4
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Priming of Anti-tumor Immune Mechanisms by Radiotherapy Is Augmented by Inhibition of Heat Shock Protein 90

      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

          Radiotherapy is an essential part of multi-modal cancer therapy. Nevertheless, for certain cancer entities such as colorectal cancer (CRC) the indications of radiotherapy are limited due to anatomical peculiarities and high radiosensitivity of the surrounding normal tissue. The development of molecularly targeted, combined modality approaches may help to overcome these limitations. Preferably, such strategies should not only enhance radiation-induced tumor cell killing and the abrogation of tumor cell clonogenicity, but should also support the stimulation of anti-tumor immune mechanisms – a phenomenon which moved into the center of interest of preclinical and clinical research in radiation oncology within the last decade. The present study focuses on inhibition of heat shock protein 90 (HSP90) whose combination with radiotherapy has previously been reported to exhibit convincing therapeutic synergism in different preclinical cancer models. By employing in vitro and in vivo analyses, we examined if this therapeutic synergism also applies to the priming of anti-tumor immune mechanisms in model systems of CRC. Our results indicate that the combination of HSP90 inhibitor treatment and ionizing irradiation induced apoptosis in colorectal cancer cells with accelerated transit into secondary necrosis in a hyperactive Kras-dependent manner. During secondary necrosis, dying cancer cells released different classes of damage-associated molecular patterns (DAMPs) that stimulated migration and recruitment of monocytic cells in vitro and in vivo. Additionally, these dying cancer cell-derived DAMPs enforced the differentiation of a monocyte-derived antigen presenting cell (APC) phenotype which potently triggered the priming of allogeneic T cell responses in vitro. In summary, HSP90 inhibition – apart from its radiosensitizing potential – obviously enables and supports the initial steps of anti-tumor immune priming upon radiotherapy and thus represents a promising partner for combined modality approaches. The therapeutic performance of such strategies requires further in-depth analyses, especially for but not only limited to CRC.

          Related collections

          Most cited references63

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

          Pancreatic cancers require autophagy for tumor growth.

          Macroautophagy (autophagy) is a regulated catabolic pathway to degrade cellular organelles and macromolecules. The role of autophagy in cancer is complex and may differ depending on tumor type or context. Here we show that pancreatic cancers have a distinct dependence on autophagy. Pancreatic cancer primary tumors and cell lines show elevated autophagy under basal conditions. Genetic or pharmacologic inhibition of autophagy leads to increased reactive oxygen species, elevated DNA damage, and a metabolic defect leading to decreased mitochondrial oxidative phosphorylation. Together, these ultimately result in significant growth suppression of pancreatic cancer cells in vitro. Most importantly, inhibition of autophagy by genetic means or chloroquine treatment leads to robust tumor regression and prolonged survival in pancreatic cancer xenografts and genetic mouse models. These results suggest that, unlike in other cancers where autophagy inhibition may synergize with chemotherapy or targeted agents by preventing the up-regulation of autophagy as a reactive survival mechanism, autophagy is actually required for tumorigenic growth of pancreatic cancers de novo, and drugs that inactivate this process may have a unique clinical utility in treating pancreatic cancers and other malignancies with a similar dependence on autophagy. As chloroquine and its derivatives are potent inhibitors of autophagy and have been used safely in human patients for decades for a variety of purposes, these results are immediately translatable to the treatment of pancreatic cancer patients, and provide a much needed, novel vantage point of attack.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Monocyte differentiation and antigen-presenting functions

            Monocytes not only serve as precursors for macrophages, but also contribute to tissue immunity by presenting antigen to T cells and producing immunomodulatory mediators. In this Review, the authors discuss some of these less well-appreciated immune functions of monocytes.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              A high-affinity conformation of Hsp90 confers tumour selectivity on Hsp90 inhibitors.

              Heat shock protein 90 (Hsp90) is a molecular chaperone that plays a key role in the conformational maturation of oncogenic signalling proteins, including HER-2/ErbB2, Akt, Raf-1, Bcr-Abl and mutated p53. Hsp90 inhibitors bind to Hsp90, and induce the proteasomal degradation of Hsp90 client proteins. Although Hsp90 is highly expressed in most cells, Hsp90 inhibitors selectively kill cancer cells compared to normal cells, and the Hsp90 inhibitor 17-allylaminogeldanamycin (17-AAG) is currently in phase I clinical trials. However, the molecular basis of the tumour selectivity of Hsp90 inhibitors is unknown. Here we report that Hsp90 derived from tumour cells has a 100-fold higher binding affinity for 17-AAG than does Hsp90 from normal cells. Tumour Hsp90 is present entirely in multi-chaperone complexes with high ATPase activity, whereas Hsp90 from normal tissues is in a latent, uncomplexed state. In vitro reconstitution of chaperone complexes with Hsp90 resulted in increased binding affinity to 17-AAG, and increased ATPase activity. These results suggest that tumour cells contain Hsp90 complexes in an activated, high-affinity conformation that facilitates malignant progression, and that may represent a unique target for cancer therapeutics.
                Bookmark

                Author and article information

                Contributors
                Journal
                Front Oncol
                Front Oncol
                Front. Oncol.
                Frontiers in Oncology
                Frontiers Media S.A.
                2234-943X
                27 August 2020
                2020
                : 10
                : 1668
                Affiliations
                [1] 1Department of Radiation Oncology, University Hospital, LMU Munich , Munich, Germany
                [2] 2Department of Otorhinolaryngology, University Hospital, LMU Munich , Munich, Germany
                [3] 3Walter Brendel Center for Experimental Medicine, Faculty of Medicine, LMU Munich , Munich, Germany
                [4] 4Department of Radiation Oncology, Universitätsklinikum Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU) , Erlangen, Germany
                [5] 5Department of Organic Chemistry, NCCR Chemical Biology, University of Geneva , Geneva, Switzerland
                [6] 6Department of Cell Biology, Faculty of Medicine Fukuoka University , Fukuoka, Japan
                [7] 7Institute for Advanced Study, Kyushu University , Fukuoka, Japan
                [8] 8Institute of Cardiovascular Physiology and Pathophysiology, Biomedical Center, LMU Munich , Munich, Germany
                [9] 9German Cancer Consortium (DKTK), Partner Site Munich , Heidelberg, Germany
                Author notes

                Edited by: Timothy F. Burns, University of Pittsburgh, United States

                Reviewed by: Phuoc T. Tran, Johns Hopkins Medicine, United States; Michele Caraglia, University of Campania Luigi Vanvitelli, Italy

                *Correspondence: Kirsten Lauber, kirsten.lauber@ 123456med.uni-muenchen.de

                Present address: Anne Ernst, Division of Radiation and Cancer Biology, Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, United States

                These authors share first authorship

                This article was submitted to Cancer Molecular Targets and Therapeutics, a section of the journal Frontiers in Oncology

                Article
                10.3389/fonc.2020.01668
                7481363
                b22dde1b-069e-456c-9e24-7d6c47a9bc0b
                Copyright © 2020 Ernst, Hennel, Krombach, Kapfhammer, Brix, Zuchtriegel, Uhl, Reichel, Frey, Gaipl, Winssinger, Shirasawa, Sasazuki, Sperandio, Belka and Lauber.

                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
                : 10 June 2020
                : 28 July 2020
                Page count
                Figures: 5, Tables: 0, Equations: 0, References: 92, Pages: 16, Words: 0
                Funding
                Funded by: Deutsche Forschungsgemeinschaft 10.13039/501100001659
                Award ID: SFB914 Project B06
                Award ID: SFB914 Project B03
                Award ID: SFB914 Project B01
                Award ID: INST 409/22-1 FUGG
                Award ID: INST 409/20-1 FUGG
                Award ID: INST 409/126-1 FUGG
                Funded by: Bundesministerium für Bildung und Forschung 10.13039/501100002347
                Award ID: ZiSS 02NUK024C
                Award ID: ZiSStrans 02NUK047C
                Funded by: Elitenetzwerk Bayern 10.13039/501100008848
                Funded by: Wilhelm Sander-Stiftung 10.13039/100008672
                Categories
                Oncology
                Original Research

                Oncology & Radiotherapy
                hsp90 inhibition,radiotherapy,anti-tumor immunity,immune priming,colorectal cancer,cancer immunology,damps,secondary necrosis

                Comments

                Comment on this article