4.7 Article

The microbiome-derived metabolite TMAO drives immune activation and boosts responses to immune checkpoint blockade in pancreatic cancer

期刊

SCIENCE IMMUNOLOGY
卷 7, 期 75, 页码 -

出版社

AMER ASSOC ADVANCEMENT SCIENCE
DOI: 10.1126/sciimmunol.abn0704

关键词

-

资金

  1. NIH [1R21CA259240-01, S10 OD023586]
  2. W. W. Smith Charitable Trust
  3. Caspar Wistar Fellows Program at the Wistar Institute [1R01CA252225-01A1, R50 CA221838]
  4. Wistar Institute Cancer Center Support Grant (CCSG) [P30 CA010815]
  5. 2022 Pancreatic Cancer Action Network Career Development Award 22-20-SHIN

向作者/读者索取更多资源

The gut microbe-derived metabolite TMAO enhances antitumor immunity in pancreatic ductal adenocarcinoma (PDAC) by promoting an immunostimulatory tumor-associated macrophage (TAM) phenotype and activating effector T cell response. TMAO exerts antitumor effects through potentiating the type I interferon pathway.
The composition of the gut microbiome can control innate and adaptive immunity and has emerged as a key regulator of tumor growth, especially in the context of immune checkpoint blockade (ICB) therapy. However, the underlying mechanisms for how the microbiome affects tumor growth remain unclear. Pancreatic ductal adenocarcinoma (PDAC) tends to be refractory to therapy, including ICB. Using a nontargeted, liquid chromatography-tandem mass spectrometry-based metabolomic screen, we identified the gut microbe-derived metabolite trimethylamine N-oxide (TMAO), which enhanced antitumor immunity to PDAC. Delivery of TMAO intraperitoneally or via a dietary choline supplement to orthotopic PDAC-bearing mice reduced tumor growth, associated with an immunostimulatory tumor-associated macrophage (TAM) phenotype, and activated effector T cell response in the tumor microenvironment. Mechanistically, TMAO potentiated the type I interferon (IFN) pathway and conferred antitumor effects in a type I IFN-dependent manner. Delivering TMAO-primed macrophages intravenously produced similar antitumor effects. Combining TMAO with ICB (anti-PD1 and/ or antiTim3) in a mouse model of PDAC significantly reduced tumor burden and improved survival beyond TMAO or ICB alone. Last, the levels of bacteria containing CutC ( an enzyme that generates trimethylamine, the TMAO precursor) correlated with long-term survival in patients with PDAC and improved response to antiPD1 in patients with melanoma. Together, our study identifies the gut microbial metabolite TMAO as a driver of antitumor immunity and lays the groundwork for potential therapeutic strategies targeting TMAO.

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