6 research outputs found

    Quantitative proteome landscape of the NCI-60 cancer cell lines

    Get PDF
    Here we describe a proteomic data resource for the NCI-60 cell lines generated by pressure cycling technology and SWATH mass spectrometry. We developed the DIA-expert software to curate and visualize the SWATH data, leading to reproducible detection of over 3,100 SwissProt proteotypic proteins and systematic quantification of pathway activities. Stoichiometric relationships of interacting proteins for DNA replication, repair, the chromatin remodeling NuRD complex, β-catenin, RNA metabolism, and prefoldins are more evident than that at the mRNA level. The data are available in CellMiner (discover.nci.nih.gov/cellminercdb and discover.nci.nih.gov/cellminer), allowing casual users to test hypotheses and perform integrative, cross-database analyses of multi-omic drug response correlations for over 20,000 drugs. We demonstrate the value of proteome data in predicting drug response for over 240 clinically relevant chemotherapeutic and targeted therapies. In summary, we present a novel proteome resource for the NCI-60, together with relevant software tools, and demonstrate the benefit of proteome analyses

    A statistical framework for assessing pharmacological responses and biomarkers using uncertainty estimates

    Get PDF
    High-throughput testing of drugs across molecular-characterised cell lines can identify candidate treatments and discover biomarkers. However, the cells’ response to a drug is typically quantified by a summary statistic from a best-fit dose-response curve, whilst neglecting the uncertainty of the curve fit and the potential variability in the raw readouts. Here, we model the experimental variance using Gaussian Processes, and subsequently, leverage uncertainty estimates to identify associated biomarkers with a new Bayesian framework. Applied to in vitro screening data on 265 compounds across 1074 cancer cell lines, our models identified 24 clinically established drug-response biomarkers, and provided evidence for six novel biomarkers by accounting for association with low uncertainty. We validated our uncertainty estimates with an additional drug screen of 26 drugs, 10 cell lines with 8 to 9 replicates. Our method is applicable to any dose-response data without replicates, and improves biomarker discovery for precision medicine

    Applying synergy metrics to oncology combination screening data: agreements, disagreements and pitfalls

    No full text
    Synergistic drug combinations are commonly sought to overcome monotherapy resistance in cancer treatment. To identify such combinations, high-throughput cancer-cell-line combination screens are performed; and synergy is quantified using competing models based on fundamentally different assumptions. Here, we compare the behaviour of four synergy models, namely Loewe additivity, Bliss independence, highest single agent and zero interaction potency, using the Merck oncology combination screen. We evaluate agreements and disagreements between the models and investigate putative artefacts of each model's assumptions. Despite at least moderate concordance between scores (Pearson's r >0.32, Spearman's ρ > 0.34), multiple instances of strong disagreement were observed. Those disagreements are driven by, among others, large differences in tested concentrations, maximum response values and median effective concentrations

    Identification of Intrinsic Drug Resistance and Its Biomarkers in High-Throughput Pharmacogenomic and CRISPR Screens

    No full text
    High-throughput drug screens in cancer cell lines test compounds at low concentrations, thereby enabling the identification of drug-sensitivity biomarkers, while resistance biomarkers remain underexplored. Dissecting meaningful drug responses at high concentrations is challenging due to cytotoxicity, i.e., off-target effects, thus limiting resistance biomarker discovery to frequently mutated cancer genes. To address this, we interrogate subpopulations carrying sensitivity biomarkers and consecutively investigate unexpectedly resistant (UNRES) cell lines for unique genetic alterations that may drive resistance. By analyzing the GDSC and CTRP datasets, we find 53 and 35 UNRES cases, respectively. For 24 and 28 of them, we highlight putative resistance biomarkers. We find clinically relevant cases such as EGFRT790M mutation in NCI-H1975 or PTEN loss in NCI-H1650 cells, in lung adenocarcinoma treated with EGFR inhibitors. Interrogating the underpinnings of drug resistance with publicly available CRISPR phenotypic assays assists in prioritizing resistance drivers, offering hypotheses for drug combinations. Cancer drug resistance is the major challenge of modern oncology. Identifying resistance and its biomarkers will empower the next generation of precision medicines. High-throughput pharmacology screens in cancer cell lines have successfully identified drug-sensitivity biomarkers, but drug-resistance biomarkers are underexplored. Intrinsic drug-resistance events are often rare and experimentally indistinguishable from cytotoxicity or artifacts without prior knowledge. To address this, we investigate cell-line populations sensitized to a drug treatment (i.e., carrying established sensitivity biomarkers) and characterize those cell lines that do not respond as expected. We highlight unique genetic features harbored by these cell lines and confirm their linkage to drug resistance using CRISPR gene essentiality data. Our analysis and results pave the way for enhanced precision medicine, guide further CRISPR screens, and identify potential drug combinations to tackle resistance. Identifying cancer drug resistance and its biomarkers will empower the next generation of anti-cancer medicines, tailoring treatments to individual patients. Detecting drug resistance in high-throughput pharmacology screens is experimentally challenging. We present a computational framework identifying rare intrinsically resistant cancer cell lines. Our observations provide hypotheses for associated drug-resistance biomarkers, which we validate with independent CRISPR essentiality screens. Our results pave the way for enhancing cancer precision medicine and effective drug combinations to overcome resistance. © 2020 The Author

    Bibliography

    No full text
    corecore