Mitotic MTH1 inhibitor TH1579 induces PD-L1 expression and inflammatory response through the cGAS-STING pathway

IF 5.9 2区 医学 Q1 ONCOLOGY Oncogenesis Pub Date : 2024-05-25 DOI:10.1038/s41389-024-00518-1
Jianyu Shen, Emilio Guillén Mancina, Shenyu Chen, Theodora Manolakou, Helge Gad, Ulrika Warpman Berglund, Kumar Sanjiv, Thomas Helleday
{"title":"Mitotic MTH1 inhibitor TH1579 induces PD-L1 expression and inflammatory response through the cGAS-STING pathway","authors":"Jianyu Shen, Emilio Guillén Mancina, Shenyu Chen, Theodora Manolakou, Helge Gad, Ulrika Warpman Berglund, Kumar Sanjiv, Thomas Helleday","doi":"10.1038/s41389-024-00518-1","DOIUrl":null,"url":null,"abstract":"<p>The mitotic MTH1 inhibitor TH1579 is a dual inhibitor that inhibits mitosis and incorporation of oxidative DNA damage and leads to cancer-specific cell death. The response to immune checkpoint inhibitor (ICI) treatment is often augmented by DNA damaging agents through the cGAS-STING pathway. This study investigates whether TH1579 can improve the efficacy of immune checkpoint blockades through its immunomodulatory properties. Various human and murine cancer cell lines were treated with mitotic MTH1i TH1579, and the expression of PD-L1 and T-cell infiltration-related chemokines was analysed by flow cytometry and real-time qPCR. Syngeneic mouse models were established to examine the combined effect of TH1579 and PD-L1 blockade. In our investigation, we found that TH1579 upregulates PD-L1 expression at both the protein and mRNA levels in human cancer cell lines. However, in murine cell lines, the increase was less pronounced. An in vivo experiment in a syngeneic mouse melanoma model showed that TH1579 treatment significantly increased the efficacy of atezolizumab, an anti-PD-L1 antibody, compared to vehicle or atezolizumab monotherapy. Furthermore, TH1579 exhibited immune-modulatory properties, elevating cytokines such as IFN-β and chemokines including CCL5 and CXCL10, in a cGAS-STING pathway-dependent manner. In conclusion, TH1579 has the potential to improve ICI treatment by modulating immune checkpoint-related proteins and pathways.</p>","PeriodicalId":19489,"journal":{"name":"Oncogenesis","volume":null,"pages":null},"PeriodicalIF":5.9000,"publicationDate":"2024-05-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Oncogenesis","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1038/s41389-024-00518-1","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

The mitotic MTH1 inhibitor TH1579 is a dual inhibitor that inhibits mitosis and incorporation of oxidative DNA damage and leads to cancer-specific cell death. The response to immune checkpoint inhibitor (ICI) treatment is often augmented by DNA damaging agents through the cGAS-STING pathway. This study investigates whether TH1579 can improve the efficacy of immune checkpoint blockades through its immunomodulatory properties. Various human and murine cancer cell lines were treated with mitotic MTH1i TH1579, and the expression of PD-L1 and T-cell infiltration-related chemokines was analysed by flow cytometry and real-time qPCR. Syngeneic mouse models were established to examine the combined effect of TH1579 and PD-L1 blockade. In our investigation, we found that TH1579 upregulates PD-L1 expression at both the protein and mRNA levels in human cancer cell lines. However, in murine cell lines, the increase was less pronounced. An in vivo experiment in a syngeneic mouse melanoma model showed that TH1579 treatment significantly increased the efficacy of atezolizumab, an anti-PD-L1 antibody, compared to vehicle or atezolizumab monotherapy. Furthermore, TH1579 exhibited immune-modulatory properties, elevating cytokines such as IFN-β and chemokines including CCL5 and CXCL10, in a cGAS-STING pathway-dependent manner. In conclusion, TH1579 has the potential to improve ICI treatment by modulating immune checkpoint-related proteins and pathways.

Abstract Image

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
有丝分裂 MTH1 抑制剂 TH1579 通过 cGAS-STING 通路诱导 PD-L1 表达和炎症反应
有丝分裂 MTH1 抑制剂 TH1579 是一种双重抑制剂,可抑制有丝分裂和氧化 DNA 损伤的结合,并导致癌症特异性细胞死亡。DNA损伤剂通常会通过cGAS-STING途径增强对免疫检查点抑制剂(ICI)治疗的反应。本研究探讨了 TH1579 能否通过其免疫调节特性提高免疫检查点阻断剂的疗效。有丝分裂 MTH1i TH1579 处理了多种人类和鼠类癌细胞系,并通过流式细胞术和实时 qPCR 分析了 PD-L1 和 T 细胞浸润相关趋化因子的表达。为了研究 TH1579 和 PD-L1 阻断的联合效应,我们建立了小鼠合成模型。我们在研究中发现,TH1579 可在蛋白和 mRNA 水平上调人癌细胞系中 PD-L1 的表达。然而,在小鼠细胞系中,这种增加并不明显。一项在合成小鼠黑色素瘤模型中进行的体内实验表明,与药物或阿特珠单抗单药治疗相比,TH1579 治疗可显著提高阿特珠单抗(一种抗 PD-L1 抗体)的疗效。此外,TH1579 还具有免疫调节特性,能以 cGAS-STING 通路依赖的方式升高 IFN-β 等细胞因子以及 CCL5 和 CXCL10 等趋化因子。总之,TH1579 有可能通过调节免疫检查点相关蛋白和通路来改善 ICI 治疗。
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
Oncogenesis
Oncogenesis ONCOLOGY-
CiteScore
11.90
自引率
0.00%
发文量
70
审稿时长
26 weeks
期刊介绍: Oncogenesis is a peer-reviewed open access online journal that publishes full-length papers, reviews, and short communications exploring the molecular basis of cancer and related phenomena. It seeks to promote diverse and integrated areas of molecular biology, cell biology, oncology, and genetics.
期刊最新文献
The branched N-glycan of PD-L1 predicts immunotherapy responses in patients with recurrent/metastatic HNSCC. DKK1 as a chemoresistant protein modulates oxaliplatin responses in colorectal cancer. NRF2 signaling plays an essential role in cancer progression through the NRF2-GPX2-NOTCH3 axis in head and neck squamous cell carcinoma. TFCP2L1 drives stemness and enhances their resistance to Sorafenib treatment by modulating the NANOG/STAT3 pathway in hepatocellular carcinoma Tumor suppressor BAP1 suppresses disulfidptosis through the regulation of SLC7A11 and NADPH levels
×
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