Skip to main content

Advertisement

Log in

Atypical Cytochrome P450 Kinetics

Implications for Drug Discovery

  • Review Article
  • Published:
Drugs in R & D Aims and scope Submit manuscript

Abstract

The Michaelis-Menten model is commonly used to estimate a drug’s potential in vivo hepatic clearance based on in vitro data obtained during drug discovery and development. This paradigm assumes that the drug obeys ‘typical’ enzyme kinetics and thus can be described by this model. However, it is increasingly being recognised that a number of drugs metabolised not only by the cytochrome P450 enzymes but also by other enzymes and transporters can exhibit atypical kinetic profiles, and thus are not accurately modeled with the Michaelis-Menten model. Application of an incorrect model can then lead to mis-estimation of in vitro intrinsic clearance and thus affect the prediction of in vivo clearance. This review discusses several types of atypical kinetic profiles that may be observed, including examples of homotropic cooperativity (i.e. sigmoidal kinetics, biphasic kinetics and substrate inhibition kinetics) as well as heterotropic cooperativity (i.e. activation). Application of the incorrect kinetic model may profoundly affect estimations of intrinsic clearance. For example, incorrectly applying the Michaelis-Menten model to a kinetic profile exhibiting substrate inhibition kinetics will result in an underestimation of Km (Michaelis-Menten constant) and Vmax (maximal velocity), whereas application of the Michaelis-Menten model to sigmoidal kinetic data typically results in an overestimation of Km and Vmax at the lower substrate concentrations that are typically therapeutically relevant. One must also be careful of potential artefactual causes of atypical kinetic profiles, such as enzyme activation by solvents, buffer dependent kinetic profiles, or altered kinetic parameter estimates due to nonspecific binding of the substrate to proteins. Despite a plethora of data on the effects of atypical kinetic profiles in vitro, only modest effects have been noted in vivo (with the exception of substrate dependent inhibition). Thus, the clinical relevance of these phenomena remains inconclusive.

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

Access this article

Subscribe and save

Springer+ Basic
$34.99 /Month
  • Get 10 units per month
  • Download Article/Chapter or eBook
  • 1 Unit = 1 Article or 1 Chapter
  • Cancel anytime
Subscribe now

Buy Now

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

Similar content being viewed by others

References

  1. Wilkinson GR, Shand DG. Commentary: a physiological approach to hepatic drug clearance. Clin Pharmacol Ther 1975; 18 (4): 377–90

    PubMed  CAS  Google Scholar 

  2. Pang KS, Rowland M. Hepatic clearance of drugs: I. Theoretical considerations of a ‘well-stirred’ model and a ‘parallel tube’ model. Influence of hepatic blood flow, plasma and blood cell binding, and the hepatocellular enzymatic activity on hepatic drug clearance. J Pharmacokinet Biopharm 1977; 5 (6): 625–53

    PubMed  CAS  Google Scholar 

  3. Houston JB. Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol 1994; 47 (9): 1469–79

    Article  PubMed  CAS  Google Scholar 

  4. Houston JB, Carlile DJ. Prediction of hepatic clearance from microsomes, hepatocytes, and liver slices. Drug Metab Rev 1997; 29 (4): 891–922

    Article  PubMed  CAS  Google Scholar 

  5. Tracy TS, Hutzler JM, Haining RL, et al. Polymorphic variants (CYP2C9*3 and CYP2C9*5) and the F114L active site mutation of CYP2C9: effect on atypical kinetic metabolism profiles. Drug Metab Dispos 2002; 30 (4): 385–90

    Article  PubMed  CAS  Google Scholar 

  6. Korzekwa KR, Krishnamachary N, Shou M, et al. Evaluation of atypical cytochrome P450 kinetics with two-substrate models: evidence that multiple substrates can simultaneously bind to cytochrome P450 active sites. Biochemistry 1998; 37 (12): 4137–47

    Article  PubMed  CAS  Google Scholar 

  7. Shou M, Mei Q, Ettore Jr MW, et al. Sigmoidal kinetic model for two co-operative substrate-binding sites in a cytochrome P450 3A4 active site: an example of the metabolism of diazepam and its derivatives. Biochem J 1999; 340 (Pt 3): 845–53

    Article  PubMed  CAS  Google Scholar 

  8. Shou M, Dai R, Cui D, et al. A kinetic model for the metabolic interaction of two substrates at the active site of cytochrome P450 3A4. J Biol Chem 2001; 276 (3): 2256–62

    Article  PubMed  CAS  Google Scholar 

  9. Galetin A, Clarke SE, Houston JB. Quinidine and haloperidol as modifiers of CYP3A4 activity: multisite kinetic model approach. Drug Metab Dispos 2002; 30 (12): 1512–22

    Article  PubMed  CAS  Google Scholar 

  10. Galetin A, Clarke SE, Houston JB. Multisite kinetic analysis of interactions between prototypical CYP3A4 subgroup substrates: midazolam, testosterone, and nifedipine. Drug Metab Dispos 2003; 31 (9): 1108–16

    Article  PubMed  CAS  Google Scholar 

  11. Kenworthy KE, Clarke SE, Andrews J, et al. Multisite kinetic models for CYP3A4: simultaneous activation and inhibition of diazepam and testosterone metabolism. Drug Metab Dispos 2001; 29 (12): 1644–51

    PubMed  CAS  Google Scholar 

  12. Witherow LE, Houston JB. Sigmoidal kinetics of CYP3A substrates: an approach for scaling dextromethorphan metabolism in hepatic microsomes and isolated hepatocytes to predict in vivo clearance in rat. J Pharmacol Exp Ther 1999; 290 (1): 58–65

    PubMed  CAS  Google Scholar 

  13. Shou M, Grogan J, Mancewicz JA, et al. Activation of CYP3A4: evidence for the simultaneous binding of two substrates in a cytochrome P450 active site. Biochemistry 1994; 33 (21): 6450–5

    Article  PubMed  CAS  Google Scholar 

  14. Fisher MB, Campanale K, Ackermann BL, et al. In vitro glucuronidation using human liver microsomes and the pore-forming peptide alamethicin. Drug Metab Dispos 2000; 28 (5): 560–6

    PubMed  CAS  Google Scholar 

  15. Bowalgaha K, Elliot DJ, Mackenzie PI, et al. S-Naproxen and desmethylnaproxen glucuronidation by human liver micro-somes and recombinant human UDP-glucuronosyltransferases (UGT): role of UGT2B7 in the elimination of naproxen. Br J Clin Pharmacol 2005; 60 (4): 423–33

    Article  PubMed  CAS  Google Scholar 

  16. Rowland A, Elliot DJ, Williams JA, et al. In vitro characterization of lamotrigine N2-glucuronidation and the lamotrigine-valproic acid interaction. Drug Metab Dispos 2006; 34 (6): 1055–62

    PubMed  CAS  Google Scholar 

  17. Stone AN, Mackenzie PI, Galetin A, et al. Isoform selectivity and kinetics of morphine 3- and 6-glucuronidation by human UDP-glucuronosyltransferases: evidence for atypical glucuronidation kinetics by UGT2B7. Drug Metab Dispos 2003; 31 (9): 1086–9

    Article  PubMed  CAS  Google Scholar 

  18. Uchaipichat V, Mackenzie PI, Guo XH, et al. Human UDP-glucuronosyltransferases: isoform selectivity and kinetics of 4-methylumbelliferone and 1-naphthol glucuronidation, effects of organic solvents, and inhibition by diclofenac and probenecid. Drug Metab Dispos 2004; 32 (4): 413–23

    Article  PubMed  CAS  Google Scholar 

  19. Tsoutsikos P, Miners JO, Stapleton A, et al. Evidence that unsaturated fatty acids are potent inhibitors of renal UDP-glucuronosyltransferases (UGT): kinetic studies using human kidney cortical microsomes and recombinant UGT1A9 and UGT2B7. Biochem Pharmacol 2004; 67 (1): 191–9

    Article  PubMed  CAS  Google Scholar 

  20. Yan Z, Caldwell GW, Gauthier D, et al. N-glucuronidation of the platelet-derived growth factor receptor tyrosine kinase inhibitor 6,7-(dimethoxy-2,4-dihydroindeno[1,2-C]pyrazol-3 -yl)-(3-fluoro-phenyl)-amine by human UDP-glucuronosyl-transferases. Drug Metab Dispos 2006; 34 (5): 748–55

    Article  PubMed  CAS  Google Scholar 

  21. Basu NK, Kole L, Kubota S, et al. Human UDP-glucuronosyltransferases show atypical metabolism of mycophenolic acid and inhibition by curcumin. Drug Metab Dispos 2004; 32 (7): 768–73

    Article  PubMed  CAS  Google Scholar 

  22. Williams JA, Ring BJ, Cantrell VE, et al. Differential modulation of UDP-glucuronosyltransferase 1A1 (UGT1A1)-catalyzed estradiol-3-glucuronidation by the addition of UGT1A1 substrates and other compounds to human liver microsomes. Drug Metab Dispos 2002; 30 (11): 1266–73

    Article  PubMed  CAS  Google Scholar 

  23. Pfeiffer E, Treiling CR, Hoehle SI, et al. Isoflavones modulate the glucuronidation of estradiol in human liver microsomes. Carcinogenesis 2005; 26 (12): 2172–8

    Article  PubMed  CAS  Google Scholar 

  24. Hummel MA, Gannett PM, Aguilar JS, et al. Effector-mediated alteration of substrate orientation in cytochrome P450 2C9. Biochemistry 2004; 43 (22): 7207–14

    Article  PubMed  CAS  Google Scholar 

  25. Rock DA, Perkins BN, Wahlstrom J, et al. A method for determining two substrates binding in the same active site of cytochrome P450BM3: an explanation of high energy omega product formation. Arch Biochem Biophys 2003; 416 (1): 9–16

    Article  PubMed  CAS  Google Scholar 

  26. Dabrowski MJ, Schrag ML, Wienkers LC, et al. Pyrene.pyrene complexes at the active site of cytochrome P450 3A4: evidence for a multiple substrate binding site. J Am Chem Soc 2002; 124 (40): 11866–7

    Article  PubMed  CAS  Google Scholar 

  27. Koley AP, Buters JT, Robinson RC, et al. Differential mechanisms of cytochrome P450 inhibition and activation by alpha-naphthoflavone. J Biol Chem 1997; 272 (6): 3149–52

    Article  PubMed  CAS  Google Scholar 

  28. Koley AP, Robinson RC, Markowitz A, et al. Drug-drug interactions: effect of quinidine on nifedipine binding to human cytochrome P450 3A4. Biochem Pharmacol 1997; 53 (4): 455–60

    Article  PubMed  CAS  Google Scholar 

  29. Koley AP, Buters JT, Robinson RC, et al. CO binding kinetics of human cytochrome P450 3A4. Specific interaction of substrates with kinetically distinguishable conformers. J Biol Chem 1995; 270 (10): 5014–8

    Article  PubMed  CAS  Google Scholar 

  30. Atkins WM, Wang RW, Lu AY. Allosteric behavior in cytochrome p450-dependent in vitro drug-drug interactions: a propective based on conformational dynamics. Chem Res Toxicol 2001; 14 (4): 338–47

    Article  PubMed  CAS  Google Scholar 

  31. Williams PA, Cosme J, Vinkovic DM, et al. Crystal structures of human cytochrome P450 3A4 bound to metyrapone and progesterone. Science 2004; 305 (5684): 683–6

    Article  PubMed  CAS  Google Scholar 

  32. Schoch GA, Yano JK, Wester MR, et al. Structure of human microsomal cytochrome P450 2C8: evidence for a peripheral fatty acid binding site. J Biol Chem 2004; 279 (10): 9497–503

    Article  PubMed  CAS  Google Scholar 

  33. Hill AV. The possible effects of the aggregation of the molecules of haemoglobin on its dissociation curves. J Physiol 1910; 40: iv–vii

    Google Scholar 

  34. Houston JB, Kenworthy KE. In vitro-in vivo scaling of CYP kinetic data not consistent with the classical Michaelis-Menten model. Drug Metab Dispos 2000; 28 (3): 246–54

    PubMed  CAS  Google Scholar 

  35. Houston JB, Galetin A. Progress towards prediction of human pharmacokinetic parameters from in vitro technologies. Drug Metab Rev 2003; 35 (4): 393–415

    Article  PubMed  CAS  Google Scholar 

  36. Shou M, Lin Y, Lu P, et al. Enzyme kinetics of cytochrome P450-mediated reactions. Curr Drug Metab 2001; 2 (1): 17–36

    Article  PubMed  CAS  Google Scholar 

  37. Hutzler JM, Wienkers LC, Wahlstrom JL, et al. Activation of cytochrome P450 2C9-mediated metabolism: mechanistic evidence in support of kinetic observations. Arch Biochem Bio-phys 2003; 410: 16–24

    Article  Google Scholar 

  38. Hutzler JM, Hauer MJ, Tracy TS. Dapsone activation of CYP2C9-mediated metabolism: evidence for activation of multiple substrates and a two-site model. Drug Metab Dispos 2001; 29 (7): 1029–34

    PubMed  CAS  Google Scholar 

  39. He YA, Roussel F, Halpert JR. Analysis of homotropic and heterotropic cooperativity of diazepam oxidation by CYP3A4 using site-directed mutagenesis and kinetic modeling. Arch Biochem Biophys 2003; 409 (1): 92–101

    Article  PubMed  CAS  Google Scholar 

  40. Kumar V, Wahlstrom JL, Rock DA, et al.. CYP2C9 inhibition: impact of probe selection and pharmacogenetics on in vitro inhibition profiles. Drug Metab Dispos. Epub 2006

    Google Scholar 

  41. Galetin A, Ito K, Hallifax D, et al. CYP3A4 substrate selection and substitution in the prediction of potential drug-drug interactions. J Pharmacol Exp Ther 2005; 314 6(1): 180–90

    Article  Google Scholar 

  42. Kenworthy KE, Bloomer JC, Clarke SE, et al. CYP3A4 drug interactions: correlation of 10 in vitro probe substrates. Br J Clin Pharmacol 1999; 48 (5): 716–27

    Article  PubMed  CAS  Google Scholar 

  43. Wienkers LC. Factors confounding the successful extrapolation of in vitro CYP3A inhibition information to the in vivo condition. Eur J Pharm Sci 2002; 15: 239–42

    Article  PubMed  CAS  Google Scholar 

  44. Venkatakrishnan K, von Moltke LL, Obach RS, et al. Microsomal binding of amitriptyline: effect on estimation of enzyme kinetic parameters in vitro. J Pharmacol Exp Ther 2000; 293 (2): 343–50

    PubMed  CAS  Google Scholar 

  45. Obach RS. Nonspecific binding to microsomes: impact on scale-up of in vitro intrinsic clearance to hepatic clearance as assessed through examination of warfarin, imipramine, and propranolol. Drug Metab Dispos 1997; 25 (12): 1359–69

    PubMed  CAS  Google Scholar 

  46. Hutzler JM, Powers FJ, Wynalda MA, et al. Effect of carbonate anion on cytochrome P450 2D6-mediated metabolism in vitro: the potential role of multiple oxygenating species. Arch Bio-chem Biophys 2003; 417 (2): 165–75

    Article  CAS  Google Scholar 

  47. Hickman D, Wang JP, Wang Y, et al. Evaluation of the selectivity of in vitro probes and suitability of organic solvents for the measurement of human cytochrome P450 monooxygenase activities. Drug Metab Dispos 1998; 26 (3): 207–15

    PubMed  CAS  Google Scholar 

  48. Tang C, Shou M, Rodrigues AD. Substrate-dependent effect of acetonitrile on human liver microsomal cytochrome P450 2C9 (CYP2C9) activity. Drug Metab Dispos 2000; 28 (5): 567–72

    PubMed  CAS  Google Scholar 

  49. Jushchyshyn MI, Hutzler JM, Schrag ML, et al. Catalytic turnover of pyrene by CYP3A4: evidence that cytochrome b5 directly induces positive cooperativity. Arch Biochem Biophys 2005; 438 (1): 21–8

    Article  PubMed  CAS  Google Scholar 

  50. Tang W, Stearns RA, Kwei GY, et al. Interaction of diclofenac and quinidine in monkeys: stimulation of diclofenac metabolism. J Pharmacol Exp Ther 1999; 291 (3): 1068–74

    PubMed  CAS  Google Scholar 

  51. Hutzler JM, Frye RF, Korzekwa KR, et al. Minimal in vivo activation of CYP2C9-mediated flurbiprofen metabolism by dapsone. Eur J Pharm Sci 2001; 14 (1): 47–52

    Article  PubMed  CAS  Google Scholar 

  52. Egnell AC, Houston B, Boyer S. In vivo CYP3A4 heteroactivation is a possible mechanism for the drug interaction between felbamate and carbamazepine. J Pharmacol Exp Ther 2003 Jun; 305 (3): 1251–62

    Article  PubMed  CAS  Google Scholar 

  53. Backman JT, Wang JS, Wen X, et al. Mibefradil but not isradipine substantially elevates the plasma concentrations of the CYP3A4 substrate triazolam. Clin Pharmacol Ther 1999; 66 (4): 401–7

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

Supported in part by Public Health Service grant #GM 063215 from the National Institutes of Health.

The author has no conflicts of interest that are directly relevant to the content of this review.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Timothy S. Tracy.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Tracy, T.S. Atypical Cytochrome P450 Kinetics. Drugs R D 7, 349–363 (2006). https://doi.org/10.2165/00126839-200607060-00004

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.2165/00126839-200607060-00004

Keywords

Navigation