1
|
Zhou Y, Zhong Y, Lauschke VM. Evaluating the synergistic use of advanced liver models and AI for the prediction of drug-induced liver injury. Expert Opin Drug Metab Toxicol 2025; 21:563-577. [PMID: 39893552 DOI: 10.1080/17425255.2025.2461484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/29/2025] [Indexed: 02/04/2025]
Abstract
INTRODUCTION Drug-induced liver injury (DILI) is a leading cause of acute liver failure. Hepatotoxicity typically occurs only in a subset of individuals after prolonged exposure and constitutes a major risk factor for the termination of drug development projects. AREAS COVERED We provide an overview of available human liver models for DILI research and discuss how they have been used to aid in early risk assessments and to mitigate the risk of project closures due to DILI in clinical stages. We summarize the different data that can be provided by such models and illustrate how these diverse data types can be interfaced with machine learning strategies to improve predictions of liver safety liabilities. EXPERT OPINION Advanced human liver models closely mimic human liver phenotypes and functions for many weeks, allowing for the recapitulation of hepatotoxicity events in vitro. Integration of the biochemical, histological, and toxicogenomic output data from these models with physicochemical compound properties using different machine learning architectures holds promise to enhance preclinical DILI predictions. However, to realize this aim, it is important to benchmark the available liver models on test sets of DILI positive and negative compounds and to carefully annotate and share the resulting data.
Collapse
Affiliation(s)
- Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
| | - Yi Zhong
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Pharmaceutical Informatics Institute, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, China
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
- Center for Molecular Medicine, Karolinska Institutet and University Hospital, Stockholm, Sweden
- Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany
- University of Tübingen, Tübingen, Germany
- Department of Pharmacy, The Second Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
2
|
Fardel O, Moreau A, Jouan E, Denizot C, Le Vée M, Parmentier Y. Human liver cell-based assays for the prediction of hepatic bile acid efflux transporter inhibition by drugs. Expert Opin Drug Metab Toxicol 2025; 21:463-480. [PMID: 39799554 DOI: 10.1080/17425255.2025.2453486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 12/11/2024] [Accepted: 01/10/2025] [Indexed: 01/15/2025]
Abstract
INTRODUCTION Drug-mediated inhibition of bile salt efflux transporters may cause liver injury. In vitro prediction of drug effects toward canalicular and/or sinusoidal efflux of bile salts from human hepatocytes is therefore a major issue, which can be addressed using liver cell-based assays. AREA COVERED This review, based on a thorough literature search in the scientific databases PubMed and Web of Science, provides key information about hepatic transporters implicated in bile salt efflux, the human liver cell models available for investigating functional inhibition of bile salt efflux, the different methodologies used for this purpose, and the modes of expression of the results. Applications of the assays to drugs are summarized, with special emphasis to the performance values of some assays for predicting hepatotoxicity/cholestatic effects of drugs. EXPERT OPINION Human liver cell-based assays for evaluating drug-mediated inhibition of bile acid efflux transporters face various limitations, such as the lack of method standardization and validation, the present poor adaptability to high throughput approaches, and some pitfalls with respect to interpretation of bile acid biliary excretion indexes. Hepatotoxicity of drugs is additionally likely multifactorial, highlighting that inhibition of hepatic bile salt efflux by drugs provides important, but not full, information about potential drug hepatotoxicity.
Collapse
Affiliation(s)
- Olivier Fardel
- Univ Rennes, CHU Rennes, Inserm, EHESP, Irset (Institut de recherche en santé, environnement et travail) - UMR_S 1085, Rennes, France
| | - Amélie Moreau
- Institut de R&D Servier, Paris-Saclay Gif-sur-Yvette, France
| | - Elodie Jouan
- Univ Rennes, Inserm, EHESP, Irset - UMR_S 1085, Rennes, France
| | - Claire Denizot
- Institut de R&D Servier, Paris-Saclay Gif-sur-Yvette, France
| | - Marc Le Vée
- Univ Rennes, Inserm, EHESP, Irset - UMR_S 1085, Rennes, France
| | | |
Collapse
|
3
|
Niu C, Xie X, Liu R, Liang X, Hu Y, Lai Y. CYP7A1 Gene Induction via SHP-Dependent or Independent Mechanisms can Increase the Risk of Drug-Induced Liver Injury Independently or in Synergy with BSEP Inhibition. Drug Metab Dispos 2024; 52:432-441. [PMID: 38485279 DOI: 10.1124/dmd.124.001675] [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: 01/26/2024] [Accepted: 03/07/2024] [Indexed: 04/18/2024] Open
Abstract
Drug-induced liver injury (DILI) is a frequent cause of clinical trial failures during drug development. While inhibiting bile salt export pump (BSEP) is a well-documented DILI mechanism, interference with genes related to bile acid (BA) metabolism and transport can further complicate DILI development. Here, the effects of twenty-eight compounds on genes associated with BA metabolism and transport were evaluated, including those with discontinued development or use, boxed warnings, and clean labels for DILI. The study also included rifampicin and omeprazole, pregnane X receptor and aryl hydrocarbon receptor ligands, and four mitogen-activated protein kinase kinase (MEK1/2) inhibitors. BSEP inhibitors with more severe DILI, notably pazopanib and CP-724714, significantly upregulated the expression of 7 alpha-hydroxylase (CYP7A1), independent of small heterodimer partner (SHP) expression. CYP7A1 expression was marginally induced by omeprazole. In contrast, its expression was suppressed by mometasone (10-fold), vinblastine (18-fold), hexachlorophene (2-fold), bosentan (2.1-fold), and rifampin (2-fold). All four MEK1/2 inhibitors that show clinical DILI were not potent BSEP inhibitors but significantly induced CYP7A1 expression, accompanied by a significant SHP gene suppression. Sulfotransferase 2A1 and BSEP were marginally upregulated, but no other genes were altered by the drugs tested. Protein levels of CYP7A1 were increased with the treatment of CYP7A1 inducers and decreased with obeticholic acid, an farnesoid X receptor ligand. CYP7A1 inducers significantly increased bile acid (BA) production in hepatocytes, indicating the overall regulatory effects of BA metabolism. This study demonstrates that CYP7A1 induction via various mechanisms can pose a risk for DILI, independently or in synergy with BSEP inhibition, and it should be evaluated early in drug discovery. SIGNIFICANCE STATEMENT: Kinase inhibitors, pazopanib and CP-724714, inhibit BSEP and induce CYP7A1 expression independent of small heterodimer partner (SHP) expression, leading to increased bile acid (BA) production and demonstrating clinically elevated drug-induced liver toxicity. MEK1/2 inhibitors that show BSEP-independent drug-induced liver injury (DILI) induced the CYP7A1 gene accompanied by SHP suppression. CYP7A1 induction via SHP-dependent or independent mechanisms can pose a risk for DILI, independently or in synergy with BSEP inhibition. Monitoring BA production in hepatocytes can reliably detect the total effects of BA-related gene regulation for de-risking.
Collapse
Affiliation(s)
- Congrong Niu
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Xiaodong Xie
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Renmeng Liu
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Xiaomin Liang
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Yiding Hu
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| |
Collapse
|
4
|
Youhanna S, Kemas AM, Preiss L, Zhou Y, Shen JX, Cakal SD, Paqualini FS, Goparaju SK, Shafagh RZ, Lind JU, Sellgren CM, Lauschke VM. Organotypic and Microphysiological Human Tissue Models for Drug Discovery and Development-Current State-of-the-Art and Future Perspectives. Pharmacol Rev 2022; 74:141-206. [PMID: 35017176 DOI: 10.1124/pharmrev.120.000238] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Accepted: 10/12/2021] [Indexed: 12/11/2022] Open
Abstract
The number of successful drug development projects has been stagnant for decades despite major breakthroughs in chemistry, molecular biology, and genetics. Unreliable target identification and poor translatability of preclinical models have been identified as major causes of failure. To improve predictions of clinical efficacy and safety, interest has shifted to three-dimensional culture methods in which human cells can retain many physiologically and functionally relevant phenotypes for extended periods of time. Here, we review the state of the art of available organotypic culture techniques and critically review emerging models of human tissues with key importance for pharmacokinetics, pharmacodynamics, and toxicity. In addition, developments in bioprinting and microfluidic multiorgan cultures to emulate systemic drug disposition are summarized. We close by highlighting important trends regarding the fabrication of organotypic culture platforms and the choice of platform material to limit drug absorption and polymer leaching while supporting the phenotypic maintenance of cultured cells and allowing for scalable device fabrication. We conclude that organotypic and microphysiological human tissue models constitute promising systems to promote drug discovery and development by facilitating drug target identification and improving the preclinical evaluation of drug toxicity and pharmacokinetics. There is, however, a critical need for further validation, benchmarking, and consolidation efforts ideally conducted in intersectoral multicenter settings to accelerate acceptance of these novel models as reliable tools for translational pharmacology and toxicology. SIGNIFICANCE STATEMENT: Organotypic and microphysiological culture of human cells has emerged as a promising tool for preclinical drug discovery and development that might be able to narrow the translation gap. This review discusses recent technological and methodological advancements and the use of these systems for hit discovery and the evaluation of toxicity, clearance, and absorption of lead compounds.
Collapse
Affiliation(s)
- Sonia Youhanna
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Aurino M Kemas
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Lena Preiss
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Yitian Zhou
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Joanne X Shen
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Selgin D Cakal
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Francesco S Paqualini
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Sravan K Goparaju
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Reza Zandi Shafagh
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Johan Ulrik Lind
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Carl M Sellgren
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden (S.Y., A.M.K., L.P., Y.Z., J.X.S., S.K.G., R.Z.S., C.M.S., V.M.L.); Department of Drug Metabolism and Pharmacokinetics (DMPK), Merck KGaA, Darmstadt, Germany (L.P.); Department of Health Technology, Technical University of Denmark, Lyngby, Denmark (S.D.C., J.U.L.); Synthetic Physiology Laboratory, Department of Civil Engineering and Architecture, University of Pavia, Pavia, Italy (F.S.P.); Division of Micro- and Nanosystems, KTH Royal Institute of Technology, Stockholm, Sweden (Z.S.); and Dr Margarete Fischer-Bosch Institute of Clinical Pharmacology, Stuttgart, Germany (V.M.L.)
| |
Collapse
|
5
|
Neuhoff S, Harwood MD, Rostami-Hodjegan A, Achour B. Application of proteomic data in the translation of in vitro observations to associated clinical outcomes. DRUG DISCOVERY TODAY. TECHNOLOGIES 2021; 39:13-22. [PMID: 34906322 DOI: 10.1016/j.ddtec.2021.06.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 04/20/2021] [Accepted: 06/11/2021] [Indexed: 12/12/2022]
Abstract
Translation of information on drug exposure and effect is facilitated by in silico models that enable extrapolation of in vitro measurements to in vivo clinical outcomes. These models integrate drug-specific data with information describing physiological processes and pathological changes, including alterations to proteins involved in drug absorption, distribution and elimination. Over the past 15 years, quantitative proteomics has contributed a wealth of protein expression data, which are currently used for a variety of systems pharmacology applications, as a complement or a surrogate for activity of the corresponding proteins. In this review, we explore current and emerging applications of targeted and global (untargeted) proteomics in translational pharmacology as well as strategies for improved integration into model-based drug development.
Collapse
Affiliation(s)
- Sibylle Neuhoff
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Matthew D Harwood
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK
| | - Amin Rostami-Hodjegan
- Certara UK Limited, Simcyp Division, 1 Concourse Way, Sheffield, S1 2BJ, UK; Centre for Applied Pharmacokinetic Research (CAPKR), School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK
| | - Brahim Achour
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Health Sciences, University of Manchester, Stopford Building, Oxford Road, Manchester, M13 9PT, UK.
| |
Collapse
|
6
|
Bowman CM, Chen B, Cheong J, Liu L, Chen Y, Mao J. Improving the Translation of Organic Anion Transporting Polypeptide Substrates using HEK293 Cell Data in the Presence and Absence of Human Plasma via Physiologically Based Pharmacokinetic Modeling. Drug Metab Dispos 2021; 49:530-539. [PMID: 33958385 DOI: 10.1124/dmd.120.000315] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Accepted: 04/13/2021] [Indexed: 11/22/2022] Open
Abstract
Accurately predicting the pharmacokinetics of compounds that are transporter substrates has been notoriously challenging using traditional in vitro systems and physiologically based pharmacokinetic (PBPK) modeling. The objective of this study was to use PBPK modeling to understand the translational accuracy of data generated with human embryonic kidney 293 (HEK293) cells overexpressing the hepatic uptake transporters organic anion transporting polypeptide (OATP) 1B1/3 with and without plasma while accounting for transporter expression. Models of four OATP substrates, two with low protein binding (pravastatin and rosuvastatin) and two with high protein binding (repaglinide and pitavastatin) were explored, and the OATP in vitro data generated in plasma incubations were used for a plasma model, and in buffer incubations for a buffer model. The pharmacokinetic parameters and concentration-time profiles of pravastatin and rosuvastatin were similar and well predicted (within 2-fold of observed values) using the plasma and buffer models without needing an empirical scaling factor, whereas the dispositions of the highly protein bound repaglinide and pitavastatin were more accurately simulated with the plasma models than the buffer models. This work suggests that data from HEK293 overexpressing transporter cells corrected for transporter expression represent a valid approach to improve bottom-up PBPK modeling for highly protein bound OATP substrates with plasma incubations and low protein binding OATP substrates with or without plasma incubations. SIGNIFICANCE STATEMENT: This work demonstrates the bottom-up approach of using in vitro data directly without employing empirical scaling factors to predict the intravenous pharmacokinetic (PK) profiles reasonably well for four organic anion transporting polypeptide (OATP) substrates. Based on these results, using HEK293 overexpressing cells, examining the impact of plasma for highly bound compounds, and incorporating transporter quantitation for the lot in which the in vitro data were generated represents a valid approach to achieve more accurate prospective PK predictions for OATP substrates.
Collapse
Affiliation(s)
- Christine M Bowman
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Buyun Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Jonathan Cheong
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Liling Liu
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Yuan Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| |
Collapse
|
7
|
Liang X, Lai Y. Overcoming the shortcomings of the extended-clearance concept: a framework for developing a physiologically-based pharmacokinetic (PBPK) model to select drug candidates involving transporter-mediated clearance. Expert Opin Drug Metab Toxicol 2021; 17:869-886. [PMID: 33793347 DOI: 10.1080/17425255.2021.1912012] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Introduction:Human pharmacokinetic (PK) prediction can be a significant challenge to drug candidates undergoing transporter-mediated clearance, when only animal data and in vitro human parameters are available in the drug discovery stage.Areas covered:The extended clearance concept (ECC) that incorporates the processes of hepatic uptake, passive diffusion, metabolism and biliary secretion has been adapted to determine the rate-determining process of hepatic clearance and drug-drug interactions (DDIs). However, since the ECC is derived from the well-stirred model and does not consider the liver as a drug distribution organ to reflect the time-dependent variation of drug concentrations between the liver and plasma, it can be misused for compound selection in drug discovery.Expert opinion:The PBPK model consists of a set of differential equations of drug mass balance, and can overcome the shortcomings of the ECC in predicting human PK. The predictability, relevance and reliability of the model and the scaling factors for IVIVE must be validated using either the measured liver concentrations or DDI data with known transporter inhibitors, or both, in monkeys. A human PBPK model that incorporates in vitro human data and SFs obtained from the validated monkey PBPK model can be used for compound selection in the drug discovery phase.
Collapse
Affiliation(s)
- Xiaomin Liang
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, CA, USA
| |
Collapse
|
8
|
Bowman CM, Ma F, Mao J, Chen Y. Examination of Physiologically-Based Pharmacokinetic Models of Rosuvastatin. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2020; 10:5-17. [PMID: 33220025 PMCID: PMC7825190 DOI: 10.1002/psp4.12571] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Accepted: 10/19/2020] [Indexed: 12/14/2022]
Abstract
Physiologically‐based pharmacokinetic (PBPK) modeling is increasingly used to predict drug disposition and drug–drug interactions (DDIs). However, accurately predicting the pharmacokinetics of transporter substrates and transporter‐mediated DDIs (tDDIs) is still challenging. Rosuvastatin is a commonly used substrate probe in DDI risk assessment for new molecular entities (NMEs) that are potential organic anion transporting polypeptide 1B or breast cancer resistance protein transporter inhibitors, and as such, several rosuvastatin PBPK models have been developed to try to predict the clinical DDI and support NME drug labeling. In this review, we examine five representative PBPK rosuvastatin models, discuss common challenges that the models have come across, and note remaining gaps. These shared learnings will help with the continuing efforts of rosuvastatin model validation, provide more information to understand transporter‐mediated drug disposition, and increase confidence in tDDI prediction.
Collapse
Affiliation(s)
- Christine M Bowman
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Fang Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Jialin Mao
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| | - Yuan Chen
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California, USA
| |
Collapse
|
9
|
Liang X, Park Y, DeForest N, Hao J, Zhao X, Niu C, Wang K, Smith B, Lai Y. In Vitro Hepatic Uptake in Human and Monkey Hepatocytes in the Presence and Absence of Serum Protein and Its In Vitro to In Vivo Extrapolation. Drug Metab Dispos 2020; 48:1283-1292. [PMID: 33037043 DOI: 10.1124/dmd.120.000163] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 09/28/2020] [Indexed: 12/19/2022] Open
Abstract
It is well documented that human hepatic clearance based on in vitro metabolism or transporter assays systematically resulted in underprediction; therefore, large empirical scalars are often needed in either static or physiologically based pharmacokinetic (PBPK) models to accurately predict human pharmacokinetics (PK). In our current investigation, we assessed hepatic uptake in hepatocyte suspension in Krebs-Henseleit buffer in the presence and absence of serum. The results showed that the unbound intrinsic active clearance (CLu,int,active) values obtained by normalizing the unbound fraction in the buffer containing 10% serum were generally higher than the CLu,int,active obtained directly from protein free buffer, suggesting "protein-facilitated" uptake. The differences of CLu,int,active in the buffer with and without protein ranged from 1- to 925-fold and negatively correlated to the unbound serum binding of organic anion transporting polypeptide substrates. When using the uptake values obtained from buffer containing serum versus serum-free buffer, the median of scaling factors (SFs) for CLu,int,active reduced from 24.2-4.6 to 22.7-7.1 for human and monkey, respectively, demonstrating the improvement of in vitro to in vivo extrapolation in a PBPK model. Furthermore, values of CLu,int,active were significantly higher in monkey hepatocytes than that in human, and the species differences appeared to be compound dependent. Scaling up in vitro uptake values derived in assays containing species-specific serum can compensate for the species-specific variabilities when using cynomolgus monkey as a probe animal model. Incorporating SFs calibrated in monkey and together with scaled in vitro data can be a reliable approach for the prospective human PK prediction in early drug discovery. SIGNIFICANCE STATEMENT: We investigated the protein effect on hepatic uptake in human and monkey hepatocytes and improved the in vitro to in vivo extrapolation using parameters obtained from the incubation in the present of serum protein. In addition, significantly higher active uptake clearances were observed in monkey hepatocytes than in human, and the species differences appeared to be compound dependent. The physiologically based pharmacokinetic model that incorporates scaling factors calibrated in monkey and together with scaled in vitro human data can be a reliable approach for the prospective human pharmacokinetics prediction.
Collapse
Affiliation(s)
- Xiaomin Liang
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Yeojin Park
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | | | - Jia Hao
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Xiaofeng Zhao
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Congrong Niu
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Kelly Wang
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Bill Smith
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., Foster City, California
| |
Collapse
|
10
|
Achour B, Al-Majdoub ZM, Rostami-Hodjegan A, Barber J. Mass Spectrometry of Human Transporters. ANNUAL REVIEW OF ANALYTICAL CHEMISTRY (PALO ALTO, CALIF.) 2020; 13:223-247. [PMID: 32084322 DOI: 10.1146/annurev-anchem-091719-024553] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Transporters are key to understanding how an individual will respond to a particular dose of a drug. Two patients with similar systemic concentrations may have quite different local concentrations of a drug at the required site. The transporter profile of any individual depends upon a variety of genetic and environmental factors, including genotype, age, and diet status. Robust models (virtual patients) are therefore required and these models are data hungry. Necessary data include quantitative transporter profiles at the relevant organ. Liquid chromatography with tandem mass spectrometry (LC-MS/MS) is currently the most powerful method available for obtaining this information. Challenges include sourcing the tissue, isolating the hydrophobic membrane-embedded transporter proteins, preparing the samples for MS (including proteolytic digestion), choosing appropriate quantification methodology, and optimizing the LC-MS/MS conditions. Great progress has been made with all of these, especially within the last few years, and is discussed here.
Collapse
Affiliation(s)
- Brahim Achour
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Zubida M Al-Majdoub
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
- Certara, Princeton, New Jersey 08540, USA
| | - Jill Barber
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PT, United Kingdom;
| |
Collapse
|
11
|
Niu C, Wang Y, Zhao X, Tep S, Murakami E, Subramanian R, Smith B, Lai Y. Organic Anion-Transporting Polypeptide Genes Are Not Induced by the Pregnane X Receptor Activator Rifampin: Studies in Hepatocytes In Vitro and in Monkeys In Vivo. Drug Metab Dispos 2019; 47:1433-1442. [PMID: 31582395 DOI: 10.1124/dmd.119.088922] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 09/27/2019] [Indexed: 12/27/2022] Open
Abstract
Induction potentials of the pregnane X receptor (PXR) activator rifampin (RIF) on transporter genes [e.g., organic anion-transporting polypeptides (OATPs)] are still in its infancy or remain controversial in the field. The present investigations characterized changes in transporter gene expression by RIF in sandwich-cultured hepatocytes from multiple donors of human and cynomolgus monkey using real-time quantitative reverse transcription polymerase chain reaction method. Three-day treatment of RIF significantly induced CYP3A4 (∼60-fold induction), but not CYP1A2 and CYP2D6 genes. SLC51B was the most highly induced uptake transporter gene (>10-fold) in both human and monkey hepatocytes. A greater induction of CYP2C9 was observed in monkey hepatocytes than that in humans. ATP-binding cassette (ABC)B1 and ABCC2 were induced slightly above 2-fold in human and monkey hepatocytes and appeared to be dose-dependent. The induction of OATP and other transporter genes was generally less than 2-fold and considered not clinically relevant. SLCO2B1 was not detectable in monkey hepatocytes. To investigate in vivo OATP induction, RIF (18 mg/kg per day) was orally dosed to cynomolgus monkeys for 7 days. Pitavastatin and antipyrine were intravenously dosed before and after RIF treatment as exogenous probes of OATP and CYP activities, respectively. Plasma coproporphyrin-I (CP-I) and coproporphyrin-III (CP-III) were measured as OATP endogenous biomarkers. Although a significant increase of antipyrine clearance (CL) was observed after RIF treatment, the plasma exposures of pitavastatin, CP-I, and CP-III remained unchanged, suggesting that OATP function was not significantly altered. The results suggested that OATP transporters were not significantly induced by PXR ligand RIF. The data are consistent with current regulatory guidances that the in vitro characterization of transporter induction during drug development is not required. SIGNIFICANCE STATEMENT: Organic anion-transporting polypeptide (OATP) genes were not induced by rifampin in sandwich-cultured human and monkey hepatocytes OATP functions measured by OATP probe pitavastatin and endogenous marker coproporphyrins were not altered in monkeys in vivo by 7-day rifampin treatment. The data suggested that OATP transporters are unlikely induced by the pregnane X receptor ligand rifampin, which are consistent with current regulatory guidances that the in vitro characterization of OATP1B induction during drug development is not required.
Collapse
Affiliation(s)
- Congrong Niu
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Yujin Wang
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Xiaofeng Zhao
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Sam Tep
- Drug Metabolism, Gilead Sciences, Foster City, California
| | | | | | - Bill Smith
- Drug Metabolism, Gilead Sciences, Foster City, California
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences, Foster City, California
| |
Collapse
|
12
|
Evaluation of Drug Biliary Excretion Using Sandwich-Cultured Human Hepatocytes. Eur J Drug Metab Pharmacokinet 2019; 44:13-30. [PMID: 30167999 DOI: 10.1007/s13318-018-0502-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Evaluation of hepatobiliary transport of drugs is an important challenge, notably during the development of new molecular identities. In this context, sandwich-cultured human hepatocytes (SCHH) have been proposed as an interesting and integrated tool for predicting in vitro biliary excretion of drugs. The present review was therefore designed to summarize key findings about SCHH, including their establishment, their main functional features and their use for the determination of canalicular transport and the prediction of in vivo biliary clearance and hepatobiliary excretion-related drug-drug interactions. Reviewed data highlight the fact that SCHH represent an original and probably unique holistic in vitro approach to predict biliary clearance in humans, through taking into account sinusoidal drug uptake, passive drug diffusion, drug metabolism and sinusoidal and canalicular drug efflux. Limits and proposed refinements for SCHH-based analysis of drug biliary excretion, as well as putative human alternative in vitro models to SCHH are also discussed.
Collapse
|
13
|
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
|
14
|
Wegler C, Gaugaz FZ, Andersson TB, Wiśniewski JR, Busch D, Gröer C, Oswald S, Norén A, Weiss F, Hammer HS, Joos TO, Poetz O, Achour B, Rostami-Hodjegan A, van de Steeg E, Wortelboer HM, Artursson P. Variability in Mass Spectrometry-based Quantification of Clinically Relevant Drug Transporters and Drug Metabolizing Enzymes. Mol Pharm 2017; 14:3142-3151. [DOI: 10.1021/acs.molpharmaceut.7b00364] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Christine Wegler
- Department
of Pharmacy, Uppsala University, Uppsala 75123, Sweden
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal 431 50, Sweden
| | | | - Tommy B. Andersson
- Cardiovascular and Metabolic Diseases, Innovative Medicines and Early Development Biotech Unit, AstraZeneca R&D, Mölndal 431 50, Sweden
| | - Jacek R. Wiśniewski
- Biochemical
Proteomics Group, Department of Proteomics and Signal Transduction, Max Planck Institute of Biochemistry, Martinsried 82152, Germany
| | - Diana Busch
- Center
of Drug Absorption and Transport, Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald 17489, Germany
| | - Christian Gröer
- Center
of Drug Absorption and Transport, Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald 17489, Germany
| | - Stefan Oswald
- Center
of Drug Absorption and Transport, Department of Clinical Pharmacology, University Medicine of Greifswald, Greifswald 17489, Germany
| | - Agneta Norén
- Department
of Surgical Sciences, Uppsala University, Uppsala 75185, Sweden
| | - Frederik Weiss
- NMI Natural
and Medical Sciences Institute, University of Tübingen, Reutlingen 72770, Germany
| | - Helen S. Hammer
- NMI Natural
and Medical Sciences Institute, University of Tübingen, Reutlingen 72770, Germany
| | - Thomas O. Joos
- NMI Natural
and Medical Sciences Institute, University of Tübingen, Reutlingen 72770, Germany
| | - Oliver Poetz
- NMI Natural
and Medical Sciences Institute, University of Tübingen, Reutlingen 72770, Germany
| | - Brahim Achour
- Centre
for Applied Pharmacokinetic Research, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Amin Rostami-Hodjegan
- Centre
for Applied Pharmacokinetic Research, University of Manchester, Manchester M13 9PL, United Kingdom
| | - Evita van de Steeg
- TNO (Netherlands Organization for Applied Scientific Research), 3700 AJ Zeist, Netherlands
| | - Heleen M. Wortelboer
- TNO (Netherlands Organization for Applied Scientific Research), 3700 AJ Zeist, Netherlands
| | - Per Artursson
- Department
of Pharmacy, Uppsala University, Uppsala 75123, Sweden
| |
Collapse
|
15
|
Zhang Y, Jackson JP, St Claire RL, Freeman K, Brouwer KR, Edwards JE. Obeticholic acid, a selective farnesoid X receptor agonist, regulates bile acid homeostasis in sandwich-cultured human hepatocytes. Pharmacol Res Perspect 2017; 5. [PMID: 28805978 PMCID: PMC5684861 DOI: 10.1002/prp2.329] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022] Open
Abstract
Farnesoid X receptor (FXR) is a master regulator of bile acid homeostasis through transcriptional regulation of genes involved in bile acid synthesis and cellular membrane transport. Impairment of bile acid efflux due to cholangiopathies results in chronic cholestasis leading to abnormal elevation of intrahepatic and systemic bile acid levels. Obeticholic acid (OCA) is a potent and selective FXR agonist that is 100‐fold more potent than the endogenous ligand chenodeoxycholic acid (CDCA). The effects of OCA on genes involved in bile acid homeostasis were investigated using sandwich‐cultured human hepatocytes. Gene expression was determined by measuring mRNA levels. OCA dose‐dependently increased fibroblast growth factor‐19 (FGF‐19) and small heterodimer partner (SHP) which, in turn, suppress mRNA levels of cholesterol 7‐alpha‐hydroxylase (CYP7A1), the rate‐limiting enzyme for de novo synthesis of bile acids. Consistent with CYP7A1 suppression, total bile acid content was decreased by OCA (1 μmol/L) to 42.7 ± 20.5% relative to control. In addition to suppressing de novo bile acids synthesis, OCA significantly increased the mRNA levels of transporters involved in bile acid homeostasis. The bile salt excretory pump (BSEP), a canalicular efflux transporter, increased by 6.4 ± 0.8‐fold, and the basolateral efflux heterodimer transporters, organic solute transporter α (OSTα) and OSTβ increased by 6.4 ± 0.2‐fold and 42.9 ± 7.9‐fold, respectively. The upregulation of BSEP and OSTα and OSTβ, by OCA reduced the intracellular concentrations of d8‐TCA, a model bile acid, to 39.6 ± 8.9% relative to control. These data demonstrate that OCA does suppress bile acid synthesis and reduce hepatocellular bile acid levels, supporting the use of OCA to treat bile acid‐induced toxicity observed in cholestatic diseases.
Collapse
|
16
|
Cantrill C, Houston JB. Understanding the Interplay Between Uptake and Efflux Transporters Within In Vitro Systems in Defining Hepatocellular Drug Concentrations. J Pharm Sci 2017; 106:2815-2825. [PMID: 28478131 DOI: 10.1016/j.xphs.2017.04.056] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2017] [Revised: 04/21/2017] [Accepted: 04/24/2017] [Indexed: 02/03/2023]
Abstract
One of the most holistic in vitro systems for prediction of intracellular drug concentrations is sandwich-cultured hepatocytes (SCH); however, a comprehensive evaluation of the utility of SCH to estimate uptake and biliary clearances and the need for additional kinetic parameters has yet to be carried out. Toward this end, we have selected 9 compounds (rosuvastatin, valsartan, fexofenadine, pravastatin, repaglinide, telmisartan, atorvastatin, saquinavir, and quinidine) to provide a range of physicochemical and hepatic disposition properties. Uptake clearances were determined in SCH and compared with conventional monolayer and suspension hepatocyte systems, previously reported by our laboratory. CLuptake ranged from 1 to 41 μL/min/106 cells in SCH which were significantly lower (1%-10%) compared with the other hepatocyte models. The hepatocyte-to-media unbound concentration ratio (Kpu) has been assessed and ranged 0.7-59, lower compared with other hepatocyte systems (8-280). Estimates of in vitro biliary clearance (CLbile) for 4 drugs were determined and were scaled to predict in vivo values using both intracellular concentration and media drug concentrations. These studies demonstrate that reduced uptake in rat SCH may limit drug access to canalicular efflux transport proteins and highlight the importance of elucidating the interplay between these proteins for accurate prediction of hepatic clearance.
Collapse
Affiliation(s)
- Carina Cantrill
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Biology, Medicine and Health Sciences, University of Manchester, UK
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Biology, Medicine and Health Sciences, University of Manchester, UK.
| |
Collapse
|
17
|
Kimoto E, Bi YA, Kosa RE, Tremaine LM, Varma MVS. Hepatobiliary Clearance Prediction: Species Scaling From Monkey, Dog, and Rat, and In Vitro-In Vivo Extrapolation of Sandwich-Cultured Human Hepatocytes Using 17 Drugs. J Pharm Sci 2017; 106:2795-2804. [PMID: 28456723 DOI: 10.1016/j.xphs.2017.04.043] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 04/19/2017] [Accepted: 04/20/2017] [Indexed: 11/26/2022]
Abstract
Hepatobiliary elimination can be a major clearance pathway dictating the pharmacokinetics of drugs. Here, we first compared the dose eliminated in bile in preclinical species (monkey, dog, and rat) with that in human and further evaluated single-species scaling (SSS) to predict human hepatobiliary clearance. Six compounds dosed in bile duct-cannulated (BDC) monkeys showed biliary excretion comparable to human; and the SSS of hepatobiliary clearance with plasma fraction unbound correction yielded reasonable predictions (within 3-fold). Although dog SSS also showed reasonable predictions, rat overpredicted hepatobiliary clearance for 13 of 24 compounds. Second, we evaluated the translatability of in vitro sandwich-cultured human hepatocytes (SCHHs) to predict human hepatobiliary clearance for 17 drugs. For drugs with no significant active uptake in SCHH studies (i.e., with or without rifamycin SV), measured intrinsic biliary clearance was directly scalable with good predictability (absolute average fold error [AAFE] = 1.6). Drugs showing significant active uptake in SCHH, however, showed improved predictability when scaled based on extended clearance term (AAFE = 2.0), which incorporated sinusoidal uptake along with a global scaling factor for active uptake and the canalicular efflux clearance. In conclusion, SCHH is a useful tool to predict human hepatobiliary clearance, whereas BDC monkey model may provide further confidence in the prospective predictions.
Collapse
Affiliation(s)
- Emi Kimoto
- Pharmacokinetics, Pharmacodynamics & Metabolism Department-New Chemical Entities, Pfizer Inc., Groton, Connecticut 06340
| | - Yi-An Bi
- Pharmacokinetics, Pharmacodynamics & Metabolism Department-New Chemical Entities, Pfizer Inc., Groton, Connecticut 06340
| | - Rachel E Kosa
- Pharmacokinetics, Pharmacodynamics & Metabolism Department-New Chemical Entities, Pfizer Inc., Groton, Connecticut 06340
| | - Larry M Tremaine
- Pharmacokinetics, Pharmacodynamics & Metabolism Department-New Chemical Entities, Pfizer Inc., Groton, Connecticut 06340
| | - Manthena V S Varma
- Pharmacokinetics, Pharmacodynamics & Metabolism Department-New Chemical Entities, Pfizer Inc., Groton, Connecticut 06340.
| |
Collapse
|
18
|
Vrana M, Whittington D, Nautiyal V, Prasad B. Database of Optimized Proteomic Quantitative Methods for Human Drug Disposition-Related Proteins for Applications in Physiologically Based Pharmacokinetic Modeling. CPT-PHARMACOMETRICS & SYSTEMS PHARMACOLOGY 2017; 6:267-276. [PMID: 28074615 PMCID: PMC5397556 DOI: 10.1002/psp4.12170] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/16/2016] [Revised: 12/28/2016] [Accepted: 12/29/2016] [Indexed: 12/16/2022]
Abstract
The purpose of this study was to create an open access repository of validated liquid chromatography tandem mass spectrometry (LC‐MS/MS) multiple reaction monitoring (MRM) methods for quantifying 284 important proteins associated with drug absorption, distribution, metabolism, and excretion (ADME). Various in silico and experimental approaches were used to select surrogate peptides and optimize instrument parameters for LC‐MS/MS quantification of the selected proteins. The final methods were uploaded to an online public database (QPrOmics; www.qpromics.uw.edu/qpromics/assay/), which provides essential information for facile method development in triple quadrupole mass spectrometry (MS) instruments. To validate the utility of the methods, the differential tissue expression of 107 key ADME proteins was characterized in the tryptic digests of the pooled subcellular fractions of human liver, kidneys, intestines, and lungs. These methods and the data are critical for development of physiologically based pharmacokinetic (PBPK) models to predict xenobiotic disposition.
Collapse
Affiliation(s)
- M Vrana
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - D Whittington
- Medicinal Chemistry, University of Washington, Seattle, Washington, USA
| | - V Nautiyal
- Indian Institute of Technology Kharagpur, Kharagpur, West Bengal, India
| | - B Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| |
Collapse
|
19
|
Sandwich-Cultured Hepatocytes as a Tool to Study Drug Disposition and Drug-Induced Liver Injury. J Pharm Sci 2016; 105:443-459. [PMID: 26869411 DOI: 10.1016/j.xphs.2015.11.008] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2015] [Revised: 11/06/2015] [Accepted: 11/09/2015] [Indexed: 12/21/2022]
Abstract
Sandwich-cultured hepatocytes (SCH) are metabolically competent and have proper localization of basolateral and canalicular transporters with functional bile networks. Therefore, this cellular model is a unique tool that can be used to estimate biliary excretion of compounds. SCH have been used widely to assess hepatobiliary disposition of endogenous and exogenous compounds and metabolites. Mechanistic modeling based on SCH data enables estimation of metabolic and transporter-mediated clearances, which can be used to construct physiologically based pharmacokinetic models for prediction of drug disposition and drug-drug interactions in humans. In addition to pharmacokinetic studies, SCH also have been used to study cytotoxicity and perturbation of biological processes by drugs and hepatically generated metabolites. Human SCH can provide mechanistic insights underlying clinical drug-induced liver injury (DILI). In addition, data generated in SCH can be integrated into systems pharmacology models to predict potential DILI in humans. In this review, applications of SCH in studying hepatobiliary drug disposition and bile acid-mediated DILI are discussed. An example is presented to show how data generated in the SCH model were used to establish a quantitative relationship between intracellular bile acids and cytotoxicity, and how this information was incorporated into a systems pharmacology model for DILI prediction.
Collapse
|
20
|
Lapham K, Novak J, Marroquin LD, Swiss R, Qin S, Strock CJ, Scialis R, Aleo MD, Schroeter T, Eng H, Rodrigues AD, Kalgutkar AS. Inhibition of Hepatobiliary Transport Activity by the Antibacterial Agent Fusidic Acid: Insights into Factors Contributing to Conjugated Hyperbilirubinemia/Cholestasis. Chem Res Toxicol 2016; 29:1778-1788. [DOI: 10.1021/acs.chemrestox.6b00262] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Affiliation(s)
| | | | | | | | - Shuzhen Qin
- Biological
Screening and Assay Development, Cyprotex, Watertown, Massachusetts 02472, United States
| | - Christopher J. Strock
- Biological
Screening and Assay Development, Cyprotex, Watertown, Massachusetts 02472, United States
| | | | | | | | | | | | - Amit S. Kalgutkar
- Pharmacokinetics,
Dynamics, and Metabolism Department, Pfizer Worldwide Research and Development, Cambridge, Massachusetts 02139, United States
| |
Collapse
|
21
|
Prasad B, Gaedigk A, Vrana M, Gaedigk R, Leeder JS, Salphati L, Chu X, Xiao G, Hop C, Evers R, Gan L, Unadkat JD. Ontogeny of Hepatic Drug Transporters as Quantified by LC-MS/MS Proteomics. Clin Pharmacol Ther 2016; 100:362-70. [PMID: 27301780 PMCID: PMC5017908 DOI: 10.1002/cpt.409] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 05/24/2016] [Accepted: 06/06/2016] [Indexed: 12/16/2022]
Abstract
Protein expression of major hepatic uptake and efflux drug transporters in human pediatric (n = 69) and adult (n = 41) livers was quantified by liquid chromatography / tandem mass spectroscopy (LC-MS/MS). Transporter protein expression of OCT1, OATP1B3, P-gp, and MRP3 was age-dependent. Particularly, significant differences were observed in transporter expression (P < 0.05) between the following age groups: neonates vs. adults (OCT1, OATP1B3, P-gp), neonates or infants vs. adolescents and/or adults (OCT1, OATP1B3, and P-gp), infants vs. children (OATP1B3 and P-gp), and adolescents vs. adults (MRP3). OCT1 showed the largest increase, of almost 5-fold, in protein expression with age. Ontogenic expression of OATP1B1 was confounded by genotype and was revealed only in livers harboring SLCO1B1*1A/*1A. In livers >1 year, tissues harboring SLCO1B1*14/*1A showed 2.5-fold higher (P < 0.05) protein expression than SLCO1B1*15/*1A. Integration of these ontogeny data in physiologically based pharmacokinetic (PBPK) models will be a crucial step in predicting hepatic drug disposition in children.
Collapse
Affiliation(s)
- B Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.
| | - A Gaedigk
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - M Vrana
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA
| | - R Gaedigk
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - J S Leeder
- Division of Clinical Pharmacology, Toxicology and Therapeutic Innovation, Children's Mercy, Kansas City, Missouri, USA
- School of Medicine, University of Missouri-Kansas City, Kansas City, Missouri, USA
| | - L Salphati
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck Sharp & Dohme, Kenilworth, New Jersey, USA
| | - X Chu
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey, USA
| | - G Xiao
- Biogen, Cambridge, Massachusetts, USA
| | - Ceca Hop
- Department of Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck Sharp & Dohme, Kenilworth, New Jersey, USA
| | - R Evers
- Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey, USA
| | - L Gan
- Biogen, Cambridge, Massachusetts, USA
| | - J D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington, USA.
| |
Collapse
|
22
|
Cheng Y, Woolf TF, Gan J, He K. In vitro model systems to investigate bile salt export pump (BSEP) activity and drug interactions: A review. Chem Biol Interact 2015; 255:23-30. [PMID: 26683212 DOI: 10.1016/j.cbi.2015.11.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Revised: 11/10/2015] [Accepted: 11/26/2015] [Indexed: 01/04/2023]
Abstract
The bile salt export pump protein (BSEP), expressed on the canalicular membranes of hepatocytes, is primarily responsible for the biliary excretion of bile salts. The inhibition of BSEP transport activity can lead to an increase in intracellular bile salt levels and liver injury. This review discusses the various in vitro assays currently available for assessing the effect of drugs or other chemical entities to modulate BSEP transport activity. BSEP transporter assays use one of the following platforms: Xenopus laevis oocytes; canalicular membrane vesicles (CMV); BSEP-expressed membrane vesicles; cell lines expressing BSEP; sandwich cultured hepatocytes (SCH); and hepatocytes in suspension. Two of these, BSEP-expressed insect membrane vesicles and sandwich cultured hepatocytes, are the most commonly used assays. BSEP membrane vesicles prepared from transfected insect cells are useful for assessing BSEP inhibition or substrate specificity and exploring mechanisms of BSEP-associated genetic diseases. This model can be applied in a high-throughput format for discovery-drug screening. However, experimental results from use of membrane vesicles may lack physiological relevance and the model does not allow for investigation of in situ metabolism in modulation of BSEP activity. Hepatocyte-based assays that use the SCH format provide results that are generally more physiologically relevant than membrane assays. The SCH model is useful in detailed studies of the biliary excretion of drugs and BSEP inhibition, but due to the complexity of SCH preparation, this model is used primarily for determining biliary clearance and BSEP inhibition in a limited number of compounds. The newly developed hepatocyte in suspension assay avoids many of the complexities of the SCH method. The use of pooled cryopreserved hepatocytes in suspension minimizes genetic variance and individual differences in BSEP activity and also provides the opportunity for higher throughput screening and cross-species comparisons.
Collapse
Affiliation(s)
- Yaofeng Cheng
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, NJ 08543, USA
| | | | - Jinping Gan
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb Company, Princeton, NJ 08543, USA
| | - Kan He
- Biotranex LLC, Monmouth Junction, NJ 08852, USA.
| |
Collapse
|
23
|
Kimoto E, Li R, Scialis RJ, Lai Y, Varma MVS. Hepatic Disposition of Gemfibrozil and Its Major Metabolite Gemfibrozil 1-O-β-Glucuronide. Mol Pharm 2015; 12:3943-52. [DOI: 10.1021/acs.molpharmaceut.5b00411] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Emi Kimoto
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Rui Li
- Systems
Modeling and Simulation, Department of Pharmacokinetics, Dynamics,
and Metabolism, Pfizer Worldwide Research and Development, Cambridge, Massachusetts, 02139, United States
| | - Renato J. Scialis
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| | - Yurong Lai
- Departments
of Metabolism and Pharmacokinetics, Bristol-Myers Squibb Research and Development, , Princeton, New Jersey 08540, United States
| | - Manthena V. S. Varma
- Department
of Pharmacokinetics, Dynamics, and Metabolism, Pfizer Worldwide Research and Development, Groton, Connecticut 06340, United States
| |
Collapse
|
24
|
Barber JA, Stahl SH, Summers C, Barrett G, Park BK, Foster JR, Kenna JG. Quantification of Drug-Induced Inhibition of Canalicular Cholyl-l-Lysyl-Fluorescein Excretion From Hepatocytes by High Content Cell Imaging. Toxicol Sci 2015. [PMID: 26220638 DOI: 10.1093/toxsci/kfv159] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
We describe the use of a commercially available high content cell imaging algorithm (Cellomics Arrayscan Spot Detector) to quantify biliary excretion of the fluorescent probe substrate cholyl-l-lysyl-fluorescein (CLF) from rat hepatocytes cultured in collagen/matrigel sandwich configuration and to explore inhibition of this process by a variety of test compounds. The method provided robust, reproducible data. Twenty-nine pharmaceuticals inhibited biliary CLF efflux from hepatocytes and a broad range of potencies of inhibition were observed (IC50 values ranged between <1 and 794 µM). Thirteen drugs that inhibited CLF efflux also inhibited hepatocellular uptake of the probe substrate [(3)H]-taurocholate. Although no clear correlation between the potencies of inhibition of the 2 processes was evident, these data highlight the need to consider possible uptake transporter inhibition when interpreting hepatocyte CLF inhibition data. It has been reported that CLF is transported by MRP2. The CLF efflux inhibition data correlated closely with published data on inhibition by the drugs of the bile salt export pump (Bsep), which suggests that the tested drugs inhibit both Bsep and Mrp2. Calculation of the ratios between the maximum human plasma concentrations of the drugs and their CLF efflux inhibition IC50 values raised the possibility that for many, but not all, of them the in vitro effects may be functionally significant in vivo and that Mrp2 inhibition might be a drug-induced liver injury (DILI) risk factor. These data indicate that imaging hepatocyte CLF inhibition is a promising new method for quantification of biliary efflux inhibition by drugs, which could aid assessment of compound-related DILI risk.
Collapse
Affiliation(s)
- Jane A Barber
- *Innovative Medicines & Early Development, Discovery Safety, Drug Safety & Metabolism, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield SK10 4TG, UK; Department of Pharmacology & Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, Merseyside, UK;
| | | | - Claire Summers
- *Innovative Medicines & Early Development, Discovery Safety, Drug Safety & Metabolism, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield SK10 4TG, UK
| | - Gillian Barrett
- Oncology iMEDs, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield, UK
| | - B Kevin Park
- Department of Pharmacology & Therapeutics, University of Liverpool, Sherrington Building, Ashton Street, Liverpool, Merseyside, UK
| | - John R Foster
- *Innovative Medicines & Early Development, Discovery Safety, Drug Safety & Metabolism, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield SK10 4TG, UK
| | - J Gerald Kenna
- *Innovative Medicines & Early Development, Discovery Safety, Drug Safety & Metabolism, AstraZeneca Pharmaceuticals, Alderley Park, Macclesfield SK10 4TG, UK
| |
Collapse
|
25
|
Li AP. Evaluation of Adverse Drug Properties with Cryopreserved Human Hepatocytes and the Integrated Discrete Multiple Organ Co-culture (IdMOC(TM)) System. Toxicol Res 2015; 31:137-49. [PMID: 26191380 PMCID: PMC4505344 DOI: 10.5487/tr.2015.31.2.137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2015] [Revised: 03/23/2015] [Accepted: 04/02/2015] [Indexed: 12/26/2022] Open
Abstract
Human hepatocytes, with complete hepatic metabolizing enzymes, transporters and cofactors, represent the gold standard for in vitro evaluation of drug metabolism, drug-drug interactions, and hepatotoxicity. Successful cryopreservation of human hepatocytes enables this experimental system to be used routinely. The use of human hepatocytes to evaluate two major adverse drug properties: drug-drug interactions and hepatotoxicity, are summarized in this review. The application of human hepatocytes in metabolism-based drug-drug interaction includes metabolite profiling, pathway identification, P450 inhibition, P450 induction, and uptake and efflux transporter inhibition. The application of human hepatocytes in toxicity evaluation includes in vitro hepatotoxicity and metabolism-based drug toxicity determination. A novel system, the Integrated Discrete Multiple Organ Co-culture (IdMOC) which allows the evaluation of nonhepatic toxicity in the presence of hepatic metabolism, is described.
Collapse
Affiliation(s)
- Albert P Li
- In Vitro ADMET Laboratories LLC, 9221 Rumsey Road Suite 8, Columbia, MD 21045
| |
Collapse
|
26
|
Heikkinen AT, Lignet F, Cutler P, Parrott N. The role of quantitative ADME proteomics to support construction of physiologically based pharmacokinetic models for use in small molecule drug development. Proteomics Clin Appl 2015; 9:732-44. [DOI: 10.1002/prca.201400147] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Revised: 01/16/2015] [Accepted: 02/05/2015] [Indexed: 01/26/2023]
Affiliation(s)
- Aki T. Heikkinen
- School of Pharmacy; Faculty of Health Sciences; University of Eastern Finland; Kuopio Finland
| | - Floriane Lignet
- Pharmaceutical Sciences; Pharmaceutical Research & Early Development; Roche Innovation Center Basel; Basel Switzerland
| | - Paul Cutler
- Pharmaceutical Sciences; Pharmaceutical Research & Early Development; Roche Innovation Center Basel; Basel Switzerland
| | - Neil Parrott
- Pharmaceutical Sciences; Pharmaceutical Research & Early Development; Roche Innovation Center Basel; Basel Switzerland
| |
Collapse
|
27
|
Wang L, Prasad B, Salphati L, Chu X, Gupta A, Hop CECA, Evers R, Unadkat JD. Interspecies variability in expression of hepatobiliary transporters across human, dog, monkey, and rat as determined by quantitative proteomics. Drug Metab Dispos 2015; 43:367-74. [PMID: 25534768 DOI: 10.1124/dmd.114.061580] [Citation(s) in RCA: 143] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
We quantified, by liquid chromatography tandem mass spectrometry, transporter protein expression of BSEP, MATE1, MRP3, MRP4, NTCP, and OCT1 in our human liver bank (n = 55) and determined the relationship between protein expression and sex, age and genotype. These data complement our previous work in the same liver bank where we quantified the protein expression of OATPs, BCRP, MDR1, and MRP2. In addition, we quantified and compared the interspecies differences in expression of the hepatobiliary transporters, corresponding to the above human transporters, in liver tissue and hepatocytes of male beagle dogs, cynomolgus monkeys, Sprague-Dawley rats, and Wistar rats. In all the species, the sinusoidal OATPs/Oatps were the most abundant hepatic transporters. However, there were notable interspecies differences in the relative abundance of the remaining transporters. For example, the next most abundant transporter in humans and monkeys was OCT1/Oct1, whereas it was Mrp2 and Ntcp in dogs/Wistar rats and Sprague-Dawley rats, respectively. In contrast, the protein expression of the efflux transporters BCRP/Bcrp, MDR1/Mdr1, MRP3/Mrp3, MRP4/Mrp4, and MATE1/Mate1 was much lower across all the species. For most transporters, the expression in the liver tissues was comparable to that in the unplated cryopreserved hepatocytes. These data on human liver transporter protein expression complete the picture of the expression of major human hepatobiliary transporters important in drug disposition and toxicity. In addition, the data on expression of the corresponding hepatobiliary transporters in preclinical species will be helpful in interpreting and extrapolating pharmacokinetic, pharmacological, and toxicological results from preclinical studies to humans.
Collapse
Affiliation(s)
- Li Wang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Laurent Salphati
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Xiaoyan Chu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Anshul Gupta
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Cornelis E C A Hop
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Raymond Evers
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (L.W., B.P., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, South San Francisco, California (L.S., C.E.C.A.H.); Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Rahway, New Jersey (X.C.); Drug Metabolism and Pharmacokinetics, Infection DMPK, AstraZeneca Pharmaceuticals LLP, Waltham, Massachusetts (A.G.); and Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co., Kenilworth, New Jersey (R.E.)
| |
Collapse
|
28
|
Stieger B, Unadkat JD, Prasad B, Langer O, Gali H. Role of (drug) transporters in imaging in health and disease. Drug Metab Dispos 2014; 42:2007-15. [PMID: 25249691 PMCID: PMC5611776 DOI: 10.1124/dmd.114.059873] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2014] [Accepted: 09/23/2014] [Indexed: 12/16/2022] Open
Abstract
This report is the summary of presentations at the symposium sponsored by the American Society for Pharmacology and Experimental Therapeutics, April 26-30, at Experimental Biology 2014 in San Diego, CA. The presentations focused on the role of transporters in imaging in health and disease and on assessing transporter function in vivo. Imaging is an important diagnostic tool in clinics and is a novel tool for in vivo visualization of transporter function. Many imaging substrates and endogenous markers for organ function are organic anions. In this symposium, the bile salt transporter sodium taurocholate cotransporting polypeptide and the liver organic anion transporting polypeptides (OATPs) as well as the renal organic anion transporters (OATs) were addressed in detail; e.g., OATPs mediate transport of contrast agents used for magnetic resonance imaging of the liver or transport agents used for hepatobiliary scintigraphy, and OATs transport substances used in renography. In addition, the symposium also focused on the multidrug-resistance transporter 1 (MDR1 or P-gp), which is the most important gatekeeper in epithelial or endothelial barriers for preventing entry of potentially harmful substances into organs. Novel substrates suitable for positron emission tomography (PET) allow the study of such transporters at the blood-brain barrier or while they are mediating uptake of drugs into hepatocytes, and, importantly, PET tracers also now allow renography. Finally, quantitative data on transporter expression in human organs allow the development of improved physiologically based pharmacokinetic (PBPK) models for drug disposition. Hence, the combined efforts using novel substrates for in vivo visualization of transporters and quantification of transporters will lead to a deeper understanding of transporter function in disease and allow development of novel PBPK models for disease states.
Collapse
Affiliation(s)
- Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland (B.S.); Department of Pharmaceutics, University of Washington, Seattle, Washington (J.D.U., B.P.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria and Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (O.L.); and the Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (H.G.)
| | - Jashvant D Unadkat
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland (B.S.); Department of Pharmaceutics, University of Washington, Seattle, Washington (J.D.U., B.P.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria and Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (O.L.); and the Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (H.G.)
| | - Bhagwat Prasad
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland (B.S.); Department of Pharmaceutics, University of Washington, Seattle, Washington (J.D.U., B.P.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria and Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (O.L.); and the Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (H.G.)
| | - Oliver Langer
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland (B.S.); Department of Pharmaceutics, University of Washington, Seattle, Washington (J.D.U., B.P.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria and Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (O.L.); and the Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (H.G.)
| | - Hariprasad Gali
- Department of Clinical Pharmacology and Toxicology, University Hospital, Zurich, Switzerland (B.S.); Department of Pharmaceutics, University of Washington, Seattle, Washington (J.D.U., B.P.); Department of Clinical Pharmacology, Medical University of Vienna, Vienna, Austria and Health and Environment Department, AIT Austrian Institute of Technology GmbH, Seibersdorf, Austria (O.L.); and the Department of Pharmaceutical Sciences, College of Pharmacy, The University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma (H.G.)
| |
Collapse
|
29
|
Harwood MD, Russell MR, Neuhoff S, Warhurst G, Rostami-Hodjegan A. Lost in centrifugation: accounting for transporter protein losses in quantitative targeted absolute proteomics. Drug Metab Dispos 2014; 42:1766-72. [PMID: 25061159 DOI: 10.1124/dmd.114.058446] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/13/2025] Open
Abstract
In drug development, considerable efforts are made to extrapolate from in vitro and preclinical findings to predict human drug disposition by using in vitro-in vivo extrapolation (IVIVE) approaches. Use of IVIVE strategies linked with physiologically based pharmacokinetic (PBPK) modeling is widespread, and regulatory agencies are accepting and occasionally requesting model analysis to support licensing submissions. Recently, there has been a drive to improve PBPK models by characterizing the absolute abundance of enzymes, transporters, and receptors within mammalian tissues and in vitro experimental systems using quantitative targeted absolute proteomics (QTAP). The absolute abundance of proteins relevant to processes governing drug disposition provided by QTAP will enable IVIVE-PBPK to incorporate terms for the abundance of enzymes and transporters in target populations. However, most studies that report absolute abundances of enzymes and transporter proteins do so in enriched membrane fractions so as to increase the abundance per sample, and thus the assay's sensitivity, rather than measuring the expected lower abundance in the more biologically meaningful whole cells or tissues. This communication discusses the balance between protein enrichment and potential loss during the preparation of membrane fractions from whole cells or tissues. Accounting for losses with recovery factors throughout the fractionation procedure provides a means to correct for procedural losses, thereby enabling the scaling of protein abundance from subcellular fractions to whole-cell or organ abundances. PBPK models based on corrected abundances will more closely resemble biological systems and facilitate development of more meaningful IVIVE scaling factors, producing more accurate quantitative predictions of drug disposition.
Collapse
Affiliation(s)
- Matthew D Harwood
- Gut Barrier Group, Inflammation & Repair, University of Manchester, Salford Royal NHS Trust, Salford (M.D.H., G.W.), Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, (M.D.H., S.N., A.R-H.), and Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, Manchester (M.R.R., A.R-H.), United Kingdom
| | - Matthew R Russell
- Gut Barrier Group, Inflammation & Repair, University of Manchester, Salford Royal NHS Trust, Salford (M.D.H., G.W.), Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, (M.D.H., S.N., A.R-H.), and Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, Manchester (M.R.R., A.R-H.), United Kingdom
| | - Sibylle Neuhoff
- Gut Barrier Group, Inflammation & Repair, University of Manchester, Salford Royal NHS Trust, Salford (M.D.H., G.W.), Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, (M.D.H., S.N., A.R-H.), and Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, Manchester (M.R.R., A.R-H.), United Kingdom
| | - Geoffrey Warhurst
- Gut Barrier Group, Inflammation & Repair, University of Manchester, Salford Royal NHS Trust, Salford (M.D.H., G.W.), Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, (M.D.H., S.N., A.R-H.), and Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, Manchester (M.R.R., A.R-H.), United Kingdom
| | - Amin Rostami-Hodjegan
- Gut Barrier Group, Inflammation & Repair, University of Manchester, Salford Royal NHS Trust, Salford (M.D.H., G.W.), Simcyp Limited (a Certara Company), Blades Enterprise Centre, Sheffield, (M.D.H., S.N., A.R-H.), and Centre for Applied Pharmacokinetic Research, Manchester Pharmacy School, Manchester (M.R.R., A.R-H.), United Kingdom
| |
Collapse
|
30
|
Prasad B, Unadkat JD. Optimized approaches for quantification of drug transporters in tissues and cells by MRM proteomics. AAPS J 2014; 16:634-48. [PMID: 24752720 PMCID: PMC4070263 DOI: 10.1208/s12248-014-9602-y] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Accepted: 03/29/2014] [Indexed: 01/12/2023] Open
Abstract
Drug transporter expression in tissues (in vivo) usually differs from that in cell lines used to measure transporter activity (in vitro). Therefore, quantification of transporter expression in tissues and cell lines is important to develop scaling factor for in vitro to in vivo extrapolation (IVIVE) of transporter-mediated drug disposition. Since traditional immunoquantification methods are semiquantitative, targeted proteomics is now emerging as a superior method to quantify proteins, including membrane transporters. This superiority is derived from the selectivity, precision, accuracy, and speed of analysis by liquid chromatography tandem mass spectrometry (LC-MS/MS) in multiple reaction monitoring (MRM) mode. Moreover, LC-MS/MS proteomics has broader applicability because it does not require selective antibodies for individual proteins. There are a number of recent research and review papers that discuss the use of LC-MS/MS for transporter quantification. Here, we have compiled from the literature various elements of MRM proteomics to provide a comprehensive systematic strategy to quantify drug transporters. This review emphasizes practical aspects and challenges in surrogate peptide selection, peptide qualification, peptide synthesis and characterization, membrane protein isolation, protein digestion, sample preparation, LC-MS/MS parameter optimization, method validation, and sample analysis. In particular, bioinformatic tools used in method development and sample analysis are discussed in detail. Various pre-analytical and analytical sources of variability that should be considered during transporter quantification are highlighted. All these steps are illustrated using P-glycoprotein (P-gp) as a case example. Greater use of quantitative transporter proteomics will lead to a better understanding of the role of drug transporters in drug disposition.
Collapse
Affiliation(s)
- Bhagwat Prasad
- Department of Pharmaceutics, University of Washington, P.O. Box 357610, Seattle, Washington, 98195, USA,
| | | |
Collapse
|
31
|
Qiu X, Zhang H, Lai Y. Quantitative targeted proteomics for membrane transporter proteins: method and application. AAPS JOURNAL 2014; 16:714-26. [PMID: 24830943 DOI: 10.1208/s12248-014-9607-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2014] [Accepted: 04/05/2014] [Indexed: 01/04/2023]
Abstract
Although global proteomics has shown promise for discovery of many new proteins, biomarkers, protein modifications, and polymorphisms, targeted proteomics is emerging in the proteomics research field as a complement to untargeted shotgun proteomics, particularly when a determined set of low-abundance functional proteins need to be measured. The function and expression of proteins related to drug absorption, distribution, metabolism, and excretion (ADME) such as cytochrome P450 enzymes and membrane transporters are of great interest in biopharmaceutical research. Since the variation in ADME-related protein expression is known to be a major complicating factor encountered during in vitro-in vivo and in vivo-in vivo extrapolations (IVIVE), the accurate quantification of the ADME proteins in complex biological systems becomes a fundamental element in establishing IVIVE for pharmacokinetic predictions. In this review, we provide an overview of relevant methodologies followed by a summary of recent applications encompassing mass spectrometry-based targeted quantifications of membrane transporters.
Collapse
Affiliation(s)
- Xi Qiu
- Pharmaceutical Candidate Optimization, Bristol-Myers Squibb, Princeton, New Jersey, 08543, USA
| | | | | |
Collapse
|
32
|
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
|
33
|
Chatterjee S, Bijsmans IT, van Mil SW, Augustijns P, Annaert P. Toxicity and intracellular accumulation of bile acids in sandwich-cultured rat hepatocytes: Role of glycine conjugates. Toxicol In Vitro 2014; 28:218-30. [DOI: 10.1016/j.tiv.2013.10.020] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Revised: 10/24/2013] [Accepted: 10/30/2013] [Indexed: 02/06/2023]
|
34
|
Abstract
The accuracy of preclinical safety evaluation to predict human toxicity is hindered by species difference in drug metabolism and toxic mechanism between human and nonhuman animals. In vitro human-based experimental systems allowing the assessment of human-specific drug properties represent a logical and practical approach to provide human-specific information. An advantage of in vitro approaches is that they require only limited amounts of time and resources, and, most importantly, do not invoke harm to human patients. Human hepatocytes, with complete hepatic metabolizing enzymes, transporters and cofactors, represent a practical and useful experimental system to assess drug metabolism. The use of human hepatocytes to evaluate two major adverse drug properties, drug–drug interactions and hepatotoxicity, are reviewed. The application of human hepatocytes in metabolism-based drug–drug interactions includes metabolite profiling, pathway identification, CYP450 inhibition, CYP450 induction, and uptake and efflux transporter inhibition. The application of human hepatocytes in toxicity evaluation includes in vitro hepatotoxicity and metabolism-based drug toxicity determination. Correlation of drug toxicity with proteomics and genomics data may allow the discovery of clinical biomarkers for early detection of liver toxicity.
Collapse
Affiliation(s)
- Albert P Li
- In Vitro ADMET Laboratories LLC, 9221 Rumsey Road Suite 8, Columbia, MD 21045, USA
| |
Collapse
|
35
|
Pfeifer ND, Bridges AS, Ferslew BC, Hardwick RN, Brouwer KLR. Hepatic basolateral efflux contributes significantly to rosuvastatin disposition II: characterization of hepatic elimination by basolateral, biliary, and metabolic clearance pathways in rat isolated perfused liver. J Pharmacol Exp Ther 2013; 347:737-45. [PMID: 24080682 PMCID: PMC3836312 DOI: 10.1124/jpet.113.208314] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2013] [Accepted: 09/30/2013] [Indexed: 01/07/2023] Open
Abstract
Basolateral efflux clearance (CLBL) contributes significantly to rosuvastatin (RSV) elimination in sandwich-cultured hepatocytes (SCH). The contribution of CLBL to RSV hepatic elimination was determined in single-pass isolated perfused livers (IPLs) from wild-type (WT) and multidrug resistance-associated protein 2 (Mrp2)-deficient (TR(-)) rats in the absence and presence of the P-glycoprotein and breast cancer resistance protein (Bcrp) inhibitor, elacridar (GF120918); clearance values were compared with SCH. RSV biliary clearance (CLBile) was ablated almost completely by GF120918 in TR(-) IPLs, confirming that Mrp2 and Bcrp primarily are responsible for RSV CLBile. RSV appearance in outflow perfusate was attributed primarily to CLBL, which was impaired in TR(-) IPLs. CLBL was ≈ 6-fold greater than CLBile in the linear range in WT IPLs in the absence of GF120918. Recovery of unchanged RSV in liver tissue increased in TR(-) compared with WT (≈ 25 versus 6% of the administered dose) due to impaired CLBL and CLBile. RSV pentanoic acid, identified by high-resolution liquid chromatography-tandem mass spectroscopy, comprised ≈ 40% of total liver content and ≈ 16% of the administered dose in TR(-) livers at the end of perfusion, compared with ≈ 30 and 3% in WT livers, consistent with impaired RSV excretion and "shunting" to the metabolic pathway. In vitro-ex vivo extrapolation between WT SCH and IPLs (without GF120918) revealed that uptake clearance and CLBL were 4.2- and 6.4-fold lower, respectively, in rat SCH compared with IPLs; CLBile translated almost directly (1.1-fold). The present IPL data confirmed the significant role of CLBL in RSV hepatic elimination, and demonstrated that both CLBL and CLBile influence RSV hepatic and systemic exposure.
Collapse
Affiliation(s)
- Nathan D Pfeifer
- Division of Pharmacotherapy and Experimental Therapeutics, UNC Eshelman School of Pharmacy, Chapel Hill, North Carolina (N.D.P., B.C.F., K.L.R.B.); and Department of Pathology (A.S.B.) and Curriculum in Toxicology (R.N.H., K.L.R.B.), School of Medicine, UNC School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | | | | | | | | |
Collapse
|
36
|
Pfeifer ND, Harris KB, Yan GZ, Brouwer KLR. Determination of intracellular unbound concentrations and subcellular localization of drugs in rat sandwich-cultured hepatocytes compared with liver tissue. Drug Metab Dispos 2013; 41:1949-56. [PMID: 23990525 PMCID: PMC3807053 DOI: 10.1124/dmd.113.052134] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 08/29/2013] [Indexed: 12/14/2022] Open
Abstract
Prediction of clinical efficacy, toxicity, and drug-drug interactions may be improved by accounting for the intracellular unbound drug concentration (C(unbound)) in vitro and in vivo. Furthermore, subcellular drug distribution may aid in predicting efficacy, toxicity, and risk assessment. The present study was designed to quantify the intracellular C(unbound) and subcellular localization of drugs in rat sandwich-cultured hepatocytes (SCH) compared with rat isolated perfused liver (IPL) tissue. Probe drugs with distinct mechanisms of hepatocellular uptake and accumulation were selected for investigation. Following drug treatment, SCH and IPL tissues were homogenized and fractionated by differential centrifugation to enrich for subcellular compartments. Binding in crude lysate and cytosol was determined by equilibrium dialysis; the C(unbound) and intracellular-to-extracellular C(unbound) ratio (K(pu,u)) were used to describe accumulation of unbound drug. Total accumulation (K(pobserved)) in whole tissue was well predicted by the SCH model (within 2- to 3-fold) for the selected drugs. Ritonavir (K(pu,u) ∼1) was evenly distributed among cellular compartments, but highly bound, which explained the observed accumulation within liver tissue. Rosuvastatin was recovered primarily in the cytosolic fraction, but did not exhibit extensive binding, resulting in a K(pu,u) >1 in liver tissue and SCH, consistent with efficient hepatic uptake. Despite extensive binding and sequestration of furamidine within liver tissue, a significant portion of cellular accumulation was attributed to unbound drug (K(pu,u) >16), as expected for a charged, hepatically derived metabolite. Data demonstrate the utility of SCH to predict quantitatively total tissue accumulation and elucidate mechanisms of hepatocellular drug accumulation such as active uptake versus binding/sequestration.
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
| | | | | | | |
Collapse
|
37
|
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
|
38
|
Shingaki T, Takashima T, Ijuin R, Zhang X, Onoue T, Katayama Y, Okauchi T, Hayashinaka E, Cui Y, Wada Y, Suzuki M, Maeda K, Kusuhara H, Sugiyama Y, Watanabe Y. Evaluation of Oatp and Mrp2 activities in hepatobiliary excretion using newly developed positron emission tomography tracer [11C]dehydropravastatin in rats. J Pharmacol Exp Ther 2013; 347:193-202. [PMID: 23926287 DOI: 10.1124/jpet.113.206425] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2025] Open
Abstract
We developed a pravastatin derivative, sodium (3R,5R)-3,5-dihydroxy-7-((1S,2S,6S,8S)-6-hydroxy-2-methyl-8-((1-[(11)C]-(E)-2-methyl-but-2-enoyl)oxy)-1,2,6,7,8,8a-hexahydronaphthalen-1-yl)heptanoate ([(11)C]DPV), as a positron emission tomography (PET) probe for noninvasive measurement of hepatobiliary transport, and conducted pharmacokinetic analysis in rats as a feasibility study for future clinical study. Transport activities of DPV in freshly isolated rat hepatocytes and rodent multidrug resistance-associated protein 2 (rMrp2; human, MRP2)-expressing membrane vesicles were similar to those of pravastatin. Rifampicin diminished the uptake of DPV and pravastatin by the hepatocytes, with similar inhibition potency. [(11)C]DPV underwent biotransformation to produce at least two metabolites in rat, but metabolism of [(11)C]DPV occurred negligibly in human hepatocytes during a 90-minute incubation. After intravenous injection, [(11)C]DPV was mainly distributed to the liver and kidneys, where the tissue uptake clearances (CLuptake,liver and CLuptake,kidney) were blood-flow-limited (73.6 ± 4.8 and 24.6 ± 0.6 ml/min per kilogram, respectively). Systemic elimination of [(11)C]DPV was delayed in rifampicin-treated rat and an Mrp2-deficient mutant rat, Eisai hyperbilirubinemic mutant rat (EHBR). Rifampicin treatment decreased both CLuptake,liver and CLuptake,kidney of [(11)C]DPV by 30% (P < 0.05), whereas these parameters were unchanged in EHBR. Meanwhile, the canalicular efflux clearance (CLint,bile) of [(11)C]DPV, which was 12.2 ± 1.5 ml/min per kilogram in the control rat, decreased by 60% and 89% in rifampicin-treated rat and EHBR (P < 0.05), respectively. These results indicate that [(11)C]DPV is taken up into the liver by organic anion-transporting polypeptides (rodent, Oatps; human, OATP) and excreted into bile by Mrp2 in rat, and that rifampicin may inhibit Mrp2 as well as Oatps, and consequently increase systemic exposure of [(11)C]DPV. PET using [(11)C]DPV is feasible for studies prior to the future clinical investigation of OATP and MRP2 functionality, especially for personalized medicine.
Collapse
Affiliation(s)
- Tomotaka Shingaki
- RIKEN Center for Life Science Technologies (T.S., Y.K., T.Ok., E.H., Y.C., Y.Wad., Y.Wat.) and RIKEN Center for Molecular Imaging Sciences, Kobe, Hyogo, Japan (T.S., T.T., R.I., T.Ok., Y.K., E.H., Y.C., Y.Wad., M.S., Y.Wat.); Laboratory of Molecular Pharmacokinetics, Graduate School of Pharmaceutical Sciences, The University of Tokyo, Tokyo, Japan (X.Z., T.On., K.M., H.K.); and Sugiyama Laboratory, RIKEN Innovation Center, Yokohama Bio Industry Center, Yokohama, Kanagawa, Japan (Y.S.)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
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]
|
40
|
Godoy P, Hewitt NJ, Albrecht U, Andersen ME, Ansari N, Bhattacharya S, Bode JG, Bolleyn J, Borner C, Böttger J, Braeuning A, Budinsky RA, Burkhardt B, Cameron NR, Camussi G, Cho CS, Choi YJ, Craig Rowlands J, Dahmen U, Damm G, Dirsch O, Donato MT, Dong J, Dooley S, Drasdo D, Eakins R, Ferreira KS, Fonsato V, Fraczek J, Gebhardt R, Gibson A, Glanemann M, Goldring CEP, Gómez-Lechón MJ, Groothuis GMM, Gustavsson L, Guyot C, Hallifax D, Hammad S, Hayward A, Häussinger D, Hellerbrand C, Hewitt P, Hoehme S, Holzhütter HG, Houston JB, Hrach J, Ito K, Jaeschke H, Keitel V, Kelm JM, Kevin Park B, Kordes C, Kullak-Ublick GA, LeCluyse EL, Lu P, Luebke-Wheeler J, Lutz A, Maltman DJ, Matz-Soja M, McMullen P, Merfort I, Messner S, Meyer C, Mwinyi J, Naisbitt DJ, Nussler AK, Olinga P, Pampaloni F, Pi J, Pluta L, Przyborski SA, Ramachandran A, Rogiers V, Rowe C, Schelcher C, Schmich K, Schwarz M, Singh B, Stelzer EHK, Stieger B, Stöber R, Sugiyama Y, Tetta C, Thasler WE, Vanhaecke T, Vinken M, Weiss TS, Widera A, Woods CG, Xu JJ, Yarborough KM, Hengstler JG. Recent advances in 2D and 3D in vitro systems using primary hepatocytes, alternative hepatocyte sources and non-parenchymal liver cells and their use in investigating mechanisms of hepatotoxicity, cell signaling and ADME. Arch Toxicol 2013; 87:1315-1530. [PMID: 23974980 PMCID: PMC3753504 DOI: 10.1007/s00204-013-1078-5] [Citation(s) in RCA: 965] [Impact Index Per Article: 80.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Accepted: 05/06/2013] [Indexed: 12/15/2022]
Abstract
This review encompasses the most important advances in liver functions and hepatotoxicity and analyzes which mechanisms can be studied in vitro. In a complex architecture of nested, zonated lobules, the liver consists of approximately 80 % hepatocytes and 20 % non-parenchymal cells, the latter being involved in a secondary phase that may dramatically aggravate the initial damage. Hepatotoxicity, as well as hepatic metabolism, is controlled by a set of nuclear receptors (including PXR, CAR, HNF-4α, FXR, LXR, SHP, VDR and PPAR) and signaling pathways. When isolating liver cells, some pathways are activated, e.g., the RAS/MEK/ERK pathway, whereas others are silenced (e.g. HNF-4α), resulting in up- and downregulation of hundreds of genes. An understanding of these changes is crucial for a correct interpretation of in vitro data. The possibilities and limitations of the most useful liver in vitro systems are summarized, including three-dimensional culture techniques, co-cultures with non-parenchymal cells, hepatospheres, precision cut liver slices and the isolated perfused liver. Also discussed is how closely hepatoma, stem cell and iPS cell-derived hepatocyte-like-cells resemble real hepatocytes. Finally, a summary is given of the state of the art of liver in vitro and mathematical modeling systems that are currently used in the pharmaceutical industry with an emphasis on drug metabolism, prediction of clearance, drug interaction, transporter studies and hepatotoxicity. One key message is that despite our enthusiasm for in vitro systems, we must never lose sight of the in vivo situation. Although hepatocytes have been isolated for decades, the hunt for relevant alternative systems has only just begun.
Collapse
Affiliation(s)
- Patricio Godoy
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | | | - Ute Albrecht
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Nariman Ansari
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Sudin Bhattacharya
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Johannes Georg Bode
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Jennifer Bolleyn
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Christoph Borner
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
| | - Jan Böttger
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Albert Braeuning
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Robert A. Budinsky
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Britta Burkhardt
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Neil R. Cameron
- Department of Chemistry, Durham University, Durham, DH1 3LE UK
| | - Giovanni Camussi
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Chong-Su Cho
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Yun-Jaie Choi
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - J. Craig Rowlands
- Toxicology and Environmental Research and Consulting, The Dow Chemical Company, Midland, MI USA
| | - Uta Dahmen
- Experimental Transplantation Surgery, Department of General Visceral, and Vascular Surgery, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - Georg Damm
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Olaf Dirsch
- Institute of Pathology, Friedrich-Schiller-University Jena, 07745 Jena, Germany
| | - María Teresa Donato
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
| | - Jian Dong
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Steven Dooley
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Dirk Drasdo
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
- INRIA (French National Institute for Research in Computer Science and Control), Domaine de Voluceau-Rocquencourt, B.P. 105, 78153 Le Chesnay Cedex, France
- UPMC University of Paris 06, CNRS UMR 7598, Laboratoire Jacques-Louis Lions, 4, pl. Jussieu, 75252 Paris cedex 05, France
| | - Rowena Eakins
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Karine Sá Ferreira
- Institute of Molecular Medicine and Cell Research, University of Freiburg, Freiburg, Germany
- GRK 1104 From Cells to Organs, Molecular Mechanisms of Organogenesis, Faculty of Biology, University of Freiburg, Freiburg, Germany
| | - Valentina Fonsato
- Department of Medical Sciences, University of Torino, 10126 Turin, Italy
| | - Joanna Fraczek
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Rolf Gebhardt
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Andrew Gibson
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Matthias Glanemann
- Department of General-, Visceral- and Transplantation Surgery, Charité University Medicine Berlin, 13353 Berlin, Germany
| | - Chris E. P. Goldring
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - María José Gómez-Lechón
- Unidad de Hepatología Experimental, IIS Hospital La Fe Avda Campanar 21, 46009 Valencia, Spain
- CIBERehd, Fondo de Investigaciones Sanitarias, Barcelona, Spain
| | - Geny M. M. Groothuis
- Department of Pharmacy, Pharmacokinetics Toxicology and Targeting, University of Groningen, A. Deusinglaan 1, 9713 AV Groningen, The Netherlands
| | - Lena Gustavsson
- Department of Laboratory Medicine (Malmö), Center for Molecular Pathology, Lund University, Jan Waldenströms gata 59, 205 02 Malmö, Sweden
| | - Christelle Guyot
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - David Hallifax
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | - Seddik Hammad
- Department of Forensic Medicine and Veterinary Toxicology, Faculty of Veterinary Medicine, South Valley University, Qena, Egypt
| | - Adam Hayward
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Dieter Häussinger
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Claus Hellerbrand
- Department of Medicine I, University Hospital Regensburg, 93053 Regensburg, Germany
| | | | - Stefan Hoehme
- Interdisciplinary Center for Bioinformatics (IZBI), University of Leipzig, 04107 Leipzig, Germany
| | - Hermann-Georg Holzhütter
- Institut für Biochemie Abteilung Mathematische Systembiochemie, Universitätsmedizin Berlin (Charité), Charitéplatz 1, 10117 Berlin, Germany
| | - J. Brian Houston
- Centre for Applied Pharmacokinetic Research (CAPKR), School of Pharmacy and Pharmaceutical Sciences, University of Manchester, Oxford Road, Manchester, M13 9PT UK
| | | | - Kiyomi Ito
- Research Institute of Pharmaceutical Sciences, Musashino University, 1-1-20 Shinmachi, Nishitokyo-shi, Tokyo, 202-8585 Japan
| | - Hartmut Jaeschke
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Verena Keitel
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | | | - B. Kevin Park
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Claus Kordes
- Clinic for Gastroenterology, Hepatology and Infectious Diseases, Heinrich-Heine-University, Moorenstrasse 5, 40225 Düsseldorf, Germany
| | - Gerd A. Kullak-Ublick
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Peng Lu
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | - Anna Lutz
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Daniel J. Maltman
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
| | - Madlen Matz-Soja
- Institute of Biochemistry, Faculty of Medicine, University of Leipzig, 04103 Leipzig, Germany
| | - Patrick McMullen
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Irmgard Merfort
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | | | - Christoph Meyer
- Department of Medicine II, Section Molecular Hepatology, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Jessica Mwinyi
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Dean J. Naisbitt
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Andreas K. Nussler
- BG Trauma Center, Siegfried Weller Institut, Eberhard Karls University Tübingen, 72076 Tübingen, Germany
| | - Peter Olinga
- Division of Pharmaceutical Technology and Biopharmacy, Department of Pharmacy, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Francesco Pampaloni
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Jingbo Pi
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Linda Pluta
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | - Stefan A. Przyborski
- Reinnervate Limited, NETPark Incubator, Thomas Wright Way, Sedgefield, TS21 3FD UK
- Biological and Biomedical Sciences, Durham University, Durham, DH13LE UK
| | - Anup Ramachandran
- Department of Pharmacology, Toxicology and Therapeutics, University of Kansas Medical Center, Kansas City, KS 66160 USA
| | - Vera Rogiers
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Cliff Rowe
- Department of Molecular and Clinical Pharmacology, Centre for Drug Safety Science, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Celine Schelcher
- Department of Surgery, Liver Regeneration, Core Facility, Human in Vitro Models of the Liver, Ludwig Maximilians University of Munich, Munich, Germany
| | - Kathrin Schmich
- Department of Pharmaceutical Biology and Biotechnology, University of Freiburg, Freiburg, Germany
| | - Michael Schwarz
- Department of Toxicology, Institute of Experimental and Clinical Pharmacology and Toxicology, Wilhelmstr. 56, 72074 Tübingen, Germany
| | - Bijay Singh
- Department of Agricultural Biotechnology and Research Institute for Agriculture and Life Sciences, Seoul National University, Seoul, 151-921 Korea
| | - Ernst H. K. Stelzer
- Buchmann Institute for Molecular Life Sciences (BMLS), Goethe University Frankfurt, Max-von-Laue-Str. 15, 60438 Frankfurt am Main, Germany
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | - Regina Stöber
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Innovation Center, RIKEN, Yokohama Biopharmaceutical R&D Center, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045 Japan
| | - Ciro Tetta
- Fresenius Medical Care, Bad Homburg, Germany
| | - Wolfgang E. Thasler
- Department of Surgery, Ludwig-Maximilians-University of Munich Hospital Grosshadern, Munich, Germany
| | - Tamara Vanhaecke
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Mathieu Vinken
- Department of Toxicology, Centre for Pharmaceutical Research, Faculty of Medicine and Pharmacy, Vrije Universiteit Brussel, 1090 Brussels, Belgium
| | - Thomas S. Weiss
- Department of Pediatrics and Juvenile Medicine, University of Regensburg Hospital, Regensburg, Germany
| | - Agata Widera
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| | - Courtney G. Woods
- The Hamner Institutes for Health Sciences, Research Triangle Park, NC USA
| | | | | | - Jan G. Hengstler
- Leibniz Research Centre for Working Environment and Human Factors (IFADO), 44139 Dortmund, Germany
| |
Collapse
|
41
|
Comment on the article "physiologically based modeling of pravastatin transporter-mediated hepatobiliary disposition and drug-drug interactions". Pharm Res 2013; 30:1467-8. [PMID: 23504339 DOI: 10.1007/s11095-013-0988-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Accepted: 01/22/2013] [Indexed: 12/31/2022]
|
42
|
De Bruyn T, Chatterjee S, Fattah S, Keemink J, Nicolaï J, Augustijns P, Annaert P. Sandwich-cultured hepatocytes: utility for in vitro exploration of hepatobiliary drug disposition and drug-induced hepatotoxicity. Expert Opin Drug Metab Toxicol 2013; 9:589-616. [PMID: 23452081 DOI: 10.1517/17425255.2013.773973] [Citation(s) in RCA: 98] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
INTRODUCTION The sandwich-cultured hepatocyte (SCH) model has become an invaluable in vitro tool for studying hepatic drug transport, metabolism, biliary excretion and toxicity. The relevant expression of many hepatocyte-specific functions together with the in vivo-like morphology favor SCHs over other preclinical models for evaluating hepatobiliary drug disposition and drug-induced hepatotoxicity. AREAS COVERED In this review, the authors highlight recommended procedures required for reproducibly culturing hepatocytes in sandwich configuration. It also provides an overview of the SCH model characteristics as a function of culture time. Lastly, the article presents a summary of the most prominent applications of the SCH model, including hepatic drug clearance prediction, drug-drug interaction potential and drug-induced hepatotoxicity. EXPERT OPINION When human (cryopreserved) hepatocytes are used to establish sandwich cultures, the model appears particularly valuable to quantitatively investigate clinically relevant mechanisms related to in vivo hepatobiliary drug disposition and hepatotoxicity. Nonetheless, the SCH model would largely benefit from better insight into the fundamental cell signaling mechanisms that are critical for long-term in vitro maintenance of the hepatocytic phenotype. Studies systematically exploring improved cell culture conditions (e.g., co-cultures or extracellular matrix modifications), as well as in vitro work identifying key transcription factors involved in hepatocyte differentiation are currently emerging.
Collapse
Affiliation(s)
- Tom De Bruyn
- Drug Delivery and Disposition, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, O&N2, Herestraat 49-bus-921, 3000 Leuven, Belgium
| | | | | | | | | | | | | |
Collapse
|
43
|
Alveroğlu E, Sözeri H, Kurtan U, Şenel M, Baykal A. Magnetic and spectroscopic properties of Polyacrylamide-CoFe2O4 magnetic hydrogel. J Mol Struct 2013. [DOI: 10.1016/j.molstruc.2012.12.009] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
44
|
Noel G, Le Vee M, Moreau A, Stieger B, Parmentier Y, Fardel O. Functional expression and regulation of drug transporters in monolayer- and sandwich-cultured mouse hepatocytes. Eur J Pharm Sci 2013; 49:39-50. [PMID: 23396053 DOI: 10.1016/j.ejps.2013.01.013] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2012] [Revised: 01/12/2013] [Accepted: 01/13/2013] [Indexed: 01/13/2023]
Abstract
Primary hepatocyte cultures are now considered as convenient models for in vitro analyzing liver drug transport. However, if primary human and rat hepatocytes have been well-characterized with respect to drug transporter expression and regulation, much less is known for primary mouse hepatocytes. The present study was therefore designed to gain insights about this point. The profile of sinusoidal and canalicular drug transporter mRNA expression in short time (4h)-cultured mouse hepatocytes was found to be highly correlated with that of freshly isolated hepatocytes; by contrast, those of counterparts cultured for a longer time (until 4 days) either in monolayer configurations on plastic or collagen or in sandwich configuration with matrigel were profoundly altered: uptake drug transporters such as Oct1, Oatps and Oat2 were thus down-regulated, whereas most of efflux transporters such as Mdr1a/b, Mrp3, Mrp4 and Bcrp were induced. Moreover, short time-cultured hepatocytes exhibited the highest levels of sinusoidal influx transporter activities. Transporter-mediated drug secretion into canalicular networks was however only observed in sandwich-cultured hepatocytes. Mouse hepatocytes cultured either in monolayer or sandwich configurations were finally shown to exhibit up-regulation of referent transporters in response to exposure to prototypical activators of the drug sensing receptors pregnane X receptor, aryl hydrocarbon receptor or constitutive androstane receptor. Taken together, these data demonstrate the feasibility of using primary mouse hepatocytes for investigating potential interactions of xenobiotics with hepatic transporter activity or regulation, provided that adequate culture conditions are retained.
Collapse
Affiliation(s)
- Gregory Noel
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Marc Le Vee
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France
| | - Amélie Moreau
- Technologie Servier, 25-27 rue Eugène Vignat, 45000 Orléans, France
| | - Bruno Stieger
- Department of Clinical Pharmacology and Toxicology, University Hospital, 8091 Zurich, Switzerland
| | | | - Olivier Fardel
- Institut de Recherches en Santé, Environnement et Travail (IRSET), UMR INSERM U1085, Faculté de Pharmacie, 2 Avenue du Pr Léon Bernard, 35043 Rennes, France; Pôle Biologie, Centre Hospitalier Universitaire, 2 rue Henri Le Guilloux, 35033 Rennes, France.
| |
Collapse
|
45
|
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
|
46
|
Ukairo O, Kanchagar C, Moore A, Shi J, Gaffney J, Aoyama S, Rose K, Krzyzewski S, McGeehan J, Andersen ME, Khetani SR, LeCluyse EL. Long-Term Stability of Primary Rat Hepatocytes in Micropatterned Cocultures. J Biochem Mol Toxicol 2013; 27:204-12. [DOI: 10.1002/jbt.21469] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2012] [Revised: 11/17/2012] [Accepted: 11/19/2012] [Indexed: 11/12/2022]
Affiliation(s)
| | | | - Amanda Moore
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | - Julianne Shi
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | | | - Simon Aoyama
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | - Kelly Rose
- The Hamner Institutes for Health Sciences; Research Triangle Park; NC; 27709; USA
| | | | - Jack McGeehan
- Hepregen Corporation, Suite 1500; Medford; MA; 2155; USA
| | - Melvin E. Andersen
- The Hamner Institutes for Health Sciences; Research Triangle Park; NC; 27709; USA
| | - Salman R. Khetani
- Colorado State University; Mechanical and Biomedical Engineering; Fort Collins; CO; 80523-1374; USA
| | - Edward L. LeCluyse
- The Hamner Institutes for Health Sciences; Research Triangle Park; NC; 27709; USA
| |
Collapse
|
47
|
Balogh LM, Lai Y. Applications of Targeted Proteomics in ADME for IVIVE. TRANSPORTERS IN DRUG DEVELOPMENT 2013. [DOI: 10.1007/978-1-4614-8229-1_5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|
48
|
Mörk LM, Isaksson B, Boran N, Ericzon BG, Strom S, Fischler B, Ellis E. Comparison of culture media for bile Acid transport studies in primary human hepatocytes. J Clin Exp Hepatol 2012; 2:315-22. [PMID: 25755453 PMCID: PMC3940529 DOI: 10.1016/j.jceh.2012.08.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2012] [Accepted: 08/16/2012] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Primary human hepatocytes are a useful in vitro model system to examine hepatic biochemical pathways, liver disorders and/or pharmacotherapies. This system can also be used for transport studies to investigate uptake and excretion of bile acids. Proper modeling of hepatic function requires careful attention to media components, and culture substrates and conditions. OBJECTIVES To examine the effects of different culture media and conditions on bile acid transport in cultured human hepatocytes. METHODS AND RESULTS Hepatocytes cultured in Williams' medium E showed an increase in both uptake and excretion of taurocholate compared to cells cultured in Dulbecco's Modified Eagle Medium (DMEM). Supplementation of DMEM with glutathione or ascorbic acid did not compensate for the lower transport. The difference can be explained by lower mRNA expression of the transporter proteins sodium taurocholate cotransporting polypeptide (NTCP) and bile salt export pump (BSEP; ABCB11) when cultured in DMEM. Hepatocytes cultured in DMEM also display fewer and smaller bile canaliculi. Following extended time in culture supplementation of Williams' medium E with dexamethasone increased the expression of NTCP and BSEP. CONCLUSION Williams' medium E is superior to DMEM for transport studies in primary human hepatocytes. Supplementation with dexamethasone increase mRNA levels of NTCP and BSEP.
Collapse
Key Words
- AA, ascorbic acid
- BSEP
- BSEP, bile salt export pump
- CgamF, cholylglycylamido-fluorescein
- DMEM, Dulbecco's Modified Eagle Medium
- GSH, glutathione
- HBSS, Hank's Balanced Salt Solution
- MRP2, multidrug resistance protein 2
- NTCP
- NTCP, sodium taurocholate cotransporting polypeptide
- OATP, organic anion-transporting polypeptide
- WE, Williams' medium E
- bile acid transport
- dexamethasone
- primary human hepatocytes
Collapse
Affiliation(s)
- Lisa-Mari Mörk
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden,Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden,Department of Laboratory Medicine, Karolinska Institutet, Stockholm 14186, Sweden
| | - Bengt Isaksson
- Division of Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden
| | - Nicola Boran
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden
| | - Bo-Göran Ericzon
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden
| | - Stephen Strom
- Department of Pathology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Björn Fischler
- Division of Pediatrics, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden
| | - Ewa Ellis
- Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden,Address for correspondence: Ewa Ellis, Liver Cell Lab F67, Division of Transplantation Surgery, Department of Clinical Science, Intervention and Technology (CLINTEC), Karolinska Institutet, Stockholm 14186, Sweden. Tel.: +46 8 58580086; fax: +46 8 7743191.
| |
Collapse
|
49
|
Kimoto E, Yoshida K, Balogh LM, Bi YA, Maeda K, El-Kattan A, Sugiyama Y, Lai Y. Characterization of Organic Anion Transporting Polypeptide (OATP) Expression and Its Functional Contribution to the Uptake of Substrates in Human Hepatocytes. Mol Pharm 2012; 9:3535-42. [DOI: 10.1021/mp300379q] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Emi Kimoto
- Pfizer Global Research and Development,
Department of Pharmacokinetics, Dynamics, and Metabolism, Groton,
Connecticut 06340, United States
| | - Kenta Yoshida
- Department of Molecular Pharmacokinetics,
Graduate School of Pharmaceutical Sciences, the University of Tokyo,
Tokyo, Japan
| | - Larissa M. Balogh
- Pfizer Global Research and Development,
Department of Pharmacokinetics, Dynamics, and Metabolism, Groton,
Connecticut 06340, United States
| | - Yi-an Bi
- Pfizer Global Research and Development,
Department of Pharmacokinetics, Dynamics, and Metabolism, Groton,
Connecticut 06340, United States
| | - Kazuya Maeda
- Department of Molecular Pharmacokinetics,
Graduate School of Pharmaceutical Sciences, the University of Tokyo,
Tokyo, Japan
| | - Ayman El-Kattan
- Pfizer Global Research and Development,
Department of Pharmacokinetics, Dynamics, and Metabolism, Groton,
Connecticut 06340, United States
| | - Yuichi Sugiyama
- Sugiyama Laboratory,
RIKEN Innovation
Center, RIKEN Research Cluster for Innovation, Yokohama, Japan
| | - Yurong Lai
- Pfizer Global Research and Development,
Department of Pharmacokinetics, Dynamics, and Metabolism, Groton,
Connecticut 06340, United States
| |
Collapse
|
50
|
Ballard P, Brassil P, Bui KH, Dolgos H, Petersson C, Tunek A, Webborn PJH. The right compound in the right assay at the right time: an integrated discovery DMPK strategy. Drug Metab Rev 2012; 44:224-52. [DOI: 10.3109/03602532.2012.691099] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|