A. Codd, S. Al-Taei, S. Tokita, E. Mizushima, P. Rizkallah, Thomas Whalley, Barbara Szomolay, K. Ladell, J. McLaren, Sian Llewellyn-Lacey, D. Price, T. Kanaseki, T. Torigoe, S. Man, Z. Tabi
{"title":"Abstract B007: Identification of prostate cancer stem cell antigens for T-cell immunotherapy by HLA ligandome analysis","authors":"A. Codd, S. Al-Taei, S. Tokita, E. Mizushima, P. Rizkallah, Thomas Whalley, Barbara Szomolay, K. Ladell, J. McLaren, Sian Llewellyn-Lacey, D. Price, T. Kanaseki, T. Torigoe, S. Man, Z. Tabi","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B007","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B007","url":null,"abstract":"Introduction: Localized prostate cancer (PCa) can be successfully treated by androgen deprivation, radiotherapy and surgery, however these may not be sufficient to eradicate cancer stem cells (CSCs). CSCs are more resistant to such treatments than the bulk of the tumor; and can contribute to disease relapse. PCa patients who relapse have a poor prognosis. We hypothesize that CSCs could be killed by T-cells in an antigen specific way, thus preventing the possibility of relapse. In this study we identified novel PCa CSC antigens by HLA ligandome analysis and isolated antigen specific CD8+ T-cells. Methods: We identified CSCs using aldehyde dehydrogenase (ALDH) activity as a CSC marker. ALDH high and low cells from the DU145 PCa cell line and from prostate adenocarcinoma primary tissue were characterized in vitro; DU145 CSCs and non-CSCs were additionally characterized in vivo. We isolated peptide-HLA complexes from DU145 cells by immunoprecipitation and analyzed the eluted peptides by mass spectrometry. We identified CSC antigens based on the gene expression in ALDH high and low DU145 cells (measured by qPCR). To select antigens for further analysis we performed homology modelling of the HLA-peptide interface using COOT software and the YASARA server for energy minimization. The interface interactions were quantified using PISA software. We additionally confirmed antigen expression in the primary cells by fluorescence microscopy and PCR. Tetramers were produced to isolate T-cells which recognized a selection of these antigens. Results: The ligandome analysis identified over 1900 peptides. We selected antigens with low gene expression in healthy tissues (www.GTexportal.org) and high predicted binding to DU145 HLA alleles (http://tools.immuneepitope.org/mhci/). ALDH high DU145 cells were more tumorigenic in vivo than ALDH low cells. ALDH high DU145 and primary prostate cancer cells grew larger colonies and spheres in vitro. We identified 11 CSC antigens by qPCR; 6 upregulated in ALDH high DU145 cells (e.g., TACSTD2) and 5 abundant in both ALDH high and ALDH low DU145 cells (e.g., XPO1). Relevant 9-mer epitopes from three antigens* induced CD8+ T-cell responses in vitro. Antigen-specific CD8+ T-cells were identified by tetramer staining at a frequency of approx. 15 per 100000 cells. These cells are currently being expanded to use in CTL assays. Conclusion: We have identified CSC antigens which could lead to specific targeting by T-cells and prevention of PCa relapse. Further epitopes restricted to the more frequent HLA alleles could additionally be predicted in silico from the novel antigens we identified. We are also investigating prediction of viral epitopes highly aligned with the self-peptides (determined in silico) to boost the immune response against CSCs. *Subject of an ongoing patent application. Citation Format: Amy S. Codd, Saly Al-Taei, Serina Tokita, Emi Mizushima, Pierre J. Rizkallah, Tom Whalley, Barbara Szomolay, Kristin Ladell, James E","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"03 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":"127189112","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 B009: New development of monoclonal antibodies targeting TRAIL agonist receptors","authors":"A. Dubuisson","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B009","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B009","url":null,"abstract":"Restoring programmed-cell death of cancer cells is a real challenge in oncology. To date, several monoclonal antibodies targeting TRAIL agonist death receptors have been developed, but none has been found to display significant clinical effects, so far. Efforts should be continued to develop novel DR4 and DR5 antibodies with improved properties. In this regard, novel monoclonal antibodies targeting DR4 and DR5 have been generated here using DNA-immunization in order to produce antibodies that can recognise DR4 or DR5 in their native forms. This cost- and time-effective technique allows the production of proteins of interest directly into the mice. Plasmids encoding DR4 or DR5 sequences have been injected into the tail veins of mice. Immunizations elicited significant humoral anti-DR4 and anti-DR5 responses and fusions of the corresponding spleens resulted in numerous hybridomas that can specifically recognise DR4 or DR5 in their native forms. After an intensive screening, anti-DR4 and anti-DR5 clones were selected for production and 4 to 5 were assessed for selectivity and affinity towards their respective targets by flow cytometry (FACS) and bio-layer interferometry (BLI). All antibodies efficiently bind to their targets with a very high affinity (nanomolar range). They were then characterized and tested for their capacity to induce apoptosis in a wide range of cancer cell lines. Of the four anti-DR4 antibodies of interest, one antibody displayed agonistic properties alone, one was able to inhibit and two were found to potentiate TRAIL-induced apoptosis. Similarly, one out of the four anti-DR5 antibodies behaved as an agonist whereas another one was found to enhance TRAIL. The most potent antibody was assessed in vivo, and preliminary results indicate that the latter is also able to inhibit tumor growth in animal models. In summary, this study is the first demonstration that DNA-based immunization method can be used to generate efficient therapeutic monoclonal antibodies targeting receptors of the TNF superfamily. Citation Format: Agathe Dubuisson. New development of monoclonal antibodies targeting TRAIL agonist receptors [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 B009.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"70 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":"127191615","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}
do Yofe, Hanjie Li, A. Leun, Yaniv Lubling, Dikla Gelbard, A. C. Akkooi, A. Tanay, T. Schumacher, I. Amit
{"title":"Abstract B054: Single-cell analysis illuminates the gradients of immune cell functional states within human melanoma tumors, and facilitates characterization of tumor-reactive T-cells","authors":"do Yofe, Hanjie Li, A. Leun, Yaniv Lubling, Dikla Gelbard, A. C. Akkooi, A. Tanay, T. Schumacher, I. Amit","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B054","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B054","url":null,"abstract":"Checkpoint blockade therapies that aim to reactivate antitumor immune responses have revolutionized cancer treatment, resulting in durable responses in a significant proportion of patients with advanced tumor progression. Nevertheless, many patients fail to reach long-term clinical benefit due to lack of response or acquired resistance. Inconsistency in therapy outcomes may be explained in part by recent findings suggesting that immune cell infiltrates in tumors are highly heterogeneous among patients. Therefore, comprehensive characterization of the diverse functional states exhibited by immune cell infiltrates is critical for the development of more effective immunotherapies. Here, we characterized immune cell infiltrates within tumors derived from 28 metastatic melanoma patients by single-cell RNA-seq of ~100,000 immune cells, and parallel T-cell receptor (TCR) sequencing, thereby generating an unbiased map of the expression signatures of immune cells, as well as clonality of T-cells within and between metastases. We identified within the tumor infiltrates naive, semi- and fully-activated effector, dysfunctional, and regulatory T-cells, as well as NK cells, and various myeloid subsets. While various immune cell types and cellular states are shared among patients, their frequency in each is highly heterogeneous even among similar tumor progression stages and treatment background. We noticed that clonally expanded T-cells predominantly adapted similar expression profiles. The frequencies of certain sub-populations were found to be correlated; notably, dysfunctional CD8 T-cell states were associated with prevalence of regulatory T-cells and follicular helper cells. The high-resolution map demonstrated gradient transitions between activation and dysfunctional states of CD8 T-cells, as well as variability within regulatory T-cell states. We used computational modeling to find the key transcription factors driving these gradients of expression states. Our analysis also puts forward novel candidate genes, which are highly correlate with known checkpoint targets within specific single-cell clusters, and may prove to be effective targets for checkpoint blockade. In order to validate our results, we performed in vitro reactivity assays (for available samples), which allowed us to determine autologous-tumor reactive and nonreactive TCRs, adding the reactive potential of T-cells as another layer of information to the single-cell expression signatures, facilitating inference of the functionality of clonally expanded T-cell populations in the tumor. Linking autologous tumor reactivity with single-cell expression profiles, we found that T-cells presenting reactive capability in vitro show a highly dysfunctional signature in the original tumor. The lack or presence of such reactive CD8 T-cells was also associated with patient response to immunotherapy (n=10). In conclusion, we present an atlas of immune cell infiltrates of human melanoma, revealing intra- and","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":"121625968","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}
Li Li, Xin Zhang, G. Maresh, L. Hellmers, Avi Patel, Ravan Moret, Sarah L. Cohen, D. Margolin
{"title":"Abstract B027: Antitumor effects of the programmed death receptor-1 and the programmed death-ligand 1 blockade in human colorectal carcinoma in a humanized orthotopic mouse model","authors":"Li Li, Xin Zhang, G. Maresh, L. Hellmers, Avi Patel, Ravan Moret, Sarah L. Cohen, D. Margolin","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B027","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B027","url":null,"abstract":"Background: The immune system plays an important role in tumor immune surveillance and progression. Immune checkpoint blockade is a new approach for cancer immunotherapy. The programmed death receptor-1 (PD-1) and the programmed death-ligand 1 (PD-L1) are immune checkpoint molecules and their expression results in negative regulation of T-cells primarily within the tumor microenvironment by preventing the killing of cancer cells by cytotoxic T-lymphocytes. Antibody blocking of the PD-1/PD-L1 signal exhibits promising therapeutic effects in non-small cell lung cancer and melanoma in patients. Due to its early success, more trials have been conducted to evaluate their efficacy for different tumors. We have developed a patient-derived orthotopic xenograft mouse model for colorectal carcinoma (CRC). Here, we investigate the potential efficacy of PD-1/PD-L1 blockade in our humanized orthotopic mouse models for human CRC. Methods: All studies were conducted under approved guidelines of the Institutional Animal Care and Use Committee and the Investigative Review Board of Ochsner Clinic Foundation. Humanized mice were established by intraperitoneal injection of donor human peripheral blood mononuclear cells (PBMCs) into recombinase activating gene 2 (Rag2) and common cytokine receptor gamma chain gene (IL2Rγ) double knockout (Rag2-/-/IL2Rγ-/-) Rag2 mice. Luciferase-tagged CRC cells were injected intra-rectally into Rag2 mice. One group of mice received a combination of anti-PD-1 and anti-PD-L1 antibodies (nivolumab, 200 µg/mouse and atezolizumab, 200 µg/mouse, intravenous injection) once a week for 3 weeks. Tumor growth was measured weekly by bioluminescent imaging (BLI). At necropsy, the CRC tumor weights were measured. Human CD45+ hematopoietic cells, CD4+ and CD8+ T-cells, and CD20+ B cells were detected in peritoneal lavage, blood, and tumor by flow cytometry. The presence of human immune cells (humanization) and tumor-infiltrating human lymphocytes was further confirmed by immunohistochemistry staining on paraffin-embedded tissue slides of mouse spleen and tumor, respectively. Results: Blood from mice receiving human PBMCs contained on average 31.4% CD45+ human cells (n=9) when tested by FACS analysis. Smaller tumor growth was observed in mice given PBMCs. This may be due to alloreactivity based on the recognition of MHC alloantigens in the transplanted tumor cells. However, mice further treated with the combination therapy of anti-PD-1 and anti-PD-L1 antibodies exhibited better antitumor response as well as less development of lung metastases compared to untreated controls. On average, tumor weight was reduced by 36% and lung metastases measured by ex-vivo BLI was reduced by 82% (n=5). In addition, circulating CD326+ tumor cells were reduced by 21% and CD3, CD4, CD8 and CD20 positive human lymphocytes were also reduced in the combination treatment group. Conclusion: Our study provides preclinical evidence of establishment of humanized Rag2 mice to ","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"15 40","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"134506145","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}
S. Haley, Charlotte Halgreen, K. Frederiksen, R. Brogaard, L. Brix
{"title":"Abstract B014: T-cell Elispot Proficiency Panel 2017/2018: Evaluating routine T-cell Elispot assays","authors":"S. Haley, Charlotte Halgreen, K. Frederiksen, R. Brogaard, L. Brix","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B014","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B014","url":null,"abstract":"Monitoring antigen-specific T-cell responses is becoming increasingly important in Immunotherapeutic research and development. Thanks to the harmonization efforts by the CIC and CIMT over many years, and the development of better reagents and protocols, MHC multimer and T-cell Elispot assays are now more reliable and accurate assays for monitoring antigen-specific T-cell immunity. Supported by CIMT and CIC, Immudex has conducted a T-cell Elispot proficiency panel Winter 2017/2018 to evaluate assay performance in immune monitoring laboratories routinely doing T-cell Elispot assays. 39 worldwide participants received two cell samples, together representing low, medium and high responses for two predefined peptide pools. A negative control reagent was also included. Participants were asked to use the Direct Human IFNγ Elispot Assay to determine spot count corresponding to each of the predefined peptide pools and the negative control reagent. After performing the Direct Human IFNγ Elispot Assay, the participants report back their results for each cell sample as “number of spots per 200.000 viable cells” for each of the predefined peptide pools and negative control reagent. The data set was analyzed, and each participating laboratory received a report detailing the individual laboratory’s performance (in an anonymized format). The anonymized report was publicly available April 2018, and data will be presented at the CRI-CIMT-EATI-AACR Fourth International Cancer Immunotherapy Conference. Citation Format: Stephen T. Haley, Charlotte Halgreen, Katrine Frederiksen, Rikke Brogaard, Liselotte Brix. T-cell Elispot Proficiency Panel 2017/2018: Evaluating routine T-cell Elispot assays [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 B014.","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":"117086412","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}
Siranush Sarkizova, Susan Klaeger, D. Keskin, K. Clauser, Hasmik Keshishian, Christina R. Hartigan, N. Hacohen, S. Carr, Catherine J. Wu
{"title":"Abstract B042: Broad analysis and more accurate predictions of HLA class I epitope binding in 92 common HLA alleles profiled by mono-allelic mass spectrometry","authors":"Siranush Sarkizova, Susan Klaeger, D. Keskin, K. Clauser, Hasmik Keshishian, Christina R. Hartigan, N. Hacohen, S. Carr, Catherine J. Wu","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B042","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B042","url":null,"abstract":"Introduction: Cancer vaccine therapies rely on accurate personalized selection of immunizing peptides in order to potentiate tumor-specific immune responses against neoepitopes derived from somatic mutations. Given the unique accumulation of mutations in each tumor as well as the patient’s particular complement of HLA class I alleles, the ability to accurately predict which epitopes will be presented by tumor cells is a fundamental prerequisite for successful vaccine design. By utilizing a mono-allelic mass spectrometry (MS) strategy for profiling the endogenous HLA class I peptidome, we recently showed that prediction of endogenous presentation can be drastically improved when model training integrates peptide sequence along with intracellular signals such as likelihood of proteasomal processing and peptide abundance. Yet the limited set of mono-allelic data did not allow for deep comparative analysis across HLA- A, B, and C alleles, which can better inform pan-allele predictor design. Moreover, the significant variability in per-allele model performance remains unexplained. Methods: We recently developed a scalable mono-allelic MS technique to profile naturally presented peptides on HLA molecules, whereby the HLA class I deficient B721.221 cell line is transfected with HLA expression vectors coding for a single allele of interest and eluted HLA peptides are analyzed by LC-MS/MS. In addition, endogenously presented antigens on primary tumor-derived cell lines from 4 melanoma patients were also identified via MS. To extract knowledge from this unique dataset, we implemented computational tools to summarize, visualize, and compare the characteristics of HLA- A, B, C, and G alleles and developed a novel approach to define allele similarity that takes into account the collection of sub-motifs per allele. We trained neural network prediction models, validated their performance on internal and external datasets, and analyzed the variability in performance across alleles. Results: To date, we have generated binding data for 92 HLA- A, B, C and G alleles, identifying more than 190,000 peptides and covering the most frequent alleles in the population. Extensive mono-allelic profiling revealed that some alleles present non-9-mer peptides with high frequency. The availability of large number of non-9-mer peptides allowed us to build length-specific models that often performed better than the corresponding non-length-specific models currently used. We observe that HLA- A and B alleles present more peptides of length 10 and 11 than C alleles, while C alleles have a higher propensity for 8-mers. Correlation-based analysis of binding motifs revealed that HLA-A and B motifs are more specific whereas C motifs are less stringent and thus share more overlapping binders. Since binding data are available only for a fraction of all known alleles, pan-allele models implicitly embed allele similarity to predict for uncharacterized alleles based on the sequence of the b","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer 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":"126462460","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 B017: Multifunctional immunomodulator capable of hypoxia-sensitive adjuvant delivery and photodynamical assistance for DC antigen presentation for cancer immunotherapy","authors":"Sooseok Im, W. Kim","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B017","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B017","url":null,"abstract":"Nanoparticle-based delivery system has been attempted for a couple of decades for cancer immunotherapy (CIT) to modulate immune responses and reduce off-target side effect. For the successful CIT, the extent of antigen presentation by dendritic cell (DC) that phagocytizes tumor associated antigen (TAA) in tumor site and migrates to tumor draining lymph node (TDLN) for an activation of T-cells. For this, recent studies have been trying to make tumor tissue releases TAAs directly, increasing the chance for antigen presenting cells (APCs) to encounter the neoantigens. Photodynamic therapy (PDT), a conventional cancer therapeutic manner that generates reactive oxygen species (ROS) from adjacent oxygen by photosensitizer (PS) under irradiation of light, could induce release of damage-associated molecular patterns (DAMPs) and promote activities of immune cells, along with release of TAAs. Furthermore, PDT mediated generation of ROS is well known as neutrophil chemotaxis, potentiating capability for DC recruitment via granule enzymatic processing the prochemerin into chemerin. Taken together, a PDT can induce both tumor cell death and recruiting DCs, followed by uptake of the as-generated cell debris by APCs which increases the antigen presentation process and further activation of antitumor immune responses. Herein, to introduce PDT-induced immunologic alterations into CIT, we devised the nanoparticle-based delivery system that fulfills a few key requirements: 1) internalized efficiently by the cells at the target site, while the cargo is protected from degradation and scavenging by macrophages during circulation; 2) induces the release of tumor proteins and deliver adjuvant to activate APCs for prolonged immune reaction. In this study, we have developed a MSN-based hypoxia-responsive PS/adjuvant nanocomplex, denoted as CAGE, to enable photodynamic therapy assisted CIT. The surface of chlorin e6 (Ce6)-doped mesoporous silica nanoparticle was decorated with glycol chitosan (GC) and PEG via azobenzene linker, a hypoxia-responsive labile linker. CpG, a short oligonucleotide immunomodulator known to activate the DCs, was loaded onto the surface of CAGE by electrostatic interaction with GC. It was designed that azobenzene linker could be cleaved under intrinsic tumor hypoxia as well as abrupt consumption of local oxygen induced by photodynamic effect, leading to both a detachment of PEG for the tumor specific retention of MSNs and release of CpG/GC complexes. Due to the photodynamic effect of PS and delivery of CpG, the population of tumor infiltrating DCs and its maturation ratio were significantly elevated. An improved activity of DCs, combined with generation of tumor debris by photodynamic effect, was synergistically resulted in increase of antigen presentation of DCs, exhibiting remarkable inhibition of tumor growth in vivo. Citation Format: Sooseok Im, Won Jong Kim. Multifunctional immunomodulator capable of hypoxia-sensitive adjuvant delivery and pho","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"103 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":"124575908","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}
A. Sasikala, A. R. Unnithan, C. Park, Cheol-Sang Kim
{"title":"Abstract B041: An injectable magnetic nanogel system for filling surgical residual cavity with effective cancer immunotherapy combined hyperthermic capability","authors":"A. Sasikala, A. R. Unnithan, C. Park, Cheol-Sang Kim","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B041","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B041","url":null,"abstract":"The advancement in nanotechnology has created a wealth of new possibilities for treating cancer with multifunctional nanosystem holding various therapeutic strategies in a single platform. It has been reported that magnetic nanoparticle hyperthermia can induce antitumor immunity whereas the immunotherapy can naturally trigger the immune system with the help of an appropriate stimulator to control cancer. Therefore the present study investigates the effectiveness of a hybrid nanocomposite system to effectively exterminating the tumor associated immune cells (TICs) as well as inducing an inflammatory immune response by activating killer T-cells by combining magnetic hyperthermia with an immunostimulatory agent. Here we report an injectable magnetic nanogels conjugating a checkpoint inhibitor (T-lymphocyte antigen-4 [CTLA4]) and a Toll-like receptor (TLR) agonists (imiquimod) for generating an effective antitumoral immune response for postsurgical glioma treatment. The injectable conductive magnetic hydrogel system enables the tracing and deterring of the recurrent tumor cells via magnetic nanoparticle-mediated hyperthermia with the strong immunologic memory effect. The superparamagnetic iron oxide nanoparticles encapsulated nanogels exhibit an induced heating ability in an alternating magnetic field (AMF) and thereby trigger the tumor cells to undergo a burst release of heat shock proteins to recruit immune cells to generate tumor-allied antigens. These antigens along with the released imiquimod from the hydrogel can generate vaccine-like functionalities. Moreover, the introduction of anti-cytotoxic T-lymphocyte antigen-4 (CTLA4) as a checkpoint-blockade will generate memory T-cells, which in turn will attack the metastatic and recurrent tumor cells. Above all, the conductive bioactive scaffold will support the neuronal regeneration and reactivation of brain cells. Thus our multifunctional novel hydrogel system will be able to fill the surgical residual cavity to prevent the glioma recurrence and will improve the local neuronal tissue reconstruction along with hyperthermic-immunotherapy. Citation Format: Arathyram Ramachandra Kururp Sasikala, Afeesh Rajan Unnithan, Chan Hee Park, Cheol Sang Kim. An injectable magnetic nanogel system for filling surgical residual cavity with effective cancer immunotherapy combined hyperthermic capability [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 B041.","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"16 1","pages":"0"},"PeriodicalIF":0.0,"publicationDate":"2019-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"121425966","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}
S. Church, Chris Merritt, Giang T Ong, A. White, Kristi Zevin, S. Warren, J. Beechem
{"title":"Abstract B006: Gridded tissue profiling strategy with digital spatial profiling for unbiased tissue sampling","authors":"S. Church, Chris Merritt, Giang T Ong, A. White, Kristi Zevin, S. Warren, J. Beechem","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B006","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B006","url":null,"abstract":"Characterization of the spatial distribution and abundance of protein and RNAs within a tissue enables deep understanding of biologic systems. The ability to interrogate multiple targets simultaneously from clinical samples enhances the potential for discovery, but has proven to be challenging for formalin fixed paraffin embedded (FFPE) tissue. NanoString Technologies® has developed the Digital Spatial Profiling (DSP) platform to enable highly multiplexed profiling of protein or RNA from FFPE tissue using a non-destructive protocol. This technology can be used to quantify the abundance of targets within user-defined regions of interest (ROI) using a variety of masking strategies to select and define those regions, including geometric, phenotypic, or automatically generated masks based on expression of certain markers within the tissue. The current technology is capable of profiling dozens of targets, and future iterations could enable hundreds or thousands of targets to be profiled simultaneously. Controlling ROI size, shape, and features defines the heterogeneity and granularity of the information generated. To date, studies on the DSP have largely focused on profiling regions of interest in a nonsystematic way. However, profiling a tissue in a gridded fashion enables a uniform sampling of protein expression either across a whole tissue section or within a smaller area of the tissue to enable nonbiased evaluation, which may aid in biomarker discovery. This study utilizes DSP and a gridded ROI selection strategy to profile protein distribution within normal and tumor tissue. FFPE tissue from tonsil or colorectal tumors are stained with a 44-plex cocktail of antibodies conjugated to unique DNA oligos via a photocleavable linker and up to 3 fluorescent antibodies. Visual images of the tissue are collected with the fluorescent antibodies, and selected regions of interest are subsequently illuminated with UV light to release the oligos for collection. ROIs are profiled serially and captured oligos are then quantitated in the standard NanoString assay. In this study, tonsil and colorectal cancer (CRC) tissue is profiled on the DSP platform using a gridded ROI selection strategy to collect protein profiles from an entire tissue section either via low-resolution sampling or from specific regions within the tissue via high-resolution sampling. Low-resolution sampling of the tissue provides an estimate of the protein distribution across a section of tonsil or CRC. It further enables identification of regions of interest within the tumor suitable for deeper profiling with the high-resolution approach. The higher resolution profiling enables reconstruction of the tissue morphology at the molecular level by looking solely at the distribution of proteins. A distinct protein profiles for the germinal centers and T-cell zones within lymph nodes. Furthermore, we see variations emerges of protein expression between different lymph nodes that may contribute to dif","PeriodicalId":352838,"journal":{"name":"Convergence of Technology and Cancer Immunotherapy","volume":"155 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":"115777679","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}
Miguel C. Sobral, Hua Wang, Alexander J. Najibi, Aileen W. Li, Catia Verbeke, D. Mooney
{"title":"Abstract B045: Antigen-free cancer vaccine to treat poorly immunogenic tumors","authors":"Miguel C. Sobral, Hua Wang, Alexander J. Najibi, Aileen W. Li, Catia Verbeke, D. Mooney","doi":"10.1158/2326-6074.CRICIMTEATIAACR18-B045","DOIUrl":"https://doi.org/10.1158/2326-6074.CRICIMTEATIAACR18-B045","url":null,"abstract":"Certain chemotherapeutic drugs can elicit immunogenic death of tumor cells and enhance antitumor immune responses. Here we explore whether immunogenic chemotherapy can be utilized for the development of antigen-free cancer vaccines, by combining it with peritumorally injected biomaterial scaffolds that recruit dendritic cells (DCs) for subsequent antigen presentation and T-cell priming. Pore-forming alginate gels containing granulocyte-macrophage colony-stimulating factor (GM-CSF) and a doxorubicin-iRGD conjugate were found to efficiently induce the apoptosis of 4T1 triple-negative breast cancer cells in vivo, while recruiting large numbers of DCs. The co-encapsulation of CpG oligodeoxynucleotides in the gel significantly enhanced the immunogenic death of 4T1 cells, increased systemic tumor-specific CD8+ T-cells and tumoral infiltration of CD8+ T-cells, repolarized tumor-associated macrophages towards an inflammatory M1-like phenotype, and resulted in significantly improved antitumor efficacy. This in situ antigen-free gel vaccine shows promise for the treatment of poorly immunogenic tumors, and more broadly, may serve as a facile platform to enable in situ personalized cancer vaccination without requiring identification of tumor-specific antigens and manufacturing of personalized vaccines. Citation Format: Miguel C. Sobral, Hua Wang, Alexander J. Najibi, Aileen Li, Catia S. Verbeke, David J. Mooney. Antigen-free cancer vaccine to treat poorly immunogenic 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 B045.","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":"127155246","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}