Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 44, 2014 - Issue 3
1,385
Views
25
CrossRef citations to date
0
Altmetric
Research Article

Species differences in intestinal glucuronidation activities between humans, rats, dogs and monkeys

, , , , , , , & show all
Pages 205-216 | Received 30 May 2013, Accepted 21 Jul 2013, Published online: 20 Aug 2013

References

  • Boersma MG, van der Woude H, Bogaards J, et al. (2002). Regioselectivity of phase II metabolism of luteolin and quercetin by UDP-glucuronosyl transferases. Chem Res Toxicol 15:662–70
  • Cayen MN, Kraml M, Ferdinandi ES, et al. (1981). The metabolic disposition of etodolac in rats, dogs, and man. Drug Metab Rev 12:339–62
  • Chiou WL, Jeong HY, Chung SM, Wu TC. (2000). Evaluation of using dog as an animal model to study the fraction of oral dose absorbed of 43 drugs in humans. Pharm Res 17:135–40
  • Court MH. (2005). Isoform-selective probe substrates for in vitro studies of human UDP-glucuronosyltransferases. Methods Enzymol 400:104–16
  • Davies B, Morris T. (1993). Physiological parameters in laboratory animals and humans. Pharm Res 10:1093–5
  • Deguchi T, Watanabe N, Kurihara A, et al. (2011). Human pharmacokinetic prediction of UDP-glucuronosyltransferase substrates with an animal scale-up approach. Drug Metab Dispos 39:820–9
  • FDA Drug Approval Package, Tasmar (Tolcapone) Tablets, Application No.20697, 1 Jan 1998, Pharmacology Reviews: 136, Available from: http://www.accessdata.fda.gov/drugsatfda_docs/nda/98/20697_Tasmar_pharmr_P3.pdf [last accessed Nov 2010]
  • Fisher MB, Labissiere G. (2007). The role of the intestine in drug metabolism and pharmacokinetics: an industry perspective. Curr Drug Metab 8:694–9
  • Furukawa T, Nakamori F, Tetsuka K, et al. (2012a). Quantitative prediction of intestinal glucuronidation of drugs in rats using in vitro metabolic clearance data. Drug Metab Pharmacokinet 27:171–80
  • Furukawa T, Yamano K, Naritomi Y, et al. (2012b). Method for predicting human intestinal first-pass metabolism of UGT substrate compounds. Xenobiotica 42:980–8
  • Ghosal A, Hapangama N, Yuan Y, et al. (2004). Identification of human UDP-glucuronosyltransferase enzyme(s) responsible for the glucuronidation of ezetimibe (Zetia). Drug Metab Dispos 32:314–20
  • 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
  • Haller S, Schuler F, Lazic SE, et al. (2012). Expression profiles of metabolic enzymes and drug transporters in the liver and along the intestine of beagle dogs. Drug Metab Dispos 40:1603–10
  • Kadono K, Akabane T, Tabata K, et al. (2010). Quantitative prediction of intestinal metabolism in humans from a simplified intestinal availability model and empirical scaling factor. Drug Metab Dispos 38:1230–7
  • 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, et al. (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 37:82–9
  • Kobayashi K, Yamamoto T, Chiba K, et al. (1998). Human buprenorphine N-dealkylation is catalyzed by cytochrome P450 3A4. Drug Metab Dispos 26:818–21
  • Komura H, Iwaki M. (2011). In vitro and in vivo small intestinal metabolism of CYP3A and UGT substrates in preclinical animals species and humans: species differences. Drug Metab Rev 43:476–98
  • Kosaka K, Sakai N, Endo Y, et al. (2011). Impact of intestinal glucuronidation on the pharmacokinetics of raloxifene. Drug Metab Dispos 39:1495–502
  • Kosoglou T, Statkevich P, Johnson-Levonas AO, et al. (2005). Ezetimibe: a review of its metabolism, pharmacokinetics and drug interactions. Clin Pharmacokinet 44:467–94
  • Lindstrom TD, Whitaker NG, Whitaker GW. (1984). Disposition and metabolism of a new benzothiophene antiestrogen in rats, dogs and monkeys. Xenobiotica 14:841–7
  • Madani S, Paine MF, Lewis L, et al. (1999). Comparison of CYP2D6 content and metoprolol oxidation between microsomes isolated from human livers and small intestines. Pharm Res 16:1199–205
  • Mano Y, Usui T, Kamimura H. (2007). Contribution of UDP-glucuronosyltransferases 1A9 and 2B7 to the glucuronidation of indomethacin in the human liver. Eur J Clin Pharmacol 63:289–96
  • Martignoni M, Groothuis GM, de Kanter R. (2006). Species differences between mouse, rat, dog, monkey and human CYP-mediated drug metabolism, inhibition and induction. Expert Opin Drug Metab Toxicol 2:875–94
  • Nakamori F, Naritomi Y, Furutani M, et al. (2011). Correlation of intrinsic in vitro and in vivo clearance for drugs metabolized by hepatic UDP-glucuronosyltransferases in rats. Drug Metab Pharmacokinet 26:465–73
  • Nakamori F, Naritomi Y, Hosoya K, et al. (2012). Quantitative prediction of human intestinal glucuronidation effects on intestinal availability of UDP-glucuronosyltransferase substrates using in vitro data. Drug Metab Dispos 40:1771–7
  • Naritomi Y, Terashita S, Kimura S, et al. (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 29:1316–24
  • Nishimura M, Koeda A, Morikawa H, et al. (2009). Tissue-specific mRNA expression profiles of drug-metabolizing enzymes and transporters in the cynomolgus monkey. Drug Metab Pharmacokinet 24:139–44
  • 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
  • Pharmaceuticals and Medical Devices Agency (PMDA) in Japan (2007). Review Report for Zetia (Ezetimebe) tablets:16, Available from: http://www.info.pmda.go.jp/shinyaku/P200700023/170050000_21900AMY00021000_A100_2.pdf [last accessed Apr 2013]
  • Peris-Ribera JE, Torres-Molina F, Garcia-Carbonell MC, et al. (1991). Pharmacokinetics and bioavailability of diclofenac in the rat. J Pharmacokinet Biopharm 19:647–65
  • Priymenko N, Ferre JP, Rascol A, et al. (1993). Migrating motor complex of the intestine and absorption of a biliary excreted drug in the dog. J Pharmacol Exp Ther 267:1161–7
  • 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
  • Rouguieg K, Picard N, Sauvage FL, et al. (2010). Contribution of the different UDP-glucuronosyltransferase (UGT) isoforms to buprenorphine and norbuprenorphine metabolism and relationship with the main UGT polymorphisms in a bank of human liver microsomes. Drug Metab Dispos 38:40–5
  • 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. (2008). 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
  • Sabolovic N, Heydel JM, Li X, et al. (2004). Carboxyl nonsteroidal anti-inflammatory drugs are efficiently glucuronidated by microsomes of the human gastrointestinal tract. Biochim Biophys Acta 1675:120–9
  • Sawada Y. (1985). Animal scale up. In: Hanano M, Umemura K, eds. Applied pharmacokinetics – theory and experiments. Tokyo: 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 31:326–33
  • Soars MG, Smith DJ, Riley RJ, Burchell B. (2001). Cloning and characterization of a canine UDP-glucuronosyltransferase. Arch Biochem Biophys 391:218–24
  • Uchaipichat V, Mackenzie PI, Elliot DJ, Miners JO. (2006). Selectivity of substrate (trifluoperazine) and inhibitor (amitriptyline, androsterone, canrenoic acid, hecogenin, phenylbutazone, quinidine, quinine, and sulfinpyrazone) “probes” for human UDP-glucuronosyltransferases. Drug Metab Dispos 34:449–56
  • Yamaoka K, Tanigawara Y, Nakagawa T, Uno T. (1981). A pharmacokinetic analysis program (multi) for microcomputer. J Pharmacobiodyn 4:879–85
  • Yoon IS, Choi MK, Kim JS, et al. (2011). Pharmacokinetics and first-pass elimination of metoprolol in rats: contribution of intestinal first-pass extraction to low bioavailability of metoprolol. Xenobiotica 41:243–51

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.