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

Author:

Mirji Gauri1ORCID,Worth Alison1,Bhat Sajad Ahmad1,El Sayed Mohamed1,Kannan Toshitha2,Goldman Aaron R.3ORCID,Tang Hsin-Yao3ORCID,Liu Qin4ORCID,Auslander Noam4,Dang Chi V.45,Abdel-Mohsen Mohamed16ORCID,Kossenkov Andrew2ORCID,Stanger Ben Z.78910ORCID,Shinde Rahul S.1ORCID

Affiliation:

1. Immunology, Microenvironment and Metastasis Program, the Wistar Institute, Philadelphia, PA, USA.

2. Bioinformatics Facility, the Wistar Institute, Philadelphia, PA, USA.

3. Proteomics and Metabolomics Facility, the Wistar Institute, Philadelphia, PA, USA.

4. Molecular and Cellular Oncogenesis Program, the Wistar Institute, Philadelphia, PA, USA.

5. Ludwig Institute for Cancer Research, New York, NY, USA.

6. Vaccine and Immunotherapy Center, the Wistar Institute, Philadelphia, PA, USA.

7. Abramson Family Cancer Research Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.

8. Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA.

9. Department of Medicine, University of Pennsylvania, Philadelphia, PA, USA.

10. Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA, USA.

Abstract

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 anti-Tim3) 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 anti-PD1 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.

Publisher

American Association for the Advancement of Science (AAAS)

Subject

General Medicine,Immunology

同舟云学术

1.学者识别学者识别

2.学术分析学术分析

3.人才评估人才评估

"同舟云学术"是以全球学者为主线,采集、加工和组织学术论文而形成的新型学术文献查询和分析系统,可以对全球学者进行文献检索和人才价值评估。用户可以通过关注某些学科领域的顶尖人物而持续追踪该领域的学科进展和研究前沿。经过近期的数据扩容,当前同舟云学术共收录了国内外主流学术期刊6万余种,收集的期刊论文及会议论文总量共计约1.5亿篇,并以每天添加12000余篇中外论文的速度递增。我们也可以为用户提供个性化、定制化的学者数据。欢迎来电咨询!咨询电话:010-8811{复制后删除}0370

www.globalauthorid.com

TOP

Copyright © 2019-2024 北京同舟云网络信息技术有限公司
京公网安备11010802033243号  京ICP备18003416号-3