SMAD7 expression in CAR-T cells improves persistence and safety for solid tumors

IF 21.8 1区 医学 Q1 IMMUNOLOGY Cellular &Molecular Immunology Pub Date : 2024-01-04 DOI:10.1038/s41423-023-01120-y
Sixin Liang, Rui Zheng, Baile Zuo, Jia Li, Yiyi Wang, Yujie Han, Hao Dong, Xiaojuan Zhao, Yiting Zhang, Pengju Wang, Ruotong Meng, Lintao Jia, Angang Yang, Bo Yan
{"title":"SMAD7 expression in CAR-T cells improves persistence and safety for solid tumors","authors":"Sixin Liang, Rui Zheng, Baile Zuo, Jia Li, Yiyi Wang, Yujie Han, Hao Dong, Xiaojuan Zhao, Yiting Zhang, Pengju Wang, Ruotong Meng, Lintao Jia, Angang Yang, Bo Yan","doi":"10.1038/s41423-023-01120-y","DOIUrl":null,"url":null,"abstract":"Despite the tremendous progress of chimeric antigen receptor T (CAR-T) cell therapy in hematological malignancies, their application in solid tumors has been limited largely due to T-cell exhaustion in the tumor microenvironment (TME) and systemic toxicity caused by excessive cytokine release. As a key regulator of the immunosuppressive TME, TGF-β promotes cytokine synthesis via the NF-κB pathway. Here, we coexpressed SMAD7, a suppressor of TGF-β signaling, with a HER2-targeted CAR in engineered T cells. These novel CAR-T cells displayed high cytolytic efficacy and were resistant to TGF-β-triggered exhaustion, which enabled sustained tumoricidal capacity after continuous antigen exposure. Moreover, SMAD7 substantially reduced the production of inflammatory cytokines by antigen-primed CAR-T cells. Mechanistically, SMAD7 downregulated TGF-β receptor I and abrogated the interplay between the TGF-β and NF-κB pathways in CAR-T cells. As a result, these CAR-T cells persistently inhibited tumor growth and promoted the survival of tumor-challenged mice regardless of the hostile tumor microenvironment caused by a high concentration of TGF-β. SMAD7 coexpression also enhanced CAR-T-cell infiltration and persistent activation in patient-derived tumor organoids. Therefore, our study demonstrated the feasibility of SMAD7 coexpression as a novel approach to improve the efficacy and safety of CAR-T-cell therapy for solid tumors.","PeriodicalId":9950,"journal":{"name":"Cellular &Molecular Immunology","volume":null,"pages":null},"PeriodicalIF":21.8000,"publicationDate":"2024-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cellular &Molecular Immunology","FirstCategoryId":"3","ListUrlMain":"https://www.nature.com/articles/s41423-023-01120-y","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

Despite the tremendous progress of chimeric antigen receptor T (CAR-T) cell therapy in hematological malignancies, their application in solid tumors has been limited largely due to T-cell exhaustion in the tumor microenvironment (TME) and systemic toxicity caused by excessive cytokine release. As a key regulator of the immunosuppressive TME, TGF-β promotes cytokine synthesis via the NF-κB pathway. Here, we coexpressed SMAD7, a suppressor of TGF-β signaling, with a HER2-targeted CAR in engineered T cells. These novel CAR-T cells displayed high cytolytic efficacy and were resistant to TGF-β-triggered exhaustion, which enabled sustained tumoricidal capacity after continuous antigen exposure. Moreover, SMAD7 substantially reduced the production of inflammatory cytokines by antigen-primed CAR-T cells. Mechanistically, SMAD7 downregulated TGF-β receptor I and abrogated the interplay between the TGF-β and NF-κB pathways in CAR-T cells. As a result, these CAR-T cells persistently inhibited tumor growth and promoted the survival of tumor-challenged mice regardless of the hostile tumor microenvironment caused by a high concentration of TGF-β. SMAD7 coexpression also enhanced CAR-T-cell infiltration and persistent activation in patient-derived tumor organoids. Therefore, our study demonstrated the feasibility of SMAD7 coexpression as a novel approach to improve the efficacy and safety of CAR-T-cell therapy for solid tumors.

Abstract Image

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
CAR-T 细胞中 SMAD7 的表达提高了治疗实体瘤的持久性和安全性
尽管嵌合抗原受体T(CAR-T)细胞疗法在血液恶性肿瘤中取得了巨大进展,但其在实体瘤中的应用却受到了限制,这主要是由于T细胞在肿瘤微环境(TME)中耗竭以及细胞因子释放过多导致的全身毒性。作为免疫抑制性 TME 的关键调节因子,TGF-β 通过 NF-κB 通路促进细胞因子的合成。在这里,我们在工程 T 细胞中将 TGF-β 信号抑制因子 SMAD7 与 HER2 靶向 CAR 共表达。这些新型CAR-T细胞显示出很高的细胞溶解效力,并能抵抗TGF-β引发的衰竭,从而在持续暴露于抗原后仍能保持杀瘤能力。此外,SMAD7 还大大减少了抗原刺激 CAR-T 细胞产生的炎性细胞因子。从机理上讲,SMAD7 下调了 TGF-β 受体 I,消减了 CAR-T 细胞中 TGF-β 和 NF-κB 通路之间的相互作用。因此,无论高浓度 TGF-β 导致的恶劣肿瘤微环境如何,这些 CAR-T 细胞都能持续抑制肿瘤生长并促进肿瘤小鼠的存活。SMAD7共表达还增强了CAR-T细胞在患者衍生肿瘤组织细胞中的浸润和持续激活。因此,我们的研究证明了SMAD7共表达作为一种新方法来提高CAR-T细胞治疗实体瘤的疗效和安全性的可行性。
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
31.20
自引率
1.20%
发文量
903
审稿时长
1 months
期刊介绍: Cellular & Molecular Immunology, a monthly journal from the Chinese Society of Immunology and the University of Science and Technology of China, serves as a comprehensive platform covering both basic immunology research and clinical applications. The journal publishes a variety of article types, including Articles, Review Articles, Mini Reviews, and Short Communications, focusing on diverse aspects of cellular and molecular immunology.
期刊最新文献
Rack1 regulates B-cell development and function by binding to and stabilizing the transcription factor Pax5 RBM25 is required to restrain inflammation via ACLY RNA splicing-dependent metabolism rewiring Fatty acid metabolism constrains Th9 cell differentiation and antitumor immunity via the modulation of retinoic acid receptor signaling. The emerging roles of B cells in cancer development. Engineered mitochondria exert potent antitumor immunity as a cancer vaccine platform.
×
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