Convergence of Technology and Cancer Immunotherapy最新文献

筛选
英文 中文
Abstract B039: Peptide-MHC-directed expansion of multifunctional antigen-responsive T-cells B039:多肽- mhc定向扩增多功能抗原反应t细胞
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B039
V. M. Rasmussen, A. Marquard, S. Jacobsen, S. Hadrup
{"title":"Abstract B039: Peptide-MHC-directed expansion of multifunctional antigen-responsive T-cells","authors":"V. M. Rasmussen, A. Marquard, S. Jacobsen, S. Hadrup","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B039","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B039","url":null,"abstract":"Functional properties and antigen specificity of expanded T-cells are crucial for the efficacy of adoptive cell transfer-based therapies in cancer. Most current strategies involve nonspecific expansion of bulk tumor-infiltrating lymphocytes, often providing growth preference to co-infiltrated virus specific T-cells and driving an exhausted phenotype of the expanded T-cell product.A potential way to resolve this challenge, is the use of artificial antigen-presenting scaffolds providing both an antigen specific stimulation through peptide-MHC interaction and additional the required co-stimulatory and growth signals through associated stimulatory molecules and cytokines. We have designed such antigen-presenting scaffolds; build on a dextran-polysaccharide, carrying both peptide-MHC and relevant stimulatory molecules. The artificial antigen-presenting scaffolds interacts specifically with T-cells based on recognition of the peptide-MHC molecule and effectively expand and functionally stimulate specific T-cells in a peptide-MHC-directed fashion, while leaving all other T-cell specificities untouched. Results from in vitro experiments have showed that antigen specific CD8 T-cells stimulated with these artificial antigen-presenting scaffolds express a less differentiated phenotype and low PD-1 expression, associated with high proliferation potential and enhanced antitumor effect in vivo. Furthermore, this expansion strategy provides a high frequency of multifunctional antigen specific CD8 T-cells expressing IFN-γ, TNF-α, and CD107a upon target recognition and provide improved cancer cell killing over IL2-driven expansion of tumor-infiltrating lymphocytes. Furthermore, the current strategy allows for simultaneous expansion of numerous different T-cell populations, required to generate T-cell products with broad recognition profiles based on the personal cancer-antigen and mutational profile. All of these characteristics are of significant importance for in vivo tumor cell killing following adoptive transfer of expanded T-cell products. Thus, the present strategy represents an optimized method for expansion of cancer-restricted T-cells for adoptive cell therapy. Citation Format: Vibeke Mindahl Rafa, Mona Bodenhofer, Amalie Kai Bentzen, Tripti Tamhane, Marco Donia, Inge Marie Stentoft Svane, Soren Nyboe Jakobsen, Christian Schmess, Sine Reker Hadrup. Peptide-MHC-directed expansion of multifunctional antigen-responsive T-cells [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B039.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"54 46 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":"125175999","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 B016: Automated ex vivo expansion of low numbers of tumor-reactive T-cells on the CliniMACS Prodigy® B016: CliniMACS Prodigy®上低数量肿瘤反应性t细胞的自动体外扩增
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B016
B. Heemskerk, C. Maeder, Elvira Criado-Moronati, L. Boettcher, A. Kaiser, M. Assenmacher, A. Dzionek
{"title":"Abstract B016: Automated ex vivo expansion of low numbers of tumor-reactive T-cells on the CliniMACS Prodigy®","authors":"B. Heemskerk, C. Maeder, Elvira Criado-Moronati, L. Boettcher, A. Kaiser, M. Assenmacher, A. Dzionek","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B016","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B016","url":null,"abstract":"Adoptive cell transfer (ACT) of tumor-infiltrating lymphocytes (TILs) has shown remarkable results in patients with metastatic melanoma. However, only a small fraction within the TIL population reacts against the tumor. Therefore, the pre-enrichment of tumor-specific T-cells and subsequent ex vivo expansion may improve the efficiency of ACT therapies. In addition, tumor-reactive T lymphocytes circulating in the blood (TRLs) have been found in low frequencies, which represents a challenge for their isolation, but also an advantage over TIL therapy since blood is a more reliable and accessible source than tumor excisions. Another impediment to the widespread application of ACT is the conventional rapid expansion protocol (REP) that constitutes a laborious and extensive process with frequent culture manipulations, and thus requires specialized personell and equipment. Our aim is to develop a fully automated large scale ex vivo T-cell isolation and expansion procedure in the CliniMACS Prodigy in order to simplify the manufacturing of tumor-reactive T-cells for ACT. The CliniMACS Prodigy instrument is a controlled system that integrates a series of cell processes, from magnetic cell separation and cell culture to final product formulation, under GMP conditions in a closed system. This process focuses on the optimization of the REP procedure on the CliniMACS Prodigy for TILs and TRLs. As a proof of concept, we used both cryopreserved outgrown TILs and magnetically isolated virus-specific T-cells from healthy donor leukapheresis via CD137 upregulation upon in vitro antigen stimulation. The first results show expansions ranging from 3,000-15,000 fold, both for TILs and CD137-expressing T-cells. From low cell numbers (2x10e5 - 1x10e6 cells) and after 14 days of cell culture in TexMACS medium and in the presence of high amounts of IL-2 and irradiated feeder cells, we were able to obtain around 3-4x10e9 cells. We also compared two different stimulation reagents, anti-CD3 antibody (OKT3) and TransAct (a soluble polymeric nanomatrix conjugated to humanized CD3 and CD28 agonist), which resulted in comparable expansion rates. Furthermore, small-scale experiments showed no differences between TexMACS and the conventional TIL culture medium (50% RPMI/50% AIM-V medium). The phenotype and reactivity of the expanded T-cells were also assessed by flow cytometry. Currently, higher starting cell numbers up to 1x10e7 cells are being assessed and first results are promising when shaking and media exchange are commenced earlier in the process. In summary, these data provide proof of concept for the expansion of low numbers of TILs and virus-specific T-cells from peripheral blood in a closed, automated manner in the CliniMACS Prodigy. In the future, this expansion process will be combined with a tumor-reactive T-cell enrichment process (e.g., via CD137-conjugated magnetic beads) to achieve the desired efficiency, simplicity and automated production of ACT therapies against","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer 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":"122502579","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 1
Abstract B024: Using single-cell paired sequencing to isolate cancer-specific T-cell receptors for cancer immunotherapy 摘要B024:利用单细胞配对测序分离肿瘤特异性t细胞受体用于肿瘤免疫治疗
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.cricimteatiaacr18-b024
Karolina Lech, Lúcia L. Correia, M. Beckmann, M. Busz, S. Collison, S. Davis, P. Mallini, S. Scaife, J. Dukes, B. Jakobsen, L. Williams, M. Teng
{"title":"Abstract B024: Using single-cell paired sequencing to isolate cancer-specific T-cell receptors for cancer immunotherapy","authors":"Karolina Lech, Lúcia L. Correia, M. Beckmann, M. Busz, S. Collison, S. Davis, P. Mallini, S. Scaife, J. Dukes, B. Jakobsen, L. Williams, M. Teng","doi":"10.1158/2326-6074.cricimteatiaacr18-b024","DOIUrl":"https://doi.org/10.1158/2326-6074.cricimteatiaacr18-b024","url":null,"abstract":"Immunocore’s ImmTAC™ (Immune Mobilising Monoclonal TCR Against Cancer) platform combines affinity-enhanced T-cell receptor (TCR)-based targeting with an anti-CD3 scFv effector function to activate a cytotoxic T-cell response against cancer cells. A key part to this process is the identification of tumour epitope specific TCRs from tumor antigen-reactive T-cells. Here, we describe an integrated in-house process leading to the isolation of TCRs specific for validated cancer epitopes, coupled with rapid identification of TCR chains from individual clones using single cell sequencing. The process involves first strand cDNA generation and universal amplification using SmartSeq2 chemistry, followed by targeted sequencing of the TCR alpha and beta chains using next-generation sequencing (NGS). We have also leveraged the 10x Genomics VDJ/5’ counting platform to label and pool multiple experimental clones for repertoire sequencing within a single run. Together with Cellular Indexing of Transcriptomes and Epitopes by sequencing (CITE-Seq), we can reliably assign each T-cell clone to its sample of origin paired with transcriptomic information of epitope specific T-cell populations, linking TCR sequences to their functional phenotype. Citation Format: Karolina Lech, Lucia Correia, Max Beckmann, Maria Busz, Sean Collison, Sterenn Davis, Paraskevi Mallini, Sarah Scaife, Joseph Dukes, Bent K. Jakobsen, Luke Williams, Michelle Teng. Using single-cell paired sequencing to isolate cancer-specific T-cell receptors for cancer immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B024.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"30 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":"116284162","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 PR13: A new high-performance HLA ligand identification strategy enables prediction of T-cell tolerance to neoepitopes PR13:一种新的高性能HLA配体鉴定策略可以预测t细胞对新表位的耐受性
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-PR13
M. Klatt, Ron S. Gejman, S. Moon, T. Korontsvit, T. Dao, D. Scheinberg
{"title":"Abstract PR13: A new high-performance HLA ligand identification strategy enables prediction of T-cell tolerance to neoepitopes","authors":"M. Klatt, Ron S. Gejman, S. Moon, T. Korontsvit, T. Dao, D. Scheinberg","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-PR13","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-PR13","url":null,"abstract":"T-cell responses against neoepitopes presented by human leukocyte antigen (HLA) complexes represent a critical effector of anticancer immunity. However, detection of neoepitopes by mass spectrometry is still challenging as is identification of the fraction of neoepitopes that elicit immune responses in vitro and in vivo. To address these problems, we developed a strategy to identify HLA ligands combining the peptide identification algorithm Byonic with the epitope binding predictor netMHCpan. We obtained up to 4-fold increases in unique HLA ligand identifications compared to standard approaches, false discovery rates below 0.6% and over 17,000 unique HLA ligand identifications in a single experiment. Even with only 10 million cancer cells we were able to detect over 5,000 unique HLA ligands, which allows our approach to be applied to small tumor samples in a clinical setting. Of note, phosphorylated as well as glycosylated HLA ligands were identified with the same accuracy and allowed a better characterization of the rules for presentation of post-translationally modified HLA ligands. Furthermore, by reanalyzing mass spectrometry samples with our novel approach, we confirmed high-confidence, rejected low-confidence and identified additional neoepitopes including the first-time detection of a phosphorylated neoepitope. Finally, we used our broadened knowledge of the immunopeptidome to create rules for prediction of non-immunogenicity of neoepitopes based on high biochemical similarity with unmutated HLA ligands, which showed high specificity and positive predictive value when validated with two datasets derived from neoepitope-based clinical studies. Altogether, our methods substantially improved sensitivity and specificity for detection of native, modified and mutated HLA ligands, propose an explanation for tolerance to neoepitopes, and should in combination facilitate the design of neoepitope-based therapies. Citation Format: Martin G. Klatt, Ron S. Gejman, Sung S. Moon, Tatyana S. Korontsvit, Tao Dao, David A. Scheinberg. A new high-performance HLA ligand identification strategy enables prediction of T-cell tolerance to neoepitopes [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 PR13.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"62 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":"126252361","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 B051: Facilitating translational research with interactive tools for immuno-oncology data B051:利用免疫肿瘤学数据的交互式工具促进转化研究
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B051
V. Thorsson, James A. Eddy, Andrew Lamb, David L. Gibbs, I. Shmulevich, J. Guinney
{"title":"Abstract B051: Facilitating translational research with interactive tools for immuno-oncology data","authors":"V. Thorsson, James A. Eddy, Andrew Lamb, David L. Gibbs, I. Shmulevich, J. Guinney","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B051","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B051","url":null,"abstract":"With the explosive growth in data and results from immuno-oncology (IO) studies, improved ways to easily share, integrate and explore available data and results are needed. The Cancer Research Institute (CRI) iAtlas (www.cri-iatlas.org) is an interactive web-based platform and set of analytic tools for studying interactions between tumors and the immune microenvironment. These tools allow researchers to explore associations among a variety of immune characterizations as well as with genomic and clinical phenotypes. The initial version of CRI iAtlas is based on an analysis performed by The Cancer Genome Atlas (TCGA) Research Network on the TCGA data set comprising over 10,000 tumor samples and 33 tumor types (Thorsson et al., 2018). The platform will be expanded to include other immunogenomic data sets and workflows. In the TCGA analysis, each tumor sample was scored for a variety of computationally estimated immune-based readouts, including immune cell composition, adaptive cell receptor repertoire, neoantigen load, and expression of genes coding for immunomodulatory proteins. Immune-based subtypes, spanning multiple tumor types, were identified. The web tool allows researchers to explore the data readouts as well as the relation between them in individual TCGA tumor types and across the global immune subtypes identified in the study. CRI iAtlas is made possible through a collaboration between CRI, Sage Bionetworks, and the Institute for Systems Biology. The main feature of the iAtlas web tool is the iAtlas Explorer, which provides several Analysis Modules to explore and visualize results. Each module presents information organized by theme, with multiple views and interactive controls to enhance and extend the information included in the original manuscript figures. Sample Group Overview: summaries of selected groups (including six immune subtypes that span cancer tissue types and molecular subtypes); Tumor Microenvironment: overall immune infiltrate and relative immune cell proportions in selected sample groups; Immune Feature Trends: distributions of immune readouts across selected groups, and associations between readouts within groups; Clinical Outcomes: trends of and associations with survival outcomes across sample groups based on immune characteristics; Immunomodulators: expression trends of genes that code for immunomodulating proteins, including checkpoint proteins. In response to community feedback, we are extending the iAtlas portal with two modules, one allowing researchers to classify their own RNAseq samples into immune subtypes, and the other allowing researchers to upload their own sample categories for analysis with the tool. As the resource evolves, we expect that the CRI iAtlas will help to accelerate discovery and improve patient outcomes by providing researchers greater access to immunogenomics data to better understand the immunologic characteristics of the tumor microenvironment and its potential impact on patient response","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"21 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":"132212223","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 B048: Proteomic approaches for the identification of druggable protein and epigenetic targets to complement melanoma immunotherapy B048:蛋白质组学方法用于鉴定可药物蛋白和表观遗传靶点,以补充黑色素瘤免疫治疗
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B048
A. Tackett
{"title":"Abstract B048: Proteomic approaches for the identification of druggable protein and epigenetic targets to complement melanoma immunotherapy","authors":"A. Tackett","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B048","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B048","url":null,"abstract":"Introduction: Immune checkpoint inhibitors (ICIs) have shown encouraging success for treatment of melanoma; however, approximately half of patients with advanced melanoma have primary ICI resistance and sub-populations of initially responding patients develop secondary resistance. To help address this, our group has used quantitative proteomic workflows to identify druggable molecular pathways in these non-responders. Specifically, we are targeting the identification of protein pathways involved in regulation of gene transcription and chromatin modification as these are upstream events in cellular programming. Experimental Procedures: We have used cutting-edge proteomic workflows to identify proteins and histone post-translational modifications that are dysregulated in ICI nonresponsive patient melanoma. Proteins were isolated from FFPE patient melanomas and analyzed by high-resolution mass spectrometry with a Thermo Fusion Orbitrap mass spectrometer. Mass spectrometric data was searched for proteins and histone post-translational modifications dysregulated in ICI nonresponding melanomas. Pathway analysis provided for the identification of molecular pathways dysregulated in ICI nonresponders. New Data: We have published a smaller-scale version of this proteomic study comparing 4 responding and 4 nonresponding melanomas (Sci Reports 2017;7:807). In new data, we have increased the sample size of this comparison to provide increased power and significance. We have also expanding our quantitative proteomic analysis by incorporating the use of Thermo Tandem Mass Tagging with FFPE samples. Furthermore, we present new workflows using fresh melanoma tumors that provide for specific enrichment of melanoma cells from human tumors. Conclusions: We have identified protein and histone epigenetic pathways that are dysregulated in ICI nonresponsive patient melanomas. These molecular pathways could be prime targets for therapeutic development for increasing responsiveness to ICI therapy. Furthermore, we have extended our proteomic capabilities to the analysis of enriched populations of melanoma cells isolated from fresh human tumors, which will provide for cell type-specific proteomic studies. Citation Format: Alan Tackett. Proteomic approaches for the identification of druggable protein and epigenetic targets to complement melanoma immunotherapy [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B048.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"284 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":"133504768","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 B013: Site specific conjugation of engineered non-native amino acids in an anti-CD3 Fab-folate bispecific antibody significantly enhances its antitumor properties in gynecologic cancers 摘要B013:在抗cd3 fab -叶酸双特异性抗体中,工程非天然氨基酸的位点特异性偶联显著增强了其抗妇科肿瘤的抗肿瘤特性
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B013
M. Gray, Wisam Barkho, B. Tipton, P. Shastri, H. Jie, J. Steen, R. Frank, Feng Tian, D. Jackson, Shawn Zhang
{"title":"Abstract B013: Site specific conjugation of engineered non-native amino acids in an anti-CD3 Fab-folate bispecific antibody significantly enhances its antitumor properties in gynecologic cancers","authors":"M. Gray, Wisam Barkho, B. Tipton, P. Shastri, H. Jie, J. Steen, R. Frank, Feng Tian, D. Jackson, Shawn Zhang","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B013","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B013","url":null,"abstract":"Ovarian cancer is the seventh most commonly diagnosed cancer in women. Standard treatments typically include surgery followed by radiation and/or chemotherapy. However, currently 5-year survival rates are 44%, underlining the need for new effective therapies. The Folate receptor 1 (FOLR1, FRα) is expressed in 80% of ovarian tumors, making it an attractive target for CD3-based bispecific antibody therapies. Bispecific engagement of activated host T-cells in the tumor microenvironment is highly effective at eliminating tumor cells. We have developed an anti-CD3 Fab-folate bispecific antibody by engineering non-native amino acids (NNAAs) at a specific region within the Fab domain and conjugated a PEGylated folate molecule to these sites. Our data demonstrate that Ambrx’s proprietary site-specific conjugation technology provides significant advantages over non-PEGylated proteins. Citation Format: Michael J. Gray, Wisam Barkho, Barbara Tipton, Prathap Shastri, Hyun-Bae Jie, Jeff Steen, Rik Frank, Feng Tian, Dowdy Jackson, Shawn Zhang. Site specific conjugation of engineered non-native amino acids in an anti-CD3 Fab-folate bispecific antibody significantly enhances its antitumor properties in gynecologic cancers [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 B013.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"52 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":"132734554","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 B058: Rapid and controlled T-cell expansion using scaffolds that mimic antigen-presenting cells B058:利用模拟抗原呈递细胞的支架快速和控制t细胞扩增
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B058
David Y. Zhang, A. Vaynrub, D. Mooney
{"title":"Abstract B058: Rapid and controlled T-cell expansion using scaffolds that mimic antigen-presenting cells","authors":"David Y. Zhang, A. Vaynrub, D. Mooney","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B058","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B058","url":null,"abstract":"Current approaches to expand T-cells ex vivo for therapeutic applications are limited by low expansion rates and often result in unpredictable expansion products of varying phenotype and function. Key attributes, such as a balanced ratio of CD4-to-CD8 T-cells, or a high central memory T-cell fraction are known to correlate with robust and durable therapeutic responses but remain difficult to reproducibly achieve. Strategies that allow one to precisely control the phenotype of expanded T-cell products may thus potentiate the effects of CAR-T-cell therapies, and understanding how T-cells respond to stimulation is a crucial step to this end. Recently, we described a 3D expansion system that mimics natural antigen presenting cells (APCs) and promotes greater polyclonal expansion of primary human T-cells than commercial expansion beads (Dynabeads). These biodegradable, APC-mimetic scaffolds (APC-ms) are composed of fluid lipid bilayers supported on mesoporous silica micro-rods, which enables precise control over the spatial organization of membrane-bound T-cell receptor (TCR) stimulatory and costimulatory cues (i.e., anti-CD3/anti-CD28). Here, we investigated the activation and memory signature of primary human T-cells over time in response to APC-ms with different densities of activating surface cues compared to Dynabead stimulation. Higher surface cue densities of anti-CD3/anti-CD28 promoted greater proportions of central memory T-cells, with a parallel increase in the CD4-to-CD8 T-cell ratio. T-cells stimulated with Dynabeads exhibited higher expression of CD25 and CD69 that peaked earlier than standard APC-ms formulations, suggesting that APC-ms promotes relatively more transient T-cell activation than Dynabeads, potentially mitigating overstimulation and exhaustion. Overall, these data demonstrate that APC-ms can be used to rapidly expand primary human T-cells and tune their phenotypic attributes (e.g., CD4-to-CD8 ratio and memory subpopulations). This technology has the potential to improve the production of T-cells for adoptive T-cell-based therapies. Citation Format: David K.Y. Zhang, Anna Vaynrub, David J. Mooney. Rapid and controlled T-cell expansion using scaffolds that mimic antigen-presenting cells [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Conference: Translating Science into Survival; Sept 30-Oct 3, 2018; New York, NY. Philadelphia (PA): AACR; Cancer Immunol Res 2019;7(2 Suppl):Abstract nr B058.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"26 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":"123908124","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 B052: Immunotherapy for melanoma by adenovirus-mediated full-length antibody, nivolumab B052:腺病毒介导的全长抗体nivolumab对黑色素瘤的免疫治疗
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B052
Xuchen Wang
{"title":"Abstract B052: Immunotherapy for melanoma by adenovirus-mediated full-length antibody, nivolumab","authors":"Xuchen Wang","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B052","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B052","url":null,"abstract":"Nivolumab, a fully human, IgG4 immune checkpoint inhibitor antibody, binds PD-1 on activated immune cells to disrupt PD-1 interaction with PD-L1 and PD-L2 ligands, thereby attenuating inhibitory signals and augmenting the host antitumour response. In 2014, nivolumab was approved by FDA and has been used to treat various types of cancer such as melanoma, non-small cell lung cancer, renal cell carcinoma, and classic Hodgkin9s lymphoma. However, due to the high price of nivolumab and the long cycle of treatment, many patients cannot afford and receive adequate treatment. Here, we generated a new, cheaper form of nivolumab for cancer immunotherapy, by cloning the full-length nivolumab antibody gene into two serotypes of adenovirus vectors, termed as AdHu5-Nivo and AdC68-Nivo. Ad vectors based on human serotype 5 (AdHu5) have been proved in a lot of previous studies to be safe and efficient. Compared with AdHu5, chimpanzee Ads exhibit much lower seroprevalence in human beings, which made them great alternative Ad gene vector. Until now, we have detected a high expression of nivolumab in vitro by Western blot and sandwich ELISA. In vivo studies showed that a single dose of AdHu5-Nivo or AdC68-Nivo can induce sustained nivolumab expression. The biologic activities of the two mAbs are analogous compared with commercial monoclonal antibody. To follow up we will verify their antitumor effects on both cellular level and melanoma transplant animal models. Citation Format: Xuchen Wang. Immunotherapy for melanoma by adenovirus-mediated full-length antibody, nivolumab [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 B052.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"23 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":"129681952","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 B012: Characterizing tumor-induced exhaustion in melanoma patients treated with neoadjuvant pembrolizumab B012:新辅助派姆单抗治疗黑色素瘤患者肿瘤诱导衰竭的特征
Convergence of Technology and Cancer Immunotherapy Pub Date : 2019-02-01 DOI: 10.1158/2326-6074.CRICIMTEATIAACR18-B012
Josephine R. Giles, Alexander C. Huang, S. Manne, Jorge Henao-Meji, E. Wherry
{"title":"Abstract B012: Characterizing tumor-induced exhaustion in melanoma patients treated with neoadjuvant pembrolizumab","authors":"Josephine R. Giles, Alexander C. Huang, S. Manne, Jorge Henao-Meji, E. Wherry","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B012","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B012","url":null,"abstract":"Immune checkpoint blockade, including anti-PD-1 therapies, has had unprecedented success in treating various forms of cancer, including melanoma. Yet the majority of patients do not achieve durable clinical remission. The limited understanding of how these therapies work at the cellular and molecular level prevents the optimization required to improve patient outcomes. We performed RNA-seq and ATAC-seq on four sorted T-cell subpopulations (naive CD8 T-cells, non-naive CD8, non-naive CD4 T-cells, T regulatory cells) from patients with metastatic melanoma before and after pembrolizumab treatment. While there were no significant changes before and after treatment in the sorted peripheral blood populations, we identified many alterations in the tumor compared to the blood following treatment. These differences may represent impediments to fully reinvigorating exhausted T-cells and be potential targets for achieving more durable clinical responses. We identified a zinc finger gene, ZC3H12C, as not only one of the most highly upregulated in non-naive CD8 T-cells in the tumor, but it also had the highest number of associated altered open chromatin regions. This gene and its locus are also significantly different in exhausted T-cells compared to effector and memory cells in a mouse model of chronic viral infection using Lymphocytic choriomeningitis (LCMV). This conservation between species suggest an important role in T-cell exhaustion and provided an avenue to test its function in vivo, so we generated ZC3H12C knockout mice using CRISPR-Cas9. Following C13 (chronic) LCMV infection, virus-specific P14 CD8 T-cells lacking ZC3H12C are unable to persist compared to wild type P14 cells. These results suggest ZC3H12C may be critical to the survival of exhausted T-cells in the tumor and their ability to persist after anti-PD1 treatment. Here, we used transcriptomic and epigenetic analysis of a clinical human cancer data set to identify a novel target and used reverse translation in a mouse model to validate its importance in T-cell exhaustion. This gene and more broadly this strategy have the potential to provide immediately useful targets for improving immunotherapies. Citation Format: Josephine R. Giles, Alexander C. Huang, Saskinath Manne, Jorge Henao-Meji, E. John Wherry. Characterizing tumor-induced exhaustion in melanoma patients treated with neoadjuvant pembrolizumab [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 B012.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"35 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":"127444131","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
0
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
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
确定
请完成安全验证×
相关产品
×
本文献相关产品
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 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信