37
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: not found

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

      research-article

      Read this article at

      ScienceOpenPublisherPMC
          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.

          Significance

          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.

          Abstract

          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.

          Related collections

          Most cited references32

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

          Myeloid-derived suppressor cells: linking inflammation and cancer.

          Many cancer immunotherapies developed in experimental animals have been tested in clinical trials. Although some have shown modest clinical effects, most have not been effective. Recent studies have identified myeloid-origin cells that are potent suppressors of tumor immunity and therefore a significant impediment to cancer immunotherapy. "Myeloid-derived suppressor cells" (MDSC) accumulate in the blood, lymph nodes, and bone marrow and at tumor sites in most patients and experimental animals with cancer and inhibit both adaptive and innate immunity. MDSC are induced by tumor-secreted and host-secreted factors, many of which are proinflammatory molecules. The induction of MDSC by proinflammatory mediators led to the hypothesis that inflammation promotes the accumulation of MDSC that down-regulate immune surveillance and antitumor immunity, thereby facilitating tumor growth. This article reviews the characterization and suppressive mechanisms used by MDSC to block tumor immunity and describes the mechanisms by which inflammation promotes tumor progression through the induction of MDSC.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Synergy of demethylation and histone deacetylase inhibition in the re-expression of genes silenced in cancer.

            Densely methylated DNA associates with transcriptionally repressive chromatin characterized by the presence of underacetylated histones. Recently, these two epigenetic processes have been dynamically linked. The methyl-CpG-binding protein MeCP2 appears to reside in a complex with histone deacetylase activity. MeCP2 can mediate formation of transcriptionally repressive chromatin on methylated promoter templates in vitro, and this process can be reversed by trichostatin A (TSA), a specific inhibitor of histone deacetylase. Little is known, however, about the relative roles of methylation and histone deacetylase activity in the stable inhibition of transcription on densely methylated endogenous promoters, such as those for silenced alleles of imprinted genes, genes on the female inactive X chromosome and tumour-suppressor genes inactivated in cancer cells. We show here that the hypermethylated genes MLH1, TIMP3 (TIMP3), CDKN2B (INK4B, p15) and CDKN2A (INK4, p16) cannot be transcriptionally reactivated with TSA alone in tumour cells in which we have shown that TSA alone can upregulate the expression of non-methylated genes. Following minimal demethylation and slight gene reactivation in the presence of low dose 5-aza-2'deoxycytidine (5Aza-dC), however, TSA treatment results in robust re-expression of each gene. TSA does not contribute to demethylation of the genes, and none of the treatments alter the chromatin structure associated with the hypermethylated promoters. Thus, although DNA methylation and histone deacetylation appear to act as synergistic layers for the silencing of genes in cancer, dense CpG island methylation is dominant for the stable maintenance of a silent state at these loci.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Combination epigenetic therapy has efficacy in patients with refractory advanced non-small cell lung cancer.

              Epigenetic alterations are strongly associated with the development of cancer. We conducted a phase I/II trial of combined epigenetic therapy with azacitidine and entinostat, inhibitors of DNA methylation and histone deacetylation, respectively, in extensively pretreated patients with recurrent metastatic non-small cell lung cancer. This therapy is well tolerated, and objective responses were observed, including a complete response and a partial response in a patient who remains alive and without disease progression approximately 2 years after completing protocol therapy. Median survival in the entire cohort was 6.4 months (95% CI 3.8-9.2), comparing favorably with existing therapeutic options. Demethylation of a set of 4 epigenetically silenced genes known to be associated with lung cancer was detectable in serial blood samples in these patients and was associated with improved progression-free (P = 0.034) and overall survival (P = 0.035). Four of 19 patients had major objective responses to subsequent anticancer therapies given immediately after epigenetic therapy. This study demonstrates that combined epigenetic therapy with low-dose azacitidine and entinostat results in objective, durable responses in patients with solid tumors and defines a blood-based biomarker that correlates with clinical benefit.
                Bookmark

                Author and article information

                Journal
                Proc Natl Acad Sci U S A
                Proc. Natl. Acad. Sci. U.S.A
                pnas
                pnas
                PNAS
                Proceedings of the National Academy of Sciences of the United States of America
                National Academy of Sciences
                0027-8424
                1091-6490
                19 December 2017
                4 December 2017
                : 114
                : 51
                : E10981-E10990
                Affiliations
                [1] aDepartment of Oncology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, MD 21287;
                [2] bDepartment of Neurosurgery, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, MD 21287;
                [3] cDepartment of Gynecology and Obstetrics, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, MD 21287;
                [4] dDepartment of Pathology, The Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins , Baltimore, MD 21287;
                [5] eDiscovery Sciences, Janssen Research & Development , Spring House, PA 19477;
                [6] fImmuno-Oncology Discovery, Janssen Research & Development , Spring House, PA 19477;
                [7] gOncology Janssen Research & Development , Spring House, PA 19477;
                [8] hLaboratory for Molecular Medicine, Department of Gynaecology and Obstetrics, University-Clinic Erlangen , 91054 Erlangen, Germany
                Author notes
                4To whom correspondence may be addressed. Email: sbaylin@ 123456jhmi.edu or zahnoci@ 123456jhmi.edu .

                Contributed by Stephen B. Baylin, November 4, 2017 (sent for review July 14, 2017; reviewed by Adam R. Karpf and Jonathan Licht)

                Author contributions: M.L.S., K.B.C., M.J.T., D.M., K.R.W., S.B.B., and C.A.Z. designed research; M.L.S., K.B.C., H.L., L.M.M., M.E.T., V.B., K.R.W., G.S.C., K.E.B., R.S., P.L.S., and C.A.Z. performed research; D.M., M.L., I.-M.S., T.-L.W., C.-F.H., and P.L.S. contributed new reagents/analytic tools; M.L.S., K.B.C., H.L., M.E.T., V.B., K.R.W., R.S., P.L.S., S.B.B., and C.A.Z. analyzed data; and M.L.S., K.B.C., S.B.B., and C.A.Z. wrote the paper.

                Reviewers: A.R.K., University of Nebraska Medical Center; and J.L., The University of Florida.

                1M.L.S. and K.B.C. contributed equally to this work.

                2Present address: Division of Hematology-Oncology, Department of Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104.

                3Present address: Department of Microbiology, Immunology, and Tropical Medicine and the George Washington University Cancer Center, George Washington University, Washington, DC 20052.

                Article
                PMC5754782 PMC5754782 5754782 201712514
                10.1073/pnas.1712514114
                5754782
                29203668
                ace888c3-5422-4b11-a37e-c1f239a8ee4f
                Copyright @ 2017

                Published under the PNAS license.

                History
                Page count
                Pages: 10
                Funding
                Funded by: NIH
                Award ID: P30CA006973
                Funded by: U.S. Department of Defense (DOD) 100000005
                Award ID: OC130454 / W81XWH-14-1-0385.
                Funded by: Dr. Miriam and Sheldon G. Adelson Medical Research Foundation (AMRF) 100005984
                Award ID: none
                Funded by: Samuel Waxman Cancer Research Foundation (SWCRF) 100001384
                Award ID: none
                Funded by: Irving Hansen fund
                Award ID: none
                Funded by: Janssen Research and Development (JRD) 100005205
                Award ID: none
                Funded by: NIH
                Award ID: F32CA183214
                Funded by: NIH
                Award ID: K99CA204592
                Categories
                PNAS Plus
                Biological Sciences
                Medical Sciences
                PNAS Plus

                5-azacytidine,histone deacetylase inhibitors,type I interferon,ovarian cancer,immunosuppression

                Comments

                Comment on this article