1
|
Ramsden D, Perloff ES, Whitcher-Johnstone A, Ho T, Patel R, Kozminski KD, Fullenwider CL, Zhang JG. Predictive In Vitro-In Vivo Extrapolation for Time Dependent Inhibition of CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6 Using Pooled Human Hepatocytes, Human Liver Microsomes, and a Simple Mechanistic Static Model. Drug Metab Dispos 2022; 50:114-127. [PMID: 34789487 DOI: 10.1124/dmd.121.000718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Accepted: 11/12/2021] [Indexed: 11/22/2022] Open
Abstract
Inactivation of Cytochrome P450 (CYP450) enzymes can lead to significant increases in exposure of comedicants. The majority of reported in vitro to in vivo extrapolation (IVIVE) data have historically focused on CYP3A, leaving the assessment of other CYP isoforms insubstantial. To this end, the utility of human hepatocytes (HHEP) and human liver microsomes (HLM) to predict clinically relevant drug-drug interactions was investigated with a focus on CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6. Evaluation of IVIVE for CYP2B6 was limited to only weak inhibition. A search of the University of Washington Drug-Drug Interaction Database was conducted to identify a clinically relevant weak, moderate, and strong inhibitor for selective substrates of CYP1A2, CYP2C8, CYP2C9, CYP2C19, and CYP2D6, resulting in 18 inhibitors for in vitro characterization against 119 clinical interaction studies. Pooled human hepatocytes and HLM were preincubated with increasing concentrations of inhibitors for designated timepoints. Time dependent inhibition was detected in HLM for four moderate/strong inhibitors, suggesting that some optimization of incubation conditions (i.e., lower protein concentrations) is needed to capture weak inhibition. Clinical risk assessment was conducted by incorporating the in vitro derived kinetic parameters maximal rate of enzyme inactivation (min-1) (kinact) and concentration of inhibitor resulting in 50% of the maximum enzyme inactivation (KI) into static equations recommended by regulatory authorities. Significant overprediction was observed when applying the basic models recommended by regulatory agencies. Mechanistic static models, which consider the fraction of metabolism through the impacted enzyme, using the unbound hepatic inlet concentration lead to the best overall prediction accuracy with 92% and 85% of data from HHEPs and HLM, respectively, within twofold of the observed value. SIGNIFICANCE STATEMENT: Coupling time-dependent inactivation parameters derived from pooled human hepatocytes and human liver microsomes (HLM) with a mechanistic static model provides an easy and quantitatively accurate means to determine clinical drug-drug interaction risk from in vitro data. Optimization is needed to evaluate time-dependent inhibition (TDI) for weak and moderate inhibitors using HLM. Recommendations are made with respect to input parameters for in vitro to in vivo extrapolation (IVIVE) of TDI with non-CYP3A enzymes using available data from HLM and human hepatocytes.
Collapse
Affiliation(s)
- Diane Ramsden
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Elke S Perloff
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Andrea Whitcher-Johnstone
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Thuy Ho
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Reena Patel
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Kirk D Kozminski
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - Cody L Fullenwider
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| | - J George Zhang
- Takeda Development Center Americas, Inc., Cambridge, Massachusetts (D.R.); Corning Gentest Contract Research Services, Corning Life Sciences, Woburn, Massachusetts (E.S.P., T.H., R.P., J.G.Z.); Takeda Development Center Americas, Inc., San Diego, California (K.D.K., C.L.F.); and Boehringer Ingelheim Pharmaceuticals Inc., Ridgefield, Connecticut (A.W.-J.)
| |
Collapse
|
2
|
Identification of Erythromycin and Clarithromycin Metabolites Formed in Chicken Liver Microsomes Using Liquid Chromatography-High-Resolution Mass Spectrometry. Foods 2021; 10:foods10071504. [PMID: 34209740 PMCID: PMC8304660 DOI: 10.3390/foods10071504] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/24/2021] [Accepted: 06/25/2021] [Indexed: 11/17/2022] Open
Abstract
Nontargeted analysis can be used for the rapid screening and confirmatory analysis of veterinary drugs and their metabolites, which are important for the comprehensive safety evaluation of animal-derived foods. Here, a novel nontargeted screening approach based on liquid chromatography coupled with electrospray ionization-high-resolution mass spectrometry (LC/ESI-HR-MS) was developed to determine erythromycin, clarithromycin, and their metabolites in chicken liver microsomes. Erythromycin and clarithromycin were incubated in vitro in the presence of NADPH for 60 min to generate metabolites in chicken liver microsomes. After the incubation, the supernatant was extracted using ultrasonic shaking, orbital shaking, and centrifugation before analysis using LC/ESI-HR-MS in positive ion mode on an Agilent Eclipse Plus C18 column (100 mm × 2.1 mm; i.d. 3.5 µm) with 0.1 percent formic acid-water and acetonitrile as the mobile phases for gradient elution at 0.4 mL/min. The results show that erythromycin can produce N-desmethyl-erythromycin A in chicken liver microsomes, but clarithromycin cannot produce N-desmethyl-clarithromycin in chicken liver microsomes. The N-desmethyl-erythromycin A and N-desmethyl-clarithromycin were tentatively identified in chicken liver microsomes using the established quick analytic method, which will provide a theoretical foundation for future research on pharmacokinetics and drug elimination in poultry.
Collapse
|
3
|
Franchetti Y, Nolin TD. Simultaneous Assessment of Hepatic Transport and Metabolism Pathways with a Single Probe Using Individualized PBPK Modeling of 14CO 2 Production Rate Data. J Pharmacol Exp Ther 2019; 371:151-161. [PMID: 31399494 PMCID: PMC6750580 DOI: 10.1124/jpet.119.257212] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Accepted: 07/31/2019] [Indexed: 12/29/2022] Open
Abstract
Erythromycin is a substrate of cytochrome P4503A4 (CYP3A4) and multiple drug transporters. Although clinical evidence suggests that uptake transport is likely to play a dominant role in erythromycin's disposition, the relative contributions of individual pathways are unclear. Phenotypic evaluation of multiple pathways generally requires a probe drug cocktail. This approach can result in ambiguous conclusions due to imprecision stemming from overlapping specificity of multiple drugs. We hypothesized that an individualized physiologically based pharmacokinetic modeling approach incorporating 14CO2 production rates (iPBPK-R) of the erythromycin breath test (ERMBT) would enable us to differentiate the contribution of metabolic and transporter pathways to erythromycin disposition. A seven-compartmental physiologically based pharmacokinetic (PBPK) model was built for 14C-erythromycin administered intravenously. Transporter clearance and CYP3A4 clearance were embedded in hepatic compartments. 14CO2 production rates were simulated taking the first derivative of by-product 14CO2 concentrations. Parameters related to nonrenal elimination pathways were estimated by model fitting the ERMBT data of 12 healthy subjects individually. Optimized iPBPK-R models fit the individual rate data well. Using one probe, nine PBPK parameters were simultaneously estimated per individual. Maximum velocity of uptake transport, CYP3A4 clearance, total passive diffusion, and others were found to collectively control 14CO2 production rates. The median CYP3A4 clearance was 12.2% of the input clearance. Male subjects had lower CYP3A4 activity than female subjects by 11.3%. We applied iPBPK-R to ERMBT data to distinguish and simultaneously estimate the activity of multiple nonrenal elimination pathways in healthy subjects. The iPBPK-R framework is a novel tool for delineating rate-limiting and non-rate-limiting elimination pathways using a single probe. SIGNIFICANCE STATEMENT: Our developed individualized physiologically based pharmacokinetic modeling approach incorporating rate data (iPBPK-R) enabled us to distinguish and simultaneously estimate the activity of multiple nonrenal elimination pathways of erythromycin in healthy subjects. A new interpretation of erythromycin breath test (ERMBT) data was also obtained via iPBPK-R. We found that rate data have rich information allowing estimation of per-person PBPK parameters. This study serves as proof of principle that the iPBPK-R framework is a novel tool for delineating rate-limiting and non-rate-limiting elimination pathways using a single probe. iPBPK-R can be applied to other rate-derived data beyond ERMBT. Potential areas of application include drug-drug interaction, pathophysiological effects on drug disposition, and the role of biomarkers on hemodialysis efficiency utilizing estimated adjustment factors with correlation analysis.
Collapse
Affiliation(s)
- Yoko Franchetti
- Departments of Pharmaceutical Sciences (Y.F.) and Pharmacy and Therapeutics (T.D.N.), Center of Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| | - Thomas D Nolin
- Departments of Pharmaceutical Sciences (Y.F.) and Pharmacy and Therapeutics (T.D.N.), Center of Clinical Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania
| |
Collapse
|
4
|
Kumar V, Salphati L, Hop CECA, Xiao G, Lai Y, Mathias A, Chu X, Humphreys WG, Liao M, Heyward S, Unadkat JD. A Comparison of Total and Plasma Membrane Abundance of Transporters in Suspended, Plated, Sandwich-Cultured Human Hepatocytes Versus Human Liver Tissue Using Quantitative Targeted Proteomics and Cell Surface Biotinylation. Drug Metab Dispos 2019; 47:350-357. [PMID: 30622164 DOI: 10.1124/dmd.118.084988] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Accepted: 01/07/2019] [Indexed: 02/13/2025] Open
Abstract
Suspended (SH), plated (PH), and sandwich-cultured hepatocytes (SCH) are commonly used models to predict in vivo transporter-mediated hepatic uptake (SH or PH) or biliary (SCH) clearance of drugs. When doing so, the total and the plasma membrane abundance (PMA) of transporter are assumed not to differ between hepatocytes and liver tissue (LT). This assumption has never been tested. In this study, we tested this assumption by measuring the total and PMA of the transporters in human hepatocyte models versus LT (total only) from which they were isolated. Total abundance of OATP1B1/2B1/1B3, OCT1, and OAT2 was not significantly different between the hepatocytes and LT. The same was true for the PMA of these transporters across the hepatocyte models. In contrast, total abundance of the sinusoidal efflux transporter, MRP3, and the canalicular efflux transporters, MRP2 and P-gp, was significantly greater (P < 0.05) in SCH versus LT. Of the transporters tested, only the percentage of PMA of OATP1B1, P-gp, and MRP3, in SCH (82.8% ± 7.3%, 57.5% ± 10.9%, 69.3% ± 5.7%) was significantly greater (P < 0.05) than in SH (73.3% ± 6.4%, 27.4% ± 6.4%, 53.6% ± 4.1%). If the transporters measured in the plasma membrane are functional and the PMA in SH is representative of that in LT, these data suggest that SH, PH, and SCH will result in equal prediction of hepatic uptake clearance of drugs mediated by the transporters tested above. However, SCH will predict higher sinusoidal efflux and biliary clearance of drugs if the change in PMA of these transporters is not taken into consideration.
Collapse
Affiliation(s)
- Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Laurent Salphati
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Cornelis E C A Hop
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Guangqing Xiao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Yurong Lai
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Anita Mathias
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Xiaoyan Chu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - W Griffith Humphreys
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Mingxiang Liao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Scott Heyward
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Drug Metabolism and Pharmacokinetics, Biogen Idec, Cambridge, Massachusetts (G.X.); Departments of Clinical Research, Clinical Pharmacology, and Drug Metabolism and Pharmacokinetics, Gilead Sciences, Foster City, California (Y.L., A.M.); Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International, Cambridge, Massachusetts (M.L.); and BioIVT, Baltimore, Maryland (S.H.)
| |
Collapse
|
5
|
Involvement of the Transporters P-Glycoprotein and Breast Cancer Resistance Protein in Dermal Distribution of the Multikinase Inhibitor Regorafenib and Its Active Metabolites. J Pharm Sci 2017; 106:2632-2641. [PMID: 28479358 DOI: 10.1016/j.xphs.2017.04.064] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2017] [Revised: 04/07/2017] [Accepted: 04/24/2017] [Indexed: 01/07/2023]
Abstract
Regorafenib is a multikinase inhibitor orally administered to colorectal cancer patients, and is known to often exhibit dermal toxicity. The purpose of this study is to clarify possible involvement of P-glycoprotein and breast cancer resistance protein (BCRP) in the dermal accumulation of regorafenib and its active metabolites M-2 and M-5. Following intravenous administration in triple knockout (Abcb1a/1b/bcrp-/-; TKO) and wild-type (WT) mice, delayed plasma clearance of M-2 and M-5, but not regorafenib, was observed in TKO mice compared to WT mice. Elacridar, an inhibitor of both transporters, also caused delayed clearance of M-2 and M-5, suggesting that these transporters are involved in their elimination. Skin-to-plasma concentration ratios of regorafenib, M-2, and M-5 were significantly higher in TKO mice than in WT mice. Elacridar increased skin-to-plasma and epidermis-to-plasma concentration ratios of regorafenib. Basal-to-apical transport of M-2 and M-5 was observed in LLC-PK1-Pgp and MDCKII/BCRP/PDZK1 cells, which was inhibited by elacridar and the BCRP inhibitor Ko143, respectively. The present findings thus indicate that P-glycoprotein and BCRP are involved in the accumulation of regorafenib and its active metabolites in the skin, by affecting either their systemic exposure or their plasma distribution in the circulating blood.
Collapse
|
6
|
Zhao M, Wang S, Li F, Dong D, Wu B. Arylsulfatase B Mediates the Sulfonation-Transport Interplay in Human Embryonic Kidney 293 Cells Overexpressing Sulfotransferase 1A3. Drug Metab Dispos 2016; 44:1441-9. [PMID: 27325375 DOI: 10.1124/dmd.116.070938] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2016] [Accepted: 06/17/2016] [Indexed: 02/13/2025] Open
Abstract
Elucidating the intricate relationships between metabolic and transport pathways contributes to improved predictions of in vivo drug disposition and drug-drug interactions. Here we reported that inhibited excretion of conjugative metabolites [i.e., hesperetin 3'-O-sulfate (H3'S) and hesperetin 7-O-sulfate (H7S)] by MK-571 led to reduced metabolism of hesperetin (a maximal 78% reduction) in human embryonic kidney 293 cells overexpressing sulfotransferase 1A3 (named SULT293 cells). The strong dependence of cellular sulfonation on the efflux transport of generated sulfated metabolites revealed an interplay of sulfonation metabolism with efflux transport (or sulfonation-transport interplay). Polymerase chain reaction (PCR) and Western blot analyses demonstrated that SULT293 cells expressed multiple sulfatases such as arylsulfatase A (ARSA), ARSB, and ARSC. Of these three desulfonation enzymes, only ARSB showed significant activities toward hesperetin sulfates. The intrinsic clearance values for the hydrolysis of H3'S and H7S were estimated at 0.6 and 0.5 μl/h/mg, respectively. Furthermore, knockdown of ARSB attenuated the regulatory effect of efflux transporter on cellular sulfonation, whereas overexpression of ABSB enhanced the transporter effect. Taken together, the results indicated that ARSB mediated the sulfonation-transport interplay in SULT293 cells.
Collapse
Affiliation(s)
- Mengjing Zhao
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, China (M.Z., S.W., B.W.); and Ocular Surface Research Center and Institute of Ophthalmology, Jinan University School of Medicine, Guangzhou, China (F.L., D.D.)
| | - Shuai Wang
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, China (M.Z., S.W., B.W.); and Ocular Surface Research Center and Institute of Ophthalmology, Jinan University School of Medicine, Guangzhou, China (F.L., D.D.)
| | - Feng Li
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, China (M.Z., S.W., B.W.); and Ocular Surface Research Center and Institute of Ophthalmology, Jinan University School of Medicine, Guangzhou, China (F.L., D.D.)
| | - Dong Dong
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, China (M.Z., S.W., B.W.); and Ocular Surface Research Center and Institute of Ophthalmology, Jinan University School of Medicine, Guangzhou, China (F.L., D.D.)
| | - Baojian Wu
- Division of Pharmaceutics, College of Pharmacy, Jinan University, Guangzhou, China (M.Z., S.W., B.W.); and Ocular Surface Research Center and Institute of Ophthalmology, Jinan University School of Medicine, Guangzhou, China (F.L., D.D.)
| |
Collapse
|
7
|
De Bruyn T, Stieger B, Augustijns PF, Annaert PP. Clearance Prediction of HIV Protease Inhibitors in Man: Role of Hepatic Uptake. J Pharm Sci 2016. [DOI: 10.1002/jps.24564] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
8
|
High-content screening imaging and real-time cellular impedance monitoring for the assessment of chemical’s bio-activation with regards hepatotoxicity. Toxicol In Vitro 2015; 29:1916-31. [DOI: 10.1016/j.tiv.2015.07.024] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2015] [Revised: 06/29/2015] [Accepted: 07/30/2015] [Indexed: 02/07/2023]
|
9
|
Zhang X, Dong D, Wang H, Ma Z, Wang Y, Wu B. Stable knock-down of efflux transporters leads to reduced glucuronidation in UGT1A1-overexpressing HeLa cells: the evidence for glucuronidation-transport interplay. Mol Pharm 2015; 12:1268-78. [PMID: 25741749 DOI: 10.1021/mp5008019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Efflux of glucuronide is facilitated by the membrane transporters including BCRP and MRPs. In this study, we aimed to determine the effects of transporter expression on glucuronide efflux and cellular glucuronidation. Single efflux transporter (i.e., BCRP, MRP1, MRP3, or MRP4) was stably knocked-down in UGT1A1-overexpressing HeLa cells. Knock-down of transporters was performed by stable transfection of short-hairpin RNA (shRNA) using lentiviral vectors. Glucuronidation and glucuronide transport in the cells were characterized using three different aglycones (i.e., genistein, apigenin, and emodin) with distinct metabolic activities. BCRP knock-down resulted in significant reductions in excretion of glucuronides (42.9% for genistein glucuronide (GG), 21.1% for apigenin glucuronide (AG) , and 33.7% for emodin glucuronide (EG); p < 0.01) and in cellular glucuronidation (38.3% for genistein, 38.6% for apigenin, and 34.7% for emodin; p < 0.01). Knock-down of a MRP transporter led to substantial decreases in excretion of GG (32.3% for MRP1, 36.7% for MRP3, and 36.6% for MRP4; p < 0.01) and AG (59.3% for MRP1, 24.7% for MRP3, and 34.1% for MRP4; p < 0.01). Also, cellular glucuronidation of genistein (38.3% for MRP1, 32.3% for MRP3, and 31.1% for MRP4; p < 0.01) and apigenin (40.6% for MRP1, 32.4% for MRP3, and 34.6% for MRP4; p < 0.001) was markedly suppressed. By contrast, silencing of MRPs did not cause any changes in either excretion of EG or cellular glucuronidation of emodin. In conclusion, cellular glucuronidation was significantly altered by decreasing expression of efflux transporters, revealing a strong interplay of glucuronidation with efflux transport.
Collapse
Affiliation(s)
- Xingwang Zhang
- †Division of Pharmaceutics, College of Pharmacy, and ‡Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | | | - Huailing Wang
- †Division of Pharmaceutics, College of Pharmacy, and ‡Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | - Zhiguo Ma
- †Division of Pharmaceutics, College of Pharmacy, and ‡Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| | | | - Baojian Wu
- †Division of Pharmaceutics, College of Pharmacy, and ‡Guangzhou Jinan Biomedicine Research and Development Center, Jinan University, 601 Huangpu Avenue West, Guangzhou 510632, China
| |
Collapse
|
10
|
Lalande L, Charpiat B, Leboucher G, Tod M. Consequences of renal failure on non-renal clearance of drugs. Clin Pharmacokinet 2015; 53:521-32. [PMID: 24861189 DOI: 10.1007/s40262-014-0146-1] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Kidney disease not only alters the renal elimination but also the non-renal disposition of drugs that are metabolized by the liver. Indeed, modifications in the expression and activity of intestinal and hepatic drug metabolism enzymes and uptake and efflux transporters have been reported. Accumulated uremic toxins, inflammatory cytokines, and parathyroid hormones may modulate these proteins either directly or by inhibiting gene expression. This can lead to important unintended variations in exposure and response when drugs are administered without dose adjustment for reduced renal function. This review summarizes our current understanding of non-renal clearance in circumstances of chronic and acute renal failure with experimental but also clinical studies. It also evaluates the clinical impact on drug disposition. Predicting the extent of the drug disposition modification is difficult first because of the complex interplay between metabolic enzymes and transport proteins but also because of the differential effects in the different organs (liver, intestines). Recommendations of the US FDA are presented as they may be potentially helpful tools to predict these modifications when no specific pharmacokinetic studies are available.
Collapse
Affiliation(s)
- Laure Lalande
- Department of Pharmacy, Groupement Hospitalier Nord, Hospices Civils de Lyon, 103 Grande Rue de la Croix Rousse, 69317, Lyon Cedex 04, France,
| | | | | | | |
Collapse
|
11
|
Huang F, Moschetti V, Lang B, Halabi A, Petersen-Sylla M, Yong CL, Elgadi M. Pharmacokinetics, safety, and tolerability of faldaprevir in patients with renal impairment. Antimicrob Agents Chemother 2015; 59:251-7. [PMID: 25348520 PMCID: PMC4291359 DOI: 10.1128/aac.03359-14] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2014] [Accepted: 10/18/2014] [Indexed: 02/06/2023] Open
Abstract
Faldaprevir is a potent hepatitis C virus (HCV) NS3/4A protease inhibitor with negligible urinary excretion. We assessed the pharmacokinetics and safety of a single oral dose of faldaprevir (480 mg) in 32 HCV-negative subjects with renal impairment or normal renal function. Compared with subjects with normal renal function, the adjusted geometric mean ratios (90% confidence intervals in parentheses) for overall exposure area under the concentration-time curve from zero to infinity (AUC0-∞) were 113.6% (41.6 to 310.2%), 178.3% (85.2 to 373.0%), and 169.2% (73.2 to 391.2%) for subjects with mild, moderate, and severe renal impairment, respectively. Overall, 5/8 (63%) subjects with normal renal function and 20/24 (83%) subjects with renal impairment reported adverse events, with gastrointestinal events being the most common. No severe or serious adverse events or deaths were reported. These results suggest that moderate or severe renal impairment can result in a modest increase in faldaprevir exposure. The increase in exposure may be related to decrease in the activity of the liver uptake transporter OATP1B1 as a result of renal impairment. Given this relatively slight increase in exposure, a dose adjustment in HCV patients with renal impairment is not warranted. (This study has been registered at ClinicalTrials.gov under registration number NCT01957657.).
Collapse
Affiliation(s)
- Fenglei Huang
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | | | - Benjamin Lang
- Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach, Germany
| | - Atef Halabi
- CRS Clinical Research Services Kiel GmbH, Kiel, Germany
| | | | - Chan-Loi Yong
- Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, Connecticut, USA
| | | |
Collapse
|
12
|
Doak B, Over B, Giordanetto F, Kihlberg J. Oral Druggable Space beyond the Rule of 5: Insights from Drugs and Clinical Candidates. ACTA ACUST UNITED AC 2014; 21:1115-42. [DOI: 10.1016/j.chembiol.2014.08.013] [Citation(s) in RCA: 282] [Impact Index Per Article: 25.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
13
|
Lundquist P, Lööf J, Fagerholm U, Sjögren I, Johansson J, Briem S, Hoogstraate J, Afzelius L, Andersson TB. Prediction of in vivo rat biliary drug clearance from an in vitro hepatocyte efflux model. Drug Metab Dispos 2014; 42:459-68. [PMID: 24396143 DOI: 10.1124/dmd.113.054155] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Well-established techniques are available to predict in vivo hepatic uptake and metabolism from in vitro data, but predictive models for biliary clearance remain elusive. Several studies have verified the expression and activity of ATP-binding cassette (ABC) efflux transporters central to biliary clearance in freshly isolated rat hepatocytes, raising the possibility of predicting biliary clearance from in vitro efflux measurements. In the present study, short-term plated rat hepatocytes were evaluated as a model to predict biliary clearance from in vitro efflux measurements before major changes in transporter expression known to take place in long-term hepatocyte cultures. The short-term cultures were carefully characterized for their uptake and metabolic properties using a set of model compounds. In vitro efflux was studied using digoxin, fexofenadine, napsagatran, and rosuvastatin, representing compounds with over 100-fold differences in efflux rates in vitro and 60-fold difference in measured in vivo biliary clearance. The predicted biliary clearances from short-term plated rat hepatocytes were within 2-fold of measured in vivo values. As in vitro efflux includes both basolateral and canalicular effluxes, pronounced basolateral efflux may introduce errors in predictions for some compounds. In addition, in vitro rat hepatocyte uptake rates corrected for simultaneous efflux predicted rat in vivo hepatic clearance of the biliary cleared compounds with less than 2-fold error. Short-term plated hepatocytes could thus be used to quantify hepatocyte uptake, metabolism, and efflux of compounds and considerably improve the prediction of hepatic clearance, especially for compounds with a large biliary clearance component.
Collapse
Affiliation(s)
- Patrik Lundquist
- CNS and Pain Innovative Medicines DMPK, AstraZeneca R&D, Södertälje (P.L., J.L., U.F., I.S., J.J., S.B., J.H., L.A.); Cardiovascular and Metabolic Diseases Innovative Medicines DMPK, AstraZeneca R&D, Mölndal (P.L., T.B.A.); Department of Pharmacy, Uppsala University, Uppsala (P.L.); and Section of Pharmacogenetics, Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm (T.B.A.), Sweden
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Lundquist P, Englund G, Skogastierna C, Lööf J, Johansson J, Hoogstraate J, Afzelius L, Andersson TB. Functional ATP-binding cassette drug efflux transporters in isolated human and rat hepatocytes significantly affect assessment of drug disposition. Drug Metab Dispos 2014; 42:448-58. [PMID: 24396144 DOI: 10.1124/dmd.113.054528] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Freshly isolated hepatocytes are considered the gold standard for in vitro studies of hepatic drug disposition. To ensure a reliable supply of cells, cryopreserved human hepatocytes are often used. ABC-superfamily drug efflux transporters are key elements in hepatic drug disposition. These transporters are often considered lost after isolation of hepatocytes. In the present study, the expression and activity of ABC transporters BCRP, BSEP, P-gp, MRP2, MRP3, and MRP4 in human and rat cryopreserved hepatocytes were investigated. In commercially available human cryopreserved hepatocytes, all drug efflux transporters except human BCRP (hBCRP) exhibited similar expression levels as in fresh liver biopsies. Expression levels of hBCRP were 60% lower in cryopreserved human hepatocytes than in liver tissue, which could lead to, at most, a 2.5-fold reduction in hBCRP-mediated efflux. Fresh rat hepatocytes showed significantly lower levels of rat BCRP compared with liver expression levels; expression levels of other ABC transporters were unchanged. ABC transporters in human cryopreserved cells were localized to the plasma membrane. Functional studies could demonstrate P-gp and BCRP activity in both human cryopreserved and fresh rat hepatocytes. Inhibiting P-gp-mediated efflux by elacridar in in vitro experiments significantly decreased fexofenadine efflux from hepatocytes, resulting in an increase in apparent fexofenadine uptake. The results from the present study clearly indicate that ABC transporter-mediated efflux in freshly isolated as well as cryopreserved rat and human hepatocytes should be taken into account in in vitro experiments used for modeling of drug metabolism and disposition.
Collapse
Affiliation(s)
- Patrik Lundquist
- CNS and Pain Innovative Medicines DMPK, AstraZeneca R&D, Södertälje, (P.L., G.E., C.S., J.L., J.J., J.H., L.A.); Cardiovascular and Gastrointestinal Innovative Medicines DMPK, AstraZeneca R&D, Mölndal, (P.L., T.B.A.); Department of Pharmacy, Uppsala University, Uppsala, (P.L.); and Section of Pharmacogenetics, Departments of Physiology and Pharmacology, Karolinska Institutet, Stockholm, (T.B.A.), Sweden
| | | | | | | | | | | | | | | |
Collapse
|
15
|
Döring B, Petzinger E. Phase 0 and phase III transport in various organs: combined concept of phases in xenobiotic transport and metabolism. Drug Metab Rev 2014; 46:261-82. [PMID: 24483608 DOI: 10.3109/03602532.2014.882353] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The historical phasing concept of drug metabolism and elimination was introduced to comprise the two phases of metabolism: phase I metabolism for oxidations, reductions and hydrolyses, and phase II metabolism for synthesis. With this concept, biological membrane barriers obstructing the accessibility of metabolism sites in the cells for drugs were not considered. The concept of two phases was extended to a concept of four phases when drug transporters were detected that guided drugs and drug metabolites in and out of the cells. In particular, water soluble or charged drugs are virtually not able to overcome the phospholipid membrane barrier. Drug transporters belong to two main clusters of transporter families: the solute carrier (SLC) families and the ATP binding cassette (ABC) carriers. The ABC transporters comprise seven families with about 20 carriers involved in drug transport. All of them operate as pumps at the expense of ATP splitting. Embedded in the former phase concept, the term "phase III" was introduced by Ishikawa in 1992 for drug export by ABC efflux pumps. SLC comprise 52 families, from which many carriers are drug uptake transporters. Later on, this uptake process was referred to as the "phase 0 transport" of drugs. Transporters for xenobiotics in man and animal are most expressed in liver, but they are also present in extra-hepatic tissues such as in the kidney, the adrenal gland and lung. This review deals with the function of drug carriers in various organs and their impact on drug metabolism and elimination.
Collapse
Affiliation(s)
- Barbara Döring
- Institute of Pharmacology and Toxicology, Biomedical Research Center Seltersberg, Justus-Liebig-University Giessen , Giessen , Germany
| | | |
Collapse
|
16
|
Pfeifer ND, Hardwick RN, Brouwer KLR. Role of hepatic efflux transporters in regulating systemic and hepatocyte exposure to xenobiotics. Annu Rev Pharmacol Toxicol 2013; 54:509-35. [PMID: 24160696 DOI: 10.1146/annurev-pharmtox-011613-140021] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Hepatic efflux transporters include numerous well-known and emerging proteins localized to the canalicular or basolateral membrane of the hepatocyte that are responsible for the excretion of drugs into the bile or blood, respectively. Altered function of hepatic efflux transporters due to drug-drug interactions, genetic variation, and/or disease states may lead to changes in xenobiotic exposure in the hepatocyte and/or systemic circulation. This review focuses on transport proteins involved in the hepatocellular efflux of drugs and metabolites, discusses mechanisms of altered transporter function as well as the interplay between multiple transport pathways, and highlights the importance of considering intracellular unbound concentrations of transporter substrates and/or inhibitors. Methods to evaluate hepatic efflux transport and predict the effects of impaired transporter function on systemic and hepatocyte exposure are discussed, and the sandwich-cultured hepatocyte model to evaluate comprehensively the role of hepatic efflux in the hepatobiliary disposition of xenobiotics is characterized.
Collapse
Affiliation(s)
- Nathan D Pfeifer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599; ,
| | | | | |
Collapse
|
17
|
Perspectives on a pharmacokinetics legend: C versus T (contributions over time). J Pharm Sci 2013; 102:2889-94. [DOI: 10.1002/jps.23628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 11/07/2022]
|
18
|
Zou P, Liu X, Wong S, Feng MR, Liederer BM. Comparison of In Vitro-In Vivo Extrapolation of Biliary Clearance Using an Empirical Scaling Factor Versus Transport-Based Scaling Factors in Sandwich-Cultured Rat Hepatocytes. J Pharm Sci 2013; 102:2837-50. [DOI: 10.1002/jps.23620] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2013] [Revised: 05/03/2013] [Accepted: 05/06/2013] [Indexed: 02/05/2023]
|
19
|
Ramboer E, Vanhaecke T, Rogiers V, Vinken M. Primary hepatocyte cultures as prominent in vitro tools to study hepatic drug transporters. Drug Metab Rev 2013; 45:196-217. [PMID: 23368091 DOI: 10.3109/03602532.2012.756010] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Before any drug can be placed on the market, drug efficacy and safety must be ensured through rigorous testing. Animal models are used for this purpose, though currently increasing attention goes to the use of alternative in vitro systems. In particular, liver-based testing platforms that allow the prediction of pharmacokinetic (PK) and pharmacotoxicological properties during the early phase of drug development are of interest. They also enable the screening of potential effects on hepatic drug transporters. The latter are known to affect drug metabolism and disposition, thereby possibly underlying drug-drug interactions, which, in turn, may result in liver toxicity. Clearly, stable in vivo-like functional expression of drug transporters in hepatic in vitro settings is a prerequisite to be applicable in routine PK and pharmacotoxicological testing. In the first part of the article, an updated overview of hepatic drug transporters is provided, followed by a state-of-the-art review of drug-transporter production and activity in primary hepatocyte cultures (PHCs), being the gold-standard in vitro system. Specific focus is hereby put on strategies to maintain long-term functional expression, in casu of drug transporters, in these systems. In the second part, the use of PHCs to assess hepatobiliary transport and transporter-mediated interactions is outlined.
Collapse
Affiliation(s)
- Eva Ramboer
- Department of Toxicology, Center for Pharmaceutical Research, Vrije Universiteit Brussel, Brussels, Belgium.
| | | | | | | |
Collapse
|
20
|
Grime K, Paine SW. Species differences in biliary clearance and possible relevance of hepatic uptake and efflux transporters involvement. Drug Metab Dispos 2013; 41:372-8. [PMID: 23139379 DOI: 10.1124/dmd.112.049312] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
From a search of the available literature, a database of 22 drugs of all charge types and several different therapeutic classes was compiled to compare rat and human biliary clearance data. Dog biliary excretion data were also found for nine of the drugs. For 19 of the 22 drugs (86%), rat unbound biliary clearance values, when normalized for body weight, exceeded those for humans by factors ranging from 9 to over 2500-fold, whereas human/dog differences were much less dramatic. It was possible to define hepatic uptake and efflux transporter involvement for many of the drugs. On the basis of the findings, it is postulated that regardless of the biliary efflux transporters implicated, when drugs do not require active hepatic uptake to access the liver there may be fairly insignificant differences in rat, dog, and human biliary clearance. Conversely, when the organic anion-transporting polypeptide drug transporters are involved, one may expect at least a 10-fold discrepancy in rat to human biliary clearance normalized for body weight and corrected for plasma protein binding.
Collapse
Affiliation(s)
- Ken Grime
- Respiratory and Inflammation Drug Metabolism and Pharmacokinetics, Astra Zeneca R&D, Mölndal, Sweden.
| | | |
Collapse
|
21
|
Lancaster CS, Bruun GH, Peer CJ, Mikkelsen TS, Corydon TJ, Gibson AA, Hu S, Orwick SJ, Mathijssen RHJ, Figg WD, Baker SD, Sparreboom A. OATP1B1 polymorphism as a determinant of erythromycin disposition. Clin Pharmacol Ther 2012; 92:642-50. [PMID: 22990751 DOI: 10.1038/clpt.2012.106] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Previous studies have demonstrated that the pharmacokinetic profile of erythromycin, a probe for CYP3A4 activity, is affected by inhibitors or inducers of hepatic solute carriers. We hypothesized that these interactions are mediated by OATP1B1 (gene symbol, SLCO1B1), a polypeptide expressed on the basolateral surface of hepatocytes. Using stably transfected Flp-In T-Rex293 cells, erythromycin was found to be a substrate for OATP1B1*1A (wild type) with a Michaelis-Menten constant of ~13 µmol/l, and that its transport was reduced by ~50% in cells expressing OATP1B1*5 (V174A). Deficiency of the ortholog transporter Oatp1b2 in mice was associated with a 52% decrease in the metabolic rate of erythromycin (P = 0.000043). In line with these observations, in humans the c.521T>C variant in SLCO1B1 (rs4149056), encoding OATP1B1*5, was associated with a decline in erythromycin metabolism (P = 0.0072). These results suggest that impairment of OATP1B1 function can alter erythromycin metabolism, independent of changes in CYP3A4 activity.
Collapse
Affiliation(s)
- C S Lancaster
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Nagar S, Korzekwa K. Commentary: nonspecific protein binding versus membrane partitioning: it is not just semantics. Drug Metab Dispos 2012; 40:1649-52. [PMID: 22711748 DOI: 10.1124/dmd.112.046599] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Nonspecific binding or sequestration results in differences between free and total drug concentrations, both in vitro and in vivo. Membrane partitioning and not protein binding is the primary mechanism of drug sequestration. Therefore, physicochemical properties, e.g., LogP can be used to predict drug sequestration in membrane and cell-based assays. The concentration of drug in a membrane is determined by the both the rate in and out of the membrane. In contrast, membrane permeability is a function of the rate in only. This commentary discusses the origins of membrane partitioning and permeability and their impact on cellular disposition.
Collapse
Affiliation(s)
- Swati Nagar
- Temple University School of Pharmacy, 3307 N. Broad Street, Philadelphia, PA, USA.
| | | |
Collapse
|
23
|
Yoon I, Han S, Choi YH, Kang HE, Cho HJ, Kim JS, Shim CK, Chung SJ, Chong S, Kim DD. Saturable sinusoidal uptake is rate-determining process in hepatic elimination of docetaxel in rats. Xenobiotica 2012; 42:1110-9. [DOI: 10.3109/00498254.2012.700139] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
24
|
Korzekwa KR, Nagar S, Tucker J, Weiskircher EA, Bhoopathy S, Hidalgo IJ. Models to predict unbound intracellular drug concentrations in the presence of transporters. Drug Metab Dispos 2012; 40:865-76. [PMID: 22279052 DOI: 10.1124/dmd.111.044289] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Knowledge of free drug intracellular concentration is necessary to predict the impacts of drugs on intracellular targets. The goal of this study was to develop models to predict free intracellular drug concentrations in the presence of apical efflux transporters. The apical efflux transporter P-glycoprotein (P-gp), encoded by human gene multidrug resistance 1 (MDR1), was studied. Apparent permeabilities for 10 compounds in Madin-Darby canine kidney (MDCK) and MDR1-MDCK cell monolayers were obtained experimentally. Six of these compounds were evaluated additionally in the presence of the P-gp inhibitor cyclosporine A. A three-compartment model was developed, and passive and apical efflux clearances (CL(d) and CL(ae), respectively) were estimated. Endogenous canine transporters also were delineated. The three-compartment model was unable to simulate experimentally observed lag times and exhibited systematic bias across the simulations. Next, a five-compartment model with explicit membrane compartments was developed. This model resulted in lower systematic errors and simulated the lag time observed experimentally. Apical efflux was modeled out of the cell or out of the membrane. The five-compartment model with apical efflux out of the membrane predicted marked differences in unbound intracellular concentrations between the apical-to-basolateral and the basolateral-to-apical directions. Upon apical drug addition, large decreases in intracellular concentrations were observed with the efflux transporter. No such difference was predicted upon basolateral drug addition. This is consistent with experimental differences in the impact of P-gp on hepatic and brain distribution and supports the hypothesis that apical efflux occurs out of the apical membrane.
Collapse
Affiliation(s)
- Ken R Korzekwa
- Department of Pharmaceutical Sciences, Temple University School of Pharmacy, 3307 N. Broad St., Philadelphia, PA 19140, USA.
| | | | | | | | | | | |
Collapse
|
25
|
Sjögren E, Bredberg U, Lennernäs H. The Pharmacokinetics and Hepatic Disposition of Repaglinide in Pigs: Mechanistic Modeling of Metabolism and Transport. Mol Pharm 2012; 9:823-41. [DOI: 10.1021/mp200218p] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Erik Sjögren
- Department of Pharmacy, Uppsala University, Box 580, S-751 23 Uppsala, Sweden
| | | | - Hans Lennernäs
- Department of Pharmacy, Uppsala University, Box 580, S-751 23 Uppsala, Sweden
| |
Collapse
|
26
|
Jin Y, Luan X, Liu H, Gao C, Li S, Cao D, Li X, Cai Z, Jiang Y. Pharmacokinetics and metabolite identification of a novel VEGFR-2 and Src dual inhibitor 6-chloro-2-methoxy-N-(2-methoxybenzyl) acridin-9-amine in rats by liquid chromatography tandem mass spectrometry. Talanta 2011; 89:70-6. [PMID: 22284461 DOI: 10.1016/j.talanta.2011.11.061] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2011] [Revised: 11/16/2011] [Accepted: 11/20/2011] [Indexed: 11/24/2022]
Abstract
A novel VEGFR-2 and Src dual inhibitor, 6-Chloro-2-methoxy-N-(2-methoxybenzyl) acridin-9-amine (MBAA), is a 9-aminoacridine derivative, but its pharmacokinetics and metabolism in body remain unknown. Using liquid chromatography tandem electrospray ionization mass spectrometry with the multiple reaction monitoring modes, we developed and validated a simple, rapid, sensitive and accurate technology for analyses of MBAA in the rat plasma, urine and bile. The micro samples were quickly prepared by 96-well plate. Chromatographic separation was performed on a C(18) column with gradient elution. High-quality linearity calibration curves were achieved over a concentration range of 1.00-3000 ng mL(-1). Intra- and inter-day precisions (RSD) were less than 8.5%, and accuracy (RE%) ranged from -2.9% to 12%. Extraction recoveries of MBAA were consistent with an average of 82.2-111.4% at three QC concentrations. When administered intravenously at a single dose of 2.0 mg kg(-1) to male SD rats, MBAA was rapidly eliminated with a T(1/2) of 0.9 ± 0.1h and AUC(0-t) of 369 ± 44.7 ng mL(-1). We identified four direct phase I and phase II metabolites by mass difference of molecular ions between metabolites and the parent compound. Various fragmentation patterns of MBAA were used to identify and characterize its metabolites. This LC-MS/MS analysis provides a useful approach to the pharmacokinetic and metabolic study of MBAA.
Collapse
Affiliation(s)
- Yibao Jin
- Department of Chemistry, Tsinghua University, Beijing 100084, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
27
|
Yin J, Meng Q. Use of primary rat hepatocytes in the gel entrapment culture to predictin vivobiliary excretion. Xenobiotica 2011; 42:417-28. [DOI: 10.3109/00498254.2011.633716] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|
28
|
Fenner KS, Jones HM, Ullah M, Kempshall S, Dickins M, Lai Y, Morgan P, Barton HA. The evolution of the OATP hepatic uptake transport protein family in DMPK sciences: from obscure liver transporters to key determinants of hepatobiliary clearance. Xenobiotica 2011; 42:28-45. [PMID: 22077101 DOI: 10.3109/00498254.2011.626464] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Over the last two decades the impact on drug pharmacokinetics of the organic anion transporting polypeptides (OATPs: OATP-1B1, 1B3 and 2B1), expressed on the sinusoidal membrane of the hepatocyte, has been increasingly recognized. OATP-mediated uptake into the hepatocyte coupled with subsequent excretion into bile via efflux proteins, such as MRP2, is often referred to as hepatobiliary excretion. OATP transporter proteins can impact some drugs in several ways including pharmacokinetic variability, pharmacodynamic response and drug-drug interactions (DDIs). The impact of transporter mediated hepatic clearance is illustrated with case examples, from the literature and also from the Pfizer portfolio. The currently available in vitro techniques to study the hepatic transporter proteins involved in the hepatobiliary clearance of drugs are reviewed herein along with recent advances in using these in vitro data to predict the human clearance of compounds recognized by hepatic uptake transporters.
Collapse
Affiliation(s)
- Katherine S Fenner
- Department of Pharmacokinetics, Dynamics and Metabolism, Pfizer Worldwide Research and Development, Sandwich, Kent, UK.
| | | | | | | | | | | | | | | |
Collapse
|
29
|
Yabe Y, Galetin A, Houston JB. Kinetic characterization of rat hepatic uptake of 16 actively transported drugs. Drug Metab Dispos 2011; 39:1808-14. [PMID: 21730030 DOI: 10.1124/dmd.111.040477] [Citation(s) in RCA: 107] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
To explore the determinants of hepatic uptake, 16 compounds were investigated with different physicochemical and disposition characteristics, including five statins, three sartans, saquinavir, ritonavir, erythromycin, clarithromycin, nateglinide, repaglinide, fexofenadine, and bosentan. Freshly isolated rat hepatocytes in suspension were used with the oil-spin method to generate kinetic parameters. Clearances, via passive diffusion (P(diff)) and active uptake (CL(active), characterized by maximal uptake rate and K(m)), were estimated from the initial uptake rate data over a 0.01 to 100 μM concentration range. The K(m) values had a range of 15-fold, with 10 of the 16 drugs with K(m) < 10 μM (median 6 μM). Both CL(active) and P(diff) ranged over 100-fold (median 188 and 14 μl/min/10⁶ cells). Assessment of the relative contribution of P(diff) and CL(active) indicated that, at low concentrations (approximately 0.1 μM), the active process contributes >80% to the overall uptake for 13 drugs. Although high P(diff) values were obtained for ritonavir and repaglinide, active process contributed predominantly to uptake; in contrast, high passive permeability dominates over transporter-mediated uptake for saquinavir over the full concentration range. For bosentan and erythromycin, active and passive processes were equally important. Hepatocyte-to-medium unbound concentration ratio was >10 for 9 of the 16 drugs, ranging from 2 to 494 for bosentan and atorvastatin, respectively. Some drugs showed extensive intracellular binding (fraction unbound range 0.01-0.6), which was not correlated with active uptake. LogD₇.₄ correlated significantly with P(diff) and the extent of intracellular binding but not with active uptake. This study provides systematic assessment of the role of active uptake relative to the passive process; implications of the findings are discussed.
Collapse
Affiliation(s)
- Yoshiyuki Yabe
- Centre for Applied Pharmacokinetic Research, School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Manchester, UK
| | | | | |
Collapse
|
30
|
Sugiyama E, Kikuchi A, Inada M, Sato H. The use of 13C-erythromycin as an in vivo probe to evaluate CYP3A-mediated drug interactions in rats. J Pharm Sci 2011; 100:3995-4005. [PMID: 21618542 DOI: 10.1002/jps.22616] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2011] [Revised: 04/16/2011] [Accepted: 04/21/2011] [Indexed: 11/05/2022]
Abstract
(14)C-erythromycin breath test has been utilized to evaluate the extent of CYP3A activity in vivo. However, its radioactivity sometimes impedes its clinical application. In this study, we employed erythromycin labeled with (13)C ((13)C-EM), a nonradioactive stable isotope, as an in vivo probe of breath test to evaluate CYP3A-mediated drug interactions in rats. A physiologically based pharmacokinetic (PBPK) model to describe (13)CO(2) exhalation altered by drug interactions was newly constructed. Rats received an intravenous or oral administration of (13)C-EM with or without a CYP3A inhibitor or inducer, that is, ketoconazole (KCZ) or dexamethasone (DEX), respectively. Breath samples were taken at designated times, measured with an infrared spectrophotometer, and the Δ(13) CO(2) value (‰) in each sample was obtained. The C(max) and AUC(0-t) of Δ(13) CO(2) were significantly decreased with KCZ and increased with DEX. The PBPK model in this study successfully described the (13)CO(2) exhalation after (13)C-EM administration in the absence and presence of drug interactions. In conclusion, this study proposed a simple and rapid in vivo methodology to utilize (13)C-EM for the quantitative analysis of CYP3A inhibition and induction. This method using small animals may be useful in early drug development processes.
Collapse
Affiliation(s)
- Erika Sugiyama
- Department of Clinical and Molecular Pharmacokinetics/Pharmacodynamics, Faculty of Pharmaceutical Sciences, Showa University, Tokyo, Japan.
| | | | | | | |
Collapse
|
31
|
Reyes M, Benet LZ. Effects of uremic toxins on transport and metabolism of different biopharmaceutics drug disposition classification system xenobiotics. J Pharm Sci 2011; 100:3831-42. [PMID: 21618544 DOI: 10.1002/jps.22640] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2011] [Revised: 04/15/2011] [Accepted: 05/06/2011] [Indexed: 12/15/2022]
Abstract
Chronic kidney disease (CKD) is recognized to cause pharmacokinetic changes in renally excreted drugs; however, pharmacokinetic changes are also reported for drugs that are nonrenally eliminated. Few studies have investigated how uremic toxins may affect drug transporters and metabolizing enzymes and how these may result in pharmacokinetic/metabolic changes in CKD. Here, we investigated the effects of uremic toxins and human uremic serum on the transport of the prototypical transporter substrate [(3) H]-estrone sulfate and three Biopharmaceutics Drug Disposition Classification System (BDDCS) drugs, propranolol, losartan, and eprosartan. We observed a significant decrease in [(3) H]-estrone sulfate, losartan, and eprosartan uptake with some uremic toxins in both transfected cells and rat hepatocytes. The uptake of losartan was decreased in rat and human hepatocytes (28% and 48%, respectively) in the presence of hemodialysis (HD) serum. Time-course studies of losartan showed a 27%, 65%, and 68% increase in area under the curve (AUC) in the presence of HD serum, rifampin, and sulfaphenazole, respectively. Intracellular losartan AUC decreased significantly in the treatment groups, and the metabolite AUC decreased by 41% and 26% in rifampin- and sulfaphenazole-treated group, respectively. The intracellular AUC of eprosartan increased 190% in the presence of HD serum. These studies indicate that the uremic toxins contained in HD serum play an important role in drug disposition through drug transporters, and that there would be differential effects depending on the BDDCS classification of the drug.
Collapse
Affiliation(s)
- Maribel Reyes
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, California 94143-0912, USA
| | | |
Collapse
|
32
|
Franke RM, Lancaster CS, Peer CJ, Gibson AA, Kosloske AM, Orwick SJ, Mathijssen RH, Figg WD, Baker SD, Sparreboom A. Effect of ABCC2 (MRP2) transport function on erythromycin metabolism. Clin Pharmacol Ther 2011; 89:693-701. [PMID: 21451505 DOI: 10.1038/clpt.2011.25] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The macrolide antiobiotic erythromycin undergoes extensive hepatic metabolism and is commonly used as a probe for cytochrome P450 (CYP) 3A4 activity. By means of a transporter screen, erythromycin was identified as a substrate for the transporter ABCC2 (MRP2) and its murine ortholog, Abcc2. Because these proteins are highly expressed on the biliary surface of hepatocytes, we hypothesized that impaired Abcc2 function may influence the rate of hepatobiliary excretion and thereby enhance erythromycin metabolism. Using Abcc2 knockout mice, we found that Abcc2 deficiency was associated with a significant increase in erythromycin metabolism, whereas murine Cyp3a protein expression and microsomal Cyp3a activity were not affected. Next, in a cohort of 108 human subjects, we observed that homozygosity for a common reduced-function variant in ABCC2 (rs717620) was also linked to an increase in erythromycin metabolism but was not correlated with the clearance of midazolam. These results suggest that impaired ABCC2 function can alter erythromycin metabolism, independent of changes in CYP3A4 activity.
Collapse
Affiliation(s)
- R M Franke
- Department of Pharmaceutical Sciences, St Jude Children's Research Hospital, Memphis, Tennessee, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Kuypers DRJ, de Jonge H, Naesens M, Vanrenterghem Y. A prospective, open-label, observational clinical cohort study of the association between delayed renal allograft function, tacrolimus exposure, and CYP3A5 genotype in adult recipients. Clin Ther 2011; 32:2012-23. [PMID: 21118736 DOI: 10.1016/j.clinthera.2010.11.010] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/16/2010] [Indexed: 02/07/2023]
Abstract
BACKGROUND Tacrolimus, a calcineurin inhibitor with a macrolide lactone structure, is currently used as a cornerstone immunosuppressive drug in solid organ transplantation. It is metabolized by hepatic and intestinal cytochrome P450 (CYP) 3A4/3A5 enzymes and is a substrate for P-glycoprotein (ABCB1). The disposition of tacrolimus might be influenced by severe renal allograft dysfunction (eg, in cases of delayed graft function [DGF]). New-onset diabetes after transplantation (NODAT) is a known adverse effect of tacrolimus therapy and has been associated with DGF. OBJECTIVES The impact of DGF on tacrolimus C(min) and dose requirements was evaluated in renal transplant recipients in the first postoperative week. The effects of the CYP3A5*3 A6986G polymorphism on initial mean tacrolimus C(min) and dose requirements in the presence and absence of DGF were assessed. This study also tested the hypothesis that if DGF influences early tacrolimus exposure, this would lead to a higher risk for NODAT (defined as the need for glucose-lowering medication for an uninterrupted period of ≥ 26 weeks). METHODS This prospective, open-label, observational clinical cohort study enrolled renal allograft recipients aged ≥ 18 years. Tacrolimus was administered as an oral loading dose of 0.2 mg/kg/d and adjusted to achieve a target mean daily tacrolimus C(min) between 12 and 15 ng/mL. C(min) values and oral dose requirements in the first postoperative week were compared between patients with and without DGF. Patients were genotyped for the CYP3A4*1B -290A>G, CYP3A5*3 A6986G, ABCB1 Exon26 C3435T, ABCB1 Exon21 G2677T, and ABCB1 Exon21 G2677A single nucleotide polymorphisms. NODAT that occurred within the first 12 weeks after transplantation was confirmed using an oral glucose tolerance test. RESULTS A total of 304 patients were enrolled (184 men, 120 women; mean [SD] age, 52.9 [14.1] years). Through day 3 after transplantation, mean (SD) 12-hour tacrolimus C(min) values were significantly higher in recipients experiencing DGF despite identical loading doses of 0.2 mg/kg. Mean tacrolimus dose requirements were significantly lower in patients with DGF during the first week. After recovery of DGF, mean tacrolimus dose requirements were not significantly different between recipients with and without DGF. In homozygous CYP3A5*3 carriers (n = 252), mean (SD) tacrolimus dose requirements remained significantly lower during DGF, while in CYP3A5*1 carriers with DGF (n = 52), lower mean dose requirements were observed only after postoperative day 4. The proportion of patients in whom NODAT developed was significantly greater in patients with DGF and tacrolimus C(min) >15 ng/mL on the first day after transplantation (27.2%) compared with recipients who remained free of DGF and had C(min) ≤15 ng/mL on day 1 (6.5%) (P = 0.016). On logistic regression analysis, greater recipient age (odds ratio [OR] = 1.044; 95% CI, 1.009-1.080), higher tacrolimus C(min) on day 1 (OR = 1.048; 95% CI, 1.017-1.080), and DGF (OR = 2.968; 95% CI, 1.107-7.959) were associated with an increased risk for NODAT. CONCLUSION In this open-label, observational study, DGF was associated with higher initial mean tacrolimus C(min) values and lower daily dose requirements predominantly in CYP3A5 nonexpressers.
Collapse
Affiliation(s)
- Dirk R J Kuypers
- Department of Nephrology and Renal Transplantation, University Hospitals Leuven, Leuven, Belgium.
| | | | | | | |
Collapse
|
34
|
Brown HS, Wilby AJ, Alder J, Houston JB. Comparative use of isolated hepatocytes and hepatic microsomes for cytochrome P450 inhibition studies: transporter-enzyme interplay. Drug Metab Dispos 2010; 38:2139-46. [PMID: 20847136 DOI: 10.1124/dmd.110.035824] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
Accurate assignment of the concentration of victim drug/inhibitor available at the enzyme active site, both in vivo and within an in vitro incubation, is an essential requirement in rationalizing and predicting drug-drug interactions. Inhibitor accumulation within the liver, whether as a result of active transport processes or intracellular binding, may best be accounted for using hepatocytes rather than hepatic microsomes to estimate in vitro inhibitory potency. The aims of this study were to compare K(i) values determined in rat liver microsomes and freshly isolated rat hepatocytes of four cytochrome P450 (P450) inhibitors (clarithromycin, enoxacin, nelfinavir, and saquinavir) with known hepatic transporter involvement and a range of uptake (cell/medium concentration ratios 20-3000) and clearance (10-1200 μl/min/10(6) cells) properties. Inhibition studies were performed using two well established P450 probe substrates (theophylline and midazolam). Comparison of unbound K(i) values showed marked differences between the two in vitro systems for inhibition of metabolism. In two cases (clarithromycin and enoxacin, both low-clearance drugs), inhibitory potency in hepatocytes markedly exceeded that in microsomes (10- to 20-fold), and this result was consistent with their high cell/medium concentration ratios. For nelfinavir and saquinavir (high-clearance, extensively metabolized drugs), the opposite trend was seen in the K(i) values: despite very high cell/medium concentration ratios, stronger inhibition was evident within microsomal preparations. Hence, the consequences of hepatic accumulation resulting from uptake transporters vary according to the clearance of the inhibitor. This study demonstrates that transporter-enzyme interplay can result in differences in inhibitory potency between microsomes and hepatocytes and hence drug-drug interaction predictions that are not always intuitive.
Collapse
Affiliation(s)
- Hayley S Brown
- Centre for Applied Pharmacokinetic Research, School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Manchester, United Kingdom
| | | | | | | |
Collapse
|
35
|
Effect of single-dose rifampin on the pharmacokinetics of warfarin in healthy volunteers. Clin Pharmacol Ther 2010; 88:540-7. [PMID: 20703222 DOI: 10.1038/clpt.2010.142] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Based on in vitro rat and human hepatocyte uptake studies showing inhibition of warfarin uptake in the presence of the nonspecific organic anion-transporting polypeptide (OATP) inhibitor rifampin, a clinical study was conducted in 10 healthy volunteers to examine the in vivo relevance of OATP hepatic uptake on the pharmacokinetics of warfarin. In a randomized, single-dose, two-period, crossover design, subjects received a 7.5-mg dose of warfarin, either alone or immediately following a 600-mg intravenous dose of rifampin. Rifampin did not significantly alter the R- or S-warfarin area under the concentration-time curves (AUCs) from 0 to 12 h (period of hepatic OATP inhibition by rifampin) or the maximum plasma concentration (C(max)) value. AUC(0-∞) was decreased on days rifampin was administered, for both R-warfarin (25% reduction; P < 0.001) and S-warfarin (15% reduction; P < 0.05). No differences were seen in the area under the international normalized ratio (INR)-time curve. Our study suggests that hepatic uptake via OATPs may not be clinically important in the pharmacokinetics of warfarin.
Collapse
|
36
|
Yang X, Morris ME. Pharmacokinetics and Biliary Excretion of Mitoxantrone in Rats. J Pharm Sci 2010; 99:2502-10. [DOI: 10.1002/jps.22011] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
37
|
Tang C, Prueksaritanont T. Use of in vivo animal models to assess pharmacokinetic drug-drug interactions. Pharm Res 2010; 27:1772-87. [PMID: 20428930 DOI: 10.1007/s11095-010-0157-z] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2010] [Accepted: 04/08/2010] [Indexed: 12/31/2022]
Abstract
Animal models are used commonly in various stages of drug discovery and development to aid in the prospective assessment of drug-drug interaction (DDI) potential and the understanding of the underlying mechanism for DDI of a drug candidate. In vivo assessments in an appropriate animal model can be very valuable, when used in combination with in vitro systems, to help verify in vivo relevance of the in vitro animal-based results, and thus substantiate the extrapolation of in vitro human data to clinical outcomes. From a pharmacokinetic standpoint, a key consideration for rational selection of an animal model is based on broad similarities to humans in important physiological and biochemical parameters governing drug absorption, distribution, metabolism or excretion (ADME) processes in question for both the perpetrator and victim drugs. Equally critical are specific in vitro and/or in vivo experiments to demonstrate those similarities, usually both qualitative and quantitative, in the ADME properties/processes under investigation. In this review, theoretical basis and specific examples are presented to illustrate the utility of the animal models in assessing the potential and understanding the mechanisms of DDIs.
Collapse
Affiliation(s)
- Cuyue Tang
- Department of Drug Metabolism and Pharmacokinetics, Merck Research Laboratories, Merck & Co., Inc., WP75A-203, West Point, Pennsylvania 19486, USA
| | | |
Collapse
|
38
|
Benet LZ. The drug transporter-metabolism alliance: uncovering and defining the interplay. Mol Pharm 2010; 6:1631-43. [PMID: 19874004 DOI: 10.1021/mp900253n] [Citation(s) in RCA: 136] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Two decades ago the importance of transporter-enzyme interplay and its effects on drug bioavailability and hepatic disposition were first recognized. Here we review the history of uncovering and defining this interplay with a primary emphasis on studies from our laboratory. We review the early 1990s oral bioavailability studies that found that the highly lipophilic, poorly water-soluble cyclosporine formulation on the market at that time did not have an absorption problem, but rather a gut metabolism problem. This led to studies of the interactive nature of CYP3A and P-glycoprotein in the intestine, and investigations of this interplay using cellular systems and isolated perfused rat organ studies. Studies investigating uptake transporter-enzyme interactions using cellular, perfused rat liver and intact rats are reviewed, followed by the human transporter-enzyme interaction studies. Work characterizing the rate limiting processes in the drug transporter-metabolism alliance is then addressed, ending with a review of areas of the interplay that require further studies and analysis.
Collapse
Affiliation(s)
- Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94143-0912, USA.
| |
Collapse
|
39
|
Endres CJ, Endres MG, Unadkat JD. Interplay of drug metabolism and transport: a real phenomenon or an artifact of the site of measurement? Mol Pharm 2010; 6:1756-65. [PMID: 19886641 DOI: 10.1021/mp9002392] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The interdependence of both transport and metabolism on the disposition of drugs has recently gained heightened attention in the literature, and has been termed the "interplay of transport and metabolism". Such "interplay" is observed when inhibition of biliary clearance of a drug results in an "apparent" increase in the metabolic clearance of the drug or vice versa. In this manuscript, we derived and explored through simulations a physiological-based pharmacokinetic model that integrates both transport and metabolism and explains the "apparent" dependence of hepatic clearance on both these processes. In addition, we show that the phenomenon of hepatic "transport-metabolism interplay" is a result of using the plasma concentration as a point of reference when calculating metabolic or biliary clearance, and this interplay is maximal when the drug is actively transported into the hepatocytes (i.e., hepatocyte sinusoidal influx clearance is greater than the sinusoidal efflux clearance). When the hepatic drug concentration is used as a reference point to calculate metabolic or biliary clearance, this interplay ceases to exist. A mechanistic understanding of this interplay phenomenon can be used to explain the somewhat paradoxical results that may be observed in drug-drug interaction studies when a drug is cleared by both metabolism and biliary excretion. That is, when one of these two pathways is inhibited, the other pathway appears to be induced or activated. This interplay results in an increase in hepatic drug concentrations and therefore has implications for the hepatic efficacy and toxicity of a drug.
Collapse
Affiliation(s)
- Christopher J Endres
- Department of Pharmaceutics, University of Washington, Seattle, Washington 98195, USA
| | | | | |
Collapse
|
40
|
Pang KS, Maeng HJ, Fan J. Interplay of transporters and enzymes in drug and metabolite processing. Mol Pharm 2010; 6:1734-55. [PMID: 19891494 DOI: 10.1021/mp900258z] [Citation(s) in RCA: 82] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This review highlights the "interplay" between enzymes and transporters, essential components of eliminating organs for drug removal. The understanding of the interplay is important in terms of deciphering the change of one eliminatory pathway on compensatory mechanisms in drug disposal, and, ultimately, their importance in drug-drug interactions. Controversy existed on the explanation underlying the interplay between transporters and enzymes in the Caco-2 cell monolayer or cell culture systems, but less so on eliminating organs such as the intestine and liver. For the Caco-2 system, the increase in the mean residence time (MRT) accompanying increased secretion had been construed as the basis for increased metabolism. We hold the opposite view and assert that increased secretion should evoke a decrease in metabolism due to the competition between the enzyme and apical efflux transporter for the drug within the cell. To illustrate this point, simulations on the MRT, fraction of dose metabolized (f(met)) and the extraction ratio (ER) as defined by various investigators under linear and nonlinear metabolic conditions were compared to observed data and the trends upon induction/inhibition of secretion. The conclusion is that the f(met) is the more appropriate index to reflect the extent of metabolism in transporter-enzyme interplay, since the parameter captures drug metabolism in the cell when its contents in the apical, cell, and basolateral compartments or the entire dose is considered to be available for metabolism. This parameter for metabolism (f(met)) bears a reciprocal relationship to the secretory intrinsic clearance and is in concordance with the notion that both the enzyme and apical transporter compete for the cellular substrate within. For the liver and intestine, several physiologically based pharmacokinetic (PBPK) models that contain transporters and enzymes were utilized, together with the solved equations for the area under the curve (AUC), metabolic, excretory, and total clearance (CL) to shed meaningful insight of how the inhibition of one pathway can result in a higher AUC and therefore a reduced total clearance for drug, but a higher apparent clearance of the alternate pathway; induction of the same pathway would lead to an increased total clearance but decreased drug AUC, and reduced clearance of the alternate pathway. The use of an increased MRT to explain increased extents of metabolism upon increased apical excretion is not tenable in these organs or "open systems" since the MRT of drug in the cell is reduced with irreversible loss from biliary excretion or hastened gastrointestinal transit of the secreted drug in the lumen. Data in the literature for the Caco-2 system, knockout animals and organ perfusion systems were discussed in relation to these concepts on clearance based on fundamental, pharmacokinetic theory. The shortcomings in data interpretation were discussed. The general conclusion is that a reciprocal relationship exists between the clearances related to enzymes and apical transporters due to their competition for the substrate within the cell, and is a relationship independent of the MRT of drug in the system.
Collapse
Affiliation(s)
- K Sandy Pang
- Leslie L. Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| | | | | |
Collapse
|
41
|
Hepatic Disposition of Ximelagatran and Its Metabolites in Pig; Prediction of the Impact of Membrane Transporters Through a Simple Disposition Model. Pharm Res 2010; 27:597-607. [DOI: 10.1007/s11095-009-0016-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2009] [Accepted: 11/20/2009] [Indexed: 10/19/2022]
|
42
|
Sun H, Frassetto LA, Huang Y, Benet LZ. Hepatic clearance, but not gut availability, of erythromycin is altered in patients with end-stage renal disease. Clin Pharmacol Ther 2010; 87:465-72. [PMID: 20090676 DOI: 10.1038/clpt.2009.247] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Nonrenal clearance of drugs can be significantly lower in patients with end-stage renal disease (ESRD) than in those with normal renal function. Using erythromycin (ER) as a probe compound, we investigated whether this decrease in nonrenal clearance is due to reduced hepatic clearance (CL(H)) and/or gut metabolism. We also examined the potential effects of the uremic toxins 3-carboxy-4-methyl-5-propyl-2-furan propanoic acid (CMPF) and indoxyl sulfate (Indox) on ER disposition. Route-randomized, two-way crossover pharmacokinetic studies of ER were conducted in 12 ESRD patients and 12 healthy controls after oral (250 mg) and intravenous (125 mg) dosing with ER. In patients with ESRD, CL(H) decreased 31% relative to baseline values (0.35 +/- 0.14 l/h/kg vs. 0.51 +/- 0.13 l/h/kg, P = 0.01), with no change in steady-state volume of distribution. With oral dosing, the bioavailability of ER increased 36% in patients with ESRD, and this increase was not related to changes in gut availability. As expected, plasma levels of CMPF and Indox were significantly higher in the patients than in the healthy controls. However, no correlation was observed between CL(H) of ER and the levels of uremic toxins.
Collapse
Affiliation(s)
- H Sun
- Department of Biopharmaceutical Sciences, School of Pharmacy, University of California, San Francisco, San Francisco, California, USA
| | | | | | | |
Collapse
|
43
|
Tanino T, Nawa A, Nakao M, Noda M, Fujiwara S, Iwaki M. Organic anion transporting polypeptide 2-mediated uptake of paclitaxel and 2′-ethylcarbonate-linked paclitaxel in freshly isolated rat hepatocytes. J Pharm Pharmacol 2010. [DOI: 10.1211/jpp.61.08.0006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Abstract
Objectives
The P-glycoprotein (P-gp) efflux pump plays an important role in paclitaxel detoxification. However, hepatic uptake of paclitaxel mediated by a solute-linked carrier transporter family is still poorly understood in animals and humans. Freshly isolated hepatocyte suspensions are a well established in-vitro model for studying drug transport and xenobiotic metabolism. Therefore, the hepatic uptake of paclitaxel and its P-gp-insensitive prodrug, 2′-ethylcarbonate-linked paclitaxel (TAX-2′-Et), has been characterized using freshly isolated and pregnenolone-16-α-carbonitrile (PCN)-treated hepatocytes in rats.
Methods
Paclitaxel and TAX-2′-Et were incubated with rat hepatocyte suspensions in the presence or absence of inhibitors.
Key findings
Paclitaxel and TAX-2′-Et showed concentration-dependent uptake in rat hepatocytes. The intrinsic transport capacity was two-fold higher for paclitaxel uptake than for TAX-2′-Et uptake. Rifampicin (a potent inhibitor of organic anion transporting polypeptide (Oatp) 2), but not indometacin (a representative inhibitor of organic anion transporter (Oat) 2 and Oatp1) treatment, significantly inhibited the uptake of paclitaxel and TAX-2′-Et. We characterized the rifampicin-sensitive uptake of paclitaxel and TAX-2′-Et using rat hepatocytes treated with PCN, which dramatically enhances hepatic Oatp2 protein levels. PCN-treated hepatocytes displayed a 1.6-fold greater uptake of paclitaxel and TAX-2′-Et than the vehicle-treated hepatocytes. The uptake of the two compounds was significantly reduced by rifampicin but not by indometacin treatment. These findings demonstrated that the rat Oatp2, but not Oatp1 orOat2, was a candidate transporter for the hepatic uptakeofpaclitaxel and TAX-2′-Et.
Conclusions
The findings have provided an important step towards identifying a key transporter in hepatic detoxification of paclitaxel and TAX-2′-Et in small animals.
Collapse
Affiliation(s)
- Tadatoshi Tanino
- School of Pharmacy, Kinki University, Higashi-Osaka, Osaka, Japan
| | - Akihiro Nawa
- Nagoya University Graduate School of Medicine, Shouwa-ku, Nagoya, Japan
| | - Mao Nakao
- School of Pharmacy, Kinki University, Higashi-Osaka, Osaka, Japan
| | - Manabu Noda
- School of Pharmacy, Kinki University, Higashi-Osaka, Osaka, Japan
| | - Sawako Fujiwara
- Nagoya University Graduate School of Medicine, Shouwa-ku, Nagoya, Japan
| | - Masahiro Iwaki
- School of Pharmacy, Kinki University, Higashi-Osaka, Osaka, Japan
| |
Collapse
|
44
|
Zhang X, Galinsky RE, Kimura RE, Quinney SK, Jones DR, Hall SD. Inhibition of CYP3A by erythromycin: in vitro-in vivo correlation in rats. Drug Metab Dispos 2010; 38:61-72. [PMID: 19797607 DOI: 10.1124/dmd.109.028290] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
The prediction of in vivo drug-drug interactions from in vitro enzyme inhibition parameters remains challenging, particularly when time-dependent inhibition occurs. This study was designed to examine the accuracy of in vitro-derived parameters for the prediction of inhibition of CYP3A by erythromycin (ERY). Chronically cannulated rats were used to estimate the reduction in in vivo and in vitro intrinsic clearance (CL(int)) of midazolam (MDZ) after single and multiple doses of ERY; in vitro recovery of CL(int) was determined at 1, 2, 3, and 4 days after discontinuation of ERY. Enzyme inhibition parameters (k(inact), K(I), and K(i)) of ERY were estimated in vitro by using untreated rat liver microsomes. In vivo enzyme kinetic analysis indicated that single and multiple doses of ERY (150 mg/kg i.v. infusion over 4 h) reduced MDZ CL(int) by reversible and irreversible mechanisms, respectively. CYP3A inactivation after multiple doses of ERY treatment reflected metabolic intermediate complex formation without a significant change in hepatic CYP3A2 mRNA. A physiologically based pharmacokinetic model of the interaction between ERY and MDZ predicted a 2.6-fold decrease in CYP3A activity after repeated ERY treatment using in vitro-estimated enzyme inhibition parameters and in vivo degradation half-life of the enzyme (20 + or - 6 h). The observed -fold decreases were 2.3-fold and 2.1-fold for the in vitro-estimated CYP3A activity and the in vivo CL(int), respectively. This study demonstrates that in vivo DDIs are predictable from in vitro data when the appropriate model and parameter estimates are available.
Collapse
Affiliation(s)
- Xin Zhang
- Departments of Pharmacy Practice, School of Pharmacy and Pharmaceutical Sciences, Purdue University, West Lafayette, Indiana.
| | | | | | | | | | | |
Collapse
|
45
|
Zhang L, Zhang Y, Huang SM. Scientific and Regulatory Perspectives on Metabolizing Enzyme−Transporter Interplay and Its Role in Drug Interactions: Challenges in Predicting Drug Interactions. Mol Pharm 2009; 6:1766-74. [DOI: 10.1021/mp900132e] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- Lei Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| | - Yuanchao Zhang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| | - Shiew-Mei Huang
- Office of Clinical Pharmacology, Office of Translational Sciences, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, Maryland 20993
| |
Collapse
|
46
|
Nolin TD, Frye RF, Le P, Sadr H, Naud J, Leblond FA, Pichette V, Himmelfarb J. ESRD impairs nonrenal clearance of fexofenadine but not midazolam. J Am Soc Nephrol 2009; 20:2269-76. [PMID: 19696225 DOI: 10.1681/asn.2009010082] [Citation(s) in RCA: 102] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
ESRD can affect the pharmacokinetic disposition of drugs subject to nonrenal clearance. Cytochrome P450 (CYP) enzymes, including CYP3A, and multiple intestinal and hepatic drug transporters are thought to mediate this process, but the extent to which kidney disease alters the function of these proteins in humans is unknown. We used midazolam and fexofenadine to assess CYP3A (intestinal and hepatic) and drug transport, respectively, in patients with ESRD and healthy control subjects. We evaluated the effect of uremia on CYP3A and transporter expression in vitro by incubating normal rat hepatocytes and enterocytes with serum drawn from study participants. ESRD dramatically reduced nonrenal transporter function, evidenced by a 63% decrease in clearance (P < 0.001) and a 2.8-fold increase in area under the plasma concentration-time curve for fexofenadine (P = 0.002), compared with control subjects. We did not observe significant differences in midazolam or 1'-hydroxymidazolam clearance or area under the curve after oral administration, suggesting that CYP3A function is not changed by ESRD. Changes in hepatocyte and enterocyte protein expression in the presence of uremic serum were consistent with in vivo results. These findings demonstrate a mechanism for altered drug disposition in kidney disease, which may partially account for the high rates of drug toxicity in this population.
Collapse
Affiliation(s)
- Thomas D Nolin
- Department of Pharmacy and Therapeutics, and Center for Clinical Pharmaceutical Research, University of Pittsburgh School of Pharmacy, Pittsburgh, Pennsylvania 15261, USA.
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Verbeeck RK, Musuamba FT. Pharmacokinetics and dosage adjustment in patients with renal dysfunction. Eur J Clin Pharmacol 2009; 65:757-73. [PMID: 19543887 DOI: 10.1007/s00228-009-0678-8] [Citation(s) in RCA: 180] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2008] [Accepted: 05/30/2009] [Indexed: 01/11/2023]
Abstract
INTRODUCTION Chronic kidney disease is a common, progressive illness that is becoming a global public health problem. In patients with kidney dysfunction, the renal excretion of parent drug and/or its metabolites will be impaired, leading to their excessive accumulation in the body. In addition, the plasma protein binding of drugs may be significantly reduced, which in turn could influence the pharmacokinetic processes of distribution and elimination. The activity of several drug-metabolizing enzymes and drug transporters has been shown to be impaired in chronic renal failure. In patients with end-stage renal disease, dialysis techniques such as hemodialysis and continuous ambulatory peritoneal dialysis may remove drugs from the body, necessitating dosage adjustment. METHODS Inappropriate dosing in patients with renal dysfunction can cause toxicity or ineffective therapy. Therefore, the normal dosage regimen of a drug may have to be adjusted in a patient with renal dysfunction. Dosage adjustment is based on the remaining kidney function, most often estimated on the basis of the patient's glomerular filtration rate (GFR) estimated by the Cockroft-Gault formula. Net renal excretion of drug is a combination of three processes: glomerular filtration, tubular secretion and tubular reabsorption. Therefore, dosage adjustment based on GFR may not always be appropriate and a re-evaluation of markers of renal function may be required. DISCUSSION According to EMEA and FDA guidelines, a pharmacokinetic study should be carried out during the development phase of a new drug that is likely to be used in patients with renal dysfunction and whose pharmacokinetics are likely to be significantly altered in these patients. This study should be carried out in carefully selected subjects with varying degrees of renal dysfunction. In addition to this two-stage pharmacokinetic approach, a population PK/PD study in patients participating in phase II/phase III clinical trials can also be used to assess the impact of renal dysfunction on the drug's pharmacokinetics and pharmacodynamics. CONCLUSION In conclusion, renal dysfunction affects more that just the renal handling of drugs and/or active drug metabolites. Even when the dosage adjustment recommended for patients with renal dysfunction are carefully followed, adverse drug reactions remain common.
Collapse
Affiliation(s)
- Roger K Verbeeck
- Faculty of Pharmacy, Rhodes University, Grahamstown, Eastern Cape, South Africa.
| | | |
Collapse
|
48
|
Soars MG, Webborn PJH, Riley RJ. Impact of Hepatic Uptake Transporters on Pharmacokinetics and Drug−Drug Interactions: Use of Assays and Models for Decision Making in the Pharmaceutical Industry. Mol Pharm 2009; 6:1662-77. [DOI: 10.1021/mp800246x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Affiliation(s)
- Mathew G. Soars
- Department of Discovery DMPK, AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, Leicestershire LE11 5RH, U.K
| | - Peter J. H. Webborn
- Department of Discovery DMPK, AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, Leicestershire LE11 5RH, U.K
| | - Robert J. Riley
- Department of Discovery DMPK, AstraZeneca R&D Charnwood, Bakewell Road, Loughborough, Leicestershire LE11 5RH, U.K
| |
Collapse
|
49
|
Hosea NA, Collard WT, Cole S, Maurer TS, Fang RX, Jones H, Kakar SM, Nakai Y, Smith BJ, Webster R, Beaumont K. Prediction of human pharmacokinetics from preclinical information: comparative accuracy of quantitative prediction approaches. J Clin Pharmacol 2009; 49:513-33. [PMID: 19299532 DOI: 10.1177/0091270009333209] [Citation(s) in RCA: 225] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Quantitative prediction of human pharmacokinetics is critical in assessing the viability of drug candidates and in determining first-in-human dosing. Numerous prediction methodologies, incorporating both in vitro and preclinical in vivo data, have been developed in recent years, each with advantages and disadvantages. However, the lack of a comprehensive data set, both preclinical and clinical, has limited efforts to evaluate the optimal strategy (or strategies) that results in quantitative predictions of human pharmacokinetics. To address this issue, the authors conducted a retrospective analysis using 50 proprietary compounds for which in vitro, preclinical pharmacokinetic data and oral single-dose human pharmacokinetic data were available. Five predictive strategies, involving either allometry or use of unbound intrinsic clearance from microsomes or hepatocytes, were then compared for their ability to predict human oral clearance, half-life through predictions of systemic clearance, volume of distribution, and bioavailability. Use of a single-species scaling approach with rat, dog, or monkey was as accurate as or more accurate than using multiple-species allometry. For those compounds cleared almost exclusively by P450-mediated pathways, scaling from human liver microsomes was as predictive as single-species scaling of clearance based on data from rat, dog, or monkey. These data suggest that use of predictive methods involving either single-species in vivo data or in vitro human liver microsomes can quantitatively predict human in vivo pharmacokinetics and suggest the possibility of streamlining the predictive methodology through use of a single species or use only of human in vitro microsomal preparations.
Collapse
Affiliation(s)
- Natilie A Hosea
- Pfizer Inc, Department of Pharmacokinetics, Dynamics & Metabolism, San Diego, CA 92121, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Fagerholm U. Presentation of a modified dispersion model (MDM) for hepatic drug extraction and a new methodology for the prediction of the rate-limiting step in hepatic metabolic clearance. Xenobiotica 2009; 39:57-71. [DOI: 10.1080/00498250802562652] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|