Comparing structural and transcriptional drug networks reveals signatures of drug activity and toxicity in transcriptional responses

We performed an integrated analysis of drug chemical structures and drug-induced transcriptional responses. We demonstrated that a network representing three-dimensional structural similarities among 5452 compounds can be used to automatically group together drugs with similar scaffolds, physicochemical parameters and mode-of-action. We compared the structural network to a network representing transcriptional similarities among a subset of 1309 drugs for which transcriptional response were available in the Connectivity Map data set. Analysis of structurally similar, but transcriptionally different drugs sharing the same MOA enabled us to detect and remove weak and noisy transcriptional responses, greatly enhancing the reliability of transcription-based approaches to drug discovery and drug repositioning. Cardiac glycosides exhibited the strongest transcriptional responses with a significant induction of pathways related to epigenetic regulation, which suggests an epigenetic mechanism of action for these drugs. Drug classes with the weakest transcriptional responses tended to induce expression of cytochrome P450 enzymes, hinting at drug-induced drug resistance. Analysis of transcriptionally similar, but structurally different drugs with unrelated MOA, led us to the identification of a 'toxic' transcriptional signature indicative of lysosomal stress (lysosomotropism) and lipid accumulation (phospholipidosis) partially masking the target-specific transcriptional effects of these drugs. We found that this transcriptional signature is shared by 258 compounds and it is associated to the activation of the transcription factor TFEB, a master regulator of lysosomal biogenesis and autophagy. Finally, we built a predictive Random Forest model of these 258 compounds based on 128 physicochemical parameters, which should help in the early identification of potentially toxic drug candidates.Systems pharmacology: the chemical-transcriptional landscape of small moleculesTranscriptional responses to drug treatment can reveal mechanism of action and off-target effects thus enabling drug repositioning, but only if measured in the appropriate cells at clinically relevant concentrations. A team led by Diego di Bernardo and Diego Medina generated a network representing structural similarities among compounds to automatically group together drugs with similar scaffolds and MOA. By comparing the structural drug network with a transcriptional drug network based on similarities in transcriptional response, the team observed broad differences between the two. This observation led to the identification of a transcriptional signature related lysosomal stress and phospholipidosis, and a physicochemical model to identify such compounds. These results provide general guidelines to prevent erroneous conclusion when using transcriptional responses of small molecules for drug discovery and drug repositioning

[1]  R. Tagliaferri,et al.  Discovery of drug mode of action and drug repositioning from transcriptional responses , 2010, Proceedings of the National Academy of Sciences.

[2]  Gabriele Cruciani,et al.  Modeling Phospholipidosis Induction: Reliability and Warnings , 2013, J. Chem. Inf. Model..

[3]  Dieter Lang,et al.  Predicting drug metabolism: experiment and/or computation? , 2015, Nature Reviews Drug Discovery.

[4]  Jürgen Borlak,et al.  Drug‐induced phospholipidosis , 2006, FEBS letters.

[5]  Alexander A. Morgan,et al.  Discovery and Preclinical Validation of Drug Indications Using Compendia of Public Gene Expression Data , 2011, Science Translational Medicine.

[6]  Luis Carrasco,et al.  Enzymic and nonenzymic translocation by yeast polysomes. Site of action of a number of inhibitors. , 1977, Biochemistry.

[7]  F. D. D. Fagagna,et al.  Crosstalk between chromatin state and DNA damage response in cellular senescence and cancer , 2012, Nature Reviews Cancer.

[8]  Justin Lamb,et al.  The Connectivity Map: a new tool for biomedical research , 2007, Nature Reviews Cancer.

[9]  F. Marceau,et al.  Cation trapping by cellular acidic compartments: beyond the concept of lysosomotropic drugs. , 2012, Toxicology and applied pharmacology.

[10]  Valerio Embrione,et al.  A Gene Network Regulating Lysosomal Biogenesis and Function , 2009, Science.

[11]  Jia Jia,et al.  Comparative analysis of machine learning methods in ligand-based virtual screening of large compound libraries. , 2009, Combinatorial chemistry & high throughput screening.

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

[13]  F. Marceau,et al.  Intense pseudotransport of a cationic drug mediated by vacuolar ATPase: procainamide-induced autophagic cell vacuolization. , 2008, Toxicology and applied pharmacology.

[14]  Johannes Kornhuber,et al.  Identification of Drugs Inducing Phospholipidosis by Novel in vitro Data , 2012, ChemMedChem.

[15]  Hanna Geppert,et al.  Current Trends in Ligand-Based Virtual Screening: Molecular Representations, Data Mining Methods, New Application Areas, and Performance Evaluation , 2010, J. Chem. Inf. Model..

[16]  S. Nordeen,et al.  Estrogen Inhibits Glucocorticoid Action via Protein Phosphatase 5 (PP5)-mediated Glucocorticoid Receptor Dephosphorylation* , 2009, The Journal of Biological Chemistry.

[17]  C. January,et al.  Block of HERG Potassium Channels by the Antihistamine Astemizole and its Metabolites Desmethylastemizole and Norastemizole , 1999, Journal of cardiovascular electrophysiology.

[18]  B. Hardesty,et al.  The mechanism of cycloheximide inhibition of protein synthesis in rabbit reticulocytes. , 1969, Biochemical and biophysical research communications.

[19]  Diego di Bernardo,et al.  Mantra 2.0: an online collaborative resource for drug mode of action and repurposing by network analysis , 2014, Bioinform..

[20]  Ruili Huang,et al.  Are hERG channel blockers also phospholipidosis inducers? , 2013, Bioorganic & medicinal chemistry letters.

[21]  P. Carrupt,et al.  Molecular fields in quantitative structure–permeation relationships: the VolSurf approach , 2000 .

[22]  L. Garren,et al.  On the mechanism of action of adrenocorticotropic hormone. The inhibitory site of cycloheximide in the pathway of steroid biosynthesis. , 1968, The Journal of biological chemistry.

[23]  C. Lipinski Lead- and drug-like compounds: the rule-of-five revolution. , 2004, Drug discovery today. Technologies.

[24]  F. Iorio,et al.  Transcriptional data: a new gateway to drug repositioning? , 2013, Drug discovery today.

[25]  Yong Wang,et al.  Network predicting drug's anatomical therapeutic chemical code , 2013, Bioinform..

[26]  I. Herr,et al.  Corticosteriod-induced chemotherapy resistance in urological cancers , 2006, Cancer biology & therapy.

[27]  Ulrich Bodenhofer,et al.  APCluster: an R package for affinity propagation clustering , 2011, Bioinform..

[28]  S. Baylin,et al.  Targeting Calcium Signaling Induces Epigenetic Reactivation of Tumor Suppressor Genes in Cancer. , 2016, Cancer Research.

[29]  Delbert Dueck,et al.  Clustering by Passing Messages Between Data Points , 2007, Science.

[30]  T. Walther,et al.  The Transcription Factor TFEB Links mTORC1 Signaling to Transcriptional Control of Lysosome Homeostasis , 2012, Science Signaling.

[31]  Pankaj Agarwal,et al.  Systematic evaluation of connectivity map for disease indications , 2014, Genome Medicine.

[32]  Peer Bork,et al.  Drug-Induced Regulation of Target Expression , 2010, PLoS Comput. Biol..

[33]  M Pastor,et al.  VolSurf: a new tool for the pharmacokinetic optimization of lead compounds. , 2000, European journal of pharmaceutical sciences : official journal of the European Federation for Pharmaceutical Sciences.

[34]  Diego di Bernardo,et al.  Drug-set enrichment analysis: a novel tool to investigate drug mode of action , 2015, Bioinform..

[35]  J. Krise,et al.  18. The Mechanisms and Therapeutic Consequences of Amine-Containing Drug Sequestration in Lysosomes , 2016 .

[36]  A. Ballabio,et al.  Transcriptional Activation of Lysosomal Exocytosis Promotes Cellular Clearance , 2011, Developmental cell.

[37]  A. Ballabio,et al.  TFEB at a glance , 2016, Journal of Cell Science.

[38]  H. Munro,et al.  Mechanism of cycloheximide inhibition of protein synthesis in a cell-free system prepared from rat liver. , 1969, The Journal of biological chemistry.

[39]  J. Auwerx,et al.  Mitochondria and Epigenetics - Crosstalk in Homeostasis and Stress. , 2017, Trends in cell biology.

[40]  Alexander D. MacKerell,et al.  Computational ligand-based rational design: Role of conformational sampling and force fields in model development. , 2011, MedChemComm.

[41]  B. Baets,et al.  Metrics and T-Equalities , 2002 .

[42]  N. D’alessandro,et al.  Dexamethasone-induced cytotoxic activity and drug resistance effects in androgen-independent prostate tumor PC-3 cells are mediated by lipocortin 1. , 1998, Oncology research.

[43]  Yvonne Will,et al.  A high content screening assay for identifying lysosomotropic compounds. , 2011, Toxicology in vitro : an international journal published in association with BIBRA.

[44]  Joel T Dudley,et al.  Mapping the effects of drugs on the immune system , 2015, Nature Biotechnology.

[45]  María Rodríguez Martínez,et al.  Elucidating Compound Mechanism of Action by Network Perturbation Analysis Graphical , 2015 .

[46]  Jürgen Bajorath,et al.  Activity Cliff Networks for Medicinal Chemistry , 2014, Drug development research.

[47]  Mathias Wawer,et al.  Navigating structure-activity landscapes. , 2009, Drug discovery today.

[48]  F. Maxfield,et al.  Lysosomes: Biology, Diseases, and Therapeutics: Biology, Diseases, and Therapeutics , 2016 .

[49]  Francesco Iorio,et al.  Identification of small molecules enhancing autophagic function from drug network analysis , 2010 .

[50]  R. Abraham,et al.  Lysosomal adaptation: How cells respond to lysosomotropic compounds , 2017, PloS one.

[51]  Shuyan Lu,et al.  The Contribution of Lysosomotropism to Autophagy Perturbation , 2013, PloS one.

[52]  Paolo Benedetti,et al.  FLAP: GRID Molecular Interaction Fields in Virtual Screening. Validation using the DUD Data Set , 2010, J. Chem. Inf. Model..

[53]  J. Bajorath,et al.  Chemoinformatics: a view of the field and current trends in method development. , 2012, Bioorganic & medicinal chemistry.

[54]  Gabriele Cruciani,et al.  A Common Reference Framework for Analyzing/Comparing Proteins and Ligands. Fingerprints for Ligands And Proteins (FLAP): Theory and Application , 2007, J. Chem. Inf. Model..

[55]  Yoshihiro Yamanishi,et al.  Predicting target proteins for drug candidate compounds based on drug-induced gene expression data in a chemical structure-independent manner , 2015, BMC Medical Genomics.

[56]  M. Gottesman,et al.  Targeting multidrug resistance in cancer , 2006, Nature Reviews Drug Discovery.

[57]  Gabriele Cruciani,et al.  Grid-derived structure-based 3D pharmacophores and their performance compared to docking. , 2010, Drug discovery today. Technologies.

[58]  Saskia Nijmeijer,et al.  Virtual Fragment Screening: Discovery of Histamine H3 Receptor Ligands Using Ligand-Based and Protein-Based Molecular Fingerprints , 2012, J. Chem. Inf. Model..

[59]  Krister Wennerberg,et al.  Identification of structural features in chemicals associated with cancer drug response: a systematic data-driven analysis , 2013, Bioinform..

[60]  Bie M. P. Verbist,et al.  Using transcriptomics to guide lead optimization in drug discovery projects: Lessons learned from the QSTAR project. , 2015, Drug discovery today.

[61]  Y. Harada,et al.  Reprofiling a classical anthelmintic, pyrvinium pamoate, as an anti-cancer drug targeting mitochondrial respiration , 2012, Front. Oncol..

[62]  A. Butte,et al.  Leveraging big data to transform target selection and drug discovery , 2016, Clinical pharmacology and therapeutics.

[63]  D. Feldman,et al.  Estradiol inhibits glucocorticoid receptor expression and induces glucocorticoid resistance in MCF-7 human breast cancer cells , 2001, The Journal of Steroid Biochemistry and Molecular Biology.

[64]  Aakash Chavan Ravindranath,et al.  Connecting gene expression data from connectivity map and in silico target predictions for small molecule mechanism-of-action analysis. , 2015, Molecular bioSystems.

[65]  A. Brown,et al.  HERG, a primary human ventricular target of the nonsedating antihistamine terfenadine. , 1996, Circulation.

[66]  J. Stenvang,et al.  Repurposing Cationic Amphiphilic Antihistamines for Cancer Treatment , 2016, EBioMedicine.

[67]  F. Lombardo,et al.  Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. , 2001, Advanced drug delivery reviews.

[68]  Yong Chen,et al.  MTORC1 functions as a transcriptional regulator of autophagy by preventing nuclear transport of TFEB , 2012, Autophagy.

[69]  G. Cruciani,et al.  Hydrogen bonding interactions of covalently bonded fluorine atoms: from crystallographic data to a new angular function in the GRID force field. , 2004, Journal of medicinal chemistry.

[70]  Ralph Mazitschek,et al.  Treatment of Obesity with Celastrol , 2015, Cell.

[71]  Robert A Newman,et al.  Cardiac glycosides as novel cancer therapeutic agents. , 2008, Molecular interventions.

[72]  N. Sonenberg,et al.  Structure-Activity Analysis of Niclosamide Reveals Potential Role for Cytoplasmic pH in Control of Mammalian Target of Rapamycin Complex 1 (mTORC1) Signaling* , 2012, The Journal of Biological Chemistry.

[73]  A. Parkinson,et al.  Lysosomal Sequestration (Trapping) of Lipophilic Amine (Cationic Amphiphilic) Drugs in Immortalized Human Hepatocytes (Fa2N-4 Cells) , 2013, Drug Metabolism and Disposition.

[74]  M. Lowe,et al.  Faculty Opinions recommendation of Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. , 2015 .

[75]  Loriano Storchi,et al.  New and Original pKa Prediction Method Using Grid Molecular Interaction Fields. , 2008 .

[76]  W H Halliwell,et al.  Cationic Amphiphilic Drug-Induced Phospholipidosis , 1997, Toxicologic pathology.

[77]  Yanli Jin,et al.  Antineoplastic mechanisms of niclosamide in acute myelogenous leukemia stem cells: inactivation of the NF-kappaB pathway and generation of reactive oxygen species. , 2010, Cancer research.

[78]  A. Ballabio,et al.  A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB , 2012, The EMBO journal.

[79]  Andrea Ballabio,et al.  TFEB Links Autophagy to Lysosomal Biogenesis , 2011, Science.

[80]  M E J Newman,et al.  Modularity and community structure in networks. , 2006, Proceedings of the National Academy of Sciences of the United States of America.

[81]  P. Goodford A computational procedure for determining energetically favorable binding sites on biologically important macromolecules. , 1985, Journal of medicinal chemistry.

[82]  Yiqun Cao,et al.  ChemMine tools: an online service for analyzing and clustering small molecules , 2011, Nucleic Acids Res..

[83]  D. di Bernardo,et al.  Computational drugs repositioning identifies inhibitors of oncogenic PI3K/AKT/P70S6K-dependent pathways among FDA-approved compounds , 2016, Oncotarget.

[84]  Christer S. Ejsing,et al.  Transformation-associated changes in sphingolipid metabolism sensitize cells to lysosomal cell death induced by inhibitors of acid sphingomyelinase. , 2013, Cancer cell.

[85]  Kathrin Heikamp,et al.  The Future of Virtual Compound Screening , 2013, Chemical biology & drug design.