Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 47, 2017 - Issue 9
1,175
Views
25
CrossRef citations to date
0
Altmetric
General Xenobiochemistry

Comparison of methods for the prediction of human clearance from hepatocyte intrinsic clearance for a set of reference compounds and an external evaluation set

, , , , &
Pages 741-751 | Received 28 Jun 2016, Accepted 07 Aug 2016, Published online: 25 Aug 2016

References

  • Armitage P, Berry G. (1994). Statistical methods in medical research. 3rd ed. Oxford: Blackwell Scientific Publications
  • Bonn B, Svanberg P, Janefeldt A, et al. (2016). Determination of human hepatocyte intrinsic clearance for slowly metabolized compounds: comparison of a primary hepatocyte/stromal cell co-culture with plated primary hepatocytes and HepaRG. Drug Metab Dispos 44:527–33
  • Brandon EF, Raap CD, Meijerman I, et al. (2003). An update on in vitro test methods in human hepatic drug biotransformation research: pros and cons. Toxicol Appl Pharmacol 189:233–46
  • Brown HS, Griffin M, Houston JB. (2007). Evaluation of cryopreserved human hepatocytes as an alternative in vitro system to microsomes for the prediction of metabolic clearance. Drug Metab Dispos 35:293–301
  • Camilleri M. (2008). Novel pharmacology: asimadoline, a kappa-opioid agonist, and visceral sensation. Neurogastroenterol Motil 20:971–9
  • Chan TS, Yu H, Moore A, et al. (2013). Meeting the challenge of predicting hepatic clearance of compounds slowly metabolized by cytochrome P450 using a novel hepatocyte model, HepatoPac. Drug Metab Dispos 41:2024–32
  • Cook D, Brown D, Alexander R, et al. (2014). Lessons learned from the fate of AstraZeneca's drug pipeline: a five-dimensional framework. Nat Rev Drug Discov 13:419–31
  • Dolgos H, Trusheim M, Gross D, et al. (2016). Translational medicine guide transforms drug development processes: the recent Merck experience. Drug Discov Today 21:517–26
  • Donato MT, Castell JV. (2003). Strategies and molecular probes to investigate the role of cytochrome P450 in drug metabolism: focus on in vitro studies. Clin Pharmacokinet 42:153–78
  • Grime K, Riley RJ. (2006). The impact of in vitro binding on in vitro-in vivo extrapolations, projections of metabolic clearance and clinical drug-drug interactions. Curr Drug Metab 7:251–64
  • Hallifax D, Galetin A, Houston JB. (2008). Prediction of metabolic clearance using fresh human hepatocytes: comparison with cryopreserved hepatocytes and hepatic microsomes for five benzodiazepines. Xenobiotica 38:353–67
  • Hansch C, Leo A, Hoekman D. (1995). In exploring QSAR. hydrophobic, electronic and steric constants. Washington (DC): American Chemical Society
  • Hewitt NJ, Lechón MJ, Houston JB, et al. (2007). Primary hepatocytes: current understanding of the regulation of metabolic enzymes and transporter proteins, and pharmaceutical practice for the use of hepatocytes in metabolism, enzyme induction, transporter, clearance, and hepatotoxicity studies. Drug Metab Rev 39:159–234
  • Hosea NA, Collard WT, Cole S, et al. (2009). Prediction of human pharmacokinetics from preclinical information: comparative accuracy of quantitative prediction approaches. J Clin Pharmacol 49:513–33
  • Hosey CM, Benet LZ. (2015). Predicting the extent of metabolism using in vitro permeability rate measurements and in silico permeability rate predictions. Mol Pharm 12:1456–66
  • Houston JB. (1994). Utility of in vitro drug metabolism data in predicting in vivo metabolic clearance. Biochem Pharmacol 47:1469–79
  • Ito K, Houston JB. (2004). Comparison of the use of liver models for predicting drug clearance using in vitro kinetic data from hepatic microsomes and isolated hepatocytes. Pharm Res 21:785–92
  • Ito K, Houston JB. (2005). Prediction of human drug clearance from in vitro and preclinical data using physiologically based and empirical approaches. Pharm Res 22:103–12
  • Kilford PJ, Gertz M, Houston JB, Galetin A. (2008). Hepatocellular binding of drugs: correction for unbound fraction in hepatocyte incubations using microsomal binding or drug lipophilicity data. Drug Metab Dispos 36:1194–7
  • Kovar A, Peters T, Beier N, Derendorf H. (2001). Pharmacokinetic/pharmacodynamic evaluation of the NHE inhibitor eniporide. J Clin Pharmacol 41:139–48
  • Krösser S, Wolna P, Fischer TZ, et al. (2015). Effect of ceralifimod (ONO-4641) on lymphocytes and cardiac function: randomized, double-blind, placebo-controlled trial with an open-label fingolimod arm. J Clin Pharmacol 55:1051–60
  • Luzier A, Morse GD. (1993). Intravascular distribution of zidovudine: role of plasma proteins and whole blood components. Antiviral Res 21:267–80
  • McGinnity DF, Soars MG, Urbanowicz RA, Riley RJ. (2004). Evaluation of fresh and cryopreserved hepatocytes as in vitro drug metabolism tools for the prediction of metabolic clearance. Drug Metab Dispos 32:1247–53
  • Nordell P, Svanberg P, Bird J, Grime K. (2013). Predicting metabolic clearance for drugs that are actively transported into hepatocytes: incubational binding as a consequence of in vitro hepatocyte concentration is a key factor. Drug Metab Dispos 41:836–43
  • Obach RS. (1999). Prediction of human clearance of twenty-nine drugs from hepatic microsomal intrinsic clearance data: an examination of in vitro half-life approach and nonspecific binding to microsomes. Drug Metab Dispos 27:1350–9
  • Poulin P, Haddad S. (2013). Toward a new paradigm for the efficient in vitro-in vivo extrapolation of metabolic clearance in humans from hepatocyte data. J Pharm Sci 102:3239–51
  • Poulin P, Cornelis Hop ECA, Ho Q, et al. (2012a). Comparative assessment of in vitro–in vivo extrapolation methods used for predicting hepatic metabolic clearance of drugs. J Pharm Sci 101:4308–26
  • Poulin P, Kenny JR, Eca H, Haddad S. (2012b). In vitro–in vivo extrapolation of clearance: modeling hepatic metabolic clearance of highly bound drugs and comparative assessment with existing methods. J Pharm Sci 101:838–51
  • Ravandi F, Pigneux A, DeAngelo DJ, et al. (2015). Clinical, pharmacokinetic and pharmacodynamic data for the MEK1/2 inhibitor pimasertib in patients with advanced hematologic malignancies. Blood Cancer J 5:e375
  • Richard B, Launay-Iliadis MC, Iliadis A, et al. (1992). Pharmacokinetics of mitoxantrone in cancer patients treated by high-dose chemotherapy and autologous bone marrow transplantation. Br J Cancer 65:399–404
  • Riley RJ, McGinnity DF, Austin RP. (2005). A unified model for predicting human hepatic, metabolic clearance from in vitro intrinsic clearance data in hepatocytes and microsomes. Drug Metab Dispos 33:1304–11
  • Ring BJ, Chien JY, Adkison KK, et al. (2011). PhRMA CPCDC initiative on predictive models of human pharmacokinetics, part 3: comparative assessement of prediction methods of human clearance. J Pharm Sci 100:4090–110
  • Schaefer M, Schänzle G, Bischoff D, Süssmuth RD. (2016). Upcyte human hepatocytes: a potent in vitro tool for the prediction of hepatic clearance of metabolically stable compounds. Drug Metab Dispos 44:435–44
  • Scheible H, Kraetzer F, Johne A, Wimmer E. (in manuscript). Metabolism of the MEK1/2 inhibitor pimasertib involves a novel conjugation with phospho ethanolamine in patients with solid tumors
  • Soars MG, Grime K, Sproston JL, et al. (2007). Use of hepatocytes to assess the contribution of hepatic uptake to clearance in vivo. Drug Metab Dispos 35:859–65
  • Sohlenius-Sternbeck AK, Afzelius L, Prusis P, et al. (2010). Evaluation of the human prediction of clearance from hepatocyte and microsome intrinsic clearance for 52 drug compounds. Xenobiotica 40:637–49
  • Sohlenius-Sternbeck AK, Jones C, Ferguson D, et al. (2012). Practical use of the regression offset approach for the prediction of in vivo intrinsic clearance from hepatocytes. Xenobiotica 42:841–53
  • Stringer R, Nicklin PL, Houston JB. (2008). Reliability of human cryopreserved hepatocytes and liver microsomes as in vitro systems to predict metabolic clearance. Xenobiotica 38:1313–29
  • Tang H, Hussain A, Leal M, et al. (2007). Interspecies prediction of human drug clearance based on scaling data from one or two animal species. Drug Metab Dispos 35:1886–93
  • Williams JA, Hyland R, Jones BC, et al. (2004). Drug-drug interactions for UDP-glucuronosyltransferase substrates: a pharmacokinetic explanation for typically observed low exposure (AUCi/AUC) ratios. Drug Metabol Dispos 32:1201–8

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.