4.7 Article

Complement downregulation promotes an inflammatory signature that renders colorectal cancer susceptible to immunotherapy

期刊

出版社

BMJ PUBLISHING GROUP
DOI: 10.1136/jitc-2022-004717

关键词

-

资金

  1. 'Lenino Fontana and Maria Lionello' AIRC fellowship
  2. Umberto Veronesi fellowship
  3. ACS-IRG
  4. NIH [P20GM130457, R01CA258882-01A1]
  5. MRC [MC_UU_00001/10]
  6. CRUK (UKRI) [MR/M016587/1]
  7. Cancer Research UK

向作者/读者索取更多资源

The dysregulation of complement anaphylatoxin C3a receptor (C3aR) in subsets of patients with CRC affects immune responses, the development of CRC, and response to immune checkpoint blockade therapy. Downregulation of C3ar1 in human CRC promotes a tumor microenvironment characterized by the accumulation of innate and adaptive immune cells supporting antitumor immunity. Lack of C3a in the colon activates a microbiota-mediated proinflammatory program promoting the development of tumors responsive to ICB therapy.
Background and aims The role of inflammatory immune responses in colorectal cancer (CRC) development and response to therapy is a matter of intense debate. While inflammation is a known driver of CRC, inflammatory immune infiltrates are a positive prognostic factor in CRC and predispose to response to immune checkpoint blockade (ICB) therapy. Unfortunately, over 85% of CRC cases are primarily unresponsive to ICB due to the absence of an immune infiltrate, and even the cases that show an initial immune infiltration can become refractory to ICB. The identification of therapy supportive immune responses in the field has been partially hindered by the sparsity of suitable mouse models to recapitulate the human disease. In this study, we aimed to understand how the dysregulation of the complement anaphylatoxin C3a receptor (C3aR), observed in subsets of patients with CRC, affects the immune responses, the development of CRC, and response to ICB therapy. Methods We use a comprehensive approach encompassing analysis of publicly available human CRC datasets, inflammation-driven and newly generated spontaneous mouse models of CRC, and multiplatform high-dimensional analysis of immune responses using microbiota sequencing, RNA sequencing, and mass cytometry. Results We found that patients' regulation of the complement C3aR is associated with epigenetic modifications. Specifically, downregulation of C3ar1 in human CRC promotes a tumor microenvironment characterized by the accumulation of innate and adaptive immune cells that support antitumor immunity. In addition, in vivo studies in our newly generated mouse model revealed that the lack of C3a in the colon activates a microbiota-mediated proinflarnrnatory program which promotes the development of tumors with an immune signature that renders them responsive to the ICB therapy. Conclusions Our findings reveal that C3aR may act as a previously unrecognized checkpoint to enhance antitumor immunity in CRC. C3aR can thus be exploited to overcome ICB resistance in a larger group of patients with CRC.

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