliated Hospital of Sun Yat-Sen University, ; No. 58 Zhongshan Road, Guangzhou, 510080 China
                Article
                1770
                10.1186/s12943-023-01770-6
                10111719
                37072770
                6c8b8bbf-8f88-4255-b20a-0381c374f3a3
                © The Author(s) 2023

                Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver ( http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

                History
                : 9 March 2023
                : 29 March 2023
                Funding
                Funded by: Guangdong Provincial Key Laboratory of Digestive Cancer Research
                Award ID: 2021B1212040006
                Award Recipient :
                Funded by: Guangdong-Hong Kong-Macau University Joint Laboratory of Digestive Cancer Research
                Award ID: 2021LSYS003
                Award Recipient :
                Funded by: Shenzhen Key Laboratory of Chinese Medicine Active Substance Screening and Translational Research
                Award ID: ZDSYS20220606100801003
                Award Recipient :
                Funded by: Guangdong Basic and Applied Basic Research Foundation
                Award ID: 2022A1515110194
                Award ID: 2021B1515120069
                Award ID: 2022A1515110156
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100017607, Shenzhen Fundamental Research Program;
                Award ID: JCYJ20220530142203008
                Award ID: JCYJ20190809142807444
                Award Recipient :
                Funded by: Shenzhen-Hong Kong-Macau Technology Research Program (Type C)
                Award ID: SGDX20201103092601008
                Award Recipient :
                Funded by: National Natural Science Foundation of China
                Award ID: U20A20379
                Award Recipient :
                Funded by: Shenzhen Sustainable Project
                Award ID: KCXFZ20200201101059392
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100012151, Sanming Project of Medicine in Shenzhen;
                Award ID: SZSM201911010
                Award Recipient :
                Categories
                Review
                Custom metadata
                © The Author(s) 2023

                Oncology & Radiotherapy
                gastrointestinal cancers,gastrointestinal stromal tumor,resistance,immunology,immunotherapy,imatinib,model,immune checkpoint inhibitors

                Comments

                Comment on this article