21 research outputs found

    HIF2alpha cooperates with RAS to promote lung tumorigenesis in mice.

    Get PDF
    Members of the hypoxia-inducible factor (HIF) family of transcription factors regulate the cellular response to hypoxia. In non-small cell lung cancer (NSCLC), high HIF2alpha levels correlate with decreased overall survival, and inhibition of either the protein encoded by the canonical HIF target gene VEGF or VEGFR2 improves clinical outcomes. However, whether HIF2alpha is causal in imparting this poor prognosis is unknown. Here, we generated mice that conditionally express both a nondegradable variant of HIF2alpha and a mutant form of Kras (KrasG12D) that induces lung tumors. Mice expressing both Hif2a and KrasG12D in the lungs developed larger tumors and had an increased tumor burden and decreased survival compared with mice expressing only KrasG12D. Additionally, tumors expressing both KrasG12D and Hif2a were more invasive, demonstrated features of epithelial- mesenchymal transition (EMT), and exhibited increased angiogenesis associated with mobilization of circulating endothelial progenitor cells. These results implicate HIF2alpha causally in the pathogenesis of lung cancer in mice, demonstrate in vivo that HIF2alpha can promote expression of markers of EMT, and define HIF2alpha as a promoter of tumor growth and progression in a solid tumor other than renal cell carcinoma. They further suggest a possible causal relationship between HIF2alpha and prognosis in patients with NSCLC

    CTC-Race: Single-Cell Motility Assay of Circulating Tumor Cells from Metastatic Lung Cancer Patients

    No full text
    Distinctive subpopulations of circulating tumor cells (CTCs) with increased motility are considered to possess enhanced tumor-initiating potential and contribute to metastasis. Single-cell analysis of the migratory CTCs may increase our understanding of the metastatic process, yet most studies are limited by technical challenges associated with the isolation and characterization of these cells due to their extreme scarcity and heterogeneity. We report a microfluidic method based on CTCs’ chemotactic motility, termed as CTC-Race assay, that can analyze migrating CTCs from metastatic non-small-cell lung cancer (NSCLC) patients with advanced tumor stages and enable concurrent biophysical and biochemical characterization of them with single-cell resolution. Analyses of motile CTCs in the CTC-Race assay, in synergy with other single cell characterization techniques, could provide insights into cancer metastasis

    Data from: Analytical and clinical validation of a digital sequencing panel for quantitative, highly accurate evaluation of cell-free circulating tumor DNA

    No full text
    Next-generation sequencing of cell-free circulating solid tumor DNA addresses two challenges in contemporary cancer care. First this method of massively parallel and deep sequencing enables assessment of a comprehensive panel of genomic targets from a single sample, and second, it obviates the need for repeat invasive tissue biopsies. Digital SequencingTM is a novel method for high-quality sequencing of circulating tumor DNA simultaneously across a comprehensive panel of over 50 cancer-related genes with a simple blood test. Here we report the analytic and clinical validation of the gene panel. Analytic sensitivity down to 0.1% mutant allele fraction is demonstrated via serial dilution studies of known samples. Near-perfect analytic specificity (> 99.9999%) enables complete coverage of many genes without the false positives typically seen with traditional sequencing assays at mutant allele frequencies or fractions below 5%. We compared digital sequencing of plasma-derived cell-free DNA to tissue-based sequencing on 165 consecutive matched samples from five outside centers in patients with stage III-IV solid tumor cancers. Clinical sensitivity of plasma-derived NGS was 85.0%, comparable to 80.7% sensitivity for tissue. The assay success rate on 1,000 consecutive samples in clinical practice was 99.8%. Digital sequencing of plasma-derived DNA is indicated in advanced cancer patients to prevent repeated invasive biopsies when the initial biopsy is inadequate, unobtainable for genomic testing, or uninformative, or when the patient’s cancer has progressed despite treatment. Its clinical utility is derived from reduction in the costs, complications and delays associated with invasive tissue biopsies for genomic testing

    Analytical and Clinical Validation of a Digital Sequencing Panel for Quantitative, Highly Accurate Evaluation of Cell-Free Circulating Tumor DNA

    No full text
    <div><p>Next-generation sequencing of cell-free circulating solid tumor DNA addresses two challenges in contemporary cancer care. First this method of massively parallel and deep sequencing enables assessment of a comprehensive panel of genomic targets from a single sample, and second, it obviates the need for repeat invasive tissue biopsies. Digital Sequencing<sup>TM</sup> is a novel method for high-quality sequencing of circulating tumor DNA simultaneously across a comprehensive panel of over 50 cancer-related genes with a simple blood test. Here we report the analytic and clinical validation of the gene panel. Analytic sensitivity down to 0.1% mutant allele fraction is demonstrated via serial dilution studies of known samples. Near-perfect analytic specificity (> 99.9999%) enables complete coverage of many genes without the false positives typically seen with traditional sequencing assays at mutant allele frequencies or fractions below 5%. We compared digital sequencing of plasma-derived cell-free DNA to tissue-based sequencing on 165 consecutive matched samples from five outside centers in patients with stage III-IV solid tumor cancers. Clinical sensitivity of plasma-derived NGS was 85.0%, comparable to 80.7% sensitivity for tissue. The assay success rate on 1,000 consecutive samples in clinical practice was 99.8%. Digital sequencing of plasma-derived DNA is indicated in advanced cancer patients to prevent repeated invasive biopsies when the initial biopsy is inadequate, unobtainable for genomic testing, or uninformative, or when the patient’s cancer has progressed despite treatment. Its clinical utility is derived from reduction in the costs, complications and delays associated with invasive tissue biopsies for genomic testing.</p></div

    Fig 7A is a comparison of tissue NGS results biopsied at five outside institutions compared to cfDNA sequencing at Guardant Health on 165 paired plasma samples from stage III-IV solid tumor cancer patients. Data summarizes diagnostic test performance for all 54 mutated tumor suppressor and oncogenes. The most commonly mutated genes were <i>ALK, APC, BRAF, CDKN2A, CTNNB1, FBXW7, KRAS, NRAS, PIK3CA, PTEN</i>, and <i>TP53</i>. Sensitivity, specificity and diagnostic accuracy are shown with 95% confidence intervals.Fig 7B illustrates the two by two contingency tables corresponding to Fig 7A. On the left cfDNA NGS results are compared to tissue-based NGS as the reference standard. On the right tissue-based NGS results are compared to cfDNA findings as the reference standard. All gene mutations found in cfDNA and tissue DNA based on NGS of 54 genes are shown in S2 Table. Both methods demonstrate similarly high sensitivity and near-perfect specificity. For cfDNA, sensitivity is limited by the

    No full text
    <p>Fig 7A is a comparison of tissue NGS results biopsied at five outside institutions compared to cfDNA sequencing at Guardant Health on 165 paired plasma samples from stage III-IV solid tumor cancer patients. Data summarizes diagnostic test performance for all 54 mutated tumor suppressor and oncogenes. The most commonly mutated genes were <i>ALK, APC, BRAF, CDKN2A, CTNNB1, FBXW7, KRAS, NRAS, PIK3CA, PTEN</i>, and <i>TP53</i>. Sensitivity, specificity and diagnostic accuracy are shown with 95% confidence intervals.Fig 7B illustrates the two by two contingency tables corresponding to Fig 7A. On the left cfDNA NGS results are compared to tissue-based NGS as the reference standard. On the right tissue-based NGS results are compared to cfDNA findings as the reference standard. All gene mutations found in cfDNA and tissue DNA based on NGS of 54 genes are shown in <a href="http://www.plosone.org/article/info:doi/10.1371/journal.pone.0140712#pone.0140712.s004" target="_blank">S2 Table</a>. Both methods demonstrate similarly high sensitivity and near-perfect specificity. For cfDNA, sensitivity is limited by the amount of tumor DNA shed into circulation and for tissue, sensitivity is likely limited by sampling error related to intra-or inter-tumor heterogeneity. The sampling error on tissue samples may be related to sub-sampling of tumor heterogeneity by needle or surgical biopsy.</p

    Analytic Sensitivity of the Guardant360 digital sequencing method.

    No full text
    <p>Twenty nine SNV samples were diluted serially until they could no longer be measured with the assay. Each column represents successively greater serial dilutions of a given sample. The limit of detection (LOD) was 0.25% mutant allele frequency or fraction) (MAF), defined as the percentage at which > 80% of samples were detected. Note that almost 30% of samples were additionally detected at 0.1% MAF or lower, where 0.1% represents a single mutated DNA fragment out of 999 wild-type (leukocyte-derived) DNA fragments overlapping the same nucleotide base.</p
    corecore