GPC3-IL7-CCL19-CAR-T primes immune microenvironment reconstitution for hepatocellular carcinoma therapy.

IF 5.3 2区 医学 Q2 CELL BIOLOGY Cell Biology and Toxicology Pub Date : 2023-12-01 Epub Date: 2023-10-19 DOI:10.1007/s10565-023-09821-w
Li-Li Lu, Shu-Xiu Xiao, Zhi-Yuan Lin, Jin-Jin Bai, Wei Li, Zheng-Qing Song, Yu-Hong Zhou, Bin Lu, Wei-Zhong Wu
{"title":"GPC3-IL7-CCL19-CAR-T primes immune microenvironment reconstitution for hepatocellular carcinoma therapy.","authors":"Li-Li Lu, Shu-Xiu Xiao, Zhi-Yuan Lin, Jin-Jin Bai, Wei Li, Zheng-Qing Song, Yu-Hong Zhou, Bin Lu, Wei-Zhong Wu","doi":"10.1007/s10565-023-09821-w","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Chimeric antigen receptor (CAR)-T-cell therapy is a revolutionary treatment that has become a mainstay of advanced cancer treatment. Conventional glypican-3 (GPC3)-CAR-T cells have not produced ideal clinical outcomes in advanced hepatocellular carcinoma (HCC), and the mechanism is unclear. This study aims to investigate the clinical utility of novel GPC3-7-19-CAR-T cells constructed by our team and to explore the mechanisms underlying their antitumor effects.</p><p><strong>Methods: </strong>We engineered a novel GPC3-targeting CAR including an anti-GPC3 scFv, CD3ζ, CD28 and 4-1BB that induces co-expression of IL-7 at a moderate level (500 pg/mL) and CCL19 at a high level (15000 pg /mL) and transduced it into human T cells. In vitro, cell killing efficacy was validated by the xCELLigence RTCA system, LDH nonradioactive cytotoxicity assay and was confirmed in primary HCC organoid models employing a 3D microfluid chip. In vivo, the antitumor capacity was assessed in a humanized NSG mouse xenograft model. Finally, we initiated a phase I clinical trial to evaluate the safety and effect of GPC3-7-19-CAR-T cells in the clinic.</p><p><strong>Results: </strong>GPC3-7-19-CAR-T cells had 1.5-2 times higher killing efficiency than GPC3-CAR-T cells. The tumor formation rates in GPC3-7-19-CAR-T cells treated model were reduced (3/5vs.5/5), and the average tumor volumes were 0.74 cm<sup>3</sup> ± 1.17 vs. 0.34 cm<sup>3</sup> ± 0.25. Of note, increased proportion of CD4<sup>+</sup> T<sub>EM</sub> and CD8<sup>+</sup> T<sub>CM</sub> cells was infiltrated in GPC3-7-19-CAR-T cells group. GPC3-7-19-CAR-T cells obviously reversed the immunosuppressive tumor microenvironment (TME) by reducing polymorphonuclear (PMN)-myeloid-derived suppressor cells (MDSCs) and regulatory T (Treg) cells infiltration and recruiting more dendritic cells (DCs) to HCC xenograft tumor tissues. In one patient with advanced HCC, GPC3-7-19-CAR-T-cell treatment resulted in tumor reduction 56 days after intravenous infusion.</p><p><strong>Conclusions: </strong>In conclusion, GPC3-7-19-CAR-T cells achieved antitumor effects superior to those of conventional GPC3-CAR-T cells by reconstructing the TME induced by the dominant CD4<sup>+</sup> T<sub>EM</sub> and CD8<sup>+</sup> T<sub>CM</sub> cell subsets. Most importantly, GPC3-7-19-CAR-T cells exhibited good safety and antitumor efficacy in HCC patients in the clinic. ► Novel GPC3-7-19-CAR-T cells designed with mediate level of IL-7 secretion and high level of CCL19 secretion, which could recruit more mature DCs to assist killing on GPC3<sup>+</sup>HCCs. ►DC cells recruited by CCL19 could interact with CD4<sup>+</sup> T cells and promote the differentiation of CD4<sup>+</sup>T<sub>EFF</sub> cells into CD4<sup>+</sup>T<sub>EM</sub> and CD8<sup>+</sup>T<sub>CM</sub> subsets, leading a better anti-tumor effect on GPC3<sup>+</sup>HCCs. ►Compared with conventional GPC3-CAR-T, GPC3-7-CCL19-CAR-T cells could reverse tumor immunosuppressive microenvironment by reducing PMN-MDSC and Treg cell infiltration.</p>","PeriodicalId":9672,"journal":{"name":"Cell Biology and Toxicology","volume":null,"pages":null},"PeriodicalIF":5.3000,"publicationDate":"2023-12-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Cell Biology and Toxicology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1007/s10565-023-09821-w","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/10/19 0:00:00","PubModel":"Epub","JCR":"Q2","JCRName":"CELL BIOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Chimeric antigen receptor (CAR)-T-cell therapy is a revolutionary treatment that has become a mainstay of advanced cancer treatment. Conventional glypican-3 (GPC3)-CAR-T cells have not produced ideal clinical outcomes in advanced hepatocellular carcinoma (HCC), and the mechanism is unclear. This study aims to investigate the clinical utility of novel GPC3-7-19-CAR-T cells constructed by our team and to explore the mechanisms underlying their antitumor effects.

Methods: We engineered a novel GPC3-targeting CAR including an anti-GPC3 scFv, CD3ζ, CD28 and 4-1BB that induces co-expression of IL-7 at a moderate level (500 pg/mL) and CCL19 at a high level (15000 pg /mL) and transduced it into human T cells. In vitro, cell killing efficacy was validated by the xCELLigence RTCA system, LDH nonradioactive cytotoxicity assay and was confirmed in primary HCC organoid models employing a 3D microfluid chip. In vivo, the antitumor capacity was assessed in a humanized NSG mouse xenograft model. Finally, we initiated a phase I clinical trial to evaluate the safety and effect of GPC3-7-19-CAR-T cells in the clinic.

Results: GPC3-7-19-CAR-T cells had 1.5-2 times higher killing efficiency than GPC3-CAR-T cells. The tumor formation rates in GPC3-7-19-CAR-T cells treated model were reduced (3/5vs.5/5), and the average tumor volumes were 0.74 cm3 ± 1.17 vs. 0.34 cm3 ± 0.25. Of note, increased proportion of CD4+ TEM and CD8+ TCM cells was infiltrated in GPC3-7-19-CAR-T cells group. GPC3-7-19-CAR-T cells obviously reversed the immunosuppressive tumor microenvironment (TME) by reducing polymorphonuclear (PMN)-myeloid-derived suppressor cells (MDSCs) and regulatory T (Treg) cells infiltration and recruiting more dendritic cells (DCs) to HCC xenograft tumor tissues. In one patient with advanced HCC, GPC3-7-19-CAR-T-cell treatment resulted in tumor reduction 56 days after intravenous infusion.

Conclusions: In conclusion, GPC3-7-19-CAR-T cells achieved antitumor effects superior to those of conventional GPC3-CAR-T cells by reconstructing the TME induced by the dominant CD4+ TEM and CD8+ TCM cell subsets. Most importantly, GPC3-7-19-CAR-T cells exhibited good safety and antitumor efficacy in HCC patients in the clinic. ► Novel GPC3-7-19-CAR-T cells designed with mediate level of IL-7 secretion and high level of CCL19 secretion, which could recruit more mature DCs to assist killing on GPC3+HCCs. ►DC cells recruited by CCL19 could interact with CD4+ T cells and promote the differentiation of CD4+TEFF cells into CD4+TEM and CD8+TCM subsets, leading a better anti-tumor effect on GPC3+HCCs. ►Compared with conventional GPC3-CAR-T, GPC3-7-CCL19-CAR-T cells could reverse tumor immunosuppressive microenvironment by reducing PMN-MDSC and Treg cell infiltration.

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
GPC3-IL7-CL19-CAR-T为肝细胞癌治疗启动免疫微环境重建。
背景:嵌合抗原受体(CAR)-T细胞疗法是一种革命性的治疗方法,已成为晚期癌症治疗的支柱。传统的糖蛋白-3(GPC3)-CAR-T细胞在晚期肝细胞癌(HCC)中没有产生理想的临床结果,其机制尚不清楚。本研究旨在研究我们团队构建的新型GPC3-7-CAR-T细胞的临床应用,并探索其抗肿瘤作用的机制。方法:我们设计了一种新的GPC3靶向CAR,包括抗GPC3 scFv、CD3ζ、CD28和4-1BB,其诱导中等水平(500pg/mL)的IL-7和高水平(15000pg/mL的CCL19的共表达,并将其转导到人T细胞中。在体外,通过xCELLigence RTCA系统、LDH非放射性细胞毒性测定验证了细胞杀伤效果,并在使用3D微流体芯片的原发性HCC类器官模型中得到了证实。在体内,在人源化NSG小鼠异种移植物模型中评估抗肿瘤能力。最后,我们启动了一项I期临床试验,以评估GPC3-7-7-CAR-T细胞在临床上的安全性和效果。结果:GPC37-19-CAR-T细胞的杀伤效率是GPC3-CAR-T的1.5~2倍。GPC3-7-7-CAR-T细胞处理模型中的肿瘤形成率降低(3/5vs.5/5),平均肿瘤体积为0.74cm3 ± 1.17与0.34 cm3 ± 0.25。值得注意的是,在GPC3-7-7-CAR-T细胞组中,CD4+TEM和CD8+TCM细胞的比例增加。GPC3-7-7-CAR-T细胞通过减少多形核细胞(PMN)-骨髓源性抑制细胞(MDSCs)和调节性T细胞(Treg)的浸润,并募集更多的树突状细胞(DC)到HCC异种移植肿瘤组织中,明显逆转了免疫抑制肿瘤微环境(TME)。在一名晚期HCC患者中,静脉输注后56天,GPC3-7-7-CAR-T细胞治疗导致肿瘤减少。结论:GPC3-7-7-CAR-T细胞通过重建由显性CD4+TEM和CD8+TCM细胞亚群诱导的TME,获得了优于常规GPC3-CAR-T的抗肿瘤效果。最重要的是,GPC3-7-7-CAR-T细胞在临床上对HCC患者表现出良好的安全性和抗肿瘤疗效。► 新型GPC3-7-7-CAR-T细胞设计具有中等水平的IL-7分泌和高水平的CCL19分泌,可以募集更成熟的DC来帮助杀死GPC3+HCC。►CCL19募集的DC细胞可与CD4+T细胞相互作用,促进CD4+TEFF细胞分化为CD4+TEM和CD8+TCM亚群,对GPC3+HCC具有较好的抗肿瘤作用。►与传统的GPC3-CAR-T相比,GPC3-7-CL19-CAR-T细胞可以通过减少PMN-MDSC和Treg细胞的浸润来逆转肿瘤免疫抑制微环境。
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
Cell Biology and Toxicology
Cell Biology and Toxicology 生物-毒理学
CiteScore
9.90
自引率
4.90%
发文量
101
审稿时长
>12 weeks
期刊介绍: Cell Biology and Toxicology (CBT) is an international journal focused on clinical and translational research with an emphasis on molecular and cell biology, genetic and epigenetic heterogeneity, drug discovery and development, and molecular pharmacology and toxicology. CBT has a disease-specific scope prioritizing publications on gene and protein-based regulation, intracellular signaling pathway dysfunction, cell type-specific function, and systems in biomedicine in drug discovery and development. CBT publishes original articles with outstanding, innovative and significant findings, important reviews on recent research advances and issues of high current interest, opinion articles of leading edge science, and rapid communication or reports, on molecular mechanisms and therapies in diseases.
期刊最新文献
G-CSFR-induced leukocyte transendothelial migration during the inflammatory response is regulated by the ICAM1-PKCa axis: based on multiomics integration analysis. PRDM1 promotes nucleus pulposus cell pyroptosis leading to intervertebral disc degeneration via activating CASP1 transcription. Three bioactive compounds from Huangqin decoction ameliorate Irinotecan-induced diarrhea via dual-targeting of Escherichia coli and bacterial β-glucuronidase. Mechanisms of HIF1A-mediated immune evasion in gastric cancer and the impact on therapy resistance. EIF4A3-mediated oncogenic circRNA hsa_circ_0001165 advances esophageal squamous cell carcinoma progression through the miR-381-3p/TNS3 pathway.
×
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