Effect of Ritonavir on (99m)Technetium-Mebrofenin Disposition in Humans: A Semi-PBPK Modeling and In Vitro Approach to Predict Transporter-Mediated DDIs

N D Pfeifer, S L Goss, B Swift, G Ghibellini, M Ivanovic, W D Heizer, L M Gangarosa, K L R Brouwer, N D Pfeifer, S L Goss, B Swift, G Ghibellini, M Ivanovic, W D Heizer, L M Gangarosa, K L R Brouwer

Abstract

A semiphysiologically based pharmacokinetic (semi-PBPK) model was developed to describe a unique blood, liver, and bile clinical data set for the hepatobiliary imaging agent (99m)Technetium-mebrofenin ((99m)Tc-mebrofenin), and to simulate sites/mechanisms of a (99m)Tc-mebrofenin-ritonavir drug-drug interaction (DDI). The transport inhibitor ritonavir (multiple-dose: 2 × 300 mg) significantly increased systemic (99m)Tc-mebrofenin exposure as compared with control (4,464 ± 1,861 vs. 1,970 ± 311 nCi min/ml; mean ± SD), without affecting overall hepatic exposure or biliary recovery. A novel extrahepatic distribution compartment was required to characterize (99m)Tc-mebrofenin disposition. Ritonavir inhibited (99m)Tc-mebrofenin accumulation in human sandwich-cultured hepatocytes (SCH) (half maximal inhibitory concentration (IC50) = 3.46 ± 1.53 µmol/l). Despite ritonavir accumulation in hepatocytes, intracellular binding was extensive (97. 6%), which limited interactions with multidrug resistance protein 2 (MRP2)-mediated biliary excretion. These in vitro data supported conclusions from modeling/simulation that ritonavir inhibited (99m)Tc-mebrofenin hepatic uptake, but not biliary excretion, at clinically relevant concentrations. This integrated approach, utilizing modeling, clinical, and in vitro data, emphasizes the importance of hepatic and extrahepatic distribution, assessment of inhibitory potential in relevant in vitro systems, and intracellular unbound concentrations to assess transporter-mediated hepatic DDIs.CPT: Pharmacometrics & Systems Pharmacology (2013) 2, e20; doi:10.1038/psp.2012.21; advance online publication 2 January 2013.

Figures

Figure 1
Figure 1
99mTc–mebrofenin (a) blood concentration vs. time curves; (b) liver scintigraphy vs. time curves by treatment group (red shaded area represents mean ± SD of control); (c) liver-to-blood ratio vs. time curves; mean data by treatment group. Data are presented as group means (±SD in (a) and (b)); red = control group, open/dashed = 200 mg ritonavir group, closed/solid = 2 × 300 mg ritonavir group.
Figure 2
Figure 2
Semi-PBPK model scheme (a) representing 99mTc–mebrofenin disposition in humans (Q denotes blood flow (ml/min), subscripts represent tissue or vascular compartments, as follows: liver (h), bile (bile), extrahepatic (“other”) tissue (o); clearance values are designated as CLuptake for influx from blood into tissue, CLefflux for efflux from tissue to blood, and CLbiliary for excretion from liver into bile). Simulations based on the semi-PBPK model and observed blood, liver, and bile curves for 99mTc–mebrofenin in subjects with quantitative scintigraphy data for (b) subject 8 (control), and (c) subject 17 (2 × 300 mg ritonavir). Circles represent observed blood samples, solid lines represent simulated blood concentrations, + symbols represent observed liver concentrations from attenuation-corrected (quantitative) scintigraphy data of the liver region of interest, dashed lines represent simulated liver concentrations, squares represent observed mass recovered in bile (corrected for gallbladder ejection fraction), and dotted lines represent simulated biliary excretion data. PBPK, physiologically based pharmacokinetic.
Figure 3
Figure 3
Sensitivity analysis of parameter estimates determined from the semiphysiologically based pharmacokinetic model (Figure 2; Table 2). Parameters were altered 10-fold in either direction of the values estimated for mean control data (Table 2), and the fold-change in the predicted study end points (99mTc–mebrofenin central compartment (blood) exposure, hepatic exposure, and biliary excretion) were examined. Solid lines represent influx clearance (CLuptake) from blood into tissue, dashed lines represent efflux clearance (CLefflux) from tissue to blood, and gray lines represent biliary excretion from liver into bile (CLbiliary). AUC, blood concentration–time curve.
Figure 4
Figure 4
Simulations based on the 99mTc–mebrofenin semiphysiologically based pharmacokinetic model scheme (Figure 2) for 99mTc–mebrofenin (a) blood, and (b) liver concentration–time data, and (c) cumulative % dose excreted in bile resulting from parameters describing mean control data (Table 2) and the following changes in transport-mediated processes, alone and in combination. Mean control data (red); 1/3 CLuptake,h + 3x CLefflux,h (blue); 1/2 CLuptake,h and CLuptake,o + 2x CLefflux,h and CLefflux,o (dashed black); 1/2 CLbiliary (solid black); mean control data without extrahepatic compartment (gray).
Figure 5
Figure 5
Human sandwich-cultured hepatocytes (SCH) were incubated for 10 min at 37 °C with 0.5 µCi/ml 99mTc–mebrofenin in standard (cells + bile) or Ca+-free (cells) HBSS (using B-CLEAR technology) alone and with various concentrations of ritonavir in SCH from three separate donors, in triplicate. (a) 99mTc–mebrofenin accumulation and biliary excretion index (BEI) in cells + bile (closed bars) and cells (open bars) in the presence of increasing concentrations of extracellular ritonavir. 99mTc–mebrofenin BEI values (above the bars) represent mean data. (b) Ritonavir accumulation in hepatocytes (Ca2+-free HBSS) when coadministered with 99mTc–mebrofenin, represented as intracellular total concentration (Ccell,total), as described in Methods section. 99mTc–mebrofenin and ritonavir accumulation data are represented as mean ± SEM of three separate donors in triplicate (5 µmol/l data are mean ± range of two donors, in triplicate).

References

    1. US Food and Drug Administration . US Department of Health and Human Services, FDA, Center for Drug Use and Evaluation (CDER), Silver Spring, MD; 2012. Draft Guidance for Industry: Drug Interaction Studies, Study Design, Data Analysis, Implications for Dosing, and Labeling Recommendations.
    1. European Medical Agency . Committee for Human Medicinal Products (CHMP), London, UK; 2012. Guideline on the Investigation of Drug Interactions.
    1. Funk C. The role of hepatic transporters in drug elimination. Expert Opin. Drug Metab. Toxicol. 2008;4:363–379.
    1. Pauli-Magnus C., &, Meier P.J. Hepatobiliary transporters and drug-induced cholestasis. Hepatology. 2006;44:778–787.
    1. Hendrikse N.H., Franssen E.J., van der Graaf W.T., Vaalburg W., &, de Vries E.G. Visualization of multidrug resistance in vivo. Eur. J. Nucl. Med. 1999;26:283–293.
    1. Michael al. Relationship of hepatic functional imaging to irinotecan pharmacokinetics and genetic parameters of drug elimination J. Clin. Oncol 244228–4235.2006
    1. Wong al. Predictors of vinorelbine pharmacokinetics and pharmacodynamics in patients with cancer J. Clin. Oncol 242448–2455.2006
    1. Chen al. Detection of in vivo P-glycoprotein inhibition by PSC 833 using Tc-99m sestamibi Clin. Cancer Res 3545–552.1997
    1. Ghibellini G., Leslie E.M., Pollack G.M., &, Brouwer K.L.R. Use of tc-99m mebrofenin as a clinical probe to assess altered hepatobiliary transport: integration of in vitro, pharmacokinetic modeling, and simulation studies. Pharm. Res. 2008;25:1851–1860.
    1. Ghibellini G., Johnson B.M., Kowalsky R.J., Heizer W.D., &, Brouwer K.L.R. A novel method for the determination of biliary clearance in humans. AAPS J. 2004;6:e33.
    1. Giacomini al. Membrane transporters in drug development Nat Rev Drug Discov 9215–236.2010
    1. Swift B., Pfeifer N.D., &, Brouwer K.L.R. Sandwich-cultured hepatocytes: an in vitro model to evaluate hepatobiliary transporter-based drug interactions and hepatotoxicity. Drug Metab. Rev. 2010;42:446–471.
    1. Balon al. Procedure guideline for hepatobiliary scintigraphy. Society of Nuclear Medicine J. Nucl. Med 381654–1657.1997
    1. Doo E., Krishnamurthy G.T., Eklem M.J., Gilbert S., &, Brown P.H. Quantification of hepatobiliary function as an integral part of imaging with technetium-99m-mebrofenin in health and disease. J. Nucl. Med. 1991;32:48–57.
    1. Loberg M.D., Cooper M., Harvey E., Callery P., &, Faith W. Development of new radiopharmaceuticals based on N-substitution of iminodiacetic acid. J. Nucl. Med. 1976;17:633–638.
    1. Swift B., Yue W., &, Brouwer K.L.R. Evaluation of (99m)technetium-mebrofenin and (99m)technetium-sestamibi as specific probes for hepatic transport protein function in rat and human hepatocytes. Pharm. Res. 2010;27:1987–1998.
    1. de Graaf al. Transporters involved in the hepatic uptake of (99m)Tc-mebrofenin and indocyanine green J. Hepatol 54738–745.2011
    1. Bhargava al. Adenosine triphosphate-binding cassette subfamily C member 2 is the major transporter of the hepatobiliary imaging agent (99m)Tc-mebrofenin J. Nucl. Med 501140–1146.2009
    1. Pinós T., Figueras C., &, Herranz R. Scintigraphic diagnosis of Dubin-Johnson syndrome: DISIDA is also useful. Am. J. Gastroenterol. 1991;86:1687–1688.
    1. Hendrikse al. In vivoimaging of hepatobiliary transport function mediated by multidrug resistance associated protein and P-glycoprotein Cancer Chemother. Pharmacol 54131–138.2004
    1. Pinós T., Constansa J.M., Palacin A., &, Figueras C. A new diagnostic approach to the Dubin-Johnson syndrome. Am. J. Gastroenterol. 1990;85:91–93.
    1. Hirsch M.S. Initiating therapy: when to start, what to use. J. Infect. Dis. 2008;197suppl. 3:S252–S260.
    1. Vourvahis M., &, Kashuba A.D. Mechanisms of pharmacokinetic and pharmacodynamic drug interactions associated with ritonavir-enhanced tipranavir. Pharmacotherapy. 2007;27:888–909.
    1. Huisman al. Multidrug resistance protein 2 (MRP2) transports HIV protease inhibitors, and transport can be enhanced by other drugs AIDS 162295–2301.2002
    1. Ye Z.W., Camus S., Augustijns P., &, Annaert P. Interaction of eight HIV protease inhibitors with the canalicular efflux transporter ABCC2 (MRP2) in sandwich-cultured rat and human hepatocytes. Biopharm. Drug Dispos. 2010;31:178–188.
    1. Gutmann H., Fricker G., Drewe J., Toeroek M., &, Miller D.S. Interactions of HIV protease inhibitors with ATP-dependent drug export proteins. Mol. Pharmacol. 1999;56:383–389.
    1. Ye Z.W., Augustijns P., &, Annaert P. Cellular accumulation of cholyl-glycylamido-fluorescein in sandwich-cultured rat hepatocytes: kinetic characterization, transport mechanisms, and effect of human immunodeficiency virus protease inhibitors. Drug Metab. Dispos. 2008;36:1315–1321.
    1. Annaert P., Ye Z.W., Stieger B., &, Augustijns P. Interaction of HIV protease inhibitors with OATP1B1, 1B3, and 2B1. Xenobiotica. 2010;40:163–176.
    1. Krishnamurthy S., &, Krishnamurthy G.T. Technetium-99m-iminodiacetic acid organic anions: review of biokinetics and clinical application in hepatology. Hepatology. 1989;9:139–153.
    1. Lee J.K., Brouwer K.R. Determination of intracellular volume of rat and human sandwich-cultured hepatocytes (Abstract ID 1595) The Toxicologist, Supplement to Toxicological Sciences. 2010;114:339.
    1. Chervu L.R., Nunn A.D., &, Loberg M.D. Radiopharmaceuticals for hepatobiliary imaging. Semin. Nucl. Med. 1982;12:5–17.
    1. Hawkins al. Radionuclide evaluation of liver transplants Semin. Nucl. Med 18199–212.1988
    1. Brown P.H., Juni J.E., Lieberman D.A., &, Krishnamurthy G.T. Hepatocyte versus biliary disease: a distinction by deconvolutional analysis of technetium-99m IDA time-activity curves. J. Nucl. Med. 1988;29:623–630.
    1. Peters A.M., Myers M.J., Mohammadtaghi S., Mubashar M., &, Mathie R.T. Bidirectional transport of iminodiacetic organic anion analogues between plasma and hepatocyte. Eur. J. Nucl. Med. 1998;25:766–773.
    1. Ito al. Which concentration of the inhibitor should be used to predict in vivo drug interactions from in vitro data AAPS PharmSci 4E25.2002
    1. van Heeswijk al. Time-dependent interaction between lopinavir/ritonavir and fexofenadine J. Clin. Pharmacol 46758–767.2006
    1. Jones al. Mechanistic pharmacokinetic modeling for the prediction of transporter-mediated disposition in humans from sandwich culture human hepatocyte data Drug Metab. Dispos 401007–1017.2012
    1. Ménochet K., Kenworthy K.E., Houston J.B., &, Galetin A. Use of mechanistic modeling to assess interindividual variability and interspecies differences in active uptake in human and rat hepatocytes. Drug Metab. Dispos. 2012;40:1744–1756.
    1. Abe K., Bridges A.S., Yue W., &, Brouwer vitrobiliary clearance of angiotensin II receptor blockers and 3-hydroxy-3-methylglutaryl-coenzyme A reductase inhibitors in sandwich-cultured rat hepatocytes: comparison with in vivo biliary clearance J. Pharmacol. Exp. Ther 326983–990.2008
    1. Higgins J.W., Bedwell D.W., &, Zamek-Gliszczynski M.J. Ablation of both organic cation transporter (OCT)1 and OCT2 alters metformin pharmacokinetics but has no effect on tissue drug exposure and pharmacodynamics. Drug Metab. Dispos. 2012;40:1170–1177.
    1. Watanabe T., Kusuhara H., Maeda K., Shitara Y., &, Sugiyama Y. Physiologically based pharmacokinetic modeling to predict transporter-mediated clearance and distribution of pravastatin in humans. J. Pharmacol. Exp. Ther. 2009;328:652–662.
    1. Poirier A., Funk C., Scherrmann J.M., &, Lavé T. Mechanistic modeling of hepatic transport from cells to whole body: application to napsagatran and fexofenadine. Mol. Pharm. 2009;6:1716–1733.
    1. Jones H.M., Parrott N., Jorga K., &, Lavé T. A novel strategy for physiologically based predictions of human pharmacokinetics. Clin. Pharmacokinet. 2006;45:511–542.
    1. Kawai R., Mathew D., Tanaka C., &, Rowland M. Physiologically based pharmacokinetics of cyclosporine A: extension to tissue distribution kinetics in rats and scale-up to human. J. Pharmacol. Exp. Ther. 1998;287:457–468.
    1. Takeda al. Evidence for a role of human organic anion transporters in the muscular side effects of HMG-CoA reductase inhibitors Eur. J. Pharmacol 483133–138.2004
    1. Knauer al. Human skeletal muscle drug transporters determine local exposure and toxicity of statins Circ. Res 106297–306.2010
    1. Rezk N.L., Crutchley R.D., Yeh R.F., &, Kashuba A.D. Full validation of an analytical method for the HIV-protease inhibitor atazanavir in combination with 8 other antiretroviral agents and its applicability to therapeutic drug monitoring. Ther. Drug Monit. 2006;28:517–525.
    1. Jung B.H., Rezk N.L., Bridges A.S., Corbett A.H., &, Kashuba A.D. Simultaneous determination of 17 antiretroviral drugs in human plasma for quantitative analysis with liquid chromatography-tandem mass spectrometry. Biomed. Chromatogr. 2007;21:1095–1104.
    1. Leslie E.M., Mao Q., Oleschuk C.J., Deeley R.G., &, Cole S.P. Modulation of multidrug resistance protein 1 (MRP1/ABCC1) transport and atpase activities by interaction with dietary flavonoids. Mol. Pharmacol. 2001;59:1171–1180.
    1. Kalvass J.C., &, Maurer T.S. Influence of nonspecific brain and plasma binding on CNS exposure: implications for rational drug discovery. Biopharm. Drug Dispos. 2002;23:327–338.

Source: PubMed

3
Subscribe