4.7 Article

MYC suppresses STING-dependent innate immunity by transcriptionally upregulating DNMT1 in triple-negative breast cancer

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2021-002528

关键词

immunity; innate; immunotherapy; tumor escape; tumor microenvironment

资金

  1. National Key Research and Development Project of China [2020YFA0112304]
  2. National Natural Science Foundation of China [82002792]
  3. Shanghai Sailing Program [19YF1409000, 20YF1408600]
  4. Innovation Team of Ministry of Education [IRT1223]
  5. Shanghai Key Laboratory of Breast Cancer [12DZ2260100]

向作者/读者索取更多资源

This study identified two distinct microenvironment phenotypes, 'inflamed' and 'non-inflamed', within the classic basal-like subtype of TNBC, and revealed that MYC amplification and overexpression led to the formation of the non-inflamed TIME. Combination therapy with a DNA methyltransferase inhibitor and immunotherapy reversed T cell exhaustion and improved T cell function, resulting in potent antitumor activity in MYC-overexpressing TNBC.
Background Triple-negative breast cancer (TNBC) is the most aggressive subtype of breast cancer and lacks definite treatment targets. Tumor immune microenvironment (TIME) heterogeneity has a profound impact on the immunotherapy response. Tumors with non-inflamed TIME derive limited benefit from immunotherapy. However, what drives the formation of the non-inflamed TIME in TNBC remains unclear. Methods Using our multiomics database of TNBC, we conducted an analysis to explore the key genomic events driving the formation of the non-inflamed TIME in TNBC. In vitro and in vivo studies further revealed potential mechanisms and the efficacy of combination treatment with immunotherapy. Results With transcriptomic and genomic data, we systematically analyzed the TIME of TNBC and revealed that the classical basal-like subtype of TNBC consisted of two distinct microenvironment phenotypes, defined as the 'inflamed' and 'non-inflamed' subtypes. We performed further screening and demonstrated that MYC amplification and overexpression led to low immune infiltration and cytolytic activity in TIME. Mechanistically, MYC bound to DNMT1 promoter and activated DNMT1 transcription in TNBC cells, thus suppressing the Cyclic GMP-AMP synthase (cGAS)-STING pathway via an epigenetic regulatory way. In MYC-overexpressing TNBC, decitabine, an Food and Drug Administration (FDA)-approved DNA methyltransferase inhibitor, converted tumors from non-inflamed to inflamed tumors by enhancing T cell infiltration. Furthermore, the combination of decitabine with programmed cell death protein 1 (PD-1) inhibitor reversed T cell exhaustion and improved T cell function in mouse models, which elicited potent antitumor activity in MYC-overexpressing TNBC. Conclusions Our work elucidates that the classic oncogene MYC induces immune evasion by repressing innate immunity. Furthermore, we provide a rationale for combining DNA methyltransferase inhibition with immunotherapy for the treatment of MYC-overexpressing TNBC.

作者

我是这篇论文的作者
点击您的名字以认领此论文并将其添加到您的个人资料中。

评论

主要评分

4.7
评分不足

次要评分

新颖性
-
重要性
-
科学严谨性
-
评价这篇论文

推荐

暂无数据
暂无数据