101
|
Pang KS, Han YR, Noh K, Lee PI, Rowland M. Hepatic clearance concepts and misconceptions: Why the well-stirred model is still used even though it is not physiologic reality? Biochem Pharmacol 2019; 169:113596. [PMID: 31398312 DOI: 10.1016/j.bcp.2019.07.025] [Citation(s) in RCA: 48] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 07/30/2019] [Indexed: 12/22/2022]
Abstract
The liver is the most important drug metabolizing organ, endowed with a plethora of metabolizing enzymes and transporters to facilitate drug entry and removal via metabolism and/or biliary excretion. For this reason, much focus surrounds the development of clearance concepts, which are based on normalizing the rate of removal to the input or arterial concentration. By so doing, some authors have recently claimed that it implies one specific model of hepatic elimination, namely, the widely used well-stirred or venous equilibration model (WSM). This commentary challenges this claim and aims to provide a comprehensive discussion of not only the WSM but other currently applied hepatic clearance models - the parallel tube model (PTM), the dispersion model (DM), the zonal liver model (ZLM), and the heterogeneous capillary transit time model of Goresky and co-workers (GM). The WSM, PTM, and DM differ in the patterns of internal blood flow, assuming bulk, plug, and dispersive flows, respectively, which render different degrees of mixing within the liver that are characterized by the magnitudes of the dispersion number (DN), resulting in different implications concerning the (unbound) substrate concentration in liver (CuH). Early models assumed perfusion rate-limited distribution, which have since been modified to include membrane-limited transport. The recent developments associated with the misconceptions and the sensitivity of the models are hereby addressed. Since the WSM has been and will likely remain widely used, the pros and cons of this model relative to physiological reality are further discussed.
Collapse
Affiliation(s)
- K Sandy Pang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada.
| | - Yi Rang Han
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Keumhan Noh
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Ping I Lee
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, Ontario, Canada
| | - Malcolm Rowland
- Centre for Applied Pharmacokinetic Research, University of Manchester, United Kingdom
| |
Collapse
|
102
|
Li AP, Amaral K, Ho MCD. A Novel In vitro Experimental System for the Evaluation of Enteric Drug Metabolism: Cofactor-Supplemented Permeabilized Cryopreserved Human Enterocytes (MetMax™ Cryopreserved Human Enterocytes). Drug Metab Lett 2019; 12:132-137. [PMID: 30124163 PMCID: PMC6350199 DOI: 10.2174/1872312812666180820142141] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Revised: 08/03/2018] [Accepted: 08/08/2018] [Indexed: 12/16/2022]
Abstract
Background: We report here an evaluation of a novel experimental system- cofactor-supplemented permeabilized cryopreserved human enterocytes (MetMax™ cryopreserved human entero-cytes (MMHE), patent pending) for applications in the evaluation of enteric drug metabolism. A major advantage of MMHE over Conventional Cryopreserved Human Enterocytes (CCHE) is the simplification of the use procedures including storage at -80°C instead of in liquid nitrogen, and use of the cells imme-diately after thawing without a need for centrifugation and microscopic evaluation of cell density and via-bility and cell density adjustment. Methods: In this study, we compared MMHE and CCHE in key phase 1 oxidation and phase 2 conjuga-tion Drug Metabolism Enzyme (DME) activities that we recently reported for cryopreserved human en-terocytes: CYP2C9 (diclofenac 4’- hydroxylation), CYP2C19 (s-mephenytoin hydroxylation), CYP3A4 (midazolam 1’-hydroxylation), CYP2J2 (astemizole O-demethylation), uridine 5'-diphospho-glucuronosyltransferase (UGT; 7-hydroxycoumarin glucuronidation), sulfotransferase (SULT; 7-hydroxycoumarin sulfation), N-acetyl transferase-1 (NAT-1; p-benzoic acid N-acetylation), and carboxy-esterase-2 (CES-2; hydrolysis of irinotecan to SN38). Both CCHE and MMHE were active in all the DME pathways evaluated, with specific activities of MMHE ranged from 142% (CYP2C9) to 1713% (UGT) of that for CCHE. β-hydroxylation and testosterone 6. Result and Conclusion: Our results suggest that the MMHE system represents a convenient and robust in vitro experimental system for the evaluation of enteric drug metabolism
Collapse
Affiliation(s)
- Albert P Li
- In vitro ADMET Laboratories Inc., 9221 Rumsey Road, Suite 8, Malden, MA 02148, USA and Boston Hepatocyte Technology Center, In vitro ADMET Laboratories, 389 Main St, Ste 301, Malden, MA 02148, United States
| | - Kirsten Amaral
- In vitro ADMET Laboratories Inc., 9221 Rumsey Road, Suite 8, Malden, MA 02148, USA and Boston Hepatocyte Technology Center, In vitro ADMET Laboratories, 389 Main St, Ste 301, Malden, MA 02148, United States
| | - Ming-Chih D Ho
- In vitro ADMET Laboratories Inc., 9221 Rumsey Road, Suite 8, Malden, MA 02148, USA and Boston Hepatocyte Technology Center, In vitro ADMET Laboratories, 389 Main St, Ste 301, Malden, MA 02148, United States
| |
Collapse
|
103
|
Henderson CJ, Kapelyukh Y, Scheer N, Rode A, McLaren AW, MacLeod AK, Lin D, Wright J, Stanley LA, Wolf CR. An Extensively Humanized Mouse Model to Predict Pathways of Drug Disposition and Drug/Drug Interactions, and to Facilitate Design of Clinical Trials. Drug Metab Dispos 2019; 47:601-615. [PMID: 30910785 PMCID: PMC6505380 DOI: 10.1124/dmd.119.086397] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2019] [Accepted: 03/04/2019] [Indexed: 02/06/2023] Open
Abstract
Species differences in drug metabolism and disposition can confound the extrapolation of in vivo PK data to man and also profoundly compromise drug efficacy studies owing to differences in pharmacokinetics, in metabolites produced (which are often pharmacologically active), and in differential activation of the transcription factors constitutive androstane receptor (CAR) and pregnane X receptor (PXR), which regulate the expression of such enzymes as P450s and drug transporters. These differences have gained additional importance as a consequence of the use of genetically modified mouse models for drug-efficacy testing and also patient-derived xenografts to predict individual patient responses to anticancer drugs. A number of humanized mouse models for cytochrome P450s, CAR, and PXR have been reported. However, the utility of these models has been compromised by the redundancy in P450 reactions across gene families, whereby the remaining murine P450s can metabolize the compounds being tested. To remove this confounding factor and create a mouse model that more closely reflects human pathways of drug disposition, we substituted 33 murine P450s from the major gene families involved in drug disposition, together with Car and Pxr, for human CAR, PXR, CYP1A1, CYP1A2, CYP2C9, CYP2D6, CYP3A4, and CYP3A7. We also created a mouse line in which 34 P450s were deleted from the mouse genome. Using model compounds and anticancer drugs, we demonstrated how these mouse lines can be applied to predict drug-drug interactions in patients and discuss here their potential application in the more informed design of clinical trials and the personalized treatment of cancer.
Collapse
Affiliation(s)
- C J Henderson
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - Y Kapelyukh
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - N Scheer
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A Rode
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A W McLaren
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - A K MacLeod
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - D Lin
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - J Wright
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - L A Stanley
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| | - C R Wolf
- Systems Medicine, School of Medicine, University of Dundee, Jacqui Wood Cancer Centre, Ninewells Hospital, Dundee, United Kingdom (C.J.H., Y.K., C.R.W., A.M., K.M., D.L.); Taconic Biosciences Inc., Rensselaer, New York (N.S., A.R.); Independent Consultant, Putley, Ledbury, Herts, United Kingdom (J.W.); and Independent Consultant, Linlithgow, West Lothian, United Kingdom (L.A.S.)
| |
Collapse
|
104
|
Bowman CM, Benet LZ. Interlaboratory Variability in Human Hepatocyte Intrinsic Clearance Values and Trends with Physicochemical Properties. Pharm Res 2019; 36:113. [PMID: 31152241 DOI: 10.1007/s11095-019-2645-0] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2019] [Accepted: 05/10/2019] [Indexed: 11/28/2022]
Abstract
PURPOSE To examine the interlaboratory variability in CLint values generated with human hepatocytes and determine trends in variability and clearance prediction accuracy using physicochemical and pharmacokinetic parameters. METHODS Data for 50 compounds from 14 papers were compiled with physicochemical and pharmacokinetic parameter values taken from various sources. RESULTS Coefficients of variation were as high as 99.8% for individual compounds and variation was not dependent on the number of prediction values included in the analysis. When examining median values, it appeared that compounds with a lower number of rotatable bonds had more variability. When examining prediction uniformity, those compounds with uniform in vivo underpredictions had higher CLint, in vivo values, while those with non-uniform predictions typically had lower CLint, in vivo values. Of the compounds with uniform predictions, only a small number were uniformly predicted accurately. Based on this limited dataset, less lipophilic, lower intrinsic clearance, and lower protein binding compounds yield more accurate clearance predictions. CONCLUSIONS Caution should be taken when compiling in vitro CLint values from different laboratories as variations in experimental procedures (such as extent of shaking during incubation) may yield different predictions for the same compound. The majority of compounds with uniform in vitro values had predictions that were inaccurate, emphasizing the need for a better mechanistic understanding of IVIVE. The non-uniform predictions, often with low turnover compounds, reaffirmed the experimental challenges for drugs in this clearance range. Separating new chemical entities by lipophilicity, intrinsic clearance, and protein binding may help instill more confidence in IVIVE predictions.
Collapse
Affiliation(s)
- Christine M Bowman
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California, 94143-0912, USA
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California, 94143-0912, USA.
| |
Collapse
|
105
|
Honda GS, Pearce RG, Pham LL, Setzer RW, Wetmore BA, Sipes NS, Gilbert J, Franz B, Thomas RS, Wambaugh JF. Using the concordance of in vitro and in vivo data to evaluate extrapolation assumptions. PLoS One 2019; 14:e0217564. [PMID: 31136631 PMCID: PMC6538186 DOI: 10.1371/journal.pone.0217564] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Accepted: 05/14/2019] [Indexed: 12/16/2022] Open
Abstract
Linking in vitro bioactivity and in vivo toxicity on a dose basis enables the use of high-throughput in vitro assays as an alternative to traditional animal studies. In this study, we evaluated assumptions in the use of a high-throughput, physiologically based toxicokinetic (PBTK) model to relate in vitro bioactivity and rat in vivo toxicity data. The fraction unbound in plasma (fup) and intrinsic hepatic clearance (Clint) were measured for rats (for 67 and 77 chemicals, respectively), combined with fup and Clint literature data for 97 chemicals, and incorporated in the PBTK model. Of these chemicals, 84 had corresponding in vitro ToxCast bioactivity data and in vivo toxicity data. For each possible comparison of in vitro and in vivo endpoint, the concordance between the in vivo and in vitro data was evaluated by a regression analysis. For a base set of assumptions, the PBTK results were more frequently better associated than either the results from a “random” model parameterization or direct comparison of the “untransformed” values of AC50 and dose (performed best in 51%, 28%, and 21% of cases, respectively). We also investigated several assumptions in the application of PBTK for IVIVE, including clearance and internal dose selection. One of the better assumptions sets–restrictive clearance and comparing free in vivo venous plasma concentration with free in vitro concentration–outperformed the random and untransformed results in 71% of the in vitro-in vivo endpoint comparisons. These results demonstrate that applying PBTK improves our ability to observe the association between in vitro bioactivity and in vivo toxicity data in general. This suggests that potency values from in vitro screening should be transformed using in vitro-in vivo extrapolation (IVIVE) to build potentially better machine learning and other statistical models for predicting in vivo toxicity in humans.
Collapse
Affiliation(s)
- Gregory S. Honda
- National Center for Computational Toxicology, U.S. EPA, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Robert G. Pearce
- National Center for Computational Toxicology, U.S. EPA, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - Ly L. Pham
- National Center for Computational Toxicology, U.S. EPA, Research Triangle Park, North Carolina, United States of America
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee, United States of America
| | - R. W. Setzer
- National Center for Computational Toxicology, U.S. EPA, Research Triangle Park, North Carolina, United States of America
| | - Barbara A. Wetmore
- National Exposure Research Laboratory, U.S. EPA, Research Triangle Park, North Carolina, United States of America
| | - Nisha S. Sipes
- Division of the National Toxicology Program, NIEHS, Research Triangle Park, North Carolina, United States of America
| | - Jon Gilbert
- Cyprotex, Watertown, MA, United States of America
| | - Briana Franz
- Cyprotex, Watertown, MA, United States of America
| | - Russell S. Thomas
- National Center for Computational Toxicology, U.S. EPA, Research Triangle Park, North Carolina, United States of America
| | - John F. Wambaugh
- National Center for Computational Toxicology, U.S. EPA, Research Triangle Park, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
106
|
Yahata M, Ishii Y, Nakagawa T, Watanabe T, Bando K. Species differences in metabolism of a new antiepileptic drug candidate, DSP-0565 [2-(2'-fluoro[1,1'-biphenyl]-2-yl)acetamide]. Biopharm Drug Dispos 2019; 40:165-175. [PMID: 30924154 DOI: 10.1002/bdd.2180] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Revised: 03/06/2019] [Accepted: 03/21/2019] [Indexed: 12/15/2022]
Abstract
The metabolism and pharmacokinetics of DSP-0565 [2-(2'-fluoro[1,1'-biphenyl]-2-yl)acetamide], an antiepileptic drug candidate, was investigated in rats, dogs, and humans. In human hepatocytes, [14 C]DSP-0565 was primarily metabolized via amide bond hydrolysis to (2'-fluoro[1,1'-biphenyl]-2-yl)acetic acid (M8), while in rat and dog hepatocytes, it was primarily metabolized via both hydrolysis to M8 and hydroxylation at the benzene ring or the benzyl site to oxidized metabolites. After single oral administration of [14 C]DSP-0565 to rats and dogs, the major radioactivity fraction was recovered in the urine (71-72% of dose) with a much smaller fraction recovered in feces (23-25% of dose). As primary metabolites in their excreta, M8, oxidized metabolites, and glucuronide of DSP-0565 were detected. The contribution of metabolic pathways was estimated from metabolite profiles in their excreta: the major metabolic pathway was oxidation (57-62%) and the next highest was the hydrolysis pathway (23-33%). These results suggest that there are marked species differences in the metabolic pathways of DSP-0565 between humans and animals. Finally, DSP-0565 human oral clearance (CL/F) was predicted using in vitro-in vivo extrapolation (IVIVE) with/without animal scaling factors (SF, in vivo intrinsic clearance/in vitro intrinsic clearance). The SF improved the underestimation of IVIVE (fold error = 0.22), but the prediction was overestimated (fold error = 2.4-3.3). In contrast, the use of SF for hydrolysis pathway was the most accurate for the prediction (fold error = 1.0-1.4). Our findings suggest that understanding of species differences in metabolic pathways between humans and animals is important for predicting human metabolic clearance when using animal SF.
Collapse
Affiliation(s)
- Masahiro Yahata
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan.,Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Yuji Ishii
- Laboratory of Molecular Life Sciences, Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka, Japan
| | - Tetsuya Nakagawa
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Takao Watanabe
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| | - Kiyoko Bando
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., Osaka, Japan
| |
Collapse
|
107
|
Liu SN, Lu JBL, Watson CJW, Lazarus P, Desta Z, Gufford BT. Mechanistic Assessment of Extrahepatic Contributions to Glucuronidation of Integrase Strand Transfer Inhibitors. Drug Metab Dispos 2019; 47:535-544. [PMID: 30804050 PMCID: PMC6474910 DOI: 10.1124/dmd.118.085035] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2018] [Accepted: 02/21/2019] [Indexed: 12/24/2022] Open
Abstract
Integrase strand transfer inhibitor (INSTI)-based regimens dominate initial human immunodeficiency virus treatment. Most INSTIs are metabolized predominantly via UDP-glucuronosyltransferases (UGTs). For drugs predominantly metabolized by UGTs, including INSTIs, in vitro data recovered from human liver microsomes (HLMs) alone often underpredict human oral clearance. While several factors may contribute, extrahepatic glucuronidation may contribute to this underprediction. Thus, we comprehensively characterized the kinetics for the glucuronidation of INSTIs (cabotegravir, dolutegravir, and raltegravir) using pooled human microsomal preparations from liver (HLMs), intestine (HIMs), and kidney (HKMs) tissues; human embryonic kidney 293 cells expressing individual UGTs; and recombinant UGTs. In vitro glucuronidation of cabotegravir (HLMs≈HKMs>>>HIMs), dolutegravir (HLMs>HIMs>>HKMs), and raltegravir (HLMs>HKMs>> HIMs) occurred in hepatic and extrahepatic tissues. The kinetic data from expression systems suggested the major enzymes in each tissue: hepatic UGT1A9 > UGT1A1 (dolutegravir and raltegravir) and UGT1A1 (cabotegravir), intestinal UGT1A3 > UGT1A8 > UGT1A1 (dolutegravir) and UGT1A8 > UGT1A1 (raltegravir), and renal UGT1A9 (dolutegravir and raltegravir). Enzymes catalyzing cabotegravir glucuronidation in the kidney and intestine could not be identified unequivocally. Using data from dolutegravir glucuronidation as a prototype, a "bottom-up" physiologically based pharmacokinetic model was developed in a stepwise approach and predicted dolutegravir oral clearance within 4.5-fold (hepatic data only), 2-fold (hepatic and intestinal data), and 32% (hepatic, intestinal, and renal data). These results suggest clinically meaningful glucuronidation of dolutegravir in tissues other than the liver. Incorporation of additional novel mechanistic and physiologic underpinnings of dolutegravir metabolism along with in silico approaches appears to be a powerful tool to accurately predict the clearance of dolutegravir from in vitro data.
Collapse
Affiliation(s)
- Stephanie N Liu
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana (S.N.L., J.B.L.L., Z.D., B.T.G.) and Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (C.J.W.W., P.L.)
| | - Jessica Bo Li Lu
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana (S.N.L., J.B.L.L., Z.D., B.T.G.) and Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (C.J.W.W., P.L.)
| | - Christy J W Watson
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana (S.N.L., J.B.L.L., Z.D., B.T.G.) and Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (C.J.W.W., P.L.)
| | - Philip Lazarus
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana (S.N.L., J.B.L.L., Z.D., B.T.G.) and Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (C.J.W.W., P.L.)
| | - Zeruesenay Desta
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana (S.N.L., J.B.L.L., Z.D., B.T.G.) and Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (C.J.W.W., P.L.)
| | - Brandon T Gufford
- Division of Clinical Pharmacology, Department of Medicine, School of Medicine, Indiana University, Indianapolis, Indiana (S.N.L., J.B.L.L., Z.D., B.T.G.) and Department of Pharmaceutical Sciences, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington (C.J.W.W., P.L.)
| |
Collapse
|
108
|
Schneider D, Oskamp A, Holschbach M, Neumaier B, Bauer A, Bier D. Relevance of In Vitro Metabolism Models to PET Radiotracer Development: Prediction of In Vivo Clearance in Rats from Microsomal Stability Data. Pharmaceuticals (Basel) 2019; 12:ph12020057. [PMID: 31013984 PMCID: PMC6631687 DOI: 10.3390/ph12020057] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2019] [Revised: 04/10/2019] [Accepted: 04/12/2019] [Indexed: 01/03/2023] Open
Abstract
The prediction of in vivo clearance from in vitro metabolism models such as liver microsomes is an established procedure in drug discovery. The potentials and limitations of this approach have been extensively evaluated in the pharmaceutical sector; however, this is not the case for the field of positron emission tomography (PET) radiotracer development. The application of PET radiotracers and classical drugs differs greatly with regard to the amount of substance administered. In typical PET imaging sessions, subnanomolar quantities of the radiotracer are injected, resulting in body concentrations that cannot be readily simulated in analytical assays. This raises concerns regarding the predictability of radiotracer clearance from in vitro data. We assessed the accuracy of clearance prediction for three prototypical PET radiotracers developed for imaging the A1 adenosine receptor (A1AR). Using the half-life (t1/2) approach and physiologically based scaling, in vivo clearance in the rat model was predicted from microsomal stability data. Actual clearance could be accurately predicted with an average fold error (AFE) of 0.78 and a root mean square error (RMSE) of 1.6. The observed slight underprediction (1.3-fold) is in accordance with the prediction accuracy reported for classical drugs. This result indicates that the prediction of radiotracer clearance is possible despite concentration differences of more than three orders of magnitude between in vitro and in vivo conditions. Consequently, in vitro metabolism models represent a valuable tool for PET radiotracer development.
Collapse
Affiliation(s)
- Daniela Schneider
- Institute of Neuroscience and Medicine-Molecular Organization of the Brain (INM-2), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Angela Oskamp
- Institute of Neuroscience and Medicine-Molecular Organization of the Brain (INM-2), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Marcus Holschbach
- Institute of Neuroscience and Medicine-Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Bernd Neumaier
- Institute of Neuroscience and Medicine-Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| | - Andreas Bauer
- Institute of Neuroscience and Medicine-Molecular Organization of the Brain (INM-2), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
- Neurological Department, Medical Faculty, Heinrich-Heine-University, Universitätsstraße 1, 40225 Düsseldorf, Germany.
| | - Dirk Bier
- Institute of Neuroscience and Medicine-Nuclear Chemistry (INM-5), Forschungszentrum Jülich GmbH, 52428 Jülich, Germany.
| |
Collapse
|
109
|
Bowman CM, Okochi H, Benet LZ. The Presence of a Transporter-Induced Protein Binding Shift: A New Explanation for Protein-Facilitated Uptake and Improvement for In Vitro-In Vivo Extrapolation. Drug Metab Dispos 2019; 47:358-363. [PMID: 30674616 PMCID: PMC6413769 DOI: 10.1124/dmd.118.085779] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2018] [Accepted: 01/18/2019] [Indexed: 12/18/2022] Open
Abstract
Accurately predicting hepatic clearance is an integral part of the drug-development process, and yet current in vitro to in vivo (IVIVE) extrapolation methods yield poor predictions, particularly for highly protein-bound transporter substrates. Explanations for error include inaccuracies in protein-binding measurements and the lack of recognition of protein-facilitated uptake, where both unbound and bound drug may be cleared, violating the principles of the widely accepted free drug theory. A new explanation for protein-facilitated uptake is proposed here, called a transporter-induced protein binding shift High-affinity binding to cell-membrane proteins may change the equilibrium of the nonspecific binding between drugs and plasma proteins, leading to greater cellular uptake and clearance than currently predicted. The uptake of two lower protein-binding organic anion transporting polypeptide substrates (pravastatin and rosuvastatin) and two higher binding substrates (atorvastatin and pitavastatin) were measured in rat hepatocytes in incubations with protein-free buffer versus 100% plasma. Decreased unbound K m values and increased intrinsic clearance values were seen in the plasma incubations for the highly bound compounds, supporting the new hypothesis and mitigating the IVIVE underprediction previously seen for highly bound transporter substrates.
Collapse
Affiliation(s)
- Christine M Bowman
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| | - Hideaki Okochi
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California
| |
Collapse
|
110
|
Hallifax D, Houston JB. Use of Segregated Hepatocyte Scaling Factors and Cross-Species Relationships to Resolve Clearance Dependence in the Prediction of Human Hepatic Clearance. Drug Metab Dispos 2019; 47:320-327. [PMID: 30610004 DOI: 10.1124/dmd.118.085191] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 12/20/2018] [Indexed: 02/13/2025] Open
Abstract
Human and rat hepatocytes have a strong tendency to underpredict hepatic intrinsic clearance (CLint) and the extent of underprediction increases with increasing observed CLint In this study, application of the log average rat hepatocyte-rat in vivo empirical scaling factor (ESF) of 4.2 to human hepatocyte prediction successfully removed bias but did not improve precision. An analogous method using individual drug rat ESFs only achieved marginal improvement in accuracy but not precision. A novel approach to resolve clearance-dependent prediction, involving rat ESFs calculated for particular (order of magnitude) ranges of observed CLint (log average range, 0.12-2.1) improved human prediction precision but only modestly reduced bias. However, rat in vivo CLint was several-fold greater than human in vivo CLint and this was reflected in greater rat hepatocyte and microsome CLint, suggesting that rat metabolic enzymes are more efficient than their human counterparts, by several-fold. By applying the segregated rat ESFs followed by the human/rat CLint ratio, which was consistent regardless of CLint (log average 3.5), both accuracy and precision were improved, providing both a means of mitigating clearance dependence and reaffirming the potential role of rat hepatocytes for prediction of human metabolic CLint These cross-species observations indicate that underprediction from human in vitro systems may be predominantly consequential of an intrinsic property of the in vitro system rather than individual drug properties.
Collapse
Affiliation(s)
- D Hallifax
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| | - J B Houston
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, Manchester Academic Health Science Centre, University of Manchester, Manchester, United Kingdom
| |
Collapse
|
111
|
Matsunaga N, Ufuk A, Morse BL, Bedwell DW, Bao J, Mohutsky MA, Hillgren KM, Hall SD, Houston JB, Galetin A. Hepatic Organic Anion Transporting Polypeptide-Mediated Clearance in the Beagle Dog: Assessing In Vitro-In Vivo Relationships and Applying Cross-Species Empirical Scaling Factors to Improve Prediction of Human Clearance. Drug Metab Dispos 2019; 47:215-226. [PMID: 30593544 DOI: 10.1124/dmd.118.084194] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Accepted: 12/17/2018] [Indexed: 02/06/2023] Open
Abstract
In the present study, the beagle dog was evaluated as a preclinical model to investigate organic anion transporting polypeptide (OATP)-mediated hepatic clearance. In vitro studies were performed with nine OATP substrates in three lots of plated male dog hepatocytes ± OATP inhibitor cocktail to determine total uptake clearance (CLuptake) and total and unbound cell-to-medium concentration ratio (Kpuu). In vivo intrinsic hepatic clearances (CLint,H) were determined following intravenous drug administration (0.1 mg/kg) in male beagle dogs. The in vitro parameters were compared with those previously reported in plated human, monkey, and rat hepatocytes; the ability of cross-species scaling factors to improve prediction of human in vivo clearance was assessed. CLuptake in dog hepatocytes ranged from 9.4 to 135 µl/min/106 cells for fexofenadine and telmisartan, respectively. Active process contributed >75% to CLuptake for 5/9 drugs. Rosuvastatin and valsartan showed Kpuu > 10, whereas cerivastatin, pitavastatin, repaglinide, and telmisartan had Kpuu < 5. The extent of hepatocellular binding in dog was consistent with other preclinical species and humans. The bias (2.73-fold) obtained from comparison of predicted versus in vivo dog CLint,H was applied as an average empirical scaling factor (ESFav) for in vitro-in vivo extrapolation of human CLint,H The ESFav based on dog reduced underprediction of human CLint,H for the same data set (geometric mean fold error = 2.1), highlighting its utility as a preclinical model to investigate OATP-mediated uptake. The ESFav from all preclinical species resulted in comparable improvement of human clearance prediction, in contrast to drug-specific empirical scalars, rationalized by species differences in expression and/or relative contribution of particular transporters to drug hepatic uptake.
Collapse
Affiliation(s)
- Norikazu Matsunaga
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Ayşe Ufuk
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Bridget L Morse
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - David W Bedwell
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Jingqi Bao
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Michael A Mohutsky
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Kathleen M Hillgren
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Stephen D Hall
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, University of Manchester, Manchester, United Kingdom (N.M., A.U., J.B.H., A.G.); Pharmacokinetic Research Laboratories, Ono Pharmaceutical Co., Ltd., Osaka, Japan (N.M.); and Lilly Research Laboratories, Eli Lilly and Company, Indianapolis, Indiana (B.L.M., D.W.B., J.B., M.A.M., K.M.H., S.D.H.)
| |
Collapse
|
112
|
Bowman CM, Benet LZ. In Vitro-In Vivo Extrapolation and Hepatic Clearance-Dependent Underprediction. J Pharm Sci 2019; 108:2500-2504. [PMID: 30817922 DOI: 10.1016/j.xphs.2019.02.009] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Revised: 02/08/2019] [Accepted: 02/14/2019] [Indexed: 12/16/2022]
Abstract
Accurately predicting the hepatic clearance of compounds using in vitro to in vivo extrapolation (IVIVE) is crucial within the pharmaceutical industry. However, several groups have recently highlighted the serious error in the process. Although empirical or regression-based scaling factors may be used to mitigate the common underprediction, they provide unsatisfying solutions because the reasoning behind the underlying error has yet to be determined. One previously noted trend was intrinsic clearance-dependent underprediction, highlighting the limitations of current in vitro systems. When applying these generated in vitro intrinsic clearance values during drug development and making first-in-human dose predictions for new chemical entities though, hepatic clearance is the parameter that must be estimated using a model of hepatic disposition, such as the well-stirred model. Here, we examine error across hepatic clearance ranges and find a similar hepatic clearance-dependent trend, with high clearance compounds not predicted to be so, demonstrating another gap in the field.
Collapse
Affiliation(s)
- Christine M Bowman
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94143
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, California 94143.
| |
Collapse
|
113
|
Nishimuta H, Watanabe T, Bando K. Quantitative Prediction of Human Hepatic Clearance for P450 and Non-P450 Substrates from In Vivo Monkey Pharmacokinetics Study and In Vitro Metabolic Stability Tests Using Hepatocytes. AAPS JOURNAL 2019; 21:20. [PMID: 30673906 DOI: 10.1208/s12248-019-0294-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Accepted: 01/02/2019] [Indexed: 01/01/2023]
Abstract
Accurate prediction of human pharmacokinetics for drugs remains challenging, especially for non-cytochrome P450 (P450) substrates. Hepatocytes might be suitable for predicting hepatic intrinsic clearance (CLint) of new chemical entities, because they can be applied to various compounds regardless of the metabolic enzymes. However, it was reported that hepatic CLint is underestimated in hepatocytes. The purpose of the present study was to confirm the predictability of human hepatic clearance for P450 and non-P450 substrates in hepatocytes and the utility of animal scaling factors for the prediction using hepatocytes. CLint values for 30 substrates of P450, UDP-glucuronosyltransferase, flavin-containing monooxygenase, esterases, reductases, and aldehyde oxidase in human microsomes, human S9 and human, rat, and monkey hepatocytes were estimated. Hepatocytes were incubated in serum of each species. Furthermore, CLint values in human hepatocytes were corrected with empirical, monkey, and rat scaling factors. CLint values in hepatocytes for most compounds were underestimated compared to observed values regardless of the metabolic enzyme, and the predictability was improved by using the scaling factors. The prediction using human hepatocytes corrected with monkey scaling factor showed the highest predictability for both P450 and non-P450 substrates among the predictions using liver microsomes, liver S9, and hepatocytes with or without scaling factors. CLint values by this method for 80% and 90% of all compounds were within 2- and 3-fold of observed values, respectively. This method is accurate and useful for estimating new chemical entities, with no need to care about cofactors, localization of metabolic enzymes, or protein binding in plasma and incubation mixture.
Collapse
Affiliation(s)
- Haruka Nishimuta
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan.
| | - Takao Watanabe
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| | - Kiyoko Bando
- Preclinical Research Unit, Sumitomo Dainippon Pharma Co., Ltd., 3-1-98 Kasugade-naka, Konohana-ku, Osaka, 554-0022, Japan
| |
Collapse
|
114
|
Gouliarmou V, Lostia AM, Coecke S, Bernasconi C, Bessems J, Dorne JL, Ferguson S, Testai E, Remy UG, Brian Houston J, Monshouwer M, Nong A, Pelkonen O, Morath S, Wetmore BA, Worth A, Zanelli U, Zorzoli MC, Whelan M. Establishing a systematic framework to characterise in vitro methods for human hepatic metabolic clearance. Toxicol In Vitro 2018; 53:233-244. [DOI: 10.1016/j.tiv.2018.08.004] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Revised: 07/17/2018] [Accepted: 08/08/2018] [Indexed: 12/26/2022]
|
115
|
Finding synergies for 3Rs – Toxicokinetics and read-across: Report from an EPAA partners' Forum. Regul Toxicol Pharmacol 2018; 99:5-21. [DOI: 10.1016/j.yrtph.2018.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 07/17/2018] [Accepted: 08/16/2018] [Indexed: 01/11/2023]
|
116
|
Li AP, Ho MCD, Amaral K, Loretz C. A Novel In Vitro Experimental System for the Evaluation of Drug Metabolism: Cofactor-Supplemented Permeabilized Cryopreserved Human Hepatocytes (MetMax Cryopreserved Human Hepatocytes). Drug Metab Dispos 2018; 46:1608-1616. [PMID: 29363498 DOI: 10.1124/dmd.117.079657] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2017] [Accepted: 01/22/2018] [Indexed: 12/14/2022] Open
Abstract
We report here a novel experimental system, cryopreserved MetMax human hepatocytes (MMHHs), for in vitro drug metabolism studies. MMHHs consist of cofactor-supplemented permeabilized cryopreserved human hepatocytes. The use procedures for MMHHs are significantly simplified from that for conventional cryopreserved human hepatocytes (CCHHs): 1) storage at -80°C instead of in liquid nitrogen and 2) usage directly after thawing without centrifugation and microscopic evaluation of cell density and viability and cell density adjustment. In this study, we compared MMHHs and CCHHs in CYP1A2, CYP2A6, CYP2B6, CYP2C8, CYP2C9, CYP2D6, CYP2E1, CYP3A4, CYP2J2, monoamine oxidase A, aldehyde oxidase, flavin-containing monooxygenase, UDP-glucuronyl transferase, SULT, N-acetyltransferase 1, and acetaminophen glutathione (GSH) conjugation activities based on liquid chromatography-tandem mass spectrometry quantification of substrate metabolism. MMHHs were prepared from CCHHs consisting of hepatocytes pooled from 10 individual donors. The drug metabolizing enzyme activities of both CCHHs and MMHHs were cell concentration and time dependent, with specific activities of MMHHs ranging from 27.2% (carboxylesterase 2) to 234.2% (acetaminophen GSH conjugation) of that for CCHHs. As observed in CCHHs, sequential oxidation and conjugation was observed in MMHHs for coumarin, 7-ethoxycoumarin, and acetaminophen. 7-Hydroxycoumarin conjugation results showed that metabolic pathways in MMHHs could be selected via the choice of cofactors, with glucuronidation but not sulfation observed in the presence of UDP-glucuronic acid and not 3-phosphoadenosine-5-phosphosulfate, and vice versa. Results with noncytotoxic and cytotoxic concentrations of acetaminophen showed that drug metabolism was compromised in CCHHs but not in MMHHs. Our results suggest that the MMHHs system represents a convenient and robust in vitro experimental system for the evaluation of drug metabolism.
Collapse
Affiliation(s)
- Albert P Li
- In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L., C.L.) and In Vitro ADMET Laboratories Inc., Malden, Massachusetts (M.-C.D.H., K.A.)
| | - Ming-Chih David Ho
- In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L., C.L.) and In Vitro ADMET Laboratories Inc., Malden, Massachusetts (M.-C.D.H., K.A.)
| | - Kirsten Amaral
- In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L., C.L.) and In Vitro ADMET Laboratories Inc., Malden, Massachusetts (M.-C.D.H., K.A.)
| | - Carol Loretz
- In Vitro ADMET Laboratories Inc., Columbia, Maryland (A.P.L., C.L.) and In Vitro ADMET Laboratories Inc., Malden, Massachusetts (M.-C.D.H., K.A.)
| |
Collapse
|
117
|
Kimoto E, Mathialagan S, Tylaska L, Niosi M, Lin J, Carlo AA, Tess DA, Varma MVS. Organic Anion Transporter 2-Mediated Hepatic Uptake Contributes to the Clearance of High-Permeability-Low-Molecular-Weight Acid and Zwitterion Drugs: Evaluation Using 25 Drugs. J Pharmacol Exp Ther 2018; 367:322-334. [PMID: 30135178 DOI: 10.1124/jpet.118.252049] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2018] [Accepted: 08/15/2018] [Indexed: 03/08/2025] Open
Abstract
High-permeability-low-molecular-weight acids/zwitterions [i.e., extended clearance classification system class 1A (ECCS 1A) drugs] are considered to be cleared by metabolism with a minimal role of membrane transporters in their hepatic clearance. However, a marked disconnect in the in vitro-in vivo (IVIV) translation of hepatic clearance is often noted for these drugs. Metabolic rates measured using human liver microsomes and primary hepatocytes tend to underpredict. Here, we evaluated the role of organic anion transporter 2 (OAT2)-mediated hepatic uptake in the clearance of ECCS 1A drugs. For a set of 25 ECCS 1A drugs, in vitro transport activity was assessed using transporter-transfected cells and primary human hepatocytes. All but two drugs showed substrate affinity to OAT2, whereas four (bromfenac, entacapone, fluorescein, and nateglinide) also showed OATP1B1 activity in transfected cells. Most of these drugs (21 of 25) showed active uptake by plated human hepatocytes, with rifamycin SV (pan-transporter inhibitor) reducing the uptake by about 25%-95%. Metabolic turnover was estimated for 19 drugs after a few showed no measurable substrate depletion in liver microsomal incubations. IVIV extrapolation using in vitro data was evaluated to project human hepatic clearance of OAT2-alone substrates considering 1) uptake transport only, 2) metabolism only, and 3) transporter-enzyme interplay (extended clearance model). The transporter-enzyme interplay approach achieved improved prediction accuracy (average fold error = 1.9 and bias = 0.93) compared with the other two approaches. In conclusion, this study provides functional evidence for the role of OAT2-mediated hepatic uptake in determining the pharmacokinetics of several clinically important ECCS 1A drugs.
Collapse
Affiliation(s)
- Emi Kimoto
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Sumathy Mathialagan
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Laurie Tylaska
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Mark Niosi
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Jian Lin
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Anthony A Carlo
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - David A Tess
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| | - Manthena V S Varma
- Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut
| |
Collapse
|
118
|
Benet LZ, Bowman CM, Liu S, Sodhi JK. The Extended Clearance Concept Following Oral and Intravenous Dosing: Theory and Critical Analyses. Pharm Res 2018; 35:242. [PMID: 30349948 PMCID: PMC6364828 DOI: 10.1007/s11095-018-2524-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2018] [Accepted: 10/15/2018] [Indexed: 12/14/2022]
Abstract
PURPOSE To derive the theoretical basis for the extended clearance model of organ elimination following both oral and IV dosing, and critically analyze the approaches previously taken. METHODS We derived from first principles the theoretical basis for the extended clearance concept of organ elimination following both oral and IV dosing and critically analyzed previous approaches. RESULTS We point out a number of critical characteristics that have either been misinterpreted or not clearly presented in previously published treatments. First, the extended clearance concept is derived based on the well-stirred model. It is not appropriate to use alternative models of hepatic clearance. In analyzing equations, clearance terms are all intrinsic clearances, not total drug clearances. Flow and protein binding parameters should reflect blood measurements, not plasma values. In calculating the AUCR-factor following oral dosing, the AUC terms do not include flow parameters. We propose that calculations of AUCR may be a more useful approach to evaluate drug-drug and pharmacogenomic interactions than evaluating rate-determining steps. Through analyses of cerivastatin and fluvastatin interactions with cyclosporine we emphasize the need to characterize volume of distribution changes resulting from transporter inhibition/induction that can affect rate constants in PBPK models. Finally, we note that for oral doses, prediction of systemic and intrahepatic drug-drug interactions do not require knowledge of fu,H or Kp,uu for substrates/victims. CONCLUSIONS The extended clearance concept is a powerful tool to evaluate drug-drug interactions, pharmacogenomic and disease state variance but evaluating the AUCR-factor may provide a more valuable approach than characterizing rate-determining steps.
Collapse
Affiliation(s)
- Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California, 94143-0912, USA.
| | - Christine M Bowman
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California, 94143-0912, USA
| | - Shufang Liu
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California, 94143-0912, USA
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California, 94143-0912, USA
| |
Collapse
|
119
|
Bowman CM, Benet LZ. An examination of protein binding and protein-facilitated uptake relating to in vitro-in vivo extrapolation. Eur J Pharm Sci 2018; 123:502-514. [PMID: 30098391 DOI: 10.1016/j.ejps.2018.08.008] [Citation(s) in RCA: 64] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2018] [Revised: 08/03/2018] [Accepted: 08/04/2018] [Indexed: 01/09/2023]
Abstract
As explained by the free drug theory, the unbound fraction of drug has long been thought to drive the efficacy of a molecule. Thus, the fraction unbound term, or fu, appears in equations for fundamental pharmacokinetic parameters such as clearance, and is used when attempting in vitro to in vivo extrapolation (IVIVE). In recent years though, it has been noted that IVIVE does not always yield accurate predictions, and that some highly protein bound ligands have more efficient uptake than can be explained by their unbound fractions. This review explores the evolution of fu terms included when implementing IVIVE, the concept of protein-facilitated uptake, and the mechanisms that have been proposed to account for facilitated uptake.
Collapse
Affiliation(s)
- C M Bowman
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - L Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA.
| |
Collapse
|
120
|
De Bruyn T, Ufuk A, Cantrill C, Kosa RE, Bi YA, Niosi M, Modi S, Rodrigues AD, Tremaine LM, Varma MVS, Galetin A, Houston JB. Predicting Human Clearance of Organic Anion Transporting Polypeptide Substrates Using Cynomolgus Monkey: In Vitro-In Vivo Scaling of Hepatic Uptake Clearance. Drug Metab Dispos 2018; 46:989-1000. [PMID: 29720472 DOI: 10.1124/dmd.118.081315] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Accepted: 04/26/2018] [Indexed: 02/13/2025] Open
Abstract
This work explores the utility of the cynomolgus monkey as a preclinical model to predict hepatic uptake clearance mediated by organic anion transporting polypeptide (OATP) transporters. Nine OATP substrates (rosuvastatin, pravastatin, repaglinide, fexofenadine, cerivastatin, telmisartan, pitavastatin, bosentan, and valsartan) were investigated in plated cynomolgus monkey and human hepatocytes. Total uptake clearance and passive diffusion were measured in vitro from initial rates in the absence and presence of the OATP inhibitor rifamycin SV , respectively. Total uptake clearance values in plated hepatocytes ranged over three orders of magnitude in both species, with a similar rank order and good agreement in the relative contribution of active transport to total uptake between cynomolgus monkey and human. In vivo hepatic clearance for these nine drugs was determined in cynomolgus monkey after intravenous dosing. Hepatic clearances showed a range similar to human parameters and good predictions from respective hepatocyte parameters (with 2.7- and 3.8-fold bias on average, respectively). The use of cross-species empirical scaling factors (determined from cynomolgus monkey data either as the data set average or individual drug values) improved prediction (less bias, better concordance) of human hepatic clearance from human hepatocyte data alone. In vitro intracellular binding in hepatocytes also correlated well between species. It is concluded that the minimal species differences observed for the current data set between cynomolgus monkey and human hepatocyte uptake, both in vitro and in vivo, support future use of this preclinical model to delineate drug hepatic uptake and enable prediction of human in vivo intrinsic hepatic clearance.
Collapse
Affiliation(s)
- Tom De Bruyn
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Ayşe Ufuk
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Carina Cantrill
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Rachel E Kosa
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Yi-An Bi
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Mark Niosi
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Sweta Modi
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - A David Rodrigues
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Larry M Tremaine
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Manthena V S Varma
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Medicine, Biology and Health Sciences, University of Manchester, Manchester, United Kingdom (T.D.B., A.U., C.C., A.G., J.B.H.) and Pharmacokinetics, Dynamics and Drug Metabolism (R.E.K., Y.B., M.N., A.D.R., L.M.T., M.V.S.V) and Research Formulations, Pharmaceutical Sciences, (S.M.) Medicines Design, Pfizer, Inc., Groton, Connecticut
| |
Collapse
|
121
|
Wambaugh JF, Hughes MF, Ring CL, MacMillan DK, Ford J, Fennell TR, Black SR, Snyder RW, Sipes NS, Wetmore BA, Westerhout J, Setzer RW, Pearce RG, Simmons JE, Thomas RS. Evaluating In Vitro-In Vivo Extrapolation of Toxicokinetics. Toxicol Sci 2018; 163:152-169. [PMID: 29385628 PMCID: PMC5920326 DOI: 10.1093/toxsci/kfy020] [Citation(s) in RCA: 103] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Prioritizing the risk posed by thousands of chemicals potentially present in the environment requires exposure, toxicity, and toxicokinetic (TK) data, which are often unavailable. Relatively high throughput, in vitro TK (HTTK) assays and in vitro-to-in vivo extrapolation (IVIVE) methods have been developed to predict TK, but most of the in vivo TK data available to benchmark these methods are from pharmaceuticals. Here we report on new, in vivo rat TK experiments for 26 non-pharmaceutical chemicals with environmental relevance. Both intravenous and oral dosing were used to calculate bioavailability. These chemicals, and an additional 19 chemicals (including some pharmaceuticals) from previously published in vivo rat studies, were systematically analyzed to estimate in vivo TK parameters (e.g., volume of distribution [Vd], elimination rate). For each of the chemicals, rat-specific HTTK data were available and key TK predictions were examined: oral bioavailability, clearance, Vd, and uncertainty. For the non-pharmaceutical chemicals, predictions for bioavailability were not effective. While no pharmaceutical was absorbed at less than 10%, the fraction bioavailable for non-pharmaceutical chemicals was as low as 0.3%. Total clearance was generally more under-estimated for nonpharmaceuticals and Vd methods calibrated to pharmaceuticals may not be appropriate for other chemicals. However, the steady-state, peak, and time-integrated plasma concentrations of nonpharmaceuticals were predicted with reasonable accuracy. The plasma concentration predictions improved when experimental measurements of bioavailability were incorporated. In summary, HTTK and IVIVE methods are adequately robust to be applied to high throughput in vitro toxicity screening data of environmentally relevant chemicals for prioritizing based on human health risks.
Collapse
Affiliation(s)
| | - Michael F Hughes
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Caroline L Ring
- National Center for Computational Toxicology
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37831
| | - Denise K MacMillan
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Jermaine Ford
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | | | - Sherry R Black
- RTI International, Research Triangle Park, North Carolina
| | | | - Nisha S Sipes
- National Institute of Environmental Health Sciences, Research Triangle Park, North Carolina 27717
| | - Barbara A Wetmore
- National Exposure Research Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | - Joost Westerhout
- The Netherlands Organisation for Applied Scientific Research (TNO), AJ Zeist 3700, The Netherlands
| | | | - Robert G Pearce
- National Center for Computational Toxicology
- Oak Ridge Institute for Science and Education, Oak Ridge, Tennessee 37831
| | - Jane Ellen Simmons
- National Health and Environmental Effects Research Laboratory, Office of Research and Development, United States Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | | |
Collapse
|
122
|
Harrison J, De Bruyn T, Darwich AS, Houston JB. Simultaneous Assessment In Vitro of Transporter and Metabolic Processes in Hepatic Drug Clearance: Use of a Media Loss Approach. Drug Metab Dispos 2018; 46:405-414. [PMID: 29439129 DOI: 10.1124/dmd.117.079590] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/29/2018] [Indexed: 12/23/2022] Open
Abstract
Hepatocyte drug depletion-time assays are well established for determination of metabolic clearance in vitro. The present study focuses on the refinement and evaluation of a "media loss" assay, an adaptation of the conventional depletion assay involving centrifugation of hepatocytes prior to sampling, allowing estimation of uptake in addition to metabolism. Using experimental procedures consistent with a high throughput, a selection of 12 compounds with a range of uptake and metabolism characteristics (atorvastatin, cerivastatin, clarithromycin, erythromycin, indinavir, pitavastatin, repaglinide, rosuvastatin, saquinavir, and valsartan, with two control compounds-midazolam and tolbutamide) were investigated in the presence and absence of the cytochrome P450 inhibitor 1-aminobenzotriazole and organic anion transporter protein inhibitor rifamycin SV in rat hepatocytes. Data were generated simultaneously for a given drug, and provided, through the use of a mechanistic cell model, clearance terms characterizing metabolism, active and passive uptake, together with intracellular binding and partitioning parameters. Results were largely consistent with the particular drug characteristics, with active uptake, passive diffusion, and metabolic clearances ranging between 0.4 and 777, 3 and 383, and 2 and 236 μl/min per milligram protein, respectively. The same experiments provided total and unbound drug cellular partition coefficients ranging between 3.8 and 254 and 2.3 and 8.3, respectively, and intracellular unbound fractions between 0.014 and 0.263. Following in vitro-in vivo extrapolation, the lowest prediction bias was noted using uptake clearance, compared with metabolic clearance or apparent clearance from the media loss assay alone. This approach allows rapid and comprehensive characterization of hepatocyte drug disposition valuable for prediction of hepatic processes in vivo.
Collapse
Affiliation(s)
- James Harrison
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom (J.H., T.D.B., A.S.D., J.B.H.) and Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (T.D.B.)
| | - Tom De Bruyn
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom (J.H., T.D.B., A.S.D., J.B.H.) and Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (T.D.B.)
| | - Adam S Darwich
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom (J.H., T.D.B., A.S.D., J.B.H.) and Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (T.D.B.)
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, United Kingdom (J.H., T.D.B., A.S.D., J.B.H.) and Department of Drug Metabolism and Pharmacokinetics, Genentech Inc., South San Francisco, California (T.D.B.)
| |
Collapse
|
123
|
Da-Silva F, Boulenc X, Vermet H, Compigne P, Gerbal-Chaloin S, Daujat-Chavanieu M, Klieber S, Poulin P. Improving Prediction of Metabolic Clearance Using Quantitative Extrapolation of Results Obtained From Human Hepatic Micropatterned Cocultures Model and by Considering the Impact of Albumin Binding. J Pharm Sci 2018. [PMID: 29524447 DOI: 10.1016/j.xphs.2018.03.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The objective was to compare, with the same data set, the predictive performance of 3 in vitro assays of hepatic clearance (CL), namely, micropatterned cocultures (also referring to HepatoPac®) and suspension as well as monolayer hepatocytes to define which assay is the most accurate. Furthermore, existing in vitro-to-in vivo extrapolation (IVIVE) methods were challenged to verify which method is the most predictive (i.e., direct scaling method without binding correction, conventional method based either on the unbound fraction in plasma (fup) according to the free-drug hypothesis, or based on an fup value adjusted for the albumin [ALB]-facilitated hepatic uptake phenomenon). Accordingly, the role of ALB binding was specifically challenged, and consequently, the ALB production was monitored in parallel to the metabolic stability. The ALB concentration data were used to compare the in vitro assays and to adjust the value of fup of each drug to mimic the ALB-facilitated hepatic uptake phenomenon. The results confirmed that the direct and conventional IVIVE methods generally overpredicted and underpredicted the CL in vivo in humans, respectively. However, the underprediction of the conventional IVIVE method based on fup was significantly reduced from data generated with the HepatoPac® system compared with the 2 other in vitro assays, which is possibly because that system is producing ALB at a rate much closer to the in vivo condition in liver. Hence, these observations suggest that the presence of more ALB molecules per hepatocyte in that HepatoPac® system may have facilitated the hepatic uptake of several bound drugs because their intrinsic CL was increased instead of being decreased by the ALB binding effect. Accordingly, the IVIVE method based on the fup value adjusted for the ALB-facilitated uptake phenomenon gave the lowest prediction bias from the statistical analyses. This study indicated that the HepatoPac® system combined with the adjusted value of fup was the most reliable IVIVE method and revealed the importance of quantifying the in vitro-to-in vivo variation of ALB concentration to improve the CL predictions, which would help any future physiologically based pharmacokinetics modeling exercise.
Collapse
Affiliation(s)
- Franck Da-Silva
- Sanofi R&D, Montpellier, France; Institute for Regenerative Medicine and Biotherapy, Université et CHU de Montpellier, INSERM, Montpellier, France
| | | | | | | | - Sabine Gerbal-Chaloin
- Institute for Regenerative Medicine and Biotherapy, Université et CHU de Montpellier, INSERM, Montpellier, France
| | - Martine Daujat-Chavanieu
- Institute for Regenerative Medicine and Biotherapy, Université et CHU de Montpellier, INSERM, Montpellier, France
| | | | - Patrick Poulin
- Consultant, Patrick Poulin Inc., Québec City, Canada; Associate professor, School of Public Health, IRSPUM, Université de Montréal, Canada
| |
Collapse
|
124
|
Wood FL, Houston JB, Hallifax D. Importance of the Unstirred Water Layer and Hepatocyte Membrane Integrity In Vitro for Quantification of Intrinsic Metabolic Clearance. Drug Metab Dispos 2018; 46:268-278. [PMID: 29233818 DOI: 10.1124/dmd.117.078949] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2017] [Accepted: 12/11/2017] [Indexed: 11/22/2022] Open
Abstract
Prediction of clearance-a vital component of drug discovery-remains in need of improvement and, in particular, requires more incisive assessment of mechanistic methodology in vitro, according to a number of recent reports. Although isolated hepatocytes have become an irreplaceable standard system for the measurement of intrinsic hepatic clearance mediated by active uptake transport and metabolism, the lack of prediction reliability appears to reflect a lack of methodological validation, especially for highly cleared drugs, as we have previously shown. Here, novel approaches were employed to explore fundamental experimental processes and associated potential limitations of in vitro predictions of clearance. Rat hepatocytes deemed nonviable by trypan blue staining showed undiminished metabolic activity for probe cytochrome P450 (P450) substrates midazolam and propranolol; supplementation with NADPH enhanced these activities. Extensive permeabilization of the plasma membrane using saponin showed either full or minimal P450 activity, depending on the presence or absence of 1 mM NADPH, respectively. The shaking of incubations facilitated P450 metabolic rates up to 5-fold greater than static incubation, depending on intrinsic clearance, indicating the critical influence of the unstirred water layer (UWL). Permeabilization allowed static incubation metabolic rates to approach those of shaking for intact cells, indicating an artificially induced breakdown of the UWL. Permeabilization combined with shaking allowed an increased metabolic rate for saquinavir, resolving the membrane permeability limitation for this drug. These findings advance the interpretation of the rate-limiting processes involved in intrinsic clearance measurements and could be critical for successful in vitro prediction.
Collapse
Affiliation(s)
- Francesca L Wood
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - J Brian Houston
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| | - David Hallifax
- Centre for Applied Pharmacokinetic Research, Division of Pharmacy and Optometry, School of Health Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, United Kingdom
| |
Collapse
|