Publication Cover
Xenobiotica
the fate of foreign compounds in biological systems
Volume 50, 2020 - Issue 8
484
Views
0
CrossRef citations to date
0
Altmetric
General Xenobiochemistry

Quantitative evaluation of hepatic and intestinal induction of CYP3A in clinical practice

, , , , &
Pages 875-884 | Received 03 Nov 2019, Accepted 27 Dec 2019, Published online: 07 Jan 2020

References

  • Actos label. Actos (pioglitazone hydrochloride) [product label]. Lincolnshire, IL: Takeda Pharmaceuticals America; 2017. Available at: https://www.accessdata.fda.gov/scripts/cder/daf/.
  • Almond LM, Mukadam S, Gardner I, et al. (2016). Prediction of drug-drug interactions arising from CYP3A induction using a physiologically based dynamic model. Drug Metab Dispos 44:821–32.
  • Asaumi R, Toshimoto K, Tobe Y, et al. (2018). Comprehensive PBPK model of rifampicin for quantitative prediction of complex drug-drug interactions: CYP3A/2C9 induction and OATP inhibition effects. CPT Pharmacometrics Syst Pharmacol 7:186–96.
  • Back DJ, Bates M, Bowden A, et al. (1980). The interaction of phenobarbital and other anticonvulsants with oral contraceptive steroid therapy. Contraception 22:495–503.
  • Bailey I, Gibson GG, Plant K, et al. (2011). A PXR-mediated negative feedback loop attenuates the expression of CYP3A in response to the PXR agonist pregnenalone-16alpha-carbonitrile. PLoS One 6:e16703.
  • Baneyx G, Parrott N, Meille C, et al. (2014). Physiologically based pharmacokinetic modeling of CYP3A4 induction by rifampicin in human: influence of time between substrate and inducer administration. Eur J Pharm Sci 56:1–15.
  • Bjorkhem-Bergman L, Backstrom T, Nylen H, et al. (2013). Comparison of endogenous 4beta-hydroxycholesterol with midazolam as markers for CYP3A4 induction by rifampicin. Drug Metab Dispos 41:1488–93.
  • Diczfalusy U, Kanebratt KP, Bredberg E, et al. (2009). 4beta-hydroxycholesterol as an endogenous marker for CYP3A4/5 activity. Stability and half-life of elimination after induction with rifampicin. Br J Clin Pharmacol 67:38–43.
  • Diczfalusy U, Miura J, Roh HK, et al. (2008). 4Beta-hydroxycholesterol is a new endogenous CYP3A marker: relationship to CYP3A5 genotype, quinine 3-hydroxylation and sex in Koreans, Swedes and Tanzanians. Pharmacogenet Genomics 18:201–8.
  • Dutreix C, Lorenzo S, Wang Y. (2014). Comparison of two endogenous biomarkers of CYP3A4 activity in a drug-drug interaction study between midostaurin and rifampicin. Eur J Clin Pharmacol 70:915–20.
  • Eeckhoudt SL, Desager JP, Robert AR, et al. (2001). Midazolam and cortisol metabolism before and after CYP3A induction in humans. Int J Clin Pharmacol Ther 39:293–9.
  • Eichelbaum M, Albrecht M, Kliems G, et al. (1980). Influence of meso-caval shunt surgery on verapamil kinetics, bioavailability and response. Br J Clin Pharmacol 10:527–30.
  • Einolf HJ, Chen L, Fahmi OA, et al. (2014). Evaluation of various static and dynamic modeling methods to predict clinical CYP3A induction using in vitro CYP3A4 mRNA induction data. Clin Pharmacol Ther 95:179–88.
  • European Medicines Agency. (2012). Guideline on the investigation of drug interactions. London: EMA.
  • Fahmi OA, Boldt S, Kish M, et al. (2008a). Prediction of drug-drug interactions from in vitro induction data: application of the relative induction score approach using cryopreserved human hepatocytes. Drug Metab Dispos 36:1971–4.
  • Fahmi OA, Hurst S, Plowchalk D, et al. (2009). Comparison of different algorithms for predicting clinical drug-drug interactions, based on the use of CYP3A4 in vitro data: predictions of compounds as precipitants of interaction. Drug Metab Dispos 37:1658–66.
  • Fahmi OA, Maurer TS, Kish M, et al. (2008b). A combined model for predicting CYP3A4 clinical net drug-drug interaction based on CYP3A4 inhibition, inactivation, and induction determined in vitro. Drug Metab Dispos 36:1698–708.
  • Fahmi OA, Ripp SL. (2010). Evaluation of models for predicting drug-drug interactions due to induction. Expert Opin Drug Metab Toxicol 6:1399–416.
  • US Food and Drug Administration. (2017). Guidance for industry-drug interaction studies-in vitro metabolism and transporter‐mediated drug‐drug interaction studies. Rockville, MD: FDA.
  • Fleishaker JC, Pearson LK, Peters GR. (1995). Phenytoin causes a rapid increase in 6 beta-hydroxycortisol urinary excretion in humans–a putative measure of CYP3A induction. J Pharm Sci 84:292–4.
  • Hassan R, Ameen SS, Al Maruf A, et al. (2013). Genotype-phenotype variability in human CYP3A locus in Nepalese people residing in Bangladesh. Int J Clin Pharmacol Ther 51:207–14.
  • Holtbecker N, Fromm MF, Kroemer HK, et al. (1996). The nifedipine-rifampin interaction. Evidence for induction of gut wall metabolism. Drug Metab Dispos 24:1121–3.
  • Horsmans Y, Desager JP, Pauwels S, et al. (1991). Lack of effect by nifedipine on hepatic mixed function oxidase in man. Fundam Clin Pharmacol 5:193–201.
  • Kanebratt KP, Andersson TB. (2008). HepaRG cells as an in vitro model for evaluation of cytochrome P450 induction in humans. Drug Metab Dispos 36:137–45.
  • Kanebratt KP, Diczfalusy U, Backstrom T, et al. (2008). Cytochrome P450 induction by rifampicin in healthy subjects: determination using the Karolinska cocktail and the endogenous CYP3A4 marker 4beta-hydroxycholesterol. Clin Pharmacol Ther 84:589–94.
  • Kaneko A, Kato M, Endo C, et al. (2010). Prediction of clinical CYP3A4 induction using cryopreserved human hepatocytes. Xenobiotica 40:791–9.
  • Kaneko A, Kato M, Sekiguchi N, et al. (2009). In vitro model for the prediction of clinical CYP3A4 induction using HepaRG cells. Xenobiotica 39:803–10.
  • Kasichayanula S, Boulton DW, Luo WL, et al. (2014). Validation of 4beta-hydroxycholesterol and evaluation of other endogenous biomarkers for the assessment of CYP3A activity in healthy subjects. Br J Clin Pharmacol 78:1122–34.
  • Kato M, Chiba K, Horikawa M, et al. (2005). The quantitative prediction of in vivo enzyme-induction caused by drug exposure from in vitro information on human hepatocytes. Drug Metab Pharmacokinet 20:236–43.
  • Keung A, Reith K, Eller MG, et al. (1999). Enzyme induction observed in healthy volunteers after repeated administration of rifapentine and its lack of effect on steady-state rifapentine pharmacokinetics: part I. Int J Tuberc Lung Dis 3:426–36.
  • Kharasch ED, Francis A, London A, et al. (2011). Sensitivity of intravenous and oral alfentanil and pupillary miosis as minimal and noninvasive probes for hepatic and first-pass CYP3A induction. Clin Pharmacol Ther 90:100–8.
  • Kharasch ED, Walker A, Hoffer C, et al. (2004). Intravenous and oral alfentanil as in vivo probes for hepatic and first-pass cytochrome P450 3A activity: noninvasive assessment by use of pupillary miosis. Clin Pharmacol Ther 76:452–66.
  • Kharasch ED, Walker A, Hoffer C, et al. (2005). Evaluation of first‐pass cytochrome P4503A (CYP3A) and P‐glycoprotein activities using alfentanil and fexofenadine in combination. J Clin Pharmacol 45:79–88.
  • Kharasch ED, Walker A, Isoherranen N, et al. (2007). Influence of CYP3A5 genotype on the pharmacokinetics and pharmacodynamics of the cytochrome P4503A probes alfentanil and midazolam. Clin Pharmacol Ther 82:410–26.
  • Kharasch ED, Whittington D, Ensign D, et al. (2012). Mechanism of efavirenz influence on methadone pharmacokinetics and pharmacodynamics. Clin Pharmacol Ther 91:673–84.
  • Kleinbloesem CH, van Harten J, Wilson JP, et al. (1986). Nifedipine: kinetics and hemodynamic effects in patients with liver cirrhosis after intravenous and oral administration. Clin Pharmacol Ther 40:21–8.
  • Koup JR, Anderson GD, Loi CM. (1998). Effect of troglitazone on urinary excretion of 6beta-hydroxycortisol. J Clin Pharmacol 38:815–8.
  • Kozawa M, Honma M, Suzuki H. (2009). Quantitative prediction of in vivo profiles of CYP3A4 induction in humans from in vitro results with a reporter gene assay. Drug Metab Dispos 37:1234–41.
  • Kuehl P, Zhang J, Lin Y, et al. (2001). Sequence diversity in CYP3A promoters and characterization of the genetic basis of polymorphic CYP3A5 expression. Nat Genet 27:383–91.
  • LeBel M, Masson E, Guilbert E, et al. (1998). Effects of rifabutin and rifampicin on the pharmacokinetics of ethinylestradiol and norethindrone. J Clin Pharmacol 38:1042–50.
  • Leil TA, Kasichayanula S, Boulton DW, et al. (2014). Evaluation of 4beta-Hydroxycholesterol as a Clinical Biomarker of CYP3A4 Drug Interactions Using a Bayesian Mechanism-Based Pharmacometric Model. CPT Pharmacometrics Syst Pharmacol 3:e120.
  • Lucas RA, Gilfillan DJ, Bergstrom RF. (1998). A pharmacokinetic interaction between carbamazepine and olanzapine: observations on possible mechanism. Eur J Clin Pharmacol 54:639–43.
  • Nowak SN, Edwards DJ, Clarke A, et al. (2002). Pioglitazone: effect on CYP3A4 activity. J Clin Pharmacol 42:1299–302.
  • Ohnhaus EE, Park BK. (1979). Measurement of urinary 6-beta-hydroxycortisol excretion as an in vivo parameter in the clinical assessment of the microsomal enzyme-inducing capacity of antipyrine, phenobarbitone and rifampicin. Eur J Clin Pharmacol 15:139–45.
  • Ohno Y, Hisaka A, Ueno M, et al. (2008). General Framework for the Prediction of Oral Drug Interactions Caused by CYP3A4 Induction from In Vivo Information. Clinical Pharmacokinetics 47:669–80.
  • Park BK, Wilson AC, Kaatz G, et al. (1984). Enzyme induction by phenobarbitone and vitamin K1 disposition in man. Br J Clin Pharmacol 18:94–7.
  • Perucca E, Grimaldi R, Frigo GM, et al. (1988). Comparative effects of rifabutin and rifampicin on hepatic microsomal enzyme activity in normal subjects. Eur J Clin Pharmacol 34:595–9.
  • Perucca E, Ruprah M, Richens A, et al. (1981). Effect of low-dose phenobarbitone on five indirect indices of hepatic microsomal enzyme induction and plasma lipoproteins in normal subjects. Br J Clin Pharmacol 12:592–6.
  • Ministry of Health, Labour and Welfare, Japan. (2019). Guideline of drug interaction studies for drug development and appropriate provision of information. Tokyo, Japan: Pharmaceuticals and Medical Device Agency (PMDA).
  • Priftin label. Priftin (rifapentine) [product label]. Bridgewater, NJ: Sanofi-Aventis; 2014. Available at: https://www.accessdata.fda.gov/scripts/cder/daf/.
  • Pukrittayakamee S, Prakongpan S, Wanwimolruk S, et al. (2003). Adverse effect of rifampin on quinine efficacy in uncomplicated falciparum malaria. Antimicrob Agents Chemother 47:1509–13.
  • Roberts MS, Rowland M. (1986a). Correlation between in-vitro microsomal enzyme activity and whole organ hepatic elimination kinetics: analysis with a dispersion model. J Pharm Pharmacol 38:177–81.
  • Roberts MS, Rowland M. (1986b). A dispersion model of hepatic elimination: 2. Steady-state considerations–influence of hepatic blood flow, binding within blood, and hepatocellular enzyme activity. J Pharmacokinet Biopharm 14:261–88.
  • Schadt S, Simon S, Kustermann S, et al. (2015). Minimizing DILI risk in drug discovery - a screening tool for drug candidates. Toxicol In Vitro 30:429–37.
  • Shou M, Hayashi M, Pan Y, et al. (2008). Modeling, prediction, and in vitro in vivo correlation of CYP3A4 induction. Drug Metab Dispos 36:2355–70.
  • Staiger C, Schlicht F, Walter E, et al. (1983). Effect of single and multiple doses of sulphinpyrazone on antipyrine metabolism and urinary excretion of 6-beta-hydroxycortisol. Eur J Clin Pharmacol 25:797–801.
  • Tebbens JD, Azar M, Friedmann E, et al. (2018). Mathematical models in the description of pregnane X receptor (PXR)-regulated cytochrome P450 enzyme induction. Int J Mol Sci 19.
  • Tomlinson B, Young RP, Ng MC, et al. (1996). Selective liver enzyme induction by carbamazepine and phenytoin in Chinese epileptics. Eur J Clin Pharmacol 50:411–5.
  • Uchimura T, Kato M, Saito T, et al. (2010). Prediction of human blood-to-plasma drug concentration ratio. Biopharm Drug Dispos 31:286–97.
  • Vermet H, Raoust N, Ngo R, et al. (2016). Evaluation of normalization methods to predict CYP3A4 induction in six fully characterized cryopreserved human hepatocyte preparations and HepaRG cells. Drug Metab Dispos 44:50–60.
  • Watanabe T, Kusuhara H, Sugiyama Y. (2010). Application of physiologically based pharmacokinetic modeling and clearance concept to drugs showing transporter-mediated distribution and clearance in humans. J Pharmacokinet Pharmacodyn 37:575–90.
  • Weber C, Schmitt R, Birnboeck H, et al. (1999). Multiple-dose pharmacokinetics, safety, and tolerability of bosentan, an endothelin receptor antagonist, in healthy male volunteers. J Clin Pharmacol 39:703–14.
  • 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 Metab Dispos 32:1201–8.
  • Williams JA, Ring BJ, Cantrell VE, et al. (2002). Comparative metabolic capabilities of CYP3A4, CYP3A5, and CYP3A7. Drug Metab Dispos 30:883–91.
  • Yamashita F, Sasa Y, Yoshida S, et al. (2013). Modeling of rifampicin-induced CYP3A4 activation dynamics for the prediction of clinical drug-drug interactions from in vitro data. PLoS One 8:e70330.
  • Zhang JG, Ho T, Callendrello AL, et al. (2014). Evaluation of calibration curve-based approaches to predict clinical inducers and noninducers of CYP3A4 with plated human hepatocytes. Drug Metab Dispos 42:1379–91.
  • Zimmerlin A, Trunzer M, Faller B. (2011). CYP3A time-dependent inhibition risk assessment validated with 400 reference drugs. Drug Metab Dispos 39:1039–46.

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.