Skip to main page content
U.S. flag

An official website of the United States government

Dot gov

The .gov means it’s official.
Federal government websites often end in .gov or .mil. Before sharing sensitive information, make sure you’re on a federal government site.

Https

The site is secure.
The https:// ensures that you are connecting to the official website and that any information you provide is encrypted and transmitted securely.

Access keys NCBI Homepage MyNCBI Homepage Main Content Main Navigation
. 2019 Jun 18;21(5):77.
doi: 10.1208/s12248-019-0344-8.

Impact of Intracellular Concentrations on Metabolic Drug-Drug Interaction Studies

Affiliations

Impact of Intracellular Concentrations on Metabolic Drug-Drug Interaction Studies

Andrea Treyer et al. AAPS J. .

Abstract

Accurate prediction of drug-drug interactions (DDI) is a challenging task in drug discovery and development. It requires determination of enzyme inhibition in vitro which is highly system-dependent for many compounds. The aim of this study was to investigate whether the determination of intracellular unbound concentrations in primary human hepatocytes can be used to bridge discrepancies between results obtained using human liver microsomes and hepatocytes. Specifically, we investigated if Kpuu could reconcile differences in CYP enzyme inhibition values (Ki or IC50). Firstly, our methodology for determination of Kpuu was optimized for human hepatocytes, using four well-studied reference compounds. Secondly, the methodology was applied to a series of structurally related CYP2C9 inhibitors from a Roche discovery project. Lastly, the Kpuu values of three commonly used CYP3A4 inhibitors-ketoconazole, itraconazole, and posaconazole-were determined and compared to compound-specific hepatic enrichment factors obtained from physiologically based modeling of clinical DDI studies with these three compounds. Kpuu obtained in suspended human hepatocytes gave good predictions of system-dependent differences in vitro. The Kpuu was also in fair agreement with the compound-specific hepatic enrichment factors in DDI models and can therefore be used to improve estimations of enrichment factors in physiologically based pharmacokinetic modeling.

Keywords: drug-drug interaction; intracellular bioavailability; physiologically based pharmacokinetic modeling; scaling factor; unbound drug concentrations.

PubMed Disclaimer

Figures

Fig. 1
Fig. 1
a Comparison of fu,mic in human vs. rat liver microsomes. Rat values are derived from Brown et al. (20). b Comparison of Kpuu in human vs. rat hepatocytes. c Log fold difference of apparent and corrected (unbound) IC50 determined in human liver microsomes and human hepatocytes or Ki determined in rat microsomes and rat hepatocytes. The dotted lines represent a 2-fold difference. Error bars represent standard deviations. Apparent and corrected Ki or IC50 values are presented in Tables S1 and S2. Enox, E: enoxacin; Clar, C: clarithromycin; Saq, S: saquinavir; Nel, N: nelfinavir
Fig. 2
Fig. 2
Kpuu as correction factor of IC50 in the RO discovery series. a Structures of RO discovery compound. be Comparison of IC50 values measured in HLM and HH. Hepatocellular IC50,app were corrected with Kpuu (= Kp · fu,cell) in suspended HH and microsomal IC50,app values were corrected with fu,mic to obtain IC50,corr. The dotted line indicates the line of unity. The labels of the data points correspond to the structure numbers in panel (a)
Fig. 3
Fig. 3
Correlation of IVIVE correction factors with Kpuu. a Overview of correction factors used for the azole antifungal compounds in in vivo DDI studies. b Illustration of hepatic uptake scalars in rat and human DDI models (error bars represent range) vs. in vitro Kpuu in suspended HH (error bars represent SD)
Fig. 4
Fig. 4
Summary of use and interpretation of Kpuu in the context of CYP-mediated DDI

References

    1. Obach RS. Predicting drug-drug interactions from in vitro drug metabolism data: challenges and recent advances. Curr Opin Drug Discov Dev. 2009;12(1):81–89. - PubMed
    1. Min JS, Bae SK. Prediction of drug-drug interaction potential using physiologically based pharmacokinetic modeling. Arch Pharm Res. 2017;40(12):1356–1379. doi: 10.1007/s12272-017-0976-0. - DOI - PubMed
    1. Sekiguchi N, Higashida A, Kato M, Nabuchi Y, Mitsui T, Takanashi K, Aso Y, Ishigai M. Prediction of drug-drug interactions based on time-dependent inhibition from high throughput screening of cytochrome P450 3A4 inhibition. Drug Metab Pharmacokinet. 2009;24(6):500–510. doi: 10.2133/dmpk.24.500. - DOI - PubMed
    1. Zhuang X, Lu C. PBPK modeling and simulation in drug research and development. Acta Pharm Sin B. 2016;6(5):430–440. doi: 10.1016/j.apsb.2016.04.004. - DOI - PMC - PubMed
    1. Hewitt NJ, Lechon MJ, Houston JB, Hallifax D, Brown HS, Maurel P, et al. 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. 2007;39(1):159–234. doi: 10.1080/03602530601093489. - DOI - PubMed

Publication types

MeSH terms

Substances

LinkOut - more resources