Skip to main content
Log in

Development of a Generalized, Quantitative Physicochemical Model of CYP3A4 Inhibition for Use in Early Drug Discovery

  • Published:
Pharmaceutical Research Aims and scope Submit manuscript

Abstract

Purpose. To examine the structure–activity relationships for the inhibition of the activity of recombinant human CYP3A4 and to establish a generalized, quantitative physicochemical model for use in early drug discovery.

Methods. Inhibition of the activity of recombinant human CYP3A4 (erythromycin N–demethylase) by 30 diverse chemicals was studied using enhanced throughput methodology.

Results. There was a general, strong correlation between the IC50 value determined against erythromycin N–demethylase activity and lipophilicity (LogD7.4) (r 2 = 0.68, p <0.0001). This relationship was strengthened further by subdividing the structures studied into two distinct subpopulations of chemistry within the dataset. These could be identified by the absence (r 2 = 0.80, p <0.0001) or presence (r 2 = 0.69, p <0.0001) of a sterically uninhindered N–containing heterocycle, more specifically a pyridine, imidazole, or triazole function. The presence of these structural motifs increased the potency of CYP3A4 inhibition by approximately 10–fold for a given lipophilicity (LogD7.4.value). More detailed analyses of AstraZeneca compounds demonstrated that the inhibitory potency of the pyridine structure can be attenuated through direct steric effects or electronic substitution resulting in a modulation of the pKa of the pyridine nitrogen, thereby influencing its ability to interact with the CYP heme.

Conclusions. A generalized, quantitative model is proposed for the inhibition of the major drug metabolizing enzyme, CYP3A4. This model indicates the importance of lipophilicity and rationalizes increased potency arising through additional interactions with the heme iron. These general relationships were shown to be applicable to a selection of compounds of interest to several early research projects.

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.

Institutional subscriptions

Similar content being viewed by others

REFERENCES

  1. T. Shimada, H. Yamazaki, M. Mimura, I. Yukiharu, and F. P. Guengerich. Interindividual variations in human liver cytochrome P450 enzymes involved in the oxidation of drugs, carcinogens and toxic chemicals: Studies with liver microsomes of 30 Japanese and 30 Caucasians. J. Pharmacol. Exp. Ther. 270:414–423 (1994).

    Google Scholar 

  2. R. J. Bertz and G. R. Granneman. Use of in vitro and in vivo data to estimate the likelihood of metabolic pharmacokinetic interactions. Clin. Pharmacokinet. 32:210–258 (1997).

    Google Scholar 

  3. J. H. Lin and A. Y. H. Lu. Inhibition and induction of cytochrome P450 and the clinical implications. Clin. Pharmacokinet. 35:361–390 (1998).

    Google Scholar 

  4. P. K. Honig, D. C. Wortham, K. Zamani, D. P. Conner, J. C. Mullin, and L. R. Cantilena. Terfenadine–ketoconazole interaction: Pharmacokinetic and electrocardiographic consequences. JAMA 269:1513–1518 (1993).

    Google Scholar 

  5. S. R. Ahmad and S. M. Wolfe. Cisapride and torsades de pointes. Lancet 345:508 (1995).

    Google Scholar 

  6. J.C. Krayenbuhl S. Vozeh. M. Kondo–Oestreicher, and P. Dayer P. Drug–drug interactions of new active substances: Mibefradil example. Eur. J. Clin. Pharmacol. 55:559–565 (1999).

    Google Scholar 

  7. C. C. Peck, R. Temple, and J. M. Collins. Understanding consequences of concurrent therapies. JAMA 269: 1550–1552 (1993).

    Google Scholar 

  8. J. Halpert, T. L. Domanski, O. Adali, C. P. Biagini, P. Cosme, E. A. Dierks, E. F. Johnson, J. P. Jones, P. Ortiz de Montellano, R. M. Philpot, O. Sibbesen, K. W. Wyatt, and Z. Zheng, Z. Structure–function of cytochromes P450 and flavin–containing monooxygenases: implications for drug metabolism. Drug Metab. Dispos. 26:1223–1231 (1998).

    Google Scholar 

  9. L. Pichard, J. Domergue, G. Fourtanier, P. Koch, H. F. Schran, and P. Maurel. Metabolism of the new immunosuppressor cyclosporin G by human liver cytochromes P450. Biochem. Pharmacol. 51:591–598 (1996).

    Google Scholar 

  10. S. Ekins, G. Bravi, S. Binkley, J. S. Gillespie, B. J. Ring, J. H. Wikel, and S. A. Wrighton. Three–and four–dimensional quantitative structure activity relationship analyses of cytochrome P450–3A4 inhibitors. J. Pharmacol. Exp. Ther. 290:429–438 (1999).

    Google Scholar 

  11. G. C. Moody, S. J. Griffin, A. N. Mather, D. F. McGinnity, and R. J. Riley. Fully automated analysis of activities catalysed by the major human liver cytochrome P450 (CYP) enzymes: Assessment of human CYP inhibition potential. Xenobiotica 29:53–75 (1999).

    Google Scholar 

  12. D. F. McGinnity, S. J. Griffin, G. C. Moody, M. Voice, S. Hanlon, T. Friedberg, and R. J. Riley. Rapid characterization of the major drug–metabolizing human hepatic cytochrome P–450 enzymes expressed in Escherichia coli. Drug Metab. Dispos. 27:1017–1023 (1999).

    Google Scholar 

  13. R. J. Riley and D. Howbrook. In vitro analysis of the activity of the major human hepatic CYP enzyme (CYP3A4) using [N–methyl 14 C]erythromycin. J. Pharmacol. Toxicol. Meth. 38:189–193 (1998).

    Google Scholar 

  14. Y. Cheng and W. H. Prusoff, WH. Relationship between the inhibition constant (Ki) and the concnetration of an inhibitor that causes a 50% inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 22:3099–3108 (1973).

    Google Scholar 

  15. L. Pichard, I. Fabre, G. Fabre, J. Domergue, B. S. Aubert, G. Mouard, and P. Maurel. Cyclosporin A drug interactions: screening for inducers and inhibitors of cytochrome P450 (cyclosporin A oxidase) in primary cultures of human hepatocytes and in liver microsomes. Drug Metab. Dispos. 18:595–606 (1990).

    Google Scholar 

  16. K.E. Kenworthy, J. C. Bloomer, S. E. Clarke, and J. B. Houston. CYP3A4 drug interactions: correlation of 10 in vitro probe substrates. Br. J. Clin. Pharmacol. 48:716–727 (1999).

    Google Scholar 

  17. R. W. Wang, D. J. Newton, N. Liu, W. M. Atkins, and A. Y. Lu. Human cytochrome P–450 3A4: in vitro drug–drug interaction patterns are substrate–dependent. Drug Metab. Dispos. 28:360–366 (2000).

    Google Scholar 

  18. D. A. Smith, S. M. Abel, R. Hyland, and B. Jones. Human cytochrome P450s: Selectivity and measurement in vivo. Xenobiotica 28:10957–1128 (1998).

    Google Scholar 

  19. C. A. Lipinski, F. Lombardo, B. W. Dominy, and P. J. Feeney. Experimental and computational approaches to estimate solubility and permeability in drug discovery and development settings. Adv. Drug Deliv. Rev. 23:3–27 (1997).

    Google Scholar 

  20. D. A. Smith. Drug metabolism: Towards the next millenium. Biomed. Health Res. 25:137–43 (1998).

    Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Rights and permissions

Reprints and permissions

About this article

Cite this article

Riley, R.J., Parker, A.J., Trigg, S. et al. Development of a Generalized, Quantitative Physicochemical Model of CYP3A4 Inhibition for Use in Early Drug Discovery. Pharm Res 18, 652–655 (2001). https://doi.org/10.1023/A:1011085411050

Download citation

  • Issue Date:

  • DOI: https://doi.org/10.1023/A:1011085411050

Navigation