GSTDTAP  > 气候变化
DOI10.1126/science.abc3421
Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy
Lukas F. Mager; Regula Burkhard; Nicola Pett; Noah C. A. Cooke; Kirsty Brown; Hena Ramay; Seungil Paik; John Stagg; Ryan A. Groves; Marco Gallo; Ian A. Lewis; Markus B. Geuking; Kathy D. McCoy
2020-09-18
发表期刊Science
出版年2020
英文摘要Checkpoint blockade immunotherapy harnesses the immune system to kill cancer cells and has been used with great success to treat certain tumors, but not all cancer patients respond. The efficacy of checkpoint blockade immunotherapy has been shown to depend on the presence of distinct, beneficial bacteria residing in the gut of patients, but how the microbiome mediates such beneficial effects is unclear. Mager et al. found that specific bacteria produce a metabolite called inosine that enhances the effect of checkpoint blockade immunotherapy (see the Perspective by Shaikh and Sears). In mouse models, inosine, together with proinflammatory stimuli and immunotherapy, strongly enhanced the antitumor capacities of T cells in multiple tumor types, including colorectal cancer, bladder cancer, and melanoma. Science , this issue p. [1481][1]; see also p. [1427][2] Several species of intestinal bacteria have been associated with enhanced efficacy of checkpoint blockade immunotherapy, but the underlying mechanisms by which the microbiome enhances antitumor immunity are unclear. In this study, we isolated three bacterial species— Bifidobacterium pseudolongum , Lactobacillus johnsonii , and Olsenella species—that significantly enhanced efficacy of immune checkpoint inhibitors in four mouse models of cancer. We found that intestinal B. pseudolongum modulated enhanced immunotherapy response through production of the metabolite inosine. Decreased gut barrier function induced by immunotherapy increased systemic translocation of inosine and activated antitumor T cells. The effect of inosine was dependent on T cell expression of the adenosine A2A receptor and required costimulation. Collectively, our study identifies a previously unknown microbial metabolite immune pathway activated by immunotherapy that may be exploited to develop microbial-based adjuvant therapies. [1]: /lookup/doi/10.1126/science.abc3421 [2]: /lookup/doi/10.1126/science.abe0709
领域气候变化 ; 资源环境
URL查看原文
引用统计
文献类型期刊论文
条目标识符http://119.78.100.173/C666/handle/2XK7JSWQ/295427
专题气候变化
资源环境科学
推荐引用方式
GB/T 7714
Lukas F. Mager,Regula Burkhard,Nicola Pett,et al. Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy[J]. Science,2020.
APA Lukas F. Mager.,Regula Burkhard.,Nicola Pett.,Noah C. A. Cooke.,Kirsty Brown.,...&Kathy D. McCoy.(2020).Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy.Science.
MLA Lukas F. Mager,et al."Microbiome-derived inosine modulates response to checkpoint inhibitor immunotherapy".Science (2020).
条目包含的文件
条目无相关文件。
个性服务
推荐该条目
保存到收藏夹
查看访问统计
导出为Endnote文件
谷歌学术
谷歌学术中相似的文章
[Lukas F. Mager]的文章
[Regula Burkhard]的文章
[Nicola Pett]的文章
百度学术
百度学术中相似的文章
[Lukas F. Mager]的文章
[Regula Burkhard]的文章
[Nicola Pett]的文章
必应学术
必应学术中相似的文章
[Lukas F. Mager]的文章
[Regula Burkhard]的文章
[Nicola Pett]的文章
相关权益政策
暂无数据
收藏/分享
所有评论 (0)
暂无评论
 

除非特别说明,本系统中所有内容都受版权保护,并保留所有权利。