Skip to main content
Log in

Physicochemical Properties, Biotransformation, and Transport Pathways of Established and Newly Approved Medications: A Systematic Review of the Top 200 Most Prescribed Drugs vs. the FDA-Approved Drugs Between 2005 and 2016

  • Systematic Review
  • Published:
Clinical Pharmacokinetics Aims and scope Submit manuscript

Abstract

Background

Enzyme-mediated biotransformation of pharmacological agents is a crucial step in xenobiotic detoxification and drug disposition. Herein, we investigated the metabolism and physicochemical properties of the top 200 most prescribed drugs (established) as well as drugs approved by the US Food and Drug Administration (FDA) between 2005 and 2016 (newly approved).

Objective

Our objective was to capture the changing trends in the routes of administration, physicochemical properties, and prodrug medications, as well as the contributions of drug-metabolizing enzymes and transporters to drug clearance.

Methods

The University of Washington Drug Interaction Database (DIDB®) as well as other online resources (e.g., CenterWatch.com, Drugs.com, DrugBank.ca, and PubChem.ncbi.nlm.nih.gov) was used to collect and stratify the dataset required for exploring the above-mentioned trends.

Results

Analyses revealed that ~ 90% of all drugs in the established and newly approved drug lists were administered systemically (oral or intravenous). Meanwhile, the portion of biologics (molecular weight > 1 kDa) was 15 times greater in the newly approved list than established drugs. Additionally, there was a 4.5-fold increase in the number of compounds with a high calculated partition coefficient (cLogP > 3) and a high total polar surface area (> 75 Å2) in the newly approved drug vs. the established category. Further, prodrugs in established or newly approved lists were found to be converted to active compounds via hydrolysis, demethylases, and kinases. The contribution of cytochrome P450 (CYP) 3A4, as the major biotransformation pathway, has increased from 40% in the established drug list to 64% in the newly approved drug list. Moreover, the role of CYP1A2, CYP2C19, and CYP2D6 were decreased as major metabolizing enzymes among the newly approved medications. Among non-CYP major metabolizers, the contribution of alcohol dehydrogenases/aldehyde dehydrogenases (ADH/ALDH) and sulfotransferases decreased in the newly approved drugs compared with the established list. Furthermore, the highest contribution among uptake and efflux transporters was found for Organic Anion Transporting Polypeptide 1B1 (OATP1B1) and P-glycoprotein (P-gp), respectively.

Conclusions

The higher portion of biologics in the newly approved drugs compared with the established list confirmed the growing demands for protein- and antibody-based therapies. Moreover, the larger number of hydrophilic drugs found in the newly approved list suggests that the probability of toxicity is likely to decrease. With regard to CYP-mediated major metabolism, CYP3A5 showed an increased involvement owing to the identification of unique probe substrates to differentiate CYP3As. Furthermore, the contribution of OATP1B1 and P-gp did not show a significant shift in the newly approved drugs as compared to the established list because of their broad substrate specificity.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6

Similar content being viewed by others

References

  1. Oda S, Fukami T, Yokoi T, Nakajima M. A comprehensive review of UDP-glucuronosyltransferase and esterases for drug development. Drug Metab Pharmacokinet. 2015;30(1):30–51.

    Article  CAS  PubMed  Google Scholar 

  2. Iyanagi T. Molecular mechanism of phase I and phase II drug-metabolizing enzymes: implications for detoxification. Int Rev Cytol. 2007;260:35–112.

    Article  CAS  PubMed  Google Scholar 

  3. Williams JA, Hyland R, Jones BC, Smith DA, Hurst S, Goosen TC, et al. Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metab Dispos. 2004;32(11):1201–8.

    Article  CAS  PubMed  Google Scholar 

  4. Nelson DR, Zeldin DC, Hoffman SM, Maltais LJ, Wain HM, Nebert DW. Comparison of cytochrome P450 (CYP) genes from the mouse and human genomes, including nomenclature recommendations for genes, pseudogenes and alternative-splice variants. Pharmacogenetics. 2004;14(1):1–18.

    Article  CAS  PubMed  Google Scholar 

  5. Nebert DW, Russell DW. Clinical importance of the cytochromes P450. Lancet. 2002;360(9340):1155–62.

    Article  CAS  PubMed  Google Scholar 

  6. Sim SC, Ingelman-Sundberg M. Update on allele nomenclature for human cytochromes P450 and the human cytochrome P450 allele (CYP-allele) nomenclature database. Methods Mol Biol. 2013;987:251–9.

    Article  CAS  PubMed  Google Scholar 

  7. Walsky RL, Obach RS. Validated assays for human cytochrome P450 activities. Drug Metab Dispos. 2004;32(6):647–60.

    Article  CAS  PubMed  Google Scholar 

  8. Wienkers LC, Heath TG. Predicting in vivo drug interactions from in vitro drug discovery data. Nat Rev Drug Discov. 2005;4(10):825–33.

    Article  CAS  PubMed  Google Scholar 

  9. Guengerich FP, Cheng Q. Orphans in the human cytochrome P450 superfamily: approaches to discovering functions and relevance in pharmacology. Pharmacol Rev. 2011;63(3):684–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Kola I, Landis J. Can the pharmaceutical industry reduce attrition rates? Nat Rev Drug Discov. 2004;3(8):711–5.

    Article  CAS  PubMed  Google Scholar 

  11. Arrowsmith J, Miller P. Trial watch: phase II and phase III attrition rates 2011–2012. Nat Rev Drug Discov. 2013;12(8):569. https://doi.org/10.1038/nrd4090.

    Article  CAS  PubMed  Google Scholar 

  12. Gan J, Ma S, Zhang D. Non-cytochrome P450-mediated bioactivation and its toxicological relevance. Drug Metab Rev. 2016;48(4):473–501.

    Article  CAS  PubMed  Google Scholar 

  13. Foti RS, Dalvie DK. Cytochrome P450 and non-cytochrome P450 oxidative metabolism: contributions to the pharmacokinetics, safety and efficacy of xenobiotics. Drug Metab Dispos. 2016;44(8):1229–45.

    Article  CAS  PubMed  Google Scholar 

  14. Cerny MA. Prevalence of non-cytochrome P450-mediated metabolism in food and drug administration-approved oral and intravenous drugs: 2006–2015. Drug Metab Dispos. 2016;44(8):1246–52.

    Article  CAS  PubMed  Google Scholar 

  15. Pryde DC, Dalvie D, Hu Q, Jones P, Obach RS, Tran TD. Aldehyde oxidase: an enzyme of emerging importance in drug discovery. J Med Chem. 2010;53(24):8441–60.

    Article  CAS  PubMed  Google Scholar 

  16. Leeson PD. Molecular inflation, attrition and the rule of five. Adv Drug Deliv Rev. 2016;101:22–33.

    Article  CAS  PubMed  Google Scholar 

  17. DeGorter MK, Xia CQ, Yang JJ, Kim RB. Drug transporters in drug efficacy and toxicity. Annu Rev Pharmacol Toxicol. 2012;52:249–73.

    Article  CAS  PubMed  Google Scholar 

  18. International Transporter Consortium, Giacomini KM, Huang SM, Tweedie DJ, Benet LZ, Brouwer KL, et al. Membrane transporters in drug development. Nat Rev Drug Discov. 2010;9(3):215–36.

  19. Kusuhara H, Sugiyama Y. In vitro-in vivo extrapolation of transporter-mediated clearance in the liver and kidney. Drug Metab Pharmacokinet. 2009;24(1):37–52.

    Article  CAS  PubMed  Google Scholar 

  20. Shitara Y, Horie T, Sugiyama Y. Transporters as a determinant of drug clearance and tissue distribution. Eur J Pharm Sci. 2006;27(5):425–46.

    Article  CAS  PubMed  Google Scholar 

  21. Symphony Health Solutions. 2015. http://symphonyhealth.com/wp-content/uploads/2015/05/Top-200-Drugs-of-2014.pdf. Accessed 18 Dec 2017.

  22. CenterWatch. 2018. Available from: http://www.centerwatch.com/drug-information/fda-approved-drugs/year. Accessed Mar 2018.

  23. Price DA, Blagg J, Jones L, Greene N, Wager T. Physicochemical drug properties associated with in vivo toxicological outcomes: a review. Expert Opin Drug Metab Toxicol. 2009;5(8):921–31.

    Article  CAS  PubMed  Google Scholar 

  24. University of Washington Metabolism and Transport Drug Interaction Database. 2019. https://sop.washington.edu/department-of-pharmaceutics/research/metabolism-and-transport-drug-interaction-database. Accessed Feb 2019.

  25. US Department of Health and Human Services, Food and Drug Administration, Center for Drug Evaluation and Research (CDER). 2017. Draft guidance for industry. In vitro metabolism and transporter mediated drug-drug interactions. https://www.fda.gov/downloads/Drugs/Guidances/UCM581965.pdf. Accessed Feb 2019.

  26. Hachad H, Ragueneau-Majlessi I, Levy RH. A useful tool for drug interaction evaluation: the University of Washington Metabolism and Transport Drug Interaction Database. Hum Genomics. 2010;5(1):61–72.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Yu J, Ritchie TK, Mulgaonkar A, Ragueneau-Majlessi I. Drug disposition and drug-drug interaction data in 2013 FDA new drug applications: a systematic review. Drug Metab Dispos. 2014;42(12):1991–2001.

    Article  PubMed  Google Scholar 

  28. Yu J, Ritchie TK, Zhou Z, Ragueneau-Majlessi I. Key findings from preclinical and clinical drug interaction studies presented in new drug and biological license applications approved by the Food and Drug Administration in 2014. Drug Metab Dispos. 2016;44(1):83–101.

    Article  CAS  PubMed  Google Scholar 

  29. Yu J, Zhou Z, Owens KH, Ritchie TK, Ragueneau-Majlessi I. What can be learned from recent new drug applications? A systematic review of drug interaction data for drugs approved by the US FDA in 2015. Drug Metab Dispos. 2017;45(1):86–108.

    Article  CAS  PubMed  Google Scholar 

  30. Tseng E, Walsky RL, Luzietti RA, Harris JJ, Kosa RE, Goosen TC, et al. Relative contributions of cytochrome CYP3A4 versus CYP3A5 for CYP3A-cleared drugs assessed in vitro using a CYP3A4-selective inactivator (CYP3cide). Drug Metab Dispos. 2014;42(7):1163–73.

    Article  PubMed  Google Scholar 

  31. Chen SY, Li JL, Meng FH, Wang XD, Liu T, Li J, et al. Individualization of tacrolimus dosage basing on cytochrome P450 3A5 polymorphism: a prospective, randomized, controlled study. Clin Transplant. 2013;27(3):E272–81.

    Article  CAS  PubMed  Google Scholar 

  32. Thervet E, Anglicheau D, King B, Schlageter MH, Cassinat B, Beaune P, et al. Impact of cytochrome P450 3A5 genetic polymorphism on tacrolimus doses and concentration-to-dose ratio in renal transplant recipients. Transplantation. 2003;76(8):1233–5.

    Article  CAS  PubMed  Google Scholar 

  33. McEuen K, Borlak J, Tong W, Chen M. Associations of drug lipophilicity and extent of metabolism with drug-induced liver injury. Int J Mol Sci. 2017;18(7):1335.

    Article  PubMed Central  Google Scholar 

  34. Leeson PD, Davis AM. Time-related differences in the physical property profiles of oral drugs. J Med Chem. 2004;47(25):6338–48.

    Article  CAS  PubMed  Google Scholar 

  35. Amidon GL, Lennernas H, Shah VP, Crison JR. A theoretical basis for a biopharmaceutic drug classification: the correlation of in vitro drug product dissolution and in vivo bioavailability. Pharm Res. 1995;12(3):413–20.

    Article  CAS  PubMed  Google Scholar 

  36. Reichert JM. Trends in development and approval times for new therapeutics in the United States. Nat Rev Drug Discov. 2003;2(9):695–702.

    Article  CAS  PubMed  Google Scholar 

  37. Yang X, Gandhi YA, Duignan DB, Morris ME. Prediction of biliary excretion in rats and humans using molecular weight and quantitative structure-pharmacokinetic relationships. AAPS J. 2009;11(3):511–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Hosey CM, Broccatelli F, Benet LZ. Predicting when biliary excretion of parent drug is a major route of elimination in humans. AAPS J. 2014;16(5):1085–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rendic S, Guengerich FP. Survey of human oxidoreductases and cytochrome P450 enzymes involved in the metabolism of xenobiotic and natural chemicals. Chem Res Toxicol. 2015;28(1):38–42.

    Article  CAS  PubMed  Google Scholar 

  40. Roth M, Obaidat A, Hagenbuch B. OATPs, OATs and OCTs: the organic anion and cation transporters of the SLCO and SLC22A gene superfamilies. Br J Pharmacol. 2012;165(5):1260–87.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kalliokoski A, Niemi M. Impact of OATP transporters on pharmacokinetics. Br J Pharmacol. 2009;158(3):693–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Romaine S, Bailey K, Hall A, Balmforth A. The influence of SLCO1B1 (OATP1B1) gene polymorphisms on response to statin therapy. Pharmacogenomics J. 2010;10(1):1–11.

    Article  CAS  PubMed  Google Scholar 

  43. Amin ML. P-glycoprotein inhibition for optimal drug delivery. Drug Target Insights. 2013;19(7):27–34.

    Google Scholar 

  44. König J, Seithel A, Gradhand U, Fromm MF. Pharmacogenomics of human OATP transporters. Naunyn Schmiedebergs Arch Pharmacol. 2006;372(6):432–43.

    Article  PubMed  Google Scholar 

  45. Lin JH, Yamazaki M. Role of P-glycoprotein in pharmacokinetics: clinical implications. Clin Pharmacokinet. 2003;42(1):59–98.

    Article  CAS  PubMed  Google Scholar 

  46. Wishart DS, Feunang YD, Guo AC, Lo EJ, Marcu A, Grant JR, et al. DrugBank 5.0: a major update to the DrugBank database for 2018. Nucleic Acids Res. 2018;46(D1):D1074–82.

    Article  PubMed Central  Google Scholar 

  47. Drugs.com. 2015. Available from: https://www.drugs.com. Accessed 18 Dec 2017.

  48. Kim S, Thiessen PA, Bolton EE, Chen J, Fu G, Gindulyte A, et al. PubChem substance and compound databases. Nucleic Acids Res. 2016;44(D1):D1202–13.

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank Mr. Timothy Lee, PharmD student for assistance in retrieving information for this study. The authors thank the University of Washington Drug Interaction Database, specifically Dr. Isabelle Ragueneau-Majlessi for providing feedback of the criteria for defining metabolism pathways. The authors also sincerely thank Dr. Kristina Ward for her contribution to the data extraction from the CenterWatch database.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Fatemeh Akhlaghi.

Ethics declarations

Funding

Support from Grant No. R15 GM101599 from the National Institutes of Health is gratefully acknowledged.

Conflict of interest

Anitha Saravanakumar, Armin Sadighi, Rachel Ryu, and Fatemeh Akhlaghi have no conflicts of interest that are directly relevant to the content of this article.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 174 kb)

Supplementary material 2 (XLS 770 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saravanakumar, A., Sadighi, A., Ryu, R. et al. Physicochemical Properties, Biotransformation, and Transport Pathways of Established and Newly Approved Medications: A Systematic Review of the Top 200 Most Prescribed Drugs vs. the FDA-Approved Drugs Between 2005 and 2016. Clin Pharmacokinet 58, 1281–1294 (2019). https://doi.org/10.1007/s40262-019-00750-8

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s40262-019-00750-8

Navigation