T. Ochi, Masaki Maruta, K. Tanimoto, H. Asai, Takashi Saitou, Y. Yakushijin, H. Fujiwara, Takeshi Imamura, K. Takenaka, M. Yasukawa
{"title":"Abstract B031: Development of antimyeloma immunotherapy by exploiting modified antibodies specific for A2/NY-ESO-1","authors":"T. Ochi, Masaki Maruta, K. Tanimoto, H. Asai, Takashi Saitou, Y. Yakushijin, H. Fujiwara, Takeshi Imamura, K. Takenaka, M. Yasukawa","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B031","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B031","url":null,"abstract":"Background: T-cell therapy can be a promising treatment option even in patients with refractory malignancies including myeloma. NY-ESO-1 is a well-known cancer-testis antigen which is expressed by refractory myeloma cells, and a NY-ESO-1_157-165 peptide presented by an HLA-A*02:01 molecule (A2/NY-ESO-1_157) has been demonstrated. Adoptive transfer therapy using T-cells modified with T-cell receptor (TCR) specific for A2/NY-ESO-1_157 successfully induced clinical responses in patients with advanced myeloma. However, TCR-transduced T-cells are laborious to generate and possess the cross-reactivity induced by mispaired and/or introduced TCRs, resulting in increase of unwanted toxicities. T-cell therapy utilizing modified antibodies containing single chain fragment variables (scFvs), such as chimeric antigen receptor (CAR) and bispecific antibody (BiTE) would overcome the issues concerning TCR-T therapy and expand clinical versatility of T-cell therapy targeting NY-ESO-1 in the treatment of refractory myeloma. In this study, we have generated both CAR and BiTE which recognize A2/NY-ESO-1_157, and assessed their anti-myeloma reactivity and cross-reactivity in vitro and in vivo. Methods: Expression of NY-ESO-1 in a panel of myeloma cell lines was examined by real-time PCR and Western blotting. Based on the structure of previously reported monoclonal antibody specific for A2/NY-ESO-1_157 (clone: 3M4E5), we newly synthesized an A2/NY-ESO-1_157-specific scFv. Second generation CAR possessing an scFv linked with CD28 and CD3z was generated. A BiTE composed of an A2/NY-ESO-1_157-specific scFv and a CD3e-binding scFv was also generated. A2/NY-ESO-1_157-specific reactivity mediated by CAR and BiTE-redirected T-cells were assessed by A2/NY-ESO-1_157 tetramer and multiple cytokine assays. Specific lysis of targeT-cells by those T-cells was measured by standard Cr-release assay. Alanine scanning of NY-ESO-1_157 peptide was performed, and nine peptides homologous to NY-ESO-1_157 were synthesized. Cross-reactivity of CAR and BiTE-redirected T-cells for these peptides and NY-ESO-1_157 peptide presented by HLA-A2 alleles was evaluated. NOG mice engrafted with a luciferase-transduced A2+NY-ESO-1+ myeloma cell line (U266/SLR) were treated with CAR-T-cells or T-cells with BiTE, and tumor sizes were measured by bioluminescence imaging assays. Results: Three out of six myeloma cell lines we tested abundantly expressed NY-ESO-1 mRNA and protein. CAR-T-cells established from five out of five donors showed A2/NY-ESO-1_157-specific reactivity. These gene-modified T-cells recognized and killed targeT-cells which naturally process and present A2/NY-ESO-1_157, resulting in anti-myeloma reactivity to A2+NY-ESO-1+ U266 myeloma cells. Newly generated BiTE successfully engaged A2/NY-ESO-1_157 expressing targeT-cells with CD3+ T-cells, thereby peripheral T-cells produced multiple cytokines against A2+NY-ESO-1+ targeT-cells, and lysed them. CAR and BiTE-redirected T-cells can possess","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer 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":"123172722","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}
Y. Kikuchi, T. Kanaseki, Ayumi Hongo, S. Tokita, T. Torigoe, K. Vitaly
{"title":"Abstract B018: Landscape of natural HLA class I ligand peptides of cancer cells","authors":"Y. Kikuchi, T. Kanaseki, Ayumi Hongo, S. Tokita, T. Torigoe, K. Vitaly","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B018","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B018","url":null,"abstract":"HLA class I ligand repertoire on cancer cells directly influences immune surveillance system against cancer. We performed large-scale mass spectrometric profiling of the natural HLA-A*2402 ligand peptides and unveiled the landscape of natural HLA class I ligand peptides derived from various cancer cells including colon cancer cells (SW480, Colo320, HCT15) and lung cancer cells (LHK2, Sq-1). Known A24-binding anchors (Y/F at P2 and F/L/I at P9) were largely conserved among the ligands; however, a subset of peptides had an unusual anchor (K/R at the C-terminal P9 or P10), suggesting diverse usage of anchors in certain types of cancer cells. Moreover, a certain amino acid residue had significantly low frequency in the N-terminal flanking sequence, suggesting that it might inhibit processing of the peptides. Profiling of the natural HLA ligands of cancer cells revealed \"neoantigen\" peptides derived from mutated genome DNA and \"aberrant antigen\" peptides derived from long noncoding RNAs as well as \"cancer-germ cell antigen\" peptides. Among the natural HLA ligands derived from cancer cells, immunogenicity of the \"neoantigen\" peptide was the highest. The landscape clarified immunogenic peptide repertoire that might be suitable for cancer vaccine as well as as-yet-unknown rules for antigen processing. Citation Format: Yasuhiro Kikuchi, Takayuki Kanaseki, Ayumi Hongo, Serina Tokita, Toshihiko Torigoe, Kochin Vitaly. Landscape of natural HLA class I ligand peptides of cancer 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 B018.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"55 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":"131425529","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}
Tripti Tamhane, S. Saini, R. Anjanappa, Ankur Saikia, S. Ramskov, M. Donia, I. Svane, S. Jakobsen, M. Garcia-Alai, M. Zacharias, R. Meijers, S. Springer, S. Hadrup
{"title":"Abstract B049: Empty MHC class I molecules for improved detection of antigen-specific T-cells","authors":"Tripti Tamhane, S. Saini, R. Anjanappa, Ankur Saikia, S. Ramskov, M. Donia, I. Svane, S. Jakobsen, M. Garcia-Alai, M. Zacharias, R. Meijers, S. Springer, S. Hadrup","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B049","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B049","url":null,"abstract":"Major histocompatibility complex (MHC) class I multimers have been widely used to identify antigen specific T-cells for immune monitoring, epitope discovery, and T-cell isolation. A bottleneck to many peptide-MHC driven applications for T-cell interrogation is the peptide ligand dependent stability of the MHC class I proteins, which thus compels high-affinity peptide dependent in-vitro folding of each MHC protein and the use of a peptide-exchange technology to investigate antigens of interest. To overcome this challenge, we demonstrate the use of empty peptide-receptive MHC class I molecule for detection of antigen specific T-cells. This strategy is based on an HLA-A*02:01 variant which is stabilized by a disulfide bond to link the alpha-1 and alpha-2 helices close to the F pocket. Determined by the crystal structure, peptide-loaded disulfide-stabilized HLA-A*02:01 show complete structural overlap to wild-type HLA-A*02:01. Following peptide loading, we used such disulfide-stabilized HLA-A*02:01 molecules to form fluorescence labeled tetramers and applied them for detections of T-cell responses against common viruses in healthy donor peripheral blood mononuclear cells. In all tested samples, disulfide-stabilized HLA-A*02:01 tetramers detected T-cell with same specificity as wild-type MHC tetramers and they consistently provide a better staining index for antigen-specific T-cell detection. Importantly, disulfide-stabilized MHC class I molecules can be loaded with peptide in the multimerized form without impacting the T-cell staining capacity. We demonstrate the value of empty loadable tetramers, converted to antigen-specific tetramers by a single-step peptide addition, for identification of T-cells specific to several neo- and cancer-associated antigens among tumor-infiltrating lymphocytes in melanoma.To evaluate if the disulfide linkage has an impact on TCR recognition of peptide-MHC complexes, we determined and compared TCR fingerprints of T-cell clones specific to a given peptide-MHC complex using both the wild-type and the disulfide-stabilized HLA-A*02:01 multimers. No differences were observed in the TCR interaction profile between the disulfide optimized and the wild-type MHC class I. In conclusion, disulfide-stabilized empty HLA class I proteins are a potentially powerful tool for interrogating T-cell recognition—offering a fast and flexible transformation from an empty peptide receptive state to a set of personalized reagents generated to match individual tumor characteristics for T-cell monitoring or selection. Citation Format: Tripti Tamhane, Sunil Kumar Saini, Raghavendra Anjanappa, Ankur Saikia, Sofie Ramskov, Marco Donia, Inge Marie Stenfoft Svane, Soren Nyboe Jakobsen, Maria Garcia-Alai, Martin Zacharias, Rob Meijers, Sebastian Springer, Sine Reker Hadrup. Empty MHC class I molecules for improved detection of antigen-specific T-cells [abstract]. In: Proceedings of the Fourth CRI-CIMT-EATI-AACR International Cancer Immunotherapy Confer","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer 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":"128844815","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}
{"title":"Abstract B025: Multiplex three-dimensional mapping of mRNA and protein in the tumor microenvironment","authors":"S. S. Lee, D. Scholten, V. Bindokas, S. Kron","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B025","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B025","url":null,"abstract":"Detecting oncogenic protein-coding messenger RNA (mRNA) in tumor tissues is a key assay for accurate cancer diagnosis. Along with cancer protein biomarker analysis, mRNA identification is critical for matching the appropriate treatment to individual patients. Likewise, cytokine mRNA localization in on-treatment biopsies enables evaluation of the antitumor response to immunotherapy by identifying the types and functional states of lymphocytes, for example distinguishing activated cytotoxic T-cells from naive T-cells. To assess mRNA localization, a biopsy is typically formalin fixed and paraffin embedded (FFPE), thin sections are cut, and then are stained for mRNA by chromogenic In situ hybridization (CISH). Analyzing samples by mRNA CISH typically takes a few days and requires considerable technical expertise. Recent progress in multiplexed fluorescence In situ hybridization (FISH) has enabled simultaneous analysis of multiple mRNA types in a single tissue section. Nonetheless, even if fully characterized, individual tissue sections cannot adequately represent the complex three-dimensional (3D) architecture of the tumor microenvironment. There remains a pressing need for a rapid sample-to-answer technology for multiplex 3D imaging of mRNA at sub-cellular resolution that is able to fully characterize the tumor microenvironment in intact biopsy tissues for cancer diagnosis and immunotherapeutic response analysis. To address these challenges, we have adapted Transparent Tissue Tomography (T3), a simple and fast tissue clearing and multiplex 3D imaging method, to analyze mRNA distribution in the tumor microenvironment. With T3, oncogenic and immunoregulatory cytokine mRNAs are readily localized in the context of cell types and other features in murine, patient derived xenograft (PDX), and head and neck tumors. This work establishes T3 as a tool for a quantitative, 3D spatial analysis of mRNA and protein distribution in the tumor microenvironment. Citation Format: Steve Seung-Young Lee, David Scholten, Vytautas P. Bindokas, Stephen Kron. Multiplex three-dimensional mapping of mRNA and protein in the tumor microenvironment [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 B025.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer 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":"129653405","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}
{"title":"Abstract B043: Enhanced T-cell immunity in vivo using injectable bioengineered scaffolds","authors":"Nisarg J. Shah, D. Mooney","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B043","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B043","url":null,"abstract":"Hematopoietic stem cell transplantation (HSCT) is a potentially curative therapy for hematological malignancies and immunologic disorders, but allogeneic HSCT is limited by deficiency and dysregulation of T-cells. We aimed to enhance post-HSCT T-cell reconstitution and hypothesized that a T-cell lymphopoietic bone marrow niche might be engineered to foster production of T-cell progenitors in vivo, which can undergo host-driven selection. To test this hypothesis, we created an injectable biomaterial-based scaffold that promotes T-cell development in vivo by recapitulating key features of the bone marrow niche. The composite device, referred to as a bone marrow cryogel (BMC), is comprised of a macroporous hydrogel-based scaffold permitting cellular infiltration. The presentation of T-lineage cues in a bone marrow-like microenvironment enhanced thymic seeding of progenitors and facilitated the enhancement of donor T-cell reconstitution after HSCT in mice. The BMC-reconstituted T-cells were functional, with a diverse T-cell receptor repertoire and modulated GVHD. BMC may represent a simple to administer, off-the-shelf approach to post-HSCT T-cell regeneration. Citation Format: Nisarg Shah, David J. Mooney. Enhanced T-cell immunity in vivo using injectable bioengineered scaffolds [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 B043.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"66 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":"133633745","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}
Luis-Alberto Pérez-Quintero, Kelly A. Pike, P. Claudia, M. Tremblay
{"title":"Abstract B035: Enhancing antitumoral activity of cell-based immunotherapies by modulating the JAK-STAT axis","authors":"Luis-Alberto Pérez-Quintero, Kelly A. Pike, P. Claudia, M. Tremblay","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B035","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B035","url":null,"abstract":"Mounting a T helper 1 (Th1) type of response is required for the successful priming of antigen specific CD8 T-cells which ultimately lead to effective antitumoral responses. Several cellular components of the immune system participate of the Th1 decision-making process. They include innate cells sensing the transformed cells, mainly dendritic cells (DCs), and the further involvement of antigen specific CD4 T-cells promoting direct activation of CD8 T-cells through cross-presentation (1). The correct stimulation of DCs by type I interferons and molecular pattern sensors as STING is required to drive their differentiation to conventional DC 1 (cDC1) which through the secretion of large amounts of IL-12 and the expression of CD40 skew the CD4 T-cells differentiation to Th1 (2). However, this system is sensitive to modifications of the tumor cytokine microenvironmen,t leading to failure. Adoptive cell transfer (ACT) cancer immunotherapies are promising treatments for advanced malignancies. These therapies attempt to supply key cellular actors missing and induce proper antitumoral immunity. They include mainly the in vitro modification of autologous cells, from DCs loaded with tumor antigens to the expression of chimeric antigen receptor (CAR) on T and NK cells. Constant development of these technologies has given successful results as clinical trials have already showed up to 90 % of complete remission in relapsed/refractory B cell acute lymphoid leukemia (B-ALL) (3), treatment now approved for commercial use. However, there is still lack of consistency in the responses obtained from different individuals and in different trials. Previously our laboratory has already shown that modulation of the JAK-STAT inhibitory protein tyrosine phosphatases (PTPs), PTPN1 and PTP-N2, enhance proinflammatory type I interferon signalling while decreases the effects of immunosuppressive cytokines acting through STAT3 signaling (4, 5). Specifically, we have demonstrated that partial inhibition of these phosphatases in immune cells enhances the secretion of IL-12p70 by DCs[6] and increases the cytotoxic activity of antigen specific CD8 T-cells [Perez-Quintero LA, Tremblay ML, unpublished results]. Moreover, the partial inhibition of these phosphatases in CD8 T-cells enhance the acquisition of a central memory (Tcm) phenotype, which has been found to be associated with better remission in CAR-T-cells therapies. Nevertheless, alternative methods to enrich this phenotype, as cytokine cocktails, have proven expensive and ineffective. Having this in mind, we propose here the use of small-molecule inhibitors specific for PTPN1 and PTPN2 as a simple and cost-effective method to enhance antitumoral responses. By treating ex vivo the cellular products in animal models for DCs and CD8 ACT therapies we show, as a proof of concept, a marked improvement on the reduction of tumor burden and remission, with the late goal of translating these findings into a clinical setup. References","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"18 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":"122392754","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}
N. Dupuis, J. Vowinckel, Daniel Heinzmann, C. Escher
{"title":"Abstract B010: A survey of circulating biomarkers in subjects with NSCLC using library-based data independent acquisition mass spectrometry reveals host immune response mechanisms","authors":"N. Dupuis, J. Vowinckel, Daniel Heinzmann, C. Escher","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B010","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B010","url":null,"abstract":"Background: Identification of circulating biomarkers in cancer has proven utility in applications for early detection, differential diagnosis, predicting pre-treatment response to therapy, and treatment monitoring. More recently, circulating proteomic biomarkers have been evaluated as surrogate endpoints for early indication of benefit for immunotherapies. This last application is especially relevant during immunotherapy development where the optimal endpoint, overall survival (OS), can take longer to mature. Here, we present an unbiased survey of the circulating proteome of subjects with NSCLC to identify candidate biomarkers that may have utility in multiple stages of patient care. Methods: Unbiased, data-independent acquisition (DIA) mass spectrometry was used to analyze plasma samples from subjects with Stage III-IV non-small cell lung cancer (NSCLC, n = 15) and age matched healthy donors (n = 15), enabling simultaneous sequencing and quantification of plasma proteins. Samples were prepared for mass spectrometry and spiked with a panel of standards covering 500 plasma proteins. All samples were analyzed using 1h gradients on a C18 column coupled to a Thermo Scientific Q Exactive HF mass spectrometer. Data were extracted using Spectronaut (Biognosys) with a sample specific spectral library and statistical analysis was conducted to identify disease associated biomarker candidates. Pathway analysis highlights dysregulated biologic functions and predicts upstream regulatory pathways. Results: A protein library was created containing 771 unique proteins. In DIA acquisition, 462 proteins were quantified across all samples. Univariate statistical testing identified 26 dysregulated proteins (20 up-regulated and 6 down-regulated; q-value > 0.05 and log2 fold change > 0.58). Multivariate (PLS-DA) analysis identified c-reactive protein (CRP) and serum amyloid a (SAA1/SAA2), complement C9, S100A8/S100A9, and leucine rich glycoprotein 1 (LRG1) as the most significantly changed proteins across sample groups. Receiver operator analysis (ROC) identified S100A8 and complement C9 as the markers with the greatest diagnostic power at 93% sensitivity and 93% specificity. Significantly enriched pathways include acute phase response, complement system as well as IL-12 and IL-6 signaling. Similarly, upstream activated candidate pathways included STAT3, IL-6, and EZ2H. Conclusions: 26 proteins were identified as candidate biomarkers and reflect the host immune response via acute phase response signaling, innate immune response (complement system), and other proinflammatory stimuli. Several of these markers have been linked to patient outcomes and poor prognosis. Accurate monitoring of these proteins offers the possibility to define surrogate, molecular-based markers with multiple modes of utility. Citation Format: Nicholas Dupuis, Jakob Vowinckel, Daniel Heinzmann, Claudia Escher. A survey of circulating biomarkers in subjects with NSCLC using library-based data inde","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"6 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":"125162966","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}
{"title":"Abstract IA35: Determinants of effective tumor immunity","authors":"N. Hacohen","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-IA35","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-IA35","url":null,"abstract":"Treatment of solid tumors has been revolutionized by immune checkpoint blockade therapies; yet even for melanoma, for which high response rates are observed, the majority of tumors continue to grow after therapy. To identify immune cell states associated with success or failure of immunotherapy, we profiled the single-cell transcriptomes of immune cells from tumor samples obtained from melanoma patients treated with checkpoint inhibitors. We defined specific activation states of CD8+ T cells that associate with tumor growth post-therapy, delineated their epigenetic landscape and clonality, and demonstrated enhanced immunity by targeting a novel combination of T cell factors. We also found that expression of a single T cell gene was predictive of tumor regression in response to therapy. Our study thus reveals immune cell states and molecules that help explain effective immunotherapy in humans. Citation Format: Nir Hacohen. Determinants of effective tumor immunity [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 IA35.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"12 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":"117132381","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}
Rami Yossef, E. Tran, A. Gros, D. Deniger, G. Cafri, S. Rosenberg
{"title":"Abstract B055: Enhanced detection of T-cells targeting unique neoantigens and shared mutated oncogenes for personalized cancer immunotherapy","authors":"Rami Yossef, E. Tran, A. Gros, D. Deniger, G. Cafri, S. Rosenberg","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B055","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B055","url":null,"abstract":"Adoptive cell transfer (ACT) of selected tumor-infiltrating lymphocytes (TILs) targeting neoantigens can mediate tumor regression in selected patients with metastatic epithelial cancer. However, effectively identifying and harnessing neoantigen-reactive T-cells for patient treatment remains a challenge and it is unknown whether current methods to detect neoantigen-reactive T-cells are missing potentially clinically relevant neoantigen reactivities. We thus investigated whether the detection of tumor-neoantigen reactive TILs in epithelial cancers could be enhanced by enriching T-cells that express PD-1 and/or T-cell activation markers (CD134, CD137) followed by microwell culturing at limiting dilution cell concentrations to avoid overgrowth of non-reactive T-cells. Using this approach, in six patients with metastatic epithelial cancer including stomach, colon pancreatic and ovarian cancers, this method led to the detection of CD4 and CD8 T-cells targeting 19 neoantigens compared to only eight neoantigens recognized using our standard TIL fragment screening approach. In two patients, no recognition of mutated peptides was observed using our conventional screen, while our high-throughput approach led to the identification of five neoantigen reactive-TCRs against five different mutations from one patient and a highly potent MHC-II-restricted KRASG12V reactive TCR from a second patient. In addition, in a metastatic tumor sample from a patient with serous ovarian cancer, we isolated three MHC class-II-restricted TCRs targeting the TP53G245S “hot-spot” mutation. In conclusion, this approach provides a highly sensitive and specific platform to isolate clinically relevant neoantigen-reactive T-cells or their TCRs for cancer treatment. Citation Format: Rami Yossef, Eric Tran, Alena Gros, Drew Deniger, Gal Cafri, Steven A. Rosenberg. Enhanced detection of T-cells targeting unique neoantigens and shared mutated oncogenes for personalized 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 B055.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer 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":"114914465","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}
{"title":"Abstract B011: Inhibition of G9a reestablishes the MHC class I loss due to EMT in lung cancer cells","authors":"H. Fukumasu, P. R. L. Pires, P. Xavier","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B011","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B011","url":null,"abstract":"The epithelial-to-mesenchymal transition (EMT) is an important phenotype for cancer cells to invade and metastasizes. In addition, cancer cells should escape from the immune system during malignant progression. Here, we aimed to demonstrate the potential of G9a inhibition of EMT in cancer cells and most importantly, the reestablishment of MHC class I expression (MHC-I), one of the important factors associated with the loss of antigenicity of cancer cells. The lung epithelial cancer cell line A549 was cultured under controlled conditions in RPMI culture media supplemented with 10% of bovine fetal serum (BFS), 1% of antibiotics (penicillin and streptomycin), 2% of glutamine in incubator at 37oC and air atmosphere containing 5% of CO2. The tumor growth factor beta (TGFb) exposure for 5 days was used to induce EMT. In addition, another group of A549 cells received TGFb plus the UNC0368 an G9a (a histone methyl transferase) inhibitor for 5 days. Then, cells were evaluated for acquiring mesenchymal cell morphology and MHC-I gene expression (relative quantification). After 5 days, A549 cells exposed to TGFb clearly have gone through EMT by changing their morphology to a fibroblast-like phenotype. Also, the MHC-I gene expression decreased significantly (p=0.0036) after EMT. The G9a inhibitor partially impeded the EMT induced changes in morphology of A549 cells and reestablished the MHC-I gene expression to the control level. In conclusion, G9a inhibition seemed a promising therapeutic target to improve the efficacy of immunotherapies depending on neoantigen expression by cancer cells. However, more studies are necessary and are under way in our laboratory. Citation Format: Heidge Fukumasu, Pedro R.L. Pires, Pedro L.P. Xavier. Inhibition of G9a reestablishes the MHC class I loss due to EMT in lung cancer 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 B011.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"78 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":"117245456","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}