Metabolic inhibitor screening identifies dihydrofolate reductase as an inducer of the tumor immune escape mediator CD24

Immunomedicine Pub Date : 2022-12-02 DOI:10.1002/imed.1041
Austin C. Boese MS, Jung Seok Hwang PhD, Isabelle Young MS, Courteney M. Malin BS, Vanessa Avalos BS, JiHoon Kang PhD, Sumin Kang PhD
{"title":"Metabolic inhibitor screening identifies dihydrofolate reductase as an inducer of the tumor immune escape mediator CD24","authors":"Austin C. Boese MS,&nbsp;Jung Seok Hwang PhD,&nbsp;Isabelle Young MS,&nbsp;Courteney M. Malin BS,&nbsp;Vanessa Avalos BS,&nbsp;JiHoon Kang PhD,&nbsp;Sumin Kang PhD","doi":"10.1002/imed.1041","DOIUrl":null,"url":null,"abstract":"<p>Immune checkpoint inhibitors (ICIs) have improved the clinical management of some cancer cases, yet patients still fail to respond to immunotherapy. Dysregulated metabolism is a common feature of many cancers, and metabolites are known to modulate functions in cancer cells. To identify potential metabolic pathways involved in anti-tumor immune response, we employed a metabolic inhibitor-based drug screen in human lung cancer cell lines and examined expression changes in a panel of immune regulator genes. Notably, pharmacologic inhibition of dihydrofolate reductase (DHFR) downregulated cancer cell expression of cluster of differentiation 24 (CD24), an anti-phagocytic surface protein. Genetic modulation of DHFR resulted in decreased CD24 expression, whereas tetrahydrofolate, the product of DHFR, enhanced CD24 expression. DHFR inhibition and the consequent CD24 decrease enhanced T cell-mediated tumor cell killing, whereas replenishment of DHFR or CD24 partially mitigated the immune-mediated tumor cell killing that resulted from methotrexate treatment in cancer cells. Moreover, publicly available clinical data analyses further revealed the link between DHFR, CD24, and the antitumor immune response in lung cancer patients. Our study highlights a novel connection between folate metabolism and the anti-tumor immune response and partially interprets how DHFR inhibitors lead to clinical benefits when combined with cancer immunotherapy agents.</p>","PeriodicalId":73348,"journal":{"name":"Immunomedicine","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2022-12-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9937563/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Immunomedicine","FirstCategoryId":"1085","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/imed.1041","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Immune checkpoint inhibitors (ICIs) have improved the clinical management of some cancer cases, yet patients still fail to respond to immunotherapy. Dysregulated metabolism is a common feature of many cancers, and metabolites are known to modulate functions in cancer cells. To identify potential metabolic pathways involved in anti-tumor immune response, we employed a metabolic inhibitor-based drug screen in human lung cancer cell lines and examined expression changes in a panel of immune regulator genes. Notably, pharmacologic inhibition of dihydrofolate reductase (DHFR) downregulated cancer cell expression of cluster of differentiation 24 (CD24), an anti-phagocytic surface protein. Genetic modulation of DHFR resulted in decreased CD24 expression, whereas tetrahydrofolate, the product of DHFR, enhanced CD24 expression. DHFR inhibition and the consequent CD24 decrease enhanced T cell-mediated tumor cell killing, whereas replenishment of DHFR or CD24 partially mitigated the immune-mediated tumor cell killing that resulted from methotrexate treatment in cancer cells. Moreover, publicly available clinical data analyses further revealed the link between DHFR, CD24, and the antitumor immune response in lung cancer patients. Our study highlights a novel connection between folate metabolism and the anti-tumor immune response and partially interprets how DHFR inhibitors lead to clinical benefits when combined with cancer immunotherapy agents.

Abstract Image

代谢抑制剂筛选确定二氢叶酸还原酶作为肿瘤免疫逃逸介质CD24的诱导剂
免疫检查点抑制剂(ICIs)已经改善了一些癌症病例的临床管理,但患者仍然无法对免疫治疗产生反应。代谢失调是许多癌症的共同特征,代谢产物被认为可以调节癌细胞的功能。为了确定参与抗肿瘤免疫反应的潜在代谢途径,我们在人肺癌细胞系中采用了基于代谢抑制剂的药物筛选,并检测了一组免疫调节基因的表达变化。值得注意的是,二氢叶酸还原酶(DHFR)的药理抑制下调了癌细胞的CD24(一种抗吞噬表面蛋白)的表达。DHFR基因调控导致CD24表达降低,而DHFR产物四氢叶酸则增强CD24表达。DHFR抑制和随后的CD24减少增强了T细胞介导的肿瘤细胞杀伤,而DHFR或CD24的补充部分减轻了甲氨蝶呤治疗对癌细胞造成的免疫介导的肿瘤细胞杀伤。此外,公开的临床数据分析进一步揭示了DHFR、CD24与肺癌患者抗肿瘤免疫反应之间的联系。我们的研究强调了叶酸代谢与抗肿瘤免疫反应之间的新联系,并部分解释了DHFR抑制剂与癌症免疫治疗药物联合使用时如何带来临床益处。
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 求助全文
来源期刊
自引率
0.00%
发文量
0
×
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
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信