191
Views
3
CrossRef citations to date
0
Altmetric
Reviews

Compound annotation with real time cellular activity profiles to improve drug discovery

Pages 269-280 | Received 10 Sep 2015, Accepted 14 Jan 2016, Published online: 03 Feb 2016

References

  • DiMasi JA, Hansen RW, Grabowski HG. The price of innovation: new estimates of drug development costs. J Health Econ. 2003;22:151–185.
  • DiMasi JA, Grabowski HG, Hansen RW. Innovation in the pharmaceutical industry: new estimates of R&D costs. Boston: Tufts Center for the Study of Drug Development, 2014 Nov 18. Available from: http://csdd.tufts.edu/files/uploads/cost_study_backgrounder.pdf.
  • Munos B. Lessons from 60 years of pharmaceutical innovation. Nat Rev Drug Discov. 2009;8:959–968.
  • Mullard A. FDA drug approvals. Nat Rev Drug Discov. 2014;2015(14):77–81.
  • Pammolli F, Magazzini L, Riccaboni M. The productivity crisis in pharmaceutical R&D. Nat Rev Drug Discov. 2011;10:428–438.
  • Hay M, Thomas DW, Craighead JL, et al. Clinical development success rates for investigational drugs. Nat Biotechnol. 2014;32:40–51.
  • Keyhani S, Diener-West M, Powe N. Are development times for pharmaceuticals increasing or decreasing? Health Aff. 2006;25:461–468.
  • Scannell JW. Diagnosing the decline in pharmaceutical R&D efficiency. Nat Rev Drug Discov. 2012;11:191–200.
  • Swinney DC, Anthony J. How were new medicines discovered? Nat Rev Drug Discov. 2011;10:507–519.
  • Lee JA, Uhlik MT, Moxham CM, et al. Modern phenotypic drug discovery is a viable, neoclassic pharma strategy. J Med Chem. 2012;55:4527–4538.
  • Eggert US. The why and how of phenotypic small-molecule screens. Nat Chem Biol. 2013;9:206–209.
  • Paolini GV, Shapland RH, Van Hoorn WP, et al. Global mapping of pharmacological space. Nat Biotechnol. 2006;24:805–815.
  • Yildirim MA, Goh K-I, Cusick ME, et al. Drug-target network. Nat Biotechnol. 2007;25:1119–1126.
  • Sirota M, Dudley JT, Kim J, et al. Discovery and preclinical validation of drug indications using compendia of public gene expression data. Sci Transl Med. 2011;3:96ra77–96ra77.
  • Szklarczyk D, Franceschini A, Wyder S, et al. STRING v10: protein-protein interaction networks, integrated over the tree of life. Nucleic Acids Res. 2015;43:D447–52.
  • Ananiadou S, Kell DB, Tsujii J. Text mining and its potential applications in systems biology. Trends Biotechnol. 2006;24:571–579.
  • Frijters R, Van Vugt M, Smeets R, et al. Literature mining for the discovery of hidden connections between drugs, genes and diseases. PLoS Comput Biol. 2010;6:e1000943.
  • Kingsmore SF, Lindquist IE, Mudge J, et al. Genome-wide association studies: progress and potential for drug discovery and development. Nat Rev Drug Discov. 2008;7:221–230.
  • Keiser MJ, Roth BL, Armbruster BN, et al. Relating protein pharmacology by ligand chemistry. Nat Biotechnol. 2007;25:197–206.
  • Keiser MJ, Setola V, Irwin JJ, et al. Predicting new molecular targets for known drugs. Nature. 2009;462:175–181.
  • Lipinski CA, Lombardo F, Dominy BW, et al. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv Drug Delivery Res. 1997;23:3–25.
  • Campillos M, Kuhn M, Gavin A-C, et al. Drug target identification using side-effect similarity. Science. 2008;321:263–266.
  • Lounkine E, Keiser MJ, Whitebread S, et al. Large-scale prediction and testing of drug activity on side-effect targets. Nature. 2012;486:361–367.
  • Lyne PD. Structure-based virtual screening: an overview. Drug Discov Today. 2002;7:1047–1055.
  • Kolb P, Ferreira RS, Irwin JJ, et al. Docking and chemoinformatic screens for new ligands and targets. Curr Opin Biotechnol. 2009;20:429–436.
  • Law V, Knox C, Djoumbou Y, et al. DrugBank 4.0: shedding new light on drug metabolism. Nucleic Acids Res. 2014;42:D1091–7.
  • Whirl-Carrillo M, McDonagh EM, Hebert JM, et al. Pharmacogenomics knowledge for personalized medicine. Clin Pharmacol Ther. 2012;92:414–417.
  • Davis AP, King BL, Mockus S, et al. The comparative toxicogenomics database: update 2011. Nucleic Acids Res. 2011;39:D1067–D1072.
  • Kuhn M, Campillos M, Letunic I, et al. A side effect resource to capture phenotypic effects of drugs. Mol Sys Biol. 2010;6:343.
  • Gaulton A, Bellis LJ, Bento AP, et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 2012;40:d1100–7.
  • Li Q, Cheng T, Wang Y, et al. PubChem as a public resource for drug discovery. Drug Discov Today. 2010;15:1052–1057.
  • Besnard J, Ruda GF, Setola V, et al. Automated design of ligands to polypharmacological profiles. Nature. 2012;492:215–220.
  • Irwin JJ, Shoichet BK. ZINC - a free database of commercially available compounds for virtual screening. J Chem Inf Model. 2005;45:177–182.
  • Fang Y. Combining label-free cell phenotypic profiling with computational approaches for novel drug discovery. Expert Opin Drug Discov. 2015;10:331–343.
  • Fang Y. The development of label-free cellular assays for drug discovery. Expert Opin Drug Discov. 2011;6:1285–1298.
  • Fang Y. Label-free biosensors for cell biology. Intl J Electrochem. 2011;2011:e460850.
  • Fang Y, editor. Label-free biosensor methods in drug discovery. Methods Pharmacol Tox. 2015;53:1–390.
  • Heller S, McNaught A, Stein S, et al. InChI - the worldwide chemical structure identifier standard. J Cheminform. 2013;5:7.
  • Chambers J, Davies M, Gaulton A, et al. UniChem: extension of InChI-based compound mapping to salt, connectivity and stereochemistry layers. J Cheminform. 2014;6:43.
  • Dolle RE. Historical overview of chemical library design. Methods Mol Biol. 2011;685:3–25.
  • Maggiora G, Vogt M, Stumpfe D, et al. Molecular similarity in medicinal chemistry. J Med Chem. 2014;57:3186–3204.
  • Keiser MJ, Irwin JJ, Shoichet BK. The chemical basis of pharmacology. Biochemistry. 2010;49:10267–10276.
  • Willett P. Similarity-based virtual screening using 2D fingerprints. Drug Discov Today. 2006;11:1046–1053.
  • Xie X-Q. Exploiting pubchem for virtual screening. Expert Opin Drug Discov. 2010;5:1205–1220.
  • Bolton EE, Kim S, Bryant SH. PubChem3D: similar conformers. J Cheminform. 201;3:13.
  • Willett P, Barnard JM, Downs GM. Chemical similarity searching. J Chem Inf Model. 1998;38:983–996.
  • Vieth M, Higgs RE, Robertson DH, et al. Kinomics-structural biology and chemogenomics of kinase inhibitors and targets. Biochim Biophys Acta. 2004;1697:243–257.
  • Izrailev S, Farnum MA. Enzyme classification by ligand binding. Proteins. 2004;57:711–724.
  • Gregori-Puigjané E, Setola V, Hert J, et al. Identifying mechanism-of-action targets for drugs and probes. Proc Natl Acad Sci USA. 2012;109:11178–11183.
  • Lipinski CA. Lead- and drug-like compounds: the rule-of-five revolution. Drug Discov Today Technol. 2004;1:337–341.
  • Veber DF, Johnson SR, Cheng H-Y, et al. Molecular properties that influence the oral bioavailability of drug candidates. J Med Chem. 2002;45:2615–2623.
  • Leeson PD, Springthorpe B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nat Rev Drug Discov. 2007;6:881–890.
  • Wang Y, Xiao J, Suzek TO, et al. PubChem: a public information system for analyzing bioactivities of small molecules. Nucleic Acids Res. 2009;37:W623–33.
  • Wang Y, Suzek T, Zhang J, et al. PubChem bioAssay: 2014 update. Nucleic Acids Res. 2014;42:D1075–82.
  • Kuhn M, Al Banchaabouchi M, Campillos M, et al. Systematic identification of proteins that elicit drug side effects. Mol Syst Biol. 2013;9:663.
  • Kuhn M, Letunic I, Jensen LJ, et al. The SIDER database of drugs and side effects. Nucleic Acids Res. 2015;pii:gkv1075. Epub ahead of print.
  • Fliri AF, Loging WT, Thadeio PF, et al. Analysis of drug-induced effect patterns to link structure and side effects of medicines. Nat Chem Biol. 2005;1:389–397.
  • Ravindranath AC, Perualila-Tan N, Kasim A, et al. Connecting gene expression data from connectivity map and in silico target predictions for small molecule mechanism-of-action analysis. Mol Biosyst. 2015;11:86–96.
  • Lum PY, Armour CD, Stepaniants SB, et al. Discovering modes of action for therapeutic compounds using a genome-wide screen of yeast heterozygotes. Cell. 2004;116:121–137.
  • Di Bernardo D, Thompson MJ, Gardner TS, et al. Chemogenomic profiling on a genome-wide scale using reverse-engineered gene networks. Nat Biotechnol. 2005;23(3):377–383.
  • Lamb J, Crawford ED, Peck D, et al. The connectivity map: using gene-expression signatures to connect small molecules, genes, and disease. Science. 2006;313:1929–1935.
  • Lamb J. The connectivity map: a new tool for biomedical research. Nat Rev Cancer. 2007;7:54–60.
  • McArt DG, Dunne PD, Blayney JK, et al. Connectivity mapping for candidate therapeutics identification using next generation sequencing rna-seq data. PLoS One. 2013;8:e66902.
  • Jain AK, Murty MN, Flynn PJ. Data clustering: a review. ACM Comput Surv. 1999;31:264–323.
  • Dalton L, Ballarin V, Brun M. Clustering algorithms: on learning, validation, performance, and applications to genomics. Curr Genomics. 2009;10:430–445.
  • Fang Y. Label-free drug discovery. Front Pharmacol. 2014;5:52.
  • Fang Y. Label-free cell phenotypic drug discovery. Comb Chem High Throughput Screen. 2014;17:566–578.
  • Shoemaker RH. The NCI60 human tumour cell line anticancer drug screen. Nat Rev Cancer. 2006;6:813–823.
  • Barretina J, Caponigro G, Stransky N, et al. The cancer cell line encyclopedia enables predictive modelling of anticancer drug sensitivity. Nature. 2012;483:603–607.
  • Febles NK, Ferrie AM, Fang Y. Label-free single cell quantification of the invasion of spheroidal colon cancer cells through 3D matrigel. Anal Chem. 2014;86:8842–8849.
  • Chandrasekaran S, Deng H, Fang Y. PTEN deletion potentiates invasion of colorectal cancer spheroidal cells through 3D matrigel. Integr Biol. 2015;7:324–4.
  • Ashburner M, Ball CA, Blake JA, et al. Gene ontology: tool for the unification of biology. The gene ontology consortium. Nat Genet. 2000;25:25–29.
  • Smith B, Ashburner M, Rosse C, et al. The OBO foundry: coordinated evolution of ontologies to support biomedical data integration. Nat Biotechnol. 2007;25:1251–1255.
  • Whetzel PL, Noy NF, Shah NH, et al. BioPortal: enhanced functionality via new web services from the National Center for Biomedical Ontology to access and use ontologies in software applications. Nucleic Acids Res. 2011;39:W541–5.
  • Natale DA, Arighi CN, Blake JA, et al. Protein ontology: a controlled structured network of protein entities. Nucleic Acids Res. 2014;42:D415–21.
  • De Matos P, Adams N, Hastings J, et al. A database for chemical proteomics: ChEBI. Methods Mol Biol. 2012;803:273–296.
  • Kibbe WA, Arze C, Felix V, et al. Disease ontology 2015 update: an expanded and updated database of human diseases for linking biomedical knowledge through disease data. Nucleic Acids Res. 2015;43:D1071–8.
  • Visser U, Abeyruwan S, Vempati U, et al. BioAssay ontology (BAO): a semantic description of bioassays and high-throughput screening results. BMC Bioinformatics. 2011;12:257.
  • Abeyruwan U, Vempati H, Küçük U, et al. Evolving bioassay ontology (BAO): modularization, integration and applications. J Biomed Semantics. 2014;5:S5.
  • Vempati UD, Przydzial MJ, Chung C, et al. Formalization, annotation and analysis of diverse drug and probe screening assay datasets using the bioassay ontology (BAO). PLoS One. 2012;7:e49198.
  • Lau J, Ioannidis JP, Schmid CH. Quantitative synthesis in systematic reviews. Ann Intern Med. 1997;127:820–826.
  • Crowther M, Lim W, Crowther MA. Systematic review and meta-analysis methodology. Blood. 2010;116:3140–3146.
  • Evangelou E, Ioannidis JP. Meta-analysis methods for genome-wide association studies and beyond. Nat Rev Genet. 2013;14:379–389.
  • Campbell SJ, Gaulton A, Marshall J, et al. Visualizing the drug target landscape. Drug Discov Today. 2010;15:3–15.
  • Kenakin T. Cellular assays as portals to seven-transmembrane receptor-based drug discovery. Nat Rev Drug Discov. 2009;8:617–626.
  • Fang Y. Troubleshooting and deconvoluting label-free cell phenotypic assays in drug discovery. J Pharmacol Tox Methods. 2013;67:69–81.
  • Fang Y, Ferrie AM. Label-free optical biosensor for ligand-directed functional selectivity acting on β2-adrenoceptor in living cells. FEBS Lett. 2008;582:558–564.
  • Ferrie AM, Goral V, Wang C, et al. Label-free functional selectivity assays. Methods Mol Biol. 2015;1272:227–246.
  • Deng H, Wang C, Su M, et al. Probing biochemical mechanisms of action of muscarinic M3 receptor antagonists with label-free whole-cell assays. Anal Chem. 2012;84:8232–8239.
  • Guo D, Mulder-Krieger T, Ijzerman AP, et al. Functional efficacy of adenosine <sub>A₂A receptor agonists is positively correlated to their receptor residence time.. Br J Pharmacol. 2012;166:1846–1959.
  • Deng H, Sun H, Fang Y. Label-free cell phenotypic assessment of the biased agonism and efficacy of agonists at the endogenous muscarinic M3 receptors. J Pharmacol Tox Methods. 2013;68:323–333.
  • Bock A, Chirinda B, Krebs F, et al. Dynamic ligand binding dictates partial agonism at a G protein-coupled receptor. Nat Chem Biol. 2014;10:18–20.
  • Fang Y, Li G, Ferrie AM. Non-invasive optical biosensor for assaying endogenous G protein-coupled receptors in adherent cells. J Pharmacol Tox Methods. 2007;55:314–322.
  • Fang Y. Probing cancer signaling with resonant waveguide grating biosensors. Expert Opin Drug Discov. 2010;5:1237–1248.
  • Schröder R, Janssen N, Schmidt J, et al. Deconvolution of complex G protein-coupled receptor signaling in live cells using dynamic mass redistribution measurements. Nat Biotechnol. 2010;28:943–949.
  • Fang Y. Label-free receptor assays. Drug Discov Today Technol. 2010;7:e5–e11.
  • Fang Y. Label-free cell phenotypic profiling and screening: techniques, experimental design and data assessment. Methods Pharmacol Tox. 2015;53:233–252.
  • Ferrie AM, Sun H, Fang Y. Label-free integrative pharmacology on-target of drugs at the β2-adrenergic receptor. Sci Rep. 2011;1:33.
  • Morse M, Tran E, Levension RL, et al. Ligand-directed functional selectivity at the mu opioid receptor revealed by label-free on-target pharmacology. PLoS One. 2011;6:e25643.
  • Morse M, Sun H, Tran E, et al. Label-free integrative pharmacology ontarget of opioid ligands at the opioid receptor family. BMC Pharmacol Tox. 2013;14:17.
  • Ferrie AM, Sun H, Zaytseva N, et al. Divergent label-free cell phenotypic pharmacology of ligands at the overexpressed β2-adrenergic receptors. Sci Rep. 2014;4:3828.
  • Ferrie AM, Wang C, Deng H, et al. A label-free optical biosensor with microfluidics identifies an intracellular signalling wave mediated through the β2-adrenergic receptor. Integr Biol. 2013;5:1253–1261.
  • Pai S, Verrier F, Sun H, et al. Dynamic mass redistribution assay decodes differentiation of a neural progenitor stem cell. J Biomol Screen. 2012;17:1180–1191.
  • Sun H, Wei Y, Xiong Q, et al. Label-free cell phenotypic profiling decodes the composition and signaling of an endogenous ATP-sensitive potassium channel. Sci Rep. 2014;4:4934.
  • Ferrie AM, Deichmann OD, Wu Q, et al. High resolution resonant waveguide grating imager for cell cluster analysis under physiological condition. Appl Phys Lett. 2012;100:223701.
  • Kholodenko BN. Cell-signalling dynamics in time and space. Nat Rev Mol Cell Biol. 2006;7:165–176.
  • Lohse MJ, Calebiro D. Cell biology: receptor signals come in waves. Nature. 2013;495:457–458.
  • Irannejad R, Tomshine JC, Tomshine JR, et al. Conformational biosensors reveal GPCR signalling from endosomes. Nature. 2013;495:534–538.
  • Abassi YA, Xi B, Li N, et al. Dynamic monitoring of beating periodicity of stem cell-derived cardiomyocytes as a predictive tool for preclinical safety assessment. Br J Pharmacol. 2012;165:1424–1441.
  • Ferrie AM, Wu Q, Deichmann O, et al. High frequency resonant waveguide grating imager for assessing drug-induced cardiotoxicity. Appl Phys Lett. 2014;104:183702.
  • Zhang X, Deng H, Xiao Y, et al. Label-free cell phenotypic profiling identifies pharmacologically active compounds in two traditional Chinese medicinal plants. RSC Adv. 2014;4:26368–26377.
  • Abassi YA, Xi B, Zhang W, et al. Kinetic cell-based morphological screening: prediction of mechanism of compound action and off-target effects. Chem Biol. 2009;16:712–723.
  • Fu H, Fu W, Sun M, et al. Kinetic cellular phenotypic profiling: prediction, identification, and analysis of bioactive natural products. Anal Chem. 2011;83:6518–6526.
  • Deng H, Hu H, He M, et al. Discovery of 2-(4-methylfuran-2(5H)-ylidene)malononitrile and thieno[3,2-b]thiophene-2-carboxylic acid derivatives as G protein-coupled receptor-35 (GPR35) agonists. J Med Chem. 2011;54:7385–7396.
  • Deng H, Hu H, Fang Y. Multiple tyrosine metabolites are GPR35 agonists. Sci Rep. 2012;2:373.
  • Deng H, Fang Y. Discovery of nitrophenols as GPR35 agonists. Med Chem Comm. 2012;3:1270–1274.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.