二肽基肽酶4抑制通过促进T细胞浸润使放射治疗增敏。

IF 6.5 2区 医学 Q1 IMMUNOLOGY Oncoimmunology Pub Date : 2023-10-12 eCollection Date: 2023-01-01 DOI:10.1080/2162402X.2023.2268257
Yu Tian, Lingyi Kong, Yan Li, Zhiyun Liao, Xing Cai, Suke Deng, Xiao Yang, Bin Zhang, Yijun Wang, Zhanjie Zhang, Bian Wu, Lu Wen, Fang Huang, Yan Hu, Chao Wan, Yifei Liao, Yajie Sun, Kunyu Yang
{"title":"二肽基肽酶4抑制通过促进T细胞浸润使放射治疗增敏。","authors":"Yu Tian,&nbsp;Lingyi Kong,&nbsp;Yan Li,&nbsp;Zhiyun Liao,&nbsp;Xing Cai,&nbsp;Suke Deng,&nbsp;Xiao Yang,&nbsp;Bin Zhang,&nbsp;Yijun Wang,&nbsp;Zhanjie Zhang,&nbsp;Bian Wu,&nbsp;Lu Wen,&nbsp;Fang Huang,&nbsp;Yan Hu,&nbsp;Chao Wan,&nbsp;Yifei Liao,&nbsp;Yajie Sun,&nbsp;Kunyu Yang","doi":"10.1080/2162402X.2023.2268257","DOIUrl":null,"url":null,"abstract":"<p><p>Radiotherapy could regulate systemic antitumor immunity, while the immune state in the tumor microenvironment (TME) also affects the efficacy of radiotherapy. We have found that higher CD8<sup>+</sup> T cell infiltration is associated with longer overall survival of lung adenocarcinoma and melanoma patients receiving radiotherapy. 8-Gray radiation increased the transcriptional levels of chemokines in tumor cells <i>in vitro</i>. However, it was not sufficient to induce significant lymphocyte infiltration <i>in vivo</i>. Dipeptidyl peptidase 4 (DPP4) has been reported to inactivate chemokines via post-translational truncation. Single-cell sequencing revealed that dendritic cells (DCs) had a higher DPP4 expression among other cells in the TME and upregulated DPP4 expression after radiation. Combining a DPP4 inhibitor with radiotherapy could promote chemokines expression and T cell infiltration in the TME, enhancing the antitumor effect of radiotherapy. Moreover, this therapy further enhanced the therapeutic efficacy of anti-PD-1. In this study, we demonstrated the underlying mechanism of why radiotherapy failed to induce sufficient T cell infiltration and proposed an effective strategy to promote T cell infiltration and sensitize radiotherapy. These findings demonstrate the translational value of DPP4 inhibition as a complementary approach to enhance the efficacy of radiotherapy and the combination of radiotherapy with immunotherapy.</p>","PeriodicalId":48714,"journal":{"name":"Oncoimmunology","volume":null,"pages":null},"PeriodicalIF":6.5000,"publicationDate":"2023-10-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/d1/96/KONI_12_2268257.PMC10578189.pdf","citationCount":"0","resultStr":"{\"title\":\"Dipeptidyl peptidase 4 inhibition sensitizes radiotherapy by promoting T cell infiltration.\",\"authors\":\"Yu Tian,&nbsp;Lingyi Kong,&nbsp;Yan Li,&nbsp;Zhiyun Liao,&nbsp;Xing Cai,&nbsp;Suke Deng,&nbsp;Xiao Yang,&nbsp;Bin Zhang,&nbsp;Yijun Wang,&nbsp;Zhanjie Zhang,&nbsp;Bian Wu,&nbsp;Lu Wen,&nbsp;Fang Huang,&nbsp;Yan Hu,&nbsp;Chao Wan,&nbsp;Yifei Liao,&nbsp;Yajie Sun,&nbsp;Kunyu Yang\",\"doi\":\"10.1080/2162402X.2023.2268257\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"<p><p>Radiotherapy could regulate systemic antitumor immunity, while the immune state in the tumor microenvironment (TME) also affects the efficacy of radiotherapy. We have found that higher CD8<sup>+</sup> T cell infiltration is associated with longer overall survival of lung adenocarcinoma and melanoma patients receiving radiotherapy. 8-Gray radiation increased the transcriptional levels of chemokines in tumor cells <i>in vitro</i>. However, it was not sufficient to induce significant lymphocyte infiltration <i>in vivo</i>. Dipeptidyl peptidase 4 (DPP4) has been reported to inactivate chemokines via post-translational truncation. Single-cell sequencing revealed that dendritic cells (DCs) had a higher DPP4 expression among other cells in the TME and upregulated DPP4 expression after radiation. Combining a DPP4 inhibitor with radiotherapy could promote chemokines expression and T cell infiltration in the TME, enhancing the antitumor effect of radiotherapy. Moreover, this therapy further enhanced the therapeutic efficacy of anti-PD-1. In this study, we demonstrated the underlying mechanism of why radiotherapy failed to induce sufficient T cell infiltration and proposed an effective strategy to promote T cell infiltration and sensitize radiotherapy. These findings demonstrate the translational value of DPP4 inhibition as a complementary approach to enhance the efficacy of radiotherapy and the combination of radiotherapy with immunotherapy.</p>\",\"PeriodicalId\":48714,\"journal\":{\"name\":\"Oncoimmunology\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":6.5000,\"publicationDate\":\"2023-10-12\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://ftp.ncbi.nlm.nih.gov/pub/pmc/oa_pdf/d1/96/KONI_12_2268257.PMC10578189.pdf\",\"citationCount\":\"0\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Oncoimmunology\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1080/2162402X.2023.2268257\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2023/1/1 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.2023.2268257","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2023/1/1 0:00:00","PubModel":"eCollection","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

摘要

放射治疗可以调节全身抗肿瘤免疫,而肿瘤微环境(TME)中的免疫状态也会影响放射治疗的疗效。我们发现,接受放疗的肺腺癌和黑色素瘤患者的CD8+T细胞浸润越高,总生存期越长。8-Gray辐射增加了体外肿瘤细胞中趋化因子的转录水平。然而,它不足以在体内诱导显著的淋巴细胞浸润。据报道,二肽基肽酶4(DPP4)通过翻译后截短使趋化因子失活。单细胞测序显示,在TME中的其他细胞中,树突状细胞(DC)具有更高的DPP4表达,并在辐射后上调DPP4表达。DPP4抑制剂与放疗结合可促进TME中趋化因子的表达和T细胞的浸润,增强放疗的抗肿瘤作用。此外,该疗法进一步增强了抗PD-1的治疗效果。在本研究中,我们证明了放射治疗未能诱导足够的T细胞浸润的潜在机制,并提出了促进T细胞浸润和增敏放射治疗的有效策略。这些发现证明了DPP4抑制作为增强放疗疗效以及放疗与免疫疗法结合的补充方法的转化价值。
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

摘要图片

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
Dipeptidyl peptidase 4 inhibition sensitizes radiotherapy by promoting T cell infiltration.

Radiotherapy could regulate systemic antitumor immunity, while the immune state in the tumor microenvironment (TME) also affects the efficacy of radiotherapy. We have found that higher CD8+ T cell infiltration is associated with longer overall survival of lung adenocarcinoma and melanoma patients receiving radiotherapy. 8-Gray radiation increased the transcriptional levels of chemokines in tumor cells in vitro. However, it was not sufficient to induce significant lymphocyte infiltration in vivo. Dipeptidyl peptidase 4 (DPP4) has been reported to inactivate chemokines via post-translational truncation. Single-cell sequencing revealed that dendritic cells (DCs) had a higher DPP4 expression among other cells in the TME and upregulated DPP4 expression after radiation. Combining a DPP4 inhibitor with radiotherapy could promote chemokines expression and T cell infiltration in the TME, enhancing the antitumor effect of radiotherapy. Moreover, this therapy further enhanced the therapeutic efficacy of anti-PD-1. In this study, we demonstrated the underlying mechanism of why radiotherapy failed to induce sufficient T cell infiltration and proposed an effective strategy to promote T cell infiltration and sensitize radiotherapy. These findings demonstrate the translational value of DPP4 inhibition as a complementary approach to enhance the efficacy of radiotherapy and the combination of radiotherapy with immunotherapy.

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