高级检索
当前位置: 首页 > 详情页

4E-BP1 Is a Tumor Suppressor Protein Reactivated by mTOR Inhibition in Head and Neck Cancer.

文献详情

资源类型:
Pubmed体系:

收录情况: ◇ 自然指数

机构: [1]Moores Cancer Center, University of California, San Diego, La Jolla, California. [2]State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China. [3]National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, Maryland. [4]National Cancer Institute, National Institutes of Health, Bethesda, Maryland. [5]International College of Dentistry, Walailak University, Nakhon Si Thammarat, Thailand. [6]Department of Pharmacology, University of California, San Diego, La Jolla, California. [7]Department of Biochemistry and Goodman Cancer Centre, McGill University, Montreal, Quebec, Canada
出处:
ISSN:

摘要:
Aberrant activation of the PI3K-mTOR signaling pathway occurs in >80% of head and neck squamous cell carcinomas (HNSCC), and overreliance on this signaling circuit may in turn represent a cancer-specific vulnerability that can be exploited therapeutically. mTOR inhibitors (mTORi) promote tumor regression in genetically defined and chemically induced HNSCC animal models, and encouraging results have been recently reported. However, the mTOR-regulated targets contributing to the clinical response have not yet been identified. Here, we focused on EIF4E-BP1 (4E-BP1), a direct target of mTOR that serves as key effector for protein synthesis. A systematic analysis of genomic alterations in the PIK3CA-mTOR pathway in HNSCC revealed that 4E-BP1 is rarely mutated, but at least one 4E-BP1 gene copy is lost in over 35% of the patients with HNSCC, correlating with decreased 4E-BP1 protein expression. 4E-BP1 gene copy number loss correlated with poor disease-free and overall survival. Aligned with a tumor-suppressive role, 4e-bp1/2 knockout mice formed larger and more lesions in models of HNSCC carcinogenesis. mTORi treatment or conditional expression of a mutant 4E-BP1 that cannot be phosphorylated by mTOR was sufficient to disrupt the translation-initiation complex and prevent tumor growth. Furthermore, CRISPR/Cas9-targeted 4E-BP1 HNSCC cells resulted in reduced sensitivity to mTORi in vitro and in vivo. Overall, these findings indicate that in HNSCC, mTOR persistently restrains 4E-BP1 via phosphorylation and that mTORi can restore the tumor-suppressive function of 4E-BP1. Our findings also support 4E-BP1 expression and phosphorylation status as a mechanistic biomarker of mTORi sensitivity in patients with HNSCC. SIGNIFICANCE: These findings suggest that EIF4E-BP1 acts as a tumor suppressor in HNSCC and that 4E-BP1 dephosphorylation mediates the therapeutic response to mTORi, providing a mechanistic biomarker for future precision oncology trials. ©2019 American Association for Cancer Research.

基金:
语种:
PubmedID:
中科院(CAS)分区:
出版当年[2019]版:
大类 | 1 区 医学
小类 | 1 区 肿瘤学
最新[2023]版:
大类 | 1 区 医学
小类 | 2 区 肿瘤学
第一作者:
第一作者机构: [1]Moores Cancer Center, University of California, San Diego, La Jolla, California. [2]State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China.
通讯作者:
通讯机构: [1]Moores Cancer Center, University of California, San Diego, La Jolla, California. [6]Department of Pharmacology, University of California, San Diego, La Jolla, California. [*1]University of California, San Diego, 3855 Health Sciences Drive, La Jolla, CA 92093. [*2]West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan 610041, China.
推荐引用方式(GB/T 7714):
APA:
MLA:

资源点击量:43389 今日访问量:0 总访问量:3120 更新日期:2024-09-01 建议使用谷歌、火狐浏览器 常见问题

版权所有©2020 四川省肿瘤医院 技术支持:重庆聚合科技有限公司 地址:成都市人民南路四段55号