Depletion of conventional CD4+ T cells is required for robust priming and dissemination of tumor antigen-specific CD8+ T cells in the setting of anti-CD4 therapy.

IF 10.3 1区 医学 Q1 IMMUNOLOGY Journal for Immunotherapy of Cancer Pub Date : 2024-11-09 DOI:10.1136/jitc-2024-010170
Delaney E Ramirez, Christo P C Dragnev, Tyler G Searles, Nathaniel Spicer, Tiffany Chen, J Louise Lines, Aaron R Hawkes, Wilson L Davis, Asmaa Mohamed, Keisuke Shirai, Joseph D Phillips, Pamela C Rosato, Yina H Huang, Mary Jo Turk
{"title":"Depletion of conventional CD4<sup>+</sup> T cells is required for robust priming and dissemination of tumor antigen-specific CD8<sup>+</sup> T cells in the setting of anti-CD4 therapy.","authors":"Delaney E Ramirez, Christo P C Dragnev, Tyler G Searles, Nathaniel Spicer, Tiffany Chen, J Louise Lines, Aaron R Hawkes, Wilson L Davis, Asmaa Mohamed, Keisuke Shirai, Joseph D Phillips, Pamela C Rosato, Yina H Huang, Mary Jo Turk","doi":"10.1136/jitc-2024-010170","DOIUrl":null,"url":null,"abstract":"<p><strong>Background: </strong>Overcoming immune suppression is a major barrier to eliciting potent CD8<sup>+</sup> T cell responses against cancer. Treatment with anti-CD4 monoclonal antibody is an effective means for eliminating CD4<sup>+</sup>Foxp3<sup>+</sup> regulatory (Treg) cells in preclinical models and has also demonstrated efficacy in early clinical trials. However, the underlying basis for treatment efficacy, more specifically the implications of codepleting other CD4-expressing cell compartments in tumor-bearing hosts, is not well understood.</p><p><strong>Methods: </strong>Tumor-bearing mice were treated with anti-CD4 versus other therapies that preserve helper T cell function, and the priming, tissue distribution, and maintenance of tumor antigen-specific CD8 T cells were assessed. Antibody blockade and transgenic mouse models were used to determine the mechanisms of CD8 T cell priming. Single-cell RNA-sequencing (scRNAseq) was used to further characterize CD8 T cells that are primed by anti-CD4 therapy and to identify immunosuppressive CD4 T cell subsets in human melanoma following immune checkpoint blockade (ICB).</p><p><strong>Results: </strong>Comparing anti-CD4 to dual ICB therapy, we show that anti-CD4 facilitates more robust priming of TCF-1<sup>+</sup>, IL-2-producing, tumor-specific CD8<sup>+</sup> T cells that disseminate to tissues and form memory. By decoupling priming from homeostatic proliferation and associated cytokines, we find that anti-CD4 functions independently of creating homeostatic space for CD8<sup>+</sup> T cells. We also show that depletion of CD4-expressing antigen-presenting cell subsets is not required for anti-CD4 efficacy. Instead, robust tumor-specific CD8<sup>+</sup> T cell priming and memory generation required the removal of total antigen-specific CD4<sup>+</sup> T cells, including both Tregs and CD4<sup>+</sup> Foxp3-negative conventional (Tconv) cells. In particular, the elimination of CD4<sup>+</sup> Tconv cells was necessary for the accumulation and maturation of conventional type-1 dendritic cells in tumor-draining LNs, which were required for CD8<sup>+</sup> T cell priming. Accordingly, anti-CD4 treatment restored CD8<sup>+</sup> T cell responses in mice cotreated with dual ICB. scRNAseq of melanoma tumors from patients who received ICB revealed the presence of Tr1 and Treg subsets, as well as CD4<sup>+</sup> Tconv subsets that lacked clear transcriptional evidence of helper differentiation.</p><p><strong>Conclusions: </strong>These findings underscore the underappreciated benefit of depleting CD4<sup>+</sup> Tconv cells to promote systemic primary and memory CD8<sup>+</sup> T cell responses against cancer.</p>","PeriodicalId":14820,"journal":{"name":"Journal for Immunotherapy of Cancer","volume":"12 11","pages":""},"PeriodicalIF":10.3000,"publicationDate":"2024-11-09","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11552015/pdf/","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal for Immunotherapy of Cancer","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1136/jitc-2024-010170","RegionNum":1,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"Q1","JCRName":"IMMUNOLOGY","Score":null,"Total":0}
引用次数: 0

Abstract

Background: Overcoming immune suppression is a major barrier to eliciting potent CD8+ T cell responses against cancer. Treatment with anti-CD4 monoclonal antibody is an effective means for eliminating CD4+Foxp3+ regulatory (Treg) cells in preclinical models and has also demonstrated efficacy in early clinical trials. However, the underlying basis for treatment efficacy, more specifically the implications of codepleting other CD4-expressing cell compartments in tumor-bearing hosts, is not well understood.

Methods: Tumor-bearing mice were treated with anti-CD4 versus other therapies that preserve helper T cell function, and the priming, tissue distribution, and maintenance of tumor antigen-specific CD8 T cells were assessed. Antibody blockade and transgenic mouse models were used to determine the mechanisms of CD8 T cell priming. Single-cell RNA-sequencing (scRNAseq) was used to further characterize CD8 T cells that are primed by anti-CD4 therapy and to identify immunosuppressive CD4 T cell subsets in human melanoma following immune checkpoint blockade (ICB).

Results: Comparing anti-CD4 to dual ICB therapy, we show that anti-CD4 facilitates more robust priming of TCF-1+, IL-2-producing, tumor-specific CD8+ T cells that disseminate to tissues and form memory. By decoupling priming from homeostatic proliferation and associated cytokines, we find that anti-CD4 functions independently of creating homeostatic space for CD8+ T cells. We also show that depletion of CD4-expressing antigen-presenting cell subsets is not required for anti-CD4 efficacy. Instead, robust tumor-specific CD8+ T cell priming and memory generation required the removal of total antigen-specific CD4+ T cells, including both Tregs and CD4+ Foxp3-negative conventional (Tconv) cells. In particular, the elimination of CD4+ Tconv cells was necessary for the accumulation and maturation of conventional type-1 dendritic cells in tumor-draining LNs, which were required for CD8+ T cell priming. Accordingly, anti-CD4 treatment restored CD8+ T cell responses in mice cotreated with dual ICB. scRNAseq of melanoma tumors from patients who received ICB revealed the presence of Tr1 and Treg subsets, as well as CD4+ Tconv subsets that lacked clear transcriptional evidence of helper differentiation.

Conclusions: These findings underscore the underappreciated benefit of depleting CD4+ Tconv cells to promote systemic primary and memory CD8+ T cell responses against cancer.

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
在抗 CD4 治疗中,常规 CD4+ T 细胞的耗竭是肿瘤抗原特异性 CD8+ T 细胞强效启动和传播的必要条件。
背景:克服免疫抑制是激发强效 CD8+ T 细胞抗癌反应的主要障碍。在临床前模型中,使用抗 CD4 单克隆抗体治疗是消除 CD4+Foxp3+ 调节(Treg)细胞的有效方法,在早期临床试验中也显示出了疗效。然而,治疗效果的根本原因,更具体地说是在肿瘤宿主体内清除其他CD4表达细胞的影响,目前还不十分清楚:方法:用抗 CD4 和其他保留辅助性 T 细胞功能的疗法治疗肿瘤小鼠,并评估肿瘤抗原特异性 CD8 T 细胞的启动、组织分布和维持情况。抗体阻断和转基因小鼠模型被用来确定CD8 T细胞的启动机制。单细胞RNA测序(scRNAseq)被用来进一步描述被抗CD4疗法激活的CD8 T细胞的特征,并确定免疫检查点阻断(ICB)后人类黑色素瘤中的免疫抑制性CD4 T细胞亚群:结果:通过比较抗CD4疗法和双重ICB疗法,我们发现抗CD4疗法能促进TCF-1+、IL-2生成、肿瘤特异性CD8+T细胞的更强引诱,这些细胞会扩散到组织中并形成记忆。通过将诱导与同源增殖及相关细胞因子分离,我们发现抗 CD4 的功能与为 CD8+ T 细胞创造同源空间无关。我们还发现,抗 CD4 的疗效并不需要消耗表达 CD4 的抗原递呈细胞亚群。相反,强大的肿瘤特异性 CD8+ T 细胞启动和记忆生成需要清除全部抗原特异性 CD4+ T 细胞,包括 Tregs 和 CD4+ Foxp3 阴性常规(Tconv)细胞。特别是,CD4+ Tconv 细胞的清除是肿瘤引流 LN 中常规 1 型树突状细胞积累和成熟的必要条件,而常规 1 型树突状细胞是 CD8+ T 细胞引物的必要条件。对接受过 ICB 治疗的患者的黑色素瘤肿瘤进行 scRNA 序列分析后发现,存在 Tr1 和 Treg 亚群以及 CD4+ Tconv 亚群,但这些亚群缺乏辅助分化的明确转录证据:这些发现强调了消耗 CD4+ Tconv 细胞以促进全身原发性和记忆性 CD8+ T 细胞抗癌反应的益处,但这一益处未得到充分重视。
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
Journal for Immunotherapy of Cancer
Journal for Immunotherapy of Cancer Biochemistry, Genetics and Molecular Biology-Molecular Medicine
CiteScore
17.70
自引率
4.60%
发文量
522
审稿时长
18 weeks
期刊介绍: The Journal for ImmunoTherapy of Cancer (JITC) is a peer-reviewed publication that promotes scientific exchange and deepens knowledge in the constantly evolving fields of tumor immunology and cancer immunotherapy. With an open access format, JITC encourages widespread access to its findings. The journal covers a wide range of topics, spanning from basic science to translational and clinical research. Key areas of interest include tumor-host interactions, the intricate tumor microenvironment, animal models, the identification of predictive and prognostic immune biomarkers, groundbreaking pharmaceutical and cellular therapies, innovative vaccines, combination immune-based treatments, and the study of immune-related toxicity.
期刊最新文献
Depletion of myeloid-derived suppressor cells sensitizes murine multiple myeloma to PD-1 checkpoint inhibitors. Early treatment discontinuation in patients with deficient mismatch repair or microsatellite instability high metastatic colorectal cancer receiving immune checkpoint inhibitors. Effect of radiotherapy exposure on fruquintinib plus sintilimab treatment in refractory microsatellite stable metastatic colorectal cancer: a prospective observation study. SIGLEC11 promotes M2 macrophage polarization through AKT-mTOR signaling and facilitates the progression of gastric cancer. Spatial and single-cell transcriptomics reveal cellular heterogeneity and a novel cancer-promoting Treg cell subset in human clear-cell renal cell carcinoma.
×
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