62 research outputs found

    MiR-17-92 Cluster Regulation in Differentiated T-cells

    Get PDF
    Data from our group and others have demonstrated that tumor-derived factors directly skew T-cell differentiation from an effective tumor fighting Th1 state to a less effective Th2 state, allowing for tumor growth. Why the Th1 response is more effective is largely still unknown. The recently discovered microRNAs (miRNAs) are a large family of small regulatory RNAs that control diverse aspects of cell functions such as cell proliferation, apoptosis, development, differentiation and immune regulation. We thereby sought to examine miRNAs differentially expressed in Th1 and Th2 cells in an effort to better understand the enhanced ability of Th1 cells in tumor immunity. MicroRNA microarray analyses revealed that the miR-17-92 cluster of microRNAs (miR-17-92) is consistently over-expressed in murine Th1 cells compared to Th2 cells. Quantitative RT-PCR confirmed that the miR-17-92 cluster expression was consistently higher in Th1 cells than Th2 cells. Furthermore, disruption of IL-4 signaling through either IL-4 neutralizing antibody or knockout of STAT6 reversed the miR-17-92 cluster suppression in Th2 cells. MiR-17-92 expression correlated with differential proliferation capacity as Th1 cells proliferated at higher levels than Th2 cells, dependent on IL-4 and STAT6. Th1 cells consistently expressed lower levels of anti-proliferative transcription factors E2F1 and E2F2, which are the known targets of miR-17-92. Collectively, our data suggests that the Th2 skewing tumor microenvironment can induce the down-regulation of miR-17-92 expression in CD4+T cells, thereby diminishing the effective proliferation of tumor-specific T cells and tumor destruction. This has significant public health relevance as we propose that therapy targeting miR-17-92 cluster may provide enhanced T-cell function and prevent tumor growth

    Novel mechanisms of cytokine signaling on T-cell and MDSC function in glioma development

    Get PDF
    Malignant gliomas are the most common primary brain tumors with dismal prognosis. A growing line of evidence supports significant roles of immunosurveillance for prevention and regulation of cancer development. For example, tumor infiltrating T-cells are capable of killing tumor cells and are a positive prognostic factor for cancer patients. T-cell immune responses are classified into distinct effector cell types, type-1 or type-2, based on their cytokine-secreting profiles. We have demonstrated that tumor-specific type-1 T-cells, but not type-2 T-cells, can efficiently traffic into CNS tumor sites and mediate effective therapeutic efficacy via a type-1 chemokine CXCL10 and an integrin receptor VLA-4. Despite the importance of the type-1 T cell response, cancers, including GBMs, secrete numerous type-2 cytokines that promote tumor proliferation and immune escape. The hallmark cytokines of type-1 and type-2 immune responses are IFNs and IL-4, respectively. We therefore sought to better understand the role of IL-4 and IFN signaling in gliomas. We herein demonstrate that the miR-17-92 cluster is down-regulated in T-cells in both human and mouse tumors, dependent on IL-4R signaling. Further, ectopic expression of miR-17-92 cluster in T-cells resulted in enhanced IFN-γ and IL-2 production and resistance to activation induced cell death (AICD) (Aim 1). We next examined IL-4Rα on immunosuppressive myeloid derived suppressor cells (MDSCs). Interestingly we found that IL-4Rα was up-regulated on human and mouse glioma infiltrating, but not peripheral, MDSCs. Additionally, IL-4Rα expression promoted arginase activity, T-cell suppressing abilities and glioma growth (Aim 2). As type I IFNs are important for anti-glioma type-1 immunity, we further examined how type I IFNs impact glioma patient prognosis. As there are multiple type I IFNs, our collaborators assisted us to identify potentially important genes by single nucleotide polymorphism (SNP) analysis. We found that IFN-pathway genes IFN- alpha receptor-1 (IFNAR1) and the IFN-alpha-8 (IFNA8) promoter both had SNPs associated with glioma prognosis. By luciferase assay and electrophoretic mobility shift assay (EMSA) we demonstrated that the A-allele, which is associated with better glioma patient survival, but not the C-allele of rs12553612 in the promoter region of IFNA8 allows for OCT-1 binding and activity of the IFNA8 promoter (Aim 3). Overall, our data suggests that type-2 promoting has a dual role in suppressing glioma immunity through decreased T-cell functioning and enhanced MDSC function. Type-2 promoted suppression of glioma immunity can thus lead to better glioma patient prognosis, a significant public health achievement

    ONO-AE3-208 inhibits myeloid derived suppressor cells and glioma growth

    Get PDF
    Myeloid Derived Suppressor Cells (MDSCs) heavily infiltrate in a variety of solid tumors and suppress anti-tumor T-cell activity. Our recent studies have demonstrated the ability of monocytic, Ly6C+ MDSCs to promote glioma growth through the activation of cyclooxygenase (COX)-2 pathway, which is responsible for prostaglandin-synthesis. ONO-AE3-208 is an antagonist of the prostaglandin E (EP)-4 receptor, which is an important positive feedback regulator of the COX-2 pathway. We thus examined the ability of ONO-AE3-208 to suppress MDSC activity in gliomas. ONO-AE3-208 treatment in mice bearing established GL261-quad glioma in the brain resulted in complete and persistent rejection of the tumors. Flow cytometric analysis revealed that gliomas in the ONO-AE3-208-treated mice were infiltrated by fewer numbers of Ly6C+ MDSCs compared with non-treated animals. We subsequently isolated glioma-infiltrating Ly6C+ MDSCs by flow-sorting to address their functions. RT-PCR analysis revealed that the Ly6C+ MDSCs derived from ONO-AE3-208 treated mice expressed lower levels of the Arg1 and Cox2 expression compared to control animals. Consistently, brain infiltrating leukocytes in ONO-AE3-208 treated tumor-bearing mice demonstrated enhanced IFN-g expression compared with control mice, suggestive of enhanced T-cell activity. Importantly, ONO-AE3-208 inhibited glioma growth and promoted immune activity in 2 additional murine glioma models: the Sleeping Beauty de novo glioma model and the SB28 glioma cell line model. Our data demonstrate that ONO-AE3-208 may be useful in the treatment of glioma patients to suppress Ly6C+ MDSCs and promote anti-tumor immunity

    Differential activity of interferon-α8 promoter is regulated by Oct-1 and a SNP that dictates prognosis of glioma

    Get PDF
    We have previously reported that the single nucleotide polymorphism (SNP) rs12553612 in IFNA8 is associated with better overall survival of glioma patients with the AA-genotype compared with patients with the AC-genotype. As rs12553612 is located in the IFNA8 promoter, we hypothesized that the A-allele allows for an enhanced IFNA8 promoter activity compared with the C-allele. Reporter assays in the human monocyte derived THP-1 cell line demonstrated a superior promoter activity of the A-allele compared with the C-allele. Electrophoretic mobility shift assays (EMSA) further demonstrated that the A-genotype specifically binds to more nuclear proteins than the C-genotype, including the transcription factor Oct-1. Further, co-transfection of plasmids encoding Oct-1 and the reporter constructs revealed that Oct-1 enhanced the promoter activity with the A- but not the C-allele. Taken together, our data demonstrate that the A-allele in the rs12553612 SNP, which is associated with better glioma patient survival, allows for IFNA8 transcription by allowing for Oct-1 binding, which is absent in patients with C allele, and suggests a molecular mechanism of IFNA8 mediated immune-surveillance of glioma progression

    IDH mutation-induced suppression of type-1 anti-glioma immune response

    Get PDF
    Isocitrate dehydrogenase (IDH) mutations are the first mutations that occur during the oncogenic process of lower-grade glioma (LGG) and confers a novel gain-of-function activity by converting α-ketoglutarate (αKG) to 2-hydroxyglutarate (2HG), promoting DNA hyper-methylation. Our analysis of LGG cases from The Cancer Genome Atlas (TCGA) database revealed that IDH-mutant (IDH-Mut) cases exhibit decreased expression of type-1 effector T cell response-related genes, which are critical for anti-glioma immunity, including: CD8A, IFNG, OAS2, GZMA, EOMES, CXCL9 and CXCL10, compared with IDH-wild type (IDH-WT) cases. On the other hand, type-2 and regulatory T cell response-related genes, such as IL5 and TGFB1, are not significantly different between IDH-Mut vs. WT cases, indicating that the observed down-regulation of type-1 response-related genes does not merely represent a possible global gene suppression. Furthermore, IDH-Mut cases exhibit increased CXCL10 promotor methylation compared with WT cases. We thus hypothesized that IDH mutation-mediated tumor intrinsic mechanisms occurring within glioma cells may inhibit anti-tumor immunity to promote tumor growth. In vitro, a normal human astrocyte (NHA) cell line transfected with IDH1-Mut cDNA expressed lower levels of CXCL10 compared to NHA cells transfected with WT IDH1. Consistently, C57Bl/6 mouse-syngeneic astrocyte and glioma cell lines transfected with IDH1-Mut expressed lower levels of CXCL10 gene and protein, compared to control cells transfected with IDH-WT, which was restored following 30 day treatment of the cells with the IDH1 inhibitor, IDH-C35. Furthermore, in vivo orthotopic IDH1-Mut gliomas at 21 days post-intracranial injection in syngeneic mice expressed lower levels of T cell chemokines CXCL9 and CXCL10 as determined by RT-PCR and ELISA and reduced infiltration of CD3+CD8+ T cells as determined by flow cytometry and quantitative immunohistochemistry compared with control IDH1-WT gliomas. Further, an in vitro migration assay demonstrated reduced migration of T cells towards culture supernatants from IDH1-Mut cell lines compared with control supernatants derived from IDH1-WT cells. Overall, our data demonstrate that IDH mutations in tumor cells lead to reduced T cell attracting chemokines and reduced T cell accumulation in gliomas. Our analyses of the TCGA 450K gene methylation database suggest that the suppressed expression of OAS2 and CXCL10 in IDH1-Mut cases is associated with hypermethylation of the promoter for these genes. Indeed, treatment of IDH-Mut cell lines with demethylating agent 5-Aza-CdR restored CXCL10 expression levels. Our data suggest that IDH inhibitors and demethylation agents may be used to enhance T cell recruitment to LGG in combination with T cell based immunotherapies

    miR-17-92 expression in differentiated T cells - implications for cancer immunotherapy

    Get PDF
    <p>Abstract</p> <p>Background</p> <p>Type-1 T cells are critical for effective anti-tumor immune responses. The recently discovered microRNAs (miRs) are a large family of small regulatory RNAs that control diverse aspects of cell function, including immune regulation. We identified miRs differentially regulated between type-1 and type-2 T cells, and determined how the expression of such miRs is regulated.</p> <p>Methods</p> <p>We performed miR microarray analyses on <it>in vitro </it>differentiated murine T helper type-1 (Th1) and T helper type-2 (Th2) cells to identify differentially expressed miRs. We used quantitative RT-PCR to confirm the differential expression levels. We also used WST-1, ELISA, and flow cytometry to evaluate the survival, function and phenotype of cells, respectively. We employed mice transgenic for the identified miRs to determine the biological impact of miR-17-92 expression in T cells.</p> <p>Results</p> <p>Our initial miR microarray analyses revealed that the miR-17-92 cluster is one of the most significantly over-expressed miR in murine Th1 cells when compared with Th2 cells. RT-PCR confirmed that the miR-17-92 cluster expression was consistently higher in Th1 cells than Th2 cells. Disruption of the IL-4 signaling through either IL-4 neutralizing antibody or knockout of signal transducer and activator of transcription (STAT)6 reversed the miR-17-92 cluster suppression in Th2 cells. Furthermore, T cells from tumor bearing mice and glioma patients had decreased levels of miR-17-92 when compared with cells from non-tumor bearing counterparts. CD4<sup>+ </sup>T cells derived from miR-17-92 transgenic mice demonstrated superior type-1 phenotype with increased IFN-γ production and very late antigen (VLA)-4 expression when compared with counterparts derived from wild type mice. Human Jurkat T cells ectopically expressing increased levels of miR-17-92 cluster members demonstrated increased IL-2 production and resistance to activation-induced cell death (AICD).</p> <p>Conclusion</p> <p>The type-2-skewing tumor microenvironment induces the down-regulation of miR-17-92 expression in T cells, thereby diminishing the persistence of tumor-specific T cells and tumor control. Genetic engineering of T cells to express miR-17-92 may represent a promising approach for cancer immunotherapy.</p

    Oncolytic HSV Vectors and Anti-Tumor Immunity

    Get PDF
    The therapeutic promise of oncolytic viruses (OVs) rests on their ability to both selectively kill tumor cells and induce anti-tumor immunity. The potential of tumors to be recognized and eliminated by an effective anti-tumor immune response has been spurred on by the discovery that immune checkpoint inhibition can overcome tumor-specific cytotoxic T cell (CTL) exhaustion and provide durable responses in multiple tumor indications. OV-mediated tumor destruction is now recognized as a powerful means to assist in the development of anti-tumor immunity for two important reasons: (i) OVs, through the elicitation of an anti-viral response and the production of type I interferon, are potent stimulators of inflammation and can be armed with transgenes to further enhance anti-tumor immune responses; and (ii) lytic activity can promote the release of tumor-associated antigens (TAAs) and tumor neoantigens that function as in situ tumor-specific vaccines to elicit adaptive immunity. Oncolytic herpes simplex viruses (oHSVs) are among the most widely studied OVs for the treatment of solid malignancies, and Amgen's oHSV Imlygic® for the treatment of melanoma is the only OV approved in major markets. Here we describe important biological features of HSV that make it an attractive OV, clinical experience with HSV-based vectors, and strategies to increase applicability to cancer treatment

    Large expert-curated database for benchmarking document similarity detection in biomedical literature search

    Get PDF
    Document recommendation systems for locating relevant literature have mostly relied on methods developed a decade ago. This is largely due to the lack of a large offline gold-standard benchmark of relevant documents that cover a variety of research fields such that newly developed literature search techniques can be compared, improved and translated into practice. To overcome this bottleneck, we have established the RElevant LIterature SearcH consortium consisting of more than 1500 scientists from 84 countries, who have collectively annotated the relevance of over 180 000 PubMed-listed articles with regard to their respective seed (input) article/s. The majority of annotations were contributed by highly experienced, original authors of the seed articles. The collected data cover 76% of all unique PubMed Medical Subject Headings descriptors. No systematic biases were observed across different experience levels, research fields or time spent on annotations. More importantly, annotations of the same document pairs contributed by different scientists were highly concordant. We further show that the three representative baseline methods used to generate recommended articles for evaluation (Okapi Best Matching 25, Term Frequency-Inverse Document Frequency and PubMed Related Articles) had similar overall performances. Additionally, we found that these methods each tend to produce distinct collections of recommended articles, suggesting that a hybrid method may be required to completely capture all relevant articles. The established database server located at https://relishdb.ict.griffith.edu.au is freely available for the downloading of annotation data and the blind testing of new methods. We expect that this benchmark will be useful for stimulating the development of new powerful techniques for title and title/abstract-based search engines for relevant articles in biomedical research.Peer reviewe

    31st Annual Meeting and Associated Programs of the Society for Immunotherapy of Cancer (SITC 2016) : part two

    Get PDF
    Background The immunological escape of tumors represents one of the main ob- stacles to the treatment of malignancies. The blockade of PD-1 or CTLA-4 receptors represented a milestone in the history of immunotherapy. However, immune checkpoint inhibitors seem to be effective in specific cohorts of patients. It has been proposed that their efficacy relies on the presence of an immunological response. Thus, we hypothesized that disruption of the PD-L1/PD-1 axis would synergize with our oncolytic vaccine platform PeptiCRAd. Methods We used murine B16OVA in vivo tumor models and flow cytometry analysis to investigate the immunological background. Results First, we found that high-burden B16OVA tumors were refractory to combination immunotherapy. However, with a more aggressive schedule, tumors with a lower burden were more susceptible to the combination of PeptiCRAd and PD-L1 blockade. The therapy signifi- cantly increased the median survival of mice (Fig. 7). Interestingly, the reduced growth of contralaterally injected B16F10 cells sug- gested the presence of a long lasting immunological memory also against non-targeted antigens. Concerning the functional state of tumor infiltrating lymphocytes (TILs), we found that all the immune therapies would enhance the percentage of activated (PD-1pos TIM- 3neg) T lymphocytes and reduce the amount of exhausted (PD-1pos TIM-3pos) cells compared to placebo. As expected, we found that PeptiCRAd monotherapy could increase the number of antigen spe- cific CD8+ T cells compared to other treatments. However, only the combination with PD-L1 blockade could significantly increase the ra- tio between activated and exhausted pentamer positive cells (p= 0.0058), suggesting that by disrupting the PD-1/PD-L1 axis we could decrease the amount of dysfunctional antigen specific T cells. We ob- served that the anatomical location deeply influenced the state of CD4+ and CD8+ T lymphocytes. In fact, TIM-3 expression was in- creased by 2 fold on TILs compared to splenic and lymphoid T cells. In the CD8+ compartment, the expression of PD-1 on the surface seemed to be restricted to the tumor micro-environment, while CD4 + T cells had a high expression of PD-1 also in lymphoid organs. Interestingly, we found that the levels of PD-1 were significantly higher on CD8+ T cells than on CD4+ T cells into the tumor micro- environment (p < 0.0001). Conclusions In conclusion, we demonstrated that the efficacy of immune check- point inhibitors might be strongly enhanced by their combination with cancer vaccines. PeptiCRAd was able to increase the number of antigen-specific T cells and PD-L1 blockade prevented their exhaus- tion, resulting in long-lasting immunological memory and increased median survival
    corecore