Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Large-scale prediction and testing of drug activity on side-effect targets

Abstract

Discovering the unintended ‘off-targets’ that predict adverse drug reactions is daunting by empirical methods alone. Drugs can act on several protein targets, some of which can be unrelated by conventional molecular metrics, and hundreds of proteins have been implicated in side effects. Here we use a computational strategy to predict the activity of 656 marketed drugs on 73 unintended ‘side-effect’ targets. Approximately half of the predictions were confirmed, either from proprietary databases unknown to the method or by new experimental assays. Affinities for these new off-targets ranged from 1 nM to 30 μM. To explore relevance, we developed an association metric to prioritize those new off-targets that explained side effects better than any known target of a given drug, creating a drug–target–adverse drug reaction network. Among these new associations was the prediction that the abdominal pain side effect of the synthetic oestrogen chlorotrianisene was mediated through its newly discovered inhibition of the enzyme cyclooxygenase-1. The clinical relevance of this inhibition was borne out in whole human blood platelet aggregation assays. This approach may have wide application to de-risking toxicological liabilities in drug discovery.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Figure 1: Predicting off-targets, and their novelty.
Figure 2: Off-target networks.
Figure 3: Target and drug promiscuity.

Similar content being viewed by others

References

  1. Giacomini, K. M. et al. When good drugs go bad. Nature 446, 975–977 (2007)

    Article  ADS  CAS  Google Scholar 

  2. Arrowsmith, J. Trial watch: phase III and submission failures: 2007–2010. Nature Rev. Drug Discov. 10, 87 (2011)

    Article  CAS  Google Scholar 

  3. Arrowsmith, J. Trial watch: phase II failures: 2008–2010. Nature Rev. Drug Discov. 10, 328–329 (2011)

    Article  CAS  Google Scholar 

  4. Boyer, S. The use of computer models in pharmaceutical safety evaluation. Altern. Lab. Anim. 37, 467–475 (2009)

    CAS  PubMed  Google Scholar 

  5. Wong, D., Wang, M., Cheng, Y. & Fitzgerald, G. A. Cardiovascular hazard and non-steroidal anti-inflammatory drugs. Curr. Opin. Pharmacol. 5, 204–210 (2005)

    Article  CAS  Google Scholar 

  6. Antunes, A. M. M. et al. Protein adducts as prospective biomarkers of nevirapine toxicity. Chem. Res. Toxicol. 23, 1714–1725 (2010)

    Article  CAS  Google Scholar 

  7. Rothman, R. B. et al. Evidence for possible involvement of 5-HT2B receptors in the cardiac valvulopathy associated with fenfluramine and other serotonergic medications. Circulation 102, 2836–2841 (2000)

    Article  CAS  Google Scholar 

  8. Roy, M., Dumaine, R. & Brown, A. M. HERG, a primary human ventricular target of the nonsedating antihistamine terfenadine. Circulation 94, 817–823 (1996)

    Article  CAS  Google Scholar 

  9. Curran, M. E. et al. A molecular basis for cardiac arrhythmia: HERG mutations cause long QT syndrome. Cell 80, 795–803 (1995)

    Article  CAS  Google Scholar 

  10. Ji, Z. L. et al. Drug Adverse Reaction Target Database (DART): proteins related to adverse drug reactions. Drug Saf. 26, 685–690 (2003)

    Article  CAS  Google Scholar 

  11. Kuhn, M., Campillos, M., Letunic, I., Jensen, L. J. & Bork, P. A side effect resource to capture phenotypic effects of drugs. Mol. Syst. Biol. 6, 343 (2010)

    Article  Google Scholar 

  12. Matthews, E. J. & Frid, A. A. Prediction of drug-related cardiac adverse effects in humans–A: Creation of a database of effects and identification of factors affecting their occurrence. Regul. Toxicol. Pharmacol. 56, 247–275 (2010)

    Article  CAS  Google Scholar 

  13. Yang, X. et al. Kinase inhibition-related adverse events predicted from in vitro kinome and clinical trial data. J. Biomed. Inform. 43, 376–384 (2010)

    Article  CAS  Google Scholar 

  14. Hopkins, A. L. Network pharmacology: the next paradigm in drug discovery. Nature Chem. Biol. 4, 682–690 (2008)

    Article  CAS  Google Scholar 

  15. Zhang, J.-X. et al. DITOP: drug-induced toxicity related protein database. Bioinformatics 23, 1710–1712 (2007)

    Article  CAS  Google Scholar 

  16. Yang, L., Luo, H., Chen, J., Xing, Q. & He, L. SePreSA: a server for the prediction of populations susceptible to serious adverse drug reactions implementing the methodology of a chemical-protein interactome. Nucleic Acids Res. 37, W406–W412 (2009)

    Article  CAS  Google Scholar 

  17. Lee, S., Lee, K. H., Song, M. & Lee, D. Building the process-drug-side effect network to discover the relationship between biological processes and side effects. BMC Bioinformatics 12 (suppl. 2). S2 (2011)

    Article  Google Scholar 

  18. Schreiber, G. & Keating, A. E. Protein binding specificity versus promiscuity. Curr. Opin. Struct. Biol. 21, 50–61 (2011)

    Article  CAS  Google Scholar 

  19. Oprea, T. I. et al. Associating drugs, targets and clinical outcomes into an integrated network affords a new platform for computer-aided drug repurposing. Mol. Inform. 30, 100–111 (2011)

    Article  CAS  Google Scholar 

  20. Paolini, G. V., Shapland, R. H. B., van Hoorn, W. P., Mason, J. S. & Hopkins, A. L. Global mapping of pharmacological space. Nature Biotechnol. 24, 805–815 (2006)

    Article  CAS  Google Scholar 

  21. Scheiber, J. et al. Mapping adverse drug reactions in chemical space. J. Med. Chem. 52, 3103–3107 (2009)

    Article  CAS  Google Scholar 

  22. Bender, A. et al. Analysis of pharmacology data and the prediction of adverse drug reactions and off-target effects from chemical structure. ChemMedChem 2, 861–873 (2007)

    Article  CAS  Google Scholar 

  23. Campillos, M., Kuhn, M., Gavin, A.-C., Jensen, L. J. & Bork, P. Drug target identification using side-effect similarity. Science 321, 263–266 (2008)

    Article  ADS  CAS  Google Scholar 

  24. Tatonetti, N. P. et al. Detecting drug interactions from adverse-event reports: interaction between paroxetine and pravastatin increases blood glucose levels. Clin. Pharmacol. Ther. 90, 133–142 (2011)

    Article  CAS  Google Scholar 

  25. Keiser, M. J. et al. Predicting new molecular targets for known drugs. Nature 462, 175–181 (2009)

    Article  ADS  CAS  Google Scholar 

  26. Keiser, M. J. et al. Relating protein pharmacology by ligand chemistry. Nature Biotechnol. 25, 197–206 (2007)

    Article  CAS  Google Scholar 

  27. Hert, J., Keiser, M. J., Irwin, J. J., Oprea, T. I. & Shoichet, B. K. Quantifying the relationships among drug classes. J. Chem. Inf. Model. 48, 755–765 (2008)

    Article  CAS  Google Scholar 

  28. Azzaoui, K. et al. Modeling promiscuity based on in vitro safety pharmacology profiling data. ChemMedChem 2, 874–880 (2007)

    Article  CAS  Google Scholar 

  29. Gaulton, A. et al. ChEMBL: a large-scale bioactivity database for drug discovery. Nucleic Acids Res. 40, D1100–D1107 10.1093/nar/gkr777. (2012)

    Article  Google Scholar 

  30. Laggner, C. et al. Chemical informatics and target identification in a zebrafish phenotypic screen. Nature Chem. Biol. 8, 144–146 (2012)

    Article  CAS  Google Scholar 

  31. Shelley, J. C. et al. Epik: a software program for pK a prediction and protonation state generation for drug-like molecules. J. Comput. Aided Mol. Des. 21, 681–691 (2007)

    Article  ADS  CAS  Google Scholar 

  32. Muchmore, S. W. et al. Application of belief theory to similarity data fusion for use in analog searching and lead hopping. J. Chem. Inf. Model. 48, 941–948 (2008)

    Article  CAS  Google Scholar 

  33. Yıldırım, M. A., Goh, K.-I., Cusick, M. E., Barabási, A.-L. & Vidal, M. Drug–target network. Nature Biotechnol. 25, 1119–1126 (2007)

    Article  Google Scholar 

  34. Marshall, V. & Grosset, D. G. Role of dopamine transporter imaging in the diagnosis of atypical tremor disorders. Mov. Disord. 18 (suppl. 7). S22–S27 (2003)

    Article  Google Scholar 

  35. Kuo, C. C., Huang, R. C. & Lou, B. S. Inhibition of Na+ current by diphenhydramine and other diphenyl compounds: molecular determinants of selective binding to the inactivated channels. Mol. Pharmacol. 57, 135–143 (2000)

    CAS  PubMed  Google Scholar 

  36. Schoen, R. T. & Vender, R. J. Mechanisms of nonsteroidal anti-inflammatory drug-induced gastric damage. Am. J. Med. 86, 449–458 (1989)

    Article  CAS  Google Scholar 

  37. Kong, S. X., Hatoum, H. T., Zhao, S. Z., Agrawal, N. M. & Geis, S. G. Prevalence and cost of hospitalization for gastrointestinal complications related to peptic ulcers with bleeding or perforation: comparison of two national databases. Am. J. Manag. Care 4, 399–409 (1998)

    CAS  PubMed  Google Scholar 

  38. G, Perrone, M., Scilimati, A., Simone, L. & Vitale, P. Selective COX-1 inhibition: A therapeutic target to be reconsidered. Curr. Med. Chem. 17, 3769–3805 (2010)

    Article  Google Scholar 

  39. Akarasereenont, P., Tripatara, P., Chotewuttakorn, S., Palo, T. & Thaworn, A. The effects of estrone, estradiol and estriol on platelet aggregation induced by adrenaline and adenosine diphosphate. Platelets 17, 441–447 (2006)

    Article  CAS  Google Scholar 

  40. Norris, L. A. & Bonnar, J. Effect of oestrogen dose on whole blood platelet activation in women taking new low dose oral contraceptives. Thromb. Haemost. 72, 926–930 (1994)

    Article  CAS  Google Scholar 

  41. Leeson, P. D. & Springthorpe, B. The influence of drug-like concepts on decision-making in medicinal chemistry. Nature Rev. Drug Discov. 6, 881–890 (2007)

    Article  CAS  Google Scholar 

  42. Peters, J.-U., Schnider, P., Mattei, P. & Kansy, M. Pharmacological promiscuity: dependence on compound properties and target specificity in a set of recent Roche compounds. ChemMedChem 4, 680–686 (2009)

    Article  CAS  Google Scholar 

  43. Cosgrove, B. D. et al. Cytokine-associated drug toxicity in human hepatocytes is associated with signaling network dysregulation. Mol. Biosyst. 6, 1195–1206 (2010)

    Article  CAS  Google Scholar 

  44. Mestres, J., Gregori-Puigjané, E., Valverde, S. & Solé, R. V. Data completeness–the Achilles heel of drug-target networks. Nature Biotechnol. 26, 983–984 (2008)

    Article  CAS  Google Scholar 

  45. Mestres, J., Gregori-Puigjané, E., Valverde, S. & Solé, R. V. The topology of drug-target interaction networks: implicit dependence on drug properties and target families. Mol. Biosyst. 5, 1051–1057 (2009)

    Article  CAS  Google Scholar 

  46. Rogers, D. & Hahn, M. Extended-connectivity fingerprints. J. Chem. Inf. Model. 50, 742–754 (2010)

    Article  CAS  Google Scholar 

  47. James, C., Weininger, D. & Delany, J. Daylight Theory Manual — Daylight 4.91 (Daylight Chemical Information Systems, 2005)

    Google Scholar 

  48. Wishart, D. S. et al. DrugBank: a knowledgebase for drugs, drug actions and drug targets. Nucleic Acids Res. 36, D901–D906 (2008)

    Article  CAS  Google Scholar 

  49. Stein, S., Heller, S. & Tchekhovski, D. An open standard for chemical structure representation — the IUPAC chemical identifier. Nimes Int. Chem. Inf. Conf. Proc. 143, 131–143 (2003)

    Google Scholar 

  50. The UniProt Consortium. Ongoing and future developments at the Universal Protein Resource. Nucleic Acids Res. 39, D214–D219 (2010)

  51. Altschul, S. F. et al. Gapped BLAST and PSI-BLAST: a new generation of protein database search programs. Nucleic Acids Res. 25, 3389–3402 (1997)

    Article  CAS  Google Scholar 

  52. Brown, E. G., Wood, L. & Wood, S. The medical dictionary for regulatory activities (MedDRA). Drug Saf. 20, 109–117 (1999)

    Article  CAS  Google Scholar 

  53. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. B 57, 289–300 (1995)

    MathSciNet  MATH  Google Scholar 

Download references

Acknowledgements

E.L. is a presidential postdoctoral fellow supported by the Education Office of the Novartis Institutes for Biomedical Research (co-mentors L.U. and B.K.S.). Supported by US National Institutes of Health grants GM71896 (to B.K.S. and J. Irwin), AG002132 (to S. Prusiner and B.K.S.), and GM93456 (to M.J.K.), and by QB3 Rogers Family Foundation ‘Bridging-the-Gap’ Award (to M.J.K.).

Author information

Authors and Affiliations

Authors

Contributions

SEA calculations were undertaken by M.J.K. Target–ADR associations, networks and promiscuity analysis were by E.L. In vitro assays were directed by S.W., J.H. and L.U. PK and PD experiments were conducted by E.W. and P.L. Platelet aggregation study was designed and carried out by L.U. and S.C. Chlorotrianisene solubility and aggregation were conducted by A.K.D. The project was conceived and planned by B.K.S., J.J., D.M. and L.U. Overall analysis and writing was largely by E.L., M.J.K., B.K.S. and L.U. All authors contributed to the manuscript.

Corresponding authors

Correspondence to Jeremy L. Jenkins, Brian K. Shoichet or Laszlo Urban.

Ethics declarations

Competing interests

B.K.S. is a founder of SeaChange Pharmaceuticals Inc, a company that uses the SEA technology. The other authors have no competing interests to declare.

Supplementary information

Supplementary Information

This file contains Supplementary Methods, Supplementary Results, Supplementary References, Supplementary Figure 1 and legends for Supplementary Tables 1-8 (see separate file for tables). (PDF 824 kb)

Supplementary Tables

This file contains Supplementary Tables 1-8 (see Supplementary Information file for legends). Each sheet in the workbook corresponds to one table. (XLS 832 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Lounkine, E., Keiser, M., Whitebread, S. et al. Large-scale prediction and testing of drug activity on side-effect targets. Nature 486, 361–367 (2012). https://doi.org/10.1038/nature11159

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature11159

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research