17
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Epigenetic treatment of multiple myeloma mediates tumor intrinsic and extrinsic immunomodulatory effects

      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

          Immune evasion is an important driver of disease progression in the plasma cell malignancy multiple myeloma. Recent work highlights the potential of epigenetic modulating agents as tool to enhance anti-tumor immunity. The immune modulating effects of the combination of a DNA methyltransferase inhibitor and a histone deacetylase inhibitor in multiple myeloma is insufficiently characterized. Therefore, we used the murine immunocompetent 5T33MM model to investigate hallmarks of immunogenic cell death as well as alterations in the immune cell constitution in the bone marrow of diseased mice in response to the DNA methyltransferase inhibitor decitabine and the histone deacetylase inhibitor quisinostat. Vaccination of mice with 5T33 cells treated with epigenetic compounds delayed tumor development upon a subsequent tumor challenge. In vitro, epigenetic treatment induced ecto-calreticulin and CD47, as well as a type I interferon response. Moreover, treated 5T33vt cells triggered dendritic cell maturation. The combination of decitabine and quisinostat in vivo resulted in combinatory anti-myeloma effects. In vivo, epigenetic treatment increased tumoral ecto-calreticulin and decreased CD47 and PD-L1 expression, increased dendritic cell maturation and reduced CD11b positive cells. Moreover, epigenetic treatment induced a temporal increase in presence of CD8-positive and CD4-positive T cells with naive and memory-like phenotypes based on CD62L and CD44 expression levels, and reduced expression of exhaustion markers PD-1 and TIM3. In conclusion, a combination of a DNA methyltransferase inhibitor and a histone deacetylase inhibitor increased the immunogenicity of myeloma cells and altered the immune cell constitution in the bone marrow of myeloma-bearing mice.

          Related collections

          Most cited references26

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

          Epigenetic therapy activates type I interferon signaling in murine ovarian cancer to reduce immunosuppression and tumor burden

          Therapies that activate the host immune system have shown tremendous promise for a variety of solid tumors. However, in most cancer types, fewer than half of patients respond to these immunotherapies. We propose epigenetic therapy as a mechanism to sensitize tumors to immune checkpoint therapy. We have shown that inhibiting DNA methylation triggers a viral defense pathway in tumors. Here we show that epigenetic therapy in a mouse model of ovarian cancer increases the numbers of activated immune cells, and that this is dependent on the interferon antiviral response. The combination of epigenetic therapy and immune checkpoint blockade leads to the greatest reduction in tumor burden and increase in survival, and may hold the greatest promise for patients. Ovarian cancer is the most lethal of all gynecological cancers, and there is an urgent unmet need to develop new therapies. Epithelial ovarian cancer (EOC) is characterized by an immune suppressive microenvironment, and response of ovarian cancers to immune therapies has thus far been disappointing. We now find, in a mouse model of EOC, that clinically relevant doses of DNA methyltransferase and histone deacetylase inhibitors (DNMTi and HDACi, respectively) reduce the immune suppressive microenvironment through type I IFN signaling and improve response to immune checkpoint therapy. These data indicate that the type I IFN response is required for effective in vivo antitumorigenic actions of the DNMTi 5-azacytidine (AZA). Through type I IFN signaling, AZA increases the numbers of CD45 + immune cells and the percentage of active CD8 + T and natural killer (NK) cells in the tumor microenvironment, while reducing tumor burden and extending survival. AZA also increases viral defense gene expression in both tumor and immune cells, and reduces the percentage of macrophages and myeloid-derived suppressor cells in the tumor microenvironment. The addition of an HDACi to AZA enhances the modulation of the immune microenvironment, specifically increasing T and NK cell activation and reducing macrophages over AZA treatment alone, while further increasing the survival of the mice. Finally, a triple combination of DNMTi/HDACi plus the immune checkpoint inhibitor α-PD-1 provides the best antitumor effect and longest overall survival, and may be an attractive candidate for future clinical trials in ovarian cancer.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found
            Is Open Access

            DNA-damage-induced type I interferon promotes senescence and inhibits stem cell function.

            Expression of type I interferons (IFNs) can be induced by DNA-damaging agents, but the mechanisms and significance of this regulation are not completely understood. We found that the transcription factor IRF3, activated in an ATM-IKKα/β-dependent manner, stimulates cell-autonomous IFN-β expression in response to double-stranded DNA breaks. Cells and tissues with accumulating DNA damage produce endogenous IFN-β and stimulate IFN signaling in vitro and in vivo. In turn, IFN acts to amplify DNA-damage responses, activate the p53 pathway, promote senescence, and inhibit stem cell function in response to telomere shortening. Inactivation of the IFN pathway abrogates the development of diverse progeric phenotypes and extends the lifespan of Terc knockout mice. These data identify DNA-damage-response-induced IFN signaling as a critical mechanism that links accumulating DNA damage with senescence and premature aging.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              High somatic mutation and neoantigen burden are correlated with decreased progression-free survival in multiple myeloma

              Tumor-specific mutations can result in immunogenic neoantigens, both of which have been correlated with responsiveness to immune checkpoint inhibitors in highly mutagenic cancers. However, early results of single-agent checkpoint inhibitors in multiple myeloma (MM) have been underwhelming. Therefore, we sought to understand the relationship between mutation and neoantigen landscape of MM patients and responsiveness to therapies. Somatic mutation burden, neoantigen load, and response to therapy were determined using interim data from the MMRF CoMMpass study (NCT01454297) on 664 MM patients. In this population, the mean somatic and missense mutation loads were 405.84(s=608.55) and 63.90(s=95.88) mutations per patient, respectively. There was a positive linear relationship between mutation and neoantigen burdens (R 2=0.862). The average predicted neoantigen load was 23.52(s=52.14) neoantigens with an average of 9.40(s=26.97) expressed neoantigens. Survival analysis revealed significantly shorter progression-free survival (PFS) in patients with greater than average somatic missense mutation load (N=163, 0.493 vs 0.726 2-year PFS, P=0.0023) and predicted expressed neoantigen load (N=214, 0.555 vs 0.729 2-year PFS, P=0.0028). This pattern is maintained when stratified by disease stage and cytogenetic abnormalities. Therefore, high mutation and neoantigen load are clinically relevant risk factors that negatively impact survival of MM patients under current standards of care.
                Bookmark

                Author and article information

                Journal
                Oncoimmunology
                Oncoimmunology
                KONI
                koni20
                Oncoimmunology
                Taylor & Francis
                2162-4011
                2162-402X
                2018
                23 July 2018
                23 July 2018
                : 7
                : 10
                : e1484981
                Affiliations
                [a ]Department of Hematology and Immunology, Myeloma Center Brussels, Vrije Universiteit Brussel , Brussel, Belgium
                [b ]Laboratory of Molecular and Cellular Therapy, Vrije Universiteit Brussel , Brussels, Belgium
                Author notes
                CONTACT Ken Maes, PhD ken.maes@ 123456vub.be Department of Hematology and Immunology, Building D, Room D0.03, Vrije Universiteit Brussel, Laarbeeklaan 103, 1090 Brussel, Belgium
                [*]

                Equal senior authors.

                Color versions of one or more of the figures in the article can be found online at www.tandfonline.com/koni.

                Author information
                http://orcid.org/0000-0002-8445-0441
                http://orcid.org/0000-0001-8906-2790
                Article
                1484981
                10.1080/2162402X.2018.1484981
                6169579
                30288346
                41b5c6e3-b4a8-4113-9a45-82da2649c576
                © 2018 Taylor & Francis Group, LLC

                This is an Open Access article distributed under the terms of the Creative Commons Attribution-NonCommercial-NoDerivatives License ( http://creativecommons.org/licenses/by-nc-nd/4.0/), which permits non-commercial re-use, distribution, and reproduction in any medium, provided the original work is properly cited, and is not altered, transformed, or built upon in any way.

                History
                : 27 April 2018
                : 29 May 2018
                : 30 May 2018
                Page count
                Figures: 7, References: 40, Pages: 13
                Funding
                Funded by: Fonds Wetenschappelijk Onderzoek 10.13039/501100003130
                Award ID: 1507917N
                Funded by: Fonds Wetenschappelijk Onderzoek 10.13039/501100003130
                Award ID: 12E3816N
                Funded by: Fonds Stimulans
                Award ID: FS-2016 Maes
                This work was supported by (1) “Fonds Wetenschappelijk Onderzoek-Vlaanderen” under grants “12E3816N” and “1507917N” for postdoctoral fellowships of Ken Maes and Kim De Veirman, respectively; (2) “Fonds Stimulans” under the research grant “FS-2016 Maes”; (3) “Vrije Universiteit Brussel” under the strategic research program scheme under grant SRP48; (4) “Kom op Tegen Kanker (Stand up to Cancer), the “Flemish cancer society” under a predoctoral fellowship of Lien De Beck, “1164918N”, and (5) ERA-NET Transcan-2 JTC 2015 under research grant “G0H7216N”.
                Categories
                Original Research

                Immunology
                multiple myeloma,dna methyltransferase inhibitor,histone deacetylase inhibitor,immunogenic cell death,calreticulin,type i interferon,dendritic cell,cytotoxic t-cell

                Comments

                Comment on this article