Intrinsic ADRB2 inhibition improves CAR-T cell therapy efficacy against prostate cancer.

IF 12.1 1区 医学 Q1 BIOTECHNOLOGY & APPLIED MICROBIOLOGY Molecular Therapy Pub Date : 2024-09-02 DOI:10.1016/j.ymthe.2024.08.028
Iqra Ajmal, Muhammad Asad Farooq, Yixin Duan, Jie Yao, Yaoxin Gao, Xinhui Hui, Yujia Ge, Yiran Chen, Yaojun Ren, Bingtan Du, Wenzheng Jiang
{"title":"Intrinsic ADRB2 inhibition improves CAR-T cell therapy efficacy against prostate cancer.","authors":"Iqra Ajmal, Muhammad Asad Farooq, Yixin Duan, Jie Yao, Yaoxin Gao, Xinhui Hui, Yujia Ge, Yiran Chen, Yaojun Ren, Bingtan Du, Wenzheng Jiang","doi":"10.1016/j.ymthe.2024.08.028","DOIUrl":null,"url":null,"abstract":"<p><p>Chimeric antigen receptor (CAR)-T cell therapy has shown limited success in patients with solid tumors. Recent in vitro and in vivo data have shown that adrenoceptor beta-2 (ADRB2) is a novel checkpoint receptor that inhibits T cell-mediated anti-tumor responses. To inhibit ADRB2-mediated inhibitory signaling, we downregulated ADRB2 in CAR-T (shβ<sub>2</sub>-CAR-T) cells via RNA interference, assessed different parameters, and compared them with conventional second-generation CAR-T cells. ADRB2 knockdown CAR-T cells exhibited enhanced cytotoxicity against prostate cancer cell lines in vitro, by increasing CD69, CD107a, GzmB, IFN-γ, T-bet, and GLUT-1. Additionally, ADRB2 deficiency led to improved proliferation, increased CD8/CD4 T cell ratio, and decreased apoptosis in CAR-T cells. shβ<sub>2</sub>-CAR-T cells expressed more Bcl-2 and led to the generation of more significant proportions of T central memory cells. Finally, the ZAP-70/NF-κB signaling axis was shown to be responsible for the improved functions of novel CAR-T cells. In tumor-bearing mice, shβ<sub>2</sub>-CAR-T cells performed better than conventional CAR-T cells in eradicating prostate tumors. The study provides the basis for future clinical and translational CAR-T cell research to focus on adrenergic stress-mediated challenges in the tumor microenvironment of stressed tumors.</p>","PeriodicalId":19020,"journal":{"name":"Molecular Therapy","volume":null,"pages":null},"PeriodicalIF":12.1000,"publicationDate":"2024-09-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Molecular Therapy","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1016/j.ymthe.2024.08.028","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"BIOTECHNOLOGY & APPLIED MICROBIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Chimeric antigen receptor (CAR)-T cell therapy has shown limited success in patients with solid tumors. Recent in vitro and in vivo data have shown that adrenoceptor beta-2 (ADRB2) is a novel checkpoint receptor that inhibits T cell-mediated anti-tumor responses. To inhibit ADRB2-mediated inhibitory signaling, we downregulated ADRB2 in CAR-T (shβ2-CAR-T) cells via RNA interference, assessed different parameters, and compared them with conventional second-generation CAR-T cells. ADRB2 knockdown CAR-T cells exhibited enhanced cytotoxicity against prostate cancer cell lines in vitro, by increasing CD69, CD107a, GzmB, IFN-γ, T-bet, and GLUT-1. Additionally, ADRB2 deficiency led to improved proliferation, increased CD8/CD4 T cell ratio, and decreased apoptosis in CAR-T cells. shβ2-CAR-T cells expressed more Bcl-2 and led to the generation of more significant proportions of T central memory cells. Finally, the ZAP-70/NF-κB signaling axis was shown to be responsible for the improved functions of novel CAR-T cells. In tumor-bearing mice, shβ2-CAR-T cells performed better than conventional CAR-T cells in eradicating prostate tumors. The study provides the basis for future clinical and translational CAR-T cell research to focus on adrenergic stress-mediated challenges in the tumor microenvironment of stressed tumors.

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
内在 ADRB2 抑制可提高 CAR-T 细胞疗法对前列腺癌的疗效。
嵌合抗原受体(CAR)-T 细胞疗法在实体瘤患者中的疗效有限。最近的体外和体内数据显示,肾上腺素受体β-2(ADRB2)是一种新型检查点受体,能抑制T细胞介导的抗肿瘤反应。为了抑制ADRB2介导的抑制信号传导,我们通过RNA干扰下调了CAR-T(shβ2-CAR-T)细胞中的ADRB2,评估了不同的参数,并与传统的第二代CAR-T细胞进行了比较。通过增加CD69、CD107a、GzmB、IFN-γ、T-bet和GLUT-1,体外敲除ADRB2的CAR-T细胞对前列腺癌细胞株表现出更强的细胞毒性。此外,ADRB2 的缺乏导致 CAR-T 细胞增殖改善、CD8/CD4 T 细胞比率增加和凋亡减少。最后,ZAP-70/NF-κB 信号轴被证明是新型 CAR-T 细胞功能改善的原因。在肿瘤小鼠体内,shβ2-CAR-T 细胞在根除前列腺肿瘤方面的表现优于传统 CAR-T 细胞。这项研究为未来的临床和转化CAR-T细胞研究奠定了基础,研究重点是受压肿瘤微环境中肾上腺素能应激介导的挑战。
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
Molecular Therapy
Molecular Therapy 医学-生物工程与应用微生物
CiteScore
19.20
自引率
3.20%
发文量
357
审稿时长
3 months
期刊介绍: Molecular Therapy is the leading journal for research in gene transfer, vector development, stem cell manipulation, and therapeutic interventions. It covers a broad spectrum of topics including genetic and acquired disease correction, vaccine development, pre-clinical validation, safety/efficacy studies, and clinical trials. With a focus on advancing genetics, medicine, and biotechnology, Molecular Therapy publishes peer-reviewed research, reviews, and commentaries to showcase the latest advancements in the field. With an impressive impact factor of 12.4 in 2022, it continues to attract top-tier contributions.
期刊最新文献
Engineering a solution for allogeneic CAR-T rejection. Targeting Rap1b signaling cascades with CDNF: Modulating Platelet Activation, Regulating Plasma Oxylipins and Mitigating Reperfusion Injury in stroke. A CD25×TIGIT bispecific antibody induces anti-tumor activity through selective intratumoral Treg cell depletion. A chimeric anti-inflammatory and anti-vascularization immunomodulator prevents high-risk corneal transplantation rejection via ex vivo gene therapy. Case study of CD19-directed chimeric antigen receptor T-cell therapy in a subject with immune-mediate necrotizing myopathy treated in the RESET-Myositis™ phase I/II trial.
×
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