947 The activation states of tumor-resident type 2 dendritic cells impact the strength of ovarian cancer immune responses

Fiona Chatterjee, Vincent Butty, Emi A Lutz, K Dane Wittrup, Stefani Spranger
{"title":"947 The activation states of tumor-resident type 2 dendritic cells impact the strength of ovarian cancer immune responses","authors":"Fiona Chatterjee, Vincent Butty, Emi A Lutz, K Dane Wittrup, Stefani Spranger","doi":"10.1136/jitc-2023-sitc2023.0947","DOIUrl":null,"url":null,"abstract":"<h3>Background</h3> Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States.<sup>1</sup> To date, checkpoint blockade therapy (CBT) has failed to be effective in ovarian cancer.<sup>2</sup> CBT efficacy typically requires a strong pre-existing tumor-specific T cell response.<sup>3 4</sup> However, analysis of patient data indicates that tumor-infiltrating T cells in ovarian cancer are often poorly activated.<sup>5</sup> Understanding the mechanisms governing this poor T cell activation could lead to novel ovarian cancer therapeutics. <h3>Methods</h3> We utilized a transplantable, syngeneic, murine ovarian cancer cell line driven by Ccne1<sup>OE</sup> p53<sup>-/-R172H</sup> Ak2<sup>OE</sup> and Kras<sup>G12V</sup> (CPAK).<sup>6</sup> CPAK tumor cells were implanted intraperitoneally to model metastatic ovarian cancer or subcutaneously to model productive systemic immunity. CPAK cells were engineered to express the model CD8<sup>+</sup> T cell antigen SIY to enable studies of tumor-specific T cells. CBT consisted of αCTLA-4 and αPD-L1 therapy. Immune cells were profiled using flow cytometry and single-cell RNA sequencing (scRNA-seq). <h3>Results</h3> Intraperitoneal (IP) CPAK-SIY tumors were unresponsive to CBT. However, mice bearing subcutaneous (SQ) CPAK-SIY tumors treated with CBT displayed delayed tumor growth compared to control animals. Flow cytometric analysis of tumor-infiltrating CD8<sup>+</sup> T cells illustrated that while tumor-reactive T cells were activated in IP and SQ tumors, tumor-reactive T cells in IP tumors failed to upregulate high levels of effector molecules such as granzyme B. Unbiased analysis of dendritic cells (DCs) within IP tumors using scRNA-seq revealed a population of DCs that expressed CD103 and CD11b and a type 2 conventional DC (cDC2) gene signature. Gene signature analysis indicated these CD103<sup>+</sup> CD11b<sup>+</sup> double-positive DCs were a suppressive state of cDC2s induced by TGFb that reside in gut tissue and induce Tregs.<sup>7</sup> Analysis of tumor-infiltrating DCs in IP tumors revealed that the proportion of double-positive DCs increased during tumor growth while the proportion of cDC2s decreased. We hypothesized that the polarization of cDC2s away from these suppressive double-positive DCs may improve ovarian cancer immunity. Previous work from our group demonstrated that cDC2s acquire an activation state characterized by an interferon-stimulated gene signature (ISG<sup>+</sup> DCs) upon exposure to interferon-beta (IFNβ) and that ISG<sup>+</sup> DCs are potent activators of CD8<sup>+</sup> T cells.<sup>8</sup> In our ovarian cancer model, addition of IFNβ to IP tumors enhanced tumor-specific T cell responses. <h3>Conclusions</h3> Metastatic ovarian tumors are refractory to CBT and are infiltrated by poorly activated CD8<sup>+</sup> T cells. Our results suggest that the poor T cell activation is a consequence of the cDC2 activation state within the tumor. <h3>Acknowledgements</h3> We would like to thank MIT’s Department of Comparative Medicine and the Koch Institute’s Swanson Biotechnology Core Facility. This work was supported by Break Through Cancer and the Margaret A. Cunningham Immune Mechanisms in Cancer Research Fellowship. <h3>References</h3> Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. <i>CA Cancer J Clin</i> 2023;<b>73:</b>17–48, doi:10.3322/caac.21763. Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? <i>J Clin Oncol</i> 2019;<b>37:</b>2460–2471, doi:10.1200/JCO.19.00508. Tumeh PC,<i> et al.</i> PD-1 blockade induces responses by inhibiting adaptive immune resistance. <i>Nature</i> 2014;<b>515:</b>568–571, doi:10.1038/nature13954. Cristescu R,<i> et al.</i> Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. <i>Science</i> 2018;<b>362</b>, doi:10.1126/science.aar3593 (2018). Vazquez-Garcia I,<i> et al.</i> Ovarian cancer mutational processes drive site-specific immune evasion. <i>Nature</i> 2022;<b>612:</b>778–786, doi:10.1038/s41586-022-05496-1. Iyer S,<i> et al.</i> Genetically Defined Syngeneic Mouse Models of Ovarian Cancer as Tools for the Discovery of Combination Immunotherapy. <i>Cancer Discov</i> 2021;<b>11:</b>384–407, doi:10.1158/2159-8290.CD-20-0818. Bain CC,<i> et al.</i> TGFbetaR signalling controls CD103(+)CD11b(+) dendritic cell development in the intestine. <i>Nat Commun</i> 2017;<b>8:</b>620, doi:10.1038/s41467-017-00658-6. Duong E,<i> et al.</i> Type I interferon activates MHC class I-dressed CD11b(+) conventional dendritic cells to promote protective anti-tumor CD8(+) T cell immunity. <i>Immunity</i> 2022;<b>55:</b>308–323 e309, doi:10.1016/j.immuni.2021.10.020. <h3>Ethics Approval</h3> All mouse experiments were approved by MIT’s Committee on Animal Care (CAC) – PHS Animal Welfare Assurance # D16–00078 (A3125–01).","PeriodicalId":500964,"journal":{"name":"Regular and Young Investigator Award Abstracts","volume":null,"pages":null},"PeriodicalIF":0.0000,"publicationDate":"2023-11-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":"0","resultStr":null,"platform":"Semanticscholar","paperid":null,"PeriodicalName":"Regular and Young Investigator Award Abstracts","FirstCategoryId":"1085","ListUrlMain":"https://doi.org/10.1136/jitc-2023-sitc2023.0947","RegionNum":0,"RegionCategory":null,"ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"","PubModel":"","JCR":"","JCRName":"","Score":null,"Total":0}
引用次数: 0

Abstract

Background

Ovarian cancer is the fifth leading cause of cancer-related death in women in the United States.1 To date, checkpoint blockade therapy (CBT) has failed to be effective in ovarian cancer.2 CBT efficacy typically requires a strong pre-existing tumor-specific T cell response.3 4 However, analysis of patient data indicates that tumor-infiltrating T cells in ovarian cancer are often poorly activated.5 Understanding the mechanisms governing this poor T cell activation could lead to novel ovarian cancer therapeutics.

Methods

We utilized a transplantable, syngeneic, murine ovarian cancer cell line driven by Ccne1OE p53-/-R172H Ak2OE and KrasG12V (CPAK).6 CPAK tumor cells were implanted intraperitoneally to model metastatic ovarian cancer or subcutaneously to model productive systemic immunity. CPAK cells were engineered to express the model CD8+ T cell antigen SIY to enable studies of tumor-specific T cells. CBT consisted of αCTLA-4 and αPD-L1 therapy. Immune cells were profiled using flow cytometry and single-cell RNA sequencing (scRNA-seq).

Results

Intraperitoneal (IP) CPAK-SIY tumors were unresponsive to CBT. However, mice bearing subcutaneous (SQ) CPAK-SIY tumors treated with CBT displayed delayed tumor growth compared to control animals. Flow cytometric analysis of tumor-infiltrating CD8+ T cells illustrated that while tumor-reactive T cells were activated in IP and SQ tumors, tumor-reactive T cells in IP tumors failed to upregulate high levels of effector molecules such as granzyme B. Unbiased analysis of dendritic cells (DCs) within IP tumors using scRNA-seq revealed a population of DCs that expressed CD103 and CD11b and a type 2 conventional DC (cDC2) gene signature. Gene signature analysis indicated these CD103+ CD11b+ double-positive DCs were a suppressive state of cDC2s induced by TGFb that reside in gut tissue and induce Tregs.7 Analysis of tumor-infiltrating DCs in IP tumors revealed that the proportion of double-positive DCs increased during tumor growth while the proportion of cDC2s decreased. We hypothesized that the polarization of cDC2s away from these suppressive double-positive DCs may improve ovarian cancer immunity. Previous work from our group demonstrated that cDC2s acquire an activation state characterized by an interferon-stimulated gene signature (ISG+ DCs) upon exposure to interferon-beta (IFNβ) and that ISG+ DCs are potent activators of CD8+ T cells.8 In our ovarian cancer model, addition of IFNβ to IP tumors enhanced tumor-specific T cell responses.

Conclusions

Metastatic ovarian tumors are refractory to CBT and are infiltrated by poorly activated CD8+ T cells. Our results suggest that the poor T cell activation is a consequence of the cDC2 activation state within the tumor.

Acknowledgements

We would like to thank MIT’s Department of Comparative Medicine and the Koch Institute’s Swanson Biotechnology Core Facility. This work was supported by Break Through Cancer and the Margaret A. Cunningham Immune Mechanisms in Cancer Research Fellowship.

References

Siegel RL, Miller KD, Wagle NS, Jemal A. Cancer statistics, 2023. CA Cancer J Clin 2023;73:17–48, doi:10.3322/caac.21763. Kandalaft LE, Odunsi K, Coukos G. Immunotherapy in Ovarian Cancer: Are We There Yet? J Clin Oncol 2019;37:2460–2471, doi:10.1200/JCO.19.00508. Tumeh PC, et al. PD-1 blockade induces responses by inhibiting adaptive immune resistance. Nature 2014;515:568–571, doi:10.1038/nature13954. Cristescu R, et al. Pan-tumor genomic biomarkers for PD-1 checkpoint blockade-based immunotherapy. Science 2018;362, doi:10.1126/science.aar3593 (2018). Vazquez-Garcia I, et al. Ovarian cancer mutational processes drive site-specific immune evasion. Nature 2022;612:778–786, doi:10.1038/s41586-022-05496-1. Iyer S, et al. Genetically Defined Syngeneic Mouse Models of Ovarian Cancer as Tools for the Discovery of Combination Immunotherapy. Cancer Discov 2021;11:384–407, doi:10.1158/2159-8290.CD-20-0818. Bain CC, et al. TGFbetaR signalling controls CD103(+)CD11b(+) dendritic cell development in the intestine. Nat Commun 2017;8:620, doi:10.1038/s41467-017-00658-6. Duong E, et al. Type I interferon activates MHC class I-dressed CD11b(+) conventional dendritic cells to promote protective anti-tumor CD8(+) T cell immunity. Immunity 2022;55:308–323 e309, doi:10.1016/j.immuni.2021.10.020.

Ethics Approval

All mouse experiments were approved by MIT’s Committee on Animal Care (CAC) – PHS Animal Welfare Assurance # D16–00078 (A3125–01).
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
肿瘤驻留的2型树突状细胞的激活状态影响卵巢癌免疫应答的强度
背景:卵巢癌是美国女性癌症相关死亡的第五大原因,迄今为止,检查点阻断疗法(CBT)对卵巢癌无效CBT的有效性通常需要一个强大的预先存在的肿瘤特异性T细胞反应。然而,对患者数据的分析表明,卵巢癌的肿瘤浸润性T细胞往往激活不良了解控制这种不良T细胞活化的机制可能会导致新的卵巢癌治疗方法。方法利用一种可移植的、同基因的小鼠卵巢癌细胞系,由Ccne1OE p53-/- r172h Ak2OE和KrasG12V (CPAK)驱动CPAK肿瘤细胞被腹腔植入以模拟转移性卵巢癌,或皮下植入以模拟产生性全身免疫。CPAK细胞被工程化表达模型CD8+ T细胞抗原SIY,使肿瘤特异性T细胞的研究成为可能。CBT包括αCTLA-4和αPD-L1治疗。利用流式细胞术和单细胞RNA测序(scRNA-seq)对免疫细胞进行分析。结果腹腔(IP) cpap - siy肿瘤对CBT无反应。然而,与对照动物相比,接受CBT治疗的皮下(SQ) cpap - siy肿瘤小鼠的肿瘤生长延迟。流式细胞术分析肿瘤浸润的CD8+ T细胞表明,虽然肿瘤反应性T细胞在IP和SQ肿瘤中被激活,但IP肿瘤中的肿瘤反应性T细胞未能上调高水平的效应分子,如颗粒酶b。使用scRNA-seq对IP肿瘤中的树突状细胞(DC)进行无偏分析,结果显示DC群体表达CD103和CD11b以及2型常规DC (cDC2)基因特征。基因标记分析表明,这些CD103+ CD11b+双阳性dc是TGFb诱导的cDC2s存在于肠道组织并诱导tregs的抑制状态。7对IP肿瘤中肿瘤浸润dc的分析显示,肿瘤生长过程中双阳性dc的比例增加,而cDC2s的比例下降。我们假设cDC2s远离这些抑制性双阳性dc的极化可能改善卵巢癌免疫。本小组之前的研究表明,cDC2s在暴露于干扰素- β (ifn - β)时获得以干扰素刺激基因特征(ISG+ DCs)为特征的激活状态,ISG+ DCs是CD8+ T细胞的有效激活剂在我们的卵巢癌模型中,在IP肿瘤中添加IFNβ增强了肿瘤特异性T细胞反应。结论卵巢转移性肿瘤CBT治疗难治性强,且有低活化CD8+ T细胞浸润。我们的研究结果表明,T细胞激活不良是肿瘤内cDC2激活状态的结果。我们要感谢麻省理工学院的比较医学系和科赫研究所的斯旺森生物技术核心设施。这项工作得到了突破癌症和玛格丽特A.坎宁安癌症研究免疫机制奖学金的支持。引用文献sigel RL, Miller KD, Wagle NS . Jemal A.癌症统计,2023。中华肿瘤杂志[J]; 2009; 33 (3): 417 - 418, doi:10.3322/caac.21763。坎达拉夫特LE, Odunsi K, Coukos G.卵巢癌的免疫治疗:我们还在那里吗?中华临床医学杂志,2019;37:2460-2471,doi:10.1200/JCO.19.00508。Tumeh PC等。PD-1阻断通过抑制适应性免疫抵抗诱导应答。Nature 2014; 515:568-571, doi:10.1038/nature13954。Cristescu R,等。基于PD-1检查点阻断免疫治疗的泛肿瘤基因组生物标志物。Science 2018;362, doi:10.1126/ Science。aar3593(2018)。巴斯克斯-加西亚等人。卵巢癌突变过程驱动位点特异性免疫逃避。Nature 2022; 612:778-786, doi:10.1038/s41586-022-05496-1。Iyer S,等。基因定义的同基因卵巢癌小鼠模型作为发现联合免疫治疗的工具。癌症发现2021;11:384-407,doi:10.1158/2159-8290.CD-20-0818。Bain CC等。TGFbetaR信号控制肠道中CD103(+)CD11b(+)树突状细胞的发育。学报,2017;8:620,doi:10.1038/s41467-017-00658-6。杜恩,等。I型干扰素激活MHC I类修饰的CD11b(+)常规树突状细胞,促进保护性抗肿瘤CD8(+) T细胞免疫。免疫2022;55:308-323 e309, doi:10.1016/ j.j immune .2021.10.020。所有小鼠实验均经麻省理工学院动物护理委员会(CAC) - PHS动物福利保证# D16-00078 (A3125-01)批准。
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
自引率
0.00%
发文量
0
期刊最新文献
1455 Characterization of gene expression signatures of tumor immunogenicity and cellular proliferation from murine cancer models grownin vitroandin vivo 1193 Synergistic targeting of multiple activating pathways with natural killer cell engagers 1413 Immunogenicity of SARS-CoV-2 mRNA vaccines in individuals with thymic epithelial tumors 1150 DM005, an EGFR × MET bispecific antibody-drug conjugate, showed robust anti-tumor activity in PDX models 1274 Infliximab for decompensated diabetes following immune checkpoint inhibitor therapy
×
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