首页 > 最新文献

Other Topics最新文献

英文 中文
Abstract B174: Co-expression of stimulators and inhibitors of T-cell activation in melanoma 摘要:黑色素瘤中t细胞激活刺激因子和抑制因子的共表达
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B174
R. Maniyar, R. Freund, A. Malhotra, Sanjukta Chakraborty, J. Geliebter, M. Wallack, R. Tiwari
Melanoma, one of the most aggressive skin cancers, has steadily been on the rise over the last three decades. With limited treatment options, the recent impetus in immuno-oncology drugs, especially checkpoint inhibitors, has changed the treatment landscape. While anti-CTLA4 and anti-PD1 demonstrated success in clinic, the search for additional targets is needed to enable potent comprehensive curative combination therapies. Our laboratory has characterized and screened five primary patient-derived melanoma cell lines, MEL-2, MEL-V, 3MM, KFM, and GLM-2. While immunomodulatory molecules are canonically present on antigen presenting cells and T-cells, increasing evidence suggests that they are not expressed in isolation. In an effort to identify immunomodulatory molecules expressed on our primary melanoma cells, a comprehensive RT-PCR screen of 29 co-inhibitory and co-stimulatory molecules was carried out. Several molecules including CD160, CD226, TIM1, HVEM, and BTLA were seen to be differentially expressed in melanoma cells compared to normal melanocytes at the mRNA level. Western blots and immunocytochemistry validated the differential expression of these molecules at the protein level. 50-80% of melanoma cases are positive for the BRAFV600E mutation, and are treated with a small molecule inhibitor of the mutated BRAF, vemurafenib (PLX4032). Treatment of these cells with PLX4032 led to an upregulation of transcription factors MITF and AP-1, as well as immunomodulatory molecules, CD160, CD226, TIM1, HVEM, and BTLA, a phenomenon seen only in cells positive for the BRAFV600E lesion. MITF and AP-1, owing to the binding sites present in the promoter regions of these molecules, can drive their expression upon treatment with PLX4032. These additional immune-regulatory molecules of T-cell activation and/or immune tolerance mechanisms are potential targets for a combination therapy with PLX4032 in melanoma patients positive for the BRAFV600E genetic lesion. Our future directions aim to elucidate the role of these molecules on the tumor cells and devise an effective combination with small-molecule inhibitors and immunotherapies. Citation Format: Rachana Maniyar, Robert Freund, Aryan Malhotra, Sanjukta Chakraborty, Jan Geliebter, Marc Wallack, Raj K. Tiwari. Co-expression of stimulators and inhibitors of T-cell activation in melanoma [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 B174.
黑色素瘤是最具侵袭性的皮肤癌之一,在过去的三十年里,它的发病率一直在稳步上升。在治疗选择有限的情况下,最近对免疫肿瘤药物,特别是检查点抑制剂的推动,已经改变了治疗前景。虽然抗ctla4和抗pd1在临床表现成功,但需要寻找其他靶点来实现有效的综合治疗联合疗法。我们的实验室已经鉴定并筛选了5种原发性患者源性黑色素瘤细胞系,MEL-2、MEL-V、3MM、KFM和GLM-2。虽然免疫调节分子通常存在于抗原提呈细胞和t细胞上,但越来越多的证据表明它们不是孤立表达的。为了鉴定在原发性黑色素瘤细胞上表达的免疫调节分子,我们对29种共抑制和共刺激分子进行了全面的RT-PCR筛选。包括CD160、CD226、TIM1、HVEM和BTLA在内的一些分子在mRNA水平上在黑色素瘤细胞中与正常黑色素细胞表达差异。Western blots和免疫细胞化学证实了这些分子在蛋白水平上的差异表达。50-80%的黑色素瘤病例BRAFV600E突变呈阳性,并使用突变BRAF的小分子抑制剂vemurafenib (PLX4032)治疗。用PLX4032处理这些细胞导致转录因子MITF和AP-1以及免疫调节分子CD160、CD226、TIM1、HVEM和BTLA的上调,这一现象仅在BRAFV600E病变阳性的细胞中可见。由于MITF和AP-1的结合位点存在于这些分子的启动子区域,因此在PLX4032处理后可以驱动它们的表达。这些额外的t细胞活化和/或免疫耐受机制的免疫调节分子是BRAFV600E遗传病变阳性的黑色素瘤患者与PLX4032联合治疗的潜在靶点。我们未来的方向是阐明这些分子在肿瘤细胞中的作用,并设计出一种有效的小分子抑制剂和免疫疗法的组合。引文格式:Rachana Maniyar, Robert Freund, Aryan Malhotra, Sanjukta Chakraborty, Jan Geliebter, Marc Wallack, Raj K. Tiwari。黑色素瘤中t细胞激活刺激因子和抑制因子的共表达[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 B174。
{"title":"Abstract B174: Co-expression of stimulators and inhibitors of T-cell activation in melanoma","authors":"R. Maniyar, R. Freund, A. Malhotra, Sanjukta Chakraborty, J. Geliebter, M. Wallack, R. Tiwari","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B174","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B174","url":null,"abstract":"Melanoma, one of the most aggressive skin cancers, has steadily been on the rise over the last three decades. With limited treatment options, the recent impetus in immuno-oncology drugs, especially checkpoint inhibitors, has changed the treatment landscape. While anti-CTLA4 and anti-PD1 demonstrated success in clinic, the search for additional targets is needed to enable potent comprehensive curative combination therapies. Our laboratory has characterized and screened five primary patient-derived melanoma cell lines, MEL-2, MEL-V, 3MM, KFM, and GLM-2. While immunomodulatory molecules are canonically present on antigen presenting cells and T-cells, increasing evidence suggests that they are not expressed in isolation. In an effort to identify immunomodulatory molecules expressed on our primary melanoma cells, a comprehensive RT-PCR screen of 29 co-inhibitory and co-stimulatory molecules was carried out. Several molecules including CD160, CD226, TIM1, HVEM, and BTLA were seen to be differentially expressed in melanoma cells compared to normal melanocytes at the mRNA level. Western blots and immunocytochemistry validated the differential expression of these molecules at the protein level. 50-80% of melanoma cases are positive for the BRAFV600E mutation, and are treated with a small molecule inhibitor of the mutated BRAF, vemurafenib (PLX4032). Treatment of these cells with PLX4032 led to an upregulation of transcription factors MITF and AP-1, as well as immunomodulatory molecules, CD160, CD226, TIM1, HVEM, and BTLA, a phenomenon seen only in cells positive for the BRAFV600E lesion. MITF and AP-1, owing to the binding sites present in the promoter regions of these molecules, can drive their expression upon treatment with PLX4032. These additional immune-regulatory molecules of T-cell activation and/or immune tolerance mechanisms are potential targets for a combination therapy with PLX4032 in melanoma patients positive for the BRAFV600E genetic lesion. Our future directions aim to elucidate the role of these molecules on the tumor cells and devise an effective combination with small-molecule inhibitors and immunotherapies. Citation Format: Rachana Maniyar, Robert Freund, Aryan Malhotra, Sanjukta Chakraborty, Jan Geliebter, Marc Wallack, Raj K. Tiwari. Co-expression of stimulators and inhibitors of T-cell activation in melanoma [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 B174.","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"1 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"130222538","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 B168: A Radiosensitivity gene signature and PD-L1 status predict clinical outcome of patients with glioblastoma multiforme in The Cancer Genome Atlas Dataset B168:肿瘤基因组图谱数据集中放射敏感性基因标记和PD-L1状态预测多形性胶质母细胞瘤患者的临床预后
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B168
I. Kim, B. Jang
Background: A combination of radiotherapy and immune checkpoint blockade, such as programmed death-1 (PD-1) or programmed death-ligand 1 (PD-L1) blockade, is being actively tested in clinical trial settings. Here, we tried to identify a subset of patients that could potentially benefit from this strategy using The Cancer Genome Atlas (TCGA) dataset for glioblastoma (GBM). Methods: A total of 399 cases were clustered into radiosensitive (RS) versus radioresistant (RR) groups based on radiosensitivity gene signature and were further stratified into PD-L1 high versus PD-L1 low groups according to the median expression of CD274 mRNA. Differential and integrated analyses with expression and methylation data were performed. CIBERSORT was used to enumerate the immune repertoire that resulted from transcriptome profiles. Results: We identified a subset of GBM patients, the “PD-L1 high-RR group” that had a worse clinical outcome compared to the other groups. In this group, differentially expressed genes (DEG) were highly enriched for an immune response and mapped into activation of PI3K-AKT and MAPK signaling pathways. Through integration of DEG and differentially methylated regions, kinase MAP3K8, which is involved in T-cell receptor signaling, was found to be upregulated, while BAI1, a factor that inhibits angiogenesis, was silenced. CIBERSORT showed that a higher infiltration of the immune repertoire, which included M2 macrophages and regulatory T-cells, contributed to an immunosuppressive tumor microenvironment. Conclusion: Looking at the results collectively, the “PD-L1-high-RR group” could potentially benefit from radiotherapy combined with PD-1/PD-L1 blockade and angiogenesis inhibition. Citation Format: Inah Kim, Bum Sup Jang. A Radiosensitivity gene signature and PD-L1 status predict clinical outcome of patients with glioblastoma multiforme in The Cancer Genome Atlas Dataset [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 B168.
背景:放疗和免疫检查点阻断的组合,如程序性死亡-1 (PD-1)或程序性死亡-配体1 (PD-L1)阻断,正在临床试验中积极进行测试。在这里,我们试图使用癌症基因组图谱(TCGA)胶质母细胞瘤(GBM)数据集确定可能从该策略中获益的患者子集。方法:根据放射敏感性基因特征将399例患者分为放射敏感组(RS)和放射耐药组(RR),并根据CD274 mRNA的中位表达水平进一步分为PD-L1高组和PD-L1低组。对表达和甲基化数据进行差异和综合分析。CIBERSORT用于列举由转录组谱产生的免疫库。结果:我们确定了GBM患者的一个亚群,即“PD-L1高rr组”,与其他组相比,其临床结果更差。在这一组中,差异表达基因(DEG)在免疫应答中高度富集,并被定位为PI3K-AKT和MAPK信号通路的激活。通过整合DEG和差异甲基化区域,发现参与t细胞受体信号传导的激酶MAP3K8上调,而抑制血管生成的因子BAI1则被沉默。CIBERSORT显示,包括M2巨噬细胞和调节性t细胞在内的免疫库的较高浸润有助于免疫抑制肿瘤微环境。结论:综合观察结果,“PD-L1-高rr组”可能从放疗联合PD-1/PD-L1阻断和血管生成抑制中获益。引文格式:Inah Kim, Bum Sup Jang。在癌症基因组图谱数据集中,放射敏感性基因标记和PD-L1状态预测多形性胶质母细胞瘤患者的临床结局。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 B168。
{"title":"Abstract B168: A Radiosensitivity gene signature and PD-L1 status predict clinical outcome of patients with glioblastoma multiforme in The Cancer Genome Atlas Dataset","authors":"I. Kim, B. Jang","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B168","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B168","url":null,"abstract":"Background: A combination of radiotherapy and immune checkpoint blockade, such as programmed death-1 (PD-1) or programmed death-ligand 1 (PD-L1) blockade, is being actively tested in clinical trial settings. Here, we tried to identify a subset of patients that could potentially benefit from this strategy using The Cancer Genome Atlas (TCGA) dataset for glioblastoma (GBM). Methods: A total of 399 cases were clustered into radiosensitive (RS) versus radioresistant (RR) groups based on radiosensitivity gene signature and were further stratified into PD-L1 high versus PD-L1 low groups according to the median expression of CD274 mRNA. Differential and integrated analyses with expression and methylation data were performed. CIBERSORT was used to enumerate the immune repertoire that resulted from transcriptome profiles. Results: We identified a subset of GBM patients, the “PD-L1 high-RR group” that had a worse clinical outcome compared to the other groups. In this group, differentially expressed genes (DEG) were highly enriched for an immune response and mapped into activation of PI3K-AKT and MAPK signaling pathways. Through integration of DEG and differentially methylated regions, kinase MAP3K8, which is involved in T-cell receptor signaling, was found to be upregulated, while BAI1, a factor that inhibits angiogenesis, was silenced. CIBERSORT showed that a higher infiltration of the immune repertoire, which included M2 macrophages and regulatory T-cells, contributed to an immunosuppressive tumor microenvironment. Conclusion: Looking at the results collectively, the “PD-L1-high-RR group” could potentially benefit from radiotherapy combined with PD-1/PD-L1 blockade and angiogenesis inhibition. Citation Format: Inah Kim, Bum Sup Jang. A Radiosensitivity gene signature and PD-L1 status predict clinical outcome of patients with glioblastoma multiforme in The Cancer Genome Atlas Dataset [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 B168.","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"28 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"130388756","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 B178: Development and preclinical efficacy characterization of a systemically administered multiple Toll-like receptor (TLR) agonist for antitumor immunotherapy B178:一种全身给药的toll样受体(TLR)激动剂的开发和临床前疗效表征
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B178
M. J. Newman
Background: The first cancer immunotherapy was developed in the 1890s by Dr. William Coley, who observed tumor regressions after injecting cancer patients with a heat-killed mixture of Gram-positive and negative pathogenic bacteria. Dr. Coley determined that the Gram-negative bacteria were the principal contributor to antitumor activity and that the product would likely be most effective when administered intravenously (i.v.), but was too toxic in this setting, limiting use to intramuscular and intratumoral administration. Coley’s toxins, as it was known, was credited with curing hundreds of late-stage cancer patients over 70 years. However, lack of knowledge regarding mechanism of action made it difficult to optimize or standardize, producing high variability. The use of multiple, local administration routes also likely contributed to variability in response, leading the FDA to refuse to grandfather in the product in 1963. We now know the mechanism of action of Coley’s toxins and the source of the i.v. toxicity. Gram-negative bacteria contain immune system danger signals, including multiple TLR agonists (activating TLRs 2, 4, 5 and 9), which directly and indirectly, via induction of cytokine and chemokine secretion, participate in the activation of most of the cellular mediators of innate and adaptive immune responses. Lipopolysaccharide (LPS), which activates TLR4, has been identified as a major contributor to both the antitumor activity and i.v. toxicity of Gram-negative bacteria. Decoy’s hypothesis is that significant reduction without complete elimination of LPS activity, in conjunction with killing and stabilization of nonpathogenic, Gram-negative bacteria, may produce a multiple TLR product that can safely and effectively induce antitumor immune responses via i.v. administration. Methods and Results: Nonpathogenic, Gram-negative E. coli were treated with polymyxin B and glutaraldehyde under conditions to kill and stabilize the cells, producing >90% reduction in LPS endotoxin activity and pyrogenicity. Endotoxin activity and pyrogenicity were quantified using Limulus amebocyte lysate (LAL) and in vivo rabbit assays. Bacterial integrity was assessed by electron and light microscopy. Antitumor activity was determined using standard syngeneic and xenograft tumor models. Decoy-treated bacteria exhibited a 3-fold reduction in acute in vivo toxicity relative to untreated bacteria. Surprisingly, induction of antitumor cytokine secretion by murine and human peripheral blood mononuclear cells (PBMCs) was not compromised, relative to untreated bacteria. Treatment with Decoy bacteria (i.v.) produced significant single-agent antitumor activity against orthotopic murine colorectal carcinoma and metastatic murine pancreatic carcinoma. Synergistic combination activity, including eradication of established tumors, with a therapeutic index of up to 10-fold, was observed in combination with IL-2 or low-dose cyclophosphamide (LDC) in murine colorectal carcino
背景:第一个癌症免疫疗法是在19世纪90年代由William Coley博士开发的,他在给癌症患者注射革兰氏阳性和阴性致病菌的热杀混合物后观察到肿瘤消退。Coley博士确定革兰氏阴性菌是抗肿瘤活性的主要因素,该产品在静脉注射(i.v.)时可能最有效,但在这种情况下毒性太大,限制了肌肉和肿瘤内给药的使用。众所周知,柯利毒素在70多年的时间里治愈了数百名晚期癌症患者。然而,由于缺乏对作用机理的了解,使其难以优化或标准化,产生了很大的可变性。使用多种地方给药途径也可能导致反应的可变性,导致FDA在1963年拒绝对该产品进行祖父处理。我们现在知道柯利毒素的作用机制和静脉注射毒性的来源。革兰氏阴性菌含有免疫系统危险信号,包括多种TLR激动剂(激活TLRs 2、4、5和9),它们通过诱导细胞因子和趋化因子的分泌,直接或间接地参与大多数先天和适应性免疫反应的细胞介质的激活。激活TLR4的脂多糖(LPS)已被确定为革兰氏阴性菌抗肿瘤活性和静脉毒性的主要因素。Decoy的假设是,在不完全消除LPS活性的情况下,显著降低LPS活性,同时杀死和稳定非致病性革兰氏阴性细菌,可能产生多重TLR产物,通过静脉注射可以安全有效地诱导抗肿瘤免疫反应。方法与结果:用多粘菌素B和戊二醛在杀死和稳定细胞的条件下处理非致病性革兰氏阴性大肠杆菌,使LPS内毒素活性和热原性降低90%以上。内毒素活性和热原性采用鲎试剂(LAL)和兔体内测定。通过电子显微镜和光镜检查细菌完整性。采用标准的同基因和异种移植肿瘤模型测定抗肿瘤活性。与未处理的细菌相比,诱骗处理的细菌表现出3倍的急性体内毒性降低。令人惊讶的是,与未经处理的细菌相比,小鼠和人外周血单核细胞(PBMCs)诱导的抗肿瘤细胞因子分泌并未受到损害。诱骗菌(诱饵菌)治疗对原位小鼠结直肠癌和转移性小鼠胰腺癌具有显著的单药抗肿瘤活性。建立协同组合活动,包括根除肿瘤,治疗指数的10倍,观察结合2或低剂量环磷酰胺(LDC)在小鼠结肠直肠癌癌模型,与LDC皮下(南),小鼠非霍奇金淋巴瘤(NHL)模型和LDC +利妥昔单抗在南卡罗来纳州,人类NHL模型。在转移性小鼠胰腺癌模型中,还观察到与低剂量非甾体抗炎药(NSAID)联合使用的协同抗肿瘤活性。此外,在sc小鼠肝细胞癌模型中,观察到NSAID联合肿瘤根除,并通过添加抗pd -1治疗增强肿瘤根除。最佳(80-100%)肿瘤根除被证明是由自然杀伤细胞(NK)、CD4+和CD8+ t细胞介导的。免疫记忆(80-100%和部分),由随后的肿瘤攻击的排斥决定,分别在免疫能力和先天条件下被证明。结论:我们已经开发出一种静脉注射的基于Coley毒素的多重TLR激动剂,它可以与多种现有的药物联合使用,以诱导治疗,抗肿瘤的免疫反应。引用格式:Michael J. Newman。一种全身给药的toll样受体(TLR)激动剂用于抗肿瘤免疫治疗的开发和临床前疗效表征[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 B178。
{"title":"Abstract B178: Development and preclinical efficacy characterization of a systemically administered multiple Toll-like receptor (TLR) agonist for antitumor immunotherapy","authors":"M. J. Newman","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B178","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B178","url":null,"abstract":"Background: The first cancer immunotherapy was developed in the 1890s by Dr. William Coley, who observed tumor regressions after injecting cancer patients with a heat-killed mixture of Gram-positive and negative pathogenic bacteria. Dr. Coley determined that the Gram-negative bacteria were the principal contributor to antitumor activity and that the product would likely be most effective when administered intravenously (i.v.), but was too toxic in this setting, limiting use to intramuscular and intratumoral administration. Coley’s toxins, as it was known, was credited with curing hundreds of late-stage cancer patients over 70 years. However, lack of knowledge regarding mechanism of action made it difficult to optimize or standardize, producing high variability. The use of multiple, local administration routes also likely contributed to variability in response, leading the FDA to refuse to grandfather in the product in 1963. We now know the mechanism of action of Coley’s toxins and the source of the i.v. toxicity. Gram-negative bacteria contain immune system danger signals, including multiple TLR agonists (activating TLRs 2, 4, 5 and 9), which directly and indirectly, via induction of cytokine and chemokine secretion, participate in the activation of most of the cellular mediators of innate and adaptive immune responses. Lipopolysaccharide (LPS), which activates TLR4, has been identified as a major contributor to both the antitumor activity and i.v. toxicity of Gram-negative bacteria. Decoy’s hypothesis is that significant reduction without complete elimination of LPS activity, in conjunction with killing and stabilization of nonpathogenic, Gram-negative bacteria, may produce a multiple TLR product that can safely and effectively induce antitumor immune responses via i.v. administration. Methods and Results: Nonpathogenic, Gram-negative E. coli were treated with polymyxin B and glutaraldehyde under conditions to kill and stabilize the cells, producing >90% reduction in LPS endotoxin activity and pyrogenicity. Endotoxin activity and pyrogenicity were quantified using Limulus amebocyte lysate (LAL) and in vivo rabbit assays. Bacterial integrity was assessed by electron and light microscopy. Antitumor activity was determined using standard syngeneic and xenograft tumor models. Decoy-treated bacteria exhibited a 3-fold reduction in acute in vivo toxicity relative to untreated bacteria. Surprisingly, induction of antitumor cytokine secretion by murine and human peripheral blood mononuclear cells (PBMCs) was not compromised, relative to untreated bacteria. Treatment with Decoy bacteria (i.v.) produced significant single-agent antitumor activity against orthotopic murine colorectal carcinoma and metastatic murine pancreatic carcinoma. Synergistic combination activity, including eradication of established tumors, with a therapeutic index of up to 10-fold, was observed in combination with IL-2 or low-dose cyclophosphamide (LDC) in murine colorectal carcino","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"41 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"133707469","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 B162: Developmental programming of long-term immunity of CD8 T-cells by perinatal glucocorticoids B162:围产期糖皮质激素对CD8 t细胞长期免疫的发育规划
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B162
J. Hong, B. Vaidyanathan, J. Cho, R. Medzhitov
Stress has been associated with various types of diseases including cancer. It was suggested that compromised antitumor immunity is often responsible for tumor progression during stress, which is caused by immunosuppressive glucocorticoids (GC), stress hormone, and sympathetic nervous system activation. Perinatal period is critical for immunity as the first major contact with the environment is made and this interaction can shape the immune system development. Epidemiologic studies found that early-life exposure to specific environment may have life-long impact on immunity, affecting the development of immune-related diseases such as inflammatory diseases, metabolic diseases, allergic diseases as well as cancer. Nevertheless, it is still largely unknown whether developmental programming of immunity exists due to the lack of mechanistic understanding on this subject. Since most of the environmental factors that are reported to cause later-life development of diseases are associated with stress, we adopted a model to directly test the effect of stress hormone. We introduced the in vivo mouse model of perinatal glucocorticoid receptor (GR) activation via dexamethasone (DEX) treatment in drinking water perinatally (embryonic day 7.5-postnatal day 1). Then, we analyzed the T-cell immunity and tumor susceptibility when the offspring became mature. We found that perinatal exposure of DEX permanently repressed the CD8 T-cell response. Consistently, accelerated tumor growth and decreased intra-tumor CD8 T-cell response were observed with perinatal DEX exposure in B16-F10 melanoma model. In OT-I T-cell receptor transgenic model, OT-I CD8 T-cells, from the mice with perinatal DEX exposure, showed suppressed ovalbumin (OVA) antigen-specific immune response as well as repressed antitumor immune response against OVA expressing E.G7 lymphoma. Furthermore, we found CD8 T-cell population expressing glucocorticoid-induced TNF receptor-related protein (GITR) was decreased with perinatal DEX exposure. Finally, we observed that endogenous corticosterone level as well as GR signaling pathway in T-cell was reduced with perinatal DEX exposure. We also identified that GR in T-cell was required for adequate CD8 T-cell activation and survival, suggesting that reduction of endogenous GC level with perinatal DEX was responsible for suppressed CD8 T-cell function. These results showed that perinatal GC exposure persistently programed the threshold for hypothalamus-pituitary-adrenal axis for regulating endogenous GC level, and that reduced systemic GC level elicited repressed CD8 T-cell activation and survival, leading to tumor susceptibility. Citation Format: Jun Young Hong, Bharat Vaidyanathan, Jen Young Cho, Ruslan Medzhitov. Developmental programming of long-term immunity of CD8 T-cells by perinatal glucocorticoids [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 20
压力与包括癌症在内的各种疾病有关。研究表明,抗肿瘤免疫功能受损通常是应激期间肿瘤进展的原因,这是由免疫抑制性糖皮质激素(GC)、应激激素和交感神经系统激活引起的。围产期对免疫至关重要,因为这是婴儿与环境的第一次主要接触,这种相互作用可以影响免疫系统的发育。流行病学研究发现,生命早期接触特定环境可能对免疫产生终身影响,影响炎症性疾病、代谢性疾病、过敏性疾病以及癌症等免疫相关疾病的发展。然而,由于缺乏对这一主题的机制理解,免疫的发育规划是否存在仍然在很大程度上是未知的。由于大多数被报道的导致疾病晚年发展的环境因素都与压力有关,我们采用了一个模型来直接测试压力激素的作用。采用地塞米松(dexamethasone, DEX)处理围生期(胚胎第7.5天至出生后第1天)饮用水,建立围生期糖皮质激素受体(GR)激活小鼠体内模型,分析后代成熟后的t细胞免疫和肿瘤易感性。我们发现,围产期暴露于DEX可永久性地抑制CD8 t细胞反应。在B16-F10黑色素瘤模型中,围产期暴露于DEX后,肿瘤生长加速,肿瘤内CD8 t细胞反应降低。在OT-I t细胞受体转基因模型中,围产期DEX暴露小鼠的OT-I CD8 t细胞表现出抑制的卵清蛋白(OVA)抗原特异性免疫反应,以及抑制的针对表达E.G7淋巴瘤的卵清蛋白抗原特异性免疫反应。此外,我们发现表达糖皮质激素诱导的TNF受体相关蛋白(GITR)的CD8 t细胞群随着围产期DEX暴露而减少。最后,我们观察到内源性皮质酮水平以及t细胞中的GR信号通路随着围产期DEX暴露而降低。我们还发现,t细胞中的GR是CD8 t细胞充分激活和存活所必需的,这表明围产期DEX降低内源性GC水平是抑制CD8 t细胞功能的原因。这些结果表明,围产期GC暴露持续编程下丘脑-垂体-肾上腺轴调节内源性GC水平的阈值,并且降低全身GC水平引起CD8 t细胞活化和存活的抑制,导致肿瘤易感性。引文格式:Jun Young Hong, Bharat Vaidyanathan, Jen Young Cho, Ruslan Medzhitov。围生期糖皮质激素对CD8 t细胞长期免疫的发育规划[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 B162。
{"title":"Abstract B162: Developmental programming of long-term immunity of CD8 T-cells by perinatal glucocorticoids","authors":"J. Hong, B. Vaidyanathan, J. Cho, R. Medzhitov","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B162","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B162","url":null,"abstract":"Stress has been associated with various types of diseases including cancer. It was suggested that compromised antitumor immunity is often responsible for tumor progression during stress, which is caused by immunosuppressive glucocorticoids (GC), stress hormone, and sympathetic nervous system activation. Perinatal period is critical for immunity as the first major contact with the environment is made and this interaction can shape the immune system development. Epidemiologic studies found that early-life exposure to specific environment may have life-long impact on immunity, affecting the development of immune-related diseases such as inflammatory diseases, metabolic diseases, allergic diseases as well as cancer. Nevertheless, it is still largely unknown whether developmental programming of immunity exists due to the lack of mechanistic understanding on this subject. Since most of the environmental factors that are reported to cause later-life development of diseases are associated with stress, we adopted a model to directly test the effect of stress hormone. We introduced the in vivo mouse model of perinatal glucocorticoid receptor (GR) activation via dexamethasone (DEX) treatment in drinking water perinatally (embryonic day 7.5-postnatal day 1). Then, we analyzed the T-cell immunity and tumor susceptibility when the offspring became mature. We found that perinatal exposure of DEX permanently repressed the CD8 T-cell response. Consistently, accelerated tumor growth and decreased intra-tumor CD8 T-cell response were observed with perinatal DEX exposure in B16-F10 melanoma model. In OT-I T-cell receptor transgenic model, OT-I CD8 T-cells, from the mice with perinatal DEX exposure, showed suppressed ovalbumin (OVA) antigen-specific immune response as well as repressed antitumor immune response against OVA expressing E.G7 lymphoma. Furthermore, we found CD8 T-cell population expressing glucocorticoid-induced TNF receptor-related protein (GITR) was decreased with perinatal DEX exposure. Finally, we observed that endogenous corticosterone level as well as GR signaling pathway in T-cell was reduced with perinatal DEX exposure. We also identified that GR in T-cell was required for adequate CD8 T-cell activation and survival, suggesting that reduction of endogenous GC level with perinatal DEX was responsible for suppressed CD8 T-cell function. These results showed that perinatal GC exposure persistently programed the threshold for hypothalamus-pituitary-adrenal axis for regulating endogenous GC level, and that reduced systemic GC level elicited repressed CD8 T-cell activation and survival, leading to tumor susceptibility. Citation Format: Jun Young Hong, Bharat Vaidyanathan, Jen Young Cho, Ruslan Medzhitov. Developmental programming of long-term immunity of CD8 T-cells by perinatal glucocorticoids [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 20","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"9 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"133952323","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 B177: Analysis of immunogenic cell death (ICD) induced in multiple myeloma cells B177:多发性骨髓瘤细胞诱导的免疫原性细胞死亡(ICD)分析
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B177
M. Matsushita, Sho Kashiwazaki, Satoshi Kamiko, Ryousuke Uozaki, D. Ichikawa, Y. Hattori
Background: Development of novel drugs, such as immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), or monoclonal antibodies, has prolonged survival of multiple myeloma (MM) patients. However, high-risk patients harboring del 17, t(14;17), or t(11;14) still have poor prognosis. Therefore, clarification of the drugs that could potentiate the effect of these treatments is necessary. Several drugs have been reported to cause cell surface expression of calreticulin (CRT), release of high mobility group box 1 (HMGB 1) and ATP, activating an immune response, which is called immunogenic cell death (ICD). However, there is almost no information on ICD in MM. The purpose of this study is to investigate whether anti-MM drugs can induce not only direct cell-killing effect but also immunomodulatory effects in high-risk MM cells. Methods: High-risk myeloma cell line, MUM24 was treated with dexamethasone, melphalan, lenalidomide, bortezomib, carfilzomib, and panobinostat. Expression of cell surface CRT and HMGB1 release were detected using flow cytometry and Western blotting, respectively. We then co-cultured DiD-labeled monocyte-derived dendritic cells with GFP-induced MUM24 treated with each drug and evaluated phagocytosis of DCs using fluorescence microscope. Results: After treatment with drugs, bortezomib and carfilzomib induced higher expression of CRT and release of HMGB1 in MUM24 compared with other drugs. We also observed that DCs could engulf MUM24 cells treated with carfilzomib. Conclusion: Our results suggest that PIs, such as bortezomib or carfilzomib, could not only kill MM cells but also induce ICD. These effects are expected to improve the prognosis of MM patients by evoking strong immune response against MM cells. Citation Format: Maiko Matsushita, Sho Kashiwazaki, Satoshi Kamiko, Ryo Uozaki, Daiju Ichikawa, Yutaka Hattori. Analysis of immunogenic cell death (ICD) induced in multiple myeloma 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 B177.
背景:新型药物的开发,如免疫调节药物(IMiDs)、蛋白酶体抑制剂(pi)或单克隆抗体,延长了多发性骨髓瘤(MM)患者的生存期。然而,携带del 17、t(14;17)、t(11;14)的高危患者预后仍较差。因此,澄清可能增强这些治疗效果的药物是必要的。一些药物引起细胞表面钙网蛋白(CRT)的表达、高迁移率组盒1 (hmgb1)和ATP的释放,激活免疫反应,称为免疫原性细胞死亡(ICD)。然而,关于MM中ICD的信息几乎没有。本研究的目的是探讨抗MM药物是否在高危MM细胞中不仅能诱导直接杀伤细胞作用,还能诱导免疫调节作用。方法:采用地塞米松、美法兰、来那度胺、硼替佐米、卡非佐米、帕比诺司他治疗高危骨髓瘤细胞株MUM24。流式细胞术检测细胞表面CRT的表达,Western blotting检测HMGB1的释放。然后,我们将did标记的单核细胞来源的树突状细胞与gfp诱导的MUM24共培养,并使用荧光显微镜观察dc的吞噬情况。结果:经药物治疗后,硼替佐米和卡非佐米在MUM24细胞中CRT的表达和HMGB1的释放均高于其他药物。我们还观察到DCs可以吞噬卡非佐米处理过的MUM24细胞。结论:硼替佐米、卡非佐米等pi不仅能杀死MM细胞,还能诱发ICD。这些作用有望通过唤起对MM细胞的强烈免疫反应来改善MM患者的预后。引文格式:Matsushita Maiko, Sho Kashiwazaki, Satoshi Kamiko, Ryo Uozaki, Daiju Ichikawa, Yutaka Hattori。多发性骨髓瘤细胞诱导的免疫原性细胞死亡(ICD)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 B177。
{"title":"Abstract B177: Analysis of immunogenic cell death (ICD) induced in multiple myeloma cells","authors":"M. Matsushita, Sho Kashiwazaki, Satoshi Kamiko, Ryousuke Uozaki, D. Ichikawa, Y. Hattori","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B177","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B177","url":null,"abstract":"Background: Development of novel drugs, such as immunomodulatory drugs (IMiDs), proteasome inhibitors (PIs), or monoclonal antibodies, has prolonged survival of multiple myeloma (MM) patients. However, high-risk patients harboring del 17, t(14;17), or t(11;14) still have poor prognosis. Therefore, clarification of the drugs that could potentiate the effect of these treatments is necessary. Several drugs have been reported to cause cell surface expression of calreticulin (CRT), release of high mobility group box 1 (HMGB 1) and ATP, activating an immune response, which is called immunogenic cell death (ICD). However, there is almost no information on ICD in MM. The purpose of this study is to investigate whether anti-MM drugs can induce not only direct cell-killing effect but also immunomodulatory effects in high-risk MM cells. Methods: High-risk myeloma cell line, MUM24 was treated with dexamethasone, melphalan, lenalidomide, bortezomib, carfilzomib, and panobinostat. Expression of cell surface CRT and HMGB1 release were detected using flow cytometry and Western blotting, respectively. We then co-cultured DiD-labeled monocyte-derived dendritic cells with GFP-induced MUM24 treated with each drug and evaluated phagocytosis of DCs using fluorescence microscope. Results: After treatment with drugs, bortezomib and carfilzomib induced higher expression of CRT and release of HMGB1 in MUM24 compared with other drugs. We also observed that DCs could engulf MUM24 cells treated with carfilzomib. Conclusion: Our results suggest that PIs, such as bortezomib or carfilzomib, could not only kill MM cells but also induce ICD. These effects are expected to improve the prognosis of MM patients by evoking strong immune response against MM cells. Citation Format: Maiko Matsushita, Sho Kashiwazaki, Satoshi Kamiko, Ryo Uozaki, Daiju Ichikawa, Yutaka Hattori. Analysis of immunogenic cell death (ICD) induced in multiple myeloma 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 B177.","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"92 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"133130842","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 B191: The impact of postoperative myeloid-derived suppressor cells on prognosis of gastric cancer patients 摘要B191:胃癌患者术后骨髓源性抑制细胞对预后的影响
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B191
S. Urakawa, H. Wada, M. Mori, Y. Doki
Background and Aim: Myeloid cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) and neutrophils have a suppressive role in the antitumor immunity. In gastric cancer (GC) patients, it has been reported that these myeloid cells in peripheral blood or tumor tissue were associated with their prognosis and increased after surgical treatments. Some reported that the tumor progression and frequent recurrence were caused by increased MDSC after surgical stress in a mouse model. However, in human GC patients, it is still unclear whether such an increase of myeloid cells after gastrectomy is related to their poor prognosis. In this study, we investigate the relationship between the increase of myeloid cells after surgery and prognosis in gastric cancer patients. Materials and Methods: For analysis of relationship between increased myeloid cells after surgery and patients’ prognosis, general archival data were obtained from 278 pStgae2-3 GC patients who received curative resection between January 2007 and December 2014. For analysis of relationship between perioperative changes of MDSCs and patients’ clinicopathologic data, peripheral blood was obtained from 75 GC patients who underwent gastrectomy between April 2016 and December 2017, and MDSCs, CD8, CD4 T-cells, and regulatory T-cells (Tregs) were analyzed by flow cytometry. For analysis of the immunosuppressive function, MDSCs were purified by FACS Aria II and the IFNγ secretion assay were performed using activated autologous T-cells as responder cells. Result and Discussion: In the analysis with 278 GC patients, short RFS was significantly correlated with increased number of monocytes after surgery (#monocytes) (P=0.0073), but not increased number of neutrophils (#neutrophils) (P=0.26). A multivariate analysis demonstrated that #monocytes (HR 1.49; 95% CI 1.01-2.21), pT (HR 2.14; 95% CI 1.26-3.83), pN (HR 2.98; 95% CI 1.81-5.18), postoperative complications (HR 1.69; 95% CI 1.03-2.68) were independently associated with RFS. In the analysis with 75 GC patients, preoperative % M-MDSCs (CD11b+ CD33+ HLA-DR- in CD14+ PBMCs) was positively associated with pathologic stages. After gastrectomy, % M-MDSCs dramatically increased in most patients. On the other hand, % Foxp3+ CD25+ in CD4+ increased but not significantly, while % CD4 and %CD8 in CD3+ were stable. By the suppression assay, smaller number of IFNγ-secreting responder cells were observed when they were co-cultured with M-MDSCs than with CD11b+ CD33+HLA-DR+ cells. Because the increase in the number of M-MDSCs (#M-MDSCs) was positively correlated with #monocytes (r2=0.57 P Citation Format: Shinya Urakawa, Hisashi Wada, Masaki Mori, Yuichiro Doki. The impact of postoperative myeloid-derived suppressor cells on prognosis of gastric cancer patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New Yor
背景与目的:髓系细胞如髓源性抑制细胞(Myeloid -derived suppressor cells, MDSCs)、肿瘤相关巨噬细胞(tumor-associated macrophages, tam)和中性粒细胞在抗肿瘤免疫中具有抑制作用。在胃癌(GC)患者中,有报道称这些外周血或肿瘤组织中的髓样细胞与预后有关,并在手术治疗后增加。一些报道称,在小鼠模型中,肿瘤进展和频繁复发是由手术应激后MDSC增加引起的。然而,在人类胃癌患者中,胃切除术后骨髓细胞的增加是否与预后不良有关尚不清楚。在本研究中,我们探讨胃癌患者术后骨髓细胞的增加与预后的关系。材料与方法:为分析术后髓样细胞增多与患者预后的关系,我们收集了2007年1月至2014年12月278例接受根治性切除的pStgae2-3型胃癌患者的一般档案资料。为了分析MDSCs围手术期变化与患者临床病理数据的关系,我们采集了2016年4月至2017年12月期间接受胃切除术的75例胃癌患者的外周血,并采用流式细胞术分析MDSCs、CD8、CD4 t细胞和调节性t细胞(Tregs)。为了分析免疫抑制功能,使用FACS Aria II纯化MDSCs,并使用活化的自体t细胞作为应答细胞进行IFNγ分泌测定。结果与讨论:在278例胃癌患者的分析中,短RFS与术后单核细胞数量(#monocytes)增加显著相关(P=0.0073),但与中性粒细胞(#neutrophils)增加无关(P=0.26)。多变量分析表明,#单核细胞(HR 1.49;95% CI 1.01-2.21), pT (HR 2.14;95% CI 1.26-3.83), pN (HR 2.98;95% CI 1.81-5.18),术后并发症(HR 1.69;95% CI 1.03-2.68)与RFS独立相关。在对75例胃癌患者的分析中,术前M-MDSCs (CD11b+ CD33+ CD14+ PBMCs中的HLA-DR-)百分比与病理分期呈正相关。胃切除术后,大多数患者的M-MDSCs显著增加。另一方面,CD4+中的% Foxp3+ CD25+增加但不显著,而CD3+中的% CD4和%CD8保持稳定。抑制实验发现,与M-MDSCs共培养时,分泌ifn γ的应答细胞数量少于与CD11b+ CD33+HLA-DR+细胞共培养的应答细胞数量。因为M-MDSCs (#M-MDSCs)数量的增加与#单核细胞呈正相关(r2=0.57 P)。胃癌患者术后骨髓源性抑制细胞对预后的影响[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 B191。
{"title":"Abstract B191: The impact of postoperative myeloid-derived suppressor cells on prognosis of gastric cancer patients","authors":"S. Urakawa, H. Wada, M. Mori, Y. Doki","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B191","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B191","url":null,"abstract":"Background and Aim: Myeloid cells such as myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages (TAMs) and neutrophils have a suppressive role in the antitumor immunity. In gastric cancer (GC) patients, it has been reported that these myeloid cells in peripheral blood or tumor tissue were associated with their prognosis and increased after surgical treatments. Some reported that the tumor progression and frequent recurrence were caused by increased MDSC after surgical stress in a mouse model. However, in human GC patients, it is still unclear whether such an increase of myeloid cells after gastrectomy is related to their poor prognosis. In this study, we investigate the relationship between the increase of myeloid cells after surgery and prognosis in gastric cancer patients. Materials and Methods: For analysis of relationship between increased myeloid cells after surgery and patients’ prognosis, general archival data were obtained from 278 pStgae2-3 GC patients who received curative resection between January 2007 and December 2014. For analysis of relationship between perioperative changes of MDSCs and patients’ clinicopathologic data, peripheral blood was obtained from 75 GC patients who underwent gastrectomy between April 2016 and December 2017, and MDSCs, CD8, CD4 T-cells, and regulatory T-cells (Tregs) were analyzed by flow cytometry. For analysis of the immunosuppressive function, MDSCs were purified by FACS Aria II and the IFNγ secretion assay were performed using activated autologous T-cells as responder cells. Result and Discussion: In the analysis with 278 GC patients, short RFS was significantly correlated with increased number of monocytes after surgery (#monocytes) (P=0.0073), but not increased number of neutrophils (#neutrophils) (P=0.26). A multivariate analysis demonstrated that #monocytes (HR 1.49; 95% CI 1.01-2.21), pT (HR 2.14; 95% CI 1.26-3.83), pN (HR 2.98; 95% CI 1.81-5.18), postoperative complications (HR 1.69; 95% CI 1.03-2.68) were independently associated with RFS. In the analysis with 75 GC patients, preoperative % M-MDSCs (CD11b+ CD33+ HLA-DR- in CD14+ PBMCs) was positively associated with pathologic stages. After gastrectomy, % M-MDSCs dramatically increased in most patients. On the other hand, % Foxp3+ CD25+ in CD4+ increased but not significantly, while % CD4 and %CD8 in CD3+ were stable. By the suppression assay, smaller number of IFNγ-secreting responder cells were observed when they were co-cultured with M-MDSCs than with CD11b+ CD33+HLA-DR+ cells. Because the increase in the number of M-MDSCs (#M-MDSCs) was positively correlated with #monocytes (r2=0.57 P Citation Format: Shinya Urakawa, Hisashi Wada, Masaki Mori, Yuichiro Doki. The impact of postoperative myeloid-derived suppressor cells on prognosis of gastric cancer patients [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New Yor","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"58 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"114993177","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 B181: Adenovirus, Semliki Forest virus and vaccinia virus-induced immunogenic cell death augments oncolytic virus immunotherapy B181:腺病毒、塞姆利基森林病毒和牛痘病毒诱导的免疫原性细胞死亡增强了溶瘤病毒免疫治疗
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B181
Mohanraj Ramachandran, M. Essand
Oncolytic viruses represent new immunotherapy agents with therapeutic effects consisting of direct oncolysis of cancer cells and anticancer immune responses induced upon oncolysis. However, the exact pathways of cell death and the subsequent immune response initiations after oncolysis are not fully elucidated. We have examined the cell death pathways and immune-stimulatory pathways involved in adenovirus (Ad), Semliki Forest virus (SFV) and vaccinia virus (VV) infection. The three viruses activate distinct but different pathways but all of them induced immunogenic cell death (ICD), which is depicted by elevated cell surface exposure of calreticulin (CRT) and extracellular secretion of ATP. Oncolysis-mediated ICD grants dying cells to be phagocytosed and it promotes DC maturation and activation with a Th1-polarized cytokine secretion profile. As a proof of concept, we validated that when DCs are incubated with Ad- or SFV-, but not VV-infected tumor cells expressing a surrogate antigen (CMVpp65), they can efficiently process and cross-present peptides of this antigen to CD8+ T-cells, as shown by specific IFN-γ production. Mice immunized with SFV- or VV-infected murine glioblastoma cells (GL261) elicit a strong and specific T-cell response against GL261. This adaptive immune response can control tumor growth and prolong mice survival after GL261 rechallenge. Citation Format: Mohanraj Ramachandran, Magnus Essand. Adenovirus, Semliki Forest virus and vaccinia virus-induced immunogenic cell death augments oncolytic virus 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 B181.
溶瘤病毒是一种新型的免疫治疗药物,其治疗效果包括直接溶瘤癌细胞和溶瘤后诱导的抗癌免疫反应。然而,肿瘤溶解后细胞死亡和随后免疫反应启动的确切途径尚未完全阐明。我们研究了腺病毒(Ad)、塞姆利基森林病毒(SFV)和牛痘病毒(VV)感染所涉及的细胞死亡途径和免疫刺激途径。这三种病毒激活不同但不同的途径,但它们都诱导免疫原性细胞死亡(ICD),这是通过细胞表面钙网蛋白(CRT)暴露升高和细胞外ATP分泌来描述的。溶瘤介导的ICD使垂死的细胞被吞噬,并通过th1极化的细胞因子分泌谱促进DC成熟和激活。作为概念验证,我们验证了当dc与表达替代抗原(CMVpp65)的Ad-或SFV-而不是vv -感染的肿瘤细胞孵育时,它们可以有效地加工和交叉呈递该抗原的肽到CD8+ t细胞,如特异性IFN-γ产生所示。用SFV或vv感染的小鼠胶质母细胞瘤细胞(GL261)免疫小鼠,可引起针对GL261的强烈特异性t细胞应答。这种适应性免疫反应可以控制肿瘤生长,延长GL261再激后小鼠的生存期。引文格式:Mohanraj Ramachandran, Magnus Essand。腺病毒、塞姆利基森林病毒和牛痘病毒诱导的免疫原性细胞死亡增强了溶瘤病毒免疫治疗。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 B181。
{"title":"Abstract B181: Adenovirus, Semliki Forest virus and vaccinia virus-induced immunogenic cell death augments oncolytic virus immunotherapy","authors":"Mohanraj Ramachandran, M. Essand","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B181","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B181","url":null,"abstract":"Oncolytic viruses represent new immunotherapy agents with therapeutic effects consisting of direct oncolysis of cancer cells and anticancer immune responses induced upon oncolysis. However, the exact pathways of cell death and the subsequent immune response initiations after oncolysis are not fully elucidated. We have examined the cell death pathways and immune-stimulatory pathways involved in adenovirus (Ad), Semliki Forest virus (SFV) and vaccinia virus (VV) infection. The three viruses activate distinct but different pathways but all of them induced immunogenic cell death (ICD), which is depicted by elevated cell surface exposure of calreticulin (CRT) and extracellular secretion of ATP. Oncolysis-mediated ICD grants dying cells to be phagocytosed and it promotes DC maturation and activation with a Th1-polarized cytokine secretion profile. As a proof of concept, we validated that when DCs are incubated with Ad- or SFV-, but not VV-infected tumor cells expressing a surrogate antigen (CMVpp65), they can efficiently process and cross-present peptides of this antigen to CD8+ T-cells, as shown by specific IFN-γ production. Mice immunized with SFV- or VV-infected murine glioblastoma cells (GL261) elicit a strong and specific T-cell response against GL261. This adaptive immune response can control tumor growth and prolong mice survival after GL261 rechallenge. Citation Format: Mohanraj Ramachandran, Magnus Essand. Adenovirus, Semliki Forest virus and vaccinia virus-induced immunogenic cell death augments oncolytic virus 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 B181.","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"41 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"116013806","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 B194: Quantifying the interaction between macrophages and deformable microparticles with cell-like mechanical properties B194:巨噬细胞与具有细胞样力学特性的可变形微粒之间相互作用的定量分析
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B194
D. Vorselen, Yifan Wang, M. Footer, W. Cai, J. Theriot
Macrophages are able to phagocytose vastly different targets, ranging from bacteria to apoptotic and cancer cells. Especially the difference in rigidity between such targets is striking, with bacteria being ~1000 fold stiffer than human cells. Phagocytosis can also be strongly affected by target rigidity and is seemingly less efficient for softer targets. However, mechanistic studies have largely focused on the uptake of stiff particles, and it is currently poorly understood how macrophages adapt the phagocytic mechanism for efficient uptake of soft targets. Here, we developed deformable hydrogel microparticles to study phagocytosis in unprecedented detail, using targets that more accurately mimic cellular physical properties. We used extrusion through Shirasu Porous glass (SPG) membranes to create emulsions with uniformly sized droplets containing acrylamide, bis(acrylamide) and acrylic acid. Subsequent polymerization yields monodisperse (CV 10 Pa) directly from the deformed particle shape. We observed highly localized force exertion by the phagocytes and 4 implied mechanically distinct steps in the phagocytic process. Initially, we observe outward-directed pushing forces from the phagocytic cup base. During subsequent pseudopod extension the majority of the deformation is localized in a ring that is initially irregular, but becomes uniform during cup closure. Surprisingly, strong localized punches at the cup base occur in these stages. After cup closure, we observe some of the strongest forces, seemingly pushing the engulfed target into the cell. Our novel approach gives us unprecedented detail on the mechanical interaction between phagocyte and target in phagocytosis. The presented method here is expected to find broad applications in the study of the immune system in vitro and in vivo. Citation Format: Daan Vorselen, Yifan Wang, Matt Footer, Wei Cai, Julie Theriot. Quantifying the interaction between macrophages and deformable microparticles with cell-like mechanical properties [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 B194.
巨噬细胞能够吞噬各种各样的目标,从细菌到凋亡细胞和癌细胞。尤其是这两种目标之间的硬度差异是惊人的,细菌的硬度是人类细胞的1000倍。吞噬作用也会受到目标刚性的强烈影响,对较软的目标似乎效率较低。然而,机制研究主要集中在硬颗粒的摄取上,目前对巨噬细胞如何适应吞噬机制以有效摄取软靶点知之甚少。在这里,我们开发了可变形的水凝胶微粒,以前所未有的细节研究吞噬作用,使用更准确地模拟细胞物理特性的靶标。我们通过Shirasu多孔玻璃(SPG)膜挤压制造出含有均匀大小的液滴的乳液,其中含有丙烯酰胺、丙烯酰胺和丙烯酸。随后的聚合直接从变形的颗粒形状产生单分散(CV 10pa)。我们观察到吞噬细胞高度局部化的力发挥,并且在吞噬过程中有4个机械上不同的步骤。最初,我们观察到从吞噬杯底向外的推力。在随后的假足延伸期间,大部分变形局限于一个环,该环最初是不规则的,但在杯闭合期间变得均匀。令人惊讶的是,在这些阶段,杯子底部会出现强烈的局部冲击。在杯口闭合后,我们观察到一些最强的力,似乎把被吞没的目标推进了细胞。我们的新方法为吞噬作用中吞噬细胞和靶标之间的机械相互作用提供了前所未有的细节。本文提出的方法有望在体外和体内免疫系统的研究中找到广泛的应用。引用格式:Daan Vorselen, Yifan Wang, Matt Footer, Wei Cai, Julie Theriot。巨噬细胞与具有细胞样力学特性的可变形微粒之间相互作用的定量研究[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 B194。
{"title":"Abstract B194: Quantifying the interaction between macrophages and deformable microparticles with cell-like mechanical properties","authors":"D. Vorselen, Yifan Wang, M. Footer, W. Cai, J. Theriot","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B194","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B194","url":null,"abstract":"Macrophages are able to phagocytose vastly different targets, ranging from bacteria to apoptotic and cancer cells. Especially the difference in rigidity between such targets is striking, with bacteria being ~1000 fold stiffer than human cells. Phagocytosis can also be strongly affected by target rigidity and is seemingly less efficient for softer targets. However, mechanistic studies have largely focused on the uptake of stiff particles, and it is currently poorly understood how macrophages adapt the phagocytic mechanism for efficient uptake of soft targets. Here, we developed deformable hydrogel microparticles to study phagocytosis in unprecedented detail, using targets that more accurately mimic cellular physical properties. We used extrusion through Shirasu Porous glass (SPG) membranes to create emulsions with uniformly sized droplets containing acrylamide, bis(acrylamide) and acrylic acid. Subsequent polymerization yields monodisperse (CV 10 Pa) directly from the deformed particle shape. We observed highly localized force exertion by the phagocytes and 4 implied mechanically distinct steps in the phagocytic process. Initially, we observe outward-directed pushing forces from the phagocytic cup base. During subsequent pseudopod extension the majority of the deformation is localized in a ring that is initially irregular, but becomes uniform during cup closure. Surprisingly, strong localized punches at the cup base occur in these stages. After cup closure, we observe some of the strongest forces, seemingly pushing the engulfed target into the cell. Our novel approach gives us unprecedented detail on the mechanical interaction between phagocyte and target in phagocytosis. The presented method here is expected to find broad applications in the study of the immune system in vitro and in vivo. Citation Format: Daan Vorselen, Yifan Wang, Matt Footer, Wei Cai, Julie Theriot. Quantifying the interaction between macrophages and deformable microparticles with cell-like mechanical properties [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 B194.","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"16 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"123685870","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 B180: Effects of EMT process under MHC class I and TAP1 gene expression related to antigen presentation 摘要:EMT过程对MHC I类和TAP1基因表达的影响
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B180
P. R. L. Pires, P. Xavier, H. Fukumasu
Metastasis is a process that involves tumor cell migration from a primary tumor and its invasion to other tissues, which in turn are believed to be driven by a process called epithelial-mesenchymal transition (EMT). EMT process in cancer cells implies increase of malignance and metastatic potential by molecular and phenotypic modifications. MHC class I (MHC-I) is a protein complex used by cells for antigen presentation, specifically to T-cell CD8. It is believed that MHC-I gene expression may fluctuate along different cancer types, biologic processes and molecular modifications which cancer cells may suffer, and it could directly impact on system immunologic responses and T-cell-dependent immunotherapy treatment. This project aimed to evaluate MHC-I and TAP1 gene expression in nonmetastatic origin and metastatic origin cells under the effect of tumor growth factor beta (TGFb), a factor that induces EMT process. Three nonmetastatic cell lineages (A549, H1703, H23) and three metastatic cell lineages (H1792, H2023, H2030) of pulmonary carcinoma were cultured under controlled conditions in RPMI culture media supplemented with 10% of bovine fetal serum (BFS), 1% of antibiotics (penicillin and streptomycin), 2% of glutamine in incubator at 37oC and air atmosphere containing 5% of CO2. Cell cultures were supplemented or not with TGFb (4µg/mL) for 5 days to induce EMT process. After 5 days, cells were evaluated for acquiring mesenchymal cell morphology and MHC-I and TAP1 gene expression (relative quantification). Pool of cells was used to obtain RNA using RNAesy Extraction Kit (QIAGEN®) followed by RT-PCR reaction using High Capacity RNA-to-cDNA Kit (Applied Biosystems®) to obtain cDNA. Relative gene expression was analyzed using Real Time PCR using Fast SYBR™ Green Master Mix (Applied Biosystems®). Except by H23 cell, TGFb incubation showed to be effective and seems to induce EMT process within 5 days culture for both cell types. The cells acquired mesenchymal morphology characteristics such as elongation and increased size. It was also possible to identify a significant gene expression pattern for MHC-I and TAP1 gene expression between control and TGFb groups. Both MHC-I and TAP1 gene expression were shown to be upregulated in the majority of non-metastatic origin cells (A549, H1703, H23; p Citation Format: Pedro Ratto Lisboa Pires, Pedro L.P. Xavier, Heidge Fukumasu. Effects of EMT process under MHC class I and TAP1 gene expression related to antigen presentation [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 B180.
转移是肿瘤细胞从原发肿瘤转移到其他组织的过程,这一过程被认为是由上皮-间质转化(EMT)过程驱动的。癌细胞中的EMT过程表明,通过分子和表型修饰,恶性和转移潜力增加。MHC I类(MHC-I)是细胞用于抗原呈递的蛋白质复合物,特别是对t细胞CD8。认为mhc - 1基因的表达随肿瘤类型、肿瘤细胞可能遭受的生物学过程和分子修饰的不同而波动,并可能直接影响系统免疫反应和t细胞依赖性免疫治疗。本项目旨在评价肿瘤生长因子β (tumor growth factor beta, TGFb)在诱导EMT过程的作用下,MHC-I和TAP1基因在非转移源细胞和转移源细胞中的表达。将3个非转移性肺癌细胞系(A549、H1703、H23)和3个转移性肺癌细胞系(H1792、H2023、H2030)在RPMI培养基中培养,培养基中添加10%的牛胎血清(BFS)、1%的抗生素(青霉素和链霉素)、2%的谷氨酰胺,37℃培养箱,空气中CO2含量为5%。细胞培养液中分别添加或不添加TGFb(4µg/mL),培养5 d诱导EMT过程。5天后,检测细胞间充质细胞形态及MHC-I和TAP1基因表达(相对定量)。细胞池采用RNAesy Extraction Kit (QIAGEN®)提取RNA,然后采用High Capacity RNA-to-cDNA Kit (Applied Biosystems®)进行RT-PCR反应获得cDNA。使用Fast SYBR™Green Master Mix (Applied Biosystems®)的Real Time PCR分析相对基因表达。除H23细胞外,TGFb孵育对两种细胞类型均有效,并在培养5天内诱导EMT过程。细胞获得间充质形态特征,如伸长和增大。在对照组和TGFb组之间,mhc - 1和TAP1基因表达也有显著的基因表达模式。MHC-I和TAP1基因表达在大多数非转移源细胞中均上调(A549, H1703, H23;p引文格式:Pedro Ratto Lisboa Pires, Pedro L.P. Xavier, Heidge Fukumasu。EMT过程对MHC I类及抗原呈递相关TAP1基因表达的影响[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 B180。
{"title":"Abstract B180: Effects of EMT process under MHC class I and TAP1 gene expression related to antigen presentation","authors":"P. R. L. Pires, P. Xavier, H. Fukumasu","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B180","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B180","url":null,"abstract":"Metastasis is a process that involves tumor cell migration from a primary tumor and its invasion to other tissues, which in turn are believed to be driven by a process called epithelial-mesenchymal transition (EMT). EMT process in cancer cells implies increase of malignance and metastatic potential by molecular and phenotypic modifications. MHC class I (MHC-I) is a protein complex used by cells for antigen presentation, specifically to T-cell CD8. It is believed that MHC-I gene expression may fluctuate along different cancer types, biologic processes and molecular modifications which cancer cells may suffer, and it could directly impact on system immunologic responses and T-cell-dependent immunotherapy treatment. This project aimed to evaluate MHC-I and TAP1 gene expression in nonmetastatic origin and metastatic origin cells under the effect of tumor growth factor beta (TGFb), a factor that induces EMT process. Three nonmetastatic cell lineages (A549, H1703, H23) and three metastatic cell lineages (H1792, H2023, H2030) of pulmonary carcinoma were cultured under controlled conditions in RPMI culture media supplemented with 10% of bovine fetal serum (BFS), 1% of antibiotics (penicillin and streptomycin), 2% of glutamine in incubator at 37oC and air atmosphere containing 5% of CO2. Cell cultures were supplemented or not with TGFb (4µg/mL) for 5 days to induce EMT process. After 5 days, cells were evaluated for acquiring mesenchymal cell morphology and MHC-I and TAP1 gene expression (relative quantification). Pool of cells was used to obtain RNA using RNAesy Extraction Kit (QIAGEN®) followed by RT-PCR reaction using High Capacity RNA-to-cDNA Kit (Applied Biosystems®) to obtain cDNA. Relative gene expression was analyzed using Real Time PCR using Fast SYBR™ Green Master Mix (Applied Biosystems®). Except by H23 cell, TGFb incubation showed to be effective and seems to induce EMT process within 5 days culture for both cell types. The cells acquired mesenchymal morphology characteristics such as elongation and increased size. It was also possible to identify a significant gene expression pattern for MHC-I and TAP1 gene expression between control and TGFb groups. Both MHC-I and TAP1 gene expression were shown to be upregulated in the majority of non-metastatic origin cells (A549, H1703, H23; p Citation Format: Pedro Ratto Lisboa Pires, Pedro L.P. Xavier, Heidge Fukumasu. Effects of EMT process under MHC class I and TAP1 gene expression related to antigen presentation [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 B180.","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"58 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"128438597","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 B185: Epithelial cancer cell-expressed genes contribute to clinically relevant immune-based classifications of breast cancer B185:上皮癌细胞表达基因有助于临床相关的基于免疫的乳腺癌分类
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B185
J. Shepherd, C. Perou
Introduction: With the development of immunotherapies for breast cancer therapy, reliable methods to evaluate the extent and type of immune involvement present in tumors and investigation of its effect on patient prognosis and treatment are needed. Identifying tumor-specific features that affect immune involvement will be a key to understand tumor-immune involvement. Therefore, we evaluated expression of immune-related mRNA signatures in TCGA breast cancer data to identify distinct immune-related tumor subsets and asociated prognostic values. We also evaluated immune cell features, including B cell and T-cell receptor richness and diversity, as well as epithelial tumor cell-specific features, including somatic mutations, copy number alterations and differential RNA expression between identified groups. Methods: More than 130 published immune related gene signatures were evaluated in 1095 breast tumors and 97 normal mammary samples. Groups were identified by consensus based hierarchical clustering of the immune signatures, using the proportion of ambiguous clustering to select the optimal number of groups. An ElasticNet model trained on TCGA data was applied to two other breast tumor datasets to predict immune group classification. RNA-sequencing (RNAseq) data from 70 breast cancer cell lines and from human tumor xenografts passaged in immune-compromised mice and processed through a human specific sequencing pipeline provided in vitro and in vivo sources of epithelial cancer cell expression with limited stromal content that was used to filter TCGA bulk RNAseq data for epithelial expressed genes. Results: We identified three distinct immune groups present in breast cancer: immune-low, immune-normal, and immune-high. The immune-high group is characterized by high T-cell scores, including both cytotoxic and regulatory T-cell signatures, and increased B cell and macrophage signatures. The immune-normal set shows signs of normal epithelia and low proliferation. The immune-low group has very low immune cell signatures. Intrinsic breast cancer subtypes (Basal, luminal A, Luminal B, Her2 and Normal-like) are present in each of the immune groups; however, enrichment of basal tumors in immune-high, luminal tumors in the immune-low, and normal mammary, normal-like tumors and luminal A tumors in the immune-normal group demonstrate an interaction between intrinsic tumor type and immune involvement. Immune groups are prognostic in TCGA, with the immune-high group having improved recurrence-free survival. Two more breast tumor datasets confirmed improved survival for basal tumors in the immune-high group relative to the immune-low tumors. Total mutation burden, unique somatic mutations, and copy number alterations did not show significant changes between immune-low and –high groups, whereas RNA expression differs between groups. Selecting for genes with evidence of expression by epithelial breast cancer cells identified over 8,000 genes differentially expressed
导论:随着乳腺癌免疫疗法的发展,需要可靠的方法来评估肿瘤中存在的免疫受累的程度和类型,并研究其对患者预后和治疗的影响。确定影响免疫受累的肿瘤特异性特征将是了解肿瘤免疫受累的关键。因此,我们评估了TCGA乳腺癌数据中免疫相关mRNA特征的表达,以识别不同的免疫相关肿瘤亚群和相关的预后价值。我们还评估了免疫细胞特征,包括B细胞和t细胞受体的丰富度和多样性,以及上皮肿瘤细胞特异性特征,包括体细胞突变、拷贝数改变和鉴定组之间的差异RNA表达。方法:对1095例乳腺肿瘤和97例正常乳腺样本的130多个已发表的免疫相关基因特征进行评价。采用基于共识的免疫特征分层聚类方法,利用模糊聚类比例选择最优组数。在TCGA数据上训练的ElasticNet模型应用于另外两个乳腺肿瘤数据集来预测免疫群分类。来自70种乳腺癌细胞系和人类肿瘤异种移植物的rna测序(RNAseq)数据在免疫受损小鼠中传代,并通过人类特异性测序管道进行处理,该管道提供了体外和体内上皮癌细胞表达来源,基质含量有限,用于过滤TCGA大量上皮表达基因的RNAseq数据。结果:我们确定了乳腺癌中存在的三种不同的免疫组:免疫低、免疫正常和免疫高。免疫高组的特点是t细胞评分高,包括细胞毒性和调节性t细胞特征,以及B细胞和巨噬细胞特征增加。免疫正常组显示正常上皮和低增殖的迹象。免疫低下组的免疫细胞特征非常低。内在乳腺癌亚型(基底、腔内A型、腔内B型、Her2型和正常样)存在于每个免疫组中;然而,免疫高组的基底肿瘤、免疫低组的管腔肿瘤以及免疫正常组的正常乳腺、正常样肿瘤和管腔A肿瘤的富集表明,内在肿瘤类型与免疫受累之间存在相互作用。免疫组是TCGA的预后,免疫高组的无复发生存率更高。另外两个乳腺肿瘤数据集证实,相对于免疫低下的肿瘤,免疫高组的基础肿瘤存活率有所提高。总突变负担、独特体细胞突变和拷贝数改变在免疫低组和免疫高组之间没有显着变化,而RNA表达在组间存在差异。选择上皮性乳腺癌细胞有表达证据的基因,在免疫低组和免疫高组之间发现了8000多个基因的差异表达,其中CCL5、ACAP1、PVRIG、SLA2、LCK和CD8A是最显著的。结论:乳腺癌可分为三个临床相关免疫相关组。免疫高具有高度的免疫参与,表现出细胞毒性和免疫抑制和衰竭的标记。这些患者的生存率有所提高,但可能仍然受益于免疫检查点抑制。免疫正常反映了正常的乳腺免疫状态,表明微环境没有被肿瘤强烈改变。免疫低下似乎表明免疫细胞从肿瘤中被排斥,即使肿瘤中含有预测的新抗原。这些患者生存率较差,需要开发新的治疗策略来激活免疫参与。上皮癌细胞表达许多免疫相关基因,包括CCL5、LCK和CD8A,这些基因可能是免疫细胞吸引的关键决定因素。引用格式:Jonathan Shepherd, Charles Perou。上皮癌细胞表达的基因有助于乳腺癌临床相关的免疫分类[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 B185。
{"title":"Abstract B185: Epithelial cancer cell-expressed genes contribute to clinically relevant immune-based classifications of breast cancer","authors":"J. Shepherd, C. Perou","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B185","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B185","url":null,"abstract":"Introduction: With the development of immunotherapies for breast cancer therapy, reliable methods to evaluate the extent and type of immune involvement present in tumors and investigation of its effect on patient prognosis and treatment are needed. Identifying tumor-specific features that affect immune involvement will be a key to understand tumor-immune involvement. Therefore, we evaluated expression of immune-related mRNA signatures in TCGA breast cancer data to identify distinct immune-related tumor subsets and asociated prognostic values. We also evaluated immune cell features, including B cell and T-cell receptor richness and diversity, as well as epithelial tumor cell-specific features, including somatic mutations, copy number alterations and differential RNA expression between identified groups. Methods: More than 130 published immune related gene signatures were evaluated in 1095 breast tumors and 97 normal mammary samples. Groups were identified by consensus based hierarchical clustering of the immune signatures, using the proportion of ambiguous clustering to select the optimal number of groups. An ElasticNet model trained on TCGA data was applied to two other breast tumor datasets to predict immune group classification. RNA-sequencing (RNAseq) data from 70 breast cancer cell lines and from human tumor xenografts passaged in immune-compromised mice and processed through a human specific sequencing pipeline provided in vitro and in vivo sources of epithelial cancer cell expression with limited stromal content that was used to filter TCGA bulk RNAseq data for epithelial expressed genes. Results: We identified three distinct immune groups present in breast cancer: immune-low, immune-normal, and immune-high. The immune-high group is characterized by high T-cell scores, including both cytotoxic and regulatory T-cell signatures, and increased B cell and macrophage signatures. The immune-normal set shows signs of normal epithelia and low proliferation. The immune-low group has very low immune cell signatures. Intrinsic breast cancer subtypes (Basal, luminal A, Luminal B, Her2 and Normal-like) are present in each of the immune groups; however, enrichment of basal tumors in immune-high, luminal tumors in the immune-low, and normal mammary, normal-like tumors and luminal A tumors in the immune-normal group demonstrate an interaction between intrinsic tumor type and immune involvement. Immune groups are prognostic in TCGA, with the immune-high group having improved recurrence-free survival. Two more breast tumor datasets confirmed improved survival for basal tumors in the immune-high group relative to the immune-low tumors. Total mutation burden, unique somatic mutations, and copy number alterations did not show significant changes between immune-low and –high groups, whereas RNA expression differs between groups. Selecting for genes with evidence of expression by epithelial breast cancer cells identified over 8,000 genes differentially expressed ","PeriodicalId":120683,"journal":{"name":"Other Topics","volume":"4 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"121597783","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
Other Topics
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