The type 2 cytokine Fc–IL-4 revitalizes exhausted CD8+ T cells against cancer

IF 50.5 1区 综合性期刊 Q1 MULTIDISCIPLINARY SCIENCES Nature Pub Date : 2024-09-25 DOI:10.1038/s41586-024-07962-4
Bing Feng, Zhiliang Bai, Xiaolei Zhou, Yang Zhao, Yu-Qing Xie, Xinyi Huang, Yang Liu, Tom Enbar, Rongrong Li, Yi Wang, Min Gao, Lucia Bonati, Mei-Wen Peng, Weilin Li, Bo Tao, Mélanie Charmoy, Werner Held, J. Joseph Melenhorst, Rong Fan, Yugang Guo, Li Tang
{"title":"The type 2 cytokine Fc–IL-4 revitalizes exhausted CD8+ T cells against cancer","authors":"Bing Feng, Zhiliang Bai, Xiaolei Zhou, Yang Zhao, Yu-Qing Xie, Xinyi Huang, Yang Liu, Tom Enbar, Rongrong Li, Yi Wang, Min Gao, Lucia Bonati, Mei-Wen Peng, Weilin Li, Bo Tao, Mélanie Charmoy, Werner Held, J. Joseph Melenhorst, Rong Fan, Yugang Guo, Li Tang","doi":"10.1038/s41586-024-07962-4","DOIUrl":null,"url":null,"abstract":"<p>Current cancer immunotherapy predominately focuses on eliciting type 1 immune responses fighting cancer; however, long-term complete remission remains uncommon<sup>1,2</sup>. A pivotal question arises as to whether type 2 immunity can be orchestrated alongside type 1-centric immunotherapy to achieve enduring response against cancer<sup>3,4</sup>. Here we show that an interleukin-4 fusion protein (Fc–IL-4), a typical type 2 cytokine, directly acts on CD8<sup>+</sup> T cells and enriches functional terminally exhausted CD8<sup>+</sup> T (CD8<sup>+</sup> T<sub>TE</sub>) cells in the tumour. Consequently, Fc–IL-4 enhances antitumour efficacy of type 1 immunity-centric adoptive T cell transfer or immune checkpoint blockade therapies and induces durable remission across several syngeneic and xenograft tumour models. Mechanistically, we discovered that Fc–IL-4 signals through both signal transducer and activator of transcription 6 (STAT6) and mammalian target of rapamycin (mTOR) pathways, augmenting the glycolytic metabolism and the nicotinamide adenine dinucleotide (NAD) concentration of CD8<sup>+</sup> T<sub>TE</sub> cells in a lactate dehydrogenase A-dependent manner. The metabolic modulation mediated by Fc–IL-4 is indispensable for reinvigorating intratumoural CD8<sup>+</sup> T<sub>TE</sub> cells. These findings underscore Fc–IL-4 as a potent type 2 cytokine-based immunotherapy that synergizes effectively with type 1 immunity to elicit long-lasting responses against cancer. Our study not only sheds light on the synergy between these two types of immune responses, but also unveils an innovative strategy for advancing next-generation cancer immunotherapy by integrating type 2 immune factors.</p>","PeriodicalId":18787,"journal":{"name":"Nature","volume":null,"pages":null},"PeriodicalIF":50.5000,"publicationDate":"2024-09-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Nature","FirstCategoryId":"103","ListUrlMain":"https://doi.org/10.1038/s41586-024-07962-4","RegionNum":1,"RegionCategory":"综合性期刊","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"MULTIDISCIPLINARY SCIENCES","Score":null,"Total":0}
引用次数: 0

Abstract

Current cancer immunotherapy predominately focuses on eliciting type 1 immune responses fighting cancer; however, long-term complete remission remains uncommon1,2. A pivotal question arises as to whether type 2 immunity can be orchestrated alongside type 1-centric immunotherapy to achieve enduring response against cancer3,4. Here we show that an interleukin-4 fusion protein (Fc–IL-4), a typical type 2 cytokine, directly acts on CD8+ T cells and enriches functional terminally exhausted CD8+ T (CD8+ TTE) cells in the tumour. Consequently, Fc–IL-4 enhances antitumour efficacy of type 1 immunity-centric adoptive T cell transfer or immune checkpoint blockade therapies and induces durable remission across several syngeneic and xenograft tumour models. Mechanistically, we discovered that Fc–IL-4 signals through both signal transducer and activator of transcription 6 (STAT6) and mammalian target of rapamycin (mTOR) pathways, augmenting the glycolytic metabolism and the nicotinamide adenine dinucleotide (NAD) concentration of CD8+ TTE cells in a lactate dehydrogenase A-dependent manner. The metabolic modulation mediated by Fc–IL-4 is indispensable for reinvigorating intratumoural CD8+ TTE cells. These findings underscore Fc–IL-4 as a potent type 2 cytokine-based immunotherapy that synergizes effectively with type 1 immunity to elicit long-lasting responses against cancer. Our study not only sheds light on the synergy between these two types of immune responses, but also unveils an innovative strategy for advancing next-generation cancer immunotherapy by integrating type 2 immune factors.

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
2 型细胞因子 Fc-IL-4 使衰竭的 CD8+ T 细胞重新焕发抗癌活力
目前的癌症免疫疗法主要集中于激发抗癌的 1 型免疫反应;然而,长期的完全缓解仍不常见1,2。一个关键问题是,能否在以 1 型免疫疗法为中心的同时协调 2 型免疫疗法,以获得持久的抗癌反应3,4。在这里,我们发现白细胞介素-4融合蛋白(Fc-IL-4)是一种典型的2型细胞因子,可直接作用于CD8+ T细胞,并在肿瘤中富集功能性终末衰竭CD8+ T(CD8+ TTE)细胞。因此,Fc-IL-4能增强以1型免疫为中心的收养性T细胞转移或免疫检查点阻断疗法的抗肿瘤疗效,并能在多种合成和异种移植肿瘤模型中诱导持久缓解。从机理上讲,我们发现Fc-IL-4通过信号转导和转录激活因子6(STAT6)和哺乳动物雷帕霉素靶标(mTOR)途径发出信号,以乳酸脱氢酶A依赖性方式增强CD8+ TTE细胞的糖代谢和烟酰胺腺嘌呤二核苷酸(NAD)浓度。Fc-IL-4 介导的新陈代谢调节对重振瘤内 CD8+ TTE 细胞不可或缺。这些研究结果表明,Fc-IL-4 是一种强效的基于 2 型细胞因子的免疫疗法,它能与 1 型免疫有效协同,激发持久的抗癌反应。我们的研究不仅揭示了这两类免疫反应之间的协同作用,还揭示了通过整合 2 型免疫因子推进下一代癌症免疫疗法的创新策略。
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
Nature
Nature 综合性期刊-综合性期刊
CiteScore
90.00
自引率
1.20%
发文量
3652
审稿时长
3 months
期刊介绍: Nature is a prestigious international journal that publishes peer-reviewed research in various scientific and technological fields. The selection of articles is based on criteria such as originality, importance, interdisciplinary relevance, timeliness, accessibility, elegance, and surprising conclusions. In addition to showcasing significant scientific advances, Nature delivers rapid, authoritative, insightful news, and interpretation of current and upcoming trends impacting science, scientists, and the broader public. The journal serves a dual purpose: firstly, to promptly share noteworthy scientific advances and foster discussions among scientists, and secondly, to ensure the swift dissemination of scientific results globally, emphasizing their significance for knowledge, culture, and daily life.
期刊最新文献
Mutant maize with a ‘smart canopy’ evades the shade at high planting density A virus-derived enzyme can destroy the membrane structures that protect bacteria How artificial intelligence can help to keep us safe Bacteria implanted into fungi offer clues to the origins of complex life Nearly 50% of researchers quit science within a decade, huge study reveals
×
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