17 research outputs found

    A Ten-microRNA Expression Signature Predicts Survival in Glioblastoma

    Get PDF
    Glioblastoma (GBM) is the most common and aggressive primary brain tumor with very poor patient median survival. To identify a microRNA (miRNA) expression signature that can predict GBM patient survival, we analyzed the miRNA expression data of GBM patients (n = 222) derived from The Cancer Genome Atlas (TCGA) dataset. We divided the patients randomly into training and testing sets with equal number in each group. We identified 10 significant miRNAs using Cox regression analysis on the training set and formulated a risk score based on the expression signature of these miRNAs that segregated the patients into high and low risk groups with significantly different survival times (hazard ratio [HR] = 2.4; 95% CI = 1.4–3.8; p<0.0001). Of these 10 miRNAs, 7 were found to be risky miRNAs and 3 were found to be protective. This signature was independently validated in the testing set (HR = 1.7; 95% CI = 1.1–2.8; p = 0.002). GBM patients with high risk scores had overall poor survival compared to the patients with low risk scores. Overall survival among the entire patient set was 35.0% at 2 years, 21.5% at 3 years, 18.5% at 4 years and 11.8% at 5 years in the low risk group, versus 11.0%, 5.5%, 0.0 and 0.0% respectively in the high risk group (HR = 2.0; 95% CI = 1.4–2.8; p<0.0001). Cox multivariate analysis with patient age as a covariate on the entire patient set identified risk score based on the 10 miRNA expression signature to be an independent predictor of patient survival (HR = 1.120; 95% CI = 1.04–1.20; p = 0.003). Thus we have identified a miRNA expression signature that can predict GBM patient survival. These findings may have implications in the understanding of gliomagenesis, development of targeted therapy and selection of high risk cancer patients for adjuvant therapy

    A DNA methylation prognostic signature of glioblastoma: identification of NPTX2-PTEN-NF-kappa B nexus

    No full text
    Glioblastoma (GBM) is the most common, malignant adult primary tumor with dismal patient survival, yet the molecular determinants of patient survival are poorly characterized. Global methylation profile of GBM samples (our cohort; n = 44) using high-resolution methylation microarrays was carried out. Cox regression analysis identified a 9-gene methylation signature that predicted survival in GBM patients. A risk-score derived from methylation signature predicted survival in univariate analysis in our and The Cancer Genome Atlas (TCGA) cohort. Multivariate analysis identified methylation risk score as an independent survival predictor in TCGA cohort. Methylation risk score stratified the patients into low-risk and high-risk groups with significant survival difference. Network analysis revealed an activated NF-kappa B pathway association with high-risk group. NF-kappa B inhibition reversed glioma chemoresistance, and RNA interference studies identified interleukin-6 and intercellular adhesion molecule-1 as key NF-kappa B targets in imparting chemoresistance. Promoter hypermethylation of neuronal pentraxin II (NPTX2), a risky methylated gene, was confirmed by bisulfite sequencing in GBMs. GBMs and glioma cell lines had low levels of NPTX2 transcripts, which could be reversed upon methylation inhibitor treatment. NPTX2 overexpression induced apoptosis, inhibited proliferation and anchorage-independent growth, and rendered glioma cells chemosensitive. Furthermore, NPTX2 repressed NF-kappa B activity by inhibiting AKT through a p53-PTEN-dependent pathway, thus explaining the hypermethylation and downregulation of NPTX2 in NF-kappa B-activated high-risk GBMs. Taken together, a 9-gene methylation signature was identified as an independent GBM prognosticator and could be used for GBM risk stratification. Prosurvival NF-kappa B pathway activation characterized high-risk patients with poor prognosis, indicating it to be a therapeutic target. (C) 2013 AACR

    A 16-Gene Signature Distinguishes Anaplastic Astrocytoma from Glioblastoma

    Get PDF
    <div><p>Anaplastic astrocytoma (AA; Grade III) and glioblastoma (GBM; Grade IV) are diffusely infiltrating tumors and are called malignant astrocytomas. The treatment regimen and prognosis are distinctly different between anaplastic astrocytoma and glioblastoma patients. Although histopathology based current grading system is well accepted and largely reproducible, intratumoral histologic variations often lead to difficulties in classification of malignant astrocytoma samples. In order to obtain a more robust molecular classifier, we analysed RT-qPCR expression data of 175 differentially regulated genes across astrocytoma using <i>P</i>rediction <i>A</i>nalysis of <i>M</i>icroarrays (PAM) and found the most discriminatory 16-gene expression signature for the classification of anaplastic astrocytoma and glioblastoma. The 16-gene signature obtained in the training set was validated in the test set with diagnostic accuracy of 89%. Additionally, validation of the 16-gene signature in <i>multiple</i> independent cohorts revealed that the signature predicted anaplastic astrocytoma and glioblastoma samples with accuracy rates of 99%, 88%, and 92% in TCGA, GSE1993 and GSE4422 datasets, respectively. The protein-protein interaction network and pathway analysis suggested that the 16-genes of the signature identified epithelial-mesenchymal transition (EMT) pathway as the most differentially regulated pathway in glioblastoma compared to anaplastic astrocytoma. In addition to identifying 16 gene classification signature, we also demonstrated that genes involved in epithelial-mesenchymal transition may play an important role in distinguishing glioblastoma from anaplastic astrocytoma.</p></div

    KEGG pathway analysis showed the upregulation of Focal adhesion pathway in GBM.

    No full text
    <p>67 differentially expressed genes between AA and GBM from GSE1993 was subjected to network analysis in Pathway Express which identified the focal adhesion as the significantly differentially regulated pathway between AA and GBM. The input genes present in the network are represented in red and blue indicating the upregulation or downregulation respectively in GBM as compared to AA.</p

    PCA and cross validated probabilities of AA and GBM samples of training set.

    No full text
    <p><b>A.</b> PCA was performed using expression values of 16- PAM identified genes between AA (n = 30) and GBM (n = 78) samples in training set. A scatter plot is generated using the first two principal components for each sample. The color code of the samples is as indicated. <b>B.</b> The detailed probabilities of 10-fold cross-validation for the samples of training set based on the expression values of 16 genes are shown. For each sample, its probability as AA (orange color) and GBM (blue color) are shown and it was predicted by the PAM program as either AA or GBM based on which grade's probability is higher. The original histological grade of the samples is shown on the top.</p

    Discordant samples do not exhibit clinical features and molecular markers of its histologic grade.

    No full text
    <p><b>A.</b> The average Age at Diagnosis along with standard deviation is plotted for Authentic AAs (n = 21), Authentic GBMs (n = 37), Discordant AAs (n = 8) and Discordant GBMs (n = 20). *** denotes that P<0.001, ** denotes that P<0.01 and * denotes that P<0.05. <b>B.</b> The Kaplan Meier survival analysis of Authentic AAs (n = 13), Authentic GBMs (n = 165) and Discordant GBMs (n = 13). The median survival was significantly different across the groups with P<0.001. <b>C.</b> The percentage of samples showing CDKN2A/2B loss, EGFR amplification and p53 mutation is plotted for Authentic AAs (n = 14), Authentic GBMs (n = 37), Discordant AAs (n = 5) and Discordant GBMs (n = 2).</p
    corecore