首页 > 最新文献

Tackling the Tumor Microenvironment: Beyond T-cells最新文献

英文 中文
Abstract A094: Characterization of pancreatic cancer endothelial cells: Approach to enhance immune cell infiltration for immunotherapy 摘要:胰腺癌内皮细胞的特征:增强免疫细胞浸润的免疫治疗方法
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A094
K. Nakajima, Y. Ino, T. Iwasaki, N. Hiraoka
The hurdles in realizing immunotherapy success for cure stem from the fact that cancer patients are either refractory to immune response and/or develop resistance. We previously proposed that these phenomena are due, in part, to the deployment of tumor-associated antigens, or employment of tumor-associated endothelium acting as a gatekeeper for immune cell infiltration into the cancer tissue (Nakajima et al, Cancer Res 2017;77:5441-4). Here, an extensive study unveiled functional/molecular differences of endothelium derived from pancreatic cancer and normal pancreas. They were isolated from fresh surgical specimen by magnet-based selection. The primary culture of tumor-associated endothelial cells was confirmed by double positive expressions of endothelial markers, CD31 and ERG1. They showed the short vessel formations and the narrow area of capillary network, indicating the low potential of angiogenesis. Further, peripheral blood–derived lymphocytes were less adhering to the tumor-associated endothelial cells. To find the molecular differences, microarray analysis was performed, and identified 2748 molecules distinct from endothelial cells of noncancerous tissues (p Citation Format: Kosei Nakajima, Yashunori Ino, Toshimitsu Iwasaki, Nobuyoshi Hiraoka. Characterization of pancreatic cancer endothelial cells: Approach to enhance immune cell infiltration for immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A094.
实现免疫疗法成功治愈的障碍源于这样一个事实,即癌症患者要么对免疫反应难治,要么产生耐药性。我们之前提出,这些现象部分是由于肿瘤相关抗原的部署,或肿瘤相关内皮作为免疫细胞浸润到癌症组织的守门人(Nakajima等人,cancer Res 2017;77:5441-4)。在这里,一项广泛的研究揭示了来自胰腺癌和正常胰腺的内皮细胞的功能/分子差异。它们是从新鲜的手术标本中通过磁基选择分离出来的。内皮标志物CD31和ERG1双阳性表达证实了肿瘤相关内皮细胞的原代培养。血管形成短,毛细血管网面积窄,表明血管生成潜力低。此外,外周血源性淋巴细胞粘附肿瘤相关内皮细胞较少。为了发现分子差异,进行了微阵列分析,并鉴定出2748个与非癌组织内皮细胞不同的分子(p引用格式:Kosei Nakajima, Yashunori Ino, Toshimitsu Iwasaki, Nobuyoshi hiroka)。胰腺癌内皮细胞的表征:增强免疫细胞浸润进行免疫治疗的方法[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A094。
{"title":"Abstract A094: Characterization of pancreatic cancer endothelial cells: Approach to enhance immune cell infiltration for immunotherapy","authors":"K. Nakajima, Y. Ino, T. Iwasaki, N. Hiraoka","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A094","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A094","url":null,"abstract":"The hurdles in realizing immunotherapy success for cure stem from the fact that cancer patients are either refractory to immune response and/or develop resistance. We previously proposed that these phenomena are due, in part, to the deployment of tumor-associated antigens, or employment of tumor-associated endothelium acting as a gatekeeper for immune cell infiltration into the cancer tissue (Nakajima et al, Cancer Res 2017;77:5441-4). Here, an extensive study unveiled functional/molecular differences of endothelium derived from pancreatic cancer and normal pancreas. They were isolated from fresh surgical specimen by magnet-based selection. The primary culture of tumor-associated endothelial cells was confirmed by double positive expressions of endothelial markers, CD31 and ERG1. They showed the short vessel formations and the narrow area of capillary network, indicating the low potential of angiogenesis. Further, peripheral blood–derived lymphocytes were less adhering to the tumor-associated endothelial cells. To find the molecular differences, microarray analysis was performed, and identified 2748 molecules distinct from endothelial cells of noncancerous tissues (p Citation Format: Kosei Nakajima, Yashunori Ino, Toshimitsu Iwasaki, Nobuyoshi Hiraoka. Characterization of pancreatic cancer endothelial cells: Approach to enhance immune cell infiltration for immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A094.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82085679","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract A048: Targeting the tumor microenvironment to enhance immunotherapy against cancer [A048]靶向肿瘤微环境增强肿瘤免疫治疗
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A048
C. Slaney, A. Oliver, M. Kershaw
Immunotherapies that harness the immune system against cancer are an attractive proposition for cancer treatment. While there have been some promising successes, only a small fraction of patients obtain clinical benefit. It has become clear that the immunosuppressive tumor microenvironment (TME) is a major obstacle for immunotherapies, because the TME suppresses immune responses, leading to reduced efficacy. We previously demonstrated that the site of tumor growth is a major determinant in sculpting the organ-specific TME, and thus predisposes treatment efficacy (1). In this project, we hypothesize that the TME of visceral tumors is more immunosuppressive than those of the tumors growing elsewhere. We investigated in murine models the difference in the TME in breast cancer growing orthotopically and the same breast cancer growing in the common metastatic sites, such as the lungs. Our findings showed that the breast cancer growing in the lungs was resistant to immunotherapies such as anti-PD1 and anti-CTLA-4, whereas the breast cancer growing orthotopically could be completely eradicated even when the cancer burden was greater. Through an institutional prospective community-based rapid autopsy program (CASCADE), we obtained genetically matched metastases and surrounding tissues from several sites in the same breast cancer patients and investigated the TME from these tumors using novel technologies such as RNAseq and multiplex immunohistochemistry. Strikingly, the TMEs from the same organs in different patients have similar immune gene expression profiles and in contrast, TMEs from the same patient differ greatly in different organs. Together, our research demonstrates an organ-specific difference between TMEs that leads to different responses to therapies. We anticipate that further study of how cancer cells sculpt the TME at different sites will greatly enhance our understanding of the TME and provide promising targets to enhance current immunotherapies, especially for patients who do not respond to existing therapies. Reference: 1. Devaud C., et al. Molecular Therapy 2014;22:18-27. Citation Format: Clare Y. Slaney, Amanda J. Oliver, Michael H. Kershaw. Targeting the tumor microenvironment to enhance immunotherapy against cancer [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A048.
利用免疫系统对抗癌症的免疫疗法是癌症治疗的一个有吸引力的提议。虽然取得了一些有希望的成功,但只有一小部分患者获得了临床益处。免疫抑制性肿瘤微环境(TME)是免疫治疗的主要障碍,因为TME抑制免疫应答,导致疗效降低。我们之前已经证明,肿瘤生长部位是塑造器官特异性TME的主要决定因素,从而影响治疗效果(1)。在本项目中,我们假设内脏肿瘤的TME比生长在其他地方的肿瘤更具有免疫抑制作用。我们在小鼠模型中研究了原位生长的乳腺癌和在常见转移部位(如肺)生长的相同乳腺癌的TME的差异。我们的研究结果表明,生长在肺部的乳腺癌对抗pd1和抗ctla -4等免疫疗法具有耐药性,而原位生长的乳腺癌即使在癌症负担更大的情况下也可以被完全根除。通过机构前瞻性社区快速尸检项目(CASCADE),我们从同一乳腺癌患者的几个部位获得遗传匹配的转移灶和周围组织,并使用RNAseq和多重免疫组织化学等新技术研究这些肿瘤的TME。引人注目的是,来自同一器官的不同患者的TMEs具有相似的免疫基因表达谱,相反,来自同一患者的不同器官的TMEs差异很大。总之,我们的研究表明,TMEs之间存在器官特异性差异,导致对治疗的不同反应。我们预计,进一步研究癌细胞如何在不同部位雕刻TME将大大提高我们对TME的理解,并提供有希望的靶点来增强当前的免疫疗法,特别是对那些对现有疗法没有反应的患者。参考:1。Devaud C.等。分子治疗2014;22:18-27。引用格式:Clare Y. Slaney, Amanda J. Oliver, Michael H. Kershaw。靶向肿瘤微环境增强肿瘤免疫治疗[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A048。
{"title":"Abstract A048: Targeting the tumor microenvironment to enhance immunotherapy against cancer","authors":"C. Slaney, A. Oliver, M. Kershaw","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A048","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A048","url":null,"abstract":"Immunotherapies that harness the immune system against cancer are an attractive proposition for cancer treatment. While there have been some promising successes, only a small fraction of patients obtain clinical benefit. It has become clear that the immunosuppressive tumor microenvironment (TME) is a major obstacle for immunotherapies, because the TME suppresses immune responses, leading to reduced efficacy. We previously demonstrated that the site of tumor growth is a major determinant in sculpting the organ-specific TME, and thus predisposes treatment efficacy (1). In this project, we hypothesize that the TME of visceral tumors is more immunosuppressive than those of the tumors growing elsewhere. We investigated in murine models the difference in the TME in breast cancer growing orthotopically and the same breast cancer growing in the common metastatic sites, such as the lungs. Our findings showed that the breast cancer growing in the lungs was resistant to immunotherapies such as anti-PD1 and anti-CTLA-4, whereas the breast cancer growing orthotopically could be completely eradicated even when the cancer burden was greater. Through an institutional prospective community-based rapid autopsy program (CASCADE), we obtained genetically matched metastases and surrounding tissues from several sites in the same breast cancer patients and investigated the TME from these tumors using novel technologies such as RNAseq and multiplex immunohistochemistry. Strikingly, the TMEs from the same organs in different patients have similar immune gene expression profiles and in contrast, TMEs from the same patient differ greatly in different organs. Together, our research demonstrates an organ-specific difference between TMEs that leads to different responses to therapies. We anticipate that further study of how cancer cells sculpt the TME at different sites will greatly enhance our understanding of the TME and provide promising targets to enhance current immunotherapies, especially for patients who do not respond to existing therapies. Reference: 1. Devaud C., et al. Molecular Therapy 2014;22:18-27. Citation Format: Clare Y. Slaney, Amanda J. Oliver, Michael H. Kershaw. Targeting the tumor microenvironment to enhance immunotherapy against cancer [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A048.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75701050","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A090: The Duffy Antigen Receptor for Chemokines (DARC) influences levels of tumor-associated leukocytes in the breast tumor microenvironment Duffy抗原趋化因子受体(DARC)影响乳腺肿瘤微环境中肿瘤相关白细胞的水平
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A090
Rachel N. Martini, Brittany D. Jenkins, C. Yates, L. Newman, M. Davis
The regulation of immune cell infiltration into the tumor microenvironment can influence disease prognosis. The specific populations that are present can inform potential treatment options. This work investigates immune cell regulation in the breast cancer tumor microenvironment, specifically how an atypical chemokine receptor, known as the Duffy Antigen Receptor for Chemokines (DARC/ACKR1) can influence levels of leukocyte populations in the breast tumor environment. DARC is a nonsignaling receptor able to bind both the CC and CXC classes of chemokines, and mainly functions to modulate levels of chemokines in circulation, and aid in chemokine transport in tissues. In this regard, DARC expression may determine the profile of immune response.To investigate DARC expression and its effects on tumor-associated leukocyte (TAL) populations, we obtained RNA-seq data from The Cancer Genome Atlas (TCGA) Breast Cancer cohort. We proceeded through our analysis with those samples denoted as primary tumor samples (n=400). To estimate the relative abundance of TAL populations, we used the CIBERSORT online platform, which estimates fractions of 22 different TAL populations based on gene expression data. After completing the CIBERSORT analysis, we proceeded with only those samples that had a significant CIBERSORT output (n=167). In our analysis, we found that the total abundance of all TALs was significantly higher in tumors that have high DARC expression (p Citation Format: Rachel Martini, Brittany D. Jenkins, Clayton Yates, Lisa Newman, Melissa Davis. The Duffy Antigen Receptor for Chemokines (DARC) influences levels of tumor-associated leukocytes in the breast tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A090.
免疫细胞对肿瘤微环境浸润的调控影响疾病预后。现有的特定人群可以为潜在的治疗方案提供信息。这项工作研究了乳腺癌肿瘤微环境中的免疫细胞调节,特别是一种非典型趋化因子受体,即达菲趋化因子抗原受体(DARC/ACKR1)如何影响乳腺癌肿瘤环境中白细胞群的水平。DARC是一种非信号受体,能够结合CC和CXC类趋化因子,主要作用是调节循环中趋化因子的水平,并帮助趋化因子在组织中的运输。在这方面,DARC的表达可能决定了免疫反应的概况。为了研究DARC表达及其对肿瘤相关白细胞(TAL)群体的影响,我们从癌症基因组图谱(TCGA)乳腺癌队列中获得了RNA-seq数据。我们继续对原发肿瘤样本(n=400)进行分析。为了估计TAL种群的相对丰度,我们使用了CIBERSORT在线平台,该平台基于基因表达数据估计了22个不同TAL种群的部分。在完成CIBERSORT分析之后,我们只处理那些具有显著CIBERSORT输出的样本(n=167)。在我们的分析中,我们发现在具有高DARC表达的肿瘤中,所有tal的总丰度明显更高(p引文格式:Rachel Martini, Brittany D. Jenkins, Clayton Yates, Lisa Newman, Melissa Davis)。达菲抗原趋化因子受体(DARC)影响乳腺肿瘤微环境中肿瘤相关白细胞的水平[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A090。
{"title":"Abstract A090: The Duffy Antigen Receptor for Chemokines (DARC) influences levels of tumor-associated leukocytes in the breast tumor microenvironment","authors":"Rachel N. Martini, Brittany D. Jenkins, C. Yates, L. Newman, M. Davis","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A090","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A090","url":null,"abstract":"The regulation of immune cell infiltration into the tumor microenvironment can influence disease prognosis. The specific populations that are present can inform potential treatment options. This work investigates immune cell regulation in the breast cancer tumor microenvironment, specifically how an atypical chemokine receptor, known as the Duffy Antigen Receptor for Chemokines (DARC/ACKR1) can influence levels of leukocyte populations in the breast tumor environment. DARC is a nonsignaling receptor able to bind both the CC and CXC classes of chemokines, and mainly functions to modulate levels of chemokines in circulation, and aid in chemokine transport in tissues. In this regard, DARC expression may determine the profile of immune response.To investigate DARC expression and its effects on tumor-associated leukocyte (TAL) populations, we obtained RNA-seq data from The Cancer Genome Atlas (TCGA) Breast Cancer cohort. We proceeded through our analysis with those samples denoted as primary tumor samples (n=400). To estimate the relative abundance of TAL populations, we used the CIBERSORT online platform, which estimates fractions of 22 different TAL populations based on gene expression data. After completing the CIBERSORT analysis, we proceeded with only those samples that had a significant CIBERSORT output (n=167). In our analysis, we found that the total abundance of all TALs was significantly higher in tumors that have high DARC expression (p Citation Format: Rachel Martini, Brittany D. Jenkins, Clayton Yates, Lisa Newman, Melissa Davis. The Duffy Antigen Receptor for Chemokines (DARC) influences levels of tumor-associated leukocytes in the breast tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A090.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"82296398","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A072: Calreticulin exposures by malignant blasts correlate with robust anticancer immunity and improved clinical outcome in AML patients 恶性细胞暴露钙网蛋白与AML患者强大的抗癌免疫和改善的临床结果相关
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A072
J. Fucikova
Cancer cell death can be perceived as immunogenic by the host only when malignant cells emit immunostimulatory signals (so-called “damage-associated molecular patterns,” DAMPs), as they die in the context of failing adaptive responses to stress. Accumulating preclinical and clinical evidence indicates that the capacity of immunogenic cell death to (re-)activate an anticancer immune response is key to the success of various chemo- and radiotherapeutic regimens. Malignant blasts from patients with acute myeloid leukemia (AML) exposed multiple DAMPs, including calreticulin (CRT), heat-shock protein 70 (HSP70), and HSP90 on their plasma membrane irrespective of treatment. In these patients, high levels of surface-exposed (ecto)-CRT correlated with an increased proportion of natural killer cells and effector memory CD4+ and CD8+ T-cells in the periphery. Moreover, CRT exposure on the plasma membrane of malignant blasts positively correlated with the frequency of circulating T-cells specific for leukemia-associated antigens, indicating that ecto-CRT favors the initiation of anticancer immunity in patients with AML. Finally, although the levels of ecto-HSP70, ecto-HSP90, and ecto-CRT were all associated with improved relapse-free survival, only CRT exposure significantly correlated with superior overall survival. Thus, CRT exposure represents a novel powerful prognostic biomarker for patients with AML, reflecting the activation of a clinically relevant AML-specific immune response. Citation Format: Jitka Fucikova. Calreticulin exposures by malignant blasts correlate with robust anticancer immunity and improved clinical outcome in AML patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A072.
只有当恶性细胞发出免疫刺激信号(所谓的“损伤相关分子模式”,DAMPs)时,宿主才会认为癌细胞死亡是免疫原性的,因为它们在对压力的适应性反应失败的情况下死亡。越来越多的临床前和临床证据表明,免疫原性细胞死亡(重新)激活抗癌免疫反应的能力是各种化疗和放疗方案成功的关键。急性髓性白血病(AML)患者的恶性细胞不论治疗方式如何,其质膜上暴露了多种DAMPs,包括钙调蛋白(CRT)、热休克蛋白70 (HSP70)和HSP90。在这些患者中,高水平的表面暴露(ecto)-CRT与外周自然杀伤细胞和效应记忆CD4+和CD8+ t细胞比例增加相关。此外,恶性原细胞质膜上的CRT暴露与白血病相关抗原特异性循环t细胞的频率呈正相关,表明体外CRT有利于AML患者抗癌免疫的启动。最后,尽管ecto-HSP70、ecto-HSP90和ecto-CRT水平均与改善无复发生存相关,但只有CRT暴露与优越的总生存显著相关。因此,CRT暴露代表了AML患者一种新的强大的预后生物标志物,反映了临床相关AML特异性免疫反应的激活。引文格式:Jitka Fucikova。恶性细胞暴露钙网蛋白与AML患者强大的抗癌免疫和改善的临床结果相关[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A072。
{"title":"Abstract A072: Calreticulin exposures by malignant blasts correlate with robust anticancer immunity and improved clinical outcome in AML patients","authors":"J. Fucikova","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A072","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A072","url":null,"abstract":"Cancer cell death can be perceived as immunogenic by the host only when malignant cells emit immunostimulatory signals (so-called “damage-associated molecular patterns,” DAMPs), as they die in the context of failing adaptive responses to stress. Accumulating preclinical and clinical evidence indicates that the capacity of immunogenic cell death to (re-)activate an anticancer immune response is key to the success of various chemo- and radiotherapeutic regimens. Malignant blasts from patients with acute myeloid leukemia (AML) exposed multiple DAMPs, including calreticulin (CRT), heat-shock protein 70 (HSP70), and HSP90 on their plasma membrane irrespective of treatment. In these patients, high levels of surface-exposed (ecto)-CRT correlated with an increased proportion of natural killer cells and effector memory CD4+ and CD8+ T-cells in the periphery. Moreover, CRT exposure on the plasma membrane of malignant blasts positively correlated with the frequency of circulating T-cells specific for leukemia-associated antigens, indicating that ecto-CRT favors the initiation of anticancer immunity in patients with AML. Finally, although the levels of ecto-HSP70, ecto-HSP90, and ecto-CRT were all associated with improved relapse-free survival, only CRT exposure significantly correlated with superior overall survival. Thus, CRT exposure represents a novel powerful prognostic biomarker for patients with AML, reflecting the activation of a clinically relevant AML-specific immune response. Citation Format: Jitka Fucikova. Calreticulin exposures by malignant blasts correlate with robust anticancer immunity and improved clinical outcome in AML patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A072.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73107856","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A079: Secreted IL-12p70 from long-term activated dendritic cells is lost concomitant with their apoptosis and release of IL-10 长期活化的树突状细胞分泌的IL-12p70随着其凋亡和IL-10的释放而丢失
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A079
M. Hansen, L. Carstensen, A. Obers, I. Svane
The intimate balance between peripheral tolerance and adaptive immunity has profound implications in several disease settings. Interleukin-12 (IL-12) plays a major role in immunity to intracellular pathogens and cancer by controlling IFNγ-dependent adaptive immunity. The transient production of the bioactive IL-12p70 heterodimer and the concurrent expression of interleukin-10 (IL-10) serves as a myeloid checkpoint to avoid immunopathology. Here, long-term exposure to inflammatory stimuli was evaluated on monocyte-derived dendritic cells (DCs) activated with lipopolysaccharide (LPS) and gamma interferon (IFNγ). The secretion of IFNγ from co-cultures with allogeneic T-cells present in peripheral blood mononuclear cells from healthy volunteers served as a measure of T-cell activation.The secretion of IFNγ from co-cultures was progressively lost as exposure of DCs to inflammatory stimuli was extended from one up to four days prior to co-culture or following IL-12p70 antibody-mediated blockade. Most pronounced was the 12-fold reduction (N = 9 donor pairs) seen with four-day activated DCs. Furthermore, at four days of activation, a significant fraction of DCs underwent apoptosis concomitant with their increased release of IL-10 and a striking 10-fold drop in levels of IL-12p70 as compared with DCs activated one, two or three days. Furthermore, after four days of activation, DC-derived IL-12p70 was inversely correlated with IL-10 and with IFNγ derived from co-cultures. It is currently an open question whether IL-12p70 naturally degrades after four days of activation or whether apoptotic DCs actively stimulate the degradation of IL-12p70. Citation Format: Morten Hansen, Laura Stentoft Carstensen, Andreas Obers, Inge Marie Stentoft Svane. Secreted IL-12p70 from long-term activated dendritic cells is lost concomitant with their apoptosis and release of IL-10 [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A079.
外周耐受性和适应性免疫之间的密切平衡在几种疾病环境中具有深远的意义。白细胞介素-12 (IL-12)通过控制ifn γ依赖性适应性免疫,在细胞内病原体和癌症的免疫中发挥重要作用。生物活性IL-12p70异二聚体的瞬时产生和白介素-10 (IL-10)的同时表达作为骨髓检查点以避免免疫病理。本研究通过脂多糖(LPS)和γ干扰素(IFNγ)激活的单核细胞来源的树突状细胞(dc),对长期暴露于炎症刺激进行了评估。与健康志愿者外周血单个核细胞中存在的同种异体t细胞共培养的IFNγ分泌可作为t细胞活化的测量指标。随着DCs暴露于炎症刺激的时间从共培养前1天延长至4天,或IL-12p70抗体介导的阻断后,共培养中IFNγ的分泌逐渐减少。最明显的是,4天激活的dc减少了12倍(N = 9对供体)。此外,在激活4天后,与激活1、2或3天的DCs相比,相当一部分DCs在IL-10释放增加的同时发生凋亡,IL-12p70水平显著下降10倍。此外,在激活4天后,dc来源的IL-12p70与IL-10和来自共培养的IFNγ呈负相关。IL-12p70在激活4天后是否会自然降解,或者凋亡的dc是否会主动刺激IL-12p70的降解,目前还是一个悬而未决的问题。引文格式:Morten Hansen, Laura Stentoft Carstensen, Andreas Obers, Inge Marie Stentoft Svane。长期活化的树突状细胞分泌的IL-12p70随着其凋亡和IL-10的释放而丢失[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A079。
{"title":"Abstract A079: Secreted IL-12p70 from long-term activated dendritic cells is lost concomitant with their apoptosis and release of IL-10","authors":"M. Hansen, L. Carstensen, A. Obers, I. Svane","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A079","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A079","url":null,"abstract":"The intimate balance between peripheral tolerance and adaptive immunity has profound implications in several disease settings. Interleukin-12 (IL-12) plays a major role in immunity to intracellular pathogens and cancer by controlling IFNγ-dependent adaptive immunity. The transient production of the bioactive IL-12p70 heterodimer and the concurrent expression of interleukin-10 (IL-10) serves as a myeloid checkpoint to avoid immunopathology. Here, long-term exposure to inflammatory stimuli was evaluated on monocyte-derived dendritic cells (DCs) activated with lipopolysaccharide (LPS) and gamma interferon (IFNγ). The secretion of IFNγ from co-cultures with allogeneic T-cells present in peripheral blood mononuclear cells from healthy volunteers served as a measure of T-cell activation.The secretion of IFNγ from co-cultures was progressively lost as exposure of DCs to inflammatory stimuli was extended from one up to four days prior to co-culture or following IL-12p70 antibody-mediated blockade. Most pronounced was the 12-fold reduction (N = 9 donor pairs) seen with four-day activated DCs. Furthermore, at four days of activation, a significant fraction of DCs underwent apoptosis concomitant with their increased release of IL-10 and a striking 10-fold drop in levels of IL-12p70 as compared with DCs activated one, two or three days. Furthermore, after four days of activation, DC-derived IL-12p70 was inversely correlated with IL-10 and with IFNγ derived from co-cultures. It is currently an open question whether IL-12p70 naturally degrades after four days of activation or whether apoptotic DCs actively stimulate the degradation of IL-12p70. Citation Format: Morten Hansen, Laura Stentoft Carstensen, Andreas Obers, Inge Marie Stentoft Svane. Secreted IL-12p70 from long-term activated dendritic cells is lost concomitant with their apoptosis and release of IL-10 [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A079.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75474703","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract PR03: Immune-based classification of soft-tissue sarcoma is associated with clinical outcome and unveils tertiary lymphoid structures as surrogate biomarker for the clinic 基于免疫的软组织肉瘤分类与临床预后相关,并揭示了三级淋巴组织结构作为临床替代生物标志物
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR03
Weikang Chen, F. Petitprez, Cheng-Ming Sun, L. Lacroix, A. Reyniès, A. Italiano, M. Toulmonde, C. Lucchesi, Y. Laizet, C. Sautès-Fridman, W. Fridman
Soft tissue sarcoma (STS) are rare mesenchymal-originated tumors with more than 50 different histologies identified. Not every histology in STS responds to immunotherapy and immunologic predictive markers are lacking. The purpose of this study is to establish an immune classification of STS by analysis of the transcriptome. This was performed by using a deconvolution method that allowed us to quantify 8 immune populations and endothelial cells. As a secondary objective, we searched for a surrogate biomarker that could be assessable in the clinic. We analyzed transcriptomic data of four publicly available datasets, accounting for 608 complex genomic STS, including leiomyosarcoma (LMS, 35.4%), dedifferentiated liposarcoma (DDLPS, 33.9%) and undifferentiated pleomorphic sarcoma (UPS, 30.8%). By using the MCP-Counter deconvolution method, we characterized the tumor microenvironment (TME) of these tumors and established a robust immune classification that is consistent through various cohorts. We classified the patients into 5 Sarcoma Immune Classes (SIC), labeled as A1, A2, B, C1 and C2. The A1 and A2 groups are associated with very low to low immune infiltrates. Conversely, SIC C1 and C2 tumors are characterized by strong to very strong expression of signatures associated to all immune cells. SIC B tumors are characterized by a high expression of endothelial cell signature, an intermediate presence of neutrophils, and a rather low infiltration by other immune cell types. Regarding functional orientation of the TME, gene signatures associated with immune cells chemotaxis activation and survival, expression of major histocompatibility complex class I, and regulatory T-cells are highly expressed in SIC C1 and C2, modestly expressed in B and A2, and very lowly expressed in A1. Interestingly, immune checkpoint genes exhibited strong expression differences between SICs. SIC C2 had a strong expression of PD-1, PD-L2, CTLA-4 and TIM-3 genes. We also found that the lymphoid structure-associated B cell chemoattractant chemokine CXCL13 is remarkably highly expressed in C2 class. CXCL13 is associated with the presence of tertiary lymphoid structures (TLS). Although all histologies are distributed in each SIC group, LMS are more commonly found in the immune low SIC A1 and A2 groups, and we also extended our analysis to other histologies such as synovial sarcoma or gastrointestinal stromal tumors. Our classification is associated with clinical outcome, and SIC group C (C1/C2) has the longest overall survival, as compared to SIC A group (A1/A2) (p = 0.015). We then validated SIC classification using STS FFPE samples (n=32). SIC classification by RNA expression was correlated with quantitative immunohistochemistry (IHC) of CD3 (T-cells), DC-Lamp (activated dendritic cells), CD20 (B cells), CD8 (CD8+ T-cells), and CD34 (endothelial cells). Densities of CD3 (p=0.0033), CD8 (p=0.004) and CD20 (p=0.00043) were significantly higher in SIC C tumors. Tumor SIC B groups ha
软组织肉瘤(STS)是一种罕见的间质肿瘤,有超过50种不同的组织学。并非所有STS的组织学都对免疫治疗有反应,缺乏免疫预测标志物。本研究的目的是通过转录组分析建立STS的免疫分类。这是通过使用反褶积方法进行的,该方法使我们能够量化免疫群体和内皮细胞。作为次要目标,我们寻找一种可在临床评估的替代生物标志物。我们分析了4个公开数据集的转录组学数据,共涉及608个复杂基因组STS,包括平滑肌肉瘤(LMS, 35.4%)、去分化脂肪肉瘤(DDLPS, 33.9%)和未分化多形性肉瘤(UPS, 30.8%)。通过使用MCP-Counter反卷积方法,我们表征了这些肿瘤的肿瘤微环境(TME),并建立了在不同队列中一致的稳健免疫分类。我们将患者分为5个肉瘤免疫级(SIC),分别标记为A1、A2、B、C1和C2。A1和A2组与非常低至低的免疫浸润有关。相反,SIC C1和C2肿瘤的特点是与所有免疫细胞相关的特征强到非常强的表达。SIC B肿瘤的特点是内皮细胞特征的高表达,中性粒细胞的中间存在,以及其他免疫细胞类型的相当低的浸润。在TME的功能取向方面,与免疫细胞趋化激活和存活、主要组织相容性复合体I类和调节性t细胞相关的基因特征在SIC C1和C2中高表达,在B和A2中适度表达,在A1中极低表达。有趣的是,免疫检查点基因在sic之间表现出强烈的表达差异。SIC C2强表达PD-1、PD-L2、CTLA-4和TIM-3基因。我们还发现淋巴样结构相关的B细胞趋化因子CXCL13在C2类中显著高表达。CXCL13与三级淋巴结构(TLS)的存在有关。虽然所有组织学分布在每个SIC组,但LMS更常见于免疫低SIC A1和A2组,我们也将分析扩展到其他组织学,如滑膜肉瘤或胃肠道间质瘤。我们的分类与临床结果相关,与SIC A组(A1/A2)相比,SIC C组(C1/C2)的总生存期最长(p = 0.015)。然后,我们使用STS FFPE样本(n=32)验证SIC分类。通过RNA表达分类SIC与CD3 (t细胞)、DC-Lamp(活化的树突状细胞)、CD20 (B细胞)、CD8 (CD8+ t细胞)和CD34(内皮细胞)的定量免疫组化(IHC)相关。CD3 (p=0.0033)、CD8 (p=0.004)和CD20 (p=0.00043)在sicc肿瘤中显著升高。肿瘤SIC B组CD34+内皮细胞表达较高,但不显著(p=0.6)。在验证队列中,SIC组C与较好的预后相关(p=0.053)。我们进一步研究了TLS是否是免疫高SIC C组的标志。不出所料,tls阳性肿瘤仅在验证队列的SIC C2组(100%,9/9)和C1组(50%,3/6)中发现。我们扩大验证队列(n = 93)来分析TLS与CD3+/CD8+/CD20+肿瘤浸润淋巴细胞的相关性。我们发现每种TILs的存在都与TLS (CD3, CD8, CD20)的存在显著相关,所有p值均为:weiwu Tom Chen, Florent Petitprez, chengming Sun, Laetitia Lacroix, Aurelien de Reynies, Antoine Italiano, Maud Toulmonde, Carlo Lucchesi, yeec9han Laizet, Catherine Sautes-Fridman, Wolf Herve Fridman。软组织肉瘤的免疫分类与临床结果相关,并揭示了三级淋巴结构作为临床替代生物标志物的作用[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫,2019;7(2增刊):摘要nr PR03。
{"title":"Abstract PR03: Immune-based classification of soft-tissue sarcoma is associated with clinical outcome and unveils tertiary lymphoid structures as surrogate biomarker for the clinic","authors":"Weikang Chen, F. Petitprez, Cheng-Ming Sun, L. Lacroix, A. Reyniès, A. Italiano, M. Toulmonde, C. Lucchesi, Y. Laizet, C. Sautès-Fridman, W. Fridman","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR03","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR03","url":null,"abstract":"Soft tissue sarcoma (STS) are rare mesenchymal-originated tumors with more than 50 different histologies identified. Not every histology in STS responds to immunotherapy and immunologic predictive markers are lacking. The purpose of this study is to establish an immune classification of STS by analysis of the transcriptome. This was performed by using a deconvolution method that allowed us to quantify 8 immune populations and endothelial cells. As a secondary objective, we searched for a surrogate biomarker that could be assessable in the clinic. We analyzed transcriptomic data of four publicly available datasets, accounting for 608 complex genomic STS, including leiomyosarcoma (LMS, 35.4%), dedifferentiated liposarcoma (DDLPS, 33.9%) and undifferentiated pleomorphic sarcoma (UPS, 30.8%). By using the MCP-Counter deconvolution method, we characterized the tumor microenvironment (TME) of these tumors and established a robust immune classification that is consistent through various cohorts. We classified the patients into 5 Sarcoma Immune Classes (SIC), labeled as A1, A2, B, C1 and C2. The A1 and A2 groups are associated with very low to low immune infiltrates. Conversely, SIC C1 and C2 tumors are characterized by strong to very strong expression of signatures associated to all immune cells. SIC B tumors are characterized by a high expression of endothelial cell signature, an intermediate presence of neutrophils, and a rather low infiltration by other immune cell types. Regarding functional orientation of the TME, gene signatures associated with immune cells chemotaxis activation and survival, expression of major histocompatibility complex class I, and regulatory T-cells are highly expressed in SIC C1 and C2, modestly expressed in B and A2, and very lowly expressed in A1. Interestingly, immune checkpoint genes exhibited strong expression differences between SICs. SIC C2 had a strong expression of PD-1, PD-L2, CTLA-4 and TIM-3 genes. We also found that the lymphoid structure-associated B cell chemoattractant chemokine CXCL13 is remarkably highly expressed in C2 class. CXCL13 is associated with the presence of tertiary lymphoid structures (TLS). Although all histologies are distributed in each SIC group, LMS are more commonly found in the immune low SIC A1 and A2 groups, and we also extended our analysis to other histologies such as synovial sarcoma or gastrointestinal stromal tumors. Our classification is associated with clinical outcome, and SIC group C (C1/C2) has the longest overall survival, as compared to SIC A group (A1/A2) (p = 0.015). We then validated SIC classification using STS FFPE samples (n=32). SIC classification by RNA expression was correlated with quantitative immunohistochemistry (IHC) of CD3 (T-cells), DC-Lamp (activated dendritic cells), CD20 (B cells), CD8 (CD8+ T-cells), and CD34 (endothelial cells). Densities of CD3 (p=0.0033), CD8 (p=0.004) and CD20 (p=0.00043) were significantly higher in SIC C tumors. Tumor SIC B groups ha","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"79775193","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A070: Virotherapy eradicates established melanoma by reprogramming the tumor microenvironment and engaging the adaptive immunity 摘要/ Abstract摘要:病毒疗法通过对肿瘤微环境进行重编程和激活适应性免疫来根除已形成的黑色素瘤
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A070
L. Flatz, S. Ring, D. Bomze, L. Onder, Jovana Cupovic, S. Schmidt, K. Orlinger, A. Bešše, L. Besse, C. Driessen, Hung-Wei Cheng, A. Lercher, D. Speiser, T. Bald, A. Bergthaler, B. Ludewig
Immunotherapy revolutionized the treatment of cancer patients. However, the lack of tumor specific T-cells and the immunosuppressive tumor microenvironment remain the major obstacles in curing treatment-resistant tumors. Here, we show that a novel, propagating noncytopathic virotherapy expressing the tumor-associated antigen TRP2 can eradicate established tumors. Interestingly, this was dependent on the route of treatment. Systemic administration of gene-based virotherapy induced a high number of tumor-infiltrating TRP2 specific CD8+ T-cells but was not able to cure established tumors. Moreover, localized tumor therapy in the periphery cured also distant metastasis in the lung, indicating that the locally induced immune response generates a systemic antitumor effect. Localized virotherapy predominantly infects tumor cells and tumor-associated fibroblasts, resulting in a proinflammatory reprogramming of the tumor microenvironment. Our data reveal that this immune activation is dependent on type I IFN signaling on the host but not on the tumor cell. These results have important clinical implications: i) our data explain why T-cell transfer or T-cell vaccines alone do not cure established tumors; ii) intratumoral gene-based cancer vaccination is superior to systemic treatment; and iii) a successful local antitumor response is associated with an efficient systemic antitumor response. Directly cancer targeting noncytopathic gene-based vaccines may be a promising approach by simultaneously supercharging the suppressive tumor microenvironment and inducing an adaptive immune response against selected tumor antigens. Citation Format: Lukas Flatz, Sandra Ring, David Bomze, Lucas Onder, Jovana Cupovic, Sarah Schmidt, Klaus Orlinger, Andrej Besse, Lenka Besse, Christoph Driessen, Hung-Wei Cheng, Alexander Lercher, Daniel Speiser, Tobias Bald, Andreas Bergthaler, Burkhard Ludewig. Virotherapy eradicates established melanoma by reprogramming the tumor microenvironment and engaging the adaptive immunity [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A070.
免疫疗法使癌症患者的治疗发生了革命性的变化。然而,缺乏肿瘤特异性t细胞和免疫抑制肿瘤微环境仍然是治疗耐药肿瘤的主要障碍。在这里,我们展示了一种表达肿瘤相关抗原TRP2的新型增殖性非细胞病变病毒疗法可以根除已建立的肿瘤。有趣的是,这取决于治疗途径。系统给予基于基因的病毒治疗诱导大量肿瘤浸润TRP2特异性CD8+ t细胞,但不能治愈已建立的肿瘤。此外,外周的局部肿瘤治疗也治愈了肺的远处转移,表明局部诱导的免疫反应产生了全身抗肿瘤作用。局部病毒治疗主要感染肿瘤细胞和肿瘤相关成纤维细胞,导致肿瘤微环境的促炎重编程。我们的数据显示,这种免疫激活依赖于宿主的I型IFN信号,而不是肿瘤细胞。这些结果具有重要的临床意义:i)我们的数据解释了为什么t细胞转移或t细胞疫苗单独不能治愈已建立的肿瘤;Ii)肿瘤内基于基因的癌症疫苗接种优于全身治疗;iii)成功的局部抗肿瘤反应与有效的全身抗肿瘤反应相关。直接靶向肿瘤的非细胞病变基因疫苗可能是一种很有前途的方法,同时增强抑制肿瘤微环境并诱导针对选定肿瘤抗原的适应性免疫反应。引文格式:Lukas Flatz, Sandra Ring, David Bomze, Lucas Onder, Jovana Cupovic, Sarah Schmidt, Klaus Orlinger, Andrej bse, Lenka bse, Christoph Driessen, hong - wei Cheng, Alexander Lercher, Daniel Speiser, Tobias Bald, Andreas Bergthaler, Burkhard Ludewig。病毒疗法通过重新编程肿瘤微环境和参与适应性免疫来根除已建立的黑色素瘤[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A070。
{"title":"Abstract A070: Virotherapy eradicates established melanoma by reprogramming the tumor microenvironment and engaging the adaptive immunity","authors":"L. Flatz, S. Ring, D. Bomze, L. Onder, Jovana Cupovic, S. Schmidt, K. Orlinger, A. Bešše, L. Besse, C. Driessen, Hung-Wei Cheng, A. Lercher, D. Speiser, T. Bald, A. Bergthaler, B. Ludewig","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A070","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A070","url":null,"abstract":"Immunotherapy revolutionized the treatment of cancer patients. However, the lack of tumor specific T-cells and the immunosuppressive tumor microenvironment remain the major obstacles in curing treatment-resistant tumors. Here, we show that a novel, propagating noncytopathic virotherapy expressing the tumor-associated antigen TRP2 can eradicate established tumors. Interestingly, this was dependent on the route of treatment. Systemic administration of gene-based virotherapy induced a high number of tumor-infiltrating TRP2 specific CD8+ T-cells but was not able to cure established tumors. Moreover, localized tumor therapy in the periphery cured also distant metastasis in the lung, indicating that the locally induced immune response generates a systemic antitumor effect. Localized virotherapy predominantly infects tumor cells and tumor-associated fibroblasts, resulting in a proinflammatory reprogramming of the tumor microenvironment. Our data reveal that this immune activation is dependent on type I IFN signaling on the host but not on the tumor cell. These results have important clinical implications: i) our data explain why T-cell transfer or T-cell vaccines alone do not cure established tumors; ii) intratumoral gene-based cancer vaccination is superior to systemic treatment; and iii) a successful local antitumor response is associated with an efficient systemic antitumor response. Directly cancer targeting noncytopathic gene-based vaccines may be a promising approach by simultaneously supercharging the suppressive tumor microenvironment and inducing an adaptive immune response against selected tumor antigens. Citation Format: Lukas Flatz, Sandra Ring, David Bomze, Lucas Onder, Jovana Cupovic, Sarah Schmidt, Klaus Orlinger, Andrej Besse, Lenka Besse, Christoph Driessen, Hung-Wei Cheng, Alexander Lercher, Daniel Speiser, Tobias Bald, Andreas Bergthaler, Burkhard Ludewig. Virotherapy eradicates established melanoma by reprogramming the tumor microenvironment and engaging the adaptive immunity [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A070.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"73967812","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract A083: Inflammasome-independent IL-1β release by myeloid cells promotes vessel destabilization and immune suppression in the tumor microenvironment 髓样细胞释放炎性小体非依赖性IL-1β促进肿瘤微环境中的血管不稳定和免疫抑制
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A083
M. Kiss, L. Walle, Helena Van Damme, Aleksandar Murgaski, Evangelia Bolli, J. Keirsse, Maria Solange Martins, Y. Elkrim, A. Fossoul, J. Serneels, M. Mazzone, M. Lamkanfi, J. A. Ginderachter, Damya Laoui
Background: Chronic inflammation in the tumor microenvironment (TME) sustained by immune cells has a crucial role both in tumor initiation and progression. One of the central cytokines of inflammation, IL-1β, is produced as a biologically inactive precursor that requires proteolytic processing by caspase-1. Activation of caspase-1 is triggered by the formation of inflammasomes, multiprotein complexes that detect microbial and endogenous danger signals primarily via NOD-like receptors, such as NLRP3 and NLRC4. Biologically active IL-1β is believed to be released through membrane pores formed by gasdermin D during a lytic form of cell death called pyroptosis. Although IL-1β-mediated inflammation has been shown to have a detrimental role in tumor progression, the signaling pathway controlling IL-1β release in the TME and the exact effect of the cytokine on antitumor T-cell responses have not been fully elucidated. A better understanding of how IL-1β release is controlled in tumors will also pave the way towards the therapeutic utilization of small-molecule inhibitors available to target NOD-like receptors and caspase-1. Methods: First, we characterized the impact of IL-1β in the TME by assessing the immune cell composition and vasculature of Lewis lung carcinomas (LLC) and E0771 breast carcinomas in IL-1β-deficient mice using flow cytometry and histologic analysis. Next, we used mice deficient in different inflammasome components, including NLRP3, NLRC4 and caspase-1, to investigate the involvement of these proteins in controlling IL-1β release in LLC and E0771 tumors. Using immunoblots and small-molecule inhibitors, we further characterized the activation of alternative enzymatic pathways and their involvement in IL-1β release by tumor-associated myeloid cells. Finally, we examined the role of pyroptosis and necroptosis in IL-1β release using gasdermin D- and MLKL-deficient mice, respectively. Release of IL-1β was assessed using ELISA and immunoblots. Results: We found that IL-1β secretion was restricted to myeloid cells and promoted tumor progression in mouse models of lung and breast carcinoma. IL-1β deletion abrogated the tumor-induced mobilization of immunosuppressive neutrophils and normalized the tumor vasculature, thereby alleviating hypoxia. Consequently, proliferation of effector T-cells in the TME was enhanced, leading to higher CD4+ and CD8+ T-cell abundance in the absence of IL-1β. We observed that, although the NLRP3 inflammasome was active in tumor-infiltrating myeloid cells, NLRP3 and caspase-1 were not essential for the proteolytic maturation of pro-IL-1β and secretion of biologically active IL-1β in the TME. Inhibition or genetic deletion of caspase-8 reduced inflammasome-independent IL-1β release, indicating that caspase-8 provides an alternative pathway for proteolytic activation and secretion of IL-1β in tumor-infiltrating myeloid cells. Moreover, IL-1β release by tumor-infiltrating myeloid cells was independent of lytic cell de
背景:肿瘤微环境(tumor microenvironment, TME)中由免疫细胞维持的慢性炎症在肿瘤的发生和发展中都起着至关重要的作用。炎症的中心细胞因子之一IL-1β是一种生物无活性的前体,需要caspase-1进行蛋白水解处理。caspase-1的激活是由炎性小体的形成触发的,炎性小体是一种多蛋白复合物,主要通过nod样受体(如NLRP3和NLRC4)检测微生物和内源性危险信号。生物活性IL-1β被认为是在细胞死亡的裂解形式(称为焦亡)中通过由气皮蛋白D形成的膜孔释放出来的。尽管IL-1β介导的炎症已被证明在肿瘤进展中具有有害作用,但在TME中控制IL-1β释放的信号通路以及细胞因子对抗肿瘤t细胞反应的确切作用尚未完全阐明。更好地了解IL-1β在肿瘤中的释放是如何被控制的,也将为靶向nod样受体和caspase-1的小分子抑制剂的治疗利用铺平道路。方法:首先,我们利用流式细胞术和组织学分析,通过评估IL-1β缺乏小鼠Lewis肺癌(LLC)和E0771乳腺癌的免疫细胞组成和血管系统,表征IL-1β对TME的影响。接下来,我们利用缺乏不同炎性小体成分(包括NLRP3、NLRC4和caspase-1)的小鼠,研究这些蛋白在LLC和E0771肿瘤中控制IL-1β释放的作用。利用免疫印迹和小分子抑制剂,我们进一步表征了替代酶途径的激活及其参与肿瘤相关骨髓细胞释放IL-1β的过程。最后,我们分别用gasdermin D-和mlkl缺陷小鼠研究了焦亡和坏死在IL-1β释放中的作用。采用ELISA和免疫印迹法检测IL-1β的释放。结果:我们发现IL-1β的分泌仅限于骨髓细胞,并促进肺癌和乳腺癌小鼠模型的肿瘤进展。IL-1β的缺失消除了肿瘤诱导的免疫抑制中性粒细胞的动员,使肿瘤血管正常化,从而缓解缺氧。因此,在缺乏IL-1β的情况下,TME中效应t细胞的增殖增强,导致CD4+和CD8+ t细胞丰度升高。我们观察到,尽管NLRP3炎性小体在肿瘤浸润的髓样细胞中具有活性,但NLRP3和caspase-1对于TME中IL-1β的蛋白水解成熟和生物活性IL-1β的分泌并不是必需的。caspase-8的抑制或基因缺失减少了炎性小体非依赖性IL-1β的释放,表明caspase-8为肿瘤浸润性骨髓细胞的蛋白水解激活和IL-1β的分泌提供了另一种途径。此外,肿瘤浸润的髓细胞释放IL-1β不依赖于溶解性细胞死亡方式,包括气真皮蛋白d介导的焦亡和mlkl介导的坏死亡,这表明该细胞因子在TME中有另一种释放机制。结论:总的来说,我们的研究结果表明,肿瘤浸润的髓样细胞能够独立于炎性小体释放IL-1β。我们发现髓细胞中通过caspase-8的IL-1β蛋白水解成熟是TME中免疫抑制的重要驱动因素,通过血管不稳定,免疫抑制中性粒细胞的募集和相应的抗肿瘤t细胞反应的抑制。我们还表明,与自身炎症不同,在肿瘤中,气皮蛋白d介导的焦亡对IL-1β的释放不是必需的。这些结果表明,由于肿瘤相关骨髓细胞中存在另一种caspase-8介导的IL-1β释放途径,治疗性抑制炎性小体或焦亡可能对某些肿瘤类型无效。引文格式:Mate Kiss, Lieselotte Vande Walle, Helena Van Damme, Aleksandar Murgaski, Evangelia Bolli, Jiri Keirsse, Maria Solange Martins, Yvon Elkrim, Amelie Fossoul, Jens Serneels, Massimiliano Mazzone, Mohamed Lamkanfi, Jo A. Van Ginderachter, Damya Laoui。髓样细胞释放炎性小体非依赖性IL-1β促进肿瘤微环境中的血管不稳定和免疫抑制[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A083。
{"title":"Abstract A083: Inflammasome-independent IL-1β release by myeloid cells promotes vessel destabilization and immune suppression in the tumor microenvironment","authors":"M. Kiss, L. Walle, Helena Van Damme, Aleksandar Murgaski, Evangelia Bolli, J. Keirsse, Maria Solange Martins, Y. Elkrim, A. Fossoul, J. Serneels, M. Mazzone, M. Lamkanfi, J. A. Ginderachter, Damya Laoui","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A083","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A083","url":null,"abstract":"Background: Chronic inflammation in the tumor microenvironment (TME) sustained by immune cells has a crucial role both in tumor initiation and progression. One of the central cytokines of inflammation, IL-1β, is produced as a biologically inactive precursor that requires proteolytic processing by caspase-1. Activation of caspase-1 is triggered by the formation of inflammasomes, multiprotein complexes that detect microbial and endogenous danger signals primarily via NOD-like receptors, such as NLRP3 and NLRC4. Biologically active IL-1β is believed to be released through membrane pores formed by gasdermin D during a lytic form of cell death called pyroptosis. Although IL-1β-mediated inflammation has been shown to have a detrimental role in tumor progression, the signaling pathway controlling IL-1β release in the TME and the exact effect of the cytokine on antitumor T-cell responses have not been fully elucidated. A better understanding of how IL-1β release is controlled in tumors will also pave the way towards the therapeutic utilization of small-molecule inhibitors available to target NOD-like receptors and caspase-1. Methods: First, we characterized the impact of IL-1β in the TME by assessing the immune cell composition and vasculature of Lewis lung carcinomas (LLC) and E0771 breast carcinomas in IL-1β-deficient mice using flow cytometry and histologic analysis. Next, we used mice deficient in different inflammasome components, including NLRP3, NLRC4 and caspase-1, to investigate the involvement of these proteins in controlling IL-1β release in LLC and E0771 tumors. Using immunoblots and small-molecule inhibitors, we further characterized the activation of alternative enzymatic pathways and their involvement in IL-1β release by tumor-associated myeloid cells. Finally, we examined the role of pyroptosis and necroptosis in IL-1β release using gasdermin D- and MLKL-deficient mice, respectively. Release of IL-1β was assessed using ELISA and immunoblots. Results: We found that IL-1β secretion was restricted to myeloid cells and promoted tumor progression in mouse models of lung and breast carcinoma. IL-1β deletion abrogated the tumor-induced mobilization of immunosuppressive neutrophils and normalized the tumor vasculature, thereby alleviating hypoxia. Consequently, proliferation of effector T-cells in the TME was enhanced, leading to higher CD4+ and CD8+ T-cell abundance in the absence of IL-1β. We observed that, although the NLRP3 inflammasome was active in tumor-infiltrating myeloid cells, NLRP3 and caspase-1 were not essential for the proteolytic maturation of pro-IL-1β and secretion of biologically active IL-1β in the TME. Inhibition or genetic deletion of caspase-8 reduced inflammasome-independent IL-1β release, indicating that caspase-8 provides an alternative pathway for proteolytic activation and secretion of IL-1β in tumor-infiltrating myeloid cells. Moreover, IL-1β release by tumor-infiltrating myeloid cells was independent of lytic cell de","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"75095153","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A062: TIM-3 plays distinct roles in different immune cells to regulate antitumor immune responses 摘要:TIM-3在不同的免疫细胞中发挥不同的作用,调节抗肿瘤免疫应答
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A062
J. Dai, J. Pei, M. Mohrs, G. Thurston, E. Ioffe
T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) was originally identified as an inhibitory receptor that is expressed on Th1 T-cells to dampen T-cell immunity upon interaction with several putative ligands. The inhibitory role of TIM-3 is supported by multiple preclinical disease models, such as viral and bacterial infections, experimental autoimmune encephalitis, and allograft rejection. Recent evidence suggests that induction of TIM-3 expression on T-cells may promote resistance to cancer immunotherapy, including in response to treatment with PD-1/PD-L1 inhibitors. Thus, TIM-3 represents a putative novel immuno-oncology target. Here we report that in naive mice TIM-3 is absent on T-cells, but is constitutively expressed on myeloid cells, including dendritic cells and macrophages. In tumor-bearing mice, TIM-3 expression is highly enriched on PD-1 positive CD4 and CD8 T-cells in the tumor microenvironment, but not on T-cells in draining lymph nodes or peripheral blood. Prophylactic or therapeutic treatment with anti-PD-1 blocking antibody delays tumor growth in wild-type mice. However, unexpectedly, TIM-3 genetic deficiency reduced overall survival of tumor-bearing mice treated with anti-PD-1 compared to wild-type control mice. Using a series of in vitro functional cell-based assays, we found that blocking TIM-3 function by either genetic knock-out or an inhibitory Ab increased proliferation of, and IFN-γ production by, effector CD8 T-cells following direct antigen stimulation. By contrast, TIM-3 gene knockout in bone marrow-derived macrophages did not impact responses to stimulation with Toll-like receptor ligands, and TIM-3 blockade reduced phagocytosis of apoptotic tumor cells by a macrophage cell line. Taken together, our results suggest TIM-3 may play opposite roles in T-cells and macrophages (inhibitory vs activating, respectively), and highlight the pleiotropic roles of TIM-3 in different immune cells in tumor immunology. Citation Format: Jie Dai, Jerry Pei, Markus Mohrs, Gavin Thurston, Ella Ioffe. TIM-3 plays distinct roles in different immune cells to regulate antitumor immune responses [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A062.
t细胞免疫球蛋白和粘蛋白结构域-3 (TIM-3)最初被认为是一种抑制受体,在Th1 t细胞上表达,通过与几种假定的配体相互作用来抑制t细胞免疫。TIM-3的抑制作用得到多种临床前疾病模型的支持,如病毒和细菌感染、实验性自身免疫性脑炎和同种异体移植排斥反应。最近的证据表明,在t细胞上诱导TIM-3表达可能会促进对癌症免疫治疗的抵抗,包括对PD-1/PD-L1抑制剂治疗的反应。因此,TIM-3代表了一个假定的新的免疫肿瘤学靶点。在这里,我们报告了在幼稚小鼠中,TIM-3在t细胞上不存在,但在骨髓细胞(包括树突状细胞和巨噬细胞)上组成性表达。在荷瘤小鼠中,TIM-3在肿瘤微环境中PD-1阳性CD4和CD8 t细胞上高度表达,而在引流淋巴结和外周血中的t细胞上则不表达。抗pd -1阻断抗体的预防性或治疗性治疗可延缓野生型小鼠的肿瘤生长。然而,出乎意料的是,与野生型对照小鼠相比,TIM-3基因缺陷降低了抗pd -1治疗的荷瘤小鼠的总存活率。通过一系列基于体外功能细胞的实验,我们发现通过基因敲除或抑制性Ab阻断TIM-3功能可增加CD8 t细胞在直接抗原刺激后的增殖和IFN-γ的产生。相比之下,在骨髓源性巨噬细胞中敲除TIM-3基因不影响toll样受体配体刺激的反应,并且TIM-3阻断减少了巨噬细胞系对凋亡肿瘤细胞的吞噬作用。综上所述,我们的研究结果表明TIM-3可能在t细胞和巨噬细胞中发挥相反的作用(分别是抑制和激活),并突出了TIM-3在不同免疫细胞中的多效性作用。引文格式:戴杰,Jerry Pei, Markus Mohrs, Gavin Thurston, Ella Ioffe。TIM-3在不同的免疫细胞中发挥不同的作用,调节抗肿瘤免疫应答[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A062。
{"title":"Abstract A062: TIM-3 plays distinct roles in different immune cells to regulate antitumor immune responses","authors":"J. Dai, J. Pei, M. Mohrs, G. Thurston, E. Ioffe","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A062","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A062","url":null,"abstract":"T-cell immunoglobulin and mucin-domain containing-3 (TIM-3) was originally identified as an inhibitory receptor that is expressed on Th1 T-cells to dampen T-cell immunity upon interaction with several putative ligands. The inhibitory role of TIM-3 is supported by multiple preclinical disease models, such as viral and bacterial infections, experimental autoimmune encephalitis, and allograft rejection. Recent evidence suggests that induction of TIM-3 expression on T-cells may promote resistance to cancer immunotherapy, including in response to treatment with PD-1/PD-L1 inhibitors. Thus, TIM-3 represents a putative novel immuno-oncology target. Here we report that in naive mice TIM-3 is absent on T-cells, but is constitutively expressed on myeloid cells, including dendritic cells and macrophages. In tumor-bearing mice, TIM-3 expression is highly enriched on PD-1 positive CD4 and CD8 T-cells in the tumor microenvironment, but not on T-cells in draining lymph nodes or peripheral blood. Prophylactic or therapeutic treatment with anti-PD-1 blocking antibody delays tumor growth in wild-type mice. However, unexpectedly, TIM-3 genetic deficiency reduced overall survival of tumor-bearing mice treated with anti-PD-1 compared to wild-type control mice. Using a series of in vitro functional cell-based assays, we found that blocking TIM-3 function by either genetic knock-out or an inhibitory Ab increased proliferation of, and IFN-γ production by, effector CD8 T-cells following direct antigen stimulation. By contrast, TIM-3 gene knockout in bone marrow-derived macrophages did not impact responses to stimulation with Toll-like receptor ligands, and TIM-3 blockade reduced phagocytosis of apoptotic tumor cells by a macrophage cell line. Taken together, our results suggest TIM-3 may play opposite roles in T-cells and macrophages (inhibitory vs activating, respectively), and highlight the pleiotropic roles of TIM-3 in different immune cells in tumor immunology. Citation Format: Jie Dai, Jerry Pei, Markus Mohrs, Gavin Thurston, Ella Ioffe. TIM-3 plays distinct roles in different immune cells to regulate antitumor immune responses [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A062.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"76927384","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Abstract A116: Lipid control of DNA-stimulated innate immunity A116: dna刺激先天免疫的脂质控制
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A116
Xiaojun Tan, Conggang Zhang, Zhijian J. Chen
Type I interferon (IFN) plays essential roles in both spontaneous and iatrogenic tumor immunogenicity. Tumor-derived DNA is recognized by 2’,3’-cyclic GMP-AMP (cGAMP) synthase (cGAS) that is important in tumor immunogenicity. Upon DNA binding, cGAS produces the endogenous second messenger cGAMP that binds to and activates stimulator of IFN genes (STING), a signaling adaptor. cGAMP binding triggers STING trafficking from the endoplasmic reticulum (ER) to perinuclear compartments, with simultaneous activation of serine/threonine-protein kinase TBK1 that in turn phosphorylates the transcription factor IRF3, leading to upregulation of type I interferons. However, where and how TBK1 is activated by STING upon cGAMP stimulation is unclear. Our study focuses on the regulation of cGAMP-stimulated STING trafficking and activation by lipid messengers with essential roles in subcellular protein/membrane trafficking and signaling. Through in vitro signaling reconstitution, we identified a cellular lipid as an essential factor of STING signaling. We found both STING and TBK1 were lipid effectors. Lipid binding not only promotes STING trafficking but also stimulates TBK1 activation. These results reveal a new component of the STING-TBK1 complex that controls cytosolic DNA-stimulated innate immune signaling. Citation Format: Xiaojun Tan, Conggang Zhang, Zhijian J. Chen. Lipid control of DNA-stimulated innate immunity [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A116.
I型干扰素(IFN)在自发性和医源性肿瘤的免疫原性中都起着重要作用。肿瘤来源的DNA被2 ',3 ' -环GMP-AMP (cGAMP)合成酶(cGAS)识别,这在肿瘤免疫原性中是重要的。DNA结合后,cGAS产生内源性第二信使cGAMP,该信使cGAMP结合并激活IFN基因刺激因子(STING),这是一种信号适配器。cGAMP结合触发STING从内质网(ER)运输到核周室,同时激活丝氨酸/苏氨酸蛋白激酶TBK1,进而磷酸化转录因子IRF3,导致I型干扰素上调。然而,在cGAMP刺激下,TBK1在何处以及如何被STING激活尚不清楚。我们的研究重点是cgamp刺激的STING运输和激活脂质信使的调节,这些信使在亚细胞蛋白/膜运输和信号传导中起重要作用。通过体外信号重构,我们发现细胞脂质是STING信号传导的重要因子。我们发现STING和TBK1都是脂质效应因子。脂质结合不仅促进STING转运,还刺激TBK1活化。这些结果揭示了STING-TBK1复合物的一个新成分,该成分控制胞质dna刺激的先天免疫信号。引用格式:谭晓军,张丛刚,陈志坚。dna刺激先天免疫的脂质调控[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志2019;7(2增刊):摘要nr A116。
{"title":"Abstract A116: Lipid control of DNA-stimulated innate immunity","authors":"Xiaojun Tan, Conggang Zhang, Zhijian J. Chen","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A116","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A116","url":null,"abstract":"Type I interferon (IFN) plays essential roles in both spontaneous and iatrogenic tumor immunogenicity. Tumor-derived DNA is recognized by 2’,3’-cyclic GMP-AMP (cGAMP) synthase (cGAS) that is important in tumor immunogenicity. Upon DNA binding, cGAS produces the endogenous second messenger cGAMP that binds to and activates stimulator of IFN genes (STING), a signaling adaptor. cGAMP binding triggers STING trafficking from the endoplasmic reticulum (ER) to perinuclear compartments, with simultaneous activation of serine/threonine-protein kinase TBK1 that in turn phosphorylates the transcription factor IRF3, leading to upregulation of type I interferons. However, where and how TBK1 is activated by STING upon cGAMP stimulation is unclear. Our study focuses on the regulation of cGAMP-stimulated STING trafficking and activation by lipid messengers with essential roles in subcellular protein/membrane trafficking and signaling. Through in vitro signaling reconstitution, we identified a cellular lipid as an essential factor of STING signaling. We found both STING and TBK1 were lipid effectors. Lipid binding not only promotes STING trafficking but also stimulates TBK1 activation. These results reveal a new component of the STING-TBK1 complex that controls cytosolic DNA-stimulated innate immune signaling. Citation Format: Xiaojun Tan, Conggang Zhang, Zhijian J. Chen. Lipid control of DNA-stimulated innate immunity [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr A116.","PeriodicalId":22141,"journal":{"name":"Tackling the Tumor Microenvironment: Beyond T-cells","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"83591098","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Tackling the Tumor Microenvironment: Beyond T-cells
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
×
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
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