首页 > 最新文献

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

英文 中文
Abstract A091: IL-33 activates antitumoral toxicity in eosinophils through stimulation of contact-dependent degranulation 摘要:IL-33通过刺激接触依赖性脱颗粒激活嗜酸性粒细胞的抗肿瘤毒性
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A091
F. Mattei, Carla Buccione, S. Andreone, F. Spadaro, A. Ninno, Jacopo Mancini, C. Zanetti, I. Parolini, F. Iosi, A. Tinari, V. Lucarini, A. Gerardino, Giovanna Ziccheddu, L. Businaro, C. Afferni, G. Schiavoni
The alarmin IL-33 plays pleiotropic roles in allergy, autoimmunity and inflammation through binding to its specific receptor ST2 expressed by most hematopoietic cells. Emerging evidences suggest an involvement of this cytokine also in cancer immunity, although its function remains ill-defined. Eosinophils (EOS) are a rare blood population playing critical roles in allergic inflammation and parasitic responses. We recently showed that EOS play an essential role in anti-tumor responses against melanoma growth and pulmonary metastasis mediated by IL-33 in vivo.In the present study we analyzed the mechanisms by which IL-33 mediates tumor infiltration and antitumoral activities of EOS. We show that IL-33 indirectly stimulates the recruitment of EOS inducing tumor-derived chemokines CCL24 and CCL5. Furthermore, IL-33 directly activates EOS inducing the expression of adhesion molecules, such as the integrin CD11b, resulting in efficient contact-dependent tumor cell killing. In co-culture experiments, IL-33 activated EOS tightly bond to tumor cells, forming increased numbers of conjugates, with respect to resting eosinophils. Confocal laser-scanning microscopy (CLSM) of eosinophil-tumor cell conjugates revealed polarization of the pore-forming eosinophilic cationic protein (ECP) and of CD11b on the cell synapses exclusively in IL-33-activated, but not resting, EOS. Furthermore, we show that IL-33 activated EOS release larger amounts of extracellular vesicles (EV) with respect to resting EOS. Transmission electron microscopy (TEM) revealed increased degranulation and EV release of IL-33-activated EOS following cell contact with target tumor cells. Our results advocate for an eosinophil-mediated tumoricidal function promoted by IL-33, thus opening perspectives for novel cancer immunotherapy strategies. Citation Format: Fabrizio Mattei, Carla Buccione, Sara Andreone, Francesca Spadaro, Adele De Ninno, Jacopo Mancini, Cristiana Zanetti, Isabella Parolini, Francesca Iosi, Antonella Tinari, Valeria Lucarini, Annamaria Gerardino, Giovanna Ziccheddu, Luca Businaro, Claudia Afferni, Giovanna Schiavoni. IL-33 activates antitumoral toxicity in eosinophils through stimulation of contact-dependent degranulation [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 A091.
警报蛋白IL-33通过与大多数造血细胞表达的特异性受体ST2结合,在过敏、自身免疫和炎症中发挥多效作用。新出现的证据表明,这种细胞因子也参与癌症免疫,尽管其功能仍不明确。嗜酸性粒细胞(EOS)是一种罕见的血液群体,在过敏性炎症和寄生反应中发挥重要作用。我们最近发现,在体内,EOS在IL-33介导的黑色素瘤生长和肺转移的抗肿瘤反应中发挥重要作用。在本研究中,我们分析了IL-33介导EOS的肿瘤浸润和抗肿瘤活性的机制。我们发现IL-33间接刺激EOS诱导肿瘤来源的趋化因子CCL24和CCL5的募集。此外,IL-33直接激活EOS,诱导粘附分子(如整合素CD11b)的表达,从而有效地杀死接触依赖性肿瘤细胞。在共培养实验中,IL-33激活的EOS与肿瘤细胞紧密结合,相对于静止的嗜酸性粒细胞形成更多的偶联物。对嗜酸性粒细胞-肿瘤细胞偶联物的共聚焦激光扫描显微镜(CLSM)显示,形成孔的嗜酸性阳离子蛋白(ECP)和CD11b在细胞突触上的极化仅在il- 33激活而非静止的EOS中发生。此外,我们发现IL-33激活的EOS比静息的EOS释放更多的细胞外囊泡(EV)。透射电镜(TEM)显示,在细胞与靶肿瘤细胞接触后,il -33激活的EOS脱颗粒和EV释放增加。我们的研究结果支持IL-33促进嗜酸性粒细胞介导的肿瘤杀伤功能,从而为新的癌症免疫治疗策略开辟了前景。引文格式:Fabrizio Mattei, Carla bucione, Sara Andreone, Francesca Spadaro, Adele De Ninno, Jacopo Mancini, Cristiana Zanetti, Isabella Parolini, Francesca Iosi, Antonella Tinari, Valeria Lucarini, Annamaria Gerardino, Giovanna Ziccheddu, Luca buciaro, Claudia Afferni, Giovanna Schiavoni。IL-33通过刺激接触依赖性脱颗粒激活嗜酸性粒细胞的抗肿瘤毒性[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 A091。
{"title":"Abstract A091: IL-33 activates antitumoral toxicity in eosinophils through stimulation of contact-dependent degranulation","authors":"F. Mattei, Carla Buccione, S. Andreone, F. Spadaro, A. Ninno, Jacopo Mancini, C. Zanetti, I. Parolini, F. Iosi, A. Tinari, V. Lucarini, A. Gerardino, Giovanna Ziccheddu, L. Businaro, C. Afferni, G. Schiavoni","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A091","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A091","url":null,"abstract":"The alarmin IL-33 plays pleiotropic roles in allergy, autoimmunity and inflammation through binding to its specific receptor ST2 expressed by most hematopoietic cells. Emerging evidences suggest an involvement of this cytokine also in cancer immunity, although its function remains ill-defined. Eosinophils (EOS) are a rare blood population playing critical roles in allergic inflammation and parasitic responses. We recently showed that EOS play an essential role in anti-tumor responses against melanoma growth and pulmonary metastasis mediated by IL-33 in vivo.In the present study we analyzed the mechanisms by which IL-33 mediates tumor infiltration and antitumoral activities of EOS. We show that IL-33 indirectly stimulates the recruitment of EOS inducing tumor-derived chemokines CCL24 and CCL5. Furthermore, IL-33 directly activates EOS inducing the expression of adhesion molecules, such as the integrin CD11b, resulting in efficient contact-dependent tumor cell killing. In co-culture experiments, IL-33 activated EOS tightly bond to tumor cells, forming increased numbers of conjugates, with respect to resting eosinophils. Confocal laser-scanning microscopy (CLSM) of eosinophil-tumor cell conjugates revealed polarization of the pore-forming eosinophilic cationic protein (ECP) and of CD11b on the cell synapses exclusively in IL-33-activated, but not resting, EOS. Furthermore, we show that IL-33 activated EOS release larger amounts of extracellular vesicles (EV) with respect to resting EOS. Transmission electron microscopy (TEM) revealed increased degranulation and EV release of IL-33-activated EOS following cell contact with target tumor cells. Our results advocate for an eosinophil-mediated tumoricidal function promoted by IL-33, thus opening perspectives for novel cancer immunotherapy strategies. Citation Format: Fabrizio Mattei, Carla Buccione, Sara Andreone, Francesca Spadaro, Adele De Ninno, Jacopo Mancini, Cristiana Zanetti, Isabella Parolini, Francesca Iosi, Antonella Tinari, Valeria Lucarini, Annamaria Gerardino, Giovanna Ziccheddu, Luca Businaro, Claudia Afferni, Giovanna Schiavoni. IL-33 activates antitumoral toxicity in eosinophils through stimulation of contact-dependent degranulation [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 A091.","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":"79766160","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 A074: Spatially resolved deep antigen profiling of single cells in FFPE tissue samples through CODEXTM 摘要/ Abstract A074:利用CODEXTM对FFPE组织样品中单细胞进行空间分辨深度抗原谱分析
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A074
M. Gallina, Atri Choksi, N. Nikulina, Jaskirat Singh, G. Dakshinamoorthy, Joseph Kim, S. Mistry, J. Kennedy‐Darling
Spatially resolved, deep antigen profiling of tissue specimens is crucial for investigating the architecture and cell diversity in complex matrices, such as the tumor microenvironment (TME). The correlation of these two parameters with the progression of a variety of pathologies, ranging from cancer to autoimmune diseases, is still vastly unknown due to the lack of technologies that enable the detection of both high content and spatial resolution. Formalin-fixed, paraffin-embedded (FFPE) tissues constitute the ideal sample for these investigations as they retain tissue morphology at room temperature for long periods of time, which is convenient and cost-effective, and are available in vast specimen archives. Akoya Biosciences, Inc., is commercializing CODEXTM (CO-Detection by indEXing), a multiparameteric imaging platform that performs the concurrent detection and quantification of dozens of antigens with single cell resolution within a tissue specimen. The CODEX platform uses a DNA-based barcode library to label antibodies and iterative cycles of adding and removing cognate dye-labeled oligonucleotides to reveal the staining pattern for a subset of the target markers per cycle. The entire data acquisition process is fully automated using the CODEX instrument, which integrates with existing microscopes. To support high-multiplexing capabilities, more than 45 different antibody clones have been screened and validated for the detection of target epitopes in human FFPE secondary lymphoid tissues for both healthy and cancerous specimens using the CODEX platform. Proof-of-concept data have also been obtained for different cancer types, including melanoma and head and neck cancer in both normal and TMA formats. Analysis software has been developed to extract meaning from CODEX datasets and is included as part of the CODEX platform. The analysis pipeline includes image drift compensation, deconvolution and segmentation to measure integrated fluorescence intensity for each cell across tens of parameters. In this manner, infiltrating lymphocytes and other immune cells have been identified within the TME. With the high multiplexing capabilities, a variety of T-cells can be identified to detect the ratios of both immunosuppressive and immune-activating subtypes. Analysis of FFPE tissues using the CODEX platform demonstrates the unparalleled opportunities for simultaneously detecting tens of antigens with single-cell resolution within a tissue specimen. Citation Format: Maria Elena Gallina, Atri Choksi, Nadya Nikulina, Jaskirat Singh, Gajalakshmi Dakshinamoorthy, Joseph Kim, Sejal Mistry, Julia Kennedy-Darling. Spatially resolved deep antigen profiling of single cells in FFPE tissue samples through CODEXTM [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 A074.
在空间分辨率上,组织标本的深层抗原谱分析对于研究复杂基质(如肿瘤微环境(TME))中的结构和细胞多样性至关重要。由于缺乏能够同时检测高含量和空间分辨率的技术,这两个参数与从癌症到自身免疫性疾病等各种病理进展的相关性仍然非常未知。福尔马林固定石蜡包埋(FFPE)组织是这些研究的理想样本,因为它们在室温下长时间保持组织形态,这是方便和经济有效的,并且可以在大量的标本档案中获得。Akoya Biosciences, Inc.正在商业化CODEXTM (CO-Detection by indEXing),这是一种多参数成像平台,可以在组织标本中以单细胞分辨率同时检测和定量数十种抗原。CODEX平台使用基于dna的条形码文库来标记抗体和添加和去除同源染料标记的寡核苷酸的迭代循环,以揭示每个循环中目标标记物子集的染色模式。整个数据采集过程使用CODEX仪器完全自动化,该仪器与现有显微镜集成。为了支持高复用能力,使用CODEX平台对超过45种不同的抗体克隆进行了筛选和验证,以检测健康和癌变标本的人FFPE二级淋巴组织中的目标表位。对于不同类型的癌症,包括正常和TMA两种形式的黑色素瘤和头颈癌,也获得了概念验证数据。已经开发了分析软件,从CODEX数据集中提取意义,并作为CODEX平台的一部分纳入其中。分析管道包括图像漂移补偿、反卷积和分割,以测量每个细胞在数十个参数中的综合荧光强度。通过这种方式,浸润淋巴细胞和其他免疫细胞已经在TME中被识别出来。由于具有高复用能力,可以识别各种t细胞以检测免疫抑制和免疫激活亚型的比例。使用CODEX平台对FFPE组织进行分析表明,在组织标本中以单细胞分辨率同时检测数十种抗原具有无与伦比的机会。引文格式:Maria Elena Gallina, Atri Choksi, Nadya Nikulina, Jaskirat Singh, Gajalakshmi Dakshinamoorthy, Joseph Kim, Sejal Mistry, Julia Kennedy-Darling。利用CODEXTM对FFPE组织样本中单细胞进行空间分辨深度抗原谱分析[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 A074。
{"title":"Abstract A074: Spatially resolved deep antigen profiling of single cells in FFPE tissue samples through CODEXTM","authors":"M. Gallina, Atri Choksi, N. Nikulina, Jaskirat Singh, G. Dakshinamoorthy, Joseph Kim, S. Mistry, J. Kennedy‐Darling","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A074","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A074","url":null,"abstract":"Spatially resolved, deep antigen profiling of tissue specimens is crucial for investigating the architecture and cell diversity in complex matrices, such as the tumor microenvironment (TME). The correlation of these two parameters with the progression of a variety of pathologies, ranging from cancer to autoimmune diseases, is still vastly unknown due to the lack of technologies that enable the detection of both high content and spatial resolution. Formalin-fixed, paraffin-embedded (FFPE) tissues constitute the ideal sample for these investigations as they retain tissue morphology at room temperature for long periods of time, which is convenient and cost-effective, and are available in vast specimen archives. Akoya Biosciences, Inc., is commercializing CODEXTM (CO-Detection by indEXing), a multiparameteric imaging platform that performs the concurrent detection and quantification of dozens of antigens with single cell resolution within a tissue specimen. The CODEX platform uses a DNA-based barcode library to label antibodies and iterative cycles of adding and removing cognate dye-labeled oligonucleotides to reveal the staining pattern for a subset of the target markers per cycle. The entire data acquisition process is fully automated using the CODEX instrument, which integrates with existing microscopes. To support high-multiplexing capabilities, more than 45 different antibody clones have been screened and validated for the detection of target epitopes in human FFPE secondary lymphoid tissues for both healthy and cancerous specimens using the CODEX platform. Proof-of-concept data have also been obtained for different cancer types, including melanoma and head and neck cancer in both normal and TMA formats. Analysis software has been developed to extract meaning from CODEX datasets and is included as part of the CODEX platform. The analysis pipeline includes image drift compensation, deconvolution and segmentation to measure integrated fluorescence intensity for each cell across tens of parameters. In this manner, infiltrating lymphocytes and other immune cells have been identified within the TME. With the high multiplexing capabilities, a variety of T-cells can be identified to detect the ratios of both immunosuppressive and immune-activating subtypes. Analysis of FFPE tissues using the CODEX platform demonstrates the unparalleled opportunities for simultaneously detecting tens of antigens with single-cell resolution within a tissue specimen. Citation Format: Maria Elena Gallina, Atri Choksi, Nadya Nikulina, Jaskirat Singh, Gajalakshmi Dakshinamoorthy, Joseph Kim, Sejal Mistry, Julia Kennedy-Darling. Spatially resolved deep antigen profiling of single cells in FFPE tissue samples through CODEXTM [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 A074.","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":"77236536","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: 2
Abstract A059: Tackling the tumor microenvironment with CD38 blockade to enhance cancer immunotherapy 摘要:利用CD38阻断物调控肿瘤微环境增强肿瘤免疫治疗
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A059
Limo Chen, L. Diao, X. Yi, B. L. Rodriguez, Yanli Li, P. Villalobos, T. Cascone, Xi Liu, L. Tan, P. Lorenzi, Jared J. Fradette, D. Peng, F. Skoulidis, Youhong Fan, J. Rodriguez-Canales, V. Papadimitrakopoulou, E. Dmitrovsky, L. Byers, Jing Wang, I. Wistuba, Jim Heymach, D. Gibbons
The single agent or the combination of anti-PD-1, anti-PD-L1, and anti-CTLA-4 is an effective strategy that is being clinically explored to treat a variety of cancer types. Some patients display primary resistance to the treatment, while others relapse after treatment. Resistance is a major issue that needs to be addressed. Using multiple immunocompetent syngeneic and K-rasLA1/+p53R172HΔg/+ spontaneous animal models of lung cancer, we have explored the mechanisms of resistance to treatments by evaluating the molecular and cellular immune profiles of the tumor microenvironment. We observed that tumor-bearing mice treated with PD-1/PD-L1 blocking antibodies developed resistance through the up-regulation of CD38. We also observed this in the combination therapy of anti-PD-1 and anti-CTLA-4, suggesting that CD38 is a major mechanism of resistance to immune checkpoint inhibitors. In vitro and in vivo studies demonstrated that CD38 impacted CD8+ T-cell function via adenosine receptor signaling and dendritic cell-mediated B7 signaling. Antibody-mediated cell depletion assays were conducted to validate the mechanisms. To determine the applicability to patients, we analyzed 793 lung cancer patients’ specimens with immunohistochemistry staining and assessed biomarker relationships in multiple large independent patient databases (~1,900 tumors). Pathologic analysis revealed positive immunohistochemical staining for CD38 on tumor cells in 15-23% of cases, and bioinformatic analyses revealed a strong correlation between CD38 expression and the immune signature. Lastly, targeting CD38 abolished the treatment resistance by modulating the adenosine levels and thereby enhancing the effector immune cell infiltrates into the tumor microenvironment. Based on our study, CD38 blockade improves the efficacy of single-agent anti-PD-1/PD-L1, or with anti-CTLA-4 combination in lung cancer. CD38 could potentially serve as a novel biomarker of resistance for immune checkpoint inhibition. The data from this study provide a unique target, biomarker, and therapeutic strategy that can be translated into the clinical practice. Citation Format: Limo Chen, Lixia Diao, Xiaohui Yi, Bertha Leticia Rodriguez, Yanli Li, Pamela Villalobos, Tina Cascone, Xi Liu, Lin Tan, Philip Lorenzi, Jared Fradette, David Peng, Ferdinandos Skoulidis, Youhong Fan, Jaime Rodriguez-Canales, Vassiliki Papadimitrakopoulou, Ethan Dmitrovsky, Lauren A Byers, Jing Wang, Ignasio Wistuba, Jim Heymach, Don Gibbons. Tackling the tumor microenvironment with CD38 blockade to enhance cancer 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 A059.
单药或联合抗pd -1、抗pd - l1、抗ctla -4是临床探索治疗多种癌症的有效策略。一些患者对治疗表现出原发性耐药性,而另一些患者在治疗后复发。耐药性是一个需要解决的主要问题。我们利用多种免疫活性同质和K-rasLA1/+p53R172HΔg/+自发肺癌动物模型,通过评估肿瘤微环境的分子和细胞免疫谱,探索了耐药机制。我们观察到,用PD-1/PD-L1阻断抗体治疗的荷瘤小鼠通过上调CD38产生耐药性。我们在抗pd -1和抗ctla -4联合治疗中也观察到这一点,这表明CD38是免疫检查点抑制剂耐药的主要机制。体外和体内研究表明,CD38通过腺苷受体信号传导和树突状细胞介导的B7信号传导影响CD8+ t细胞功能。进行了抗体介导的细胞耗竭试验来验证其机制。为了确定对患者的适用性,我们分析了793例肺癌患者的免疫组织化学染色标本,并评估了多个大型独立患者数据库(约1,900例肿瘤)中的生物标志物关系。病理分析显示15-23%的肿瘤细胞免疫组化染色CD38阳性,生物信息学分析显示CD38表达与免疫特征之间有很强的相关性。最后,靶向CD38通过调节腺苷水平来消除治疗耐药性,从而增强效应免疫细胞对肿瘤微环境的浸润。根据我们的研究,CD38阻断可提高单药抗pd -1/PD-L1或联合抗ctla -4治疗肺癌的疗效。CD38有可能作为免疫检查点抑制耐药性的一种新的生物标志物。这项研究的数据提供了一个独特的靶点、生物标志物和治疗策略,可以转化为临床实践。利用CD38阻断物处理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 A059。
{"title":"Abstract A059: Tackling the tumor microenvironment with CD38 blockade to enhance cancer immunotherapy","authors":"Limo Chen, L. Diao, X. Yi, B. L. Rodriguez, Yanli Li, P. Villalobos, T. Cascone, Xi Liu, L. Tan, P. Lorenzi, Jared J. Fradette, D. Peng, F. Skoulidis, Youhong Fan, J. Rodriguez-Canales, V. Papadimitrakopoulou, E. Dmitrovsky, L. Byers, Jing Wang, I. Wistuba, Jim Heymach, D. Gibbons","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A059","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A059","url":null,"abstract":"The single agent or the combination of anti-PD-1, anti-PD-L1, and anti-CTLA-4 is an effective strategy that is being clinically explored to treat a variety of cancer types. Some patients display primary resistance to the treatment, while others relapse after treatment. Resistance is a major issue that needs to be addressed. Using multiple immunocompetent syngeneic and K-rasLA1/+p53R172HΔg/+ spontaneous animal models of lung cancer, we have explored the mechanisms of resistance to treatments by evaluating the molecular and cellular immune profiles of the tumor microenvironment. We observed that tumor-bearing mice treated with PD-1/PD-L1 blocking antibodies developed resistance through the up-regulation of CD38. We also observed this in the combination therapy of anti-PD-1 and anti-CTLA-4, suggesting that CD38 is a major mechanism of resistance to immune checkpoint inhibitors. In vitro and in vivo studies demonstrated that CD38 impacted CD8+ T-cell function via adenosine receptor signaling and dendritic cell-mediated B7 signaling. Antibody-mediated cell depletion assays were conducted to validate the mechanisms. To determine the applicability to patients, we analyzed 793 lung cancer patients’ specimens with immunohistochemistry staining and assessed biomarker relationships in multiple large independent patient databases (~1,900 tumors). Pathologic analysis revealed positive immunohistochemical staining for CD38 on tumor cells in 15-23% of cases, and bioinformatic analyses revealed a strong correlation between CD38 expression and the immune signature. Lastly, targeting CD38 abolished the treatment resistance by modulating the adenosine levels and thereby enhancing the effector immune cell infiltrates into the tumor microenvironment. Based on our study, CD38 blockade improves the efficacy of single-agent anti-PD-1/PD-L1, or with anti-CTLA-4 combination in lung cancer. CD38 could potentially serve as a novel biomarker of resistance for immune checkpoint inhibition. The data from this study provide a unique target, biomarker, and therapeutic strategy that can be translated into the clinical practice. Citation Format: Limo Chen, Lixia Diao, Xiaohui Yi, Bertha Leticia Rodriguez, Yanli Li, Pamela Villalobos, Tina Cascone, Xi Liu, Lin Tan, Philip Lorenzi, Jared Fradette, David Peng, Ferdinandos Skoulidis, Youhong Fan, Jaime Rodriguez-Canales, Vassiliki Papadimitrakopoulou, Ethan Dmitrovsky, Lauren A Byers, Jing Wang, Ignasio Wistuba, Jim Heymach, Don Gibbons. Tackling the tumor microenvironment with CD38 blockade to enhance cancer 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 A059.","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":"86493192","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 A061: STING, an immune biomarker for colorectal cancer 摘要/ Abstract摘要:STING是结直肠癌的免疫生物标志物
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A061
H. Chon, Chan Kim
Background: STING is an innate immune sensor for cytosolic DNA. The activation of STING signaling is indispensable in Type I interferon response and the evocation of anticancer immune response by CD8+ T-cells. The aim of this study is to characterize intratumoral STING expression pattern and its clinical implication in colorectal cancer (CRC). Methods: We analyzed STING and CD8 expression in 225 CRC patients who underwent surgical resection. Clinicopathologic variables and survival outcomes were analyzed according to STING expression level. Results: Distinct STING expression was observed in tumor specimens of CRCs. Patients with higher STING expression seemed to have early stage cancer (P = 0.001) with increased intratumoral CD8+ T-cell infiltration (P Citation Format: Hongjae Chon, Chan Kim. STING, an immune biomarker for colorectal 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 A061.
背景:STING是细胞质DNA的先天免疫传感器。在I型干扰素应答和CD8+ t细胞抗癌免疫应答中,STING信号的激活是必不可少的。本研究的目的是表征结直肠癌(CRC)肿瘤内STING表达模式及其临床意义。方法:我们分析了225例手术切除的结直肠癌患者中STING和CD8的表达。根据STING表达水平分析临床病理指标及生存结果。结果:在肿瘤标本中观察到明显的STING表达。STING表达较高的患者似乎患有早期癌症(P = 0.001),肿瘤内CD8+ t细胞浸润增加(P引用格式:Hongjae Chon, Chan Kim)。结直肠癌的免疫生物标志物STING[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;癌症免疫杂志2019;7(2增刊):摘要nr A061。
{"title":"Abstract A061: STING, an immune biomarker for colorectal cancer","authors":"H. Chon, Chan Kim","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A061","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A061","url":null,"abstract":"Background: STING is an innate immune sensor for cytosolic DNA. The activation of STING signaling is indispensable in Type I interferon response and the evocation of anticancer immune response by CD8+ T-cells. The aim of this study is to characterize intratumoral STING expression pattern and its clinical implication in colorectal cancer (CRC). Methods: We analyzed STING and CD8 expression in 225 CRC patients who underwent surgical resection. Clinicopathologic variables and survival outcomes were analyzed according to STING expression level. Results: Distinct STING expression was observed in tumor specimens of CRCs. Patients with higher STING expression seemed to have early stage cancer (P = 0.001) with increased intratumoral CD8+ T-cell infiltration (P Citation Format: Hongjae Chon, Chan Kim. STING, an immune biomarker for colorectal 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 A061.","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":"87424577","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 A051: Melanoma displays an evolutionarily conserved resistance to upregulation of prophagocytic signals and to CD47 blockade 摘要:黑色素瘤对前吞噬细胞信号上调和CD47阻断表现出进化上保守的抗性
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A051
K. L. Anderson, K. Snyder, D. Ito, Debra C. Lins, L. Mills, K. Weiskopf, N. G. Ring, A. Ring, Y. Shimizu, M. Mescher, I. Weissman, J. Modiano
Therapeutic activation of macrophage phagocytosis has the ability to restrain tumor growth through phagocytic clearance of tumor cells and activation of the adaptive immune response. Our objective for this study was to evaluate the effects of modulating pro- and anti-phagocytic pathways in malignant melanoma. In order to identify evolutionarily conserved mechanisms of resistance that may be important for melanoma cell survival, we utilized a multispecies approach and examined the phagocytosis of human, mouse, and dog melanoma cells in vitro. We blocked the interaction between CD47 on tumor cells and SIRPα on macrophages using anti-CD47 monoclonal antibodies, SIRPα mimotopes, and a soluble fusion protein of the SIRPα extracellular domain. In all cases, we confirmed that these reagents blocked >85% of the maximum binding of CD47 to recombinant SIRPα. We observed that melanoma cells from all three species displayed unexpected resistance to phagocytosis that could not be fully mitigated by blockade of the “don’t eat me” signal CD47 or by chemotherapeutic enhancement of known “eat me” signals. In vitro, CD47 blockade minimally enhanced phagocytosis of melanoma cells, and loss of CD47 expression did not increase sensitivity to phagocytosis. In vivo, CD47 blockade enhanced the proliferation of tumor-specific CD8+ T-cells, but this response failed to inhibit tumor growth. Resistance to phagocytosis was not mediated by soluble factors, as phagocytosis of melanoma cells was not enhanced by inhibition of secretory pathways, and phagocytosis of sensitive lymphoma tumor cells was not impaired in the presence of melanoma cells or melanoma culture supernatants. siRNA-mediated knockdown of 47 prospective “don’t eat me” signals similarly did not enhance melanoma cell phagocytosis. Interestingly, in lymphoma cells, CD47 knockout alone did not enhance phagocytosis, suggesting that at least part of the pro-phagocytic effect of CD47 blockade is due to Fc receptor engagement. Restoration of CD47 expression in lymphoma cells re-established sensitivity to CD47 blockade. We conclude that melanoma cells possess an evolutionarily conserved resistance to macrophage phagocytosis. Further investigation will be needed to define and overcome the mechanisms that mediate melanoma cell resistance to innate immunity. Citation Format: Katie L. Anderson, Kristin M. Snyder, Daisuke Ito, Debra C. Lins, Lauren J. Mills, Kipp Weiskopf, Nan G. Ring, Aaron M Ring, Yoji Shimizu, Matthew F. Mescher, Irving L. Weissman, Jaime F. Modiano. Melanoma displays an evolutionarily conserved resistance to upregulation of prophagocytic signals and to CD47 blockade [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 A051.
治疗性激活巨噬细胞吞噬作用可以通过吞噬清除肿瘤细胞和激活适应性免疫反应来抑制肿瘤生长。我们这项研究的目的是评估调节促和抗吞噬通路在恶性黑色素瘤中的作用。为了确定可能对黑色素瘤细胞存活很重要的进化保守的耐药机制,我们采用多物种方法,并在体外检测了人类、小鼠和狗黑色素瘤细胞的吞噬作用。我们利用抗CD47单克隆抗体、SIRPα模位和SIRPα胞外结构域的可溶性融合蛋白阻断肿瘤细胞上的CD47与巨噬细胞上的SIRPα的相互作用。在所有病例中,我们证实这些试剂阻断了>85%的CD47与重组SIRPα的最大结合。我们观察到,来自这三个物种的黑色素瘤细胞都表现出意想不到的对吞噬的抵抗,这种抵抗不能通过阻断“不要吃我”信号CD47或通过化疗增强已知的“吃我”信号来完全减轻。在体外,CD47阻断对黑色素瘤细胞的吞噬作用有最低限度的增强,CD47表达的缺失并没有增加对吞噬的敏感性。在体内,CD47阻断增强了肿瘤特异性CD8+ t细胞的增殖,但这种反应未能抑制肿瘤生长。对吞噬的抵抗不是由可溶性因子介导的,因为黑素瘤细胞的吞噬作用不会通过抑制分泌途径而增强,并且在黑素瘤细胞或黑素瘤培养上清存在时,敏感淋巴瘤肿瘤细胞的吞噬作用不会受损。sirna介导的47个“不要吃我”信号的敲除同样没有增强黑色素瘤细胞的吞噬作用。有趣的是,在淋巴瘤细胞中,单独敲除CD47并没有增强吞噬作用,这表明CD47阻断的促吞噬作用至少部分是由于Fc受体的参与。淋巴瘤细胞中CD47表达的恢复重建了对CD47阻断的敏感性。我们得出结论,黑色素瘤细胞具有对巨噬细胞吞噬的进化保守抗性。需要进一步的研究来确定和克服介导黑色素瘤细胞抵抗先天免疫的机制。引文格式:Katie L. Anderson, Kristin M. Snyder, Daisuke Ito, Debra C. Lins, Lauren J. Mills, Kipp Weiskopf, Nan G. Ring, Aaron M Ring, Yoji Shimizu, Matthew F. Mescher, Irving L. Weissman, Jaime F. Modiano。黑色素瘤对前吞噬细胞信号上调和CD47阻断表现出进化上保守的抗性[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 A051。
{"title":"Abstract A051: Melanoma displays an evolutionarily conserved resistance to upregulation of prophagocytic signals and to CD47 blockade","authors":"K. L. Anderson, K. Snyder, D. Ito, Debra C. Lins, L. Mills, K. Weiskopf, N. G. Ring, A. Ring, Y. Shimizu, M. Mescher, I. Weissman, J. Modiano","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A051","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A051","url":null,"abstract":"Therapeutic activation of macrophage phagocytosis has the ability to restrain tumor growth through phagocytic clearance of tumor cells and activation of the adaptive immune response. Our objective for this study was to evaluate the effects of modulating pro- and anti-phagocytic pathways in malignant melanoma. In order to identify evolutionarily conserved mechanisms of resistance that may be important for melanoma cell survival, we utilized a multispecies approach and examined the phagocytosis of human, mouse, and dog melanoma cells in vitro. We blocked the interaction between CD47 on tumor cells and SIRPα on macrophages using anti-CD47 monoclonal antibodies, SIRPα mimotopes, and a soluble fusion protein of the SIRPα extracellular domain. In all cases, we confirmed that these reagents blocked >85% of the maximum binding of CD47 to recombinant SIRPα. We observed that melanoma cells from all three species displayed unexpected resistance to phagocytosis that could not be fully mitigated by blockade of the “don’t eat me” signal CD47 or by chemotherapeutic enhancement of known “eat me” signals. In vitro, CD47 blockade minimally enhanced phagocytosis of melanoma cells, and loss of CD47 expression did not increase sensitivity to phagocytosis. In vivo, CD47 blockade enhanced the proliferation of tumor-specific CD8+ T-cells, but this response failed to inhibit tumor growth. Resistance to phagocytosis was not mediated by soluble factors, as phagocytosis of melanoma cells was not enhanced by inhibition of secretory pathways, and phagocytosis of sensitive lymphoma tumor cells was not impaired in the presence of melanoma cells or melanoma culture supernatants. siRNA-mediated knockdown of 47 prospective “don’t eat me” signals similarly did not enhance melanoma cell phagocytosis. Interestingly, in lymphoma cells, CD47 knockout alone did not enhance phagocytosis, suggesting that at least part of the pro-phagocytic effect of CD47 blockade is due to Fc receptor engagement. Restoration of CD47 expression in lymphoma cells re-established sensitivity to CD47 blockade. We conclude that melanoma cells possess an evolutionarily conserved resistance to macrophage phagocytosis. Further investigation will be needed to define and overcome the mechanisms that mediate melanoma cell resistance to innate immunity. Citation Format: Katie L. Anderson, Kristin M. Snyder, Daisuke Ito, Debra C. Lins, Lauren J. Mills, Kipp Weiskopf, Nan G. Ring, Aaron M Ring, Yoji Shimizu, Matthew F. Mescher, Irving L. Weissman, Jaime F. Modiano. Melanoma displays an evolutionarily conserved resistance to upregulation of prophagocytic signals and to CD47 blockade [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 A051.","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":"85450414","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 A067: Cognate interaction with CD4+ T-cells instructs M2-like macrophages to acquire M1-like phenotype A067:与CD4+ t细胞同源相互作用可指导m2样巨噬细胞获得m1样表型
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A067
D. Eisel, W. Osen, K. Das, Franziska Hoerhold, R. König, S. Eichmüller
Current tumor immunotherapy approaches are based on application of checkpoint inhibitors, such as monoclonal antibodies against PD-1/PD-L1 and CTLA-4 or adoptive T-cell therapy using ex vivo expanded TILs or genetically modified autologous T-cells expressing recombinant T-cell receptors (TCRs) or chimeric antigen receptors (CARs). However, the success of these therapeutic strategies is often limited by various inhibitory immune cell types accumulating in the tumor. In particular, tumor-associated macrophages (TAMs) contribute to the immune suppressive tumor micro-milieu, since so-called M2-like macrophages suppress the function of T effector cells and promote the differentiation of regulatory T-cells (Treg) through secretion of inhibitory cytokines such as TGF-β and IL10. Tumor antigen-specific CD4+ T effector cells, on the other hand, can essentially sustain antitumoral immune responses as shown for various tumor entities. In fact, using peritoneal exudate cells (PECs) as source for macrophages we demonstrate that MHC II restricted interaction between ovalbumin (OVA) specific CD4+ T-cells and M2-like macrophages drives M1 polarization. This was confirmed by detailed gene and protein expression analyses as well as functional assays testing phagocytic and pinocytic activities of repolarized macrophages. Moreover, in a set of preclinical experiments, adoptive transfer of CD4+, OVA-specific OT-II cells into C57BL/6 mice bearing OVA expressing IAb-/- tumors resulted in increased intratumoral number of M1-like TAMs as determined by gene expression analysis and flow cytometry. Furthermore, we observed a significant survival benefit of mice treated with a combination of OVA-specific CD4+ (OT-II) and CD8+ (OT-I) cells after transplantation of B16F10-Ova tumors and a complete response in some mice that rejected the tumor cells also upon a later re-challenge. While the antitumoral effect of this adoptive transfer experiment has been already described, we now offer a possible explanation for the supportive effect of specific CD4+ cells on co-transferred CD8+ cells. Taken together, the instructive impact of CD4+ T-cells on M2-like macrophages described in this presentation points towards a so far underestimated function of the CD4+ T-cell / TAM axis in reconditioning the immunosuppressive tumor micro-milieu. Citation Format: David Eisel, Wolfram Osen, Krishna Das, Franziska Marie-Claire Hoerhold, Rainer Konig, Stefan B. Eichmuller. Cognate interaction with CD4+ T-cells instructs M2-like macrophages to acquire M1-like phenotype [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 A067.
目前的肿瘤免疫治疗方法基于检查点抑制剂的应用,如针对PD-1/PD-L1和CTLA-4的单克隆抗体,或使用体外扩增til或表达重组t细胞受体(tcr)或嵌合抗原受体(CARs)的转基因自体t细胞的过继t细胞治疗。然而,这些治疗策略的成功往往受到肿瘤中积累的各种抑制性免疫细胞类型的限制。特别是肿瘤相关巨噬细胞(tumor-associated macrophages, tam)参与了免疫抑制性肿瘤微环境,所谓的m2样巨噬细胞通过分泌TGF-β、IL10等抑制性细胞因子抑制T效应细胞的功能,促进调节性T细胞(regulatory T cells, Treg)的分化。另一方面,肿瘤抗原特异性CD4+ T效应细胞基本上可以维持各种肿瘤实体的抗肿瘤免疫反应。事实上,利用腹膜渗出细胞(PECs)作为巨噬细胞的来源,我们证明MHC II限制了卵白蛋白(OVA)特异性CD4+ t细胞和m2样巨噬细胞之间的相互作用,从而驱动M1极化。通过详细的基因和蛋白表达分析以及重极化巨噬细胞吞噬和吞噬活性的功能分析证实了这一点。此外,在一系列临床前实验中,通过基因表达分析和流式细胞术检测,将CD4+、OVA特异性的OT-II细胞过继转移到携带表达IAb-/-的OVA的C57BL/6小鼠体内,可导致瘤内m1样tam数量增加。此外,我们观察到,在B16F10-Ova肿瘤移植后,接受ova特异性CD4+ (OT-II)和CD8+ (OT-I)细胞联合治疗的小鼠的生存率显著提高,并且在一些拒绝肿瘤细胞的小鼠中,在随后的再次攻击中也有完全应答。虽然这种过继性转移实验的抗肿瘤作用已经被描述过,但我们现在为特异性CD4+细胞对共转移的CD8+细胞的支持作用提供了一种可能的解释。综上所述,本报告中描述的CD4+ t细胞对m2样巨噬细胞的指导性影响表明,CD4+ t细胞/ TAM轴在修复免疫抑制性肿瘤微环境中的功能迄今被低估。引用格式:David Eisel, Wolfram Osen, Krishna Das, Franziska Marie-Claire Hoerhold, Rainer Konig, Stefan B. Eichmuller。与CD4+ t细胞的同源相互作用指导m2样巨噬细胞获得m1样表型[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 A067。
{"title":"Abstract A067: Cognate interaction with CD4+ T-cells instructs M2-like macrophages to acquire M1-like phenotype","authors":"D. Eisel, W. Osen, K. Das, Franziska Hoerhold, R. König, S. Eichmüller","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A067","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A067","url":null,"abstract":"Current tumor immunotherapy approaches are based on application of checkpoint inhibitors, such as monoclonal antibodies against PD-1/PD-L1 and CTLA-4 or adoptive T-cell therapy using ex vivo expanded TILs or genetically modified autologous T-cells expressing recombinant T-cell receptors (TCRs) or chimeric antigen receptors (CARs). However, the success of these therapeutic strategies is often limited by various inhibitory immune cell types accumulating in the tumor. In particular, tumor-associated macrophages (TAMs) contribute to the immune suppressive tumor micro-milieu, since so-called M2-like macrophages suppress the function of T effector cells and promote the differentiation of regulatory T-cells (Treg) through secretion of inhibitory cytokines such as TGF-β and IL10. Tumor antigen-specific CD4+ T effector cells, on the other hand, can essentially sustain antitumoral immune responses as shown for various tumor entities. In fact, using peritoneal exudate cells (PECs) as source for macrophages we demonstrate that MHC II restricted interaction between ovalbumin (OVA) specific CD4+ T-cells and M2-like macrophages drives M1 polarization. This was confirmed by detailed gene and protein expression analyses as well as functional assays testing phagocytic and pinocytic activities of repolarized macrophages. Moreover, in a set of preclinical experiments, adoptive transfer of CD4+, OVA-specific OT-II cells into C57BL/6 mice bearing OVA expressing IAb-/- tumors resulted in increased intratumoral number of M1-like TAMs as determined by gene expression analysis and flow cytometry. Furthermore, we observed a significant survival benefit of mice treated with a combination of OVA-specific CD4+ (OT-II) and CD8+ (OT-I) cells after transplantation of B16F10-Ova tumors and a complete response in some mice that rejected the tumor cells also upon a later re-challenge. While the antitumoral effect of this adoptive transfer experiment has been already described, we now offer a possible explanation for the supportive effect of specific CD4+ cells on co-transferred CD8+ cells. Taken together, the instructive impact of CD4+ T-cells on M2-like macrophages described in this presentation points towards a so far underestimated function of the CD4+ T-cell / TAM axis in reconditioning the immunosuppressive tumor micro-milieu. Citation Format: David Eisel, Wolfram Osen, Krishna Das, Franziska Marie-Claire Hoerhold, Rainer Konig, Stefan B. Eichmuller. Cognate interaction with CD4+ T-cells instructs M2-like macrophages to acquire M1-like phenotype [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 A067.","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":"89703261","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 A085: High infiltration of NK cells expressing elevated LAG-3 in a subgroup of renal cell carcinoma patients 摘要/ Abstract A085:表达LAG-3升高的NK细胞在肾细胞癌患者亚组中高浸润
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A085
M. Lee, P. Järvinen, H. Nísen, O. Brück, Mette Ilander, S. Mustjoki, K. Anna
Background: Renal cell carcinoma (RCC) is considered one of the most immunogenic cancers with the highest number of indel mutations, frequent infiltration of T-cells, presence of many antigen-specific T-cell clones, and high immuno-oncologic (IO) sensitivity. Since less is known about natural killer (NK) cells in RCC, our primary aim was to investigate the intratumoral phenotype of NK cells as well as further assess the overall immune landscape of the tumors, peripheral blood (PB) and adjacent healthy kidney tissue, which may be critical for patient prognosis and predictions to targeted immunotherapies. Methods: We used multi-parameter flow cytometry together with a comprehensive immunostaining panel containing a total of 56 fundamental markers to cancer immunology to immunophenotype the tumor, adjacent healthy kidney tissue and presurgical peripheral blood (PB) samples from 31 RCC patients who underwent partial or radical nephrectomies. To study the intratumoral T-cell clonalities, T-cell receptor beta (TCRβ) deep sequencing was carried out with eight tumors. Results: Using hierarchical clustering (Spearman correlation distance and Ward linkage) and correlation analyses (Spearman correlation), we discovered that our patient cohort clustered into two distinct subgroups defined by a high (NKhigh, n=11; mean 29.7%) and low (NKlow, n=20; mean 9.4%) percentage of NK cells among the intratumoral lymphocyte population. Accordingly, the NKhigh subgroup had a lower percentage of T-cells (mean 36.9%) than the NKlow group (mean 65.7%), and overall, a significant negative correlation between T and NK cells was discovered. Our TCRβ sequencing results revealed a positive correlation between T-cell clonality and the intratumoral T-cell percentage, whereas the higher proportion of tumor NK cells associated with low T-cell clonality, possibly due to a polyclonal T-cell population. When we compared the expressions of the most clinically relevant IO markers (LAG-3 and PD-1) on the NK cells, LAG-3 was more expressed in the NKhigh group than in the NKlow group (21.3% vs 9.8%; p=0.08). In contrast, no differences were observed with PD-1. Clinical parameters such as tumor grade (Fuhrman), weight, size (diameter), the presence of necrosis, gender, or age of the patients did not differ between the two subgroups. To examine the overall immune landscape of RCC, we compared the cells from the tumor, PB, and healthy kidney tissue of seven patients. Our results showed that tumors have more NK cells compared to their corresponding T-cell-rich PB and healthy tissue counterparts, supporting our findings that some tumors accumulate NK cells. Compared to the adjacent healthy tissue, PD-1 and LAG-3 expressions were higher in the intratumoral CD8 cells. The expressions of PD-1 and LAG-3 on PB CD8+ T-cells or NK cells did not correlate with their intratumoral counterparts, whereas a positive correlation was found between the PB and tumor CD4+ T-cells for both LAG-3 and PD-1. Conclus
背景:肾细胞癌(RCC)被认为是免疫原性最强的癌症之一,其indel突变数量最多,t细胞浸润频繁,存在许多抗原特异性t细胞克隆,免疫肿瘤学(IO)敏感性高。由于对RCC中自然杀伤(NK)细胞的了解较少,我们的主要目的是研究NK细胞的瘤内表型,并进一步评估肿瘤、外周血(PB)和邻近健康肾组织的整体免疫景观,这可能对患者预后和靶向免疫治疗的预测至关重要。方法:采用多参数流式细胞术和包含56种肿瘤免疫学基本标志物的综合免疫染色面板,对31例接受部分或根治性肾切除术的RCC患者的肿瘤、邻近健康肾组织和手术前外周血(PB)样本进行免疫表型分析。为了研究肿瘤内t细胞的克隆性,我们对8个肿瘤进行了t细胞受体β (TCRβ)深度测序。结果:通过分层聚类(Spearman相关距离和Ward连锁)和相关分析(Spearman相关),我们发现我们的患者队列聚为两个不同的亚组,由高(NKhigh, n=11;平均29.7%)和低(NKlow, n=20;平均9.4%)NK细胞在瘤内淋巴细胞群中的百分比。因此,NKhigh亚组的T细胞百分比(平均36.9%)低于NKlow组(平均65.7%),总体而言,T细胞和NK细胞之间存在显著的负相关。我们的TCRβ测序结果显示,t细胞克隆性与肿瘤内t细胞百分比呈正相关,而较高的肿瘤NK细胞比例与较低的t细胞克隆性相关,可能是由于多克隆t细胞群。当我们比较与临床最相关的IO标志物(LAG-3和PD-1)在NK细胞上的表达时,LAG-3在NKhigh组的表达量高于NKlow组(21.3% vs 9.8%;p = 0.08)。相比之下,PD-1组无差异。临床参数,如肿瘤分级(Fuhrman)、体重、大小(直径)、坏死的存在、性别或患者的年龄在两个亚组之间没有差异。为了研究肾细胞癌的整体免疫景观,我们比较了7例患者的肿瘤、PB和健康肾组织的细胞。我们的研究结果表明,与相应的富含t细胞的PB和健康组织相比,肿瘤具有更多的NK细胞,这支持了我们的发现,即一些肿瘤积聚NK细胞。与邻近健康组织相比,PD-1和LAG-3在瘤内CD8细胞中的表达更高。PD-1和LAG-3在PB CD8+ t细胞或NK细胞上的表达与肿瘤内的表达不相关,而LAG-3和PD-1在PB和肿瘤CD4+ t细胞之间呈正相关。结论:我们的研究发现了两种不同的RCC肿瘤亚组,它们在临床先导分子中具有差异表达。这些结果表明,免疫分型RCC患者可以有效地帮助选择那些将从免疫检查点抑制疗法(如抗pd1和-LAG3)中获益最多的患者。接下来将评估多重免疫组织化学空间免疫谱和外显子组测序突变负荷的前瞻性分析,以找到为什么一些肿瘤是nk显性的答案,并期望进一步了解两种肿瘤类型之间的生物学差异。引文格式:Moon Hee Lee, Petrus Jarvinen, Harry Nisen, Oscar Bruck, Mette Ilander, Satu Mustjoki, Kreutzman Anna。肾细胞癌患者亚组中表达LAG-3升高的NK细胞高浸润[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 A085。
{"title":"Abstract A085: High infiltration of NK cells expressing elevated LAG-3 in a subgroup of renal cell carcinoma patients","authors":"M. Lee, P. Järvinen, H. Nísen, O. Brück, Mette Ilander, S. Mustjoki, K. Anna","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A085","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A085","url":null,"abstract":"Background: Renal cell carcinoma (RCC) is considered one of the most immunogenic cancers with the highest number of indel mutations, frequent infiltration of T-cells, presence of many antigen-specific T-cell clones, and high immuno-oncologic (IO) sensitivity. Since less is known about natural killer (NK) cells in RCC, our primary aim was to investigate the intratumoral phenotype of NK cells as well as further assess the overall immune landscape of the tumors, peripheral blood (PB) and adjacent healthy kidney tissue, which may be critical for patient prognosis and predictions to targeted immunotherapies. Methods: We used multi-parameter flow cytometry together with a comprehensive immunostaining panel containing a total of 56 fundamental markers to cancer immunology to immunophenotype the tumor, adjacent healthy kidney tissue and presurgical peripheral blood (PB) samples from 31 RCC patients who underwent partial or radical nephrectomies. To study the intratumoral T-cell clonalities, T-cell receptor beta (TCRβ) deep sequencing was carried out with eight tumors. Results: Using hierarchical clustering (Spearman correlation distance and Ward linkage) and correlation analyses (Spearman correlation), we discovered that our patient cohort clustered into two distinct subgroups defined by a high (NKhigh, n=11; mean 29.7%) and low (NKlow, n=20; mean 9.4%) percentage of NK cells among the intratumoral lymphocyte population. Accordingly, the NKhigh subgroup had a lower percentage of T-cells (mean 36.9%) than the NKlow group (mean 65.7%), and overall, a significant negative correlation between T and NK cells was discovered. Our TCRβ sequencing results revealed a positive correlation between T-cell clonality and the intratumoral T-cell percentage, whereas the higher proportion of tumor NK cells associated with low T-cell clonality, possibly due to a polyclonal T-cell population. When we compared the expressions of the most clinically relevant IO markers (LAG-3 and PD-1) on the NK cells, LAG-3 was more expressed in the NKhigh group than in the NKlow group (21.3% vs 9.8%; p=0.08). In contrast, no differences were observed with PD-1. Clinical parameters such as tumor grade (Fuhrman), weight, size (diameter), the presence of necrosis, gender, or age of the patients did not differ between the two subgroups. To examine the overall immune landscape of RCC, we compared the cells from the tumor, PB, and healthy kidney tissue of seven patients. Our results showed that tumors have more NK cells compared to their corresponding T-cell-rich PB and healthy tissue counterparts, supporting our findings that some tumors accumulate NK cells. Compared to the adjacent healthy tissue, PD-1 and LAG-3 expressions were higher in the intratumoral CD8 cells. The expressions of PD-1 and LAG-3 on PB CD8+ T-cells or NK cells did not correlate with their intratumoral counterparts, whereas a positive correlation was found between the PB and tumor CD4+ T-cells for both LAG-3 and PD-1. Conclus","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":"89846966","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: 2
Abstract A073: CODEXTM: A novel platform for spatially resolved deep antigen profiling of single cells in tissue samples 摘要:CODEXTM:一种用于组织样本中单细胞的空间分辨深度抗原谱分析的新平台
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A073
M. Gallina, Atri Choksi, N. Nikulina, Jaskirat Singh, G. Dakshinamoorthy, Joseph Kim, S. Mistry, J. Kennedy‐Darling
The tumor microenvironment (TME) comprises a multitude of cell types that collectively create an immunosuppressive environment, enabling tumor growth. Understanding the spatial organization of these tissues requires a technology that can identify the presence of multiple markers and correlate it to their specific spatial location within the same tissue. Characteristics of the TME including infiltrating immune cells, angiogenesis and the presence of other nonmalignant cells influence important phenomena like drug delivery, treatment effectiveness and, ultimately, clinical outcomes. Current methodologies for analyzing the spatial dimension of tissues, e.g., traditional immunofluorescence (IF) and immunohistochemistry (IHC), are limited to measuring a few parameters simultaneously, thereby restricting the number of phenotypes that can be identified. Conversely, single-cell technologies like mass cytometry and next-generation sequencing-based tools provide multiplexing capabilities, but at the expense of the associated spatial information. Akoya Biosciences, Inc., is commercializing CODEXTM (CO-Detection by indEXing), a multiparameteric imaging platform that allows the simultaneous detection and quantification of dozens of target epitopes in single cells within a single tissue section. This innovative platform is an end-to-end solution that comprises three components: 1) the CODEX fluidics instrument, 2) a suite of specialized CODEX reagents, and 3) an analysis pipeline. The CODEX workflow involves a barcoding system such that each antibody moiety is conjugated to a proprietary tag. Panels of CODEX antibodies are used to stain tissue specimens en masse in a single step. Staining data for sets of antibodies are revealed across iterative cycles using corresponding dye-labeled barcodes. The CODEX fluidics instrument integrates with existing microscope units for a fully automated data collection process. CODEX data are processed to achieve noise reduction and analyzed at the single-cell level through a segmentation algorithm based on nuclear staining.More than 80 antibodies have been validated on the CODEX platform for analysis of human and mouse fresh-frozen and FFPE tissues. Preliminary studies on fresh-frozen tissues with developed antibodies demonstrate an unprecedented capability for revealing the spatial correlation between a variety of target epitopes. These results demonstrate the enormous potential of the CODEX technology to identify spatial correlations in the TME through highly multiplexed detection with single-cell resolution. Citation Format: Maria Elena Gallina, Atri Choksi, Nadya Nikulina, Jaskirat Singh, Gajalakshmi Dakshinamoorthy, Joseph Kim, Sejal Mistry, Julia Kennedy-Darling. CODEXTM: A novel platform for spatially resolved deep antigen profiling of single cells in tissue samples [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3
肿瘤微环境(TME)包括多种细胞类型,它们共同创造免疫抑制环境,使肿瘤生长。了解这些组织的空间组织需要一种技术,可以识别多个标记的存在,并将其与同一组织中特定的空间位置相关联。TME的特征包括浸润性免疫细胞、血管生成和其他非恶性细胞的存在,影响药物输送、治疗效果以及最终的临床结果等重要现象。目前用于分析组织空间维度的方法,例如传统的免疫荧光(IF)和免疫组织化学(IHC),仅限于同时测量几个参数,从而限制了可以识别的表型的数量。相反,单细胞技术,如大规模细胞术和下一代基于测序的工具提供了多路复用功能,但以牺牲相关的空间信息为代价。Akoya Biosciences, Inc.正在商业化CODEXTM (CO-Detection by indEXing),这是一种多参数成像平台,可以同时检测和定量单个组织切片内单个细胞中的数十个目标表位。这一创新平台是一个端到端解决方案,由三个部分组成:1)CODEX流体仪器,2)一套专门的CODEX试剂,以及3)分析管道。CODEX工作流程涉及条形码系统,使每个抗体片段与专有标签结合。CODEX抗体组用于一次性对组织标本进行染色。通过使用相应的染料标记条形码,揭示了抗体组的染色数据。CODEX流体仪器与现有的显微镜单元集成,实现全自动数据收集过程。对CODEX数据进行处理以实现降噪,并通过基于核染色的分割算法在单细胞水平上进行分析。已有80多种抗体在CODEX平台上得到验证,用于分析人和小鼠新鲜冷冻组织和FFPE组织。对已开发抗体的新鲜冷冻组织的初步研究表明,在揭示各种靶表位之间的空间相关性方面具有前所未有的能力。这些结果证明了CODEX技术通过单细胞分辨率的高复用检测来识别TME中的空间相关性的巨大潜力。引文格式:Maria Elena Gallina, Atri Choksi, Nadya Nikulina, Jaskirat Singh, Gajalakshmi Dakshinamoorthy, Joseph Kim, Sejal Mistry, Julia Kennedy-Darling。CODEXTM:用于组织样本中单细胞的空间分解深度抗原谱分析的新平台[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 A073。
{"title":"Abstract A073: CODEXTM: A novel platform for spatially resolved deep antigen profiling of single cells in tissue samples","authors":"M. Gallina, Atri Choksi, N. Nikulina, Jaskirat Singh, G. Dakshinamoorthy, Joseph Kim, S. Mistry, J. Kennedy‐Darling","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A073","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A073","url":null,"abstract":"The tumor microenvironment (TME) comprises a multitude of cell types that collectively create an immunosuppressive environment, enabling tumor growth. Understanding the spatial organization of these tissues requires a technology that can identify the presence of multiple markers and correlate it to their specific spatial location within the same tissue. Characteristics of the TME including infiltrating immune cells, angiogenesis and the presence of other nonmalignant cells influence important phenomena like drug delivery, treatment effectiveness and, ultimately, clinical outcomes. Current methodologies for analyzing the spatial dimension of tissues, e.g., traditional immunofluorescence (IF) and immunohistochemistry (IHC), are limited to measuring a few parameters simultaneously, thereby restricting the number of phenotypes that can be identified. Conversely, single-cell technologies like mass cytometry and next-generation sequencing-based tools provide multiplexing capabilities, but at the expense of the associated spatial information. Akoya Biosciences, Inc., is commercializing CODEXTM (CO-Detection by indEXing), a multiparameteric imaging platform that allows the simultaneous detection and quantification of dozens of target epitopes in single cells within a single tissue section. This innovative platform is an end-to-end solution that comprises three components: 1) the CODEX fluidics instrument, 2) a suite of specialized CODEX reagents, and 3) an analysis pipeline. The CODEX workflow involves a barcoding system such that each antibody moiety is conjugated to a proprietary tag. Panels of CODEX antibodies are used to stain tissue specimens en masse in a single step. Staining data for sets of antibodies are revealed across iterative cycles using corresponding dye-labeled barcodes. The CODEX fluidics instrument integrates with existing microscope units for a fully automated data collection process. CODEX data are processed to achieve noise reduction and analyzed at the single-cell level through a segmentation algorithm based on nuclear staining.More than 80 antibodies have been validated on the CODEX platform for analysis of human and mouse fresh-frozen and FFPE tissues. Preliminary studies on fresh-frozen tissues with developed antibodies demonstrate an unprecedented capability for revealing the spatial correlation between a variety of target epitopes. These results demonstrate the enormous potential of the CODEX technology to identify spatial correlations in the TME through highly multiplexed detection with single-cell resolution. Citation Format: Maria Elena Gallina, Atri Choksi, Nadya Nikulina, Jaskirat Singh, Gajalakshmi Dakshinamoorthy, Joseph Kim, Sejal Mistry, Julia Kennedy-Darling. CODEXTM: A novel platform for spatially resolved deep antigen profiling of single cells in tissue samples [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3","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":"91482794","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 A077: Microenvironmental factors shape resistance patterns to immune checkpoint blockade 微环境因素影响免疫检查点阻断的抵抗模式
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A077
Sheng Gu, Xihao Hu, Xiaoqing Wang, Ziyi Li, Nicole Traugh, Xia Bu, Xiaofang Xing, G. Freeman, Myles A. Brown, Xiaole Shirley Liu
Immune checkpoint blockade (ICB) therapy has dramatically improved the prognosis of several types of cancer. However, only a small proportion of patients respond. Although multiple biomarkers/mechanisms of resistance to ICB have been identified, it remains elusive to what extent the cellular/molecular mechanisms contribute to the heterogeneity of ICB response. To this end, we applied clonal barcoding for lineage tracing of cancer cells following control IgG or ICB treatment in a transplantation mouse model. We identified significant clonal heterogeneity within cancer cells in response to ICB, suggestive of a minority of pre-existing ICB-resistant clones prior to treatment. Furthermore, counter-intuitively, ICB-responding tumors harbored a higher proportion of ICB-resistant clones than nonresponding tumors post-treatment, indicating that the tumor microenvironment might dictate the initial response to ICB. Integrated gene expression and immune repertoire analyses of the tumor microenvironment identified more T-cell and B cell infiltration in the ICB responders, and found that BCR class switch is associated with ICB response. Our study established a system to assess the contributions of cancer cell-intrinsic and microenvironmental factors in response to ICB treatment, and identified B cell infiltration and repertoire constitution as a novel biomarker for ICB response. Citation Format: Shengquing Stan Gu, Xihao Sherlock Hu, Xiaoqing Shawn Wang, Ziyi Li, Nicole Traugh, Xia Bu, Xiaofang Xing, Gordon Freeman, Myles Brown, Xiaole Shirley Liu. Microenvironmental factors shape resistance patterns to immune checkpoint blockade [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 A077.
免疫检查点阻断(ICB)疗法显著改善了几种类型癌症的预后。然而,只有一小部分患者有反应。虽然已经确定了多种对ICB耐药的生物标志物/机制,但细胞/分子机制在多大程度上促成了ICB反应的异质性仍然是难以捉摸的。为此,我们在移植小鼠模型中应用克隆条形码对对照IgG或ICB治疗后的癌细胞进行谱系追踪。我们发现了肿瘤细胞对ICB反应的显著克隆异质性,提示在治疗前存在少数ICB耐药克隆。此外,与直觉相反,治疗后对ICB有反应的肿瘤比无反应的肿瘤含有更高比例的ICB耐药克隆,这表明肿瘤微环境可能决定了对ICB的初始反应。整合基因表达和肿瘤微环境的免疫库分析发现,ICB应答者中有更多的t细胞和B细胞浸润,并发现BCR类转换与ICB应答有关。我们的研究建立了一个系统来评估癌细胞内在和微环境因素对ICB治疗反应的贡献,并确定了B细胞浸润和库构成作为ICB反应的一个新的生物标志物。引文格式:顾圣青、胡希豪、王晓青、李子怡、Nicole Traugh、布霞、邢晓芳、Gordon Freeman、Myles Brown、Xiaole Shirley Liu。微环境因素塑造免疫检查点封锁的抵抗模式[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 A077。
{"title":"Abstract A077: Microenvironmental factors shape resistance patterns to immune checkpoint blockade","authors":"Sheng Gu, Xihao Hu, Xiaoqing Wang, Ziyi Li, Nicole Traugh, Xia Bu, Xiaofang Xing, G. Freeman, Myles A. Brown, Xiaole Shirley Liu","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A077","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A077","url":null,"abstract":"Immune checkpoint blockade (ICB) therapy has dramatically improved the prognosis of several types of cancer. However, only a small proportion of patients respond. Although multiple biomarkers/mechanisms of resistance to ICB have been identified, it remains elusive to what extent the cellular/molecular mechanisms contribute to the heterogeneity of ICB response. To this end, we applied clonal barcoding for lineage tracing of cancer cells following control IgG or ICB treatment in a transplantation mouse model. We identified significant clonal heterogeneity within cancer cells in response to ICB, suggestive of a minority of pre-existing ICB-resistant clones prior to treatment. Furthermore, counter-intuitively, ICB-responding tumors harbored a higher proportion of ICB-resistant clones than nonresponding tumors post-treatment, indicating that the tumor microenvironment might dictate the initial response to ICB. Integrated gene expression and immune repertoire analyses of the tumor microenvironment identified more T-cell and B cell infiltration in the ICB responders, and found that BCR class switch is associated with ICB response. Our study established a system to assess the contributions of cancer cell-intrinsic and microenvironmental factors in response to ICB treatment, and identified B cell infiltration and repertoire constitution as a novel biomarker for ICB response. Citation Format: Shengquing Stan Gu, Xihao Sherlock Hu, Xiaoqing Shawn Wang, Ziyi Li, Nicole Traugh, Xia Bu, Xiaofang Xing, Gordon Freeman, Myles Brown, Xiaole Shirley Liu. Microenvironmental factors shape resistance patterns to immune checkpoint blockade [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 A077.","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":"90420433","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 A095: Discovering the immune profiles of a novel antifolate receptor alpha IgE antibody associated with monocyte-mediated antitumor functions 摘要:发现一种与单核细胞介导的抗肿瘤功能相关的新型抗叶酸受体α - IgE抗体的免疫谱
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A095
Mano Nakamura, H. J. Bax, J. Spicer, K. Lacy, S. Tsoka, D. Josephs, S. Karagiannis
Monoclonal antibodies are an established modality for the clinical management of many cancers. To date, all antibodies approved for cancer treatment are of the IgG class, and many are targeted towards non-solid tumors. We hypothesized that antibodies of the IgE class may be able to recruit and activate immune cells against tissue resident tumors based on very high affinity of IgE for cognate Fcϵ Receptors on potent effector cells such as masT-cells, monocytes and macrophages. A novel IgE-based antibody against folate receptor alpha (FRα) for ovarian carcinoma was developed and demonstrated to engender superior anti-tumor effector functions compared with those triggered by its IgG counterpart in different in vivo models of cancer. We have now translated this concept to a first-in-man clinical trial. We have previously reported that monocytes and macrophages are important IgE effector cells against tumors. However, the underlying mechanisms of how IgE exerts antitumor immunity by engaging these effector cells are insufficiently characterized. Here, we sought to investigate the immune profile signatures triggered by the engagement of tumor antigen-specific IgE with Fc-receptors on the surface of monocytes, a key immune effector cell recruited by IgE antibodies. In order to investigate the above aim, the following methods were utilized: Antibody dependenT-cell-mediated cytotoxicity (ADCC) and phagocytosis (ADCP) were measured by flow cytometry. Cell stimulation studies with cytokines or by cross-linking of IgE monoclonal antibodies on the surface of monocytes were conducted. Expression and secretion of immune mediators were assessed using quantitative-PCR and ELISA. Toxicity of cytokines towards immune and tumor cells were investigated by cell viability (MTS) assays. As a result, tumor antigen-specific IgE-mediated tumor cell cytotoxicity mediated by human monocytes correlated with increased levels of TNFα, MCP-1 and IL-10. Cross-linking of IgE, but not IgG, on the monocyte surface, triggered up-regulation of the proinflammatory mediator TNFα. TNFα stimulation of monocytes and of tumor cells triggered stimulation of MCP-1 by both cells. Neither cross-linking of IgE or IgG antibodies on tumor cells or monocytes, nor stimulation of these cells with TNFα or MCP-1, was sufficient to upregulate IL-10. However, IL-10 expression was induced by monocytic cells when stimulated by TNFα and MCP-1 in combination, and by stimulation with IL-10 in an autocrine manner. None of these stimulation conditions was sufficient to upregulate IL-10 in cancer cell lines of different origins. None of the three cytokines had direct cytotoxic effects towards immune or tumor cells. However, blockade of TNFα receptor on monocytes reduced ADCC and increased levels of the chemoattractant MCP-1 recruited monocytes in chemotaxis assays in vitro and macrophages in tumor lesions in a rat model of cancer treated with anti-tumor IgE. These findings suggest that IgE-dependent monocyte-med
单克隆抗体是许多癌症临床治疗的既定模式。迄今为止,所有被批准用于癌症治疗的抗体都是IgG类抗体,其中许多抗体针对非实体肿瘤。我们假设IgE类抗体可能能够招募和激活针对组织驻留肿瘤的免疫细胞,基于IgE对同源fcef受体在强效效应细胞(如mat细胞、单核细胞和巨噬细胞)上的非常高的亲和力。一种新的针对卵巢癌叶酸受体α (FRα)的基于ige的抗体被开发出来,并在不同的体内癌症模型中显示出比其IgG对应抗体更优越的抗肿瘤效应。我们现在已经将这一概念转化为首次人体临床试验。我们以前报道过单核细胞和巨噬细胞是重要的抗肿瘤IgE效应细胞。然而,IgE如何通过这些效应细胞发挥抗肿瘤免疫的潜在机制尚未充分表征。在这里,我们试图研究肿瘤抗原特异性IgE与单核细胞表面的fc受体结合引发的免疫谱特征,单核细胞是IgE抗体募集的关键免疫效应细胞。为了探讨上述目的,采用以下方法:流式细胞术检测抗体依赖细胞介导的细胞毒性(ADCC)和吞噬作用(ADCP)。用细胞因子或在单核细胞表面交联IgE单克隆抗体进行细胞刺激研究。采用定量pcr和ELISA检测免疫介质的表达和分泌情况。采用细胞活力(MTS)法研究细胞因子对免疫细胞和肿瘤细胞的毒性。因此,由人单核细胞介导的肿瘤抗原特异性ige介导的肿瘤细胞毒性与TNFα、MCP-1和IL-10水平升高相关。单核细胞表面的IgE而非IgG交联可引发促炎介质TNFα的上调。tnf - α对单核细胞和肿瘤细胞的刺激均触发MCP-1的刺激。无论是在肿瘤细胞或单核细胞上的IgE或IgG抗体交联,还是用TNFα或MCP-1刺激这些细胞,都不足以上调IL-10。然而,TNFα和MCP-1联合刺激和IL-10自分泌刺激均可诱导单核细胞表达IL-10。在不同来源的癌细胞系中,这些刺激条件都不足以上调IL-10。这三种细胞因子中没有一种对免疫细胞或肿瘤细胞具有直接的细胞毒性作用。然而,在体外趋化实验中,单核细胞上TNFα受体的阻断降低了ADCC,增加了趋化剂MCP-1募集单核细胞的水平,并在抗肿瘤IgE治疗的大鼠肿瘤模型中增加了肿瘤病变中的巨噬细胞。这些发现表明,依赖ige的单核细胞介导的肿瘤细胞毒性可触发TNFα、MCP-1和IL-10,后者通常与ige介导的抗寄生虫免疫功能相关。这种级联似乎是单核细胞和巨噬细胞向肿瘤细胞募集的主要原因,但可能不是ige介导的抗肿瘤细胞毒功能的唯一原因。确定参与IgE介导的单核细胞抗肿瘤机制的免疫谱可能有助于确定效应细胞激活的标记物,并支持IgE类作为一种新的免疫肿瘤学策略的发展。引文格式:Mano Nakamura, Heather J. Bax, James F. Spicer, Katie E. Lacy, Sophia Tsoka, Debra H. Josephs, Sophia Karagiannis。发现一种与单核细胞介导的抗肿瘤功能相关的新型抗叶酸受体- IgE抗体的免疫谱[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 A095。
{"title":"Abstract A095: Discovering the immune profiles of a novel antifolate receptor alpha IgE antibody associated with monocyte-mediated antitumor functions","authors":"Mano Nakamura, H. J. Bax, J. Spicer, K. Lacy, S. Tsoka, D. Josephs, S. Karagiannis","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A095","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A095","url":null,"abstract":"Monoclonal antibodies are an established modality for the clinical management of many cancers. To date, all antibodies approved for cancer treatment are of the IgG class, and many are targeted towards non-solid tumors. We hypothesized that antibodies of the IgE class may be able to recruit and activate immune cells against tissue resident tumors based on very high affinity of IgE for cognate Fcϵ Receptors on potent effector cells such as masT-cells, monocytes and macrophages. A novel IgE-based antibody against folate receptor alpha (FRα) for ovarian carcinoma was developed and demonstrated to engender superior anti-tumor effector functions compared with those triggered by its IgG counterpart in different in vivo models of cancer. We have now translated this concept to a first-in-man clinical trial. We have previously reported that monocytes and macrophages are important IgE effector cells against tumors. However, the underlying mechanisms of how IgE exerts antitumor immunity by engaging these effector cells are insufficiently characterized. Here, we sought to investigate the immune profile signatures triggered by the engagement of tumor antigen-specific IgE with Fc-receptors on the surface of monocytes, a key immune effector cell recruited by IgE antibodies. In order to investigate the above aim, the following methods were utilized: Antibody dependenT-cell-mediated cytotoxicity (ADCC) and phagocytosis (ADCP) were measured by flow cytometry. Cell stimulation studies with cytokines or by cross-linking of IgE monoclonal antibodies on the surface of monocytes were conducted. Expression and secretion of immune mediators were assessed using quantitative-PCR and ELISA. Toxicity of cytokines towards immune and tumor cells were investigated by cell viability (MTS) assays. As a result, tumor antigen-specific IgE-mediated tumor cell cytotoxicity mediated by human monocytes correlated with increased levels of TNFα, MCP-1 and IL-10. Cross-linking of IgE, but not IgG, on the monocyte surface, triggered up-regulation of the proinflammatory mediator TNFα. TNFα stimulation of monocytes and of tumor cells triggered stimulation of MCP-1 by both cells. Neither cross-linking of IgE or IgG antibodies on tumor cells or monocytes, nor stimulation of these cells with TNFα or MCP-1, was sufficient to upregulate IL-10. However, IL-10 expression was induced by monocytic cells when stimulated by TNFα and MCP-1 in combination, and by stimulation with IL-10 in an autocrine manner. None of these stimulation conditions was sufficient to upregulate IL-10 in cancer cell lines of different origins. None of the three cytokines had direct cytotoxic effects towards immune or tumor cells. However, blockade of TNFα receptor on monocytes reduced ADCC and increased levels of the chemoattractant MCP-1 recruited monocytes in chemotaxis assays in vitro and macrophages in tumor lesions in a rat model of cancer treated with anti-tumor IgE. These findings suggest that IgE-dependent monocyte-med","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":"87195424","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