16
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Plasmacytoid dendritic cells orchestrate innate and adaptive anti-tumor immunity induced by oncolytic coxsackievirus A21

      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

          Background

          The oncolytic virus, coxsackievirus A21 (CVA21), has shown promise as a single agent in several clinical trials and is now being tested in combination with immune checkpoint blockade. Combination therapies offer the best chance of disease control; however, the design of successful combination strategies requires a deeper understanding of the mechanisms underpinning CVA21 efficacy, in particular, the role of CVA21 anti-tumor immunity. Therefore, this study aimed to examine the ability of CVA21 to induce human anti-tumor immunity, and identify the cellular mechanism responsible.

          Methods

          This study utilized peripheral blood mononuclear cells from i) healthy donors, ii) Acute Myeloid Leukemia (AML) patients, and iii) patients taking part in the STORM clinical trial, who received intravenous CVA21; patients receiving intravenous CVA21 were consented separately in accordance with local institutional ethics review and approval. Collectively, these blood samples were used to characterize the development of innate and adaptive anti-tumor immune responses following CVA21 treatment.

          Results

          An Initial characterization of peripheral blood mononuclear cells, collected from cancer patients following intravenous infusion of CVA21, confirmed that CVA21 activated immune effector cells in patients. Next, using hematological disease models which were sensitive (Multiple Myeloma; MM) or resistant (AML) to CVA21-direct oncolysis, we demonstrated that CVA21 stimulated potent anti-tumor immune responses, including: 1) cytokine-mediated bystander killing; 2) enhanced natural killer cell-mediated cellular cytotoxicity; and 3) priming of tumor-specific cytotoxic T lymphocytes, with specificity towards known tumor-associated antigens. Importantly, immune-mediated killing of both MM and AML, despite AML cells being resistant to CVA21-direct oncolysis, was observed. Upon further examination of the cellular mechanisms responsible for CVA21-induced anti-tumor immunity we have identified the importance of type I IFN for NK cell activation, and demonstrated that both ICAM-1 and plasmacytoid dendritic cells were key mediators of this response.

          Conclusion

          This work supports the development of CVA21 as an immunotherapeutic agent for the treatment of both AML and MM. Additionally, the data presented provides an important insight into the mechanisms of CVA21-mediated immunotherapy to aid the development of clinical biomarkers to predict response and rationalize future drug combinations.

          Electronic supplementary material

          The online version of this article (10.1186/s40425-019-0632-y) contains supplementary material, which is available to authorized users.

          Related collections

          Most cited references44

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

          CD69: from activation marker to metabolic gatekeeper.

          CD69 is a membrane-bound, type II C-lectin receptor. It is a classical early marker of lymphocyte activation due to its rapid appearance on the surface of the plasma membrane after stimulation. CD69 is expressed by several subsets of tissue resident immune cells, including resident memory T (TRM) cells and gamma delta (γδ) T cells, and is therefore considered a marker of tissue retention. Recent evidence has revealed that CD69 regulates some specific functions of selected T-cell subsets, determining the migration-retention ratio as well as the acquisition of effector or regulatory phenotypes. Specifically, CD69 regulates the differentiation of regulatory T (Treg) cells as well as the secretion of IFN-γ, IL-17 and IL-22. The identification of putative CD69 ligands, such as Galectin-1 (Gal-1), suggests that CD69-induced signaling can be regulated not only during cognate contacts between T cells and antigen-presenting cells in lymphoid organs, but also in the periphery, where cytokines and other metabolites control the final outcome of the immune response. Here, we will discuss new aspects of the molecular signaling mediated by CD69, and its involvement in the metabolic reprogramming regulating TH-effector lineages and provide their ramifications and possible significance in homeostasis and pathological scenarios. This article is protected by copyright. All rights reserved.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Activated granulocytes and granulocyte-derived hydrogen peroxide are the underlying mechanism of suppression of t-cell function in advanced cancer patients.

            Impaired T-cell function in patients with advanced cancer has been a widely acknowledged finding, but mechanisms reported thus far are those primarily operating in the tumor microenvironment. Very few mechanisms have been put forth to explain several well-described defects in peripheral blood T cells, such as reduction in expression of signaling molecules, decreased production of cytokines, or increased apoptosis. We have closely examined the peripheral blood mononuclear cell (PBMC) samples derived from patients and healthy individuals, and we have observed an important difference that may underlie the majority of reported defects. We observed that in samples from patients only, an unusually large number of granulocytes copurify with low density PBMCs on a density gradient rather than sediment, as expected, to the bottom of the gradient. We also show that activating granulocytes from a healthy donor with N-formyl-L-methionyl-L-leucyl-L-phenylalanine could also cause them to sediment aberrantly and copurify with PBMCs, suggesting that density change is a marker of their activation. To confirm this, we looked for other evidence of in vivo granulocyte activation and found it in drastically elevated plasma levels of 8-isoprostane, a product of lipid peroxidation and a marker of oxidative stress. Reduced T-cell receptor zeta chain expression and decreased cytokine production by patients' T cells correlated with the presence of activated granulocytes in their PBMCs. We showed that freshly obtained granulocytes from healthy donors, if activated, can also inhibit cytokine production by T cells. This action is abrogated by the addition of the hydrogen peroxide (H(2)O(2)) scavenger, catalase, implicating H(2)O(2) as the effector molecule. Indeed, when added alone, H(2)O(2) could suppress cytokine production of normal T cells. These findings indicate that granulocytes are activated in advanced cancer patients and that granulocyte-derived H(2)O(2) is the major cause of severe systemic T-cell suppression.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A phase I study of OncoVEXGM-CSF, a second-generation oncolytic herpes simplex virus expressing granulocyte macrophage colony-stimulating factor.

              To conduct a phase I clinical trial with a second-generation oncolytic herpes simplex virus (HSV) expressing granulocyte macrophage colony-stimulating factor (Onco VEXGM-CSF) to determine the safety profile of the virus, look for evidence of biological activity, and identify a dosing schedule for later studies. The virus was administered by intratumoral injection in patients with cutaneous or s.c. deposits of breast, head and neck and gastrointestinal cancers, and malignant melanoma who had failed prior therapy. Thirteen patients were in a single-dose group, where doses of 10(6), 10(7), and 10(8) plaque-forming units (pfu)/mL were tested, and 17 patients were in a multidose group testing a number of dose regimens. The virus was generally well tolerated with local inflammation, erythema, and febrile responses being the main side effects. The local reaction to injection was dose limiting in HSV-seronegative patients at 10(7) pfu/mL. The multidosing phase thus tested seroconverting HSV-seronegative patients with 10(6) pfu/mL followed by multiple higher doses (up to 10(8) pfu/mL), which was well tolerated by all patients. Biological activity (virus replication, local reactions, granulocyte macrophage colony-stimulating factor expression, and HSV antigen-associated tumor necrosis), was observed. The duration of local reactions and virus replication suggested that dosing every 2 to 3 weeks was appropriate. Nineteen of 26 patient posttreatment biopsies contained residual tumor of which 14 showed tumor necrosis, which in some cases was extensive, or apoptosis. In all cases, areas of necrosis also strongly stained for HSV. The overall responses to treatment were that three patients had stable disease, six patients had tumors flattened (injected and/or uninjected lesions), and four patients showed inflammation of uninjected as well as the injected tumor, which, in nearly all cases, became inflamed. Onco VEXGM-CSF is well tolerated and can be safely administered using the multidosing protocol described. Evidence of an antitumor effect was seen.
                Bookmark

                Author and article information

                Contributors
                medlmul@leeds.ac.uk
                holmes.matthew22@yahoo.com
                jonanne_hopper@hotmail.co.uk
                g.b.scott@leeds.ac.uk
                e.j.west@leeds.ac.uk
                k.j.scott@leeds.ac.uk
                christopherparrish@nhs.net
                kat83df@googlemail.com
                s.staeble@dkfz-heidelberg.de
                vicki.jennings@icr.ac.uk
                matthewcullen@nhs.net
                stewart.mcconnnell@nhs.net
                Catherine.Langton@leedsth.nhs.uk
                e.l.tidswell@leeds.ac.uk
                Darren.Shafren@newcastle.edu.au
                a.samson@leeds.ac.uk
                Kevin.Harrington@icr.ac.uk
                H.Pandha@surrey.ac.uk
                christy.ralph@nhs.net
                richardkelly@nhs.net
                gordoncook@nhs.net
                alan.melcher@icr.ac.uk
                f.errington@leeds.ac.uk
                Journal
                J Immunother Cancer
                J Immunother Cancer
                Journal for Immunotherapy of Cancer
                BioMed Central (London )
                2051-1426
                1 July 2019
                1 July 2019
                2019
                : 7
                : 164
                Affiliations
                [1 ]Section of Infection and Immunity, Leeds Institute of Medical Research (LIMR), University of Leeds, St. James’s University Hospital, Level 5, Wellcome Trust Brenner Building (WTBB), Leeds, LS9 7TF UK
                [2 ]GRID grid.443984.6, Haematological Malignancy Diagnostics Service, , St. James’s University Hospital, ; Leeds, UK
                [3 ]GRID grid.443984.6, Department of Haematology, , St. James’s University Hospital, ; Leeds, UK
                [4 ]ISNI 0000 0000 8831 109X, GRID grid.266842.c, School of Biomedical Science and Pharmacy, , University of Newcastle, ; Newcastle, Australia
                [5 ]ISNI 0000 0001 1271 4623, GRID grid.18886.3f, Translational Immunotherapy Team, , The Institute of Cancer Research and Royal Marsden Hospital/Institute of Cancer Research NIHR Biomedical Research Centre, ; London, UK
                [6 ]ISNI 0000 0004 0407 4824, GRID grid.5475.3, Surrey Cancer Research Institute, Leggett Building, Faculty of Health and Medical Sciences, , University of Surrey, ; Guildford, UK
                [7 ]Section of Experimental Haematology, LIMR, University of Leeds, St. James’s University Hospital, Leeds, UK
                Author information
                http://orcid.org/0000-0003-2155-534X
                Article
                632
                10.1186/s40425-019-0632-y
                6604201
                31262361
                c7fe36a9-0837-4249-a425-a25ede3e9d2a
                © The Author(s). 2019

                Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License ( http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. 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.

                History
                : 13 February 2019
                : 6 June 2019
                Funding
                Funded by: Cancer research UK
                Award ID: A13244 and C16708
                Funded by: Kay Kendall Leukaemia Fund
                Award ID: KKL1071
                Award Recipient :
                Funded by: FundRef http://dx.doi.org/10.13039/501100002653, Yorkshire Cancer Research;
                Award ID: L374RA
                Award Recipient :
                Categories
                Research Article
                Custom metadata
                © The Author(s) 2019

                oncolytic viruses,coxsackievirus a21,plasmacytoid dc,innate immunity,adaptive immunity

                Comments

                Comment on this article