Drug mechanism‐of‐action discovery through the integration of pharmacological and CRISPR screens

Low success rates during drug development are due in part to the difficulty of defining drug mechanism-of-action and molecular markers of therapeutic activity. Here, we integrated 199,219 drug sensitivity measurements for 397 unique anti-cancer drugs and genome-wide CRISPR loss-of-function screens in 484 cell lines to systematically investigate in cellular drug mechanism-of-action. We observed an enrichment for positive associations between drug sensitivity and knockout of their nominal targets, and by leveraging protein-protein networks we identified pathways that mediate drug response. This revealed an unappreciated role of mitochondrial E3 ubiquitin-protein ligase MARCH5 in sensitivity to MCL1 inhibitors. We also estimated drug on-target and off-target activity, informing on specificity, potency and toxicity. Linking drug and gene dependency together with genomic datasets uncovered contexts in which molecular networks when perturbed mediate cancer cell loss-of-fitness, and thereby provide independent and orthogonal evidence of biomarkers for drug development. This study illustrates how integrating cell line drug sensitivity with CRISPR loss-of-function screens can elucidate mechanism-of-action to advance drug development.

[1]  Lei Liu,et al.  MARCH5 requires MTCH2 to coordinate proteasomal turnover of the MCL1:NOXA complex , 2020, Cell Death & Differentiation.

[2]  B. Al-Lazikani,et al.  The kinase polypharmacology landscape of clinical PARP inhibitors , 2020, Scientific Reports.

[3]  A. Villunger,et al.  MARCH5-dependent degradation of MCL1/NOXA complexes defines susceptibility to antimitotic drug treatment , 2020, Cell Death & Differentiation.

[4]  A. Villunger,et al.  MARCH5-dependent degradation of MCL1/NOXA complexes defines susceptibility to anti-mitotic drug treatment , 2020, Cell Death & Differentiation.

[5]  Eiru Kim,et al.  Biases and Blind-Spots in Genome-Wide CRISPR Knockout Screens , 2020, bioRxiv.

[6]  A. Lin,et al.  Off-target toxicity is a common mechanism of action of cancer drugs undergoing clinical trials , 2019, Science Translational Medicine.

[7]  D. Durocher,et al.  Control of homologous recombination by the HROB–MCM8–MCM9 pathway , 2019, Genes & Development.

[8]  Johannes L. Schönberger,et al.  SciPy 1.0: fundamental algorithms for scientific computing in Python , 2019, Nature Methods.

[9]  Peter C. DeWeirdt,et al.  Genetic screens in isogenic mammalian cell lines without single cell cloning , 2019, Nature Communications.

[10]  Joshua M. Dempster,et al.  Agreement between two large pan-cancer CRISPR-Cas9 gene dependency data sets , 2019, Nature Communications.

[11]  Emanuel J. V. Gonçalves,et al.  Prioritization of cancer therapeutic targets using CRISPR–Cas9 screens , 2019, Nature.

[12]  Eiru Kim,et al.  A network of human functional gene interactions from knockout fitness screens in cancer cells , 2019, Life Science Alliance.

[13]  B. Kuster,et al.  Chemoproteomic Selectivity Profiling of PIKK and PI3K Kinase Inhibitors. , 2019, ACS chemical biology.

[14]  D. Durocher,et al.  A consensus set of genetic vulnerabilities to ATR inhibition , 2019, bioRxiv.

[15]  Emanuel J. V. Gonçalves,et al.  Functional linkage of gene fusions to cancer cell fitness assessed by pharmacological and CRISPR-Cas9 screening , 2019, Nature Communications.

[16]  Emanuel J. V. Gonçalves,et al.  Structural rearrangements generate cell-specific, gene-independent CRISPR-Cas9 loss of fitness effects , 2019, Genome Biology.

[17]  P. Pourquier,et al.  [Genetic and transcriptional evolution alters cancer cell line drug response]. , 2019, Bulletin du cancer.

[18]  Andrew R. Leach,et al.  ChEMBL: towards direct deposition of bioassay data , 2018, Nucleic Acids Res..

[19]  Howard Lightfoot,et al.  Cell Model Passports—a hub for clinical, genetic and functional datasets of preclinical cancer models , 2018, Nucleic Acids Res..

[20]  Kyoung-Mee Kim,et al.  Pharmacogenomic landscape of patient-derived tumor cells informs precision oncology therapy , 2018, Nature Genetics.

[21]  J. Pritchard,et al.  High‐resolution mapping of cancer cell networks using co‐functional interactions , 2018, bioRxiv.

[22]  Martin A. M. Reijns,et al.  CRISPR screens identify genomic ribonucleotides as a source of PARP-trapping lesions , 2018, Nature.

[23]  Evert Bosdriesz,et al.  An Acquired Vulnerability of Drug-Resistant Melanoma with Therapeutic Potential , 2018, Cell.

[24]  Aviad Tsherniak,et al.  Interrogation of Mammalian Protein Complex Structure, Function, and Membership Using Genome-Scale Fitness Screens. , 2018, Cell systems.

[25]  J. Weissman,et al.  CRISPR Approaches to Small Molecule Target Identification. , 2017, ACS chemical biology.

[26]  Emanuel J. V. Gonçalves,et al.  Unsupervised correction of gene-independent cell responses to CRISPR-Cas9 targeting , 2017, BMC Genomics.

[27]  B. Kuster,et al.  The target landscape of clinical kinase drugs , 2017, Science.

[28]  C. Myers,et al.  Pathway-based discovery of genetic interactions in breast cancer , 2017, PLoS genetics.

[29]  Phillip G. Montgomery,et al.  Defining a Cancer Dependency Map , 2017, Cell.

[30]  Ann E. Sizemore,et al.  Computational correction of copy-number effect improves specificity of CRISPR-Cas9 essentiality screens in cancer cells , 2017, Nature Genetics.

[31]  S. Kazmirski,et al.  Abstract DDT01-02: AZD5991: A potent and selective macrocyclic inhibitor of Mcl-1 for treatment of hematologic cancers , 2017 .

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

[33]  Thomas Hielscher,et al.  Toward an integrated map of genetic interactions in cancer cells , 2017, bioRxiv.

[34]  Yiling Lu,et al.  Characterization of Human Cancer Cell Lines by Reverse-phase Protein Arrays. , 2017, Cancer cell.

[35]  Eric S. Lander,et al.  Gene Essentiality Profiling Reveals Gene Networks and Synthetic Lethal Interactions with Oncogenic Ras , 2017, Cell.

[36]  O. Stegle,et al.  Joint genetic analysis using variant sets reveals polygenic gene-context interactions , 2016, bioRxiv.

[37]  Tudor I. Oprea,et al.  A comprehensive map of molecular drug targets , 2016, Nature Reviews Drug Discovery.

[38]  Damian Szklarczyk,et al.  The STRING database in 2017: quality-controlled protein–protein association networks, made broadly accessible , 2016, Nucleic Acids Res..

[39]  Julio Saez-Rodriguez,et al.  A CRISPR Dropout Screen Identifies Genetic Vulnerabilities and Therapeutic Targets in Acute Myeloid Leukemia , 2016, Cell reports.

[40]  Joshua M. Korn,et al.  CRISPR Screens Provide a Comprehensive Assessment of Cancer Vulnerabilities but Generate False-Positive Hits for Highly Amplified Genomic Regions. , 2016, Cancer discovery.

[41]  T. Golub,et al.  Genomic Copy Number Dictates a Gene-Independent Cell Response to CRISPR/Cas9 Targeting. , 2016, Cancer discovery.

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

[43]  Francesco Iorio,et al.  Multilevel models improve precision and speed of IC50 estimates. , 2016, Pharmacogenomics.

[44]  U. McDermott,et al.  Identification of differential PI3K pathway target dependencies in T-cell acute lymphoblastic leukemia through a large cancer cell panel screen , 2016, Oncotarget.

[45]  Max A. Horlbeck,et al.  Parallel shRNA and CRISPR-Cas9 screens enable antiviral drug target identification , 2016, Nature chemical biology.

[46]  Gary D Bader,et al.  Functional Genomic Landscape of Human Breast Cancer Drivers, Vulnerabilities, and Resistance , 2016, Cell.

[47]  P. Pinton,et al.  Mcl-1 involvement in mitochondrial dynamics is associated with apoptotic cell death , 2016, Molecular biology of the cell.

[48]  D. Durocher,et al.  High-Resolution CRISPR Screens Reveal Fitness Genes and Genotype-Specific Cancer Liabilities , 2015, Cell.

[49]  Meagan E. Sullender,et al.  Optimized sgRNA design to maximize activity and minimize off-target effects of CRISPR-Cas9 , 2015, Nature Biotechnology.

[50]  M. Stratton,et al.  Combinations of PARP Inhibitors with Temozolomide Drive PARP1 Trapping and Apoptosis in Ewing’s Sarcoma , 2015, PloS one.

[51]  B. Kuster,et al.  Optimized chemical proteomics assay for kinase inhibitor profiling. , 2015, Journal of proteome research.

[52]  Jean J. Zhao,et al.  PI3K in cancer: divergent roles of isoforms, modes of activation and therapeutic targeting , 2014, Nature Reviews Cancer.

[53]  G. Drewes,et al.  Tracking cancer drugs in living cells by thermal profiling of the proteome , 2014, Science.

[54]  M. Pangalos,et al.  Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework , 2014, Nature Reviews Drug Discovery.

[55]  S. Desagher,et al.  Mcl-1 Ubiquitination: Unique Regulation of an Essential Survival Protein , 2014, Cells.

[56]  C. Lippert,et al.  LIMIX: genetic analysis of multiple traits , 2014, bioRxiv.

[57]  G. Smyth,et al.  voom: precision weights unlock linear model analysis tools for RNA-seq read counts , 2014, Genome Biology.

[58]  Neville E. Sanjana,et al.  Genome-Scale CRISPR-Cas9 Knockout Screening in Human Cells , 2014, Science.

[59]  Yilong Li,et al.  Genome-wide recessive genetic screening in mammalian cells with a lentiviral CRISPR-guide RNA library , 2013, Nature Biotechnology.

[60]  Peter E. Czabotar,et al.  Control of apoptosis by the BCL-2 protein family: implications for physiology and therapy , 2013, Nature Reviews Molecular Cell Biology.

[61]  Tony Pawson,et al.  Temporal regulation of EGF signaling networks by the scaffold protein Shc1 , 2013, Nature.

[62]  P. Clemons,et al.  Target identification and mechanism of action in chemical biology and drug discovery. , 2013, Nature chemical biology.

[63]  Sridhar Ramaswamy,et al.  Genomics of Drug Sensitivity in Cancer (GDSC): a resource for therapeutic biomarker discovery in cancer cells , 2012, Nucleic Acids Res..

[64]  R. Korneluk,et al.  Modulation of immune signalling by inhibitors of apoptosis. , 2012, Trends in immunology.

[65]  J. Doudna,et al.  A Programmable Dual-RNA–Guided DNA Endonuclease in Adaptive Bacterial Immunity , 2012, Science.

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

[67]  Adam A. Margolin,et al.  The Cancer Cell Line Encyclopedia enables predictive modeling of anticancer drug sensitivity , 2012, Nature.

[68]  Gaël Varoquaux,et al.  The NumPy Array: A Structure for Efficient Numerical Computation , 2011, Computing in Science & Engineering.

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

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

[71]  Derek Y. Chiang,et al.  The landscape of somatic copy-number alteration across human cancers , 2010, Nature.

[72]  Bernhard Kuster,et al.  Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors , 2007, Nature Biotechnology.

[73]  R. Youle,et al.  The mitochondrial E3 ubiquitin ligase MARCH5 is required for Drp1 dependent mitochondrial division , 2007, The Journal of cell biology.

[74]  John D. Hunter,et al.  Matplotlib: A 2D Graphics Environment , 2007, Computing in Science & Engineering.

[75]  T. Golub,et al.  Gene expression-based chemical genomics identifies rapamycin as a modulator of MCL1 and glucocorticoid resistance. , 2006, Cancer cell.

[76]  J. Baselga,et al.  The Epidermal Growth Factor Receptor Pathway: A Model for Targeted Therapy , 2006, Clinical Cancer Research.

[77]  R. Youle,et al.  Mitochondrial fission in apoptosis , 2005, Nature Reviews Molecular Cell Biology.

[78]  R. Bataille,et al.  Mcl-1 is overexpressed in multiple myeloma and associated with relapse and shorter survival , 2005, Leukemia.

[79]  G. Thomas,et al.  Furin at the cutting edge: From protein traffic to embryogenesis and disease , 2002, Nature Reviews Molecular Cell Biology.

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

[81]  Y. Pommier,et al.  Classification of PARP Inhibitors Based on PARP Trapping and Catalytic Inhibition, and Rationale for Combinations with Topoisomerase I Inhibitors and Alkylating Agents , 2015 .

[82]  Y. Miki,et al.  [PARP inhibitors for cancer therapy]. , 2011, Gan to kagaku ryoho. Cancer & chemotherapy.

[83]  Wes McKinney,et al.  Data Structures for Statistical Computing in Python , 2010, SciPy.

[84]  Gábor Csárdi,et al.  The igraph software package for complex network research , 2006 .

[85]  T. A. Connors Alkylating agents. , 1990, Cancer chemotherapy and biological response modifiers.

[86]  Mulin Jun Li,et al.  Nature Genetics Advance Online Publication a N a Ly S I S the Support of Human Genetic Evidence for Approved Drug Indications , 2022 .

[87]  the original work is properly cited. , 2022 .