Signatures of cell death and proliferation in perturbation transcriptomics data—from confounding factor to effective prediction

Transcriptomics perturbation signatures are valuable data sources for functional genomic studies. They can be effectively used to identify mechanism of action for new compounds and to infer functional activity of different cellular processes. Linking perturbation signatures to phenotypic studies opens up the possibility to model selected cellular phenotypes from gene expression data and to predict drugs interfering with the phenotype. At the same time, close association of transcriptomics changes with phenotypes can potentially mask the compound specific signatures. By linking perturbation transcriptomics data from the LINCS-L1000 project with cell viability phenotypic information upon genetic (from Achilles project) and chemical (from CTRP screen) perturbations for more than 90,000 signature - cell viability pairs, we show here that a cell death signature is a major factor behind perturbation signatures. We use this relationship to effectively predict cell viability from transcriptomics signatures, and identify compounds that induce either cell death or proliferation. We also show that cellular toxicity can lead to an unexpected similarity of toxic compound signatures confounding the mechanism of action discovery. Consensus compound signatures predict cell-specific anti-cancer drug sensitivity, even if the drug signature is not measured in the same cell line. These signatures outperform conventional drug-specific features like nominal target and chemical fingerprints. Our results can help removing confounding factors of large scale transcriptomics perturbation screens and show that expression signatures boost prediction of drug sensitivity.

[1]  Gurmit Singh,et al.  Doxycycline and other tetracyclines in the treatment of bone metastasis. , 2003, Anti-cancer drugs.

[2]  J. Massagué,et al.  Cyclin-dependent Kinase Inhibitors Uncouple Cell Cycle Progression from Mitochondrial Apoptotic Functions in DNA-damaged Cancer Cells* , 2005, Journal of Biological Chemistry.

[3]  Paul A Clemons,et al.  The Connectivity Map: Using Gene-Expression Signatures to Connect Small Molecules, Genes, and Disease , 2006, Science.

[4]  R. Shoemaker The NCI60 human tumour cell line anticancer drug screen , 2006, Nature Reviews Cancer.

[5]  Geoffrey E. Hinton,et al.  Visualizing Data using t-SNE , 2008 .

[6]  Xavier Robin,et al.  pROC: an open-source package for R and S+ to analyze and compare ROC curves , 2011, BMC Bioinformatics.

[7]  D. di Bernardo,et al.  Identification of small molecules enhancing autophagic function from drug network analysis. , 2010, Autophagy.

[8]  Tjerk P. Straatsma,et al.  NWChem: A comprehensive and scalable open-source solution for large scale molecular simulations , 2010, Comput. Phys. Commun..

[9]  Bertram Klinger,et al.  Discovering causal signaling pathways through gene-expression patterns , 2010, Nucleic Acids Res..

[10]  Gaël Varoquaux,et al.  Scikit-learn: Machine Learning in Python , 2011, J. Mach. Learn. Res..

[11]  S. Ramaswamy,et al.  Systematic identification of genomic markers of drug sensitivity in cancer cells , 2012, Nature.

[12]  T. Cheng,et al.  An Opposite Effect of the CDK Inhibitor, p18INK4c on Embryonic Stem Cells Compared with Tumor and Adult Stem Cells , 2012, PloS one.

[13]  Sean R. Davis,et al.  NCBI GEO: archive for functional genomics data sets—update , 2012, Nucleic Acids Res..

[14]  I. Nookaew,et al.  Enriching the gene set analysis of genome-wide data by incorporating directionality of gene expression and combining statistical hypotheses and methods , 2013, Nucleic acids research.

[15]  P. McGettigan Transcriptomics in the RNA-seq era. , 2013, Current opinion in chemical biology.

[16]  Benjamin Haibe-Kains,et al.  Inconsistency in large pharmacogenomic studies , 2013, Nature.

[17]  L. Trojan,et al.  Testosterone boosts for treatment of castration resistant prostate cancer: An experimental implementation of intermittent androgen deprivation , 2013, The Prostate.

[18]  W. Oh,et al.  Roles of Matrix Metalloproteinases and Their Natural Inhibitors in Prostate Cancer Progression , 2014, Cancers.

[19]  Nci Dream Community A community effort to assess and improve drug sensitivity prediction algorithms , 2014 .

[20]  L. Wodicka,et al.  Dual kinase-bromodomain inhibitors for rationally designed polypharmacology , 2014, Nature chemical biology.

[21]  Michael P. Morrissey,et al.  Pharmacogenomic agreement between two cancer cell line data sets , 2015, Nature.

[22]  Joshua A. Bittker,et al.  Harnessing Connectivity in a Large-Scale Small-Molecule Sensitivity Dataset. , 2015, Cancer discovery.

[23]  Emanuel J. V. Gonçalves,et al.  A Landscape of Pharmacogenomic Interactions in Cancer , 2016, Cell.

[24]  M. Kusaka,et al.  Growth Inhibition by Testosterone in an Androgen Receptor Splice Variant‐Driven Prostate Cancer Model , 2016, The Prostate.

[25]  Y. Hu,et al.  ERK5 kinase activity is dispensable for cellular immune response and proliferation , 2016, Proceedings of the National Academy of Sciences.

[26]  Kathleen M Jagodnik,et al.  Extraction and analysis of signatures from the Gene Expression Omnibus by the crowd , 2016, Nature Communications.

[27]  Defining a Cancer Dependency Map , 2017, Cell.

[28]  Evan O. Paull,et al.  A Community Challenge for Inferring Genetic Predictors of Gene Essentialities through Analysis of a Functional Screen of Cancer Cell Lines. , 2017, Cell systems.

[29]  Angela N. Brooks,et al.  A Next Generation Connectivity Map: L1000 Platform and the First 1,000,000 Profiles , 2017, Cell.

[30]  Ted Natoli,et al.  Evaluation of RNAi and CRISPR technologies by large-scale gene expression profiling in the Connectivity Map , 2017, bioRxiv.

[31]  J. Sáez-Rodríguez,et al.  Perturbation-response genes reveal signaling footprints in cancer gene expression , 2016, Nature Communications.

[32]  Marc Hafner,et al.  Common and cell-type specific responses to anti-cancer drugs revealed by high throughput transcript profiling , 2017, Nature Communications.

[33]  Jacob K. Asiedu,et al.  The Drug Repurposing Hub: a next-generation drug library and information resource , 2017, Nature Medicine.

[34]  Tero Aittokallio,et al.  Machine learning and feature selection for drug response prediction in precision oncology applications , 2018, Biophysical Reviews.

[35]  Nuno A. Fonseca,et al.  Transcription Factor Activities Enhance Markers of Drug Sensitivity in Cancer. , 2018, Cancer research.

[36]  Rajiv Narayan,et al.  The GCTx format and cmap{Py, R, M} packages: resources for the optimized storage and integrated traversal of dense matrices of data and annotations , 2018, bioRxiv.

[37]  Doheon Lee,et al.  Deconvoluting essential gene signatures for cancer growth from genomic expression in compound-treated cells , 2018, Bioinform..