TIGIT 或 PD-1 基因缺失对黑色素瘤特异性 T 淋巴细胞的不同影响。

IF 6.5 2区 医学 Q1 IMMUNOLOGY Oncoimmunology Pub Date : 2024-07-08 eCollection Date: 2024-01-01 DOI:10.1080/2162402X.2024.2376782
Gwenann Cadiou, Tiffany Beauvais, Lucine Marotte, Sylvia Lambot, Cécile Deleine, Caroline Vignes, Malika Gantier, Melanie Hussong, Samuel Rulli, Anne Jarry, Sylvain Simon, Bernard Malissen, Nathalie Labarriere
{"title":"TIGIT 或 PD-1 基因缺失对黑色素瘤特异性 T 淋巴细胞的不同影响。","authors":"Gwenann Cadiou, Tiffany Beauvais, Lucine Marotte, Sylvia Lambot, Cécile Deleine, Caroline Vignes, Malika Gantier, Melanie Hussong, Samuel Rulli, Anne Jarry, Sylvain Simon, Bernard Malissen, Nathalie Labarriere","doi":"10.1080/2162402X.2024.2376782","DOIUrl":null,"url":null,"abstract":"<p><p>Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of <i>PDCD1</i> or <i>TIGIT</i> genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1<sup>KO</sup> T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGIT<sup>KO</sup> T-cells. Functional analyses showed that PD-1<sup>KO</sup> and TIGIT<sup>KO</sup> T-cells displayed superior antitumor reactivity than their wild-type counterpart <i>in vitro</i> and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGIT<sup>KO</sup> T-cells were more effective at inhibiting tumor cell proliferation <i>in vivo</i>, and persist longer within tumors than PD-1<sup>KO</sup> T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic strategies.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5000,"publicationDate":"2024-07-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11232637/pdf/","citationCount":"0","resultStr":"{\"title\":\"Differential impact of genetic deletion of TIGIT or PD-1 on melanoma-specific T-lymphocytes.\",\"authors\":\"Gwenann Cadiou, Tiffany Beauvais, Lucine Marotte, Sylvia Lambot, Cécile Deleine, Caroline Vignes, Malika Gantier, Melanie Hussong, Samuel Rulli, Anne Jarry, Sylvain Simon, Bernard Malissen, Nathalie Labarriere\",\"doi\":\"10.1080/2162402X.2024.2376782\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of <i>PDCD1</i> or <i>TIGIT</i> genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1<sup>KO</sup> T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGIT<sup>KO</sup> T-cells. Functional analyses showed that PD-1<sup>KO</sup> and TIGIT<sup>KO</sup> T-cells displayed superior antitumor reactivity than their wild-type counterpart <i>in vitro</i> and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGIT<sup>KO</sup> T-cells were more effective at inhibiting tumor cell proliferation <i>in vivo</i>, and persist longer within tumors than PD-1<sup>KO</sup> T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic strategies.</p>\",\"PeriodicalId\":48714,\"journal\":{\"name\":\"Oncoimmunology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":6.5000,\"publicationDate\":\"2024-07-08\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11232637/pdf/\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Oncoimmunology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1080/2162402X.2024.2376782\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2024/1/1 0:00:00\",\"PubModel\":\"eCollection\",\"JCR\":\"Q1\",\"JCRName\":\"IMMUNOLOGY\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Oncoimmunology","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1080/2162402X.2024.2376782","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2024/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

免疫检查点(IC)阻断和肿瘤特异性 T 细胞(ACT)的采纳性转移是治疗转移性黑色素瘤的两种主要策略。二者结合可在抑制性肿瘤微环境中增强T细胞活化,但全身注射IC阻断剂带来的自身免疫不良反应在这一策略中依然存在。对肿瘤有反应的 T 细胞表达 IC 缺陷的 ACT 可解决这一问题。为此,PD-1 和 TIGIT 似乎是相关的候选者,因为它们在高肿瘤反应性淋巴细胞上的共同表达限制了它们在肿瘤微环境中的疗效。我们的研究比较了 PDCD1 或 TIGIT 基因缺失对黑色素瘤特异性 T 淋巴细胞抗肿瘤特性和 T 细胞适应性的影响。转录组分析表明,PD-1KO T细胞中细胞周期相关基因下调,这与生物学观察结果一致,而TIGITKO T细胞中增殖途径得以保留。功能分析显示,在体外和使用免疫缺陷小鼠的临床前黑色素瘤模型中,PD-1KO 和 TIGITKO T 细胞的抗肿瘤反应性优于野生型细胞。有趣的是,与 PD-1KO T 细胞相比,TIGITKO T 细胞似乎能更有效地抑制体内肿瘤细胞的增殖,在肿瘤内存活的时间也更长,这与 TIGIT 基因缺失对 T 细胞适应性没有影响是一致的。综上所述,这些结果表明,黑色素瘤特异性T细胞的TIGIT缺失比PD-1缺失更有吸引力,是未来免疫治疗策略的一种选择。
本文章由计算机程序翻译,如有差异,请以英文原文为准。
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
Differential impact of genetic deletion of TIGIT or PD-1 on melanoma-specific T-lymphocytes.

Immune checkpoint (IC) blockade and adoptive transfer of tumor-specific T-cells (ACT) are two major strategies to treat metastatic melanoma. Their combination can potentiate T-cell activation in the suppressive tumor microenvironment, but the autoimmune adverse effects associated with systemic injection of IC blockers persist with this strategy. ACT of tumor-reactive T-cells defective for IC expression would overcome this issue. For this purpose, PD-1 and TIGIT appear to be relevant candidates, because their co-expression on highly tumor-reactive lymphocytes limits their therapeutic efficacy within the tumor microenvironme,nt. Our study compares the consequences of PDCD1 or TIGIT genetic deletion on anti-tumor properties and T-cell fitness of melanoma-specific T lymphocytes. Transcriptomic analyses revealed down-regulation of cell cycle-related genes in PD-1KO T-cells, consistent with biological observations, whereas proliferative pathways were preserved in TIGITKO T-cells. Functional analyses showed that PD-1KO and TIGITKO T-cells displayed superior antitumor reactivity than their wild-type counterpart in vitro and in a preclinical melanoma model using immunodeficient mice. Interestingly, it appears that TIGITKO T-cells were more effective at inhibiting tumor cell proliferation in vivo, and persist longer within tumors than PD-1KO T-cells, consistent with the absence of impact of TIGIT deletion on T-cell fitness. Taken together, these results suggest that TIGIT deletion, over PD-1 deletion, in melanoma-specific T-cells is a compelling option for future immunotherapeutic strategies.

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
Oncoimmunology
Oncoimmunology ONCOLOGYIMMUNOLOGY-IMMUNOLOGY
CiteScore
12.50
自引率
2.80%
发文量
276
审稿时长
24 weeks
期刊介绍: OncoImmunology is a dynamic, high-profile, open access journal that comprehensively covers tumor immunology and immunotherapy. As cancer immunotherapy advances, OncoImmunology is committed to publishing top-tier research encompassing all facets of basic and applied tumor immunology. The journal covers a wide range of topics, including: -Basic and translational studies in immunology of both solid and hematological malignancies -Inflammation, innate and acquired immune responses against cancer -Mechanisms of cancer immunoediting and immune evasion -Modern immunotherapies, including immunomodulators, immune checkpoint inhibitors, T-cell, NK-cell, and macrophage engagers, and CAR T cells -Immunological effects of conventional anticancer therapies.
期刊最新文献
High-grade serous ovarian cancer development and anti-PD-1 resistance is driven by IRE1α activity in neutrophils. Clinical prognosticators and targets in the immune microenvironment of intrahepatic cholangiocarcinoma. Enhancing T-cell recruitment in renal cell carcinoma with cytokine-armed adenoviruses. Immune priming and induction of tertiary lymphoid structures in a cord-blood humanized mouse model of gastrointestinal stromal tumor. CD4+ tumor-infiltrating lymphocytes secreting T cell-engagers induce regression of autologous patient-derived non-small cell lung cancer xenografts.
×
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