首页 > 最新文献

Mutational Analysis and Predicting Response to Immunotherapy最新文献

英文 中文
Abstract B092: Tumor infiltrating T-cells from renal cell carcinoma patients recognize neoantigens derived from point and frameshift mutations B092:肾细胞癌患者肿瘤浸润性t细胞可识别源自点移突变和移码突变的新抗原
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B092
S. Ramskov, U. Hansen, Anne-Mette Bjerregaard, A. Bentzen, M. Donia, R. Andersen, Z. Szallasi, I. Svane, A. Eklund, S. Hadrup
Mutation-derived neoantigens are important targets of T-cell mediated reactivity towards tumors. Their unique tumor-restriction poses an advantage compared to shared tumor antigens in that they are in principle both foreign and tumor specific, hence presumably less impacted by T-cell tolerance and for therapeutic applications less prone to mediate immune-related destruction of noncancerous tissue. Moreover, the mutational burden and predicted number of neoantigens correlate to favorable clinical outcome and benefit from immune checkpoint therapy. Neoantigen-reactive T-cells have been detected across a number of solid cancers, ranging from immunogenic tumors such as melanoma and non-small cell lung cancer to less immunogenic tumors such as breast cancer. Renal cell carcinomas (RCCs) are among medium-range mutational burden tumors and present with the highest pan-cancer number and proportion of frameshift mutations, a mutation type considered to be highly immunogenic. However, to our knowledge, yet no reports have described neoantigen-specific T-cells in this malignancy. In this study, the mutational landscape and HLA (human leukocyte antigen) profile of tumors from six renal cell carcinoma patients were analyzed by whole-exome sequencing (WXS) of DNA from tumor fragments (TFs), autologous tumor cell lines (TCLs) and tumor-infiltrating lymphocytes (TILs, germline reference). Hereafter the online MuPeXi tool was used to predict binding of mutated peptide sequences of 9-11mer length to the HLAs of each patient, using a rank score Citation Format: Sofie Ramskov, Ulla Kring Hansen, Anne-Mette Bjerregaard, Amalie Kai Bentzen, Marco Donia, Rikke Andersen, Zoltan Szallasi, Inge Marie Stentoft Svane, Aron Charles Eklund, Sine Reker Hadrup. Tumor infiltrating T-cells from renal cell carcinoma patients recognize neoantigens derived from point and frameshift mutations [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 B092.
突变衍生的新抗原是t细胞介导的肿瘤反应性的重要靶标。与共享的肿瘤抗原相比,它们独特的肿瘤限制具有优势,因为它们原则上既具有外源性又具有肿瘤特异性,因此可能较少受到t细胞耐受性的影响,并且在治疗应用中不易介导非癌组织的免疫相关破坏。此外,突变负担和预测的新抗原数量与良好的临床结果相关,并受益于免疫检查点治疗。新抗原反应性t细胞已经在许多实体癌症中被检测到,从免疫原性肿瘤如黑色素瘤和非小细胞肺癌到免疫原性较低的肿瘤如乳腺癌。肾细胞癌(rcc)是一种中等范围的突变负担肿瘤,具有最高的泛癌数量和移码突变比例,移码突变被认为是一种高度免疫原性的突变类型。然而,据我们所知,还没有报道描述新抗原特异性t细胞在这种恶性肿瘤。本研究采用肿瘤片段(TFs)、自体肿瘤细胞系(TCLs)和肿瘤浸润淋巴细胞(TILs,种系参考)DNA全外显子组测序(WXS)方法,分析了6例肾癌患者肿瘤的突变格局和HLA(人白细胞抗原)谱。随后,使用在线MuPeXi工具预测9-11mer长度的突变肽序列与每个患者hla的结合,使用排名评分引用格式:Sofie Ramskov, Ulla Kring Hansen, Anne-Mette Bjerregaard, Amalie Kai Bentzen, Marco Donia, Rikke Andersen, Zoltan Szallasi, Inge Marie Stentoft Svane, Aron Charles Eklund, Sine Reker Hadrup。肾细胞癌患者的肿瘤浸润性t细胞可识别源自点突变和移码突变的新抗原[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 B092。
{"title":"Abstract B092: Tumor infiltrating T-cells from renal cell carcinoma patients recognize neoantigens derived from point and frameshift mutations","authors":"S. Ramskov, U. Hansen, Anne-Mette Bjerregaard, A. Bentzen, M. Donia, R. Andersen, Z. Szallasi, I. Svane, A. Eklund, S. Hadrup","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B092","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B092","url":null,"abstract":"Mutation-derived neoantigens are important targets of T-cell mediated reactivity towards tumors. Their unique tumor-restriction poses an advantage compared to shared tumor antigens in that they are in principle both foreign and tumor specific, hence presumably less impacted by T-cell tolerance and for therapeutic applications less prone to mediate immune-related destruction of noncancerous tissue. Moreover, the mutational burden and predicted number of neoantigens correlate to favorable clinical outcome and benefit from immune checkpoint therapy. Neoantigen-reactive T-cells have been detected across a number of solid cancers, ranging from immunogenic tumors such as melanoma and non-small cell lung cancer to less immunogenic tumors such as breast cancer. Renal cell carcinomas (RCCs) are among medium-range mutational burden tumors and present with the highest pan-cancer number and proportion of frameshift mutations, a mutation type considered to be highly immunogenic. However, to our knowledge, yet no reports have described neoantigen-specific T-cells in this malignancy. In this study, the mutational landscape and HLA (human leukocyte antigen) profile of tumors from six renal cell carcinoma patients were analyzed by whole-exome sequencing (WXS) of DNA from tumor fragments (TFs), autologous tumor cell lines (TCLs) and tumor-infiltrating lymphocytes (TILs, germline reference). Hereafter the online MuPeXi tool was used to predict binding of mutated peptide sequences of 9-11mer length to the HLAs of each patient, using a rank score Citation Format: Sofie Ramskov, Ulla Kring Hansen, Anne-Mette Bjerregaard, Amalie Kai Bentzen, Marco Donia, Rikke Andersen, Zoltan Szallasi, Inge Marie Stentoft Svane, Aron Charles Eklund, Sine Reker Hadrup. Tumor infiltrating T-cells from renal cell carcinoma patients recognize neoantigens derived from point and frameshift mutations [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 B092.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"87 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":"114545680","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 B083: Defective transcription elongation in a subset of cancers confers immunotherapy resistance 摘要B083:癌症亚群中有缺陷的转录延伸会导致免疫治疗耐药性
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B083
V. Modur
Immunogenicity of most cancer types is a result of either neoantigenic mutational load, which stokes up an adaptive immune response, or due to oncogenic stress pathways, which elicit an innate antitumor response. There have been exponential gains made in recent times using numerous strategies to reactivate the host antitumor immunity including the development of immune checkpoint inhibitors. Nonetheless, the promise of a cure and durable response in select patients does not refute the low response rates in advanced cases, most of which also relapse. These observations shore up the possibility of other mechanisms beyond the inactivation of local lymphocyte infiltrates that might also play in tumor cell evasion of antitumor immune response. Through comprehensive computational and follow-up experimental validations, we have found that a subset of cancers (~15-20% of all cancers) are characterized by severe defects in almost the entire epigenetic and transcriptional apparatus. These defects result in genome-wide deregulation of histone modifications, mRNA transcription elongation, and mRNA processing and nuclear export, especially in the long genes which we term as Transcriptional Elongation Deficient: TEdef. As such, cancer cells with TEdef suppressed the expression of the pathways enriched for long genes, such as proinflammatory signaling pathways (FasL response, TNF/NF-kB signaling, interferon signaling) at both mRNA and protein levels, and had diminished response to interferon and TNF stimuli. Remarkably, in renal cell carcinoma and metastatic melanoma patients in four cohorts, the TEdef phenotype significantly correlated with poor response and unfavorable outcome to immunotherapy, but not to chemo- or targeted therapy. Importantly, forced induction of TEdef in tumor cells impaired the expression of, and signaling through, the proinflammatory pathways, and imposed a resistance to the innate and adaptive antitumor immune responses and to immune checkpoint inhibitor therapy in vivo. Tumor lymphocyte infiltration (TIL) and somatic neoepitope load (SNL) are some of the best markers of immunotherapy response in the clinic. However, TEdef tumors were characterized by a higher rate of immune cell infiltration, but paradoxically, less immune-mediated local tumor cell lysis, further supporting the notion that TEdef is a tumor cell-autonomous mechanism of immune resistance. As such, combining TIL or SNL with TEdef had superior power in predicting the progression-free and overall survival of melanoma patients treated with the anti-CTLA4 and anti-PD-1 therapy. Overall, TEdef is a novel epigenetic mechanism of resistance to antitumor immune attack, which warrants its assessment in cancer patients undergoing immunotherapy. Citation Format: Vishnu Modur. Defective transcription elongation in a subset of cancers confers immunotherapy resistance [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating
大多数癌症类型的免疫原性要么是新抗原突变负荷的结果,引发适应性免疫反应,要么是由于致癌应激途径,引发先天抗肿瘤反应。近年来,利用多种策略重新激活宿主抗肿瘤免疫,包括免疫检查点抑制剂的开发,取得了指数级的进展。尽管如此,在某些患者中,治愈和持久反应的承诺并不能反驳晚期病例的低反应率,其中大多数也会复发。这些观察结果支持了局部淋巴细胞浸润失活之外的其他机制的可能性,这些机制也可能在肿瘤细胞逃避抗肿瘤免疫反应中发挥作用。通过全面的计算和后续的实验验证,我们发现癌症的一个子集(约占所有癌症的15-20%)几乎在整个表观遗传和转录装置中都存在严重缺陷。这些缺陷导致组蛋白修饰的全基因组失调,mRNA转录延伸,mRNA加工和核输出,特别是在我们称之为转录延伸缺陷的长基因中:TEdef。因此,具有TEdef的癌细胞在mRNA和蛋白质水平上抑制了富含长基因的通路的表达,如促炎信号通路(FasL反应、TNF/NF-kB信号传导、干扰素信号传导),并且对干扰素和TNF刺激的反应减弱。值得注意的是,在四个队列中的肾细胞癌和转移性黑色素瘤患者中,TEdef表型与免疫治疗的不良反应和不良结果显著相关,而与化疗或靶向治疗无关。重要的是,在肿瘤细胞中强制诱导TEdef会破坏促炎途径的表达和信号传导,并在体内对先天和适应性抗肿瘤免疫反应以及免疫检查点抑制剂治疗施加抵抗。肿瘤淋巴细胞浸润(TIL)和体细胞新表位负荷(SNL)是临床免疫治疗反应的最佳标志物。然而,TEdef肿瘤的特点是免疫细胞浸润率较高,但矛盾的是,免疫介导的局部肿瘤细胞裂解较少,这进一步支持了TEdef是肿瘤细胞自主免疫抵抗机制的观点。因此,TIL或SNL联合TEdef在预测接受抗ctla4和抗pd -1治疗的黑色素瘤患者的无进展和总生存期方面具有更强的能力。总之,TEdef是抗肿瘤免疫攻击抵抗的一种新的表观遗传机制,值得在接受免疫治疗的癌症患者中进行评估。引文格式:毗湿奴模式。在癌症的一个子集中,有缺陷的转录延伸会导致免疫治疗耐药性[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志,2019;7(2增刊):摘要nr B083。
{"title":"Abstract B083: Defective transcription elongation in a subset of cancers confers immunotherapy resistance","authors":"V. Modur","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B083","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B083","url":null,"abstract":"Immunogenicity of most cancer types is a result of either neoantigenic mutational load, which stokes up an adaptive immune response, or due to oncogenic stress pathways, which elicit an innate antitumor response. There have been exponential gains made in recent times using numerous strategies to reactivate the host antitumor immunity including the development of immune checkpoint inhibitors. Nonetheless, the promise of a cure and durable response in select patients does not refute the low response rates in advanced cases, most of which also relapse. These observations shore up the possibility of other mechanisms beyond the inactivation of local lymphocyte infiltrates that might also play in tumor cell evasion of antitumor immune response. Through comprehensive computational and follow-up experimental validations, we have found that a subset of cancers (~15-20% of all cancers) are characterized by severe defects in almost the entire epigenetic and transcriptional apparatus. These defects result in genome-wide deregulation of histone modifications, mRNA transcription elongation, and mRNA processing and nuclear export, especially in the long genes which we term as Transcriptional Elongation Deficient: TEdef. As such, cancer cells with TEdef suppressed the expression of the pathways enriched for long genes, such as proinflammatory signaling pathways (FasL response, TNF/NF-kB signaling, interferon signaling) at both mRNA and protein levels, and had diminished response to interferon and TNF stimuli. Remarkably, in renal cell carcinoma and metastatic melanoma patients in four cohorts, the TEdef phenotype significantly correlated with poor response and unfavorable outcome to immunotherapy, but not to chemo- or targeted therapy. Importantly, forced induction of TEdef in tumor cells impaired the expression of, and signaling through, the proinflammatory pathways, and imposed a resistance to the innate and adaptive antitumor immune responses and to immune checkpoint inhibitor therapy in vivo. Tumor lymphocyte infiltration (TIL) and somatic neoepitope load (SNL) are some of the best markers of immunotherapy response in the clinic. However, TEdef tumors were characterized by a higher rate of immune cell infiltration, but paradoxically, less immune-mediated local tumor cell lysis, further supporting the notion that TEdef is a tumor cell-autonomous mechanism of immune resistance. As such, combining TIL or SNL with TEdef had superior power in predicting the progression-free and overall survival of melanoma patients treated with the anti-CTLA4 and anti-PD-1 therapy. Overall, TEdef is a novel epigenetic mechanism of resistance to antitumor immune attack, which warrants its assessment in cancer patients undergoing immunotherapy. Citation Format: Vishnu Modur. Defective transcription elongation in a subset of cancers confers immunotherapy resistance [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"39 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":"116486789","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 B093: Development of prediction software PrDx, trained on peptide-MHC stability assays, shows new important positions in the binding patterns of the peptides-MHC I and II complexes 摘要:基于肽- mhc稳定性分析的预测软件PrDx的开发,在肽- mhc I和II复合物的结合模式中显示了新的重要位置
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B093
Stephan Thorgrimsen, S. Justesen, N. Rapin
Mutations in cancer cells may lead to the formation of neo-epitopes potentially presented by both major histocompatibility complex (MHC) class I or II. These neo-epitopes may be recognized by CD8+ or CD4+ T-cells, and trigger an immune response. Only a small fraction of the neoepitopes will be displayed by the MHC class I or II. One of the challenges of cancer immunotherapy is therefore to predict which neoepitopes are susceptible to elicit a T-cell response. Software tools such as netMHC, MHC Flury and many others are over-predictive as the bulk part of the data used to train these methods are based on affinity assays. Several publications have indicated that stability assays may provide data that better correlate with epitope presentation by MHC. Prediction tools trained on stability assays may therefore be better at selecting neoepitopes resulting in more effective cancer vaccine design. We performed stability assay measurements for 10 MHC class I and 10 MHC class II alleles using a peptide scan library approach. In brief, random 9-mers where one position is known were used to measure stability of the peptide MHC complex. Next, the data were used to train a prediction tool, PrDx, that relies on a combination of different machine learning methods (random forest, feed forward neural networks and recurrent neural networks), of which the outputs are gathered in an assemble model. The model was then further trained with peptides predicted to bind with high stability, to the MHC alleles, until satisfactory performances were attained. To our surprise, PrDx showed new binding patterns for the alleles we trained. Although mostly similar to the binding patterns seen with affinity data trained method, the stability trained method is able to show new important positions in the binding patterns of the peptide-MHC complexes. Through retrospective analysis, our method seems able to select more accurately peptides susceptible to elicit a T-cell response, compared to state-of-the-art epitope prediction methods. Our results suggest that PrDx may be an attractive prediction tool for neo-epitopes discovery. Citation Format: Stephan Thorgrimsen, Sune Justesen, Nicolas Rapin. Development of prediction software PrDx, trained on peptide-MHC stability assays, shows new important positions in the binding patterns of the peptides-MHC I and II complexes [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 B093.
癌细胞中的突变可能导致新表位的形成,这些新表位可能由主要组织相容性复合体(MHC) I类或II类呈现。这些新表位可能被CD8+或CD4+ t细胞识别,并引发免疫反应。只有一小部分新表位会被MHC I类或II类所显示。因此,癌症免疫治疗的挑战之一是预测哪些新表位容易引起t细胞反应。netMHC、MHC Flury等软件工具的预测能力太强,因为用于训练这些方法的大部分数据都是基于亲和性分析。一些出版物表明,稳定性测定可以提供更好地与MHC表位呈递相关的数据。因此,经过稳定性试验训练的预测工具可能更擅长选择新表位,从而设计出更有效的癌症疫苗。我们使用肽扫描文库方法对10个MHC I类和10个MHC II类等位基因进行了稳定性测定。简而言之,随机9-mers,其中一个位置是已知的,用于测量肽MHC复合物的稳定性。接下来,这些数据被用来训练一个预测工具PrDx,它依赖于不同机器学习方法(随机森林、前馈神经网络和循环神经网络)的组合,其中的输出被收集在一个组装模型中。然后用预测与MHC等位基因结合具有高稳定性的肽进一步训练模型,直到获得满意的性能。令我们惊讶的是,PrDx显示了我们训练的等位基因的新结合模式。虽然与亲和数据训练方法所见的结合模式大致相似,但稳定性训练方法能够在肽- mhc复合物的结合模式中显示新的重要位置。通过回顾性分析,与最先进的表位预测方法相比,我们的方法似乎能够更准确地选择易引起t细胞反应的肽。我们的研究结果表明,PrDx可能是发现新表位的一个有吸引力的预测工具。引文格式:Stephan Thorgrimsen, Sune Justesen, Nicolas Rapin。基于肽- mhc稳定性分析的预测软件PrDx的开发,显示了肽- mhc I和II复合物结合模式中新的重要位置[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 B093。
{"title":"Abstract B093: Development of prediction software PrDx, trained on peptide-MHC stability assays, shows new important positions in the binding patterns of the peptides-MHC I and II complexes","authors":"Stephan Thorgrimsen, S. Justesen, N. Rapin","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B093","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B093","url":null,"abstract":"Mutations in cancer cells may lead to the formation of neo-epitopes potentially presented by both major histocompatibility complex (MHC) class I or II. These neo-epitopes may be recognized by CD8+ or CD4+ T-cells, and trigger an immune response. Only a small fraction of the neoepitopes will be displayed by the MHC class I or II. One of the challenges of cancer immunotherapy is therefore to predict which neoepitopes are susceptible to elicit a T-cell response. Software tools such as netMHC, MHC Flury and many others are over-predictive as the bulk part of the data used to train these methods are based on affinity assays. Several publications have indicated that stability assays may provide data that better correlate with epitope presentation by MHC. Prediction tools trained on stability assays may therefore be better at selecting neoepitopes resulting in more effective cancer vaccine design. We performed stability assay measurements for 10 MHC class I and 10 MHC class II alleles using a peptide scan library approach. In brief, random 9-mers where one position is known were used to measure stability of the peptide MHC complex. Next, the data were used to train a prediction tool, PrDx, that relies on a combination of different machine learning methods (random forest, feed forward neural networks and recurrent neural networks), of which the outputs are gathered in an assemble model. The model was then further trained with peptides predicted to bind with high stability, to the MHC alleles, until satisfactory performances were attained. To our surprise, PrDx showed new binding patterns for the alleles we trained. Although mostly similar to the binding patterns seen with affinity data trained method, the stability trained method is able to show new important positions in the binding patterns of the peptide-MHC complexes. Through retrospective analysis, our method seems able to select more accurately peptides susceptible to elicit a T-cell response, compared to state-of-the-art epitope prediction methods. Our results suggest that PrDx may be an attractive prediction tool for neo-epitopes discovery. Citation Format: Stephan Thorgrimsen, Sune Justesen, Nicolas Rapin. Development of prediction software PrDx, trained on peptide-MHC stability assays, shows new important positions in the binding patterns of the peptides-MHC I and II complexes [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 B093.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"19 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":"121987433","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 B094: Successful identification of neoantigen-specific T-cell responses in low mutation burden colorectal cancers for personalized cancer vaccine development B094:成功鉴定低突变负担结直肠癌中新抗原特异性t细胞应答,用于个体化癌症疫苗的开发
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B094
Jitske van den Bulk, D. Ruano, M. Ijsselsteijn, Marten Visser, R. V. D. Breggen, K. Peeters, T. Duhen, Rebekka Duhen, A. Weinberg, S. V. D. Burg, E. Verdegaal, N. Miranda
Innovative treatment options are required to improve cure rates in advanced colorectal cancer patients. Immune checkpoint blockade therapy (anti-PD-1) was shown to be effective in colorectal cancers with high mutation burden (e.g., mismatch repair deficient) as antitumor reactivity is largely explained by the recognition of somatically mutated antigens (neoantigens). No immunotherapeutic strategies are currently available for patients diagnosed with low mutation burden colorectal cancer. We hypothesized that if neoantigen-reactive T-cells are present in low mutation burden patients, the latter could benefit from immunotherapeutic interventions that stimulate neoantigen recognition and the onset of a robust antitumor immune response.In order to detect neoantigens, whole exome and RNA next-generation sequencing analyses were performed in cancer and healthy tissues from five colorectal cancer patients. Corresponding neoepitopes were synthesized and tested for their ability to induce immune cell activation in T-cells isolated from the tumor tissue (TIL) and from peripheral blood. Neoantigen-specific T-cell responses were identified in the majority of patients that presented with tumors carrying 25 to 36 transcribed, non-synonymous variants. Up to six different neoantigens were recognized per tumor, which resulted in a higher detection rate than anticipated based on published data. Moreover, we discovered the merits of isolating CD39+CD103+CD8+ T-cells for detection of a broad recognition of HLA class I-restricted neoantigens. This CD39+CD103+CD8+ T-cell subset comprises the majority and a broader repertoire of neoantigen-specific T-cells compared to bulk TIL populations or lymphocytes derived from peripheral blood. In conclusion, we developed a neoantigen screening pipeline to unlock the immunogenic potential of colorectal cancers with low mutation burden. We have detected a relatively high number of neoantigens that are recognized by tumor- and/or PBMC-derived T-cells in mismatch repair proficient, low mutation burden colorectal cancer patients, and show the importance of the CD39+CD103+CD8+ T-cell subset for neoantigen-based immunotherapies. These findings warrant the further exploration of the potential to employ neoantigen-targeted therapies to improve clinical outcomes of colorectal cancer patients. Citation Format: Jitske van den Bulk, Dina Ruano, Marieke E. Ijsselsteijn, Marten Visser, Ruud van der Breggen, Koen C.M.J. Peeters, Thomas Duhen, Rebekka Duhen, Andrew D Weinberg, Sjoerd S.H. van der Burg, Els M.E. Verdegaal, Noel F. de Miranda. Successful identification of neoantigen-specific T-cell responses in low mutation burden colorectal cancers for personalized cancer vaccine development [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 B094.
需要创新的治疗方案来提高晚期结直肠癌患者的治愈率。免疫检查点阻断疗法(抗pd -1)被证明对具有高突变负担(例如错配修复缺陷)的结直肠癌有效,因为抗肿瘤反应性在很大程度上可以通过识别体细胞突变抗原(新抗原)来解释。目前尚无免疫治疗策略可用于诊断为低突变负担结直肠癌的患者。我们假设,如果新抗原反应性t细胞存在于低突变负担患者中,后者可能受益于免疫治疗干预,刺激新抗原识别和强大的抗肿瘤免疫反应的发生。为了检测新抗原,对5例结直肠癌患者的癌组织和健康组织进行了全外显子组和RNA新一代测序分析。合成了相应的新表位,并测试了它们在从肿瘤组织(TIL)和外周血中分离的t细胞中诱导免疫细胞激活的能力。在大多数携带25 - 36个转录的非同义变体的肿瘤患者中,发现了新抗原特异性t细胞反应。每个肿瘤可识别多达六种不同的新抗原,这使得检出率高于基于已发表数据的预期。此外,我们发现分离CD39+CD103+CD8+ t细胞用于检测广泛识别的HLA i类限制性新抗原的优点。与大量TIL群体或来自外周血的淋巴细胞相比,CD39+CD103+CD8+ t细胞亚群包括大多数和更广泛的新抗原特异性t细胞库。总之,我们开发了一种新抗原筛选管道,以解锁低突变负担结直肠癌的免疫原性潜力。我们已经在错配修复熟练、低突变负担的结直肠癌患者中检测到相对大量的肿瘤和/或pbmc衍生的t细胞识别的新抗原,并显示了CD39+CD103+CD8+ t细胞亚群对基于新抗原的免疫治疗的重要性。这些发现为进一步探索利用新抗原靶向治疗来改善结直肠癌患者的临床结果提供了依据。引用格式:Jitske van den Bulk, Dina Ruano, Marieke E. Ijsselsteijn, Marten Visser, Ruud van der Breggen, Koen C.M.J. Peeters, Thomas Duhen, Rebekka Duhen, Andrew D Weinberg, Sjoerd S.H. van der Burg, Els M.E. Verdegaal, Noel F. de Miranda。成功鉴定低突变负担结直肠癌中新抗原特异性t细胞反应,用于个体化癌症疫苗的开发[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 B094。
{"title":"Abstract B094: Successful identification of neoantigen-specific T-cell responses in low mutation burden colorectal cancers for personalized cancer vaccine development","authors":"Jitske van den Bulk, D. Ruano, M. Ijsselsteijn, Marten Visser, R. V. D. Breggen, K. Peeters, T. Duhen, Rebekka Duhen, A. Weinberg, S. V. D. Burg, E. Verdegaal, N. Miranda","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B094","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B094","url":null,"abstract":"Innovative treatment options are required to improve cure rates in advanced colorectal cancer patients. Immune checkpoint blockade therapy (anti-PD-1) was shown to be effective in colorectal cancers with high mutation burden (e.g., mismatch repair deficient) as antitumor reactivity is largely explained by the recognition of somatically mutated antigens (neoantigens). No immunotherapeutic strategies are currently available for patients diagnosed with low mutation burden colorectal cancer. We hypothesized that if neoantigen-reactive T-cells are present in low mutation burden patients, the latter could benefit from immunotherapeutic interventions that stimulate neoantigen recognition and the onset of a robust antitumor immune response.In order to detect neoantigens, whole exome and RNA next-generation sequencing analyses were performed in cancer and healthy tissues from five colorectal cancer patients. Corresponding neoepitopes were synthesized and tested for their ability to induce immune cell activation in T-cells isolated from the tumor tissue (TIL) and from peripheral blood. Neoantigen-specific T-cell responses were identified in the majority of patients that presented with tumors carrying 25 to 36 transcribed, non-synonymous variants. Up to six different neoantigens were recognized per tumor, which resulted in a higher detection rate than anticipated based on published data. Moreover, we discovered the merits of isolating CD39+CD103+CD8+ T-cells for detection of a broad recognition of HLA class I-restricted neoantigens. This CD39+CD103+CD8+ T-cell subset comprises the majority and a broader repertoire of neoantigen-specific T-cells compared to bulk TIL populations or lymphocytes derived from peripheral blood. In conclusion, we developed a neoantigen screening pipeline to unlock the immunogenic potential of colorectal cancers with low mutation burden. We have detected a relatively high number of neoantigens that are recognized by tumor- and/or PBMC-derived T-cells in mismatch repair proficient, low mutation burden colorectal cancer patients, and show the importance of the CD39+CD103+CD8+ T-cell subset for neoantigen-based immunotherapies. These findings warrant the further exploration of the potential to employ neoantigen-targeted therapies to improve clinical outcomes of colorectal cancer patients. Citation Format: Jitske van den Bulk, Dina Ruano, Marieke E. Ijsselsteijn, Marten Visser, Ruud van der Breggen, Koen C.M.J. Peeters, Thomas Duhen, Rebekka Duhen, Andrew D Weinberg, Sjoerd S.H. van der Burg, Els M.E. Verdegaal, Noel F. de Miranda. Successful identification of neoantigen-specific T-cell responses in low mutation burden colorectal cancers for personalized cancer vaccine development [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 B094.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"47 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":"127925979","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 B087: MHC class II on cancer cells—Role in response to BCG therapy of bladder cancer B087: MHC II类在膀胱癌卡介苗治疗应答中的作用
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B087
Gil Redelman-Sidi, M. Glickman, A. Binyamin, A. Antonelli
BCG, a live mycobacterium also used worldwide as a vaccine against tuberculosis, is one of the first successful immunotherapies of cancer. Despite being in clinical use for the treatment of non-muscle-invasive bladder cancer (NMIBC) for 4 decades, the mechanism by which BCG activates the immune system to destroy cancer cells is poorly understood and there are no reliable methods to predict a patient’s response to BCG treatment. Current data suggest that the efficacy of BCG depends on initial attachment of BCG to bladder cancer cells leading to activation of an immune response that is T-cell dependent. It is not known whether bladder cancer cells have a direct role in activating T-cells. Using an orthotopic mouse model of BCG treatment of bladder cancer we show that: 1) BCG induces tumor-specific immunity; 2) CD4+ T-cells are the main requirement for BCG efficacy and for the tumor-specific immunity engendered by BCG therapy;3) bladder cancer cells can directly activate CD4+ T-cells; 4) the efficacy of BCG depends on MHC class II expression on the surface of bladder cancer cells. These data suggest that bladder cancer cells directly activate and drive a CD4+ T-cell dependent immune response that is required for the efficacy of BCG. Current work in the lab is focused on characterizing MHC class II expression on human NMIBC specimens and determining whether expression of MHC class II on bladder cancer cells predicts clinical outcomes after BCG therapy. Citation Format: Gil Redelman-Sidi, Michael Glickman, Anna Binyamin, Anthony Antonelli. MHC class II on cancer cells—Role in response to BCG therapy of bladder 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 B087.
卡介苗是一种活的分枝杆菌,在世界范围内也被用作结核病疫苗,它是最早成功的癌症免疫疗法之一。尽管卡介苗用于治疗非肌肉侵袭性膀胱癌(NMIBC)已有40年的临床应用,但人们对卡介苗激活免疫系统破坏癌细胞的机制知之甚少,也没有可靠的方法来预测患者对卡介苗治疗的反应。目前的数据表明,卡介苗的疗效取决于卡介苗与膀胱癌细胞的初始附着,导致t细胞依赖性免疫反应的激活。目前尚不清楚膀胱癌细胞是否在激活t细胞中起直接作用。利用卡介苗治疗膀胱癌的原位小鼠模型,我们发现:1)卡介苗诱导肿瘤特异性免疫;2) CD4+ t细胞是卡介苗发挥作用的主要条件,也是卡介苗治疗产生肿瘤特异性免疫的主要条件;3)膀胱癌细胞可直接激活CD4+ t细胞;4)卡介苗的疗效取决于膀胱癌细胞表面MHC II类的表达。这些数据表明,膀胱癌细胞直接激活并驱动CD4+ t细胞依赖的免疫反应,这是卡介苗疗效所必需的。实验室目前的工作重点是表征人类NMIBC标本中MHC II类的表达,并确定膀胱癌细胞中MHC II类的表达是否能预测卡介苗治疗后的临床结果。引用格式:Gil Redelman-Sidi, Michael Glickman, Anna Binyamin, Anthony Antonelli。MHCⅱ类对癌细胞的作用——在膀胱癌卡介苗治疗应答中的作用[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志,2019;7(2增刊):摘要nr B087。
{"title":"Abstract B087: MHC class II on cancer cells—Role in response to BCG therapy of bladder cancer","authors":"Gil Redelman-Sidi, M. Glickman, A. Binyamin, A. Antonelli","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B087","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B087","url":null,"abstract":"BCG, a live mycobacterium also used worldwide as a vaccine against tuberculosis, is one of the first successful immunotherapies of cancer. Despite being in clinical use for the treatment of non-muscle-invasive bladder cancer (NMIBC) for 4 decades, the mechanism by which BCG activates the immune system to destroy cancer cells is poorly understood and there are no reliable methods to predict a patient’s response to BCG treatment. Current data suggest that the efficacy of BCG depends on initial attachment of BCG to bladder cancer cells leading to activation of an immune response that is T-cell dependent. It is not known whether bladder cancer cells have a direct role in activating T-cells. Using an orthotopic mouse model of BCG treatment of bladder cancer we show that: 1) BCG induces tumor-specific immunity; 2) CD4+ T-cells are the main requirement for BCG efficacy and for the tumor-specific immunity engendered by BCG therapy;3) bladder cancer cells can directly activate CD4+ T-cells; 4) the efficacy of BCG depends on MHC class II expression on the surface of bladder cancer cells. These data suggest that bladder cancer cells directly activate and drive a CD4+ T-cell dependent immune response that is required for the efficacy of BCG. Current work in the lab is focused on characterizing MHC class II expression on human NMIBC specimens and determining whether expression of MHC class II on bladder cancer cells predicts clinical outcomes after BCG therapy. Citation Format: Gil Redelman-Sidi, Michael Glickman, Anna Binyamin, Anthony Antonelli. MHC class II on cancer cells—Role in response to BCG therapy of bladder 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 B087.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","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":"116625403","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 IA34: Mapping immune recognition of non-self neoantigens in human pancreatic cancer 摘要:人类胰腺癌非自体新抗原的免疫识别图谱
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA34
M. Luksza, Joanne Leung, Alexander Solovyov, D. Redmond, M. Merad, S. Gnjatic, Christine Iacubuzio-Donohue, R. DeMatteo, T. Chan, J. Wolchock, S. Leach, B. Greenbaum, T. Merghoub, V. Balachandran
Endogenous T-cell responses to cancer-specific mutations (neoantigens) are a common denominator of successful immunotherapies. However, endogenous neoantigen-specific T cell immunity in immunotherapy refractory tumors remains poorly characterized. Pancreatic ductal adenocarcinoma (PDAC) is the prototypical immunotherapy refractory cancer: response rates to single-agent checkpoint blockade are 5 years, T cell immunity has been linked to their exceptional outcome however the relevant antigens remained unknown. We sought to identify the landscape, antigenic potential, and T cell functional states induced by neoantigens in rare long-term pancreatic cancer survivors. Using genetic, immunologic, and computational techniques, we recently reported that tumors with the highest predicted neoantigen number and the greatest density of CD3+CD8+ infiltrates together, but neither parameter alone, could identify long-term survivors, suggesting differential inherent neoantigen immunogenicity. To investigate the specific neoantigen qualities promoting differential immunogenicity, we developed a fitness model that quantifies neoantigen immunogenicity based on estimations of the relative MHC binding affinity of each neoantigen to its wild type counterpart, as well as a nonlinear dependence on sequence similarity of neoantigens to known antigens. Our neoantigen quality fitness model identified long-term survivors in two independent PDAC datasets, as well as predicted survival in anti-CTLA4 treated melanoma patients, and anti-PD-1 treated lung cancer patients. In long-term PDAC survivors, we detected high quality neoantigen-specific intratumoral T cell clones persisting in the blood up to 12 years after primary tumor removal, with both unique clonal and phenotypic profiles. Our results identify neoantigens with unique qualities as T cell targets in both endogenous and immunotherapy treated cancers. Neoantigen quality can therefore inform rational neoantigen immunogenicity predictions that may guide patient and target selection for immunotherapies. Citation Format: Marta Luksza, Joanne P. Leung, Alexander Solovyov, David Redmond, Miriam Merad, Sacha Gnjatic, Christine Iacubuzio-Donohue, Ronald P. DeMatteo, Timothy A. Chan, Jedd Wolchock, Steven D. Leach, Benjamin D. Greenbaum, Taha Merghoub, Vinod P. Balachandran. Mapping immune recognition of non-self neoantigens in human pancreatic 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 IA34.
内源性t细胞对癌症特异性突变(新抗原)的反应是成功免疫疗法的共同特征。然而,免疫治疗难治性肿瘤的内源性新抗原特异性T细胞免疫特性仍然很差。胰腺导管腺癌(PDAC)是一种典型的免疫治疗难治性癌症:单药检查点阻断的应答率为5年,T细胞免疫与它们的特殊结果有关,但相关抗原仍然未知。我们试图在罕见的长期胰腺癌幸存者中确定新抗原诱导的景观、抗原潜能和T细胞功能状态。利用遗传学、免疫学和计算技术,我们最近报道了具有最高预测新抗原数量和最高CD3+CD8+浸润密度的肿瘤,但这两个参数都不能单独识别长期幸存者,这表明不同的固有新抗原免疫原性。为了研究促进差异免疫原性的特异性新抗原质量,我们建立了一个适应度模型,该模型基于每个新抗原与其野生型对应物的相对MHC结合亲和力的估计,以及新抗原与已知抗原序列相似性的非线性依赖,来量化新抗原的免疫原性。我们的新抗原质量适应度模型在两个独立的PDAC数据集中确定了长期幸存者,并预测了抗ctla4治疗的黑色素瘤患者和抗pd -1治疗的肺癌患者的生存。在长期的PDAC幸存者中,我们检测到高质量的新抗原特异性肿瘤内T细胞克隆在原发性肿瘤切除后持续存在长达12年,具有独特的克隆和表型谱。我们的研究结果确定了具有独特品质的新抗原,作为内源性和免疫治疗癌症的T细胞靶标。因此,新抗原的质量可以为合理的新抗原免疫原性预测提供信息,从而指导患者和免疫治疗靶点的选择。引用格式:Marta Luksza, Joanne P. Leung, Alexander Solovyov, David Redmond, Miriam Merad, Sacha Gnjatic, Christine Iacubuzio-Donohue, Ronald P. DeMatteo, Timothy A. Chan, Jedd Wolchock, Steven D. Leach, Benjamin D. Greenbaum, Taha Merghoub, Vinod P. Balachandran人胰腺癌非自体新抗原的免疫识别图谱[摘要]。第四届CRI-CIMT-EATI-AACR国际癌症免疫治疗会议:将科学转化为生存;2018年9月30日至10月3日;纽约,纽约。费城(PA): AACR;癌症免疫学杂志,2019;7(2增刊):摘要nr - IA34。
{"title":"Abstract IA34: Mapping immune recognition of non-self neoantigens in human pancreatic cancer","authors":"M. Luksza, Joanne Leung, Alexander Solovyov, D. Redmond, M. Merad, S. Gnjatic, Christine Iacubuzio-Donohue, R. DeMatteo, T. Chan, J. Wolchock, S. Leach, B. Greenbaum, T. Merghoub, V. Balachandran","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA34","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA34","url":null,"abstract":"Endogenous T-cell responses to cancer-specific mutations (neoantigens) are a common denominator of successful immunotherapies. However, endogenous neoantigen-specific T cell immunity in immunotherapy refractory tumors remains poorly characterized. Pancreatic ductal adenocarcinoma (PDAC) is the prototypical immunotherapy refractory cancer: response rates to single-agent checkpoint blockade are 5 years, T cell immunity has been linked to their exceptional outcome however the relevant antigens remained unknown. We sought to identify the landscape, antigenic potential, and T cell functional states induced by neoantigens in rare long-term pancreatic cancer survivors. Using genetic, immunologic, and computational techniques, we recently reported that tumors with the highest predicted neoantigen number and the greatest density of CD3+CD8+ infiltrates together, but neither parameter alone, could identify long-term survivors, suggesting differential inherent neoantigen immunogenicity. To investigate the specific neoantigen qualities promoting differential immunogenicity, we developed a fitness model that quantifies neoantigen immunogenicity based on estimations of the relative MHC binding affinity of each neoantigen to its wild type counterpart, as well as a nonlinear dependence on sequence similarity of neoantigens to known antigens. Our neoantigen quality fitness model identified long-term survivors in two independent PDAC datasets, as well as predicted survival in anti-CTLA4 treated melanoma patients, and anti-PD-1 treated lung cancer patients. In long-term PDAC survivors, we detected high quality neoantigen-specific intratumoral T cell clones persisting in the blood up to 12 years after primary tumor removal, with both unique clonal and phenotypic profiles. Our results identify neoantigens with unique qualities as T cell targets in both endogenous and immunotherapy treated cancers. Neoantigen quality can therefore inform rational neoantigen immunogenicity predictions that may guide patient and target selection for immunotherapies. Citation Format: Marta Luksza, Joanne P. Leung, Alexander Solovyov, David Redmond, Miriam Merad, Sacha Gnjatic, Christine Iacubuzio-Donohue, Ronald P. DeMatteo, Timothy A. Chan, Jedd Wolchock, Steven D. Leach, Benjamin D. Greenbaum, Taha Merghoub, Vinod P. Balachandran. Mapping immune recognition of non-self neoantigens in human pancreatic 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 IA34.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"68 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":"126341385","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 PR11: CX3CR1+CD8+ T-cells are responsible to the clinical benefit of chemoimmunotherapy in metastatic melanoma patients after disease progression on PD-1 blockade PR11: CX3CR1+CD8+ t细胞是PD-1阻断后疾病进展的转移性黑色素瘤患者化疗免疫治疗的临床获益的原因
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR11
Yiyi Yan, Haidong Dong, R. Dronca, S. Markovic
Background: Clinical management of metastatic melanoma (MM) after PD-1 blockade failure remains a challenge and lacks a standard of care. Immunotherapy or chemotherapy alone provides limited benefit in this setting. Chemo-immunotherapy (CIT) combinations have demonstrated favorable efficacy and safety profiles in lung cancer patients. Our preclinical study in MM has shown that the addition of chemotherapy to PD-1 blockade can reshape a subset of therapy-responsive CD8+ T-cells with resultant enhanced antitumor activities, suggesting the potential clinical benefits of CIT in MM patients whose disease have progressed after anti-PD-1 therapy. Further understanding of the clinical benefits and immunoregulatory mechanisms of CIT in this setting is crucial for the development of optimal combinatorial chemo-immunotherapeutic strategies to improve clinical outcomes in patients with advanced cancer. Methods: MM patients (n=22) who have failed PD-1 blockade therapy were subsequently treated with CIT (paclitaxel and carboplatin in combination with pembrolizumab). The overall survival (OS), objective response rate (ORR), time-to-next therapy (TTNT), and toxicities were assessed. Using peripheral blood (PB) from MM patients, the phenotypic and functional changes induced by chemotherapy in therapy-responsive T-cells, in the setting of anti-PD1 therapy, were examined. The immunoregulatory effects of CIT were also examined in melanoma mouse model. Results: MM patients who have received subsequent CIT had a median OS of 5 years (95% CI: 2-NR) (median follow up of 3.9 years), with ORR of 61% (CR of 23%). The median TTNT was 8 months (95% CI: 6-15). No additional toxicities were identified. CX3CR1+CD8+ therapy-responsive T-cells are low in MM patients who have failed to respond to anti-PD-1 monotherapy. However, in MM patients who responded to subsequent CIT, this subset of therapy-responsive T-cells survived the chemotherapy with increased frequency and enhanced function. The clinical benefit of CIT is only observed in CX3CR1 wild type mice, not in KO mice, and ongoing PD-1 blockade is necessary to improve its anti-tumor activities. Conclusion: In MM patients who have failed anti-PD-1 therapy, the chemo-immunotherapy combination showed favorable clinical outcomes and an acceptable toxicity profile. CX3CR1+ CD8+ effector T-cells are responsible for the clinical benefit of CIT. This novel therapy-responsive population underlies the key cellular and molecular immunoregulatory mechanisms of chemotherapy. It serves as a meaningful marker to measure these collaborative effects and to develop the optimal chemo-immunotherapy strategy to improve clinical responses to current immune checkpoint blocking agents. Citation Format: Yiyi Yan, Haidong Dong, Roxana Dronca, Svetomir Markovic. CX3CR1+CD8+ T-cells are responsible to the clinical benefit of chemoimmunotherapy in metastatic melanoma patients after disease progression on PD-1 blockade [abstract]. In: Proceedings of the F
背景:PD-1阻断失败后转移性黑色素瘤(MM)的临床管理仍然是一个挑战,缺乏标准的护理。在这种情况下,单独免疫治疗或化疗的疗效有限。化疗-免疫治疗(CIT)联合治疗在肺癌患者中显示出良好的疗效和安全性。我们在MM的临床前研究表明,在PD-1阻断的基础上增加化疗可以重塑治疗应答的CD8+ t细胞亚群,从而增强抗肿瘤活性,这表明CIT在抗PD-1治疗后疾病进展的MM患者中具有潜在的临床益处。在这种情况下,进一步了解CIT的临床益处和免疫调节机制对于制定最佳的化疗-免疫治疗组合策略以改善晚期癌症患者的临床结果至关重要。方法:PD-1阻断治疗失败的MM患者(n=22)随后接受CIT(紫杉醇和卡铂联合派姆单抗)治疗。评估总生存期(OS)、客观缓解率(ORR)、下一次治疗时间(TTNT)和毒副反应。利用MM患者外周血(PB),在抗pd1治疗的背景下,检测化疗引起的治疗反应性t细胞的表型和功能变化。在黑素瘤小鼠模型中检测了CIT的免疫调节作用。结果:接受后续CIT治疗的MM患者的中位OS为5年(95% CI: 2-NR)(中位随访时间为3.9年),ORR为61% (CR为23%)。中位TTNT为8个月(95% CI: 6-15)。没有发现其他的毒性。在抗pd -1单药治疗无效的MM患者中,CX3CR1+CD8+治疗反应性t细胞较低。然而,在对随后的CIT有反应的MM患者中,这种治疗反应性t细胞亚群在化疗中存活下来,频率增加,功能增强。CIT的临床获益仅在CX3CR1野生型小鼠中观察到,未在KO小鼠中观察到,需要持续阻断PD-1以提高其抗肿瘤活性。结论:在抗pd -1治疗失败的MM患者中,化疗-免疫联合治疗显示出良好的临床结果和可接受的毒性特征。CX3CR1+ CD8+效应t细胞负责CIT的临床获益,这种新型治疗反应性群体是化疗关键的细胞和分子免疫调节机制的基础。它可以作为一个有意义的标志物来衡量这些协同效应,并制定最佳的化学免疫治疗策略,以提高对当前免疫检查点阻断剂的临床反应。引文格式:闫依依,董海东,Roxana Dronca, Svetomir Markovic。CX3CR1+CD8+ t细胞是PD-1阻断后疾病进展的转移性黑色素瘤患者化疗免疫治疗的临床获益的原因[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 PR11。
{"title":"Abstract PR11: CX3CR1+CD8+ T-cells are responsible to the clinical benefit of chemoimmunotherapy in metastatic melanoma patients after disease progression on PD-1 blockade","authors":"Yiyi Yan, Haidong Dong, R. Dronca, S. Markovic","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR11","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR11","url":null,"abstract":"Background: Clinical management of metastatic melanoma (MM) after PD-1 blockade failure remains a challenge and lacks a standard of care. Immunotherapy or chemotherapy alone provides limited benefit in this setting. Chemo-immunotherapy (CIT) combinations have demonstrated favorable efficacy and safety profiles in lung cancer patients. Our preclinical study in MM has shown that the addition of chemotherapy to PD-1 blockade can reshape a subset of therapy-responsive CD8+ T-cells with resultant enhanced antitumor activities, suggesting the potential clinical benefits of CIT in MM patients whose disease have progressed after anti-PD-1 therapy. Further understanding of the clinical benefits and immunoregulatory mechanisms of CIT in this setting is crucial for the development of optimal combinatorial chemo-immunotherapeutic strategies to improve clinical outcomes in patients with advanced cancer. Methods: MM patients (n=22) who have failed PD-1 blockade therapy were subsequently treated with CIT (paclitaxel and carboplatin in combination with pembrolizumab). The overall survival (OS), objective response rate (ORR), time-to-next therapy (TTNT), and toxicities were assessed. Using peripheral blood (PB) from MM patients, the phenotypic and functional changes induced by chemotherapy in therapy-responsive T-cells, in the setting of anti-PD1 therapy, were examined. The immunoregulatory effects of CIT were also examined in melanoma mouse model. Results: MM patients who have received subsequent CIT had a median OS of 5 years (95% CI: 2-NR) (median follow up of 3.9 years), with ORR of 61% (CR of 23%). The median TTNT was 8 months (95% CI: 6-15). No additional toxicities were identified. CX3CR1+CD8+ therapy-responsive T-cells are low in MM patients who have failed to respond to anti-PD-1 monotherapy. However, in MM patients who responded to subsequent CIT, this subset of therapy-responsive T-cells survived the chemotherapy with increased frequency and enhanced function. The clinical benefit of CIT is only observed in CX3CR1 wild type mice, not in KO mice, and ongoing PD-1 blockade is necessary to improve its anti-tumor activities. Conclusion: In MM patients who have failed anti-PD-1 therapy, the chemo-immunotherapy combination showed favorable clinical outcomes and an acceptable toxicity profile. CX3CR1+ CD8+ effector T-cells are responsible for the clinical benefit of CIT. This novel therapy-responsive population underlies the key cellular and molecular immunoregulatory mechanisms of chemotherapy. It serves as a meaningful marker to measure these collaborative effects and to develop the optimal chemo-immunotherapy strategy to improve clinical responses to current immune checkpoint blocking agents. Citation Format: Yiyi Yan, Haidong Dong, Roxana Dronca, Svetomir Markovic. CX3CR1+CD8+ T-cells are responsible to the clinical benefit of chemoimmunotherapy in metastatic melanoma patients after disease progression on PD-1 blockade [abstract]. In: Proceedings of the F","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"42 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":"130848537","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 B076: Pan-cancer analysis to identify which cancers may benefit most from LAG-3 blockade B076:泛癌症分析确定哪些癌症可能从LAG-3阻断中获益最多
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B076
S. Ganesan, Anshuman Panda, G. Bhanot
Immune checkpoint blockade using antibodies that target programmed-cell-death-1 (PD-1) or its ligand programmed-cell-death-ligand 1 (PD-L1), and/or cytotoxic-T-lymphocyte-associated-protein-4 (CTLA-4) have led to dramatic responses, but only in a subset of human cancers. An antibody targeting lymphocyte-activation-gene-3 (LAG-3), relatlimab (BMS-986016), recently showed significant clinical activity in melanoma, and early clinical data showed that LAG-3 expression is a biomarker of response to this agent. We performed a pan-cancer analysis of The Cancer Genome Atlas data to identify genomic and immunologic correlates of LAG-3 expression. Hyper-mutation, and exogenous (Epstein-Barr virus, human papillomavirus) or endogenous (endogenous retrovirus group 3 member 2) viral expression in tumor were associated with increased expression of LAG-3 in multiple cancer types. LAG-3 expression was also correlated with CD8A and PD-L1 expression in most cancer types; however, there were notable exceptions, including head-neck squamous-cell cancer (HNSC), renal cell cancer (RCC) and glioblastoma. In HNSC, LAG-3 expression, but not PD-L1, was significantly increased in HPV+ cancers. In RCC and gliobastoma, LAG-3 was more strongly associated with CD8A expression than PD-L1. These results may have implications on the design of future clinical trials of LAG-3 blockade. Citation Format: Shridar Ganesan, Anshuman Panda, Gyan Bhanot. Pan-cancer analysis to identify which cancers may benefit most from LAG-3 blockade [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B076.
使用靶向程序性细胞死亡-1 (PD-1)或其配体程序性细胞死亡-配体1 (PD-L1)和/或细胞毒性t淋巴细胞相关蛋白-4 (CTLA-4)的抗体阻断免疫检查点已经导致了戏剧性的反应,但仅在人类癌症的一小部分中。一种靶向淋巴细胞激活基因3 (LAG-3)的抗体relatlimab (BMS-986016)最近在黑色素瘤中显示出显著的临床活性,早期临床数据显示LAG-3的表达是对该药物反应的生物标志物。我们对癌症基因组图谱数据进行了泛癌症分析,以确定LAG-3表达的基因组和免疫学相关性。在多种癌症类型中,超突变、外源性(eb病毒、人乳头瘤病毒)或内源性(内源性逆转录病毒组3成员2)病毒在肿瘤中的表达与LAG-3表达增加相关。在大多数癌症类型中,LAG-3的表达也与CD8A和PD-L1的表达相关;然而,也有明显的例外,包括头颈部鳞状细胞癌(HNSC)、肾细胞癌(RCC)和胶质母细胞瘤。在HNSC中,LAG-3的表达在HPV+癌中显著增加,而PD-L1的表达没有显著增加。在RCC和胶质母细胞瘤中,LAG-3与CD8A表达的相关性强于PD-L1。这些结果可能对未来LAG-3阻断剂临床试验的设计有启示意义。引文格式:Shridar Ganesan, Anshuman Panda, Gyan Bhanot。泛癌症分析,以确定哪些癌症可能从LAG-3阻断中获益最多[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 B076。
{"title":"Abstract B076: Pan-cancer analysis to identify which cancers may benefit most from LAG-3 blockade","authors":"S. Ganesan, Anshuman Panda, G. Bhanot","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B076","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B076","url":null,"abstract":"Immune checkpoint blockade using antibodies that target programmed-cell-death-1 (PD-1) or its ligand programmed-cell-death-ligand 1 (PD-L1), and/or cytotoxic-T-lymphocyte-associated-protein-4 (CTLA-4) have led to dramatic responses, but only in a subset of human cancers. An antibody targeting lymphocyte-activation-gene-3 (LAG-3), relatlimab (BMS-986016), recently showed significant clinical activity in melanoma, and early clinical data showed that LAG-3 expression is a biomarker of response to this agent. We performed a pan-cancer analysis of The Cancer Genome Atlas data to identify genomic and immunologic correlates of LAG-3 expression. Hyper-mutation, and exogenous (Epstein-Barr virus, human papillomavirus) or endogenous (endogenous retrovirus group 3 member 2) viral expression in tumor were associated with increased expression of LAG-3 in multiple cancer types. LAG-3 expression was also correlated with CD8A and PD-L1 expression in most cancer types; however, there were notable exceptions, including head-neck squamous-cell cancer (HNSC), renal cell cancer (RCC) and glioblastoma. In HNSC, LAG-3 expression, but not PD-L1, was significantly increased in HPV+ cancers. In RCC and gliobastoma, LAG-3 was more strongly associated with CD8A expression than PD-L1. These results may have implications on the design of future clinical trials of LAG-3 blockade. Citation Format: Shridar Ganesan, Anshuman Panda, Gyan Bhanot. Pan-cancer analysis to identify which cancers may benefit most from LAG-3 blockade [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B076.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"34 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":"134524820","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 B071: Validating sequence similarity-driven neoepitope fitness models via immunogenomics on TCGA and multiregional tumor data B071:基于TCGA和多区域肿瘤数据的免疫基因组学验证序列相似性驱动的新表位适应度模型
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B071
A. Bubie, N. Akers, A. Villanueva, B. Losic
Clonal fitness and survival models for checkpoint blockade response prediction have recently been proposed on the basis of ranking neoepitopes via an interaction between a proxy of T-cell recognition, a nonlinear function of neoepitope sequence similarity to known antigens, and neoantigen relative MHC binding affinity. In this work we examine the SKCM (n = 337) and NSCLC (n = 305) TCGA datasets to explicitly test if sequence similarity to known antigens from the IEDB is associated with tumor infiltrating lymphocyte (TIL) burden in patients, as measured by TCR sequencing and CDR reconstruction in RNA-seq data. We find that there is no statistically significant association between either the inferred clonality or magnitude of TIL response and neoepitope sequence similarity to known antigens. We do, however, find significant, moderate associations of TIL response to neoepitope burden. Further, we examined the LIHC (n = 193) and UCEC (n = 245) TCGA cohorts to explicitly derive tumor and virally derived epitopes (HepB, HPV respectively) and ranked their relative predicted MHC binding affinity profiles. We find a greater MHC binding affinity bias exists towards neoepitopes compared to virally derived peptides in a natural setting where both viral and tumor antigens are simultaneously present. Moreover, we find low and significant associations between TIL burden and overall neoepitope burden, but no association with overall viral epitope or expression burden. Finally, we used multiregionally sampled data (12 patients, 72 regions) from HepB-positive HCC liver cancer patients to confirm preferential MHC binding affinity and TIL response bias towards neoepitopes still holds and is significant. Our results suggest that neoepitopes dominate in their recruitment potential of, and association with, TIL burden compared to viral-cofactors. They also suggest that neoepitope sequence similarity to known antigens does not recapitulate patient TIL burden to first approximation. Citation Format: Adrian Bubie, Nicholas Akers, Augusto Villanueva, Bojan Losic. Validating sequence similarity-driven neoepitope fitness models via immunogenomics on TCGA and multiregional tumor data [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 B071.
最近提出了用于检查点阻断反应预测的克隆适应度和生存模型,该模型基于t细胞识别代理、新表位序列与已知抗原相似性的非线性函数和新抗原相对MHC结合亲和力之间的相互作用对新表位进行排序。在这项工作中,我们检查了SKCM (n = 337)和NSCLC (n = 305)的TCGA数据集,通过TCR测序和RNA-seq数据中的CDR重建,明确测试了与来自IEDB的已知抗原的序列相似性是否与患者的肿瘤浸润性淋巴细胞(TIL)负担相关。我们发现,TIL反应的推断克隆性或大小与新表位序列与已知抗原的相似性之间没有统计学上的显著关联。然而,我们确实发现TIL对新表位负荷的反应有显著的、适度的关联。进一步,我们检查了LIHC (n = 193)和UCEC (n = 245) TCGA队列,明确推导出肿瘤和病毒衍生的表位(分别为HepB和HPV),并对它们的相对预测MHC结合亲和力谱进行了排序。我们发现,在病毒和肿瘤抗原同时存在的自然环境中,与病毒衍生肽相比,MHC结合亲和力偏向于新表位存在更大的差异。此外,我们发现TIL负担与总体新表位负担之间存在低而显著的相关性,但与总体病毒表位或表达负担无关。最后,我们使用来自hepb阳性HCC肝癌患者的多区域采样数据(12例患者,72个区域)来证实MHC的优先结合亲和力和TIL对新表位的反应偏倚仍然存在并且是显著的。我们的研究结果表明,与病毒辅助因子相比,新表位在TIL负荷的招募潜力和关联方面占主导地位。他们还表明,新表位序列与已知抗原的相似性并不能将患者的TIL负担概括为第一近似。引文格式:Adrian Bubie, Nicholas Akers, Augusto Villanueva, Bojan Losic。基于TCGA和多区域肿瘤数据的免疫基因组学验证序列相似性驱动的新表位适应度模型[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 B071。
{"title":"Abstract B071: Validating sequence similarity-driven neoepitope fitness models via immunogenomics on TCGA and multiregional tumor data","authors":"A. Bubie, N. Akers, A. Villanueva, B. Losic","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B071","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B071","url":null,"abstract":"Clonal fitness and survival models for checkpoint blockade response prediction have recently been proposed on the basis of ranking neoepitopes via an interaction between a proxy of T-cell recognition, a nonlinear function of neoepitope sequence similarity to known antigens, and neoantigen relative MHC binding affinity. In this work we examine the SKCM (n = 337) and NSCLC (n = 305) TCGA datasets to explicitly test if sequence similarity to known antigens from the IEDB is associated with tumor infiltrating lymphocyte (TIL) burden in patients, as measured by TCR sequencing and CDR reconstruction in RNA-seq data. We find that there is no statistically significant association between either the inferred clonality or magnitude of TIL response and neoepitope sequence similarity to known antigens. We do, however, find significant, moderate associations of TIL response to neoepitope burden. Further, we examined the LIHC (n = 193) and UCEC (n = 245) TCGA cohorts to explicitly derive tumor and virally derived epitopes (HepB, HPV respectively) and ranked their relative predicted MHC binding affinity profiles. We find a greater MHC binding affinity bias exists towards neoepitopes compared to virally derived peptides in a natural setting where both viral and tumor antigens are simultaneously present. Moreover, we find low and significant associations between TIL burden and overall neoepitope burden, but no association with overall viral epitope or expression burden. Finally, we used multiregionally sampled data (12 patients, 72 regions) from HepB-positive HCC liver cancer patients to confirm preferential MHC binding affinity and TIL response bias towards neoepitopes still holds and is significant. Our results suggest that neoepitopes dominate in their recruitment potential of, and association with, TIL burden compared to viral-cofactors. They also suggest that neoepitope sequence similarity to known antigens does not recapitulate patient TIL burden to first approximation. Citation Format: Adrian Bubie, Nicholas Akers, Augusto Villanueva, Bojan Losic. Validating sequence similarity-driven neoepitope fitness models via immunogenomics on TCGA and multiregional tumor data [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 B071.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"600 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":"132481484","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 B074: A recall antigen-based potency assay for immunomodulatory biologics that could discriminate responders from nonresponders B074:一种基于召回抗原的免疫调节生物制剂效价测定方法,可以区分应答者和无应答者
Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B074
M. Delcommenne, E. Kleiman, Wushouer Ouerkaxi, P. Daftarian
The surge of therapeutic regimens involving immune checkpoint antagonists and co-stimulatory pathways (ACP) antagonists used as single agents or in combination necessitates better functional screening assays. In vitro functional screening assays need to be non-invasive and high throughput. The recall antigen assay is arguably the one that, in an antigen specific controlled system, directly assesses the activity of potential immune checkpoint inhibitors (ICI) candidates, in a relatively physiologically relevant manner. The recall antigen assay has been often used in the past to prove the potency of ICI or ACP as indicated in publications and submissions to regulatory bodies. We have conducted a thorough optimization combined with modifications to the recall antigen assay to enable the need for a more effective and efficient assay.Our recall antigen assay has been optimized across multiple parameters including donor selection based on expression of co-inhibitory/co-stimulatory molecules. This facilitates the selection of the right donor for the right candidate. We have optimized both stimulation and readout components to enhance signal to noise ratio. For example, we reduced the duration of in vitro antigen specific CD8+ T-cell clonotype expansion from 2 weeks to 5 to 7 days. To maximize the direct readout, we have used an MHC Tetramer guided analysis, using a cocktail of MHC tetramers made with stimulating peptides. To further empower the tetramer assay with functionality, we also have co-stained tetramer positive cells with activation markers. Finally, we verified the assay using various activity-confirmed ICIs and controls. The assay uses characterized PBMCs stimulated with a pool of peptides with positive and negative treatments (e.g., PD-1 blockades and IgG4) and “test samples,” which are the candidates to be tested. Depending on the donor’s recall responses, a typical assay results in two to 6-fold increase of the antigen specific MHC tetramer positive population. Furthermore, after developing this recall antigen assay, we validated it by performing the assay in different laboratories, using different reagents lots, different instrumentations, and different operators. Recall antigen assay data indicated that not all donors respond to approved immune checkpoint blockades. Healthy donors stimulated with anti CD3 or cytokine cocktails varied showed a wide a wide variability of immune checkpoint molecules (ICM) cell surface levels. Further studies using characterized PBMCs in our recall antigen potency assay revealed that the expression of immune checkpoint molecules may contribute in unresponsiveness to a given ICI. In conclusion, we have developed an improved in vitro recall antigen potency assay for immunomodulatory biologics functional screening including ICIs and APCs. Furthermore, we observed that in our studies this potency assay divides hosts into responders and nonresponders. Interestingly, we also showed significant variation of persons
免疫检查点拮抗剂和共刺激途径(ACP)拮抗剂作为单一药物或联合使用的治疗方案的激增,需要更好的功能筛选分析。体外功能筛选试验需要非侵入性和高通量。召回抗原试验可以说是在抗原特异性控制系统中,以相对生理相关的方式直接评估潜在免疫检查点抑制剂(ICI)候选物的活性。召回抗原测定在过去经常用于证明ICI或ACP的效力,如出版物和提交给监管机构所示。我们进行了彻底的优化,并对召回抗原测定进行了修改,以实现更有效和高效的测定。我们的召回抗原试验已经在多个参数上进行了优化,包括基于共抑制/共刺激分子表达的供体选择。这有助于为合适的候选人选择合适的供体。我们优化了激励和读出组件,以提高信噪比。例如,我们将体外抗原特异性CD8+ t细胞克隆扩增的持续时间从2周减少到5至7天。为了最大化直接读数,我们使用MHC四聚体引导分析,使用由刺激肽制成的MHC四聚体鸡尾酒。为了进一步增强四聚体检测的功能,我们还用激活标记物对四聚体阳性细胞进行了共染色。最后,我们使用各种活性确认的ICIs和对照验证了该分析。该检测方法使用经过阳性和阴性处理(例如,PD-1阻断剂和IgG4)的肽池和“测试样品”刺激的特征pbmc,这些样品是待测试的候选物。根据供体的回忆反应,典型的测定结果抗原特异性MHC四聚体阳性人群增加2至6倍。此外,在开发了这种召回抗原测定法之后,我们通过在不同的实验室、使用不同的试剂批次、不同的仪器和不同的操作员进行测定来验证它。回忆抗原测定数据表明,并非所有供体对批准的免疫检查点阻断反应。受抗CD3或细胞因子鸡尾酒刺激的健康供体表现出免疫检查点分子(ICM)细胞表面水平的广泛变异性。在我们的回忆抗原效价测定中,使用表征pbmc的进一步研究表明,免疫检查点分子的表达可能导致对给定ICI的无反应。总之,我们开发了一种改进的体外召回抗原效价法,用于免疫调节生物制剂的功能筛选,包括ICIs和APCs。此外,我们观察到,在我们的研究中,这种效价测定将宿主分为应答者和无应答者。有趣的是,我们还发现人的PBMC对ICIs的反应存在显著差异,这可能与他们固有的免疫检查点分子表达水平有关。我们打算通过使用注释良好的临床样本的进一步研究来证实这一发现。引文格式:Marc Delcommenne, Eden Kleiman, Wushouer Ouerkaxi, Pirouz Daftarian。一种基于召回抗原的免疫调节生物制剂效价测定,可以区分应答者和无应答者[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 B074。
{"title":"Abstract B074: A recall antigen-based potency assay for immunomodulatory biologics that could discriminate responders from nonresponders","authors":"M. Delcommenne, E. Kleiman, Wushouer Ouerkaxi, P. Daftarian","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B074","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B074","url":null,"abstract":"The surge of therapeutic regimens involving immune checkpoint antagonists and co-stimulatory pathways (ACP) antagonists used as single agents or in combination necessitates better functional screening assays. In vitro functional screening assays need to be non-invasive and high throughput. The recall antigen assay is arguably the one that, in an antigen specific controlled system, directly assesses the activity of potential immune checkpoint inhibitors (ICI) candidates, in a relatively physiologically relevant manner. The recall antigen assay has been often used in the past to prove the potency of ICI or ACP as indicated in publications and submissions to regulatory bodies. We have conducted a thorough optimization combined with modifications to the recall antigen assay to enable the need for a more effective and efficient assay.Our recall antigen assay has been optimized across multiple parameters including donor selection based on expression of co-inhibitory/co-stimulatory molecules. This facilitates the selection of the right donor for the right candidate. We have optimized both stimulation and readout components to enhance signal to noise ratio. For example, we reduced the duration of in vitro antigen specific CD8+ T-cell clonotype expansion from 2 weeks to 5 to 7 days. To maximize the direct readout, we have used an MHC Tetramer guided analysis, using a cocktail of MHC tetramers made with stimulating peptides. To further empower the tetramer assay with functionality, we also have co-stained tetramer positive cells with activation markers. Finally, we verified the assay using various activity-confirmed ICIs and controls. The assay uses characterized PBMCs stimulated with a pool of peptides with positive and negative treatments (e.g., PD-1 blockades and IgG4) and “test samples,” which are the candidates to be tested. Depending on the donor’s recall responses, a typical assay results in two to 6-fold increase of the antigen specific MHC tetramer positive population. Furthermore, after developing this recall antigen assay, we validated it by performing the assay in different laboratories, using different reagents lots, different instrumentations, and different operators. Recall antigen assay data indicated that not all donors respond to approved immune checkpoint blockades. Healthy donors stimulated with anti CD3 or cytokine cocktails varied showed a wide a wide variability of immune checkpoint molecules (ICM) cell surface levels. Further studies using characterized PBMCs in our recall antigen potency assay revealed that the expression of immune checkpoint molecules may contribute in unresponsiveness to a given ICI. In conclusion, we have developed an improved in vitro recall antigen potency assay for immunomodulatory biologics functional screening including ICIs and APCs. Furthermore, we observed that in our studies this potency assay divides hosts into responders and nonresponders. Interestingly, we also showed significant variation of persons","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"102 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":"115893835","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
Mutational Analysis and Predicting Response to Immunotherapy
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