13
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      BET inhibitors rescue anti-PD1 resistance by enhancing TCF7 accessibility in leukemia-derived terminally exhausted CD8 + T cells

      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

          Many acute myeloid leukemia (AML) patients exhibit hallmarks of immune exhaustion, such as increased myeloid-derived suppressor cells, suppressive regulatory T cells and dysfunctional T cells. Similarly, we have identified the same immune-related features, including exhausted CD8 + T cells (TEx) in a mouse model of AML. Here we show that inhibitors that target bromodomain and extra-terminal domain (BET) proteins affect tumor-intrinsic factors but also rescue T cell exhaustion and ICB resistance. Ex vivo treatment of cells from AML mice and AML patients with BET inhibitors (BETi) reversed CD8 + T cell exhaustion by restoring proliferative capacity and expansion of the more functional precursor-exhausted T cells. This reversal was enhanced by combined BETi and anti-PD1 treatment. BETi synergized with anti-PD1 in vivo, resulting in the reduction of circulating leukemia cells, enrichment of CD8 + T cells in the bone marrow, and increase in expression of Tcf7, Slamf6, and Cxcr5 in CD8 + T cells. Finally, we profiled the epigenomes of in vivo JQ1-treated AML-derived CD8 + T cells by single-cell ATAC-seq and found that JQ1 increases Tcf7 accessibility specifically in Tex cells, suggesting that BETi likely acts mechanistically by relieving repression of progenitor programs in Tex CD8 + T cells and maintaining a pool of anti-PD1 responsive CD8 + T cells.

          Related collections

          Most cited references62

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

          Subsets of exhausted CD8+ T cells differentially mediate tumor control and respond to checkpoint blockade

          T cell dysfunction is a hallmark of many cancers, but the basis for T cell dysfunction and the mechanisms by which antibody blockade of the inhibitory receptor PD-1 (anti-PD-1) reinvigorates T cells are not fully understood. Here we show that such therapy acts on a specific subpopulation of exhausted CD8 + tumor-infiltrating lymphocytes (TILs). Dysfunctional CD8 + TILs possess canonical epigenetic and transcriptional features of exhaustion that mirror those seen in chronic viral infection. Exhausted CD8 + TILs include a subpopulation of ‘progenitor exhausted’ cells that retain polyfunctionality, persist long term and differentiate into ‘terminally exhausted’ TILs. Consequently, progenitor exhausted CD8 + TILs are better able to control tumor growth than are terminally exhausted T cells. Progenitor exhausted TILs can respond to anti-PD-1 therapy, but terminally exhausted TILs cannot. Patients with melanoma who have a higher percentage of progenitor exhausted cells experience a longer duration of response to checkpoint-blockade therapy. Thus, approaches to expand the population of progenitor exhausted CD8 + T cells might be an important component of improving the response to checkpoint blockade.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Defining T Cell States Associated with Response to Checkpoint Immunotherapy in Melanoma

            Treatment of cancer has been revolutionized by immune checkpoint blockade therapies. Despite the high rate of response in advanced melanoma, the majority of patients succumb to disease. To identify factors associated with success or failure of checkpoint therapy, we profiled transcriptomes of 16,291 individual immune cells from 48 tumor samples of melanoma patients treated with checkpoint inhibitors. Two distinct states of CD8 + T cells were defined by clustering, and associated with patient tumor regression or progression. A single transcription factor, TCF7 , was visualized within CD8 + T cells in fixed tumor samples and predicted positive clinical outcome in an independent cohort of checkpoint-treated patients. We delineated the epigenetic landscape and clonality of these T cell states, and demonstrated enhanced anti-tumor immunity by targeting novel combinations of factors in exhausted cells. Our study of immune cell transcriptomes from tumors demonstrates a strategy for identifying predictors, mechanisms and targets for enhancing checkpoint immunotherapy. Single cell analysis of immune cells from melanoma uncovers a TCF7+ memory-like state in the cytotoxic T cell population, and demonstrates its association with a positive outcome
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Selective inhibition of BET bromodomains

              Epigenetic proteins are intently pursued targets in ligand discovery. To date, successful efforts have been limited to chromatin modifying enzymes, or so-called epigenetic “writers” and “erasers”. Potent inhibitors of histone binding modules have not yet been described. Here we report a cell-permeable small molecule (JQ1) which binds competitively to acetyl-lysine recognition motifs, or bromodomains. High potency and specificity toward a subset of human bromodomains is explained by co-crystal structures with BRD4, revealing excellent shape complementarity with the acetyl-lysine binding cavity. Recurrent translocation of BRD4 is observed in a genetically-defined, incurable subtype of human squamous carcinoma. Competitive binding by JQ1 displaces the BRD4 fusion oncoprotein from chromatin, prompting squamous differentiation and specific anti-proliferative effects in BRD4-dependent cell lines and patient-derived xenograft models. These data establish proof of concept for targeting protein-protein interactions of epigenetic “readers” and provide a versatile chemical scaffold for the development of chemical probes more broadly throughout the bromodomain family.
                Bookmark

                Author and article information

                Contributors
                linde@ohsu.edu
                Journal
                Leukemia
                Leukemia
                Leukemia
                Nature Publishing Group UK (London )
                0887-6924
                1476-5551
                21 January 2023
                21 January 2023
                2023
                : 37
                : 3
                : 580-592
                Affiliations
                [1 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, Department of Cell, Developmental & Cancer Biology, , Oregon Health & Science University, ; Portland, OR USA
                [2 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, Department of Molecular & Medical Genetics, , Oregon Health & Science University, ; Portland, OR USA
                [3 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, Department of Molecular Microbiology and Immunology, , Oregon Health & Science University, ; Portland, OR USA
                [4 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, School of Medicine, , Oregon Health & Science University, ; Portland, OR USA
                [5 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, Knight Cancer Institute, , Oregon Health & Science University, ; Portland, OR USA
                [6 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, Knight Cardiovascular Institute, , Oregon Health & Science University, ; Portland, OR USA
                [7 ]GRID grid.5288.7, ISNI 0000 0000 9758 5690, Center for Early Detection Advanced Research, , Oregon Health & Science University, ; Portland, OR USA
                Author information
                http://orcid.org/0000-0003-3757-7690
                http://orcid.org/0000-0002-2091-8869
                http://orcid.org/0000-0001-6110-6608
                http://orcid.org/0000-0001-7026-2012
                Article
                1808
                10.1038/s41375-023-01808-0
                9991923
                36681742
                1f329591-f565-4727-b32a-f423ef0e8fa6
                © The Author(s) 2023

                Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as 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 images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this license, visit http://creativecommons.org/licenses/by/4.0/.

                History
                : 26 May 2022
                : 8 December 2022
                : 4 January 2023
                Funding
                Funded by: FundRef https://doi.org/10.13039/100000092, U.S. Department of Health & Human Services | NIH | U.S. National Library of Medicine (NLM);
                Award ID: 5T15LM7088-28
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000057, U.S. Department of Health & Human Services | NIH | National Institute of General Medical Sciences (NIGMS);
                Award ID: R35GM124704
                Award Recipient :
                Funded by: FundRef https://doi.org/10.13039/100000054, U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI);
                Award ID: 1R01CA262145-01A1
                Award ID: U54CA224019
                Award ID: U01CA217862
                Award Recipient :
                Funded by: U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
                Funded by: U.S. Department of Health & Human Services | NIH | National Cancer Institute (NCI)
                Categories
                Article
                Custom metadata
                © Springer Nature Limited 2023

                Oncology & Radiotherapy
                tumour immunology,cancer models
                Oncology & Radiotherapy
                tumour immunology, cancer models

                Comments

                Comment on this article