内源性旁观者杀伤机制增强了新型 FAP 特异性 CAR-T 细胞对抗胶质母细胞瘤的活性

IF 4.6 2区 医学 Q2 IMMUNOLOGY Clinical & Translational Immunology Pub Date : 2024-07-05 DOI:10.1002/cti2.1519
Wenbo Yu, Nga TH Truong, Ruhi Polara, Tessa Gargett, Melinda N Tea, Stuart M Pitson, Michaelia P Cockshell, Claudine S Bonder, Lisa M Ebert, Michael P Brown
{"title":"内源性旁观者杀伤机制增强了新型 FAP 特异性 CAR-T 细胞对抗胶质母细胞瘤的活性","authors":"Wenbo Yu,&nbsp;Nga TH Truong,&nbsp;Ruhi Polara,&nbsp;Tessa Gargett,&nbsp;Melinda N Tea,&nbsp;Stuart M Pitson,&nbsp;Michaelia P Cockshell,&nbsp;Claudine S Bonder,&nbsp;Lisa M Ebert,&nbsp;Michael P Brown","doi":"10.1002/cti2.1519","DOIUrl":null,"url":null,"abstract":"<div>\n \n \n <section>\n \n <h3> Objectives</h3>\n \n <p>CAR-T cells are being investigated as a novel immunotherapy for glioblastoma, but clinical success has been limited. We recently described fibroblast activation protein (FAP) as an ideal target antigen for glioblastoma immunotherapy, with expression on both tumor cells and tumor blood vessels. However, CAR-T cells targeting FAP have never been investigated as a therapy for glioblastoma.</p>\n </section>\n \n <section>\n \n <h3> Methods</h3>\n \n <p>We generated a novel FAP targeting CAR with CD3ζ and CD28 signalling domains and tested the resulting CAR-T cells for their lytic activity and cytokine secretion function <i>in vitro</i> (using real-time impedance, flow cytometry, imaging and bead-based cytokine assays), and <i>in vivo</i> (using a xenograft mimicking the natural heterogeneity of human glioblastoma).</p>\n </section>\n \n <section>\n \n <h3> Results</h3>\n \n <p>FAP-CAR-T cells exhibited target specificity against model cell lines and potent cytotoxicity against patient-derived glioma neural stem cells, even when only a subpopulation expressed FAP, indicating a bystander killing mechanism. Using co-culture assays, we confirmed FAP-CAR-T cells mediate bystander killing of antigen-negative tumor cells, but only after activation by FAP-positive target cells. This bystander killing was at least partially mediated by soluble factors and amplified by IL-2 which activated the non-transduced fraction of the CAR-T product. Finally, a low dose of intravenously administered FAP-CAR-T cells controlled, without overt toxicity, the growth of subcutaneous tumors created using a mixture of antigen-negative and antigen-positive glioblastoma cells.</p>\n </section>\n \n <section>\n \n <h3> Conclusions</h3>\n \n <p>Our findings advance FAP as a leading candidate for clinical CAR-T therapy of glioblastoma and highlight under-recognised antigen nonspecific mechanisms that may contribute meaningfully to the antitumor activity of CAR-T cells.</p>\n </section>\n </div>","PeriodicalId":152,"journal":{"name":"Clinical & Translational Immunology","volume":null,"pages":null},"PeriodicalIF":4.6000,"publicationDate":"2024-07-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cti2.1519","citationCount":"0","resultStr":"{\"title\":\"Endogenous bystander killing mechanisms enhance the activity of novel FAP-specific CAR-T cells against glioblastoma\",\"authors\":\"Wenbo Yu,&nbsp;Nga TH Truong,&nbsp;Ruhi Polara,&nbsp;Tessa Gargett,&nbsp;Melinda N Tea,&nbsp;Stuart M Pitson,&nbsp;Michaelia P Cockshell,&nbsp;Claudine S Bonder,&nbsp;Lisa M Ebert,&nbsp;Michael P Brown\",\"doi\":\"10.1002/cti2.1519\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<div>\\n \\n \\n <section>\\n \\n <h3> Objectives</h3>\\n \\n <p>CAR-T cells are being investigated as a novel immunotherapy for glioblastoma, but clinical success has been limited. We recently described fibroblast activation protein (FAP) as an ideal target antigen for glioblastoma immunotherapy, with expression on both tumor cells and tumor blood vessels. However, CAR-T cells targeting FAP have never been investigated as a therapy for glioblastoma.</p>\\n </section>\\n \\n <section>\\n \\n <h3> Methods</h3>\\n \\n <p>We generated a novel FAP targeting CAR with CD3ζ and CD28 signalling domains and tested the resulting CAR-T cells for their lytic activity and cytokine secretion function <i>in vitro</i> (using real-time impedance, flow cytometry, imaging and bead-based cytokine assays), and <i>in vivo</i> (using a xenograft mimicking the natural heterogeneity of human glioblastoma).</p>\\n </section>\\n \\n <section>\\n \\n <h3> Results</h3>\\n \\n <p>FAP-CAR-T cells exhibited target specificity against model cell lines and potent cytotoxicity against patient-derived glioma neural stem cells, even when only a subpopulation expressed FAP, indicating a bystander killing mechanism. Using co-culture assays, we confirmed FAP-CAR-T cells mediate bystander killing of antigen-negative tumor cells, but only after activation by FAP-positive target cells. This bystander killing was at least partially mediated by soluble factors and amplified by IL-2 which activated the non-transduced fraction of the CAR-T product. Finally, a low dose of intravenously administered FAP-CAR-T cells controlled, without overt toxicity, the growth of subcutaneous tumors created using a mixture of antigen-negative and antigen-positive glioblastoma cells.</p>\\n </section>\\n \\n <section>\\n \\n <h3> Conclusions</h3>\\n \\n <p>Our findings advance FAP as a leading candidate for clinical CAR-T therapy of glioblastoma and highlight under-recognised antigen nonspecific mechanisms that may contribute meaningfully to the antitumor activity of CAR-T cells.</p>\\n </section>\\n </div>\",\"PeriodicalId\":152,\"journal\":{\"name\":\"Clinical & Translational Immunology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":4.6000,\"publicationDate\":\"2024-07-05\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://onlinelibrary.wiley.com/doi/epdf/10.1002/cti2.1519\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Clinical & Translational Immunology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://onlinelibrary.wiley.com/doi/10.1002/cti2.1519\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"\",\"PubModel\":\"\",\"JCR\":\"Q2\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Clinical & Translational Immunology","FirstCategoryId":"3","ListUrlMain":"https://onlinelibrary.wiley.com/doi/10.1002/cti2.1519","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q2","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

目的 CAR-T 细胞作为一种治疗胶质母细胞瘤的新型免疫疗法正在接受研究,但临床成功率有限。我们最近描述了成纤维细胞活化蛋白(FAP),它是胶质母细胞瘤免疫疗法的理想靶抗原,在肿瘤细胞和肿瘤血管上都有表达。然而,以 FAP 为靶点的 CAR-T 细胞从未作为胶质母细胞瘤的疗法进行过研究。 方法 我们生成了一种带有 CD3ζ 和 CD28 信号结构域的新型 FAP 靶向 CAR,并在体外(使用实时阻抗、流式细胞仪、成像和基于微珠的细胞因子测定)和体内(使用模拟人类胶质母细胞瘤天然异质性的异种移植)测试了所生成的 CAR-T 细胞的溶解活性和细胞因子分泌功能。 结果 FAP-CAR-T 细胞对模型细胞系具有靶向特异性,对源自患者的胶质瘤神经干细胞具有强大的细胞毒性,即使只有亚群表达 FAP,这表明存在旁观者杀伤机制。通过共培养试验,我们证实了 FAP-CAR-T 细胞对抗原阴性肿瘤细胞的旁观者杀伤作用,但只有在 FAP 阳性靶细胞激活后才能起作用。这种旁观者杀伤作用至少部分由可溶性因子介导,并由 IL-2 激活 CAR-T 产物的非转化部分而放大。最后,静脉注射低剂量的 FAP-CAR-T 细胞可控制使用抗原阴性和抗原阳性胶质母细胞瘤细胞混合物生成的皮下肿瘤的生长,且无明显毒性。 结论 我们的研究结果推动了 FAP 成为胶质母细胞瘤临床 CAR-T 疗法的主要候选者,并强调了未被充分认识的抗原非特异性机制,这种机制可能会对 CAR-T 细胞的抗肿瘤活性做出有意义的贡献。
本文章由计算机程序翻译,如有差异,请以英文原文为准。

摘要图片

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
Endogenous bystander killing mechanisms enhance the activity of novel FAP-specific CAR-T cells against glioblastoma

Objectives

CAR-T cells are being investigated as a novel immunotherapy for glioblastoma, but clinical success has been limited. We recently described fibroblast activation protein (FAP) as an ideal target antigen for glioblastoma immunotherapy, with expression on both tumor cells and tumor blood vessels. However, CAR-T cells targeting FAP have never been investigated as a therapy for glioblastoma.

Methods

We generated a novel FAP targeting CAR with CD3ζ and CD28 signalling domains and tested the resulting CAR-T cells for their lytic activity and cytokine secretion function in vitro (using real-time impedance, flow cytometry, imaging and bead-based cytokine assays), and in vivo (using a xenograft mimicking the natural heterogeneity of human glioblastoma).

Results

FAP-CAR-T cells exhibited target specificity against model cell lines and potent cytotoxicity against patient-derived glioma neural stem cells, even when only a subpopulation expressed FAP, indicating a bystander killing mechanism. Using co-culture assays, we confirmed FAP-CAR-T cells mediate bystander killing of antigen-negative tumor cells, but only after activation by FAP-positive target cells. This bystander killing was at least partially mediated by soluble factors and amplified by IL-2 which activated the non-transduced fraction of the CAR-T product. Finally, a low dose of intravenously administered FAP-CAR-T cells controlled, without overt toxicity, the growth of subcutaneous tumors created using a mixture of antigen-negative and antigen-positive glioblastoma cells.

Conclusions

Our findings advance FAP as a leading candidate for clinical CAR-T therapy of glioblastoma and highlight under-recognised antigen nonspecific mechanisms that may contribute meaningfully to the antitumor activity of CAR-T cells.

求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
Clinical & Translational Immunology
Clinical & Translational Immunology Medicine-Immunology and Allergy
CiteScore
12.00
自引率
1.70%
发文量
77
审稿时长
13 weeks
期刊介绍: Clinical & Translational Immunology is an open access, fully peer-reviewed journal devoted to publishing cutting-edge advances in biomedical research for scientists and physicians. The Journal covers fields including cancer biology, cardiovascular research, gene therapy, immunology, vaccine development and disease pathogenesis and therapy at the earliest phases of investigation.
期刊最新文献
Pulmonary complications post allogeneic haematopoietic stem cell transplant in children Biomarkers to predict and diagnose pulmonary complications in children post haematopoietic stem cell transplant The Omega-6 Lipid pathway shift is associated with neutrophil influx and structural lung damage in early cystic fibrosis lung disease Siglec-7 and Siglec-9 expression in primary triple negative and oestrogen receptor positive breast cancer and in vitro signalling Schistosoma excretory/secretory products: an untapped library of tolerogenic immunotherapeutics against food allergy
×
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