15
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      TIGIT and PD1 Co-blockade Restores ex vivo Functions of Human Tumor-Infiltrating CD8 + T Cells in Hepatocellular Carcinoma

      research-article
      1 , 1 , 1 , 1 , 1 , 1 , 2 , 3 , 3 , 1 , 4 , 5 , 6 , 3 , 1 , 7 , 1 , b , 1 , , b
      Cellular and Molecular Gastroenterology and Hepatology
      Elsevier
      TIGIT, CD226, TOX, HCC, Immunotherapy, AFP, alpha fetoprotein, APC, antigen-presenting cell, cDC, conventional dendritic cells, HCC, hepatocellular carcinoma, IFN, interferon, LAG3, lymphocyte-activation gene 3, MFI, median fluorescent intensity, PD1, programmed cell death protein 1, PD-L1, programmed death-ligand 1, PMA, phorbol 12-myristate 13-acetate, SEM, standard error of the mean, TCF1, transcription factor 1, TFL, tumor-free liver tissue, TIGIT, T-cell immune receptor with Ig and ITIM domains, TIL, tumor-infiltrating leukocyte, TIM3, T-cell immunoglobulin and mucin-domain containing-3, TMA, tissue microarray, TNF, tumor necrosis factor, TOX, thymocyte selection-associated high mobility group box protein, Treg, regulatory T cells

      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

          Background & Aims

          TIGIT is a co-inhibitory receptor, and its suitability as a target for cancer immunotherapy in HCC is unknown. PD1 blockade is clinically effective in about 20% of advanced HCC patients. Here we aim to determine whether co-blockade of TIGIT/PD1 has added value to restore functionality of HCC tumor-infiltrating T cells (TILs).

          Methods

          Mononuclear leukocytes were isolated from tumors, paired tumor-free liver tissues (TFL) and peripheral blood of HCC patients, and used for flow cytometric phenotyping and functional assays. CD3/CD28 T-cell stimulation and antigen-specific assays were used to study the ex vivo effects of TIGIT/PD1 single or dual blockade on T-cell functions.

          Results

          TIGIT was enriched, whereas its co-stimulatory counterpart CD226 was down-regulated on PD1 high CD8 + TILs. PD1 high TIGIT + CD8 + TILs co-expressed exhaustion markers TIM3 and LAG3 and demonstrated higher TOX expression. Furthermore, this subset showed decreased capacity to produce IFN-γ and TNF-α. Expression of TIGIT-ligand CD155 was up-regulated on tumor cells compared with hepatocytes in TFL. Whereas single PD1 blockade preferentially enhanced ex vivo functions of CD8 + TILs from tumors with PD1 high CD8 + TILs (high PD1 expressers), co-blockade of TIGIT and PD1 improved proliferation and cytokine production of CD8 + TILs from tumors enriched for PD1 int CD8 + TILs (low PD1 expressers). Importantly, ex vivo co-blockade of TIGIT/PD1 improved proliferation, cytokine production, and cytotoxicity of CD8 + TILs compared with single PD1 blockade.

          Conclusions

          Ex vivo , co-blockade of TIGIT/PD1 improves functionality of CD8 + TILs that do not respond to single PD1 blockade. Therefore co-blockade of TIGIT/PD1 could be a promising immune therapeutic strategy for HCC patients.

          Graphical abstract

          Related collections

          Most cited references50

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

          Global Cancer Statistics 2018: GLOBOCAN Estimates of Incidence and Mortality Worldwide for 36 Cancers in 185 Countries

          This article provides a status report on the global burden of cancer worldwide using the GLOBOCAN 2018 estimates of cancer incidence and mortality produced by the International Agency for Research on Cancer, with a focus on geographic variability across 20 world regions. There will be an estimated 18.1 million new cancer cases (17.0 million excluding nonmelanoma skin cancer) and 9.6 million cancer deaths (9.5 million excluding nonmelanoma skin cancer) in 2018. In both sexes combined, lung cancer is the most commonly diagnosed cancer (11.6% of the total cases) and the leading cause of cancer death (18.4% of the total cancer deaths), closely followed by female breast cancer (11.6%), prostate cancer (7.1%), and colorectal cancer (6.1%) for incidence and colorectal cancer (9.2%), stomach cancer (8.2%), and liver cancer (8.2%) for mortality. Lung cancer is the most frequent cancer and the leading cause of cancer death among males, followed by prostate and colorectal cancer (for incidence) and liver and stomach cancer (for mortality). Among females, breast cancer is the most commonly diagnosed cancer and the leading cause of cancer death, followed by colorectal and lung cancer (for incidence), and vice versa (for mortality); cervical cancer ranks fourth for both incidence and mortality. The most frequently diagnosed cancer and the leading cause of cancer death, however, substantially vary across countries and within each country depending on the degree of economic development and associated social and life style factors. It is noteworthy that high-quality cancer registry data, the basis for planning and implementing evidence-based cancer control programs, are not available in most low- and middle-income countries. The Global Initiative for Cancer Registry Development is an international partnership that supports better estimation, as well as the collection and use of local data, to prioritize and evaluate national cancer control efforts. CA: A Cancer Journal for Clinicians 2018;0:1-31. © 2018 American Cancer Society.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Combined Nivolumab and Ipilimumab or Monotherapy in Untreated Melanoma

            New England Journal of Medicine, 373(1), 23-34
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Nivolumab in patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, non-comparative, phase 1/2 dose escalation and expansion trial

              For patients with advanced hepatocellular carcinoma, sorafenib is the only approved drug worldwide, and outcomes remain poor. We aimed to assess the safety and efficacy of nivolumab, a programmed cell death protein-1 (PD-1) immune checkpoint inhibitor, in patients with advanced hepatocellular carcinoma with or without chronic viral hepatitis. We did a phase 1/2, open-label, non-comparative, dose escalation and expansion trial (CheckMate 040) of nivolumab in adults (≥18 years) with histologically confirmed advanced hepatocellular carcinoma with or without hepatitis C or B (HCV or HBV) infection. Previous sorafenib treatment was allowed. A dose-escalation phase was conducted at seven hospitals or academic centres in four countries or territories (USA, Spain, Hong Kong, and Singapore) and a dose-expansion phase was conducted at an additional 39 sites in 11 countries (Canada, UK, Germany, Italy, Japan, South Korea, Taiwan). At screening, eligible patients had Child-Pugh scores of 7 or less (Child-Pugh A or B7) for the dose-escalation phase and 6 or less (Child-Pugh A) for the dose-expansion phase, and an Eastern Cooperative Oncology Group performance status of 1 or less. Patients with HBV infection had to be receiving effective antiviral therapy (viral load <100 IU/mL); antiviral therapy was not required for patients with HCV infection. We excluded patients previously treated with an agent targeting T-cell costimulation or checkpoint pathways. Patients received intravenous nivolumab 0·1–10 mg/kg every 2 weeks in the dose-escalation phase (3+3 design). Nivolumab 3 mg/kg was given every 2 weeks in the dose-expansion phase to patients in four cohorts: sorafenib untreated or intolerant without viral hepatitis, sorafenib progressor without viral hepatitis, HCV infected, and HBV infected. Primary endpoints were safety and tolerability for the escalation phase and objective response rate (Response Evaluation Criteria In Solid Tumors version 1.1) for the expansion phase. This study is registered with ClinicalTrials.gov , number NCT01658878 . Between Nov 26, 2012, and Aug 8, 2016, 262 eligible patients were treated (48 patients in the dose-escalation phase and 214 in the dose-expansion phase). 202 (77%) of 262 patients have completed treatment and follow-up is ongoing. During dose escalation, nivolumab showed a manageable safety profile, including acceptable tolerability. In this phase, 46 (96%) of 48 patients discontinued treatment, 42 (88%) due to disease progression. Incidence of treatment-related adverse events did not seem to be associated with dose and no maximum tolerated dose was reached. 12 (25%) of 48 patients had grade 3/4 treatment-related adverse events. Three (6%) patients had treatment-related serious adverse events (pemphigoid, adrenal insufficiency, liver disorder). 30 (63%) of 48 patients in the dose-escalation phase died (not determined to be related to nivolumab therapy). Nivolumab 3 mg/kg was chosen for dose expansion. The objective response rate was 20% (95% CI 15–26) in patients treated with nivolumab 3 mg/kg in the dose-expansion phase and 15% (95% CI 6–28) in the dose-escalation phase. Nivolumab had a manageable safety profile and no new signals were observed in patients with advanced hepatocellular carcinoma. Durable objective responses show the potential of nivolumab for treatment of advanced hepatocellular carcinoma. Bristol-Myers Squibb.
                Bookmark

                Author and article information

                Contributors
                Journal
                Cell Mol Gastroenterol Hepatol
                Cell Mol Gastroenterol Hepatol
                Cellular and Molecular Gastroenterology and Hepatology
                Elsevier
                2352-345X
                2021
                27 March 2021
                : 12
                : 2
                : 443-464
                Affiliations
                [1 ]Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
                [2 ]Department of Pathology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
                [3 ]Department of Surgery, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
                [4 ]Department of Gastroenterology and Hepatology, Amsterdam UMC location AMC, Amsterdam, the Netherlands
                [5 ]Department of Pathology, Amsterdam UMC location AMC, Amsterdam, the Netherlands
                [6 ]Department of Surgery, Amsterdam UMC location AMC, Amsterdam, the Netherlands
                [7 ]Department of Medical Oncology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
                Author notes
                [] Correspondence Address correspondence to: Dave Sprengers, MD, PhD, Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center Rotterdam, Wytemaweg 80, 3015 CN Rotterdam, the Netherlands. fax: +31 10 7030352. d.sprengers@ 123456erasmusmc.nl
                [b]

                Authors share co-senior authorship.

                Article
                S2352-345X(21)00055-2
                10.1016/j.jcmgh.2021.03.003
                8255944
                33781741
                0e7c093a-51d2-4e6f-b1f7-501ba76c5159
                © 2021 The Authors

                This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

                History
                : 2 June 2020
                : 2 March 2021
                Categories
                Original Research

                tigit,cd226,tox,hcc,immunotherapy,afp, alpha fetoprotein,apc, antigen-presenting cell,cdc, conventional dendritic cells,hcc, hepatocellular carcinoma,ifn, interferon,lag3, lymphocyte-activation gene 3,mfi, median fluorescent intensity,pd1, programmed cell death protein 1,pd-l1, programmed death-ligand 1,pma, phorbol 12-myristate 13-acetate,sem, standard error of the mean,tcf1, transcription factor 1,tfl, tumor-free liver tissue,tigit, t-cell immune receptor with ig and itim domains,til, tumor-infiltrating leukocyte,tim3, t-cell immunoglobulin and mucin-domain containing-3,tma, tissue microarray,tnf, tumor necrosis factor,tox, thymocyte selection-associated high mobility group box protein,treg, regulatory t cells

                Comments

                Comment on this article