Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B096
S. Warren, T. Hood, P. Danaher, A. Cesano
Introduction: Numerous immune checkpoint inhibitors are being developed for the clinic, but identifying the population of patients most likely to respond remains a significant challenge. PD-(L)1 blocking antibodies have been approved for multiple indications, but even in those indications the majority of patients fail to respond to PD-(L)1 monotherapy. Consequently, diagnostic assays have been developed to identify patients with a higher likelihood of response. PD-L1 immunohistochemistry is the platform for multiple assays currently being used in the clinical as companion and complementary diagnostics for the PD-(L)1 checkpoint inhibitors, but those assays have limited sensitivity and selectivity and have inherent risk of subjective interpretation bias. Tumor mutation burden is in development as a proxy readout for a tumor’s potential to prime immune responses, but it does not measure the actual presence of an immune response, and it is not able to inform treatment decisions if there is the option of more than one immunomodulatory intervention. Gene expression assays have the advantage of being a sensitive, selective, and quantitative assay which can directly measure immune biology, and may overcome many of the limitations of the other assay platforms. The Tumor Inflammation Signature (TIS) has been developed on the NanoString® platform as an 18-gene signature of a suppressed immune response within the tumor and has been developed as a clinically validated assay which enriches for response to anti-PD-1 (Ayers, JCI 2017). We have recently evaluated the distribution of TIS in The Cancer Genome Atlas (TCGA) database to understand the prevalence and distribution of immune “hot” vs “cold” tumors by indication (Danaher, JITC 2018). We now extend that work to evaluate the expression of individual immune checkpoint molecules after segregating tumors by TIS to understand the distribution of immune checkpoints across indications and within the context of a preexisting immune response. Methods: We leverage biostatistical analysis of the RNA-seq data in the TCGA database to evaluate the expression of the TIS signature and individual immune checkpoints. Results: We observe that the expression of many immune checkpoint molecules is directly proportional to the degree of immune infiltrate within the tumor as measured by TIS. As such, there is a distribution of IO targets across indications, with inflamed tumors expressing greater median levels of immune checkpoints vs noninflamed tumors. Within individual indication, we also see a distribution of hot and cold tumors, and a corresponding distribution of checkpoint molecules, indicating that there may be some subpopulations of patients with the potential to respond to immune checkpoint blockade even in an indication that is nonresponsive in an unselected population. Furthermore, we also observe increased expression of particular immune checkpoints in subpopulations of certain tumors. For example, certain bladder
{"title":"Abstract B096: Dissecting the flames from the fire: Distribution of immune checkpoints in hot and cold tumors","authors":"S. Warren, T. Hood, P. Danaher, A. Cesano","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B096","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B096","url":null,"abstract":"Introduction: Numerous immune checkpoint inhibitors are being developed for the clinic, but identifying the population of patients most likely to respond remains a significant challenge. PD-(L)1 blocking antibodies have been approved for multiple indications, but even in those indications the majority of patients fail to respond to PD-(L)1 monotherapy. Consequently, diagnostic assays have been developed to identify patients with a higher likelihood of response. PD-L1 immunohistochemistry is the platform for multiple assays currently being used in the clinical as companion and complementary diagnostics for the PD-(L)1 checkpoint inhibitors, but those assays have limited sensitivity and selectivity and have inherent risk of subjective interpretation bias. Tumor mutation burden is in development as a proxy readout for a tumor’s potential to prime immune responses, but it does not measure the actual presence of an immune response, and it is not able to inform treatment decisions if there is the option of more than one immunomodulatory intervention. Gene expression assays have the advantage of being a sensitive, selective, and quantitative assay which can directly measure immune biology, and may overcome many of the limitations of the other assay platforms. The Tumor Inflammation Signature (TIS) has been developed on the NanoString® platform as an 18-gene signature of a suppressed immune response within the tumor and has been developed as a clinically validated assay which enriches for response to anti-PD-1 (Ayers, JCI 2017). We have recently evaluated the distribution of TIS in The Cancer Genome Atlas (TCGA) database to understand the prevalence and distribution of immune “hot” vs “cold” tumors by indication (Danaher, JITC 2018). We now extend that work to evaluate the expression of individual immune checkpoint molecules after segregating tumors by TIS to understand the distribution of immune checkpoints across indications and within the context of a preexisting immune response. Methods: We leverage biostatistical analysis of the RNA-seq data in the TCGA database to evaluate the expression of the TIS signature and individual immune checkpoints. Results: We observe that the expression of many immune checkpoint molecules is directly proportional to the degree of immune infiltrate within the tumor as measured by TIS. As such, there is a distribution of IO targets across indications, with inflamed tumors expressing greater median levels of immune checkpoints vs noninflamed tumors. Within individual indication, we also see a distribution of hot and cold tumors, and a corresponding distribution of checkpoint molecules, indicating that there may be some subpopulations of patients with the potential to respond to immune checkpoint blockade even in an indication that is nonresponsive in an unselected population. Furthermore, we also observe increased expression of particular immune checkpoints in subpopulations of certain tumors. For example, certain bladder ","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","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":"134138725","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B080
Julia Kodysh, Tim O’Donnell, A. Blázquez, J. Finnigan, N. Bhardwaj, A. Rubinsteyn
The OpenVax group has helped initiate two neoantigen vaccine clinical trials (NCT02721043, NCT02721043) at Mount Sinai based on a simple multiplicative ranking criterion which assigns equal weight to expression and predicted Class I MHC binding affinity of mutated peptides (1). This poster seeks to better ground our ranking method for selecting the contents of neoantigen vaccines in several sources of immunological data. We built a better model of MHC-I presentation on the cell surface by relating RNA expression and MHC affinity to pMHC ligands identified with mass spectrometry (2). Secondly, we trained a model of overall T-cell immunogenicity whose primary input is the predicted pMHC presentation score of any peptide-MHC combination, alongside other features such as similarity to the self proteome. This model is trained on T-cell response data deposited in the Immune Epitope Database (3). Lastly, we assembled a small dataset of peptide sequences used in neoantigen vaccine trials (1,4,5), which are labeled by whether they achieved a CD8+ or CD4+ T-cell response. This dataset allows us to explore several hypotheses about the relationship between immunogenic response and sequence similarity to both the self proteome and pathogenic proteomes. References: 1. Rubinsteyn A, Kodysh J, …, Hammerbacher J. Computational pipeline for the PGV-001 Neoantigen Vaccine Trial. Frontiers in Immunology 2018. 2. Abelin JG, Keskin DB,..., Wu CJ. Mass spectrometry profiling of HLA-associated peptidomes in mono-allelic cells enables more accurate epitope prediction. Immunity 2017. 3. Vita R, Overton JA, …, Peters B. The immune epitope database (IEDB) 3.0. Nucleic Acids Res 2014. [4. Sahin U, Derhovanessian E, …, Tureci O. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017. 5. Ott P, Hu Z, …, Wu CJ. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017. Citation Format: Julia Kodysh, Tim O9Donnell, Ana B. Blazquez, John Finnigan, Nina Bhardwaj, Alex Rubinsteyn. Improved neoantigen vaccine selection by combining prediction of pMHC presentation and T-cell epitopes [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 B080.
OpenVax小组在西奈山帮助启动了两项新抗原疫苗临床试验(NCT02721043, NCT02721043),该试验基于一个简单的乘法排序标准,该标准赋予表达同等权重,并预测突变肽的I类MHC结合亲和力(1)。该海报旨在更好地为我们的排序方法奠定基础,以便在多个免疫学数据来源中选择新抗原疫苗的内容。通过将RNA表达和MHC亲和力与质谱鉴定的pMHC配体联系起来,我们建立了一个更好的MHC- i在细胞表面呈递的模型(2)。其次,我们训练了一个整体t细胞免疫原性模型,其主要输入是任何肽-MHC组合的预测pMHC呈递评分,以及其他特征,如与自身蛋白质组的相似性。该模型是根据储存在免疫表位数据库(Immune Epitope Database)中的t细胞应答数据进行训练的(3)。最后,我们收集了一个用于新抗原疫苗试验(1,4,5)的肽序列的小数据集,这些序列通过它们是否达到CD8+或CD4+ t细胞应答来标记。该数据集允许我们探索免疫原性反应与自身蛋白质组和致病性蛋白质组序列相似性之间关系的几个假设。引用:1。李建军,李建军,李建军,等。ppv001新抗原疫苗的临床研究进展。免疫学前沿2018。2. 艾柏林JG,凯斯金DB,…吴俊杰。质谱分析hla相关肽在单等位细胞使更准确的表位预测。2017年的免疫力。3.李建军,李建军,李建军,等。免疫表位数据库(IEDB) 3.0。核酸学报,2014。(4。李建军,张建军,张建军,等。RNA突变体疫苗可激活肿瘤多特异性免疫。2017年自然。5. 赵鹏,胡忠,吴俊杰。黑色素瘤患者的免疫原性个人新抗原疫苗。2017年自然。引文格式:Julia kodhh, Tim O9Donnell, Ana B. Blazquez, John Finnigan, Nina Bhardwaj, Alex rubinstein。结合pMHC呈递和t细胞表位预测改进新抗原疫苗选择[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 B080。
{"title":"Abstract B080: Improved neoantigen vaccine selection by combining prediction of pMHC presentation and T-cell epitopes","authors":"Julia Kodysh, Tim O’Donnell, A. Blázquez, J. Finnigan, N. Bhardwaj, A. Rubinsteyn","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B080","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B080","url":null,"abstract":"The OpenVax group has helped initiate two neoantigen vaccine clinical trials (NCT02721043, NCT02721043) at Mount Sinai based on a simple multiplicative ranking criterion which assigns equal weight to expression and predicted Class I MHC binding affinity of mutated peptides (1). This poster seeks to better ground our ranking method for selecting the contents of neoantigen vaccines in several sources of immunological data. We built a better model of MHC-I presentation on the cell surface by relating RNA expression and MHC affinity to pMHC ligands identified with mass spectrometry (2). Secondly, we trained a model of overall T-cell immunogenicity whose primary input is the predicted pMHC presentation score of any peptide-MHC combination, alongside other features such as similarity to the self proteome. This model is trained on T-cell response data deposited in the Immune Epitope Database (3). Lastly, we assembled a small dataset of peptide sequences used in neoantigen vaccine trials (1,4,5), which are labeled by whether they achieved a CD8+ or CD4+ T-cell response. This dataset allows us to explore several hypotheses about the relationship between immunogenic response and sequence similarity to both the self proteome and pathogenic proteomes. References: 1. Rubinsteyn A, Kodysh J, …, Hammerbacher J. Computational pipeline for the PGV-001 Neoantigen Vaccine Trial. Frontiers in Immunology 2018. 2. Abelin JG, Keskin DB,..., Wu CJ. Mass spectrometry profiling of HLA-associated peptidomes in mono-allelic cells enables more accurate epitope prediction. Immunity 2017. 3. Vita R, Overton JA, …, Peters B. The immune epitope database (IEDB) 3.0. Nucleic Acids Res 2014. [4. Sahin U, Derhovanessian E, …, Tureci O. Personalized RNA mutanome vaccines mobilize poly-specific therapeutic immunity against cancer. Nature 2017. 5. Ott P, Hu Z, …, Wu CJ. An immunogenic personal neoantigen vaccine for patients with melanoma. Nature 2017. Citation Format: Julia Kodysh, Tim O9Donnell, Ana B. Blazquez, John Finnigan, Nina Bhardwaj, Alex Rubinsteyn. Improved neoantigen vaccine selection by combining prediction of pMHC presentation and T-cell epitopes [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 B080.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","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":"130689032","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-IA31
M. Luksza, Alexander Solovyov, N. Vabret, V. Balachandran, N. Riaz, V. Makarov, M. Hellmann, A. Snyder, S. Funt, R. Remark, M. Merad, S. Gnjatic, D. Bajorin, J. Rosenberg, S. Leach, A. Levine, T. Chan, N. Bhardwaj, J. Wolchock, B. Greenbaum
Molecules generated by mutational and epigenetic processes in tumors have been associated with recognition of tumors by the innate and adaptive immune system. For example, neoantigens have been implicated in response to checkpoint blockade therapies. Likewise, the display of pathogen-associated patterns by nucleic acids unsilenced by epigenetic alterations have been implicated in activation of the innate immune system. Here we determine molecular features which place a tumor at a selective advantage or disadvantage, and how these selective pressures depend on the tumor’s environment. We have proposed general frameworks to address these questions. In the case of tumor neoantigens, we present a fitness model of candidate immunogenic neoantigens distributed across a tumor’s subclonal structure in a given microenvironment. We show how our approach can be used to characterize response to checkpoint-blockade therapies and apply it to the general problem of immune-driven tumor evolution in a unique cohort of long-term survivors of pancreatic cancer. In the case of immunostimulatory RNA, we proposed a method of calculating entropic forces for determining the likelihood of tumoral RNA being recognized as pathogen-associated and characterizing classes of pathogen mimicry. Citation Format: Marta Luksza, Alexander Solovyov, Nicolas Vabret, Vinod Balachandran, Nadeem Riaz, Vladimir Makarov, Matthew D. Hellmann , Alexandra Snyder, Samuel Funt, Romain Remark, Miriam Merad, Sacha Gnjatic, Dean F. Bajorin, Jonathan Rosenberg, Steven Leach, Arnold J. Levine, Timothy A. Chan, Nina Bhardwaj, Jedd Wolchock, Benjamin D. Greenbaum. Measuring the emergence of non-self in tumors [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 IA31.
肿瘤中由突变和表观遗传过程产生的分子与先天和适应性免疫系统对肿瘤的识别有关。例如,新抗原与检查点阻断疗法的反应有关。同样,通过表观遗传改变而沉默的核酸所显示的病原体相关模式与先天免疫系统的激活有关。在这里,我们确定了使肿瘤处于选择性优势或劣势的分子特征,以及这些选择压力如何取决于肿瘤的环境。我们提出了解决这些问题的一般框架。在肿瘤新抗原的情况下,我们提出了在给定微环境中分布在肿瘤亚克隆结构中的候选免疫原性新抗原的适应度模型。我们展示了我们的方法如何用于描述对检查点阻断疗法的反应,并将其应用于胰腺癌长期幸存者的独特队列中免疫驱动肿瘤进化的一般问题。在免疫刺激RNA的情况下,我们提出了一种计算熵力的方法,以确定肿瘤RNA被识别为病原体相关的可能性,并表征病原体模仿的类别。引文格式:Marta Luksza、Alexander Solovyov、Nicolas Vabret、Vinod Balachandran、Nadeem Riaz、Vladimir Makarov、Matthew D. Hellmann、Alexandra Snyder、Samuel Funt、Romain Remark、Miriam Merad、Sacha Gnjatic、Dean F. Bajorin、Jonathan Rosenberg、Steven Leach、Arnold J. Levine、Timothy A. Chan、Nina Bhardwaj、Jedd Wolchock、Benjamin D. Greenbaum。测量肿瘤中非自我的出现[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1 - 31。
{"title":"Abstract IA31: Measuring the emergence of non-self in tumors","authors":"M. Luksza, Alexander Solovyov, N. Vabret, V. Balachandran, N. Riaz, V. Makarov, M. Hellmann, A. Snyder, S. Funt, R. Remark, M. Merad, S. Gnjatic, D. Bajorin, J. Rosenberg, S. Leach, A. Levine, T. Chan, N. Bhardwaj, J. Wolchock, B. Greenbaum","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA31","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA31","url":null,"abstract":"Molecules generated by mutational and epigenetic processes in tumors have been associated with recognition of tumors by the innate and adaptive immune system. For example, neoantigens have been implicated in response to checkpoint blockade therapies. Likewise, the display of pathogen-associated patterns by nucleic acids unsilenced by epigenetic alterations have been implicated in activation of the innate immune system. Here we determine molecular features which place a tumor at a selective advantage or disadvantage, and how these selective pressures depend on the tumor’s environment. We have proposed general frameworks to address these questions. In the case of tumor neoantigens, we present a fitness model of candidate immunogenic neoantigens distributed across a tumor’s subclonal structure in a given microenvironment. We show how our approach can be used to characterize response to checkpoint-blockade therapies and apply it to the general problem of immune-driven tumor evolution in a unique cohort of long-term survivors of pancreatic cancer. In the case of immunostimulatory RNA, we proposed a method of calculating entropic forces for determining the likelihood of tumoral RNA being recognized as pathogen-associated and characterizing classes of pathogen mimicry. Citation Format: Marta Luksza, Alexander Solovyov, Nicolas Vabret, Vinod Balachandran, Nadeem Riaz, Vladimir Makarov, Matthew D. Hellmann , Alexandra Snyder, Samuel Funt, Romain Remark, Miriam Merad, Sacha Gnjatic, Dean F. Bajorin, Jonathan Rosenberg, Steven Leach, Arnold J. Levine, Timothy A. Chan, Nina Bhardwaj, Jedd Wolchock, Benjamin D. Greenbaum. Measuring the emergence of non-self in tumors [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 IA31.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"52 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":"129615267","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B095
Guangchuan Wang, Ryan D. Chow, Z. Bai, Lupeng Ye, Sidi Chen
Immune checkpoint blockade has achieved tremendous clinical success across many tumor types, but fails to induce clinical responses in many patients. The mechanisms underlying checkpoint resistance remain poorly characterized. Recent studies have applied next generation sequencing techniques to catalog the mutational burden of patient tumors, which provides a wealth of data to determine common mutations. To map the genetic features of response to checkpoint blockade immunotherapy as well as correlating the clinical efficacy with certain mutations, we developed a novel direct in vivo CRISPR screening approach for high-throughput profiling of functional cancer drivers in an autochthonous manner by injecting AAVs carrying an sgRNA library targeting the top 50 TCGA pan-cancer recurrently mutated tumor suppressor genes (mTSG) into the immunocompetent Cas9 transgenic mice. All mice that received the AAV-mTSG library developed liver cancer and died within four months. We then utilized MIP sequencing of sgRNA target sites to chart the mutational landscape of these tumors, revealing the functional consequence of multiple variants in driving liver tumorigenesis as well as identifying specific gene pairs that were co-occurring across mice. Using this approach, we also mapped the mutation landscape changes under the pressures of immune checkpoint inhibitors, anti-PD1 or anti-CTLA4. We monitored liver tumor growth in AAV-mTSG injected LSL-Cas9;LSL-Fluc mice by using intravital bioluminescent imaging system (IVIS) in combination with dissection check before drug administration. Using IVIS data, we grouped them into 3 size-matched cohorts to receive anti-PD1 or anti-CTLA4 treatments or PBS control. According to the survival data, the mice with mTSG-induced liver tumor benefit from anti-PD1 or anti-CLTA4 treatment. By comparing the mutation frequencies of liver tumors in the mice receiving either checkpoint inhibitors or PBS treatment, we mapped the mutation landscape changes associated with anti-PD1 or anti-CTLA4 treatment. We are performing validation studies on top targets such as Arid1a, Stk11, and B2M. Using this approach, we systematically mapped the correlation of these top 50 driver mutations with cancer immune evasion and immunotherapy responsiveness, providing a valuable reference for patient stratification when considering immunotherapy as well as novel targets for synergistic interventions. Citation Format: Guangchuan Wang, Ryan Chow, Zhigang Bai, Lupeng Ye, Sidi Chen. Mapping the genetic features of immune checkpoint responsiveness using AAV-CRISPR mediated in vivo screen [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 B095.
{"title":"Abstract B095: Mapping the genetic features of immune checkpoint responsiveness using AAV-CRISPR mediated in vivo screen","authors":"Guangchuan Wang, Ryan D. Chow, Z. Bai, Lupeng Ye, Sidi Chen","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B095","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B095","url":null,"abstract":"Immune checkpoint blockade has achieved tremendous clinical success across many tumor types, but fails to induce clinical responses in many patients. The mechanisms underlying checkpoint resistance remain poorly characterized. Recent studies have applied next generation sequencing techniques to catalog the mutational burden of patient tumors, which provides a wealth of data to determine common mutations. To map the genetic features of response to checkpoint blockade immunotherapy as well as correlating the clinical efficacy with certain mutations, we developed a novel direct in vivo CRISPR screening approach for high-throughput profiling of functional cancer drivers in an autochthonous manner by injecting AAVs carrying an sgRNA library targeting the top 50 TCGA pan-cancer recurrently mutated tumor suppressor genes (mTSG) into the immunocompetent Cas9 transgenic mice. All mice that received the AAV-mTSG library developed liver cancer and died within four months. We then utilized MIP sequencing of sgRNA target sites to chart the mutational landscape of these tumors, revealing the functional consequence of multiple variants in driving liver tumorigenesis as well as identifying specific gene pairs that were co-occurring across mice. Using this approach, we also mapped the mutation landscape changes under the pressures of immune checkpoint inhibitors, anti-PD1 or anti-CTLA4. We monitored liver tumor growth in AAV-mTSG injected LSL-Cas9;LSL-Fluc mice by using intravital bioluminescent imaging system (IVIS) in combination with dissection check before drug administration. Using IVIS data, we grouped them into 3 size-matched cohorts to receive anti-PD1 or anti-CTLA4 treatments or PBS control. According to the survival data, the mice with mTSG-induced liver tumor benefit from anti-PD1 or anti-CLTA4 treatment. By comparing the mutation frequencies of liver tumors in the mice receiving either checkpoint inhibitors or PBS treatment, we mapped the mutation landscape changes associated with anti-PD1 or anti-CTLA4 treatment. We are performing validation studies on top targets such as Arid1a, Stk11, and B2M. Using this approach, we systematically mapped the correlation of these top 50 driver mutations with cancer immune evasion and immunotherapy responsiveness, providing a valuable reference for patient stratification when considering immunotherapy as well as novel targets for synergistic interventions. Citation Format: Guangchuan Wang, Ryan Chow, Zhigang Bai, Lupeng Ye, Sidi Chen. Mapping the genetic features of immune checkpoint responsiveness using AAV-CRISPR mediated in vivo screen [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 B095.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","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":"126182720","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B084
C. Ock, Changhee Park, Kyeonghun Jeong, Sohee Jung, J. Bae, Kwangsoo Kim
The most reliable predictive biomarker of cancer immunotherapy is gene expression profile (GEP) of tumor microenvironment. GEPs such as local immune cytolytic activity, interferon-gamma signature, and immune signature score have been reported to represent anti-tumor immune signature. Previously, we reported that immune signature score was positively correlated with tumor mutational burden, but negatively correlated with chromosomal instability (CIN) score, since tumors with high CIN score had significantly low neoantigen burden. However, methylation signature or burden of tumor would also affect antitumor immunogenicity, there has been no analysis reported so far. In the current study, we investigated if methylation landscape of tumor would be associated with GEPs of anti-tumor immune signature using The Cancer Genome Atlas (TCGA) pan-cancer database. In TCGA, 8269 pan-cancer samples had both RNA sequencing data and methylation data using Infinium HumanMethylation450K BeadChip, which were included in the main analysis. Although tumors with high mutational burden (Mu-type) and high CIN burden (C-type) were exclusively classified with negative correlation, methylation burden was not correlated with mutational burden or CIN burden in any pattern. Interestingly, antitumor immune signature measured by local immune cytolytic activity (CytAct) was clearly decreased with high methylational burden, as seen in high CIN burden. Hypermethylation of promoter of genes related to tumor antigen recognition by T-cell such as HLA family, B2M, CD74, and CD274 (PD-L1) were negatively associated with CytAct in pan-cancer analysis. In conclusion, methylation signature of tumor is also associated with antitumor immunogenicity with a negative correlation in general. Further study of whether specific methylation pattern would be associated with anti-PD-1/PD-L1 inhibitors in clinical study would be warranted. Citation Format: Chan-Young Ock, Changhee Park, Kyeonghun Jeong, Sohee Jung, Jeong Mo Bae, Kwangsoo Kim. Methylation landscape of tumors associated with antitumor immune signature [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 B084.
肿瘤微环境基因表达谱(GEP)是预测肿瘤免疫治疗最可靠的生物标志物。gep如局部免疫细胞溶解活性、干扰素γ信号和免疫信号评分已被报道为抗肿瘤免疫信号。之前,我们报道了免疫标记评分与肿瘤突变负荷呈正相关,但与染色体不稳定性(CIN)评分负相关,因为CIN评分高的肿瘤新抗原负荷明显低。然而,甲基化特征或肿瘤负荷也会影响抗肿瘤免疫原性,目前尚无相关分析报道。在本研究中,我们利用癌症基因组图谱(TCGA)泛癌症数据库研究肿瘤的甲基化景观是否与抗肿瘤免疫特征的GEPs相关。在TCGA中,使用Infinium HumanMethylation450K BeadChip的8269份泛癌样本同时具有RNA测序数据和甲基化数据,并被纳入主分析。虽然高突变负担(mu型)和高CIN负担(c型)的肿瘤被单独归类为负相关,但甲基化负担与突变负担或CIN负担没有任何模式的相关性。有趣的是,通过局部免疫细胞溶解活性(CytAct)测量的抗肿瘤免疫特征在高甲基化负荷下明显降低,正如在高CIN负荷中所见。HLA家族、B2M、CD74、CD274 (PD-L1)等t细胞肿瘤抗原识别相关基因启动子的高甲基化与泛癌分析中发现CytAct呈负相关。综上所述,肿瘤甲基化特征也与抗肿瘤免疫原性相关,总体上呈负相关。在临床研究中,特异性甲基化模式是否与抗pd -1/PD-L1抑制剂相关有待进一步研究。引文格式:occhanyoung, Changhee Park, Kyeonghun Jeong Sohee Jung, Jeong Mo Bae, Kwangsoo Kim。与抗肿瘤免疫特征相关的肿瘤甲基化景观[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 B084。
{"title":"Abstract B084: Methylation landscape of tumors associated with antitumor immune signature","authors":"C. Ock, Changhee Park, Kyeonghun Jeong, Sohee Jung, J. Bae, Kwangsoo Kim","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B084","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B084","url":null,"abstract":"The most reliable predictive biomarker of cancer immunotherapy is gene expression profile (GEP) of tumor microenvironment. GEPs such as local immune cytolytic activity, interferon-gamma signature, and immune signature score have been reported to represent anti-tumor immune signature. Previously, we reported that immune signature score was positively correlated with tumor mutational burden, but negatively correlated with chromosomal instability (CIN) score, since tumors with high CIN score had significantly low neoantigen burden. However, methylation signature or burden of tumor would also affect antitumor immunogenicity, there has been no analysis reported so far. In the current study, we investigated if methylation landscape of tumor would be associated with GEPs of anti-tumor immune signature using The Cancer Genome Atlas (TCGA) pan-cancer database. In TCGA, 8269 pan-cancer samples had both RNA sequencing data and methylation data using Infinium HumanMethylation450K BeadChip, which were included in the main analysis. Although tumors with high mutational burden (Mu-type) and high CIN burden (C-type) were exclusively classified with negative correlation, methylation burden was not correlated with mutational burden or CIN burden in any pattern. Interestingly, antitumor immune signature measured by local immune cytolytic activity (CytAct) was clearly decreased with high methylational burden, as seen in high CIN burden. Hypermethylation of promoter of genes related to tumor antigen recognition by T-cell such as HLA family, B2M, CD74, and CD274 (PD-L1) were negatively associated with CytAct in pan-cancer analysis. In conclusion, methylation signature of tumor is also associated with antitumor immunogenicity with a negative correlation in general. Further study of whether specific methylation pattern would be associated with anti-PD-1/PD-L1 inhibitors in clinical study would be warranted. Citation Format: Chan-Young Ock, Changhee Park, Kyeonghun Jeong, Sohee Jung, Jeong Mo Bae, Kwangsoo Kim. Methylation landscape of tumors associated with antitumor immune signature [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 B084.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"8 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":"115035367","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B079
Julia Kodysh, J. Finnigan, A. Rubinsteyn
Neoantigen vaccination is an emerging modality of cancer immunotherapy with many ongoing trials. One central question of neoantigen vaccination is the method for selecting which mutated tumor-specific antigens to include in a patient’s vaccine. Many in-silico pipelines for neoantigen selection have been published in the past few years, but no comprehensive evaluation has compared them directly on the same tumor/normal sequencing data. We evaluate several publicly available commonly used neoantigen pipelines (pVACtools [1], MuPeXI [2], TIminer [3], OpenVax [4]) on both murine and human cancer samples. Our evaluation highlights the salient differences between these pipelines and shows the divergent results they achieve. References: 1. Kiwala S, Hundal J, …, Griffith M. pVACtools: Computational selection and visualization of neoantigens for personalized cancer vaccine design. Cancer Genetics 2018. 2. Bjerregaard A-M, Nielsen M, ..., Eklund AC. MuPeXI: Prediction of neo-epitopes from tumor sequencing data. Cancer Immunology Immunotherapy 2018. 3. Tappeiner E, Finotello F, ..., Trajanoski Z. TIminer: NGS data mining pipeline for cancer immunology and immunotherapy. Bioinformatics 2017. 4. Rubinsteyn A, Kodysh J, …, Hammerbacher J. Computational pipeline for the PGV-001 Neoantigen Vaccine Trial. Frontiers in Immunology 2018. Citation Format: Julia Kodysh, John P. Finnigan, Alex Rubinsteyn. Evaluation of tools for predicting mutated tumor antigens from exome and RNA sequencing [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 B079.
新抗原疫苗接种是一种新兴的癌症免疫治疗方式,有许多正在进行的试验。新抗原疫苗接种的一个中心问题是选择将哪些突变的肿瘤特异性抗原包括在患者疫苗中的方法。在过去的几年中,已经发表了许多用于新抗原选择的计算机管道,但没有对它们在相同的肿瘤/正常测序数据上进行直接比较的综合评估。我们评估了几种公开可用的常用新抗原管道(pVACtools [1], MuPeXI [2], TIminer [3], OpenVax[4])对小鼠和人类癌症样本的影响。我们的评估强调了这些管道之间的显著差异,并显示了它们实现的不同结果。引用:1。张建军,张建军,张建军,等。肿瘤疫苗抗原筛选方法的研究进展。癌症遗传学2018。2. bjerregard A-M, Nielsen M,…MuPeXI:基于肿瘤测序数据的新表位预测。癌症免疫学-免疫疗法2018。3.Tappeiner E, Finotello F,…(1)基于NGS数据挖掘的肿瘤免疫与免疫治疗研究。2017年生物信息学。4. 李建军,李建军,李建军,等。ppv001新抗原疫苗的临床研究进展。免疫学前沿2018。引用格式:Julia kodhh, John P. Finnigan, Alex rubinstein。外显子组和RNA测序预测肿瘤抗原突变的工具评价[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 B079。
{"title":"Abstract B079: Evaluation of tools for predicting mutated tumor antigens from exome and RNA sequencing","authors":"Julia Kodysh, J. Finnigan, A. Rubinsteyn","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B079","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B079","url":null,"abstract":"Neoantigen vaccination is an emerging modality of cancer immunotherapy with many ongoing trials. One central question of neoantigen vaccination is the method for selecting which mutated tumor-specific antigens to include in a patient’s vaccine. Many in-silico pipelines for neoantigen selection have been published in the past few years, but no comprehensive evaluation has compared them directly on the same tumor/normal sequencing data. We evaluate several publicly available commonly used neoantigen pipelines (pVACtools [1], MuPeXI [2], TIminer [3], OpenVax [4]) on both murine and human cancer samples. Our evaluation highlights the salient differences between these pipelines and shows the divergent results they achieve. References: 1. Kiwala S, Hundal J, …, Griffith M. pVACtools: Computational selection and visualization of neoantigens for personalized cancer vaccine design. Cancer Genetics 2018. 2. Bjerregaard A-M, Nielsen M, ..., Eklund AC. MuPeXI: Prediction of neo-epitopes from tumor sequencing data. Cancer Immunology Immunotherapy 2018. 3. Tappeiner E, Finotello F, ..., Trajanoski Z. TIminer: NGS data mining pipeline for cancer immunology and immunotherapy. Bioinformatics 2017. 4. Rubinsteyn A, Kodysh J, …, Hammerbacher J. Computational pipeline for the PGV-001 Neoantigen Vaccine Trial. Frontiers in Immunology 2018. Citation Format: Julia Kodysh, John P. Finnigan, Alex Rubinsteyn. Evaluation of tools for predicting mutated tumor antigens from exome and RNA sequencing [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 B079.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"13 6","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"133077137","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR12
S. Schoenberger, Aaron M. Miller, Luise Sternberg, Leslie Montero Cuencac, Milad Bahmanof, Zeynep Koasaloglu-Yalcin, Manasa Lanka, A. Premlal, P. Vijayanand, J. Greenbaum, Allesandro Seatte, Ezra E. W. Cohen, Bjoern Peters
Accurate identification of tumor-specific neoantigens (NeoAg) is essential for the development of effective personalized cancer vaccines and cellular immunotherapies. The success rates for purely computational approaches which rely on predicted HLA-binding have been disappointing, as these generally ignore 85-90% of total mutations and find less than 5% of those selected can be confirmed as T-cell targets. We have developed a novel NeoAg identification platform in which WES and RNAseq metadata is used to nominate mutations for subsequent functional T-cell analysis using autologous PBMC and/or TIL. Applying this platform to tumors of low mutational burden including PDAC, HNSCC, and MSS-CRC, we report that an average of 35% of expressed mutations selected for functional testing can be verified as neoantigens, and that a significant number of these would be missed by HLA-binding algorithms. Responses comprise both type I and type 2 CD4+ and CD8+ effector T-cells recognizing both “passenger” mutations and known activating mutations in driver oncogenes such as KRAS, PIK3CA, and NRAS. Additionally, we have established a single-cell platform for isolation of T-cell receptors (TCR) against these shared recurrent mutations, and have opened a phase 1b clinical trial to evaluate the efficacy of personalized NeoAg vaccination in solid tumors. Citation Format: Stephen Phillip Schoenberger, Aaron M. Miller, Luise A. Sternberg, Leslie Montero Cuencac, Milad Bahmanof, Zeynep Koasaloglu-Yalcin, Manasa Lanka, Ashmitaa Premlal, Pandurangan Vijayanand, Jason Greenbaum, Allesandro Seatte, Ezra E.W. Cohen, Bjoern Peters. Functional identification and therapeutic targeting of tumor neoantigens [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 PR12.
准确识别肿瘤特异性新抗原(NeoAg)对于开发有效的个性化癌症疫苗和细胞免疫疗法至关重要。依靠预测hla结合的纯计算方法的成功率令人失望,因为这些方法通常忽略了总突变的85-90%,并且发现只有不到5%的被选中的突变可以被确认为t细胞靶标。我们开发了一种新的NeoAg鉴定平台,其中使用WES和RNAseq元数据来指定突变,以便随后使用自体PBMC和/或TIL进行功能性t细胞分析。将该平台应用于低突变负担的肿瘤,包括PDAC、HNSCC和MSS-CRC,我们报告了平均35%的用于功能测试的表达突变可以被验证为新抗原,并且hla结合算法会错过其中的很大一部分。应答包括I型和2型CD4+和CD8+效应t细胞,识别驱动癌基因(如KRAS、PIK3CA和NRAS)中的“乘客”突变和已知的激活突变。此外,我们已经建立了一个针对这些共同复发突变的t细胞受体(TCR)分离的单细胞平台,并已经开启了一项1b期临床试验,以评估个性化NeoAg疫苗接种在实体瘤中的疗效。引文格式:Stephen Phillip Schoenberger, Aaron M. Miller, louise A. Sternberg, Leslie Montero Cuencac, Milad Bahmanof, Zeynep Koasaloglu-Yalcin, Manasa Lanka, Ashmitaa Premlal, Pandurangan Vijayanand, Jason Greenbaum, Allesandro Seatte, Ezra E.W. Cohen, Bjoern Peters。肿瘤新抗原的功能鉴定及治疗靶向[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 PR12。
{"title":"Abstract PR12: Functional identification and therapeutic targeting of tumor neoantigens","authors":"S. Schoenberger, Aaron M. Miller, Luise Sternberg, Leslie Montero Cuencac, Milad Bahmanof, Zeynep Koasaloglu-Yalcin, Manasa Lanka, A. Premlal, P. Vijayanand, J. Greenbaum, Allesandro Seatte, Ezra E. W. Cohen, Bjoern Peters","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR12","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR12","url":null,"abstract":"Accurate identification of tumor-specific neoantigens (NeoAg) is essential for the development of effective personalized cancer vaccines and cellular immunotherapies. The success rates for purely computational approaches which rely on predicted HLA-binding have been disappointing, as these generally ignore 85-90% of total mutations and find less than 5% of those selected can be confirmed as T-cell targets. We have developed a novel NeoAg identification platform in which WES and RNAseq metadata is used to nominate mutations for subsequent functional T-cell analysis using autologous PBMC and/or TIL. Applying this platform to tumors of low mutational burden including PDAC, HNSCC, and MSS-CRC, we report that an average of 35% of expressed mutations selected for functional testing can be verified as neoantigens, and that a significant number of these would be missed by HLA-binding algorithms. Responses comprise both type I and type 2 CD4+ and CD8+ effector T-cells recognizing both “passenger” mutations and known activating mutations in driver oncogenes such as KRAS, PIK3CA, and NRAS. Additionally, we have established a single-cell platform for isolation of T-cell receptors (TCR) against these shared recurrent mutations, and have opened a phase 1b clinical trial to evaluate the efficacy of personalized NeoAg vaccination in solid tumors. Citation Format: Stephen Phillip Schoenberger, Aaron M. Miller, Luise A. Sternberg, Leslie Montero Cuencac, Milad Bahmanof, Zeynep Koasaloglu-Yalcin, Manasa Lanka, Ashmitaa Premlal, Pandurangan Vijayanand, Jason Greenbaum, Allesandro Seatte, Ezra E.W. Cohen, Bjoern Peters. Functional identification and therapeutic targeting of tumor neoantigens [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 PR12.","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"7 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":"133683027","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}
Pub Date : 2019-02-01DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B089
Guilhem Richard, R. Sweis, L. Moise, M. Ardito, W. Martin, Gad Berdugo, G. Steinberg, A. Groot
Precision cancer immunotherapy targeting mutations expressed by cancer cells has proven to effectively control the tumor of patients in multiple clinical trials (Sahin et al., Nature 2017; Ott et al., Nature 2017). However, the selection of immunogenic T-cell neo-epitopes remains challenging and many epitopes selected using traditional methodologies fail to induce effector T-cell responses. Poor performance may partially be due to inclusion of mutated epitopes cross-conserved with self-epitopes recognized by regulatory (Treg), anergic, or deleted T-cells. Vaccination with self-epitopes can lead to weak effector responses, active immune suppression, and toxicity due to immune-mediated adverse effects. In addition, most cancer vaccine studies focus on the selection of CD8 T-cell neo-epitopes due to an apparent lack of robust and accurate CD4 T-cell epitope prediction tools. We have developed Ancer, an integrated and streamlined neo-epitope selection pipeline, that accelerates the selection of both CD4 and CD8 T-cell neo-epitopes from next-generation sequencing (NGS) data. Ancer leverages EpiMatrix and JanusMatrix, predictive algorithms that have been extensively validated in prospective vaccine studies for infectious diseases (Moise et al., Hum Vaccines Immunother 2015; Wada et al., Sci Rep 2017). Distinctive features of Ancer are its ability to accurately predict Class II HLA ligands, or CD4 epitopes, with EpiMatrix, and to identify tolerated or Treg epitopes with JanusMatrix. In addition, screening candidate sequences with JanusMatrix enables to the removal of neo-epitopes that may trigger off-target events, which have in some cases abruptly halted the development of promising cancer therapies. Ancer was applied to NGS data derived from the BLCA bladder cancer cohort from The Cancer Genome Atlas (TCGA) database. On average, 55 out of 204 missense mutations in bladder cancer patients’ tumors met Ancer’s quality control standards, in an initial analysis carried out for a representative set of 11 patients. This subset of high-quality missense variants was then screened using Ancer settings defined by the unique HLA of each patient, to derive the best vaccine candidate sequences encompassing these mutations. A median number of 24 (interquartile range: 15-64) candidate sequences were generated for each patient under study. The time required to select sequences for all of the patients in this study was less than two days. This initial analysis of eleven BLCA bladder cancer cohort patients demonstrates the capacity of Ancer to define a sufficient number of candidate sequences for vaccinating bladder cancer patients in a precision immunotherapy setting. We also assessed Ancer’s ability to predict patient outcomes on a larger subset of 58 individuals. While the disease-free status of BLCA patients could not be explained by their tumor mutational burden (AUC = 0.55, p-value = 0.1328), nor by their load of missense mutations (AUC = 0.54, p-value = 0.1740),
针对癌细胞表达突变的精准癌症免疫治疗在多个临床试验中被证明可以有效控制患者的肿瘤(Sahin et al., Nature 2017;Ott et al., Nature 2017)。然而,免疫原性t细胞新表位的选择仍然具有挑战性,使用传统方法选择的许多表位无法诱导效应t细胞反应。表现不佳的部分原因可能是包含突变的表位与调节性(Treg)、无能或缺失的t细胞识别的自我表位交叉保守。使用自身抗原表位的疫苗接种可导致弱效应反应、主动免疫抑制和由于免疫介导的不良反应而产生的毒性。此外,由于缺乏可靠和准确的CD4 t细胞表位预测工具,大多数癌症疫苗研究都集中在CD8 t细胞新表位的选择上。我们开发了一种集成的流线型新表位选择管道Ancer,可从下一代测序(NGS)数据中加速CD4和CD8 t细胞新表位的选择。Ancer利用EpiMatrix和JanusMatrix,这两种预测算法已在传染病的前瞻性疫苗研究中得到广泛验证(Moise等人,Hum Vaccines Immunother 2015;Wada et al., Sci Rep 2017)。Ancer的独特之处在于它能够使用epimmatrix准确预测II类HLA配体或CD4表位,并使用JanusMatrix识别耐受或Treg表位。此外,使用JanusMatrix筛选候选序列可以去除可能引发脱靶事件的新表位,这些事件在某些情况下会突然停止有希望的癌症治疗的发展。Ancer应用于来自癌症基因组图谱(TCGA)数据库中BLCA膀胱癌队列的NGS数据。在对11名具有代表性的患者进行的初步分析中,平均而言,膀胱癌患者肿瘤中204个错义突变中有55个符合Ancer的质量控制标准。然后使用每个患者独特的HLA定义的Ancer设置筛选高质量错义变体子集,以获得包含这些突变的最佳候选疫苗序列。为研究中的每个患者生成的候选序列中位数为24(四分位数范围:15-64)。在这项研究中,为所有患者选择序列所需的时间少于两天。这项对11名BLCA膀胱癌队列患者的初步分析表明,Ancer有能力确定足够数量的候选序列,用于在精确免疫治疗环境中接种膀胱癌患者。我们还评估了Ancer在58个个体的更大子集中预测患者预后的能力。虽然BLCA患者的无病状态不能用其肿瘤突变负荷(AUC = 0.55, p值= 0.1328)来解释,也不能用其错义突变负荷(AUC = 0.54, p值= 0.1740)来解释,但根据Ancer的定义,与自身高度不同的新表位数量显著地将无病患者与复发或进展患者区分开来(AUC = 0.68, p值= 0.0214)。这些结果表明,使用Ancer来定义真正的新表位的数量可能代表一种新的生物标志物,用于更强大的抗肿瘤免疫反应和更高的无病生存可能性。我们对TCGA数据库中BLCA队列的分析显示了Ancer在临床环境中的价值。Ancer可用于鉴定高价值的候选序列,以纳入个性化治疗,同时从考虑中去除潜在耐受或耐受性的自我表位。我们的下一步将是研究癌症定义的新表位负荷是否可以作为整个BLCA队列中预后和治疗反应的生物标志物。引文格式:Guilhem Richard, Randy F. Sweis, Leonard Moise, Matthew Ardito, William A. Martin, Gad Berdugo, Gary D. Steinberg, Anne S. De Groot。精准癌症免疫治疗设计工具在膀胱癌中的应用:非自我样新表位作为预后生物标志物[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 B089。
{"title":"Abstract B089: Application of precision cancer immunotherapy design tools to bladder cancer: Non-self-like neoepitopes as a prognostic biomarker","authors":"Guilhem Richard, R. Sweis, L. Moise, M. Ardito, W. Martin, Gad Berdugo, G. Steinberg, A. Groot","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B089","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B089","url":null,"abstract":"Precision cancer immunotherapy targeting mutations expressed by cancer cells has proven to effectively control the tumor of patients in multiple clinical trials (Sahin et al., Nature 2017; Ott et al., Nature 2017). However, the selection of immunogenic T-cell neo-epitopes remains challenging and many epitopes selected using traditional methodologies fail to induce effector T-cell responses. Poor performance may partially be due to inclusion of mutated epitopes cross-conserved with self-epitopes recognized by regulatory (Treg), anergic, or deleted T-cells. Vaccination with self-epitopes can lead to weak effector responses, active immune suppression, and toxicity due to immune-mediated adverse effects. In addition, most cancer vaccine studies focus on the selection of CD8 T-cell neo-epitopes due to an apparent lack of robust and accurate CD4 T-cell epitope prediction tools. We have developed Ancer, an integrated and streamlined neo-epitope selection pipeline, that accelerates the selection of both CD4 and CD8 T-cell neo-epitopes from next-generation sequencing (NGS) data. Ancer leverages EpiMatrix and JanusMatrix, predictive algorithms that have been extensively validated in prospective vaccine studies for infectious diseases (Moise et al., Hum Vaccines Immunother 2015; Wada et al., Sci Rep 2017). Distinctive features of Ancer are its ability to accurately predict Class II HLA ligands, or CD4 epitopes, with EpiMatrix, and to identify tolerated or Treg epitopes with JanusMatrix. In addition, screening candidate sequences with JanusMatrix enables to the removal of neo-epitopes that may trigger off-target events, which have in some cases abruptly halted the development of promising cancer therapies. Ancer was applied to NGS data derived from the BLCA bladder cancer cohort from The Cancer Genome Atlas (TCGA) database. On average, 55 out of 204 missense mutations in bladder cancer patients’ tumors met Ancer’s quality control standards, in an initial analysis carried out for a representative set of 11 patients. This subset of high-quality missense variants was then screened using Ancer settings defined by the unique HLA of each patient, to derive the best vaccine candidate sequences encompassing these mutations. A median number of 24 (interquartile range: 15-64) candidate sequences were generated for each patient under study. The time required to select sequences for all of the patients in this study was less than two days. This initial analysis of eleven BLCA bladder cancer cohort patients demonstrates the capacity of Ancer to define a sufficient number of candidate sequences for vaccinating bladder cancer patients in a precision immunotherapy setting. We also assessed Ancer’s ability to predict patient outcomes on a larger subset of 58 individuals. While the disease-free status of BLCA patients could not be explained by their tumor mutational burden (AUC = 0.55, p-value = 0.1328), nor by their load of missense mutations (AUC = 0.54, p-value = 0.1740), ","PeriodicalId":433681,"journal":{"name":"Mutational Analysis and Predicting Response to Immunotherapy","volume":"53 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":"116458793","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}