Targeting the NLRP3 inflammasome abrogates cardiotoxicity of immune checkpoint blockers.

IF 10.3 1区 医学 Q1 IMMUNOLOGY
Yang Lu, Jiamin Gao, Yachen Hou, Han Yang, Dashuai Wang, Ge Zhang, Zhen Qin, Pengchong Du, Zhenwei Wang, Yunzhe Wang, Quanzhou Chen, Zhaowei Sun, Ping Li, Jinying Zhang, Junnan Tang
{"title":"Targeting the NLRP3 inflammasome abrogates cardiotoxicity of immune checkpoint blockers.","authors":"Yang Lu, Jiamin Gao, Yachen Hou, Han Yang, Dashuai Wang, Ge Zhang, Zhen Qin, Pengchong Du, Zhenwei Wang, Yunzhe Wang, Quanzhou Chen, Zhaowei Sun, Ping Li, Jinying Zhang, Junnan Tang","doi":"10.1136/jitc-2024-010127","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many malignant tumors. However, ICI-induced hyper-immune activation causes cardiotoxicity. Traditional treatments such as glucocorticoids and immunosuppressants have limited effectiveness and may even accelerate tumor growth. This study aimed to identify approaches that effectively reduce cardiotoxicity and simultaneously preserve or enhance the antitumor immunity of ICI therapy.</p><p><strong>Methods: </strong>ICI injection in melanoma-bearing C57BL/6J female mice was used to simulate cardiotoxicity in patients with tumor undergoing immune therapy. MCC950 was used to block nod-like receptor protein 3 (NLRP3) inflammasome activity. Echocardiography, immunofluorescence, flow cytometry, and reverse transcription quantitative polymerase chain reaction were used to assess cardiac function, immune cell populations, and inflammatory factor levels. Bulk and single-cell RNA sequencing was used to detect the changes in cardiac transcriptome and immunological network.</p><p><strong>Results: </strong>NLRP3 inhibition reduced inflammatory response and improved cardiac function. Notably, NLRP3 inhibition also resulted in a pronounced suppression of tumor growth. Single-cell RNA sequencing elucidated that MCC950 treatment reduced the cardiac infiltration of pathogenic macrophages, cytotoxic T cells, activated T cells, and their production of inflammatory cytokines, while enhancing the presence of reparative macrophages and naive T cells. In addition, MCC950 attenuated cardiotoxicity induced by dual programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immunotherapy and promoted tumor regression, and showed efficacy in treating established cardiotoxicity.</p><p><strong>Conclusions: </strong>Our findings provide a promising clinical approach for preventing and treating cardiotoxicity induced by ICIs, dissociating the antitumor efficacy of ICI-based therapies from their cardiotoxic side effects.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"13 1","pages":""},"PeriodicalIF":10.3000,"publicationDate":"2025-01-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11749606/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal for Immunotherapy of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1136/jitc-2024-010127","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Immune checkpoint inhibitors (ICIs) have revolutionized the treatment of many malignant tumors. However, ICI-induced hyper-immune activation causes cardiotoxicity. Traditional treatments such as glucocorticoids and immunosuppressants have limited effectiveness and may even accelerate tumor growth. This study aimed to identify approaches that effectively reduce cardiotoxicity and simultaneously preserve or enhance the antitumor immunity of ICI therapy.

Methods: ICI injection in melanoma-bearing C57BL/6J female mice was used to simulate cardiotoxicity in patients with tumor undergoing immune therapy. MCC950 was used to block nod-like receptor protein 3 (NLRP3) inflammasome activity. Echocardiography, immunofluorescence, flow cytometry, and reverse transcription quantitative polymerase chain reaction were used to assess cardiac function, immune cell populations, and inflammatory factor levels. Bulk and single-cell RNA sequencing was used to detect the changes in cardiac transcriptome and immunological network.

Results: NLRP3 inhibition reduced inflammatory response and improved cardiac function. Notably, NLRP3 inhibition also resulted in a pronounced suppression of tumor growth. Single-cell RNA sequencing elucidated that MCC950 treatment reduced the cardiac infiltration of pathogenic macrophages, cytotoxic T cells, activated T cells, and their production of inflammatory cytokines, while enhancing the presence of reparative macrophages and naive T cells. In addition, MCC950 attenuated cardiotoxicity induced by dual programmed cell death protein 1 (PD-1) and cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) immunotherapy and promoted tumor regression, and showed efficacy in treating established cardiotoxicity.

Conclusions: Our findings provide a promising clinical approach for preventing and treating cardiotoxicity induced by ICIs, dissociating the antitumor efficacy of ICI-based therapies from their cardiotoxic side effects.

靶向NLRP3炎性体可消除免疫检查点阻滞剂的心脏毒性。
背景:免疫检查点抑制剂(ICIs)已经彻底改变了许多恶性肿瘤的治疗。然而,ici诱导的超免疫激活会引起心脏毒性。传统的治疗方法,如糖皮质激素和免疫抑制剂的效果有限,甚至可能加速肿瘤的生长。本研究旨在确定有效降低心脏毒性的方法,同时保持或增强ICI治疗的抗肿瘤免疫。方法:采用注射ICI对携带黑素瘤的C57BL/6J雌性小鼠进行免疫治疗,模拟肿瘤患者的心脏毒性。MCC950用于阻断淋巴结样受体蛋白3 (NLRP3)炎性体活性。超声心动图、免疫荧光、流式细胞术和逆转录定量聚合酶链反应用于评估心功能、免疫细胞群和炎症因子水平。采用大体积和单细胞RNA测序检测心脏转录组和免疫网络的变化。结果:抑制NLRP3可减轻炎症反应,改善心功能。值得注意的是,NLRP3的抑制也导致了肿瘤生长的明显抑制。单细胞RNA测序表明,MCC950治疗减少了致病性巨噬细胞、细胞毒性T细胞、活化T细胞的心脏浸润及其炎症细胞因子的产生,同时增强了修复性巨噬细胞和幼稚T细胞的存在。此外,MCC950可减轻双程序性细胞死亡蛋白1 (PD-1)和细胞毒性t淋巴细胞相关蛋白4 (CTLA-4)免疫治疗诱导的心脏毒性,促进肿瘤消退,并对已建立的心脏毒性有疗效。结论:我们的研究结果为预防和治疗ICIs诱导的心脏毒性提供了一种有希望的临床方法,将基于ICIs的治疗方法的抗肿瘤疗效与其心脏毒性副作用分离开来。
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
Journal for Immunotherapy of Cancer
Journal for Immunotherapy of Cancer Biochemistry, Genetics and Molecular Biology-Molecular Medicine
CiteScore
17.70
自引率
4.60%
发文量
522
审稿时长
18 weeks
期刊介绍: The Journal for ImmunoTherapy of Cancer (JITC) is a peer-reviewed publication that promotes scientific exchange and deepens knowledge in the constantly evolving fields of tumor immunology and cancer immunotherapy. With an open access format, JITC encourages widespread access to its findings. The journal covers a wide range of topics, spanning from basic science to translational and clinical research. Key areas of interest include tumor-host interactions, the intricate tumor microenvironment, animal models, the identification of predictive and prognostic immune biomarkers, groundbreaking pharmaceutical and cellular therapies, innovative vaccines, combination immune-based treatments, and the study of immune-related toxicity.
×
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