6
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Tumor Cell-Based Vaccine Generated With High Hydrostatic Pressure Synergizes With Radiotherapy by Generating a Favorable Anti-tumor Immune Microenvironment

      research-article

      Read this article at

          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

          Dendritic cell (DC)-based vaccines pulsed with high hydrostatic pressure (HHP)-inactivated tumor cells have been demonstrated to be a promising immunotherapy for solid tumors. We focused on sole injection of tumor cells that were inactivated by HHP and their combination with local radiotherapy (RTx) for in vivo induction of anti-tumor immune responses. HHP-treatment of tumor cells resulted in pre-dominantly necrotic cells with degraded DNA. We confirmed that treatments at 200 MPa or higher completely inhibited the formation of tumor cell colonies in vitro. No tumor growth was seen in vivo after injection of HHP-treated tumor cells. Single vaccination with HHP-killed tumor cells combined with local RTx significantly retarded tumor growth and improved the survival as shown in B16-F10 and CT26 tumor models. In B16-F10 tumors that were irradiated with 2 × 5Gy and vaccinated once with HHP-killed tumor cells, the amount of natural killer (NK) cells, monocytes/macrophages, CD4+ T cells and NKT cells was significantly increased, while the amount of B cells was significantly decreased. In both models, a trend of increased CD8+ T cell infiltration was observed. Generally, in irradiated tumors high amounts of CD4+ and CD8+ T cells expressing PD-1 were found. We conclude that HHP generates inactivated tumor cells that can be used as a tumor vaccine. Moreover, we show for the first time that tumor cell-based vaccine acts synergistically with RTx to significantly retard tumor growth by generating a favorable anti-tumor immune microenvironment.

          Related collections

          Most cited references39

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

          A novel assay for apoptosis Flow cytometric detection of phosphatidylserine expression on early apoptotic cells using fluorescein labelled Annexin V

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

            Clinical use of dendritic cells for cancer therapy.

            Since the mid-1990s, dendritic cells have been used in clinical trials as cellular mediators for therapeutic vaccination of patients with cancer. Dendritic cell-based immunotherapy is safe and can induce antitumour immunity, even in patients with advanced disease. However, clinical responses have been disappointing, with classic objective tumour response rates rarely exceeding 15%. Paradoxically, findings from emerging research indicate that dendritic cell-based vaccination might improve survival, advocating implementation of alternative endpoints to assess the true clinical potency of dendritic cell-based vaccination. We review the clinical effectiveness of dendritic cell-based vaccine therapy in melanoma, prostate cancer, malignant glioma, and renal cell carcinoma, and summarise the most important lessons from almost two decades of clinical studies of dendritic cell-based immunotherapy in these malignant disorders. We also address how the specialty is evolving, and which new therapeutic concepts are being translated into clinical trials to leverage the clinical effectiveness of dendritic cell-based cancer immunotherapy. Specifically, we discuss two main trends: the implementation of the next-generation dendritic cell vaccines that have improved immunogenicity, and the emerging paradigm of combination of dendritic cell vaccination with other cancer therapies. Copyright © 2014 Elsevier Ltd. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Local radiotherapy and granulocyte-macrophage colony-stimulating factor to generate abscopal responses in patients with metastatic solid tumours: a proof-of-principle trial.

              An abscopal response describes radiotherapy-induced immune-mediated tumour regression at sites distant to the irradiated field. Granulocyte-macrophage colony-stimulating factor is a potent stimulator of dendritic cell maturation. We postulated that the exploitation of the pro-immunogenic effects of radiotherapy with granulocyte-macrophage colony-stimulating factor might result in abscopal responses among patients with metastatic cancer.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                28 August 2019
                2019
                : 9
                : 805
                Affiliations
                [1] 1Department of Radiation Oncology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen , Erlangen, Germany
                [2] 2Department of Psychiatry and Psychotherapy, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen , Erlangen, Germany
                [3] 3Institute of Process Machinery and Systems Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg , Erlangen, Germany
                [4] 4Department of Cardiac Surgery, Friedrich-Alexander-Universität Erlangen-Nürnberg, Universitätsklinikum Erlangen , Erlangen, Germany
                Author notes

                Edited by: Jin S. Im, University of Texas MD Anderson Cancer Center, United States

                Reviewed by: Irena Adkins, SOTIO a.s., Czechia; Amorette Barber, Longwood University, United States

                *Correspondence: Udo S. Gaipl udo.gaipl@ 123456uk-erlangen.de

                This article was submitted to Cancer Immunity and Immunotherapy, a section of the journal Frontiers in Oncology

                †These authors have contributed equally to this work as first authors

                ‡These authors have contributed equally to this work as senior authors

                Article
                10.3389/fonc.2019.00805
                6722191
                518f6816-11ca-42b6-8c01-0accb97227dd
                Copyright © 2019 Seitz, Rückert, Deloch, Weiss, Utz, Izydor, Ebel, Schlücker, Fietkau, Gaipl and Frey.

                This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.

                History
                : 31 January 2019
                : 07 August 2019
                Page count
                Figures: 5, Tables: 0, Equations: 0, References: 55, Pages: 12, Words: 8360
                Funding
                Funded by: Deutsche Forschungsgemeinschaft 10.13039/501100001659
                Funded by: Bundesministerium für Bildung, Wissenschaft, Forschung und Technologie 10.13039/501100010571
                Funded by: European Commission 10.13039/501100000780
                Categories
                Oncology
                Original Research

                Oncology & Radiotherapy
                radiotherapy,immunotherapy,tumor cell-based vaccine,high hydrostatic pressure,malignant melanoma,colorectal carcinoma,tumor-infiltrating leukocytes,tumor microenvironment

                Comments

                Comment on this article