Riluzole Enhancing anti-PD-1 Efficacy by Activating cGAS/STING Signaling in Colorectal Cancer.

IF 5.3 2区 医学 Q1 ONCOLOGY Molecular Cancer Therapeutics Pub Date : 2024-10-09 DOI:10.1158/1535-7163.MCT-24-0289
Beiyuan Liang, Misbah Khan, Hayden Storts, Evan H Zhang, Xinru Zheng, Xuanxuan Xing, Hazel Claybon, Jenna Wilson, Chunjie Li, Ning Jin, Richard Fishel, Wayne O Miles, Jing J Wang
{"title":"Riluzole Enhancing anti-PD-1 Efficacy by Activating cGAS/STING Signaling in Colorectal Cancer.","authors":"Beiyuan Liang, Misbah Khan, Hayden Storts, Evan H Zhang, Xinru Zheng, Xuanxuan Xing, Hazel Claybon, Jenna Wilson, Chunjie Li, Ning Jin, Richard Fishel, Wayne O Miles, Jing J Wang","doi":"10.1158/1535-7163.MCT-24-0289","DOIUrl":null,"url":null,"abstract":"<p><p>Colorectal cancer is the second leading cause of cancer mortality in the US. Although immune checkpoint blockade therapies including anti-PD-1/PD-L1 have been successful in treating a subset of colorectal cancer patients, response rates remain low. We have found that riluzole, a well-tolerated FDA-approved oral medicine for treating amyotrophic lateral sclerosis, increased intratumoral CD8+ T cells and suppressed tumor growth of colon cancer cells in syngeneic immune competent mice. Riluzole-mediated tumor suppression was dependent on the presence of CD8+ T cells. Riluzole activates the cytosolic DNA sensing cGAS/STING pathway in colon cancer cells, resulting in increased expression of interferon β (IFNβ) and IFNβ-regulated genes including CXCL10. Inhibition of ATM, but not ATR, resulted in a synergistic increase in IFNβ expression, suggesting that riluzole induces ATM-mediated damage response that contribute to cGAS/STING activation. Depletion of cGAS or STING significantly attenuated riluzole-induced expression of IFNβ and CXCL10 as well as increase of intratumoral CD8+ T cells and suppression of tumor growth. These results indicate that riluzole-mediated tumor infiltration of CD8+ T cells and attenuation of tumor growth is dependent on tumor cell intrinsic STING activation. To determine whether riluzole treatment primes the tumor microenvironment for immune checkpoint modulation, riluzole was combined with anti-PD-1 treatment. This combination showed greater efficacy than either single agent, and strongly suppressed tumor growth in vivo. Taken together, our studies indicate that riluzole activates cGAS/STING-mediated innate immune responses, which might be exploited to sensitize colorectal tumors to anti-PD-1/PD-L1 therapies. .</p>","PeriodicalId":18791,"journal":{"name":"Molecular Cancer Therapeutics","volume":null,"pages":null},"PeriodicalIF":5.3000,"publicationDate":"2024-10-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Cancer Therapeutics","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1158/1535-7163.MCT-24-0289","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"ONCOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Colorectal cancer is the second leading cause of cancer mortality in the US. Although immune checkpoint blockade therapies including anti-PD-1/PD-L1 have been successful in treating a subset of colorectal cancer patients, response rates remain low. We have found that riluzole, a well-tolerated FDA-approved oral medicine for treating amyotrophic lateral sclerosis, increased intratumoral CD8+ T cells and suppressed tumor growth of colon cancer cells in syngeneic immune competent mice. Riluzole-mediated tumor suppression was dependent on the presence of CD8+ T cells. Riluzole activates the cytosolic DNA sensing cGAS/STING pathway in colon cancer cells, resulting in increased expression of interferon β (IFNβ) and IFNβ-regulated genes including CXCL10. Inhibition of ATM, but not ATR, resulted in a synergistic increase in IFNβ expression, suggesting that riluzole induces ATM-mediated damage response that contribute to cGAS/STING activation. Depletion of cGAS or STING significantly attenuated riluzole-induced expression of IFNβ and CXCL10 as well as increase of intratumoral CD8+ T cells and suppression of tumor growth. These results indicate that riluzole-mediated tumor infiltration of CD8+ T cells and attenuation of tumor growth is dependent on tumor cell intrinsic STING activation. To determine whether riluzole treatment primes the tumor microenvironment for immune checkpoint modulation, riluzole was combined with anti-PD-1 treatment. This combination showed greater efficacy than either single agent, and strongly suppressed tumor growth in vivo. Taken together, our studies indicate that riluzole activates cGAS/STING-mediated innate immune responses, which might be exploited to sensitize colorectal tumors to anti-PD-1/PD-L1 therapies. .

查看原文
分享 分享
微信好友 朋友圈 QQ好友 复制链接
本刊更多论文
利鲁唑通过激活结直肠癌中的cGAS/STING信号增强抗PD-1疗效
结直肠癌是美国癌症死亡的第二大原因。尽管包括抗PD-1/PD-L1在内的免疫检查点阻断疗法已成功治疗了一部分结直肠癌患者,但反应率仍然很低。我们发现,经 FDA 批准用于治疗肌萎缩侧索硬化症的利鲁唑是一种耐受性良好的口服药物,它能增加瘤内 CD8+ T 细胞,抑制合成免疫小鼠结肠癌细胞的肿瘤生长。利鲁唑介导的肿瘤抑制依赖于 CD8+ T 细胞的存在。利鲁唑可激活结肠癌细胞中的细胞DNA感应cGAS/STING通路,导致干扰素β(IFNβ)和IFNβ调控基因(包括CXCL10)的表达增加。抑制ATM(而非ATR)会导致IFNβ表达的协同增加,这表明利鲁唑诱导了ATM介导的损伤反应,从而促进了cGAS/STING的激活。cGAS 或 STING 的耗竭可显著降低利鲁唑诱导的 IFNβ 和 CXCL10 的表达以及瘤内 CD8+ T 细胞的增加和肿瘤生长的抑制。这些结果表明,利鲁唑介导的 CD8+ T 细胞肿瘤浸润和肿瘤生长抑制依赖于肿瘤细胞内在的 STING 激活。为了确定利鲁唑治疗是否为免疫检查点调控提供了肿瘤微环境,利鲁唑与抗PD-1治疗相结合。这种联合疗法的疗效优于任何一种单药,并能强烈抑制体内肿瘤的生长。总之,我们的研究表明,利鲁唑能激活 cGAS/STING 介导的先天性免疫反应,可以利用这种反应使结直肠肿瘤对抗 PD-1/PD-L1 疗法敏感。.
本文章由计算机程序翻译,如有差异,请以英文原文为准。
求助全文
约1分钟内获得全文 去求助
来源期刊
CiteScore
11.20
自引率
1.80%
发文量
331
审稿时长
3 months
期刊介绍: Molecular Cancer Therapeutics will focus on basic research that has implications for cancer therapeutics in the following areas: Experimental Cancer Therapeutics, Identification of Molecular Targets, Targets for Chemoprevention, New Models, Cancer Chemistry and Drug Discovery, Molecular and Cellular Pharmacology, Molecular Classification of Tumors, and Bioinformatics and Computational Molecular Biology. The journal provides a publication forum for these emerging disciplines that is focused specifically on cancer research. Papers are stringently reviewed and only those that report results of novel, timely, and significant research and meet high standards of scientific merit will be accepted for publication.
期刊最新文献
A small molecule BCL6 inhibitor effectively suppresses diffuse large B cell lymphoma cells growth. Riluzole Enhancing anti-PD-1 Efficacy by Activating cGAS/STING Signaling in Colorectal Cancer. Tumor integrin-targeted glucose oxidase enzyme promotes ROS-mediated cell death that combines with interferon alpha therapy for tumor control. Tumor-specific antigen delivery for T-cell therapy via a pH-sensitive peptide conjugate. Zelenirstat Inhibits N-Myristoyltransferases to Disrupt Src Family Kinase Signalling and Oxidative Phosphorylation Killing Acute Myeloid Leukemia Cells.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
已复制链接
已复制链接
快去分享给好友吧!
我知道了
×
扫码分享
扫码分享
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1