Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 43, 2013 - Issue 12
652
Views
18
CrossRef citations to date
0
Altmetric
General Xenobiochemistry

Using human recombinant UDP-glucuronosyltransferase isoforms and a relative activity factor approach to model total body clearance of laropiprant (MK-0524) in humans

, , , , , , , & show all
Pages 1027-1036 | Received 22 Jan 2013, Accepted 28 Mar 2013, Published online: 03 May 2013

References

  • Al-Jahdari WS, Yamamoto K, Hiraoka H, et al. (2006). Prediction of total propofol clearance based on enzyme activities in microsomes from human kidney and liver. Eur J Clin Pharmacol 62:527–33
  • Barter ZE, Bayliss MK, Beaune PH, et al. (2007). Scaling factors for the extrapolation of in vivo metabolic drug clearance from in vitro data: reaching a consensus on values of human microsomal protein and hepatocellularity per gram of liver. Curr Drug Metab 8:33–45
  • Chang SW, Reddy V, Pereira T, et al. (2007). The pharmacokinetics and disposition of MK-0524, a Prosglandin D2 Receptor 1 antagonist, in rats, dogs and monkeys. Xenobiotica 37:514–33
  • Chen Y, Liu L, Nguyen K, Fretland AJ. (2011). Utility of intersystem extrapolation factors in early reaction phenotyping and the quantitative extrapolation of human liver microsomal intrinsic clearance using recombinant cytochromes P450. Drug Metab Dispos 39:373–82
  • Crespi CL, Miller VP. (1999). The use of heterologously expressed drug metabolizing enzymes – state of the art and prospects for the future. Pharmacol Ther 84:121–31
  • Cubitt HE, Houston JB, Galetin A. (2009). Relative importance of intestinal and hepatic glucuronidation-impact on the prediction of drug clearance. Pharm Res 26:1073–83
  • Davies B, Morris T. (1993). Physiological parameters in laboratory animals and humans. Pharm Res 10:1093–5
  • Dean BJ, Chang S, Silva Elipe MV, et al. (2007). Metabolism of MK-0524, a prostaglandin D2 receptor 1 antagonist, in microsomes and hepatocytes from preclinical species and humans. Drug Metab Dispos 35:283–92
  • Fisher MB, Paine MF, Strelevitz TJ, Wrighton SA. (2001). The role of hepatic and extrahepatic UDP-glucuronosyltransferases in human drug metabolism. Drug Metab Rev 33:273–97
  • Gibson CR, Bergman A, Lu P, et al. (2009). Prediction of phase I single-dose pharmacokinetics using recombinant cytochromes P450 and physiologically based modelling. Xenobiotica 39:637–48
  • Gill KL, Houston JB, Galetin A. (2012). Characterization of in vitro glucuronidation clearance of a range of drugs in human kidney microsomes: comparison with liver and intestinal glucuronidation and impact of albumin. Drug Metab Dispos 40:825–35
  • Harbourt DE, Fallon JK, Ito S, et al. (2011). Quantification of human uridine-diphosphate glucuronosyl transferase 1A isoforms in liver, intestine, and kidney using nanobore liquid chromatography-tandem mass spectrometry. Anal Chem 84:98–105
  • Houston JB, Kenworthy KE. (2000). In vitro--in vivo scaling of CYP kinetic data not consistent with the classical Michaelis--Menten model. Drug Metab Dispos 28:246–54
  • Karanam B, Madeira M, Bradley S, et al. (2007). Absorption, metabolism, and excretion of [(14)C]MK-0524, a prostaglandin D(2) receptor antagonist, in humans. Drug Metab Dispos 35:1196–202
  • Kato Y, Nakajima M, Oda S, et al. (2012). Human UDP-glucuronosyltransferase isoforms involved in haloperidol glucuronidation and quantitative estimation of their contribution. Drug Metab Dispos 40:240–8
  • Mei Q, Tang C, Assang C, et al. (1999). Role of a potent inhibitory monoclonal antibody to cytochrome P-450 3A4 in assessment of human drug metabolism. J Pharmacol Exp Ther 291:749–59
  • Miners JO, Knights KM, Houston JB, Mackenzie PI. (2006). In vitro--in vivo correlation for drugs and other compounds eliminated by glucuronidation in humans: pitfalls and promises. Biochem Pharmacol 71:1531–9
  • Miners JO, Mackenzie PI, Knights KM. (2010). The prediction of drug-glucuronidation parameters in humans: UDP-glucuronosyltransferase enzyme-selective substrate and inhibitor probes for reaction phenotyping and in vitro--in vivo extrapolation of drug clearance and drug--drug interaction potential. Drug Metab Rev 42:189–201
  • Nakajima M, Nakamura S, Tokudome S, et al. (1999). Azelastine N-demethylation by cytochrome P-450 (CYP)3A4, CYP2D6, and CYP1A2 in human liver microsomes: evaluation of approach to predict the contribution of multiple CYPs. Drug Metab Dispos 27:1381–91
  • Nicoll-Griffith DA, Seto C, Aubin Y, et al. (2007). In vitro biotransformations of the prostaglandin D2 (DP) antagonist MK-0524 and synthesis of metabolites. Bioorg Med Chem Lett 17:301–4
  • Obach RS, Baxter JG, Liston TE, et al. (1997). The prediction of human pharmacokinetic parameters from preclinical and in vitro metabolism data. J Pharmacol Exp Ther 283:46–58
  • Ohno S, Nakajin S. (2009). Determination of mRNA expression of human UDP-glucuronosyltransferases and application for localization in various human tissues by real-time reverse transcriptase-polymerase chain reaction. Drug Metab Dispos 37:32–40
  • Proctor NJ, Tucker GT, Rostami-Hodjegan A. (2004). Predicting drug clearance from recombinantly expressed CYPs: intersystem extrapolation factors. Xenobiotica 34:151–78
  • Rowland A, Elliot DJ, Knights KM, et al. (2008a). The “albumin effect” and in vitro-in vivo extrapolation: sequestration of long-chain unsaturated fatty acids enhances phenytoin hydroxylation by human liver microsomal and recombinant cytochrome P450 2C9. Drug Metab Dispos 36:870–7
  • Rowland A, Gaganis P, Elliot DJ, et al. (2007). Binding of inhibitory fatty acids is responsible for the enhancement of UDP-glucuronosyltransferase 2B7 activity by albumin: implications for in vitro--in vivo extrapolation. J Pharmacol Exp Ther 321:137–47
  • Rowland A, Knights KM, Mackenzie PI, Miners JO. (2008b). The “albumin effect” and drug glucuronidation: bovine serum albumin and fatty acid-free human serum albumin enhance the glucuronidation of UDP-glucuronosyltransferase (UGT) 1A9 substrates but not UGT1A1 and UGT1A6 activities. Drug Metab Dispos 36:1056–62
  • Rowland M, Benet LZ, Graham GG. (1973). Clearance concepts in pharmacokinetics. J Pharmacokinet Biopharm 1:123–36
  • Soars MG, Burchell B, Riley RJ. (2002). In vitro analysis of human drug glucuronidation and prediction of in vivo metabolic clearance. J Pharmacol Exp Ther 301:382–90
  • Soars MG, Ring BJ, Wrighton SA. (2003). The effect of incubation conditions on the enzyme kinetics of udp-glucuronosyltransferases. Drug Metab Dispos 31:762–7
  • Sturino CF, O'Neill G, Lachance N, et al. (2007). Discovery of a potent and selective prostaglandin D2 receptor antagonist, [(3R)-4-(4-chloro-benzyl)-7-fluoro-5-(methylsulfonyl)-1,2,3,4-tetrahydrocy clopenta[b]indol-3-yl]-acetic acid (MK-0524). J Med Chem 50:794–806
  • Tsoutsikos P, Miners JO, Stapleton A, et al. (2004). 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 67:191–9
  • Zhang H, Davis CD, Sinz MW, Rodrigues AD. (2007). Cytochrome P450 reaction-phenotyping: an industrial perspective. Expert Opin Drug Metab Toxicol 3:667–87

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.