4.7 Article

Tumor Cell IDO Enhances Immune Suppression and Decreases Survival Independent of Tryptophan Metabolism in Glioblastoma

期刊

CLINICAL CANCER RESEARCH
卷 27, 期 23, 页码 6514-6528

出版社

AMER ASSOC CANCER RESEARCH
DOI: 10.1158/1078-0432.CCR-21-1392

关键词

-

类别

资金

  1. NIH [R01NS102669, P50CA221747, R01NS097851, K02AG068617]
  2. UK Medical Research Council [MR/P025838/1, MR/M008959/1]
  3. BrainUp grant [2136]
  4. American Cancer Society [RSG-21058-01-CCE]

向作者/读者索取更多资源

This study reveals that nonenzymatic tumor cell IDO activity decreases survival and increases CFH and FHL-1 expression in human GBM independent of Trp metabolism. The increased intra-tumoral CFH and FHL-1 levels are associated with poorer survival in glioma patients. Like IDO effects, GBM cell FHL-1 expression increases intratumoral Treg and myeloid-derived suppressor cells while decreasing overall survival in mice with GBM, providing a new therapeutic target for GBM patients.
Purpose: Glioblastoma (GBM) is an incurable primary brain tumor that has not benefited from immunotherapy to date. More than 90% of GBM expresses the tryptophan (Trp) metabolic enzyme, indoleamine 2,3-dioxygenase 1 (IDO). This observation supported the historical hypothesis that IDO suppresses the antitumor immune response solely through a mechanism that requires intratumoral Trp depletion. However, recent findings led us to investigate the alter-native hypothesis that IDO suppresses the anti-GBM immune response independent of its association with Trp metabolism. Experimental Design: IDO-deficient GBM cell lines reconsti-tuted with IDO wild-type or IDO enzyme-null cDNA were created and validated in vitro and in vivo. Microarray analysis was con-ducted to search for genes that IDO regulates, followed by the analysis of human GBM cell lines, patient GBM and plasma, and The Cancer Genome Atlas (TCGA) database. Ex vivo cell coculture assays, syngeneic and humanized mouse GBM models, were used to test the alternative hypothesis. Results: Nonenzymic tumor cell IDO activity decreased the survival of experimental animals and increased the expression of complement factor H (CFH) and its isoform, factor H like protein 1 (FHL-1) in human GBM. Tumor cell IDO increased CFH and FHL-1 expression independent of Trp metabolism. Increased intra-tumoral CFH and FHL-1 levels were associated with poorer survival among patients with glioma. Similar to IDO effects, GBM cell FHL-1 expression increased intratumoral regulatory T cells (Treg) and myeloid-derived suppressor cells while it decreased overall survival in mice with GBM. Conclusions: Our study reveals a nonmetabolic IDO-mediated enhancement of CFH expression and provides a new therapeutic target for patients with GBM.

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