首页 > 最新文献

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

英文 中文
Abstract A069: NIM15 blockade – A new stroma-targeting approach for the treatment of epithelial ovarian cancer 摘要:NIM15阻断-一种新的基质靶向治疗上皮性卵巢癌的方法
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A069
D. Ferber, M. Suarez-Carmona, F. Momburg, Marten Meyer, Rebecca Rothenheber, B. Lenoir, S. Schott, I. Zoernig, D. Jäger, N. Halama
Despite the immense research over the past decade in the cancer immunology field, which has led to several clinical trials and FDA and EMA approvals of biologicals for the reinvigoration of T-cell-mediated cancer cell killing in diverse tumor entities, the long-term survival of patients with advanced epithelial ovarian cancer is still devastating. These results therefore imply the need for a more intensive investigation of the tumor microenvironment in this cancer type in order to enhance disease outcome and improve the effectiveness of current immunotherapeutics. We herein show for the first time efficacy data of a novel treatment approach for the specific targeting of the stromal tumor compartment in a human tissue explant culture model of high-grade serous ovarian cancer. Antibody-mediated blockade of NIM15, a protein suspected to be predominantly expressed by tumor-associated macrophages and cancer-associated-fibroblasts in ovarian cancer, has the potential to polarize the immune landscape in a subset of patients from a stromal-dense and immunosuppressive one into a Th1-M1-supportive microenvironment, as measured by cytokine pattern analyses and semiautomated immune cell quantification. Abrogating the effects of secreted NIM15 unleashes in vitro proliferation of T-cell subsets and increases the production of cytokines and chemokines involved in innate and adaptive antitumor immune responses in our tissue culture explant model. In order to unravel the mechanistic relations behind the observed effects, we plan further experiments to prove whether these might be due to a disruption of the collagen-dense tumor stroma and a repolarization of the secretory profile of tumor-associated macrophages and fibroblasts. In summary, we hope to develop a pharmacologic tool that converts immune-depleted, “cold” cancer types into T-cell infiltrated ones and therewith provide a rationale for combination treatment approaches, like anti-PD1 blockade or adoptive cell transfer, to further ameliorate the so far poor response of metastasized, refractory ovarian cancer. Citation Format: Dyke Ferber, Meggy Suarez-Carmona, Frank Momburg, Marten Meyer, Rebecca Rothenheber, Benedicte M.A. Lenoir, Sarah Schott, Inka Zoernig, Dirk Jager, Niels Halama. NIM15 blockade – A new stroma-targeting approach for the treatment of epithelial ovarian 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 A069.
尽管在过去的十年中,癌症免疫学领域进行了大量的研究,导致了一些临床试验,FDA和EMA批准了用于在各种肿瘤实体中振兴t细胞介导的癌细胞杀伤的生物制剂,但晚期上皮性卵巢癌患者的长期生存仍然是毁灭性的。因此,这些结果意味着需要对这种癌症类型的肿瘤微环境进行更深入的研究,以增强疾病预后并提高当前免疫治疗的有效性。我们在此首次展示了一种新的治疗方法的疗效数据,该方法可以在高级别浆液性卵巢癌的人组织外植体培养模型中特异性靶向间质肿瘤室。抗体介导的NIM15阻断(一种被怀疑主要由卵巢癌肿瘤相关巨噬细胞和癌症相关成纤维细胞表达的蛋白质)有可能使一部分患者的免疫景观从基质密集和免疫抑制变为th1 - m1支持微环境,这是通过细胞因子模式分析和半自动免疫细胞定量测量的结果。在我们的组织培养外植体模型中,去除分泌的NIM15的作用可以释放t细胞亚群的体外增殖,并增加参与先天和适应性抗肿瘤免疫反应的细胞因子和趋化因子的产生。为了揭示观察到的效应背后的机制关系,我们计划进一步的实验来证明这些是否可能是由于胶原密集肿瘤基质的破坏和肿瘤相关巨噬细胞和成纤维细胞分泌谱的复极化。总之,我们希望开发一种药物工具,将免疫衰竭的“冷”癌症类型转化为t细胞浸润的癌症类型,从而为联合治疗方法提供理论依据,如抗pd1阻断或过继细胞转移,以进一步改善迄今为止转移性难治性卵巢癌的不良反应。引用格式:Dyke Ferber, Meggy Suarez-Carmona, Frank Momburg, Marten Meyer, Rebecca Rothenheber, Benedicte M.A. Lenoir, Sarah Schott, Inka Zoernig, Dirk Jager, Niels Halama。NIM15阻断-一种新的基质靶向治疗上皮性卵巢癌的方法[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 A069。
{"title":"Abstract A069: NIM15 blockade – A new stroma-targeting approach for the treatment of epithelial ovarian cancer","authors":"D. Ferber, M. Suarez-Carmona, F. Momburg, Marten Meyer, Rebecca Rothenheber, B. Lenoir, S. Schott, I. Zoernig, D. Jäger, N. Halama","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A069","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A069","url":null,"abstract":"Despite the immense research over the past decade in the cancer immunology field, which has led to several clinical trials and FDA and EMA approvals of biologicals for the reinvigoration of T-cell-mediated cancer cell killing in diverse tumor entities, the long-term survival of patients with advanced epithelial ovarian cancer is still devastating. These results therefore imply the need for a more intensive investigation of the tumor microenvironment in this cancer type in order to enhance disease outcome and improve the effectiveness of current immunotherapeutics. We herein show for the first time efficacy data of a novel treatment approach for the specific targeting of the stromal tumor compartment in a human tissue explant culture model of high-grade serous ovarian cancer. Antibody-mediated blockade of NIM15, a protein suspected to be predominantly expressed by tumor-associated macrophages and cancer-associated-fibroblasts in ovarian cancer, has the potential to polarize the immune landscape in a subset of patients from a stromal-dense and immunosuppressive one into a Th1-M1-supportive microenvironment, as measured by cytokine pattern analyses and semiautomated immune cell quantification. Abrogating the effects of secreted NIM15 unleashes in vitro proliferation of T-cell subsets and increases the production of cytokines and chemokines involved in innate and adaptive antitumor immune responses in our tissue culture explant model. In order to unravel the mechanistic relations behind the observed effects, we plan further experiments to prove whether these might be due to a disruption of the collagen-dense tumor stroma and a repolarization of the secretory profile of tumor-associated macrophages and fibroblasts. In summary, we hope to develop a pharmacologic tool that converts immune-depleted, “cold” cancer types into T-cell infiltrated ones and therewith provide a rationale for combination treatment approaches, like anti-PD1 blockade or adoptive cell transfer, to further ameliorate the so far poor response of metastasized, refractory ovarian cancer. Citation Format: Dyke Ferber, Meggy Suarez-Carmona, Frank Momburg, Marten Meyer, Rebecca Rothenheber, Benedicte M.A. Lenoir, Sarah Schott, Inka Zoernig, Dirk Jager, Niels Halama. NIM15 blockade – A new stroma-targeting approach for the treatment of epithelial ovarian 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 A069.","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":"74817347","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 A050: Intratumoral delivery of a novel STING agonist synergizes with checkpoint blockade to regress multifocal pancreatic cancer 摘要:肿瘤内递送一种新型STING激动剂与检查点阻断协同作用可治疗多灶性胰腺癌
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A050
C. Ager, M. D. Francesco, Philip D. Jones, M. Curran
Immunosuppressive myeloid populations including tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) are abundant within pancreatic adenocarcinoma (PDAC) tumors and play critical roles in constraining cytotoxic T-cell function in the tumor microenvironment. We hypothesized that intratumoral engagement of innate pathogen recognition receptors such as Stimulator of Interferon Genes (STING) could induce proinflammatory polarization of the myeloid stroma and liberate the antitumor T-cell response to regress refractory PDAC tumors in the presence of checkpoint blockade.We developed and characterized a novel cyclic dinucleotide (CDN) STING agonist IACS-8803, and found that 8803 activates downstream STING signaling in both human (THP-1) and murine (J774) myeloid reporter cells with over 10-fold greater potency than ML-RR-S2-CDA, the first-in-class CDN currently undergoing clinical evaluation (NCT02675439, NCT03172936). Intratumoral delivery of 8803 into subcutaneous B16 melanoma and PDAC tumors additionally revealed a greater capacity to induce tumor regression relative to ML-RR-S2-CDA. In order to evaluate the specific effects of 8803 on the phenotype and function of suppressive myeloid populations, we generated in vitro polarized human M2 macrophages and murine bone marrow-derived MDSC. Upon exposure to 8803, we observe downregulation of M2 markers CD163, LAP/TGF-β, and Arginase on human M2 macrophages, concomitant with upregulation of M1 markers CD86, CD80, and IL-6. Additionally, 8803-stimulated murine MDSC exhibit reduced T-cell suppressive capacity compared to unstimulated MDSC. In these studies, we consistently observe that the magnitude of phenotypic and functional repolarization by 8803 is superior to that of ML-RR-S2-CDA as well as other known CDN, 2’3’-cGAMP and c-di-GMP. Therefore, we describe IACS-8803 as a novel, highly potent STING agonist with the capacity to induce inflammatory repolarization in suppressive myeloid cells of both human and murine origin. We next investigated the capacity for intratumoral delivery of IACS-8803 to sensitize murine pancreatic cancer to checkpoint blockade and to mobilize systemic immunity against disseminated lesions. We utilized mT4-2D, a novel pancreatic cancer cell line from Kras+/LSL-G12D Tp53+/LSL-R172H Pdx1-Cre tumor organoids. We isolateda single cell clone of mT4-2D with reduced in vivo growth kinetics (termed mT4-LS), as well as a clone that maintains the aggressive nature of the parental line (termed mT4-LA). Mice bearing 10-day established orthotopic and subcutaneous mT4-LS tumors received standard regimens of αCTLA-4, αPD-1, or combined αCTLA-4/αPD-1 in the presence or absence of 8803 CDN injected into the orthotopic pancreatic tumor. We find single-agent treatment with 8803, αCTLA-4, αPD-1, or αCTLA-4/αPD-1 can cure 40-60% of mice of both orthotopic and subcutaneous tumors in this system; however, combining 8803 with checkpoint blockade completely eradicates both injec
包括肿瘤相关巨噬细胞(TAM)和骨髓源性抑制细胞(MDSC)在内的免疫抑制髓系细胞群在胰腺腺癌(PDAC)肿瘤中大量存在,并在肿瘤微环境中抑制细胞毒性t细胞功能发挥关键作用。我们假设肿瘤内天然病原体识别受体(如干扰素基因刺激因子(STING))的参与可以诱导髓样基质的促炎极化,并在检查点阻断存在的情况下释放抗肿瘤t细胞应答以消退难治性PDAC肿瘤。我们开发并鉴定了一种新型环二核苷酸(CDN) STING激动剂IACS-8803,并发现8803在人(THP-1)和鼠(J774)髓系报告细胞中激活下游STING信号,其效力比目前正在临床评估的同类首个CDN (NCT02675439, NCT03172936) ml - rr - s1 - cda高10倍以上。与ML-RR-S2-CDA相比,8803在皮下B16黑色素瘤和PDAC肿瘤中的瘤内递送也显示出更大的诱导肿瘤消退的能力。为了评估8803对抑制性骨髓群体表型和功能的特异性影响,我们在体外培养了极化的人M2巨噬细胞和小鼠骨髓来源的MDSC。暴露于8803后,我们观察到M2巨噬细胞中M2标记物CD163、LAP/TGF-β和精氨酸酶下调,同时M1标记物CD86、CD80和IL-6上调。此外,与未刺激的MDSC相比,8803刺激的小鼠MDSC表现出降低的t细胞抑制能力。在这些研究中,我们一致观察到8803在表型和功能上的复极化程度优于ML-RR-S2-CDA以及其他已知的CDN, 2 ' 3 ' -cGAMP和c-di-GMP。因此,我们将IACS-8803描述为一种新型的,高效的STING激动剂,具有诱导人类和小鼠来源的抑制性骨髓细胞炎症再极化的能力。接下来,我们研究了肿瘤内递送IACS-8803的能力,使小鼠胰腺癌对检查点阻断敏感,并动员全身免疫对抗弥散性病变。我们利用了一种来自Kras+/LSL-G12D Tp53+/LSL-R172H Pdx1-Cre肿瘤类器官的新型胰腺癌细胞系mT4-2D。我们分离出体内生长动力学降低的mT4-2D单细胞克隆(称为mT4-LS),以及保持亲本系侵袭性的克隆(称为mT4-LA)。移植10天的原位和皮下mT4-LS肿瘤小鼠接受αCTLA-4、αPD-1或αCTLA-4/αPD-1联合治疗方案,同时向原位胰腺肿瘤注射8803 CDN或不注射。我们发现8803、αCTLA-4、αPD-1或αCTLA-4/αPD-1单药治疗可治愈40-60%的小鼠原位和皮下肿瘤;然而,将8803与检查点阻断联合使用,可以完全根除所有小鼠的注射和远端mT4-LS肿瘤。我们使用高侵袭性和难治性mT4-LA模型重复了这些研究,发现与单独使用8803或αCTLA-4/αPD-1相比,胰腺内8803和全身αCTLA-4/αPD-1联合治疗可显著延长生存期(p=0.001, p=0.0086)。流式细胞术分析治疗后的mT4-LA肿瘤显示,联合治疗增强了注射和未注射病变处CD8 t细胞的细胞毒性潜能,并促进胰腺环境中的树突状细胞增殖。这些研究为寻求使用sting激活CDN作为一种局部方法使难治性PDAC肿瘤对检查点阻断免疫治疗敏感提供了临床前理论依据。引文格式:Casey R. Ager, Maria E. Di Francesco, Philip Jones, Michael A. Curran。肿瘤内递送一种新型STING激动剂与检查点阻断协同治疗多灶性胰腺癌[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 A050。
{"title":"Abstract A050: Intratumoral delivery of a novel STING agonist synergizes with checkpoint blockade to regress multifocal pancreatic cancer","authors":"C. Ager, M. D. Francesco, Philip D. Jones, M. Curran","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A050","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A050","url":null,"abstract":"Immunosuppressive myeloid populations including tumor-associated macrophages (TAM) and myeloid-derived suppressor cells (MDSC) are abundant within pancreatic adenocarcinoma (PDAC) tumors and play critical roles in constraining cytotoxic T-cell function in the tumor microenvironment. We hypothesized that intratumoral engagement of innate pathogen recognition receptors such as Stimulator of Interferon Genes (STING) could induce proinflammatory polarization of the myeloid stroma and liberate the antitumor T-cell response to regress refractory PDAC tumors in the presence of checkpoint blockade.We developed and characterized a novel cyclic dinucleotide (CDN) STING agonist IACS-8803, and found that 8803 activates downstream STING signaling in both human (THP-1) and murine (J774) myeloid reporter cells with over 10-fold greater potency than ML-RR-S2-CDA, the first-in-class CDN currently undergoing clinical evaluation (NCT02675439, NCT03172936). Intratumoral delivery of 8803 into subcutaneous B16 melanoma and PDAC tumors additionally revealed a greater capacity to induce tumor regression relative to ML-RR-S2-CDA. In order to evaluate the specific effects of 8803 on the phenotype and function of suppressive myeloid populations, we generated in vitro polarized human M2 macrophages and murine bone marrow-derived MDSC. Upon exposure to 8803, we observe downregulation of M2 markers CD163, LAP/TGF-β, and Arginase on human M2 macrophages, concomitant with upregulation of M1 markers CD86, CD80, and IL-6. Additionally, 8803-stimulated murine MDSC exhibit reduced T-cell suppressive capacity compared to unstimulated MDSC. In these studies, we consistently observe that the magnitude of phenotypic and functional repolarization by 8803 is superior to that of ML-RR-S2-CDA as well as other known CDN, 2’3’-cGAMP and c-di-GMP. Therefore, we describe IACS-8803 as a novel, highly potent STING agonist with the capacity to induce inflammatory repolarization in suppressive myeloid cells of both human and murine origin. We next investigated the capacity for intratumoral delivery of IACS-8803 to sensitize murine pancreatic cancer to checkpoint blockade and to mobilize systemic immunity against disseminated lesions. We utilized mT4-2D, a novel pancreatic cancer cell line from Kras+/LSL-G12D Tp53+/LSL-R172H Pdx1-Cre tumor organoids. We isolateda single cell clone of mT4-2D with reduced in vivo growth kinetics (termed mT4-LS), as well as a clone that maintains the aggressive nature of the parental line (termed mT4-LA). Mice bearing 10-day established orthotopic and subcutaneous mT4-LS tumors received standard regimens of αCTLA-4, αPD-1, or combined αCTLA-4/αPD-1 in the presence or absence of 8803 CDN injected into the orthotopic pancreatic tumor. We find single-agent treatment with 8803, αCTLA-4, αPD-1, or αCTLA-4/αPD-1 can cure 40-60% of mice of both orthotopic and subcutaneous tumors in this system; however, combining 8803 with checkpoint blockade completely eradicates both injec","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":"79559243","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 A066: Expanding insights into the colorectal cancer tumor proteome; unbiased protein profiling reveals multiple proteomic-based tumor subtypes [摘要]A066:扩大对结直肠癌肿瘤蛋白质组的认识;无偏蛋白质分析揭示了多种基于蛋白质组学的肿瘤亚型
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A066
N. Dupuis, J. Muntel, R. Bruderer, L. Reiter
Introduction: Recent approvals of microsatellite instability (MSI) or mismatch repair (MMR) testing, in addition to PD-L1 expression, expand the tools available to identify tumor characteristics that may help predict the likelihood of patient response to immunotherapy treatment. However, even in MSI positive subgroups, not all subjects achieve a durable response and research continues to identify tumor characteristics that further predict the likelihood of patient response. To support and advance this area of research, new tools are being developed that provide deeper and unbiased views of the tumor proteome. Here, we characterize the protein expression profiles of 95 colorectal cancer tumors (CRC) using SWATH acquisition mass spectrometry (SWATH MS) to further probe tumor phenotypic characteristics. Experimental Methods: FFPE colon tissue samples (10 healthy, 95 cancer) were obtained from commercial biobanks. Proteins were extracted from the tissue, processed to peptides with trypsin, and prepared for LC-MS analysis. Peptides for each sample were injected on a Triart C18 column (YMC) coupled to a NanoLC 425 system (SCIEX) using a 43min gradient at a flow rate of 5µl/min. The eluted peptides were then analyzed with a TripleTOF® 6600 system (SCIEX) operated in SWATH mode. Total run time per sample was 1 hour. Data were analyzed in Spectronaut Pulsar X (Biognosys) with a project specific library. All data were filtered with a 1% FDR on peptide and protein level. Results: Across all samples, >4,500 protein groups were quantified (approximately 3,600 per sample). Data analysis revealed a large number of proteins (~1,000) were differentially expressed in the cancer cohort. Consistent with increased tumor cell proliferation, proteins involved in protein translation were upregulated in the tumor samples. Unsupervised clustering of the data separated the healthy and the cancer cohort. Clustering also revealed three main proteomic subtypes within in the cancer cohort (A, B and C), which were largely distinguished by expression of cell adhesion proteins, including neuronal growth regulator 1 (NEGR1), a potential tumor suppressor. Interestingly, hepatocyte nuclear factor 4-alpha (HNF4A), a transcription factor which is known to be elevated in CRC, was only overexpressed in subtype B. Further analysis of key protein networks related to CRC treatment and immunotherapy development will be presented. Conclusions: High-throughput proteomic profiling of FFPE tissues using SWATH-MS enables the deepest phenotypic characterization of tumor tissue. Ultimately, analyses of this type will enable a functional understanding of interplay between the tumor microenvironment, expression of protein networks and response to immune-directed therapies. Citation Format: Nicholas Dupuis, Jan Muntel, Roland Bruderer, Lukas Reiter. Expanding insights into the colorectal cancer tumor proteome; unbiased protein profiling reveals multiple proteomic-based tumor subtypes [abstract]. In:
最近批准的微卫星不稳定性(MSI)或错配修复(MMR)测试,除了PD-L1表达,扩展了可用的工具来识别肿瘤特征,可能有助于预测患者对免疫治疗的反应的可能性。然而,即使在MSI阳性亚组中,也不是所有的受试者都能获得持久的反应,研究继续确定肿瘤特征,进一步预测患者反应的可能性。为了支持和推进这一领域的研究,人们正在开发新的工具,以提供对肿瘤蛋白质组更深入和公正的看法。在这里,我们使用SWATH获取质谱(SWATH MS)表征95个结直肠癌肿瘤(CRC)的蛋白质表达谱,以进一步探索肿瘤表型特征。实验方法:从商业生物库获得FFPE结肠组织样本(健康10例,癌95例)。从组织中提取蛋白质,用胰蛋白酶处理成多肽,并准备用于LC-MS分析。每个样品的多肽在Triart C18色谱柱(YMC)上以5 μ l/min的流速以43分钟的梯度注入到nanoc 425系统(SCIEX)上。然后用TripleTOF®6600系统(SCIEX)在SWATH模式下分析洗脱的肽。每个样品的总运行时间为1小时。数据在Spectronaut Pulsar X (Biognosys)中使用项目特定的库进行分析。所有数据在肽和蛋白质水平上用1%的FDR过滤。结果:在所有样品中,超过4,500个蛋白质组被量化(每个样品约3,600个)。数据分析显示,大量蛋白(约1000个)在癌症队列中存在差异表达。与肿瘤细胞增殖增加一致,参与蛋白质翻译的蛋白质在肿瘤样本中上调。数据的无监督聚类将健康组和癌症组分开。聚类还揭示了癌症队列中的三个主要蛋白质组亚型(A, B和C),它们在很大程度上通过细胞粘附蛋白的表达来区分,包括神经生长调节剂1 (NEGR1),一种潜在的肿瘤抑制因子。有趣的是,已知在结直肠癌中升高的转录因子肝细胞核因子4- α (HNF4A)仅在b亚型中过表达,将进一步分析与结直肠癌治疗和免疫疗法发展相关的关键蛋白网络。结论:利用SWATH-MS对FFPE组织进行高通量蛋白质组学分析,可以对肿瘤组织进行最深入的表型表征。最终,这种类型的分析将有助于了解肿瘤微环境、蛋白质网络表达和免疫定向治疗反应之间的相互作用。引文格式:Nicholas Dupuis, Jan Muntel, Roland Bruderer, Lukas Reiter。扩大对结直肠癌肿瘤蛋白质组的认识;无偏蛋白质分析揭示了多种基于蛋白质组学的肿瘤亚型[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 A066。
{"title":"Abstract A066: Expanding insights into the colorectal cancer tumor proteome; unbiased protein profiling reveals multiple proteomic-based tumor subtypes","authors":"N. Dupuis, J. Muntel, R. Bruderer, L. Reiter","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A066","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A066","url":null,"abstract":"Introduction: Recent approvals of microsatellite instability (MSI) or mismatch repair (MMR) testing, in addition to PD-L1 expression, expand the tools available to identify tumor characteristics that may help predict the likelihood of patient response to immunotherapy treatment. However, even in MSI positive subgroups, not all subjects achieve a durable response and research continues to identify tumor characteristics that further predict the likelihood of patient response. To support and advance this area of research, new tools are being developed that provide deeper and unbiased views of the tumor proteome. Here, we characterize the protein expression profiles of 95 colorectal cancer tumors (CRC) using SWATH acquisition mass spectrometry (SWATH MS) to further probe tumor phenotypic characteristics. Experimental Methods: FFPE colon tissue samples (10 healthy, 95 cancer) were obtained from commercial biobanks. Proteins were extracted from the tissue, processed to peptides with trypsin, and prepared for LC-MS analysis. Peptides for each sample were injected on a Triart C18 column (YMC) coupled to a NanoLC 425 system (SCIEX) using a 43min gradient at a flow rate of 5µl/min. The eluted peptides were then analyzed with a TripleTOF® 6600 system (SCIEX) operated in SWATH mode. Total run time per sample was 1 hour. Data were analyzed in Spectronaut Pulsar X (Biognosys) with a project specific library. All data were filtered with a 1% FDR on peptide and protein level. Results: Across all samples, >4,500 protein groups were quantified (approximately 3,600 per sample). Data analysis revealed a large number of proteins (~1,000) were differentially expressed in the cancer cohort. Consistent with increased tumor cell proliferation, proteins involved in protein translation were upregulated in the tumor samples. Unsupervised clustering of the data separated the healthy and the cancer cohort. Clustering also revealed three main proteomic subtypes within in the cancer cohort (A, B and C), which were largely distinguished by expression of cell adhesion proteins, including neuronal growth regulator 1 (NEGR1), a potential tumor suppressor. Interestingly, hepatocyte nuclear factor 4-alpha (HNF4A), a transcription factor which is known to be elevated in CRC, was only overexpressed in subtype B. Further analysis of key protein networks related to CRC treatment and immunotherapy development will be presented. Conclusions: High-throughput proteomic profiling of FFPE tissues using SWATH-MS enables the deepest phenotypic characterization of tumor tissue. Ultimately, analyses of this type will enable a functional understanding of interplay between the tumor microenvironment, expression of protein networks and response to immune-directed therapies. Citation Format: Nicholas Dupuis, Jan Muntel, Roland Bruderer, Lukas Reiter. Expanding insights into the colorectal cancer tumor proteome; unbiased protein profiling reveals multiple proteomic-based tumor subtypes [abstract]. In: ","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":"79750649","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 A109: Matrix metalloproteinase-2 stimulates Toll-like receptor-2 on melanoma cells to induce immunosuppressive inflammation in the tumor microenvironment 摘要A109:基质金属蛋白酶-2刺激黑色素瘤细胞上的toll样受体-2诱导肿瘤微环境中的免疫抑制性炎症
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A109
Mansi Saxena, Keerthi Caroline Sadanala, L. Muniz-Bongers, N. Bhardwaj
Extracellular proteinases, such as matrix metalloproteinases (MMPs), support tumor progression through modulation of the tumor microenvironment (TME). MMP-2, in particular, is overexpressed in several cancers and high MMP-2 levels are associated with advanced tumor stages, increased dissemination and poorer survival/prognosis. Our lab has previously demonstrated that upon antigenic stimulation, MMP-2-specific CD4+ T-cells, derived from patients with melanoma, secrete inflammatory TH2 cytokines. We subsequently showed that active MMP-2 drives the differentiation of TH2 responses by inhibiting IL-12 production and up-regulating OX40L expression on dendritic cells (DCs). We published our novel discovery identifying MMP-2 as a ligand for TLR-2 and showed that MMP-2 mediated TLR-2 stimulation lead to up-regulation OX40L on DCs (Cell Reports 2014). This is particularly interesting as TLR-2 stimulating adjuvants are being tested for immunotherapy. However, the full spectrum of how TLR-2 activation affects tumor cells or immune cells remains unclear.The main purpose of this study is to characterize the role of TLR-2-MMP-2 axis in shaping the TME through its influence on tumor cells and tumor infiltrating immune cells. Towards this end we performed RNA sequencing to identify genes induced in human DCs upon MMP-2 stimulation. One of these targets is an atypical member of the canonical NFκB family, IkappaBzeta (NFKBIZ or IκBζ). We show that MMP-2 secreted by melanoma cells upregulates IκBζ in DCs through TLR-2 and promotes secretion of Th2 and Th17 inducing cytokines. Furthermore, we screened several human melanoma cell lines for high and low MMP-2 and TLR-2 expression. CRISPR/Cas9 technology was used to stably knock out TLR-2, TLR-4 and MMP-2 in tumor cell lines. Early data indicates a role for tumor cell intrinsic MMP-2 in promoting secretion of protumorigenic cytokines and chemokines from tumor cells that support immune evasion and tumor growth, both constitutively and upon TLR-2 stimulation. Moreover, IκBζ was found to positively regulate MMP-2 dependent protumorigenic inflammation. In summary, we have identified a novel role for MMP-2 as a TLR-2 alarmin with particular emphasis on induction of atypical signaling modulator IκBζ and have uncovered a new role for MMP-2 in modulating tumor cell-induced inflammation. Taken together, our previous research and current data indicate that MMP-2 acts simultaneously as an endogenous T-cell differentiation "conditioner" and a tumor-associated antigen. Therefore, delving into MMP-2 signaling mechanisms in the TME holds a strong potential for discovering novel therapeutic options for treating melanoma. Citation Format: Mansi Saxena, Keerthi Caroline Sadanala, Luciana Rebiero Muniz-Bongers, Nina Bhardwaj. Matrix metalloproteinase-2 stimulates Toll-like receptor-2 on melanoma cells to induce immunosuppressive inflammation in the tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR Intern
细胞外蛋白酶,如基质金属蛋白酶(MMPs),通过调节肿瘤微环境(TME)支持肿瘤进展。特别是,MMP-2在几种癌症中过度表达,高MMP-2水平与肿瘤晚期、传播增加和生存/预后较差相关。我们的实验室之前已经证明,在抗原刺激下,来自黑色素瘤患者的mmp -2特异性CD4+ t细胞会分泌炎症性TH2细胞因子。我们随后发现,活性MMP-2通过抑制IL-12的产生和上调树突状细胞(dc)上OX40L的表达来驱动TH2反应的分化。我们发表了我们的新发现,确定MMP-2是TLR-2的配体,并表明MMP-2介导的TLR-2刺激导致dc上OX40L的上调(Cell Reports 2014)。这是特别有趣的,因为TLR-2刺激佐剂正在测试用于免疫治疗。然而,TLR-2激活如何影响肿瘤细胞或免疫细胞的全谱仍不清楚。本研究的主要目的是通过TLR-2-MMP-2轴对肿瘤细胞和肿瘤浸润免疫细胞的影响来表征其在形成TME中的作用。为此,我们进行了RNA测序,以鉴定在MMP-2刺激下人类dc中诱导的基因。其中一个靶点是典型NFκB家族的非典型成员,IkappaBzeta (NFKBIZ或IκBζ)。我们发现黑色素瘤细胞分泌的MMP-2通过TLR-2上调dc中的IκBζ,并促进Th2和Th17诱导细胞因子的分泌。此外,我们筛选了几种人类黑色素瘤细胞系,检测MMP-2和TLR-2的高表达和低表达。利用CRISPR/Cas9技术稳定敲除肿瘤细胞系中的TLR-2、TLR-4和MMP-2。早期数据表明,肿瘤细胞内在的MMP-2在促进肿瘤细胞分泌支持免疫逃避和肿瘤生长的致瘤细胞因子和趋化因子方面的作用,包括组成性和TLR-2刺激。此外,发现IκBζ正调节MMP-2依赖性蛋白原性炎症。总之,我们已经确定了MMP-2作为TLR-2报警蛋白的新作用,特别强调了非典型信号调节剂IκBζ的诱导,并揭示了MMP-2在调节肿瘤细胞诱导炎症中的新作用。综上所述,我们之前的研究和目前的数据表明,MMP-2同时作为内源性t细胞分化“调节剂”和肿瘤相关抗原。因此,深入研究TME中的MMP-2信号机制,对于发现治疗黑色素瘤的新治疗方案具有很大的潜力。引文格式:Mansi Saxena, Keerthi Caroline Sadanala, Luciana Rebiero Muniz-Bongers, Nina Bhardwaj。基质金属蛋白酶-2刺激黑色素瘤细胞上toll样受体-2在肿瘤微环境中诱导免疫抑制性炎症[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 A109。
{"title":"Abstract A109: Matrix metalloproteinase-2 stimulates Toll-like receptor-2 on melanoma cells to induce immunosuppressive inflammation in the tumor microenvironment","authors":"Mansi Saxena, Keerthi Caroline Sadanala, L. Muniz-Bongers, N. Bhardwaj","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A109","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A109","url":null,"abstract":"Extracellular proteinases, such as matrix metalloproteinases (MMPs), support tumor progression through modulation of the tumor microenvironment (TME). MMP-2, in particular, is overexpressed in several cancers and high MMP-2 levels are associated with advanced tumor stages, increased dissemination and poorer survival/prognosis. Our lab has previously demonstrated that upon antigenic stimulation, MMP-2-specific CD4+ T-cells, derived from patients with melanoma, secrete inflammatory TH2 cytokines. We subsequently showed that active MMP-2 drives the differentiation of TH2 responses by inhibiting IL-12 production and up-regulating OX40L expression on dendritic cells (DCs). We published our novel discovery identifying MMP-2 as a ligand for TLR-2 and showed that MMP-2 mediated TLR-2 stimulation lead to up-regulation OX40L on DCs (Cell Reports 2014). This is particularly interesting as TLR-2 stimulating adjuvants are being tested for immunotherapy. However, the full spectrum of how TLR-2 activation affects tumor cells or immune cells remains unclear.The main purpose of this study is to characterize the role of TLR-2-MMP-2 axis in shaping the TME through its influence on tumor cells and tumor infiltrating immune cells. Towards this end we performed RNA sequencing to identify genes induced in human DCs upon MMP-2 stimulation. One of these targets is an atypical member of the canonical NFκB family, IkappaBzeta (NFKBIZ or IκBζ). We show that MMP-2 secreted by melanoma cells upregulates IκBζ in DCs through TLR-2 and promotes secretion of Th2 and Th17 inducing cytokines. Furthermore, we screened several human melanoma cell lines for high and low MMP-2 and TLR-2 expression. CRISPR/Cas9 technology was used to stably knock out TLR-2, TLR-4 and MMP-2 in tumor cell lines. Early data indicates a role for tumor cell intrinsic MMP-2 in promoting secretion of protumorigenic cytokines and chemokines from tumor cells that support immune evasion and tumor growth, both constitutively and upon TLR-2 stimulation. Moreover, IκBζ was found to positively regulate MMP-2 dependent protumorigenic inflammation. In summary, we have identified a novel role for MMP-2 as a TLR-2 alarmin with particular emphasis on induction of atypical signaling modulator IκBζ and have uncovered a new role for MMP-2 in modulating tumor cell-induced inflammation. Taken together, our previous research and current data indicate that MMP-2 acts simultaneously as an endogenous T-cell differentiation \"conditioner\" and a tumor-associated antigen. Therefore, delving into MMP-2 signaling mechanisms in the TME holds a strong potential for discovering novel therapeutic options for treating melanoma. Citation Format: Mansi Saxena, Keerthi Caroline Sadanala, Luciana Rebiero Muniz-Bongers, Nina Bhardwaj. Matrix metalloproteinase-2 stimulates Toll-like receptor-2 on melanoma cells to induce immunosuppressive inflammation in the tumor microenvironment [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR Intern","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":"79326731","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 A097: Complement system mutations in cancer: Uncovering new relationships between tumor hypoxia and complement 摘要:肿瘤中补体系统突变:揭示肿瘤缺氧与补体之间的新关系
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A097
M. Olcina, N. Balanis, Ryan K. Kim, M. Thompson, T. Graeber, A. Giaccia
The complement system has been proposed to facilitate cancer hallmarks such as increased metastatic potential, proliferation and apoptosis evasion. Despite the association between complement and tumor progression, a detailed characterization of cancer genetic alterations in the complement system has not been performed to date. Here, we report a number of previously unappreciated mutations in complement system genes. Taken together as a pathway, mutations in complement genes occur at a relatively high frequency and across a number of cancer types. Notably, when grouping complement mutations into functionally relevant subgroups according to gene function, mutations and copy number alterations in genes within these subgroups are associated with changes in overall survival outcomes in a range of cancers. We use specific complement component mutations in colorectal cancer to uncover and experimentally validate crosstalk between complement and hypoxia, providing new associations between this innate immunity pathway and a prevalent component of the tumor microenvironment. Our data highlight the complex mechanism employed by cancers to manipulate the innate immune system and point to the potential use of complement system mutations in successful patient stratification into clinically and biologically relevant groups. Citation Format: Monica M. Olcina, Nikolas G. Balanis, Ryan K. Kim, Michael J. Thompson, Thomas G. Graeber, Amato J. Giaccia. Complement system mutations in cancer: Uncovering new relationships between tumor hypoxia and complement [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 A097.
补体系统已被提出促进癌症特征,如增加转移潜力,增殖和细胞凋亡逃避。尽管补体与肿瘤进展之间存在关联,但迄今为止还没有对补体系统中癌症遗传改变的详细描述。在这里,我们报告了补体系统基因中一些以前未被发现的突变。作为一种途径,补体基因的突变以相对较高的频率发生,并在许多癌症类型中发生。值得注意的是,当根据基因功能将补体突变分组为功能相关的亚组时,这些亚组中基因的突变和拷贝数改变与一系列癌症的总体生存结果的变化有关。我们在结直肠癌中使用特定的补体成分突变来揭示和实验验证补体和缺氧之间的串串,提供这种先天免疫途径与肿瘤微环境的普遍成分之间的新关联。我们的数据强调了癌症操纵先天免疫系统的复杂机制,并指出补体系统突变在成功地将患者分层为临床和生物学相关组中的潜在用途。引文格式:Monica M. Olcina, Nikolas G. Balanis, Ryan K. Kim, Michael J. Thompson, Thomas G. Graeber, Amato J. Giaccia。肿瘤中补体系统突变:揭示肿瘤缺氧与补体之间的新关系[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 A097。
{"title":"Abstract A097: Complement system mutations in cancer: Uncovering new relationships between tumor hypoxia and complement","authors":"M. Olcina, N. Balanis, Ryan K. Kim, M. Thompson, T. Graeber, A. Giaccia","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A097","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A097","url":null,"abstract":"The complement system has been proposed to facilitate cancer hallmarks such as increased metastatic potential, proliferation and apoptosis evasion. Despite the association between complement and tumor progression, a detailed characterization of cancer genetic alterations in the complement system has not been performed to date. Here, we report a number of previously unappreciated mutations in complement system genes. Taken together as a pathway, mutations in complement genes occur at a relatively high frequency and across a number of cancer types. Notably, when grouping complement mutations into functionally relevant subgroups according to gene function, mutations and copy number alterations in genes within these subgroups are associated with changes in overall survival outcomes in a range of cancers. We use specific complement component mutations in colorectal cancer to uncover and experimentally validate crosstalk between complement and hypoxia, providing new associations between this innate immunity pathway and a prevalent component of the tumor microenvironment. Our data highlight the complex mechanism employed by cancers to manipulate the innate immune system and point to the potential use of complement system mutations in successful patient stratification into clinically and biologically relevant groups. Citation Format: Monica M. Olcina, Nikolas G. Balanis, Ryan K. Kim, Michael J. Thompson, Thomas G. Graeber, Amato J. Giaccia. Complement system mutations in cancer: Uncovering new relationships between tumor hypoxia and complement [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 A097.","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":"89539039","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 A054: Tumor cell intrinsic factors dictate immune cell infiltration and response to immunotherapy 肿瘤细胞的内在因子决定了免疫细胞的浸润和对免疫治疗的反应
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A054
Katelyn T. Byrne, Jinyang Li, R. Vonderheide, B. Stanger
The establishment of immune heterogeneity in the tumor microenvironment (TME) is poorly understood, despite recent data that the success of immunotherapies is dictated by the immune environment of the tumor site. Pancreatic ductal adenocarcinoma (PDA) is characteristically devoid of CD8 T-cells and resistant to therapeutic intervention. However, a small subset of patients (15%) have tumors highly infiltrated by CD8 T-cells, correlating with improved overall survival. To better elucidate the determinants of immune heterogeneity in the PDA TME, we generated clones from spontaneous tumors harvested from KrasG12D+/-;Trp53R172H+/-;Pdx-1 Cre (KPC) mice, a genetically engineered mouse model of PDA. Using a panel of 17 tumor clones, we found the clones segregated in to two groups with differential immune cell infiltration upon implantation in congenic C57BL/6 mice. 7/17 tumor clones were categorized as “T-cell high,” with an immune infiltrate comprising CD8 T-cells and CD103+ dendritic cells (DCs). In contrast, the remaining 10 tumor clones were categorized as “T-cell low” lines, with the TME dominated by myeloid cells and macrophages, especially granulocytic myeloid-derived suppressor cells. Hypothesizing that increased T-cell infiltrate would render PDA sensitive to therapy, we treated two T-cell high and two T-cell low tumor clones with combination immunotherapy. Mice bearing T-cell high clones responded to therapy (7/7 and 4/7 mice cured) and formed protective memory responses against secondary tumor challenge, while none of the mice bearing T-cell low tumors responded to treatment (0/7 and 0/7 mice cured). At baseline, T-cell high tumors had similar proportions of functional CD8 T-cells as in T-cell low tumors. However, the proportion of activated CD44hiPD-1+ CD8 T-cells was significantly increased in T-cell high tumors (62.2% vs. 35.1% in T-cell low clones, p Citation Format: Katelyn T. Byrne, Jinyang Li, Robert H. Vonderheide, Ben Stanger. Tumor cell intrinsic factors dictate immune cell infiltration and response to 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 A054.
尽管最近的数据表明免疫疗法的成功取决于肿瘤部位的免疫环境,但人们对肿瘤微环境(TME)中免疫异质性的建立知之甚少。胰腺导管腺癌(PDA)的特点是缺乏CD8 t细胞,并且对治疗干预具有抗性。然而,一小部分患者(15%)的肿瘤被CD8 t细胞高度浸润,这与改善的总生存率相关。为了更好地阐明PDA TME中免疫异质性的决定因素,我们从KrasG12D+/-、Trp53R172H+/-、Pdx-1 Cre (KPC)小鼠(一种PDA基因工程小鼠模型)的自发肿瘤中获得克隆。使用17个肿瘤克隆,我们发现克隆在基因C57BL/6小鼠体内植入后分化为两组,免疫细胞浸润差异。7/17的肿瘤克隆被归类为“高t细胞”,免疫浸润包括CD8 t细胞和CD103+树突状细胞(dc)。相比之下,其余10个肿瘤克隆被归类为“低t细胞”系,TME以骨髓细胞和巨噬细胞为主,特别是粒细胞骨髓源性抑制细胞。假设增加的t细胞浸润会使PDA对治疗敏感,我们用联合免疫疗法治疗了两个t细胞高和两个t细胞低的肿瘤克隆。携带t细胞高克隆的小鼠对治疗有反应(7/7和4/7小鼠治愈),并形成针对继发性肿瘤攻击的保护性记忆反应,而携带t细胞低克隆的小鼠对治疗没有反应(0/7和0/7小鼠治愈)。在基线时,高t细胞肿瘤具有与低t细胞肿瘤相似的功能性CD8 t细胞比例。然而,激活CD44hiPD-1+ CD8 t细胞的比例在t细胞高克隆肿瘤中显著增加(62.2%比在t细胞低克隆中增加35.1%,p引文格式:Katelyn T. Byrne, Jinyang Li, Robert H. Vonderheide, Ben Stanger。肿瘤细胞的内在因素决定了免疫细胞的浸润和对免疫治疗的反应[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 A054。
{"title":"Abstract A054: Tumor cell intrinsic factors dictate immune cell infiltration and response to immunotherapy","authors":"Katelyn T. Byrne, Jinyang Li, R. Vonderheide, B. Stanger","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A054","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A054","url":null,"abstract":"The establishment of immune heterogeneity in the tumor microenvironment (TME) is poorly understood, despite recent data that the success of immunotherapies is dictated by the immune environment of the tumor site. Pancreatic ductal adenocarcinoma (PDA) is characteristically devoid of CD8 T-cells and resistant to therapeutic intervention. However, a small subset of patients (15%) have tumors highly infiltrated by CD8 T-cells, correlating with improved overall survival. To better elucidate the determinants of immune heterogeneity in the PDA TME, we generated clones from spontaneous tumors harvested from KrasG12D+/-;Trp53R172H+/-;Pdx-1 Cre (KPC) mice, a genetically engineered mouse model of PDA. Using a panel of 17 tumor clones, we found the clones segregated in to two groups with differential immune cell infiltration upon implantation in congenic C57BL/6 mice. 7/17 tumor clones were categorized as “T-cell high,” with an immune infiltrate comprising CD8 T-cells and CD103+ dendritic cells (DCs). In contrast, the remaining 10 tumor clones were categorized as “T-cell low” lines, with the TME dominated by myeloid cells and macrophages, especially granulocytic myeloid-derived suppressor cells. Hypothesizing that increased T-cell infiltrate would render PDA sensitive to therapy, we treated two T-cell high and two T-cell low tumor clones with combination immunotherapy. Mice bearing T-cell high clones responded to therapy (7/7 and 4/7 mice cured) and formed protective memory responses against secondary tumor challenge, while none of the mice bearing T-cell low tumors responded to treatment (0/7 and 0/7 mice cured). At baseline, T-cell high tumors had similar proportions of functional CD8 T-cells as in T-cell low tumors. However, the proportion of activated CD44hiPD-1+ CD8 T-cells was significantly increased in T-cell high tumors (62.2% vs. 35.1% in T-cell low clones, p Citation Format: Katelyn T. Byrne, Jinyang Li, Robert H. Vonderheide, Ben Stanger. Tumor cell intrinsic factors dictate immune cell infiltration and response to 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 A054.","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":"85228412","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 A060: Targeting glioma-macrophage interplay via LOX in PTEN-deficient glioblastoma 摘要:在pten缺陷胶质母细胞瘤中,通过LOX靶向胶质瘤-巨噬细胞相互作用
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A060
Peiwen Chen, Alan Wang, R. DePinho
Glioblastoma multiforme (GBM) is the most lethal form of brain cancer in adults. The median survival of GBM patients is only about one year after initial diagnosis. Genomic profiling has stratified GBM into various subgroups, which are driven by specific genetic alternations of core signaling pathways, including RTK/RAS/PI3K/PTEN, P53/ARF/MDM2 and RB/CDKN2A pathways. However, targeted therapies, such as therapy against EGFR, have failed in the clinic, and no effective therapeutic drugs are available to target tumor suppressors. A key reason for therapeutic failure is inter- and intra-tumoral cancer cell genetic instability and heterogeneity, resulting in aberrant activation of multiple signaling pathways within and across tumors. Stromal/immune cells in the tumor microenvironment (TME) are genetically stable, which not only play a pivotal role in GBM progression by affecting multiple cancer hallmarks, but can also be educated by cancer cells. However, whether and how the behavior and function of specific stromal/immune cells in the TME are regulated by cancer cell with specific genetic alterations in GBM remain relatively undefined. Utilizing a large scale of bioinformatic analysis in TCGA GBM patients, we revealed that genetic alteration (deletion/mutation) of PTEN in GBM patients specifically triggers immune response by promoting macrophage recruitment, without affecting macroglia and other immune cells. Using unbiased transcriptome profiling following functional validation, we identified that lysyl oxidase (LOX) is preferentially secreted by PTEN-deficient cancer cells. In vitro transwell migration assay and in vivo Matrigel Plug assay demonstrated that LOX is a potent macrophage chemoattractant. Transcriptome profiling following Gene Set Enrichment Analysis (GSEA) and functional validation demonstrated that activation of SRC and AKT signaling pathways drives LOX upregulation in PTEN-deficient cancer cells. Genetic and pharmacologic inhibition of LOX in PTEN-deficient cancer cells does not affect tumor cell proliferation in vitro, but markedly inhibits macrophage density and tumor growth in vivo. Using the bioinformatics analysis in clinical GBM samples, we demonstrated that LOX is enriched in GBM patients with higher macrophage density, and that these patients show lower survival. Together, our findings highlight the significance of PTEN-LOX axis in macrophage infiltration in GBM, and demonstrate a possibility of improving GBM treatment by targeting this axis-mediated macrophage recruitment. Citation Format: Peiwen Chen, Alan Wang, Ronald DePinho. Targeting glioma-macrophage interplay via LOX in PTEN-deficient glioblastoma [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 A060.
多形性胶质母细胞瘤(GBM)是成人中最致命的脑癌。GBM患者在初次诊断后的中位生存期仅为一年左右。基因组分析将GBM分为不同的亚组,这些亚组由核心信号通路的特定遗传改变驱动,包括RTK/RAS/PI3K/PTEN, P53/ARF/MDM2和RB/CDKN2A通路。然而,靶向治疗,如针对EGFR的治疗,在临床中失败了,并且没有有效的治疗药物可用于靶向肿瘤抑制因子。治疗失败的一个关键原因是肿瘤间和肿瘤内癌细胞遗传不稳定性和异质性,导致肿瘤内和肿瘤间多种信号通路的异常激活。肿瘤微环境(tumor microenvironment, TME)中的基质/免疫细胞在遗传上是稳定的,它们不仅通过影响多种癌症特征在GBM的进展中起关键作用,而且还可以被癌细胞教育。然而,在GBM中,特异性基质/免疫细胞的行为和功能是否以及如何受到具有特异性遗传改变的癌细胞的调节,仍然相对不明确。利用TCGA GBM患者的大规模生物信息学分析,我们揭示了GBM患者PTEN的遗传改变(缺失/突变)通过促进巨噬细胞募集特异性触发免疫反应,而不影响大胶质细胞和其他免疫细胞。通过功能验证后的无偏转录组分析,我们发现赖氨酸氧化酶(LOX)优先由pten缺陷癌细胞分泌。体外transwell迁移实验和体内Matrigel Plug实验表明,LOX是一种有效的巨噬细胞化学引诱剂。基因集富集分析(GSEA)和功能验证后的转录组分析表明,SRC和AKT信号通路的激活驱动pten缺陷癌细胞中LOX的上调。在pten缺失的癌细胞中,LOX的遗传和药理学抑制在体外不影响肿瘤细胞的增殖,但在体内明显抑制巨噬细胞密度和肿瘤生长。通过对临床GBM样本的生物信息学分析,我们发现巨噬细胞密度较高的GBM患者富含LOX,并且这些患者的生存率较低。总之,我们的研究结果强调了PTEN-LOX轴在GBM巨噬细胞浸润中的重要性,并证明了通过靶向这种轴介导的巨噬细胞募集来改善GBM治疗的可能性。引用格式:陈培文,Alan Wang, Ronald DePinho。通过LOX靶向pten缺陷胶质母细胞瘤中的胶质瘤-巨噬细胞相互作用[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;癌症免疫学杂志2019;7(2增刊):摘要nr A060。
{"title":"Abstract A060: Targeting glioma-macrophage interplay via LOX in PTEN-deficient glioblastoma","authors":"Peiwen Chen, Alan Wang, R. DePinho","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A060","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A060","url":null,"abstract":"Glioblastoma multiforme (GBM) is the most lethal form of brain cancer in adults. The median survival of GBM patients is only about one year after initial diagnosis. Genomic profiling has stratified GBM into various subgroups, which are driven by specific genetic alternations of core signaling pathways, including RTK/RAS/PI3K/PTEN, P53/ARF/MDM2 and RB/CDKN2A pathways. However, targeted therapies, such as therapy against EGFR, have failed in the clinic, and no effective therapeutic drugs are available to target tumor suppressors. A key reason for therapeutic failure is inter- and intra-tumoral cancer cell genetic instability and heterogeneity, resulting in aberrant activation of multiple signaling pathways within and across tumors. Stromal/immune cells in the tumor microenvironment (TME) are genetically stable, which not only play a pivotal role in GBM progression by affecting multiple cancer hallmarks, but can also be educated by cancer cells. However, whether and how the behavior and function of specific stromal/immune cells in the TME are regulated by cancer cell with specific genetic alterations in GBM remain relatively undefined. Utilizing a large scale of bioinformatic analysis in TCGA GBM patients, we revealed that genetic alteration (deletion/mutation) of PTEN in GBM patients specifically triggers immune response by promoting macrophage recruitment, without affecting macroglia and other immune cells. Using unbiased transcriptome profiling following functional validation, we identified that lysyl oxidase (LOX) is preferentially secreted by PTEN-deficient cancer cells. In vitro transwell migration assay and in vivo Matrigel Plug assay demonstrated that LOX is a potent macrophage chemoattractant. Transcriptome profiling following Gene Set Enrichment Analysis (GSEA) and functional validation demonstrated that activation of SRC and AKT signaling pathways drives LOX upregulation in PTEN-deficient cancer cells. Genetic and pharmacologic inhibition of LOX in PTEN-deficient cancer cells does not affect tumor cell proliferation in vitro, but markedly inhibits macrophage density and tumor growth in vivo. Using the bioinformatics analysis in clinical GBM samples, we demonstrated that LOX is enriched in GBM patients with higher macrophage density, and that these patients show lower survival. Together, our findings highlight the significance of PTEN-LOX axis in macrophage infiltration in GBM, and demonstrate a possibility of improving GBM treatment by targeting this axis-mediated macrophage recruitment. Citation Format: Peiwen Chen, Alan Wang, Ronald DePinho. Targeting glioma-macrophage interplay via LOX in PTEN-deficient glioblastoma [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 A060.","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":"91345935","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 A099: Using high-throughput phenotypic screening to identify therapeutic targets for the inhibition of myeloid-derived suppressor cells 摘要:利用高通量表型筛选确定髓源性抑制细胞抑制的治疗靶点
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-A099
E. Petrova, Sandra Schäffner, J. Pieck, C. Herhaus, F. Rippmann, Oliver Pöschke, L. Helming
Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature myeloid cells with immunosuppressive function, which inhibit the antitumor activity of T-cells and natural killer (NK) cells. MDSC number is greatly increased in tumor-bearing mice and in cancer patients, and in the clinic, MDSC accumulation is associated with cancer progression, recurrence, and poor response to chemo-, radio- and immunotherapies. The increasing evidence for the clinical significance of MDSC has triggered a strong interest in the therapeutic modulation of their function. To date, however, limited progress has been made in this direction, as a major challenge in the field remains the identification of suitable therapeutic targets for the development of novel drugs. Here, we describe a systematic approach in which a small-molecule high-throughput phenotypic screen was used to identify MDSC targets and pathways of therapeutic relevance. This screen was based on a validated in vitro mouse mononuclear MDSC (M-MDSC) model, in which hematopoietic progenitors, immortalized using a NUP98/HOXB4 transgene, were differentiated into immunosuppressive MDSC. Using this model, we developed a 384-well-based phenotypic screening assay, in which the suppressive effect of mouse M-MDSC on CD8+ T-cell proliferation and cytokine secretion was monitored. We screened a small molecule library, comprising 5000+ biologically active compounds with known target(s), and identified 116 compounds that potently disrupted MDSC suppression of T-cell function. With the help of chemoinformatics methods, reported target activities associated with the compounds were annotated, and a set of targets and pathways of potential significance for MDSC-driven immunosuppression was identified. Altogether, this work provides insight into the signaling nodes that could be of relevance for MDSC function, and offers a path forward for the therapeutic targeting of MDSC. Citation Format: Elissaveta Petrova, Sandra Schaffner, Jan-Carsten Pieck, Christian Herhaus, Friedrich Rippmann, Oliver Poschke, Laura Helming. Using high-throughput phenotypic screening to identify therapeutic targets for the inhibition of myeloid-derived suppressor cells [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 A099.
髓源性抑制细胞(myeloid -derived suppressor cells, MDSC)是一类具有免疫抑制功能的未成熟髓细胞,其抑制t细胞和自然杀伤细胞(natural killer, NK)的抗肿瘤活性。在荷瘤小鼠和癌症患者中,MDSC的数量大大增加,在临床上,MDSC的积累与癌症的进展、复发以及对化疗、放疗和免疫治疗的不良反应有关。越来越多的证据表明MDSC的临床意义已经引发了对其功能的治疗调节的强烈兴趣。然而,到目前为止,在这个方向上取得的进展有限,因为该领域的一个主要挑战仍然是确定合适的治疗靶点来开发新药。在这里,我们描述了一种系统的方法,其中使用小分子高通量表型筛选来识别MDSC靶点和治疗相关的途径。该筛选基于体外验证的小鼠单核MDSC (M-MDSC)模型,其中使用NUP98/HOXB4转基因永生化的造血祖细胞分化为免疫抑制MDSC。利用该模型,我们建立了一种基于384个孔的表型筛选实验,在该实验中,我们监测了小鼠M-MDSC对CD8+ t细胞增殖和细胞因子分泌的抑制作用。我们筛选了一个小分子文库,包括5000多种已知靶点的生物活性化合物,并鉴定出116种可能破坏MDSC抑制t细胞功能的化合物。在化学信息学方法的帮助下,对已报道的与这些化合物相关的靶点活性进行了注释,并确定了一组对mdsc驱动的免疫抑制具有潜在意义的靶点和途径。总之,这项工作提供了可能与MDSC功能相关的信号节点的见解,并为MDSC的治疗靶向提供了一条前进的道路。引文格式:Elissaveta Petrova, Sandra Schaffner, Jan-Carsten Pieck, Christian Herhaus, Friedrich Rippmann, Oliver Poschke, Laura Helming。利用高通量表型筛选确定髓源性抑制细胞抑制的治疗靶点[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 A099。
{"title":"Abstract A099: Using high-throughput phenotypic screening to identify therapeutic targets for the inhibition of myeloid-derived suppressor cells","authors":"E. Petrova, Sandra Schäffner, J. Pieck, C. Herhaus, F. Rippmann, Oliver Pöschke, L. Helming","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-A099","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-A099","url":null,"abstract":"Myeloid-derived suppressor cells (MDSC) are a heterogeneous population of immature myeloid cells with immunosuppressive function, which inhibit the antitumor activity of T-cells and natural killer (NK) cells. MDSC number is greatly increased in tumor-bearing mice and in cancer patients, and in the clinic, MDSC accumulation is associated with cancer progression, recurrence, and poor response to chemo-, radio- and immunotherapies. The increasing evidence for the clinical significance of MDSC has triggered a strong interest in the therapeutic modulation of their function. To date, however, limited progress has been made in this direction, as a major challenge in the field remains the identification of suitable therapeutic targets for the development of novel drugs. Here, we describe a systematic approach in which a small-molecule high-throughput phenotypic screen was used to identify MDSC targets and pathways of therapeutic relevance. This screen was based on a validated in vitro mouse mononuclear MDSC (M-MDSC) model, in which hematopoietic progenitors, immortalized using a NUP98/HOXB4 transgene, were differentiated into immunosuppressive MDSC. Using this model, we developed a 384-well-based phenotypic screening assay, in which the suppressive effect of mouse M-MDSC on CD8+ T-cell proliferation and cytokine secretion was monitored. We screened a small molecule library, comprising 5000+ biologically active compounds with known target(s), and identified 116 compounds that potently disrupted MDSC suppression of T-cell function. With the help of chemoinformatics methods, reported target activities associated with the compounds were annotated, and a set of targets and pathways of potential significance for MDSC-driven immunosuppression was identified. Altogether, this work provides insight into the signaling nodes that could be of relevance for MDSC function, and offers a path forward for the therapeutic targeting of MDSC. Citation Format: Elissaveta Petrova, Sandra Schaffner, Jan-Carsten Pieck, Christian Herhaus, Friedrich Rippmann, Oliver Poschke, Laura Helming. Using high-throughput phenotypic screening to identify therapeutic targets for the inhibition of myeloid-derived suppressor cells [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 A099.","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":"86185365","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 IA09: Harnessing natural and engineered properties of NKT cells for cancer immunotherapy 利用NKT细胞的自然和工程特性进行癌症免疫治疗
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA09
A. Heczey, Amy N. Courtney, Ho Ngai, Gengwen Tian, S. Robinson, G. Dotti, L. Metelitsa
Due to their natural anti-tumor properties and ability to preferentially localize to the neuroblastoma (NB) tumor site, Vα24-invariant natural killer T cells (NKTs) are promising candidate immune effectors for chimeric antigen receptor (CAR)-based immunotherapies targeting NB and other solid tumors. We have previously demonstrated that human NKTs expressing a CAR specific for ganglioside GD2 (CAR.GD2) mediated potent anti-tumor activity in a xenogeneic NB model in NOD/SCID/IL-2Rγnull mice. In comparison with CAR.GD2 T cells, CAR.GD2 NKTs localized more effectively to the tumor tissues and did not induce graft-versus-host disease (GvHD). Clinical development of NKT cell-based therapeutics requires overcoming two fundamental challenges: 1) the low frequency of NKTs in human peripheral blood, and 2) the limited ability of adoptively transfered NKTs/CAR-NKTs to persist in tumor-bearing animals. To address the first limitation, we have developed a cGMP protocol to isolate NKT cells from leukapheresis products using NKT-specific magnetic beads with the CliniMAX® system (Miltenyi). Isolated NKTs then undergo stimulation with CD1d-expressing antigen-presenting cells pulsed with α-galactosylceramide, retroviral transduction with a CAR-expressing vector, and rapid numeric expansion in cytokine-supplemented culture. This protocol routinely produces more than 109 CAR-NKTs within 17 days with average NKT cell purity and CAR expression of 96% and 54%, respectively. To overcome the second limitation, we incorporated the primary NKT homeostatic cytokine, IL-15, into the CAR.GD2 construct and evaluated its ability to enhance NKT cell in vivo persistence and therapeutic efficacy. Following adoptive transfer into mice bearing human NB xenografts, NKTs expressing CAR.GD2 were undetectable by three weeks whereas CAR.GD2/IL-15 NKTs underwent progressive expansion at sites of NB metastasis, reaching 32% of bone marrow cells two months after a single injection. Treatment with CAR.GD2/IL-15 NKTs resulted in a median survival of 70 days versus 48 days for mice treated with CAR.GD2 NKTs (P Citation Format: Andras Heczey, Amy N. Courtney, Ho Ngai, Gengwen Tian, Simon N. Robinson, Gianpietro Dotti, Leonid S. Metelitsa. Harnessing natural and engineered properties of NKT cells for 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 IA09.
由于其天然的抗肿瘤特性和优先定位于神经母细胞瘤(NB)肿瘤部位的能力,v α24不变性自然杀伤T细胞(nkt)是基于嵌合抗原受体(CAR)的针对NB和其他实体肿瘤的免疫疗法的有希望的候选免疫效应物。我们之前已经证明,在NOD/SCID/ il - 2r - γ缺失小鼠的异种NB模型中,表达CAR特异性神经节苷脂GD2 (CAR.GD2)的人nkt介导了有效的抗肿瘤活性。与CAR相比。GD2 T细胞,CARGD2 NKTs更有效地定位于肿瘤组织,不会诱导移植物抗宿主病(GvHD)。基于NKT细胞的治疗方法的临床发展需要克服两个基本挑战:1)NKT在人外周血中的低频率,以及2)过继性转移NKT / car -NKT在载瘤动物中的持续能力有限。为了解决第一个限制,我们开发了一种cGMP方案,使用CliniMAX®系统(Miltenyi)的NKT特异性磁珠从白细胞分离产品中分离NKT细胞。然后用α-半乳糖神经酰胺脉冲的表达cd1的抗原呈递细胞刺激分离的nkt,用car表达载体进行逆转录病毒转导,并在细胞因子补充培养中快速扩增。该方案通常在17天内产生超过109个CAR-NKT,平均NKT细胞纯度和CAR表达分别为96%和54%。为了克服第二个限制,我们将主要的NKT稳态细胞因子IL-15加入到CAR中。构建GD2并评价其增强NKT细胞体内持久性和治疗效果的能力。通过过继转移到携带人NB异种移植物的小鼠中,表达CAR的nkt。GD2在三周内检测不到,而CAR。GD2/IL-15 nkt在NB转移部位进行性扩增,单次注射2个月后达到骨髓细胞的32%。CAR治疗。GD2/IL-15 NKTs导致CAR治疗小鼠的中位生存期为70天,而CAR治疗小鼠为48天。GD2 NKTs (P引文格式:Andras Heczey, Amy N. Courtney, Ho Ngai, Gengwen Tian, Simon N. Robinson, Gianpietro Dotti, Leonid S. Metelitsa。利用NKT细胞的自然和工程特性进行癌症免疫治疗[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 - IA09。
{"title":"Abstract IA09: Harnessing natural and engineered properties of NKT cells for cancer immunotherapy","authors":"A. Heczey, Amy N. Courtney, Ho Ngai, Gengwen Tian, S. Robinson, G. Dotti, L. Metelitsa","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA09","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA09","url":null,"abstract":"Due to their natural anti-tumor properties and ability to preferentially localize to the neuroblastoma (NB) tumor site, Vα24-invariant natural killer T cells (NKTs) are promising candidate immune effectors for chimeric antigen receptor (CAR)-based immunotherapies targeting NB and other solid tumors. We have previously demonstrated that human NKTs expressing a CAR specific for ganglioside GD2 (CAR.GD2) mediated potent anti-tumor activity in a xenogeneic NB model in NOD/SCID/IL-2Rγnull mice. In comparison with CAR.GD2 T cells, CAR.GD2 NKTs localized more effectively to the tumor tissues and did not induce graft-versus-host disease (GvHD). Clinical development of NKT cell-based therapeutics requires overcoming two fundamental challenges: 1) the low frequency of NKTs in human peripheral blood, and 2) the limited ability of adoptively transfered NKTs/CAR-NKTs to persist in tumor-bearing animals. To address the first limitation, we have developed a cGMP protocol to isolate NKT cells from leukapheresis products using NKT-specific magnetic beads with the CliniMAX® system (Miltenyi). Isolated NKTs then undergo stimulation with CD1d-expressing antigen-presenting cells pulsed with α-galactosylceramide, retroviral transduction with a CAR-expressing vector, and rapid numeric expansion in cytokine-supplemented culture. This protocol routinely produces more than 109 CAR-NKTs within 17 days with average NKT cell purity and CAR expression of 96% and 54%, respectively. To overcome the second limitation, we incorporated the primary NKT homeostatic cytokine, IL-15, into the CAR.GD2 construct and evaluated its ability to enhance NKT cell in vivo persistence and therapeutic efficacy. Following adoptive transfer into mice bearing human NB xenografts, NKTs expressing CAR.GD2 were undetectable by three weeks whereas CAR.GD2/IL-15 NKTs underwent progressive expansion at sites of NB metastasis, reaching 32% of bone marrow cells two months after a single injection. Treatment with CAR.GD2/IL-15 NKTs resulted in a median survival of 70 days versus 48 days for mice treated with CAR.GD2 NKTs (P Citation Format: Andras Heczey, Amy N. Courtney, Ho Ngai, Gengwen Tian, Simon N. Robinson, Gianpietro Dotti, Leonid S. Metelitsa. Harnessing natural and engineered properties of NKT cells for 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 IA09.","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":"81423731","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 PR10: Reprogramming myeloid cells in TME with pepinemab, first-in-class semaphorin 4D MAb, enhances combination immunotherapy 摘要:用pepinemab(一流的信号素4D单抗)重编程TME中的骨髓细胞,增强联合免疫治疗
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR10
E. Evans, H. Bussler, C. Mallow, C. Reilly, Sebold Torno, Maria Scrivens, Alan P. Howell, Leslie Balch, J. Leonard, T. Fisher, C. Allen, Paúl E. Clavijo, Gregory Lesiniski, Christina Wu, S. Hu-Lieskovan, A. Ribas, Emily G Greengard, Ernest S. Smith, M. Zauderer
Purpose: Tumor growth inhibition by anti-semaphorin 4D (SEMA4D, CD100) blocking antibody is enhanced when combined with various immunotherapies in preclinical animal models. Immune checkpoint combinations with pepinemab (VX15/2503), a humanized anti-SEMA4D antibody, are currently being evaluated in several clinical trials. Methods: Expanded mechanistic studies in syngeneic preclinical models investigated the effect of SEMA4D blockade on immune contexture within the tumor microenvironment, as a single agent and in combination with various immunotherapy agents. Antitumor activity and immune response was characterized by immunohistochemistry, flow cytometry, functional assays, and cytokine, chemokine and gene expression analysis. Pepinemab (VX15/2503) is currently being evaluated as single agent or in combination with other immunotherapies in four clinical trials: (i) a phase 1b/2a combination trial of pepinemab with avelumab in NSCLC (CLASSICAL-Lung) (NCT03268057); (ii) a phase 1 combination trial of pepinemab with nivolumab or ipilimumab in melanoma patients who have progressed on any anti-PD-1/PD-L1 (NCT03373188); (iii) a neoadjuvant integrated biomarker trial in patients with metastatic colorectal and pancreatic cancers treated with pepinemab in combination with nivolumab or ipilimumab (NCT03373188); and (iv) a phase 1/2 trial of pepinemab in children with solid tumors and children and young adults with osteosarcoma (NCT03320330). Results: SEMA4D exerts multifaceted effects within the tumor microenvironment by creating a barrier at the tumor-stroma margin to restrict immune cell infiltration and promoting immunosuppressive activity of myeloid-derived cells. Blocking antibody to SEMA4D directly enhanced M1/M2 ratio and both reduced expression of chemokines that recruit MDSC and the ability of MDSC to suppress T-cell proliferation. Antibody blockade reduced the function of immunosuppressive myeloid and regulatory T-cells in the TME while simultaneously restoring the ability of dendritic cells and cytotoxic T-cells to migrate into the tumor in several syngeneic tumor models. Importantly, anti-SEMA4D MAb enhanced the activity of co-administered immunotherapies in murine colon, head and neck (HNSCC), and melanoma models. For example, anti-SEMA4D plus anti-CTLA-4 resulted in 100% survival and 90% complete tumor rejection (CR) (p Citation Format: Elizabeth E. Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Alan Howell, Leslie Balch, John E. Leonard, Terrence L. Fisher, Clint Allen, Paul Clavijo, Gregory Lesiniski, Christina Wu, Siwen Hu-Lieskovan, Antoni Ribas, Emily Greengard, Ernest S. Smith, Maurice Zauderer. Reprogramming myeloid cells in TME with pepinemab, first-in-class semaphorin 4D MAb, enhances combination 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. Philadelphi
目的:在临床前动物模型中,抗信号蛋白4D (SEMA4D, CD100)阻断抗体联合多种免疫疗法可增强肿瘤生长抑制。免疫检查点联合pepinemab (VX15/2503),一种人源抗sema4d抗体,目前正在几个临床试验中进行评估。方法:在同基因临床前模型中扩展机制研究SEMA4D阻断剂作为单一药物和与各种免疫治疗药物联合使用对肿瘤微环境内免疫环境的影响。通过免疫组织化学、流式细胞术、功能分析、细胞因子、趋化因子和基因表达分析来表征抗肿瘤活性和免疫应答。Pepinemab (VX15/2503)目前正在四项临床试验中作为单药或与其他免疫疗法联合进行评估:(i)在NSCLC (classic - lung) (NCT03268057)中,Pepinemab与avelumab的1b/2a期联合试验;(ii)在任何抗pd -1/PD-L1治疗进展的黑色素瘤患者中,pepinemab与nivolumab或ipilimumab的一期联合试验(NCT03373188);(iii)在pepinemab联合nivolumab或ipilimumab治疗的转移性结直肠癌和胰腺癌患者中进行的新辅助综合生物标志物试验(NCT03373188);(iv) pepinemab在患有实体瘤的儿童和患有骨肉瘤的儿童和年轻人中的1/2期试验(NCT03320330)。结果:SEMA4D通过在肿瘤-基质边缘形成屏障,限制免疫细胞浸润,促进髓源性细胞的免疫抑制活性,在肿瘤微环境中发挥多方面的作用。SEMA4D阻断抗体直接提高M1/M2比值,同时降低募集MDSC趋化因子的表达和MDSC抑制t细胞增殖的能力。在几种同基因肿瘤模型中,抗体阻断降低了TME中免疫抑制性骨髓细胞和调节性t细胞的功能,同时恢复了树突状细胞和细胞毒性t细胞迁移到肿瘤中的能力。重要的是,anti-SEMA4D MAb增强了小鼠结肠、头颈部(HNSCC)和黑色素瘤模型中共给药免疫疗法的活性。例如,抗sema4d +抗ctla -4可获得100%的存活率和90%的肿瘤完全排斥(CR) (p引用格式:Elizabeth E. Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Alan Howell, Leslie Balch, John E. Leonard, Terrence L. Fisher, Clint Allen, Paul Clavijo, Gregory Lesiniski, Christina Wu, Siwen ho - lieskovan, Antoni Ribas, Emily greenard, Ernest S. Smith, Maurice Zauderer)。用pepinemab(一流的信号素4D单抗)重编程TME中的骨髓细胞,增强联合免疫治疗[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 PR10。
{"title":"Abstract PR10: Reprogramming myeloid cells in TME with pepinemab, first-in-class semaphorin 4D MAb, enhances combination immunotherapy","authors":"E. Evans, H. Bussler, C. Mallow, C. Reilly, Sebold Torno, Maria Scrivens, Alan P. Howell, Leslie Balch, J. Leonard, T. Fisher, C. Allen, Paúl E. Clavijo, Gregory Lesiniski, Christina Wu, S. Hu-Lieskovan, A. Ribas, Emily G Greengard, Ernest S. Smith, M. Zauderer","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR10","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR10","url":null,"abstract":"Purpose: Tumor growth inhibition by anti-semaphorin 4D (SEMA4D, CD100) blocking antibody is enhanced when combined with various immunotherapies in preclinical animal models. Immune checkpoint combinations with pepinemab (VX15/2503), a humanized anti-SEMA4D antibody, are currently being evaluated in several clinical trials. Methods: Expanded mechanistic studies in syngeneic preclinical models investigated the effect of SEMA4D blockade on immune contexture within the tumor microenvironment, as a single agent and in combination with various immunotherapy agents. Antitumor activity and immune response was characterized by immunohistochemistry, flow cytometry, functional assays, and cytokine, chemokine and gene expression analysis. Pepinemab (VX15/2503) is currently being evaluated as single agent or in combination with other immunotherapies in four clinical trials: (i) a phase 1b/2a combination trial of pepinemab with avelumab in NSCLC (CLASSICAL-Lung) (NCT03268057); (ii) a phase 1 combination trial of pepinemab with nivolumab or ipilimumab in melanoma patients who have progressed on any anti-PD-1/PD-L1 (NCT03373188); (iii) a neoadjuvant integrated biomarker trial in patients with metastatic colorectal and pancreatic cancers treated with pepinemab in combination with nivolumab or ipilimumab (NCT03373188); and (iv) a phase 1/2 trial of pepinemab in children with solid tumors and children and young adults with osteosarcoma (NCT03320330). Results: SEMA4D exerts multifaceted effects within the tumor microenvironment by creating a barrier at the tumor-stroma margin to restrict immune cell infiltration and promoting immunosuppressive activity of myeloid-derived cells. Blocking antibody to SEMA4D directly enhanced M1/M2 ratio and both reduced expression of chemokines that recruit MDSC and the ability of MDSC to suppress T-cell proliferation. Antibody blockade reduced the function of immunosuppressive myeloid and regulatory T-cells in the TME while simultaneously restoring the ability of dendritic cells and cytotoxic T-cells to migrate into the tumor in several syngeneic tumor models. Importantly, anti-SEMA4D MAb enhanced the activity of co-administered immunotherapies in murine colon, head and neck (HNSCC), and melanoma models. For example, anti-SEMA4D plus anti-CTLA-4 resulted in 100% survival and 90% complete tumor rejection (CR) (p Citation Format: Elizabeth E. Evans, Holm Bussler, Crystal Mallow, Christine Reilly, Sebold Torno, Maria Scrivens, Alan Howell, Leslie Balch, John E. Leonard, Terrence L. Fisher, Clint Allen, Paul Clavijo, Gregory Lesiniski, Christina Wu, Siwen Hu-Lieskovan, Antoni Ribas, Emily Greengard, Ernest S. Smith, Maurice Zauderer. Reprogramming myeloid cells in TME with pepinemab, first-in-class semaphorin 4D MAb, enhances combination 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. Philadelphi","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":"74387619","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