Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 42, 2012 - Issue 10
605
Views
8
CrossRef citations to date
0
Altmetric
General Xenobiochemistry

Method for predicting human intestinal first-pass metabolism of UGT substrate compounds

, , , , &
Pages 980-988 | Received 03 Feb 2012, Accepted 26 Mar 2012, Published online: 27 Apr 2012

References

  • Bock KW, Bock-Hennig BS, Münzel PA, Brandenburg JO, Köhle CT, Soars MG, Riley RJ, Burchell B, von Richter O, Eichelbaum MF, Swedmark S, Orzechowski A. (2002). Tissue-specific regulation of canine intestinal and hepatic phenol and morphine UDP-glucuronosyltransferases by beta-naphthoflavone in comparison with humans. Biochem Pharmacol 63:1683–1690.
  • Cubitt HE, Houston JB, Galetin A. (2009). Relative importance of intestinal and hepatic glucuronidation-impact on the prediction of drug clearance. Pharm Res 26:1073–1083.
  • Furukawa T, Nakamori F, Tetsuka K, Naritomi Y, Moriguchi H, Yamano K, Terashita S, Teramura T. (2012). Quantitative prediction of intestinal glucuronidation of drugs in rats using in vitro metabolic clearance data. Drug Metab Pharmacokinet 27:171–180.
  • Gong QH, Cho JW, Huang T, Potter C, Gholami N, Basu NK, Kubota S, Carvalho S, Pennington MW, Owens IS, Popescu NC. (2001). Thirteen UDPglucuronosyltransferase genes are encoded at the human UGT1 gene complex locus. Pharmacogenetics 11:357–368.
  • Harbourt DE, Fallon JK, Ito S, Baba T, Ritter JK, Glish GL, Smith PC. (2012). 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.
  • Jeong EJ, Liu Y, Lin H, Hu M. (2005). Species- and disposition model-dependent metabolism of raloxifene in gut and liver: role of UGT1A10. Drug Metab Dispos 33:785–794.
  • Kadono K, Akabane T, Tabata K, Gato K, Terashita S, Teramura T. (2010). Quantitative prediction of intestinal metabolism in humans from a simplified intestinal availability model and empirical scaling factor. Drug Metab Dispos 38:1230–1237.
  • Kato M, Chiba K, Hisaka A, Ishigami M, Kayama M, Mizuno N, Nagata Y, Takakuwa S, Tsukamoto Y, Ueda K, Kusuhara H, Ito K, Sugiyama Y. (2003). The intestinal first-pass metabolism of substrates of CYP3A4 and P-glycoprotein-quantitative analysis based on information from the literature. Drug Metab Pharmacokinet 18:365–372.
  • Kemp DC, Fan PW, Stevens JC. (2002). Characterization of raloxifene glucuronidation in vitro: contribution of intestinal metabolism to presystemic clearance. Drug Metab Dispos 30:694–700.
  • Kilford PJ, Stringer R, Sohal B, Houston JB, Galetin A. (2009). Prediction of drug clearance by glucuronidation from in vitro data: use of combined cytochrome P450 and UDP-glucuronosyltransferase cofactors in alamethicin-activated human liver microsomes. Drug Metab Dispos 37:82–89.
  • Kosaka K, Sakai N, Endo Y, Fukuhara Y, Tsuda-Tsukimoto M, Ohtsuka T, Kino I, Tanimoto T, Takeba N, Takahashi M, Kume T. (2011). Impact of intestinal glucuronidation on the pharmacokinetics of raloxifene. Drug Metab Dispos 39:1495–1502.
  • Kosoglou T, Statkevich P, Johnson-Levonas AO, Paolini JF, Bergman AJ, Alton KB. (2005). Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions. Clin Pharmacokinet 44:467–494.
  • Lindstrom TD, Whitaker NG, Whitaker GW. (1984). Disposition and metabolism of a new benzothiophene antiestrogen in rats, dogs and monkeys. Xenobiotica 14:841–847.
  • Naritomi Y, Terashita S, Kimura S, Suzuki A, Kagayama A, Sugiyama Y. (2001). Prediction of human hepatic clearance from in vivo animal experiments and in vitro metabolic studies with liver microsomes from animals and humans. Drug Metab Dispos 29:1316–1324.
  • Nishimura M, Koeda A, Morikawa H, Satoh T, Narimatsu S, Naito S. (2009). Tissue-specific mRNA expression profiles of drug-metabolizing enzymes and transporters in the cynomolgus monkey. Drug Metab Pharmacokinet 24:139–144.
  • Nishimuta H, Sato K, Yabuki M, Komuro S. (2011). Prediction of the intestinal first-pass metabolism of CYP3A and UGT substrates in humans from in vitro data. Drug Metab Pharmacokinet 26:592–601.
  • 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.
  • Ohtsuki S, Schaefer O, Kawakami H, Inoue T, Liehner S, Saito A, Ishiguro N, Kishimoto W, Ludwig-Schwellinger E, Ebner T, Terasaki T. (2012). Simultaneous absolute protein quantification of transporters, cytochromes P450, and UDP-glucuronosyltransferases as a novel approach for the characterization of individual human liver: comparison with mRNA levels and activities. Drug Metab Dispos 40:83–92.
  • Ritter JK. (2007). Intestinal UGTs as potential modifiers of pharmacokinetics and biological responses to drugs and xenobiotics. Expert Opin Drug Metab Toxicol 3:93–107.
  • Rowland A, Knights KM, Mackenzie PI, Miners JO. (2008). The “albumin effectâ 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 36:1056–1062.
  • Rowland A, Knights KM, Mackenzie PI, Miners JO. (2009). Characterization of the binding of drugs to human intestinal fatty acid binding protein (IFABP): potential role of IFABP as an alternative to albumin for in vitro-in vivo extrapolation of drug kinetic parameters. Drug Metab Dispos 37:1395–1403.
  • Sakaguchi K, Green M, Stock N, Reger TS, Zunic J, King C. (2004). Glucuronidation of carboxylic acid containing compounds by UDP-glucuronosyltransferase isoforms. Arch Biochem Biophys 424:219–225.
  • Sawada Y. (1985). Animal scale up. In: Hanano MUmemura K, eds. Applied Pharmacokinetics: Theory and Experiments. Tokyo, Japan: Soft Science, Inc., 477.
  • Shelby MK, Cherrington NJ, Vansell NR, Klaassen CD. (2003). Tissue mRNA expression of the rat UDP-glucuronosyltransferase gene family. Drug Metab Dispos 31:326–333.
  • Shiratani H, Katoh M, Nakajima M, Yokoi T. (2008). Species differences in UDP-glucuronosyltransferase activities in mice and rats. Drug Metab Dispos 36:1745–1752.
  • Soars MG, Riley RJ, Findlay KA, Coffey MJ, Burchell B. (2001). Evidence for significant differences in microsomal drug glucuronidation by canine and human liver and kidney. Drug Metab Dispos 29:121–126.
  • Tong Z, Li H, Goljer I, McConnell O, Chandrasekaran A. (2007). In vitro glucuronidation of thyroxine and triiodothyronine by liver microsomes and recombinant human UDP-glucuronosyltransferases. Drug Metab Dispos 35:2203–2210.
  • van Heek M, Farley C, Compton DS, Hoos L, Alton KB, Sybertz EJ, Davis HR Jr. (2000). Comparison of the activity and disposition of the novel cholesterol absorption inhibitor, SCH58235, and its glucuronide, SCH60663. Br J Pharmacol 129:1748–1754.
  • Webb LJ, Miles KK, Auyeung DJ, Kessler FK, Ritter JK. (2005). Analysis of substrate specificities and tissue expression of rat UDP-glucuronosyltransferases UGT1A7 and UGT1A8. Drug Metab Dispos 33:77–82.
  • Wu B, Kulkarni K, Basu S, Zhang S, Hu M. (2011). First-pass metabolism via UDP-glucuronosyltransferase: a barrier to oral bioavailability of phenolics. J Pharm Sci 100:3655–3681.

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.