1
|
Tsuchitani T, Kato M, Tomaru A, Aoki Y, Sugiyama Y. Trends of in vitro pharmacological potency and in vivo pharmacokinetics parameters of modern drugs: Can the therapeutic/subtherapeutic dose be estimated from in vitro K i and pharmacokinetic parameters? Clin Transl Sci 2024; 17:e70034. [PMID: 39600105 PMCID: PMC11599425 DOI: 10.1111/cts.70034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/15/2024] [Accepted: 09/07/2024] [Indexed: 11/29/2024] Open
Abstract
In drug discovery and development, estimating therapeutic and subtherapeutic doses is crucial for designing early-phase clinical trials, particularly first-in-human (FIH) studies. While increasing the in vitro pharmacological potency (lowering Ki) of a compound to its target is expected to decrease the therapeutic dose, its benefit is not necessarily clarified. We analyzed in vitro Ki, human in vivo pharmacokinetics (PK) parameters, and therapeutics doses of 144 oral small-molecule drugs approved in Japan (2010-2021). The data on classic drugs were obtained from Goodman and Gilman's textbook, 9th ed. (published in 1996). Modern drugs had lower Ki (2.5 nM) and daily doses (88 μmol/day) than classic drugs (33 nM and 313 μmol/day), but 3.6-fold higher intrinsic clearance (CLint; 171 vs. 47 L/h), suggest that increasing potency over the years has not resulted in a reduction in dosage as expected. Estimating therapeutic doses using target receptor occupancy (tRO)-optimized approach improved estimation accuracy (63% within 10-fold of observed values) compared with tRO-fixed approaches. Subtherapeutic dose estimations revealed a risk of overdosing in FIH trials, indicating that these estimates are not necessarily as conservative as is generally understood. Notably, the unbound average concentration-to-Ki ratio (Cave,u/Ki) varied among drugs and correlated negatively with Ki, suggesting the possible necessity of incorporating it into dose estimation methods. This study provides insights into the relationship between in vitro Ki, in vivo PK parameters, and therapeutic dose of modern drugs, proposing strategies and revealing the risk for dose estimation and drug development in the era of highly potent small molecules.
Collapse
Affiliation(s)
- Toshiaki Tsuchitani
- iHuman Institute, ShanghaiTech UniversityShanghaiChina
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| | - Motohiro Kato
- Research Institute of Pharmaceutical Sciences, Musashino UniversityTokyoJapan
| | - Atsuko Tomaru
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| | - Yasunori Aoki
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
- Drug Metabolism and Pharmacokinetics, Research and Early Development, CardiovascularRenal and Metabolism (CVRM), BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Yuichi Sugiyama
- iHuman Institute, ShanghaiTech UniversityShanghaiChina
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| |
Collapse
|
2
|
Savaryn JP, Coe K, Cerny MA, Colizza K, Moliner P, King L, Ma B, Atherton J, Auclair A, Cancilla MT, Eno M, Jurva U, Yue Q, Zhu SX, Freiberger E, Zhong G, Barlock B, Nachtigall J, Laboureur L, Pusalkar S, Guo R, Niehues M, Hauri S, Carreras ET, Maurer C, Prakash C, Jenkins GJ. The Current State of Biotransformation Science - Industry Survey of In Vitro and In Vivo Practices, Clinical Translation, and Future Trends. Pharm Res 2024; 41:2079-2093. [PMID: 39496990 PMCID: PMC11599300 DOI: 10.1007/s11095-024-03787-y] [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: 08/01/2024] [Accepted: 10/17/2024] [Indexed: 11/06/2024]
Abstract
Embedded within the field of drug metabolism and pharmacokinetics (DMPK), biotransformation is a discipline that studies the origins, disposition, and structural identity of metabolites to provide a comprehensive safety assessment, including the assessment of exposure coverage in toxicological species. Spanning discovery and development, metabolite identification (metID) scientists employ various strategies and tools to address stage-specific questions aimed at guiding the maturation of early chemical matter into drug candidates. During this process, the identity of major (and minor) circulating human metabolites is ascertained to comply with the regulatory requirements such as the Metabolites in Safety Testing (MIST) guidance. Through the International Consortium for Innovation and Quality in Pharmaceutical Development (IQ), the "Translatability of MetID In Vitro Systems Working Group" was created within the Translational and ADME Sciences Leadership Group. The remit of this group was to objectively determine how accurate commonly employed in vitro systems have been with respect to prediction of circulating human metabolites, both qualitatively and quantitatively. A survey composed of 34 questions was conducted across 26 pharmaceutical companies to obtain a foundational understanding of current metID practices, preclinically and clinically, as well as to provide perspective on how successful these practices have been at predicting circulating human metabolites. The results of this survey are presented as an initial snapshot of current industry-based metID practices, including our perspective on how a harmonized framework for the conduct of in vitro metID studies could be established. Future perspectives from current practices to emerging advances with greater translational capability are also provided.
Collapse
Affiliation(s)
- John P Savaryn
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA.
| | - Kevin Coe
- J&J, Translational PKPD & Investigational Toxicology, San Diego, CA, USA
| | | | - Kevin Colizza
- GSK, DMPK Disposition and Biotransformation, Collegeville, PA, USA.
| | | | - Lloyd King
- UCB Biopharma, Dept. of DMPK, Slough, UK
| | - Bin Ma
- Genentech, Inc., Department of Drug Metabolism and Pharmacokinetics, South San Francisco, CA, USA
| | - Jim Atherton
- Incyte Research Institute, Translational Sciences, Wilmington, DE, USA
| | - Adam Auclair
- Boehringer Ingelheim Pharmaceuticals, Inc.,Drug Metabolism and Pharmacokinetics, Ridgefield, CT, USA
| | - Mark T Cancilla
- Merck & Co., Inc., Pharmacokinetics, Dynamics, Metabolism, and Bioanalysis, Rahway, NJ, USA
| | - Marsha Eno
- Eisai Inc., Global Drug Metabolism and Pharmacokinetics, Cambridge, MA, USA
| | - Ulrik Jurva
- AstraZeneca, Drug Metabolism and Pharmacokinetics (DMPK), Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM), BioPharmaceuticals R&D, Gothenburg, Sweden
| | - Qin Yue
- Gilead Sciences, Inc., Drug Metabolism Dept, Foster City, CA, USA
| | - Sean Xiaochun Zhu
- Takeda Development Center Americas, Inc., Drug Metabolism and Pharmacokinetics & Modeling, Cambridge, MA, USA
| | - Elyse Freiberger
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA
| | - Guo Zhong
- Amgen, Pharmacokinetics and Drug Metabolism Department, South San Francisco, CA, USA
| | | | | | | | | | - Runcong Guo
- Beigene, DMPK, Department of Biology, Shanghai, China
| | - Michael Niehues
- Bayer AG, In Vitro ADME & Isotope Chemistry, Berlin, Germany
| | - Simon Hauri
- Roche Pharma Research and Early Development, Pharmaceutical Sciences, Roche Innovation Center Basel, F. Hoffmann-La Roche, Basel, Switzerland
| | - Ester Tor Carreras
- Novartis Pharma AG, Novartis Institute for Biomedical Research, Basel, Switzerland
| | | | - Chandra Prakash
- DMPK/Clinical Pharmacology, Agios Pharmaceuticals, Cambridge, MA, USA
| | - Gary J Jenkins
- AbbVie, Quantitative, Translational & ADME Sciences, North Chicago, IL, USA
| |
Collapse
|
3
|
Chothe PP, Argikar UA, Mitra P, Nakakariya M, Ramsden D, Rotter CJ, Sandoval P, Tohyama K. Drug transporters in drug disposition - highlights from the year 2023. Drug Metab Rev 2024; 56:318-348. [PMID: 39221672 DOI: 10.1080/03602532.2024.2399523] [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: 05/06/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Drug transporter field is rapidly evolving with significant progress in in vitro and in vivo tools and, computational models to assess transporter-mediated drug disposition and drug-drug interactions (DDIs) in humans. On behalf of all coauthors, I am pleased to share the fourth annual review highlighting articles published and deemed influential in the field of drug transporters in the year 2023. Each coauthor independently selected peer-reviewed articles published or available online in the year 2023 and summarized them as shown previously (Chothe et al. 2021; Chothe et al. 2022, 2023) with unbiased perspectives. Based on selected articles, this review was categorized into four sections: (1) transporter structure and in vitro evaluation, (2) novel in vitro/ex vivo models, (3) endogenous biomarkers, and (4) PBPK modeling for evaluating transporter DDIs (Table 1). As the scope of this review is not to comprehensively review each article, readers are encouraged to consult original paper for specific details. Finally, I appreciate all the authors for their time and continued support in writing this review.
Collapse
Affiliation(s)
- Paresh P Chothe
- Drug Metabolism and Pharmacokinetics, Oncology Research and Development, AstraZeneca, Waltham, MA, USA
| | - Upendra A Argikar
- Non-clinical Development, Bill and Melinda Gates Medical Research Institute, Cambridge, MA, USA
| | - Pallabi Mitra
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc, Ridgefield, CT, USA
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda irinote Pharmaceutical Company Limited, Fujisawa, Japan
| | - Diane Ramsden
- Preclinical Development, Korro Bio, Inc. One Kendall Square, Cambridge, MA, USA
| | - Charles J Rotter
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), San Diego, CA, USA
| | - Philip Sandoval
- Global Drug Metabolism and Pharmacokinetics, Takeda Development Center Americas, Inc. (TDCA), Lexington, MA, USA
| | - Kimio Tohyama
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda irinote Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
4
|
Tan SPF, Tillmann A, Murby SJ, Rostami-Hodjegan A, Scotcher D, Galetin A. Albumin-Mediated Drug Uptake by Organic Anion Transporter 1/3 Is Real: Implications for the Prediction of Active Renal Secretion Clearance. Mol Pharm 2024; 21:4603-4617. [PMID: 39166754 PMCID: PMC11372837 DOI: 10.1021/acs.molpharmaceut.4c00504] [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] [Indexed: 08/23/2024]
Abstract
Modulation of the transport-mediated active uptake by human serum albumin (HSA) for highly protein-bound substrates has been reported and improved the in vitro-to-in vivo extrapolation (IVIVE) of hepatic clearance. However, evidence for the relevance of such a phenomenon in the case of renal transporters is sparse. In this study, transport of renal organic anion transporter 1 or 3 (OAT1/3) substrates into conditionally immortalized proximal tubular epithelial cells transduced with OAT1/3 was measured in the presence and absence of 1 and 4% HSA while keeping the unbound substrate concentration constant (based on measured fraction unbound, fu,inc). In the presence of 4% HSA, the unbound intrinsic active uptake clearance (CLint,u,active) of six highly protein-bound substrates increased substantially relative to the HSA-free control (3.5- to 122-fold for the OAT1 CLint,u,active, and up to 28-fold for the OAT3 CLint,u,active). The albumin-mediated uptake effect (fold increase in CLint,u,active) was more pronounced with highly bound substrates compared to no effect seen for weakly protein-bound substrates adefovir (OAT1-specific) and oseltamivir carboxylate (OAT3-specific). The relationship between OAT1/3 CLint,u,active and fu,inc agreed with the facilitated-dissociation model; a relationship was established between the albumin-mediated fold change in CLint,u,active and fu,inc for both the OAT1 and OAT3, with implications for IVIVE modeling. The relative activity factor and the relative expression factor based on global proteomic quantification of in vitro OAT1/3 expression were applied for IVIVE of renal clearance. The inclusion of HSA improved the bottom-up prediction of the level of OAT1/3-mediated secretion and renal clearance (CLsec and CLr), in contrast to the underprediction observed with the control (HSA-free) scenario. For the first time, this study confirmed the presence of the albumin-mediated uptake effect with renal OAT1/3 transporters; the extent of the effect was more pronounced for highly protein-bound substrates. We recommend the inclusion of HSA in routine in vitro OAT1/3 assays due to considerable improvements in the IVIVE of CLsec and CLr.
Collapse
Affiliation(s)
- Shawn Pei Feng Tan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PL, U.K
| | - Annika Tillmann
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PL, U.K
| | - Susan J Murby
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PL, U.K
| | - Amin Rostami-Hodjegan
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PL, U.K
- Certara Predictive Technologies (CPT), Certara Inc., 1 Concourse Way, Sheffield S1 2BJ, U.K
| | - Daniel Scotcher
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PL, U.K
| | - Aleksandra Galetin
- Centre for Applied Pharmacokinetic Research, School of Health Sciences, University of Manchester, Manchester M13 9PL, U.K
| |
Collapse
|
5
|
Schulz Pauly JA, Kalvass JC. How predictive are isolated perfused liver data of in vivo hepatic clearance? A meta-analysis of isolated perfused rat liver data. Xenobiotica 2024; 54:658-669. [PMID: 39279675 DOI: 10.1080/00498254.2024.2404170] [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: 06/27/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/18/2024]
Abstract
Isolated perfused rat liver (IPRL) experiments have been used to answer clearance-related questions, including evaluating the impact of pathological and physiological processes on hepatic clearance (CLH). However, to date, IPRL data has not been evaluated for in vivo CLH prediction accuracy.In addition to a detailed overview of available IPRL literature, we present an in-depth analysis of the performance of IPRL in CLH prediction.While the entire dataset poorly predicted CLH (GAFE = 3.2; 64% within 3-fold), IPRL conducted under optimal experimental conditions, such as in the presence of plasma proteins and with a perfusion rate within 2-fold of physiological liver blood flow and corrected for unbound fraction in the presence of red blood cells, can accurately predict rat CLH (GAFE = 2.0; 78% within 3-fold). Careful consideration of experimental conditions is needed to allow proper data analysis.Further, isolated perfused liver experiments in other species, including human livers, may allow us to address the current in vitro-in vivo disconnects of hepatic metabolic clearance and improve our methodology for CLH predictions.
Collapse
Affiliation(s)
- Julia A Schulz Pauly
- Quantitative, Translational, & ADME Sciences (QTAS), Abbvie Inc., North Chicago, IL, USA
| | - J Cory Kalvass
- Quantitative, Translational, & ADME Sciences (QTAS), Abbvie Inc., North Chicago, IL, USA
| |
Collapse
|
6
|
Poulin P. First-in-Human Predictions of Hepatic Clearance for Drugs With the Well-Stirred Model: Comparative Assessment Between Models of Fraction Unbound Based Either on the Free Drug Hypothesis, Albumin-Facilitated Hepatic Uptake or Dynamic Binding Kinetics. J Pharm Sci 2024; 113:2641-2650. [PMID: 38796154 DOI: 10.1016/j.xphs.2024.05.021] [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: 02/27/2024] [Revised: 05/16/2024] [Accepted: 05/16/2024] [Indexed: 05/28/2024]
Abstract
The well-stirred model (WSM) is commonly used to predict the hepatic clearance in vivo (CLH) of drugs. The necessary intrinsic clearance of the unbound drug (CLint-in vitro-unbound) is generated in the in vitro assays in the presence of microsomes or hepatocytes but in the absence of plasma proteins. The value of CLint-in vitro-unbound can be extrapolated with the fraction unbound determined in vitro in plasma (fup) only if the fraction unbound in vivo in liver is the same. However, this approach resulted to a systematic underprediction bias of CLH. With the goal of reducing this bias, two new models of fraction unbound were published in this journal. These models estimate the binding kinetics of the rates of association and dissociation of the drug-protein complex and propose that more dissociation in the liver compared to plasma will increase the fraction unbound available for the metabolism. Consequently, these two models generated higher values of fraction unbound, implying a lower underprediction bias of CLH with the WSM. The first model was developed by Poulin et al. and is referring to the value of fup that is adjusted (fu-adjusted) to quantify the effect of a full dissociation of the drug-protein complex at the hepatocyte membrane in accordance with the theory of the albumin-facilitated hepatic uptake. A second model was developed by Yan et al. who presented a dynamic fraction unbound (fu-dynamic) measuring the real dissociation kinetics of the drug-protein complex with a new in vitro assay in the presence and absence of a recombinant liver enzyme in plasma. Therefore, the objective of this study was to make the first comparative assessment between these two models. The results indicate that, in general, the WSM combined with the values of fu-adjusted was the most accurate approach for predicting CLH. The WSM combined with the values of fu-dynamic has underperformed particularly with the acidic and neutral drugs binding to the albumin and presenting a low metabolic turnover in vitro. Therefore, the new in vitro assay for fu-dynamic gave an underprediction bias of CLH for these drug properties. However, the values of fu-adjusted are significantly higher than those values of fu-dynamic, and, this resulted to no underprediction bias, which is reinforcing the theory of the ALB-facilitated hepatic uptake. For the other neutral and acidic drugs, the models of fu-dynamic and fu-adjusted are in closer agreement. Finally, for the basic drugs, the models of fu-adjusted and fu-dynamic as well as a third model only considering a pH gradient effect on fup are almost accurately equivalent.
Collapse
Affiliation(s)
- Patrick Poulin
- Consultant Patrick Poulin Inc., Québec City, Québec, Canada; School of Public Health, Université de Montréal, Montréal, Québec, Canada.
| |
Collapse
|
7
|
Li R, Kimoto E, Bi YA, Tess D, Varma MVS. Physiologically Based Pharmacokinetic Model of OATP1B Substrates with a Nonlinear Mixed Effect Approach: Estimating Empirical In Vitro-to-In Vivo Scaling Factors. Clin Pharmacokinet 2024; 63:1177-1189. [PMID: 39158814 DOI: 10.1007/s40262-024-01408-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/28/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND AND OBJECTIVE Physiologically based pharmacokinetic (PBPK) models are valuable for translating in vitro absorption, distribution, metabolism, and excretion (ADME) data to predict clinical pharmacokinetics, and can enable discovery and early clinical stages of pharmaceutical research. However, in predicting pharmacokinetics of organic anion transporting polypeptide (OATP) 1B substrates based on in vitro transport and metabolism data, PBPK models typically require additional empirical in vitro-to-in vivo scaling factors (ESFs) in order to accurately recapitulate observed clinical profiles. As model simulation is very sensitive to ESFs, a critical evaluation of ESF estimation is prudent. Previously studies have applied classic 'two-stage' and 'naïve pooled data' approaches in identifying a set of compound independent ESFs. However, the 'two-stage' approach has the parameter identification issue in separately fitting data for individual compounds, while the 'naïve pooled data' approach ignores interstudy variability, leading to potentially biased ESF estimates. METHODS In this study, we have applied a nonlinear mixed-effect approach in estimating ESF of the PBPK model and incorporated additional data from 86 runs of in vitro uptake assay and 49 clinical studies of 12 training compounds in model development to further enhance the translation of in vitro data to predict the pharmacokinetics of OATP1B substrate drugs. To test predication accuracy of the model, a 'leave-one-out' analysis has been performed. RESULTS The established model can reasonably describe the clinical observations, with both mean values and interstudy variabilities quantified for ESF and volume of distribution parameters. The mean estimates are largely consistent with values in the previous reports. The interstudy variabilities of these parameters are estimated to be at least 50% (as coefficient of variation). Most compounds can be reasonably predicted in the 'leave-one-out' analysis. CONCLUSION This study improves the confidence in predicting the pharmacokinetics of OATP1B substrates in individual studies of small sample sizes, and quantifies the variability associated with the prediction.
Collapse
Affiliation(s)
- Rui Li
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Cambridge, MA, 02139, USA.
| | - Emi Kimoto
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Groton, CT, USA
| | - Yi-An Bi
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Groton, CT, USA
| | - David Tess
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Cambridge, MA, 02139, USA
| | - Manthena V S Varma
- Pharmacokinetics Dynamics and Metabolism, Pfizer Global Research and Development, Groton, CT, USA
| |
Collapse
|
8
|
Pang KS, Lu WI, Mulder GJ. After 50 Years of Hepatic Clearance Models, Where Should We Go from Here? Improvements and Implications for Physiologically Based Pharmacokinetic Modeling. Drug Metab Dispos 2024; 52:919-931. [PMID: 39013583 DOI: 10.1124/dmd.124.001649] [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: 01/09/2024] [Accepted: 04/25/2024] [Indexed: 07/18/2024] Open
Abstract
There is overwhelming preference for application of the unphysiologic, well-stirred model (WSM) over the parallel tube model (PTM) and dispersion model (DM) to predict hepatic drug clearance, CLH , despite that liver blood flow is dispersive and closer to the DM in nature. The reasoning is the ease in computation relating the hepatic intrinsic clearance ( CLint ), hepatic blood flow ( QH ), unbound fraction in blood ( fub ) and the transmembrane clearances ( CLin and CLef ) to CLH for the WSM. However, the WSM, being the least efficient liver model, predicts a lower EH that is associated with the in vitro CLint ( Vmax / Km ), therefore requiring scale-up to predict CLH in vivo. By contrast, the miniPTM, a three-subcompartment tank-in-series model of uniform enzymes, closely mimics the DM and yielded similar patterns for CLint versus EH , substrate concentration [S] , and KL / B , the tissue to outflow blood concentration ratio. We placed these liver models nested within physiologically based pharmacokinetic models to describe the kinetics of the flow-limited, phenolic substrate, harmol, using the WSM (single compartment) and the miniPTM and zonal liver models (ZLMs) of evenly and unevenly distributed glucuronidation and sulfation activities, respectively, to predict CLH For the same, given CLint ( Vmax and Km ), the WSM again furnished the lowest extraction ratio ( EH,WSM = 0.5) compared with the miniPTM and ZLM (>0.68). Values of EH,WSM were elevated to those for EH, PTM and EH, ZLM when the Vmax s for sulfation and glucuronidation were raised 5.7- to 1.15-fold. The miniPTM is easily manageable mathematically and should be the new normal for liver/physiologic modeling. SIGNIFICANCE STATEMENT: Selection of the proper liver clearance model impacts strongly on CLH predictions. The authors recommend use of the tank-in-series miniPTM (3 compartments mini-parallel tube model), which displays similar properties as the dispersion model (DM) in relating CLint and [ S ] to CLH as a stand-in for the DM, which best describes the liver microcirculation. The miniPTM is readily modified to accommodate enzyme and transporter zonation.
Collapse
Affiliation(s)
- K Sandy Pang
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P., W.I.L.) and Department of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (G.J.M.)
| | - Weijia Ivy Lu
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P., W.I.L.) and Department of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (G.J.M.)
| | - Gerard J Mulder
- Leslie Dan Faculty of Pharmacy, University of Toronto, Ontario, Canada (K.S.P., W.I.L.) and Department of Toxicology, Leiden Academic Centre for Drug Research, Leiden University, The Netherlands (G.J.M.)
| |
Collapse
|
9
|
Tsuchitani T, Tomaru A, Aoki Y, Ishiguro N, Tsuda Y, Sugiyama Y. Elucidating nonlinear pharmacokinetics of telmisartan: Integration of target-mediated drug disposition and OATP1B3-mediated hepatic uptake in a physiologically based model. CPT Pharmacometrics Syst Pharmacol 2024; 13:1224-1237. [PMID: 38745377 PMCID: PMC11247111 DOI: 10.1002/psp4.13154] [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: 01/10/2024] [Revised: 03/25/2024] [Accepted: 04/16/2024] [Indexed: 05/16/2024] Open
Abstract
Telmisartan, a selective inhibitor of angiotensin II receptor type 1 (AT1), demonstrates nonlinear pharmacokinetics (PK) when orally administered in ascending doses to healthy volunteers, but the underlying mechanisms remain unclear. This study presents a physiologically based pharmacokinetic model integrated with target-mediated drug disposition (TMDD-PBPK model) to explore the mechanism of its nonlinear PK. We employed the Cluster-Gauss Newton method for top-down analysis, estimating the in vivo Km,OATP1B3 (Michaelis-Menten constant for telmisartan hepatic uptake via Organic Anion Transporting Polypeptide 1B3) to be 2.0-5.7 nM. This range is significantly lower than the reported in vitro value of 810 nM, obtained in 0.3% human serum albumin (HSA) conditions. Further validation was achieved through in vitro assessment in plated human hepatocytes with 4.5% HSA, showing a Km of 4.5 nM. These results underscore the importance of albumin-mediated uptake effect for the hepatic uptake of telmisartan. Our TMDD-PBPK model, developed through a "middle-out" approach, underwent sensitivity analysis to identify key factors in the nonlinear PK of telmisartan. We found that the nonlinearity in the area under the concentration-time curve (AUC) and/or maximum concentration (Cmax) of telmisartan is sensitive to Km,OATP1B3 across all dosages. Additionally, the dissociation constant (Kd) for telmisartan binding to the AT1 receptor, along with its receptor abundance, notably influences PK at lower doses (below 20 mg). In conclusion, the nonlinear PK of telmisartan appears primarily driven by hepatic uptake saturation across all dose ranges and by AT1-receptor binding saturation, notably at lower doses.
Collapse
Affiliation(s)
- Toshiaki Tsuchitani
- iHuman InstituteShanghaiTech UniversityShanghaiChina
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| | - Atsuko Tomaru
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| | - Yasunori Aoki
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
- Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism (CVRM)BioPharmaceuticals R&D, AstraZenecaGothenburgSweden
| | - Naoki Ishiguro
- Pharmacokinetics and Non‐Clinical Safety DepartmentNippon Boehringer Ingelheim Co., Ltd.KobeHyogoJapan
| | - Yasuhiro Tsuda
- Clinical Pharmacology DepartmentNippon Boehringer Ingelheim Co., Ltd.KobeHyogoJapan
| | - Yuichi Sugiyama
- iHuman InstituteShanghaiTech UniversityShanghaiChina
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, School of PharmacyJosai International University (JIU)TokyoJapan
| |
Collapse
|
10
|
Yan Z, Ma L, Hwang N, Huang J, Kenny JR, Hop CECA. Using the Dynamic Well-Stirred Model to Extrapolate Hepatic Clearance of Organic Anion-Transporting Polypeptide Transporter Substrates without Assuming Albumin-Mediated Hepatic Drug Uptake. Drug Metab Dispos 2024; 52:548-554. [PMID: 38604729 DOI: 10.1124/dmd.124.001645] [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: 01/04/2024] [Revised: 03/13/2024] [Accepted: 03/27/2024] [Indexed: 04/13/2024] Open
Abstract
Extrapolating in vivo hepatic clearance from in vitro uptake data is a known challenge, especially for organic anion-transporting polypeptide transporter (OATP) substrates, and the well-stirred model (WSM) commonly yields systematic underpredictions for those anionic drugs, hypothetically due to "albumin-mediated hepatic drug uptake". In the present study, we demonstrate that the WSM incorporating the dynamic free fraction (f D), a measure of drug protein binding affinity, performs reasonably well in predicting hepatic clearance of OATP substrates. For a selection of anionic drugs, including atorvastatin, fluvastatin, pravastatin, rosuvastatin, pitavastatin, cerivastatin, and repaglinide, this dynamic well-stirred model (dWSM) correctly predicts hepatic plasma clearance within 2-fold error for six out of seven OATP substrates examined. The geometric mean of clearance ratios between the predicted and the observed values falls in the range of 1.21-1.38. As expected, the WSM with unbound fraction (f u) systematically underpredicts hepatic clearance with greater than 2-fold error for five out of seven drugs, and the geometric mean of clearance ratios between the predicted and the observed values is in the range of 0.20-0.29. The results suggest that, despite its simplicity, the dWSM operates well for transporter-mediated uptake clearance, and that clearance under-prediction of OATP substrates may not necessarily be associated with the chemical class of the anionic drugs, nor is it a result of albumin-mediated hepatic drug uptake as currently hypothesized. Instead, the superior prediction power of the dWSM confirms the utility of the dynamic free fraction in clearance prediction and the importance of drug plasma binding kinetics in hepatic uptake clearance. SIGNIFICANCE STATEMENT: The traditional well-stirred model (WSM) consistently underpredicts organin anion-transporting polypeptide transporter (OATP)-mediated hepatic uptake clearance, hypothetically due to the albumin-mediated hepatic drug uptake. In this manuscript, we apply the dynamic WSM to extrapolate hepatic clearance of the OATP substrates, and our results show significant improvements in clearance prediction without assuming albumin-mediated hepatic drug uptake.
Collapse
Affiliation(s)
- Zhengyin Yan
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Li Ma
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Nicky Hwang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Julie Huang
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Jane R Kenny
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| | - Cornelis E C A Hop
- Department of Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California
| |
Collapse
|
11
|
Handa K, Yoshimura S, Kageyama M, Iijima T. Development of Novel Methods for QSAR Modeling by Machine Learning Repeatedly: A Case Study on Drug Distribution to Each Tissue. J Chem Inf Model 2024; 64:3662-3669. [PMID: 38639496 DOI: 10.1021/acs.jcim.4c00046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/20/2024]
Abstract
Artificial intelligence is expected to help identify excellent candidates in drug discovery. However, we face a lack of data, as it is time-consuming and expensive to acquire raw data perfectly for many compounds. Hence, we tried to develop a novel quantitative structure-activity relationship (QSAR) method to predict a parameter more precisely from an incomplete data set via optimizing data handling by making use of predicted explanatory variables. As a case study we focused on the tissue-to-plasma partition coefficient (Kp), which is an important parameter for understanding drug distribution in tissues and building the physiologically based pharmacokinetic model and is a representative of small and sparse data sets. In this study, we predicted the Kp values of 119 compounds in nine tissues (adipose, brain, gut, heart, kidney, liver, lung, muscle, and skin), although some of these were not available. To fill the missing values in Kp for each tissue, first we predicted those Kp values by the nonmissing data set using a random forest (RF) model with in vitro parameters (log P, fu, Drug Class, and fi) like a classical prediction by a QSAR model. Next, to predict the tissue-specific Kp values in a test data set, we constructed a second RF model with not only in vitro parameters but also the Kp values of other tissues (i.e., other than target tissues) predicted by the first RF model as explanatory variables. Furthermore, we tested all possible combinations of explanatory variables and selected the model with the highest predictability from the test data set as the final model. The evaluation of Kp prediction accuracy based on the root-mean-square error and R2 value revealed that the proposed models outperformed other machine learning methods such as the conventional RF and message-passing neural networks. Significant improvements were observed in the Kp values of adipose tissue, brain, kidney, liver, and skin. These improvements indicated that the Kp information on other tissues can be used to predict the same for a specific tissue. Additionally, we found a novel relationship between each tissue by evaluating all combinations of explanatory variables. In conclusion, we developed a novel RF model to predict Kp values. We hope that this method will be applied to various problems in the field of experimental biology which often contains missing values in the near future.
Collapse
Affiliation(s)
- Koichi Handa
- Toxicology & DMPK Research Department, Teijin Institute for Bio-medical Research, Teijin Pharma Limited, 4-3-2 Asahigaoka, Hino-shi, Tokyo 191-8512, Japan
| | - Saki Yoshimura
- Toxicology & DMPK Research Department, Teijin Institute for Bio-medical Research, Teijin Pharma Limited, 4-3-2 Asahigaoka, Hino-shi, Tokyo 191-8512, Japan
| | - Michiharu Kageyama
- Toxicology & DMPK Research Department, Teijin Institute for Bio-medical Research, Teijin Pharma Limited, 4-3-2 Asahigaoka, Hino-shi, Tokyo 191-8512, Japan
| | - Takeshi Iijima
- Toxicology & DMPK Research Department, Teijin Institute for Bio-medical Research, Teijin Pharma Limited, 4-3-2 Asahigaoka, Hino-shi, Tokyo 191-8512, Japan
| |
Collapse
|
12
|
Liang X, Koleske ML, Yang J, Lai Y. Building a Predictive PBPK Model for Human OATP Substrates: a Strategic Framework for Early Evaluation of Clinical Pharmacokinetic Variations Using Pitavastatin as an Example. AAPS J 2024; 26:13. [PMID: 38182946 DOI: 10.1208/s12248-023-00882-7] [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: 08/16/2023] [Accepted: 12/07/2023] [Indexed: 01/07/2024] Open
Abstract
To select a drug candidate for clinical development, accurately and promptly predicting human pharmacokinetic (PK) profiles, assessing drug-drug interactions (DDIs), and anticipating potential PK variations in disease populations are crucial steps in drug discovery. The complexity of predicting human PK significantly increases when hepatic transporters are involved in drug clearance (CL) and volume of distribution (Vss). A strategic framework is developed here, utilizing pitavastatin as an example. The framework includes the construction of a monkey physiologically-based PK (PBPK) model, model calibration to obtain scaling factors (SF) of in vitro-in vivo extrapolation (IVIVE) for various clearance parameters, human model development and validation, and assessment of DDIs and PK variations in disease populations. Through incorporating in vitro human parameters and calibrated SFs from the monkey model of 3.45, 0.14, and 1.17 for CLint,active, CLint,passive, and CLint,bile, respectively, and together with the relative fraction transported by individual transporters obtained from in vitro studies and the optimized Ki values for OATP inhibition, the model reasonably captured observed pitavastatin PK profiles, DDIs and PK variations in human subjects carrying genetic polymorphisms, i.e., AUC within 20%. Lastly, when applying the functional reduction based on measured OATP1B biomarkers, the model adequately predicted PK changes in the hepatic impairment population. The present study presents a strategic framework for early-stage drug development, enabling the prediction of PK profiles and assessment of PK variations in scenarios like DDIs, genetic polymorphism, and hepatic impairment-related disease states, specifically focusing on OATP substrates.
Collapse
Affiliation(s)
- Xiaomin Liang
- Drug Metabolism, Gilead Sciences Inc., 333 Lakeside Dr., Foster City, California, 94404, USA
| | - Megan L Koleske
- Drug Metabolism, Gilead Sciences Inc., 333 Lakeside Dr., Foster City, California, 94404, USA
| | - Jesse Yang
- Drug Metabolism, Gilead Sciences Inc., 333 Lakeside Dr., Foster City, California, 94404, USA
| | - Yurong Lai
- Drug Metabolism, Gilead Sciences Inc., 333 Lakeside Dr., Foster City, California, 94404, USA.
| |
Collapse
|
13
|
Jeong YS, Jusko WJ. A Complete Extension of Classical Hepatic Clearance Models Using Fractional Distribution Parameter f d in Physiologically Based Pharmacokinetics. J Pharm Sci 2024; 113:95-117. [PMID: 37279835 PMCID: PMC10902797 DOI: 10.1016/j.xphs.2023.05.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/30/2023] [Accepted: 05/30/2023] [Indexed: 06/08/2023]
Abstract
The classical organ clearance models have been proposed to relate the plasma clearance CLp to probable mechanism(s) of hepatic clearance. However, the classical models assume the intrinsic capability of drug elimination (CLu,int) that is physically segregated from the vascular blood but directly acts upon the unbound drug concentration in the blood (fubCavg), and do not handle the transit-time delay between the inlet/outlet concentrations in their closed-form clearance equations. Therefore, we propose unified model structures that can address the internal blood concentration patterns of clearance organs in a more mechanistic/physiological manner, based on the fractional distribution parameter fd operative in PBPK. The basic partial/ordinary differential equations for four classical models are revisited/modified to yield a more complete set of extended clearance models, i.e., the Rattle, Sieve, Tube, and Jar models, which are the counterparts of the dispersion, series-compartment, parallel-tube, and well-stirred models. We demonstrate the feasibility of applying the resulting extended models to isolated perfused rat liver data for 11 compounds and an example dataset for in vitro-in vivo extrapolation of the intrinsic to the systemic clearances. Based on their feasibilities to handle such real data, these models may serve as an improved basis for applying clearance models in the future.
Collapse
Affiliation(s)
- Yoo-Seong Jeong
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA
| | - William J Jusko
- Department of Pharmaceutical Sciences, School of Pharmacy and Pharmaceutical Sciences, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
14
|
Mitra P, Kasliwala R, Iboki L, Madari S, Williams Z, Takahashi R, Taub ME. Mechanistic Static Model based Prediction of Transporter Substrate Drug-Drug Interactions Utilizing Atorvastatin and Rifampicin. Pharm Res 2023; 40:3025-3042. [PMID: 37821766 DOI: 10.1007/s11095-023-03613-x] [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: 06/30/2023] [Accepted: 09/19/2023] [Indexed: 10/13/2023]
Abstract
OBJECTIVE An in vitro relative activity factor (RAF) technique combined with mechanistic static modeling was examined to predict drug-drug interaction (DDI) magnitude and analyze contributions of different clearance pathways in complex DDIs involving transporter substrates. Atorvastatin and rifampicin were used as a model substrate and inhibitor pair. METHODS In vitro studies were conducted with transfected HEK293 cells, hepatocytes and human liver microsomes. Prediction success was defined as predictions being within twofold of observations. RESULTS The RAF method successfully translated atorvastatin uptake from transfected cells to hepatocytes, demonstrating its ability to quantify transporter contributions to uptake. Successful translation of atorvastatin's in vivo intrinsic hepatic clearance (CLint,h,in vivo) from hepatocytes to liver was only achieved through consideration of albumin facilitated uptake or through application of empirical scaling factors to transporter-mediated clearances. Transporter protein expression differences between hepatocytes and liver did not affect CLint,h,in vivo predictions. By integrating cis and trans inhibition of OATP1B1/OATP1B3, atorvastatin-rifampicin (single dose) DDI magnitude could be accurately predicted (predictions within 0.77-1.0 fold of observations). Simulations indicated that concurrent inhibition of both OATP1B1 and OATP1B3 caused approximately 80% of atorvastatin exposure increases (AUCR) in the presence of rifampicin. Inhibiting biliary elimination, hepatic metabolism, OATP2B1, NTCP, and basolateral efflux are predicted to have minimal to no effect on AUCR. CONCLUSIONS This study demonstrates the effective application of a RAF-based translation method combined with mechanistic static modeling for transporter substrate DDI predictions and subsequent mechanistic interpretation.
Collapse
Affiliation(s)
- Pallabi Mitra
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals Inc., 900 Old Ridgebury Road, Ridgefield, CT, 06877, USA.
| | - Rumanah Kasliwala
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Laeticia Iboki
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Shilpa Madari
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Zachary Williams
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| | - Ryo Takahashi
- Pharmacokinetics and Non-Clinical Safety Department, Nippon Boehringer Ingelheim Co., Ltd., Kobe, Hyogo, Japan
| | - Mitchell E Taub
- Department of Drug Metabolism and Pharmacokinetics, Boehringer Ingelheim Pharmaceuticals, Inc., Ridgefield, CT, USA
| |
Collapse
|
15
|
Francis L, Ogungbenro K, De Bruyn T, Houston JB, Hallifax D. Exploring the Boundaries for In Vitro-In Vivo Extrapolation: Use of Isolated Rat Hepatocytes in Co-culture and Impact of Albumin Binding Properties in the Prediction of Clearance of Various Drug Types. Drug Metab Dispos 2023; 51:1463-1473. [PMID: 37580106 DOI: 10.1124/dmd.123.001309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 07/15/2023] [Accepted: 08/08/2023] [Indexed: 08/16/2023] Open
Abstract
Prediction of hepatic clearance of drugs (via uptake or metabolism) from in vitro systems continues to be problematic, particularly when plasma protein binding is high. The following work explores simultaneous assessment of both clearance processes, focusing on a commercial hepatocyte-fibroblast co-culture system (HμREL) over a 24-hour period using six probe drugs (ranging in metabolic and transporter clearance and low-to-high plasma protein binding). A rat hepatocyte co-culture assay was established using drug depletion (measuring both medium and total concentrations) and cell uptake kinetic analysis, both in the presence and absence of plasma protein (1% bovine serum albumin). Secretion of endogenous albumin was monitored as a marker of viability, and this reached 0.004% in incubations (at a rate similar to in vivo synthesis). Binding to stromal cells was substantial and required appropriate correction factors. Drug concentration-time courses were analyzed both by conventional methods and a mechanistic cell model prior to in vivo extrapolation. Clearance assayed by drug depletion in conventional suspended rat hepatocytes provided a benchmark to evaluate co-culture value. Addition of albumin appeared to improve predictions for some compounds (where fraction unbound in the medium is less than 0.1); however, for high-binding drugs, albumin significantly limited quantification and thus predictions. Overall, these results highlight ongoing challenges concerning in vitro hepatocyte system complexity and limitations of practical expediency. Considering this, more reliable measurement of hepatically cleared compounds seems possible through judicious use of available hepatocyte systems, including co-culture systems, as described herein; this would include those compounds with low metabolic turnover but high active uptake clearance. SIGNIFICANCE STATEMENT: Co-culture systems offer a more advanced tool than standard hepatocytes, with the ability to be cultured for longer periods of time, yet their potential as an in vitro tool has not been extensively assessed. We evaluate the strengths and limitations of the HμREL system using six drugs representing various metabolic and transporter-mediated clearance pathways with various degrees of albumin binding. Studies in the presence/absence of albumin allow in vitro-in vivo extrapolation and a framework to maximize their utility.
Collapse
Affiliation(s)
- Laura Francis
- 1Centre of 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 (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - Kayode Ogungbenro
- 1Centre of 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 (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - Tom De Bruyn
- 1Centre of 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 (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - J Brian Houston
- 1Centre of 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 (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| | - David Hallifax
- 1Centre of 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 (L.F., K.O., J.B.H., D.H.) and Genentech, Inc., South San Francisco, California (T.D.B.)
| |
Collapse
|
16
|
Yin M, Ishida K, Liang X, Lai Y, Unadkat JD. Interpretation of Protein-Mediated Uptake of Statins by Hepatocytes Is Confounded by the Residual Statin-Protein Complex. Drug Metab Dispos 2023; 51:1381-1390. [PMID: 37429727 DOI: 10.1124/dmd.123.001386] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 06/29/2023] [Accepted: 07/05/2023] [Indexed: 07/12/2023] Open
Abstract
Inclusion of plasma (or plasma proteins) in human hepatocyte uptake studies narrows, but does not close, the gap in in vitro to in vivo extrapolation (IVIVE) of organic anion transporting polypeptide (OATP)-mediated hepatic clearance (CLh) of statins. We have previously shown that this "apparent" protein-mediated uptake effect (PMUE) of statins by OATP1B1-expressing cells, in the presence of 5% human serum albumin (HSA), is mostly an artifact caused by residual statin-HSA complex remaining in the uptake assay. We determined if the same was true with plated human hepatocytes (PHH) and if this artifact can be reduced using suspended human hepatocytes (SHH) and the oil-spin method. We quantified the uptake of a cocktail of five statins by PHH and SHH in the absence and presence of 5% HSA. After terminating the uptake assay, the amount of residual HSA was quantified by quantitative targeted proteomics. For both PHH and SHH, except for atorvastatin and cerivastatin, the increase in total, active, and passive uptake of the statins, in the presence of 5% HSA, was explained by the estimated residual stain-HSA complex. In addition, the increase in active statin uptake by SHH, where present, was marginal (<50%), much smaller than that observed with PHH. Such a marginal increase cannot bridge the gap in IVIVE of CLh of statins. These data disprove the prevailing hypotheses for the in vitro PMUE. A true PMUE should be evaluated using the uptake data corrected for the residual drug-protein complex. SIGNIFICANCE STATEMENT: We show that the apparent protein-mediated uptake (PMUE) of statins by human hepatocytes is largely confounded by residual statin when plated or suspended human hepatocytes are used. Therefore, mechanisms other than PMUE need to be explored to explain the underprediction of the in vivo human hepatic clearance of statins by human hepatocyte uptake assays.
Collapse
Affiliation(s)
- Mengyue Yin
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.Y., J.D.U.); and Drug Metabolism, Gilead Sciences, Inc., Foster City, California (K.I., X.L., Y.L.)
| | - Kazuya Ishida
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.Y., J.D.U.); and Drug Metabolism, Gilead Sciences, Inc., Foster City, California (K.I., X.L., Y.L.)
| | - Xiaomin Liang
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.Y., J.D.U.); and Drug Metabolism, Gilead Sciences, Inc., Foster City, California (K.I., X.L., Y.L.)
| | - Yurong Lai
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.Y., J.D.U.); and Drug Metabolism, Gilead Sciences, Inc., Foster City, California (K.I., X.L., Y.L.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.Y., J.D.U.); and Drug Metabolism, Gilead Sciences, Inc., Foster City, California (K.I., X.L., Y.L.)
| |
Collapse
|
17
|
Cheong EJY, Chin SY, Ng ZW, Yap TJ, Cheong EZB, Wang Z, Chan ECY. Unraveling Complexities in the Absorption and Disposition Kinetics of Abiraterone via Iterative PBPK Model Development and Refinement. Clin Pharmacokinet 2023; 62:1243-1261. [PMID: 37405634 DOI: 10.1007/s40262-023-01266-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/12/2023] [Indexed: 07/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Abiraterone is a first-in-class inhibitor of cytochrome P450 17A1 (CYP17A1), and its pharmacokinetic (PK) profile is susceptible to intrinsic and extrinsic variabilities. Potential associations between abiraterone concentrations and pharmacodynamic consequences in prostate cancer may demand further dosage optimization to balance therapeutic outcomes. Consequently, we aim to develop a physiologically based pharmacokinetic (PBPK) model for abiraterone via a middle-out approach to prospectively interrogate the untested, albeit clinically relevant, scenarios. METHODS To characterize in vivo hydrolysis of prodrug abiraterone acetate (AA) and supersaturation of abiraterone, in vitro aqueous solubility data, biorelevant measurements, and supersaturation and precipitation parameters were utilized for mechanistic absorption simulation. CYP3A4-mediated N-oxidation and sulfotransferase 2A1-catalyzed sulfation of abiraterone were subsequently quantified in human liver subcellular systems. Iterative PBPK model refinement involved evaluation of potential organic anion transporting polypeptide (OATP)-mediated abiraterone uptake in transfected cells in the absence and presence of albumin. RESULTS The developed PBPK model recapitulated the duodenal concentration-time profile of both AA and abiraterone after simulated AA administration. Our findings established abiraterone as a substrate of hepatic OATP1B3 to recapitulate its unbound metabolic intrinsic clearance. Further consideration of a transporter-induced protein-binding shift established accurate translational scaling factors and extrapolated the sinusoidal uptake process. Subsequent simulations effectively predicted the PK of abiraterone upon single and multiple dosing. CONCLUSION Our systematic development of the abiraterone PBPK model has demonstrated its application for the prospective interrogation of the individual or combined influences of potential interindividual variabilities influencing the systemic exposure of abiraterone.
Collapse
Affiliation(s)
- Eleanor Jing Yi Cheong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Sheng Yuan Chin
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Zheng Wei Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Ting Jian Yap
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Ervin Zhi Bin Cheong
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Ziteng Wang
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, 18 Science Drive 4, Singapore, 117543, Singapore.
| |
Collapse
|
18
|
Sugiyama Y, Aoki Y. A 20-Year Research Overview: Quantitative Prediction of Hepatic Clearance Using the In Vitro-In Vivo Extrapolation Approach Based on Physiologically Based Pharmacokinetic Modeling and Extended Clearance Concept. Drug Metab Dispos 2023; 51:1067-1076. [PMID: 37407092 DOI: 10.1124/dmd.123.001344] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 06/20/2023] [Accepted: 06/27/2023] [Indexed: 07/07/2023] Open
Abstract
Understanding the extended clearance concept and establishing a physiologically based pharmacokinetic (PBPK) model are crucial for investigating the impact of changes in transporter and metabolizing enzyme abundance/functions on drug pharmacokinetics in blood and tissues. This mini-review provides an overview of the extended clearance concept and a PBPK model that includes transporter-mediated uptake processes in the liver. In general, complete in vitro and in vivo extrapolation (IVIVE) poses challenges due to missing factors that bridge the gap between in vitro and in vivo systems. By considering key in vitro parameters, we can capture in vivo pharmacokinetics, a strategy known as the top-down or middle-out approach. We present the latest progress, theory, and practice of the Cluster Gauss-Newton method, which is used for middle-out analyses. As examples of poor IVIVE, we discuss "albumin-mediated hepatic uptake" and "time-dependent inhibition" of OATP1Bs. The hepatic uptake of highly plasma-bound drugs is more efficient than what can be accounted for by their unbound concentration alone. This phenomenon is referred to as "albumin-mediated" hepatic uptake. IVIVE was improved by measuring hepatic uptake clearance in vitro in the presence of physiologic albumin concentrations. Lastly, we demonstrate the application of Cluster Gauss-Newton method-based analysis to the target-mediated drug disposition of bosentan. Incorporating saturable target binding and OATP1B-mediated hepatic uptake into the PBPK model enables the consideration of nonlinear kinetics across a wide dose range and the prediction of receptor occupancy over time. SIGNIFICANCE STATEMENT: There have been multiple instances where researchers' endeavors to unravel the underlying mechanism of poor in vitro-in vivo extrapolation have led to the discovery of previously undisclosed truths. These include 1) albumin-mediated hepatic uptake, 2) the target-mediated drug disposition in small molecules, and 3) the existence of a trans-inhibition mechanism by inhibitors for OATP1B-mediated hepatic uptake of drugs. Consequently, poor in vitro-in vivo extrapolation and the subsequent inquisitiveness of scientists may serve as a pivotal gateway to uncover hidden mechanisms.
Collapse
Affiliation(s)
- Yuichi Sugiyama
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Chiyoda-ku, Tokyo, Japan (Y.A., Y.S.); ShanghaiTech University, iHuman Institute, Pudong, Shanghai, China (Y.S.); and Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Y.A.)
| | - Yasunori Aoki
- Laboratory of Quantitative System Pharmacokinetics/Pharmacodynamics, Josai International University, Chiyoda-ku, Tokyo, Japan (Y.A., Y.S.); ShanghaiTech University, iHuman Institute, Pudong, Shanghai, China (Y.S.); and Drug Metabolism and Pharmacokinetics, Research and Early Development, Cardiovascular, Renal and Metabolism, BioPharmaceuticals R&D, AstraZeneca, Gothenburg, Sweden (Y.A.)
| |
Collapse
|
19
|
Schulz JA, Stresser DM, Kalvass JC. Plasma Protein-Mediated Uptake and Contradictions to the Free Drug Hypothesis: A Critical Review. Drug Metab Rev 2023:1-34. [PMID: 36971325 DOI: 10.1080/03602532.2023.2195133] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/29/2023]
Abstract
According to the free drug hypothesis (FDH), only free, unbound drug is available to interact with biological targets. This hypothesis is the fundamental principle that continues to explain the vast majority of all pharmacokinetic and pharmacodynamic processes. Under the FDH, the free drug concentration at the target site is considered the driver of pharmacodynamic activity and pharmacokinetic processes. However, deviations from the FDH are observed in hepatic uptake and clearance predictions, where observed unbound intrinsic hepatic clearance (CLint,u) is larger than expected. Such deviations are commonly observed when plasma proteins are present and form the basis of the so-called plasma protein-mediated uptake effect (PMUE). This review will discuss the basis of plasma protein binding as it pertains to hepatic clearance based on the FDH, as well as several hypotheses that may explain the underlying mechanisms of PMUE. Notably, some, but not all, potential mechanisms remained aligned with the FDH. Finally, we will outline possible experimental strategies to elucidate PMUE mechanisms. Understanding the mechanisms of PMUE and its potential contribution to clearance underprediction is vital to improving the drug development process.
Collapse
|
20
|
Plasma Protein Binding Refinement of the Extended Clearance Classification System: Subclasses for Predicting Hepatic Uptake or Renal Clearance for Classes 1B and 3B. Eur J Drug Metab Pharmacokinet 2023; 48:63-73. [PMID: 36441468 DOI: 10.1007/s13318-022-00806-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/02/2022] [Indexed: 11/29/2022]
Abstract
BACKGROUND AND OBJECTIVES The Extended Clearance Classification System (ECCS) was established to facilitate the timely anticipation of clearance rate determination according to the physicochemical characteristics of a given compound and in vitro passive membrane permeability. Unfortunately, distinguishing between renal and hepatic uptake clearance mechanisms using ECCS class 3B is not possible. We determined the effects of plasma protein binding (PPB) on major hepatic organic anion transporting polypeptide (OATP) and renal organic anion transporter (OAT) substrates. A modified ECCS could predict when renal or hepatic uptake mechanisms were the main clearance rate determinants (accounting for ≥ 70% of total clearance). METHODS A dataset of 66 human OATP and 41 OAT substrates was analyzed to determine the effect of PPB. A total of 63 acidic and zwitterionic, and high-molecular-weight (MW > 400 Da) compounds, including 50 drugs in ECCS classes 1B and 3B, were reanalyzed considering their PPB. RESULTS Statistical analyses revealed that hepatic uptake transporter (OATP1B1 and OATP1B3) substrates possess a high PPB rate of ≥ 90%, whereas OAT1 and/or OAT3 substrates possess low PPB rates of < 90%. By analyzing the 63 drugs on the basis of their PPB, the active hepatic uptakes of acids and zwitterions were determined to be the main clearance mechanisms, with PPB ≥ 90%, whereas renally eliminated drugs exhibited limited PPB (< 90%). CONCLUSIONS Therefore, PPB is an effective parameter for defining clearance rate determination for acidic and zwitterionic drugs with high MWs. Using PPB as an additional parameter in ECCS, clearance mechanisms for class 1B and 3B compounds can be predicted, and OATP and OAT substrates may be readily distinguished.
Collapse
|
21
|
The next frontier in ADME science: Predicting transporter-based drug disposition, tissue concentrations and drug-drug interactions in humans. Pharmacol Ther 2022; 238:108271. [DOI: 10.1016/j.pharmthera.2022.108271] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2022] [Revised: 08/05/2022] [Accepted: 08/17/2022] [Indexed: 12/25/2022]
|
22
|
Evidence of the Need for Modified Well-stirred Model in In Vitro to In Vivo Extrapolation. Eur J Pharm Sci 2022; 177:106268. [PMID: 35901930 DOI: 10.1016/j.ejps.2022.106268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Accepted: 07/01/2022] [Indexed: 11/20/2022]
Abstract
In vitro to in vivo extrapolation (IVIVE), an approach for hepatic clearance (CLH) prediction used worldwide, remains controversial due to systematic underprediction. Among the various probable factors, the original assumption of the hepatic mathematical model (i.e., the well-stirred model, WSM) may become problematic, leading to the underestimation of drug CLH. Having a similar prerequisite that the well-stirred conditions are homogenous with perfectly mixed reactants, but using a different driving concentration, the modified well-stirred model (MWSM) stands apart from the WSM. However, we believe that both models should coexist so that the entire well-stirred scenario can be completely illustrated. Consequently, we collected published data from the literature and employed a logistic regression method to differentiate the optimal timing of use between WSM and MWSM in drug CLH prediction. Generally, variances adopted in the regression, including partition coefficient (logP), fraction unbound (fu), volumes of distribution at steady-state (Vss), and mean residence time (MRT), corresponded to our assumption when protein-facilitated uptake was considered. Furthermore, a new empirical approach was introduced to allow practical use of the MWSM. The results showed that this model could provide a more precise prediction compared to previous empirical approaches. Therefore, these preliminary results not only delineated a more detailed structure and mechanism of MWSM but also highlighted its necessity and potential.
Collapse
|
23
|
Jones RS, Leung C, Chang JH, Brown S, Liu N, Yan Z, Kenny JR, Broccatelli F. Application of empirical scalars to enable early prediction of human hepatic clearance using IVIVE in drug discovery: an evaluation of 173 drugs. Drug Metab Dispos 2022; 50:DMD-AR-2021-000784. [PMID: 35636770 DOI: 10.1124/dmd.121.000784] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 04/20/2022] [Accepted: 05/12/2022] [Indexed: 11/22/2022] Open
Abstract
The utilization of in vitro data to predict drug pharmacokinetics (PK) in vivo has been a consistent practice in early drug discovery for decades. However, its success is hampered by mispredictions attributed to uncharacterized biological phenomena/experimental artifacts. Predicted drug clearance (CL) from experimental data (i.e. hepatocyte intrinsic clearance: CLint, fraction unbound in plasma: fu,p) is often systematically underpredicted using the well-stirred model (WSM). The objective of this study was to evaluate using empirical scalars in the WSM to correct for CL mispredictions. Drugs (N=28) were used to generate numerical scalars on CLint (α), and fu,p (β) to minimize the error (AAFE) for CL predictions. These scalars were validated using an additional dataset (N=28 drugs) and applied to a non-redundant AstraZeneca (AZ) dataset available in the literature (N=117 drugs) for a total of 173 compounds. CL predictions using the WSM were improved for most compounds using an α value of 3.66 (~64%<2-fold) compared to no scaling (~46%<2-fold). Similarly, using a β value of 0.55 or combination of α and β scalars (values of 1.74 and 0.66, respectively) resulted in a similar improvement in predictions (~64%<2-fold and ~65%<2-fold, respectively). For highly bound compounds (fu,p{less than or equal to}0.01), AAFE was substantially reduced across all scaling methods. Using the β scalar alone or a combination of α and β appeared optimal; and produce larger magnitude corrections for highly-bound compounds. Some drugs are still disproportionally mispredicted, however the improvements in prediction error and simplicity of applying these scalars suggests its utility for early-stage CL predictions. Significance Statement In early drug discovery, prediction of human clearance using in vitro experimental data plays an essential role in triaging compounds prior to in vivo studies. These predictions have been systematically underestimated. Here we introduce empirical scalars calibrated on the extent of plasma protein binding that appear to improve clearance prediction across multiple datasets. This approach can be used in early phases of drug discovery prior to the availability of pre-clinical data for early quantitative predictions of human clearance.
Collapse
Affiliation(s)
| | | | - Jae H Chang
- Preclinical Development Sciences, ORIC Pharmaceuticals, United States
| | | | | | | | - Jane R Kenny
- Drug Metabolism & Pharmacokinetics, Genentech Inc, United States
| | | |
Collapse
|
24
|
Kameyama T, Sodhi JK, Benet LZ. Does Addition of Protein to Hepatocyte or Microsomal In Vitro Incubations Provide a Useful Improvement in In Vitro-In Vivo Extrapolation Predictability? Drug Metab Dispos 2022; 50:401-412. [PMID: 35086847 PMCID: PMC11022888 DOI: 10.1124/dmd.121.000677] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Accepted: 01/21/2022] [Indexed: 11/22/2022] Open
Abstract
Accurate prediction of in vivo hepatic clearance is an essential part of successful and efficient drug development; however, many investigators have recognized that there are significant limitations in the predictability of clearance with a tendency for underprediction for primarily metabolized drugs. Here, we examine the impact of adding serum or albumin into hepatocyte and microsomal incubations on the predictability of in vivo hepatic clearance. The addition of protein into hepatocyte incubations has been reported to improve the predictability for high clearance (extraction ratio) drugs and highly protein-bound drugs. Analyzing published data for 60 different drugs and 97 experimental comparisons (with 17 drugs being investigated from two to seven) we confirmed the marked underprediction of clearance. However, we could not validate any relevant improved predictability within twofold by the addition of serum to hepatocyte incubations or albumin to microsomal incubations. This was the case when investigating all measurements, or when subdividing analyses by extraction ratio, degree of protein binding, Biopharmaceutics Drug Disposition Classification System class, examining Extended Clearance Classification System class 1B drugs only, or drug charge. Manipulating characteristics of small data sets of like compounds and adding scaling factors can appear to yield good predictability, but the carryover of these methods to alternate drug classes and different laboratories is not evident. Improvement in predictability of poorly soluble compounds is greater than that for soluble compounds, but not to a meaningful extent. Overall, we cannot confirm that protein addition improves in vitro-in vivo extrapolation predictability to any clinically meaningful degree when considering all drugs and different subsets. SIGNIFICANCE STATEMENT: The addition of protein into microsomal or hepatocyte incubations has been widely proposed to improve hepatic clearance predictions. To date, studies examining this phenomenon have not included appropriate negative controls where predictability is achieved without protein addition and have been conducted with small data sets of similar compounds that don't apply to alternate drug classes. Here, an extensive analysis of published data for 60 drugs and 97 experimental comparisons couldn't validate any relevant clinically improved clearance predictability with protein addition.
Collapse
Affiliation(s)
- Tsubasa Kameyama
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| | - Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California
| |
Collapse
|
25
|
Yin M, Storelli F, Unadkat JD. Is The Protein-Mediated Uptake Of Drugs By OATPs A Real Phenomenon Or An Artifact?. Drug Metab Dispos 2022; 50:1132-1141. [PMID: 35351775 DOI: 10.1124/dmd.122.000849] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Accepted: 03/24/2022] [Indexed: 11/22/2022] Open
Abstract
Plasma proteins or human serum albumin (HSA) have been reported to increase the in vitro intrinsic uptake clearance (CLint,uptake) of drugs by hepatocytes or organic anion transporting polypeptide (OATP)-transfected cell lines. This, so called protein-mediated uptake effect (PMUE), is thought to be due to an interaction between the drug-protein complex and the cell membrane causing an increase in the unbound drug concentration at the cell surface resulting in an increase in the apparent CLint,uptake of the drug. To determine if the PMUE on OATP-mediated drug uptake is an artifact or a real phenomenon, we determined the effect of 1%, 2% and 5% HSA on OATP1B1-mediated (HEK293 transfected cells) and passive CLint,uptake (MOCK HEK293 cells) of a cocktail of five statins. In addition, we determined the non-specific binding (NSB) of the statin-HSA complex to the cells/labware. The increase in uptake of atorvastatin, fluvastatin and rosuvastatin in the presence of HSA was completely explained by the extent of NSB of the statin-HSA complex, indicating that the PMUE for these statins is an artifact. In contrast, this was not the case for OATP1B1-mediated uptake of pitavastatin and passive uptake of cerivastatin suggesting that the PMUE is a real phenomenon for these drugs. Additionally, the PMUE on OATP1B1-mediated uptake of pitavastatin was confirmed by a decrease in its unbound IC50 in the presence of 5% HSA vs. HBSS buffer. These data question the utility of routinely including plasma proteins or HSA in uptake experiments and the previous findings on PMUE on OATP-mediated drug uptake. Significance Statement Here we report, for the first time, that the protein-mediated uptake effect (PMUE) on OATP-transported drugs could be an artifact of the non-specific binding (NSB) of the drug-albumin complex to cells/labware. Future experiments on PMUE must take into consideration such NSB. In addition, mechanisms other than PMUE need to be explored to explain the underprediction of in vivo OATP-mediated hepatic drug CL from in vitro uptake studies.
Collapse
|
26
|
Cheong EJY, Ng DZW, Chin SY, Wang Z, Chan ECY. Application of a PBPK Model of Rivaroxaban to Prospective Simulations of Drug-Drug-Disease Interactions with Protein Kinase Inhibitors in CA-VTE. Br J Clin Pharmacol 2021; 88:2267-2283. [PMID: 34837258 DOI: 10.1111/bcp.15158] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2021] [Revised: 10/24/2021] [Accepted: 11/08/2021] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND AND PURPOSE Rivaroxaban is a viable anticoagulant for the management of cancer associated venous thromboembolism (CA-VTE). A previously verified physiologically-based pharmacokinetic (PBPK) model of rivaroxaban established how its multiple pathways of elimination via both CYP3A4/2J2-mediated hepatic metabolism and organic anion transporter 3 (OAT3)/P-glycoprotein-mediated renal secretion predisposes rivaroxaban to drug-drug-disease interactions (DDDIs) with clinically relevant protein kinase inhibitors (PKIs). We proposed the application of PBPK modelling to prospectively interrogate clinically significant DDIs between rivaroxaban and PKIs (erlotinib and nilotinib) for dose adjustments in CA-VTE. EXPERIMENTAL APPROACH The inhibitory potencies of the PKIs on CYP3A4/2J2-mediated metabolism of rivaroxaban were characterized. Using prototypical OAT3 inhibitor ketoconazole, in vitro OAT3 inhibition assays were optimized to ascertain the in vivo relevance of derived transport inhibitory constants (Ki ). Untested DDDIs between rivaroxaban and erlotinib or nilotinib were simulated. KEY RESULTS Mechanism-based inactivation (MBI) of CYP3A4-mediated rivaroxaban metabolism by both PKIs and MBI of CYP2J2 by erlotinib were established. The importance of substrate specificity and nonspecific binding to derive OAT3-inhibitory Ki values of ketoconazole and nilotinib for the accurate prediction of interactions was illustrated. When simulated rivaroxaban exposure variations with concomitant erlotinib and nilotinib therapy were evaluated using published dose-exposure equivalence metrics and bleeding risk analyses, dose reductions from 20 mg to 15 mg and 10 mg in normal and mild renal dysfunction, respectively, were warranted. CONCLUSION AND IMPLICATIONS We established a PBPK-DDDI model to prospectively evaluate clinically relevant interactions between rivaroxaban and PKIs for the safe and efficacious management of CA-VTE.
Collapse
Affiliation(s)
- Eleanor Jing Yi Cheong
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Daniel Zhi Wei Ng
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Sheng Yuan Chin
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Ziteng Wang
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| | - Eric Chun Yong Chan
- Department of Pharmacy, Faculty of Science, National University of Singapore, Singapore, Singapore
| |
Collapse
|
27
|
Chothe PP, Nakakariya M, Rotter CJ, Sandoval P, Tohyama K. Recent Advances in Drug Transporter Sciences: Highlights From the Year 2020. Drug Metab Rev 2021; 53:321-349. [PMID: 34346798 DOI: 10.1080/03602532.2021.1963270] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
Drug Metabolism Reviews has an impressive track record of providing scientific reviews in the area of xenobiotic biotransformation over 47 years. It has consistently proved to be resourceful to many scientists from pharmaceutical industry, academia, regulatory agencies working in diverse areas including enzymology, pharmacology, pharmacokinetics and toxicology. Over the last 5 years Drug metabolism Reviews has annually published an industry commentary aimed to highlight novel insights and approaches that have made significant impacts on the field of biotransformation (led by Cyrus Khojasteh). We hope to continue this tradition by providing an overview of advances made in the field of drug transporters during 2020. The field of drug transporters is rapidly evolving as they play an essential role in drug absorption, distribution, clearance and elimination. In this review we have selected outstanding drug transporter articles that have significantly contributed to moving forward the field of transporter science with respect to translation and improved understanding of diverse aspects including uptake clearance, clinical biomarkers, induction, proteomics, emerging transporters and tissue targeting.The theme of this review consists of synopsis that summarizes each article followed by our commentary. The objective of this work is not to provide a comprehensive review but rather exemplify novel insights and state-of-the-art highlights of recent research that have advanced our understanding of drug transporters in drug disposition. We are hopeful that this effort will prove useful to the scientific community and as such request feedback, and further extend an invitation to anyone interested in contributing to future reviews.
Collapse
Affiliation(s)
- Paresh P Chothe
- Global Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| | - Masanori Nakakariya
- Drug Metabolism and Pharmacokinetics Research Laboratories, Takeda Pharmaceutical Company Limited, 26-1, Muraoka-Higashi 2-Chrome, Fujisawa, Kanagawa, 251-8555, Japan
| | - Charles J Rotter
- Global Drug Metabolism and Pharmacokinetics, Takeda California Incorporated, 9625 Towne Centre Drive, San Diego, California, 92121, USA
| | - Philip Sandoval
- Global Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| | - Kimio Tohyama
- Global Drug Metabolism and Pharmacokinetics, Takeda Pharmaceutical Company Limited, 35 Landsdowne Street, Cambridge, Massachusetts, 02139, USA
| |
Collapse
|
28
|
Miyauchi S, Kim SJ, Lee W, Sugiyama Y. Consideration of albumin-mediated hepatic uptake for highly protein-bound anionic drugs: Bridging the gap of hepatic uptake clearance between in vitro and in vivo. Pharmacol Ther 2021; 229:107938. [PMID: 34171335 DOI: 10.1016/j.pharmthera.2021.107938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Accepted: 03/22/2021] [Indexed: 10/21/2022]
Abstract
The accuracy in predicting in vivo hepatic clearance of drugs from in vitro data (often termed as in vitro-to-in vivo extrapolation, IVIVE) has improved in part by applying the extended-clearance concept that considers the interplay between hepatic metabolism and uptake/efflux processes. However, the IVIVE-based prediction performs poorly in predicting the hepatic uptake clearance of highly albumin-bound anionic drugs. Their hepatic uptake clearances tend to be much higher than expected based on the free-drug theory. Such observation can be attributable to a phenomenon called albumin-mediated hepatic uptake, for which various models have been thus far proposed. Our group has been applying a facilitated-dissociation model, which assumes the enhanced dissociation of the drug-albumin complex upon interaction with the cell surface. By considering the albumin-mediated hepatic uptake (using the facilitated-dissociation model or alternative kinetic models), a number of investigations demonstrated the improvement in the prediction accuracy for the hepatic clearance of highly protein-bound anionic drugs that are substrates for hepatic uptake transporters. This review summarizes the reported kinetic analyses of the albumin-mediated hepatic uptake of highly albumin-bound drugs concerning the IVIVE and the clinical and physiological relevance.
Collapse
Affiliation(s)
- Seiji Miyauchi
- Faculty of Pharmaceutical Sciences, Toho University, 2-2-1 Miyama, Funabashi, Chiba, Japan
| | - Soo-Jin Kim
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, Technology and Innovation Hub, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama, Kanagawa 230-0045, Japan.
| |
Collapse
|
29
|
Yadav J, El Hassani M, Sodhi J, Lauschke VM, Hartman JH, Russell LE. Recent developments in in vitro and in vivo models for improved translation of preclinical pharmacokinetics and pharmacodynamics data. Drug Metab Rev 2021; 53:207-233. [PMID: 33989099 DOI: 10.1080/03602532.2021.1922435] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Improved pharmacokinetics/pharmacodynamics (PK/PD) prediction in the early stages of drug development is essential to inform lead optimization strategies and reduce attrition rates. Recently, there have been significant advancements in the development of new in vitro and in vivo strategies to better characterize pharmacokinetic properties and efficacy of drug leads. Herein, we review advances in experimental and mathematical models for clearance predictions, advancements in developing novel tools to capture slowly metabolized drugs, in vivo model developments to capture human etiology for supporting drug development, limitations and gaps in these efforts, and a perspective on the future in the field.
Collapse
Affiliation(s)
- Jaydeep Yadav
- Department of Pharmacokinetics, Pharmacodynamics, and Drug Metabolism, Merck & Co., Inc., Boston, MA, USA
| | | | - Jasleen Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, CA, USA
| | - Volker M Lauschke
- Department of Physiology and Pharmacology, Karolinska Institutet, Stockholm, Sweden
| | - Jessica H Hartman
- Department of Biochemistry and Molecular Biology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
30
|
Sodhi JK, Benet LZ. Successful and Unsuccessful Prediction of Human Hepatic Clearance for Lead Optimization. J Med Chem 2021; 64:3546-3559. [PMID: 33765384 PMCID: PMC8504179 DOI: 10.1021/acs.jmedchem.0c01930] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Development of new chemical entities is costly, time-consuming, and has a low success rate. Accurate prediction of pharmacokinetic properties is critical to progress compounds with favorable drug-like characteristics in lead optimization. Of particular importance is the prediction of hepatic clearance, which determines drug exposure and contributes to projection of dose, half-life, and bioavailability. The most commonly employed methodology to predict hepatic clearance is termed in vitro to in vivo extrapolation (IVIVE) that involves measuring drug metabolism in vitro, scaling-up this in vitro intrinsic clearance to a prediction of in vivo intrinsic clearance by reconciling the enzymatic content between the incubation and an average human liver, and applying a model of hepatic disposition to account for limitations of protein binding and blood flow to predict in vivo clearance. This manuscript reviews common in vitro techniques used to predict hepatic clearance as well as current challenges and recent theoretical advancements in IVIVE.
Collapse
Affiliation(s)
- Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94143, United States
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California 94143, United States
| |
Collapse
|
31
|
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
|
32
|
Investigation of the arcane inhibition of human organic anion transporter 3 by benzofuran antiarrhythmic agents. Drug Metab Pharmacokinet 2021; 38:100390. [PMID: 33836300 DOI: 10.1016/j.dmpk.2021.100390] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Revised: 02/08/2021] [Accepted: 03/07/2021] [Indexed: 12/16/2022]
Abstract
The combination of antiarrhythmic agents, amiodarone or dronedarone, with the anticoagulant rivaroxaban is used clinically in the management of atrial fibrillation for rhythm control and secondary stroke prevention respectively. Renal drug-drug interactions (DDIs) between amiodarone or dronedarone and rivaroxaban were previously ascribed to inhibition of rivaroxaban secretion by P-glycoprotein at the apical membrane of renal proximal tubular epithelial cells. Benzbromarone, a known inhibitor of organic anion transporter 3 (OAT3), shares a benzofuran scaffold with amiodarone and dronedarone. However, inhibitory activity of amiodarone and dronedarone against OAT3 remains arcane. Here, we conducted in vitro transporter inhibition assays in OAT3-transfected HEK293 cells which revealed amiodarone, dronedarone and their respective major pharmacologically-active metabolites N-desethylamiodarone and N-desbutyldronedarone possess inhibitory activity against OAT3, with corrected Ki values of 0.042, 0.019, 0.028 and 0.0046 μM respectively. Protein binding effects and probe substrate dependency were accounted for in our assays. Static modelling predicted 1.29-, 1.01-, 1.29- and 1.16-fold increase in rivaroxaban exposure, culminating in a predicted 1.29-, 1.01-, 1.28- and 1.15-fold increase in major bleeding risk respectively, suggesting potential OAT3-mediated DDI between amiodarone and rivaroxaban. Future work involving physiologically-based pharmacokinetic modelling is crucial in holistically predicting the complex DDIs between the benzofuran antiarrhythmic agents and rivaroxaban.
Collapse
|
33
|
Eng H, Bi YA, West MA, Ryu S, Yamaguchi E, Kosa RE, Tess DA, Griffith DA, Litchfield J, Kalgutkar AS, Varma MVS. Organic Anion-Transporting Polypeptide 1B1/1B3-Mediated Hepatic Uptake Determines the Pharmacokinetics of Large Lipophilic Acids: In Vitro-In Vivo Evaluation in Cynomolgus Monkey. J Pharmacol Exp Ther 2021; 377:169-180. [PMID: 33509903 DOI: 10.1124/jpet.120.000457] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 01/25/2021] [Indexed: 12/22/2022] Open
Abstract
It is generally presumed that uptake transport mechanisms are of limited significance in hepatic clearance for lipophilic or high passive-permeability drugs. In this study, we evaluated the mechanistic role of the hepato-selective organic anion-transporting polypeptides (OATPs) 1B1/1B3 in the pharmacokinetics of compounds representing large lipophilic acid space. Intravenous pharmacokinetics of 16 compounds with molecular mass ∼400-730 Da, logP ∼3.5-8, and acid pKa <6 were obtained in cynomolgus monkey after dosing without and with a single-dose rifampicin-OATP1B1/1B3 probe inhibitor. Rifampicin (30 mg/kg oral) significantly (P < 0.05) reduced monkey clearance and/or steady-state volume of distribution (VDss) for 15 of 16 acids evaluated. Additionally, clearance of danoprevir was reduced by about 35%, although statistical significance was not reached. A significant linear relationship was noted between the clearance ratio (i.e., ratio of control to treatment groups) and VDss ratio, suggesting hepatic uptake contributes to the systemic clearance and distribution simultaneously. In vitro transport studies using primary monkey and human hepatocytes showed uptake inhibition by rifampicin (100 µM) for compounds with logP ≤6.5 but not for the very lipophilic acids (logP > 6.5), which generally showed high nonspecific binding in hepatocyte incubations. In vitro uptake clearance and fraction transported by OATP1B1/1B3 (ft,OATP1B) were found to be similar in monkey and human hepatocytes. Finally, for compounds with logP ≤6.5, good agreement was noted between in vitro ft,OATP1B and clearance ratio (as well as VDss ratio) in cynomolgus monkey. In conclusion, this study provides mechanistic evidence for the pivotal role of OATP1B-mediated hepatic uptake in the pharmacokinetics across a wide, large lipophilic acid space. SIGNIFICANCE STATEMENT: This study provides mechanistic insight into the pharmacokinetics of a broad range of large lipophilic acids. Organic anion-transporting polypeptides 1B1/1B3-mediated hepatic uptake is of key importance in the pharmacokinetics and drug-drug interactions of almost all drugs and new molecular entities in this space. Diligent in vitro and in vivo transport characterization is needed to avoid the false negatives often noted because of general limitations in the in vitro assays while handling compounds with such physicochemical attributes.
Collapse
Affiliation(s)
- Heather Eng
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Yi-An Bi
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Mark A West
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Sangwoo Ryu
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Emi Yamaguchi
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Rachel E Kosa
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - David A Tess
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - David A Griffith
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - John Litchfield
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Amit S Kalgutkar
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| | - Manthena V S Varma
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, Connecticut (H.E., Y.B., M.A.W., S.R., E.Y., R.E.K., M.V.S.V.), and PDM (D.A.T., J.L., A.S.K.) and Medicinal Chemistry, Medicine Design, Worldwide Research and Development (D.A.G.), Pfizer Inc., Cambridge, Massachusetts
| |
Collapse
|
34
|
Izat N, Sahin S. Hepatic transporter-mediated pharmacokinetic drug-drug interactions: Recent studies and regulatory recommendations. Biopharm Drug Dispos 2021; 42:45-77. [PMID: 33507532 DOI: 10.1002/bdd.2262] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 12/16/2020] [Accepted: 01/13/2021] [Indexed: 12/13/2022]
Abstract
Transporter-mediated drug-drug interactions are one of the major mechanisms in pharmacokinetic-based drug interactions and correspondingly affecting drugs' safety and efficacy. Regulatory bodies underlined the importance of the evaluation of transporter-mediated interactions as a part of the drug development process. The liver is responsible for the elimination of a wide range of endogenous and exogenous compounds via metabolism and biliary excretion. Therefore, hepatic uptake transporters, expressed on the sinusoidal membranes of hepatocytes, and efflux transporters mediating the transport from hepatocytes to the bile are determinant factors for pharmacokinetics of drugs, and hence, drug-drug interactions. In parallel with the growing research interest in this area, regulatory guidances have been updated with detailed assay models and criteria. According to well-established preclinical results, observed or expected hepatic transporter-mediated drug-drug interactions can be taken into account for clinical studies. In this paper, various methods including in vitro, in situ, in vivo, in silico approaches, and combinational concepts and several clinical studies on the assessment of transporter-mediated drug-drug interactions were reviewed. Informative and effective evaluation by preclinical tools together with the integration of pharmacokinetic modeling and simulation can reduce unexpected clinical outcomes and enhance the success rate in drug development.
Collapse
Affiliation(s)
- Nihan Izat
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - Selma Sahin
- Department of Pharmaceutical Technology, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| |
Collapse
|
35
|
Abstract
Accurate estimation of in vivo clearance in human is pivotal to determine the dose and dosing regimen for drug development. In vitro-in vivo extrapolation (IVIVE) has been performed to predict drug clearance using empirical and physiological scalars. Multiple in vitro systems and mathematical modeling techniques have been employed to estimate in vivo clearance. The models for predicting clearance have significantly improved and have evolved to become more complex by integrating multiple processes such as drug metabolism and transport as well as passive diffusion. This chapter covers the use of conventional as well as recently developed methods to predict metabolic and transporter-mediated clearance along with the advantages and disadvantages of using these methods and the associated experimental considerations. The general approaches to improve IVIVE by use of appropriate scalars, incorporation of extrahepatic metabolism and transport and application of physiologically based pharmacokinetic (PBPK) models with proteomics data are also discussed. The chapter also provides an overview of the advantages of using such dynamic mechanistic models over static models for clearance predictions to improve IVIVE.
Collapse
|
36
|
Francis LJ, Houston JB, Hallifax D. Impact of Plasma Protein Binding in Drug Clearance Prediction: A Data Base Analysis of Published Studies and Implications for In Vitro-In Vivo Extrapolation. Drug Metab Dispos 2020; 49:188-201. [PMID: 33355201 DOI: 10.1124/dmd.120.000294] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 12/07/2020] [Indexed: 11/22/2022] Open
Abstract
Plasma protein-mediated uptake (PMU) and its effect on clearance (CL) prediction have been studied in various formats; however, a comprehensive analysis of the overall impact of PMU on CL parameters from hepatocyte assays (routinely used for IVIVE) has not previously been performed. The following work collated data reflecting the effect of PMU for 26 compounds with a wide variety of physicochemical, drug, and in vivo CL properties. PMU enhanced the unbound intrinsic clearance in vitro (CLint,u in vitro) beyond that conventionally calculated using fraction unbound and was correlated with the unbound fraction of drug in vitro and in plasma (fup) and absolute unbound intrinsic clearance in vivo (CLint,u in vivo) in both rat and human hepatocytes. PMU appeared to be more important for highly bound (fup < 0.1) and high CLint,u in vivo drugs. These trends were independent of species, assay conditions, ionization, and extended clearance classification system group, although the type of plasma protein used in in vitro assays may require further investigation. Such generalized trends (spanning fup 0.0008-0.99) may suggest a generic mechanism behind PMU; however, multiple drug-dependent mechanisms are also possible. Using the identified relationship between the impact of PMU on CLint,u in vitro and fup, PMU-enhanced predictions of CLint,u in vivo were calculated for both transporter substrates and metabolically cleared drugs. PMU was accurately predicted, and incorporation of predicted PMU improved the IVIVE of hepatic CL, with an average fold error of 1.17 and >50% of compounds predicted within a 2-fold error for both rat and human data sets (n ≥ 100). SIGNIFICANCE STATEMENT: Current strategies for prediction of hepatic clearance from in vitro data are recognized to be inaccurate, but they do not account for PMU. The impact of PMU on CLint,u in vitro is wide ranging and can be predicted based on fraction unbound in plasma and applied to CLint,u in vitro values obtained by standard procedures in the absence of plasma protein. Such PMU-enhanced predictions improved IVIVE, and future studies may easily incorporate this PMU relationship to provide more accurate IVIVE.
Collapse
Affiliation(s)
- L J Francis
- 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 B 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
| | - D 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
|
37
|
Comparative Assessment of Extrapolation Methods Based on the Conventional Free Drug Hypothesis and Plasma Protein-Mediated Hepatic Uptake Theory for the Hepatic Clearance Predictions of Two Drugs Extensively Bound to Both the Albumin And Alpha-1-Acid Glycoprotein. J Pharm Sci 2020; 110:1385-1391. [PMID: 33217427 DOI: 10.1016/j.xphs.2020.11.009] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/10/2020] [Accepted: 11/10/2020] [Indexed: 11/22/2022]
Abstract
Bteich and coworkers recently demonstrated in a companion manuscript (J Pharm Sci 109: https://doi.org/10.1016/j.xphs.2020.07.003) that a protein-mediated hepatic uptake have occurred in an isolated perfused rat liver (IPRL) model for two drugs (Perampanel; PER and Fluoxetine; FLU) that bind extensively to the albumin (ALB) and alpha-1-acid glycoprotein (AGP). However, to our knowledge, there is no quantitative model available to predict the impact of a plasma protein-mediated hepatic uptake on the extent of hepatic clearance (CLh) for a drug binding extensively to these two proteins. Therefore, the main objective was to predict the corresponding CLh, which is an extension of the companion manuscript. The method consisted of extrapolating the intrinsic clearance from the unbound fraction measured in the perfusate or the unbound fraction extrapolated to the surface of the hepatocyte membrane by adapting an existing model of protein-mediated hepatic uptake (i.e., the fup-adjusted model) to include a binding ratio between the ALB and AGP. This new approach showed a relevant improvement compared to the free drug hypothesis particularly for FLU that showed the highest degree of ALB-mediated uptake. Overall, this study is a first step towards the development of predictive methods of CLh by considering the binding to ALB and AGP.
Collapse
|
38
|
Li N, Badrinarayanan A, Ishida K, Li X, Roberts J, Wang S, Hayashi M, Gupta A. Albumin-Mediated Uptake Improves Human Clearance Prediction for Hepatic Uptake Transporter Substrates Aiding a Mechanistic In Vitro-In Vivo Extrapolation (IVIVE) Strategy in Discovery Research. AAPS JOURNAL 2020; 23:1. [DOI: 10.1208/s12248-020-00528-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2020] [Accepted: 10/16/2020] [Indexed: 01/09/2023]
|
39
|
Bi YA, Ryu S, Tess DA, Rodrigues AD, Varma MVS. Effect of Human Plasma on Hepatic Uptake of Organic Anion–Transporting Polypeptide 1B Substrates: Studies Using Transfected Cells and Primary Human Hepatocytes. Drug Metab Dispos 2020; 49:72-83. [DOI: 10.1124/dmd.120.000134] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2020] [Accepted: 10/19/2020] [Indexed: 12/27/2022] Open
|
40
|
Park JE, Shitara Y, Lee W, Morita S, Sahi J, Toshimoto K, Sugiyama Y. Improved Prediction of the Drug-Drug Interactions of Pemafibrate Caused by Cyclosporine A and Rifampicin via PBPK Modeling: Consideration of the Albumin-Mediated Hepatic Uptake of Pemafibrate and Inhibition Constants With Preincubation Against OATP1B. J Pharm Sci 2020; 110:517-528. [PMID: 33058894 DOI: 10.1016/j.xphs.2020.10.016] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/27/2020] [Accepted: 10/07/2020] [Indexed: 11/17/2022]
Abstract
Pemafibrate (PMF) is highly albumin-bound (>99.8%) and a substrate for hepatic uptake transporters (OATP1B) and CYP enzymes. Here, we developed a PBPK model of PMF to capture drug-drug interactions (DDI) incurred by cyclosporine (CsA) and rifampicin (RIF), the two OATP1B inhibitors. Initial PBPK modeling of PMF utilized in vitro hepatic uptake clearance (PSinf) obtained in the absence of albumin, but failed in capturing the blood PMF pharmacokinetic (PK) profiles. Based on the results that in vitro PSinf of unbound PMF was enhanced in the presence of albumin, we applied the albumin-facilitated dissociation model and the resulting PSinf parameters improved the prediction of the blood PMF PK profiles. In refining our PBPK model toward improved prediction of the observed DDI data (PMF co-administered with single dosing of CsA or RIF; PMF following multiple RIF dosing), we adjusted the previously obtained in vivo OATP1B inhibition constants (Ki,OATP1B) of CsA or RIF for pitavastatin by correcting for substrate-dependency. We also incorporated the induction of OATP1B and CYP enzymes after multiple RIF dosing. Sensitivity analysis informed that the higher gastrointestinal absorption rate constant could further improve capturing the observed DDI data, suggesting the possible inhibition of intestinal ABC transporter(s) by CsA or RIF.
Collapse
Affiliation(s)
- Ji Eun Park
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan; Pharmacokinetics, Dynamics and Metabolism, Translational Medicine and Early Development, R&D, Sanofi K.K., 3 Chome-20-2, Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Yoshihisa Shitara
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine and Early Development, R&D, Sanofi K.K., 3 Chome-20-2, Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Wooin Lee
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Bldg 21 Rm 309, 1 Gwanak-ro, Gwanak-gu, Seoul 08826, S. Korea
| | - Shigemichi Morita
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine and Early Development, R&D, Sanofi K.K., 3 Chome-20-2, Nishishinjuku, Shinjuku-ku, Tokyo, 160-0023, Japan
| | - Jasminder Sahi
- Pharmacokinetics, Dynamics and Metabolism, Translational Medicine and Early Development, R&D, Sanofi China, 1228 Yan'an Middle Road, Jing'an District, Shanghai, China
| | - Kota Toshimoto
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan
| | - Yuichi Sugiyama
- Sugiyama Laboratory, RIKEN Baton Zone Program, RIKEN Cluster for Science, RIKEN, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama 230-0045, Japan.
| |
Collapse
|
41
|
Kumar V, Yin M, Ishida K, Salphati L, Hop CECA, Rowbottom C, Xiao G, Lai Y, Mathias A, Chu X, Humphreys WG, Liao M, Nerada Z, Szilvásy N, Heyward S, Unadkat JD. Prediction of Transporter-Mediated Rosuvastatin Hepatic Uptake Clearance and Drug Interaction in Humans Using Proteomics-Informed REF Approach. Drug Metab Dispos 2020; 49:159-168. [PMID: 33051248 DOI: 10.1124/dmd.120.000204] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2020] [Accepted: 09/24/2020] [Indexed: 01/08/2023] Open
Abstract
Suspended, plated, or sandwich-cultured human hepatocytes are routinely used for in vitro to in vivo extrapolation (IVIVE) of transporter-mediated hepatic clearance (CL) of drugs. However, these hepatocyte models have been reported to underpredict transporter-mediated in vivo hepatic uptake CL (CL uptake,in vivo ) of some drugs. Therefore, we determined whether transporter-expressing cells (TECs) can accurately predict the CL uptake,in vivo of drugs. To do so, we determined the uptake CL (CL int,uptake,cells ) of rosuvastatin (RSV) by TECs (organic anion transporting polypeptides/Na+-taurocholate cotransporting polypeptide) and then scaled it to that in vivo by relative expression factor (REF) (the ratio of transporter abundance in human livers and TEC) determined by liquid chromatography tandem mass spectrometry-based quantitative proteomics. Both the TEC and hepatocyte models did not meet our predefined success criteria of predicting within 2-fold the RSV CL uptake,in vivo value obtained from our positron emission tomography (PET) imaging. However, the TEC performed better than the hepatocyte models. Interestingly, using REF, TECs successfully predicted RSV CL int,uptake,hep obtained by the hepatocyte models, suggesting that the underprediction of RSV CL uptake,in vivo by TECs and hepatocytes is due to endogenous factor(s) not present in these in vitro models. Therefore, we determined whether inclusion of plasma (or albumin) in TEC uptake studies improved IVIVE of RSV CL uptake,in vivo It did, and our predictions were close to or just fell above our lower 2-fold acceptance boundary. Despite this success, additional studies are needed to improve transporter-mediated IVIVE of hepatic uptake CL of drugs. However, using REF and TEC, we successfully predicted the magnitude of PET-imaged inhibition of RSV CL uptake,in vivo by cyclosporine A. SIGNIFICANCE STATEMENT: We showed that the in vivo transporter-mediated hepatic uptake CL of rosuvastatin, determined by PET imaging, can be predicted (within 2-fold) from in vitro studies in transporter-expressing cells (TECs) (scaled using REF), but only when plasma proteins were included in the in vitro studies. This conclusion did not hold when plasma proteins were absent in the TEC or human hepatocyte studies. Thus, additional studies are needed to improve in vitro to in vivo extrapolation of transporter-mediated drug CL.
Collapse
Affiliation(s)
- Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Mengyue Yin
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Kazuya Ishida
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Laurent Salphati
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Cornelis E C A Hop
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Christopher Rowbottom
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Guangqing Xiao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Yurong Lai
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Anita Mathias
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Xiaoyan Chu
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - W Griffith Humphreys
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Mingxiang Liao
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Zsuzsanna Nerada
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Nóra Szilvásy
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Scott Heyward
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (V.K., M.Y., K.I., J.D.U.); Drug Metabolism and Pharmacokinetics, Genentech, Inc., South San Francisco, California (L.S., C.E.C.A.H.); DMPK, Biogen Idec, Cambridge, Massachusetts (C.R., G.X.); Clinical Pharmacology (A.M.) and Drug Metabolism (Y.L.), Gilead Sciences, Inc., Foster City, California; Pharmacokinetics, Pharmacodynamics and Drug Metabolism, Merck & Co. Inc., Kenilworth, New Jersey (X.C.); Bristol-Myers Squibb Company, Princeton, New Jersey (W.G.H.); Takeda Pharmaceuticals International Co., Cambridge, Massachusetts (M.L.); SOLVO Biotechnology, Budaörs, Hungary (Z.N., N.S.); and BioIVT, Baltimore, Maryland (S.H.)
| |
Collapse
|
42
|
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: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [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
|
43
|
Sachar M, Kumar V, Gormsen LC, Munk OL, Unadkat JD. Successful Prediction of Positron Emission Tomography-Imaged Metformin Hepatic Uptake Clearance in Humans Using the Quantitative Proteomics-Informed Relative Expression Factor Approach. Drug Metab Dispos 2020; 48:1210-1216. [PMID: 32843330 DOI: 10.1124/dmd.120.000156] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
Predicting transporter-mediated in vivo hepatic drug clearance (CL) from in vitro data (IVIVE) is important in drug development to estimate first-in-human dose and the impact of drug interactions and pharmacogenetics on hepatic drug CL. For IVIVE, one can use human hepatocytes and the traditional milligrams of protein content per gram of liver tissue (MGPGL) approach. However, this approach has been found to consistently underpredict the observed in vivo hepatic drug CL. Therefore, we hypothesized that using transporter-expressing cells and the relative expression factor (REF), determined using targeted quantitative proteomics, will accurately predict in vivo hepatic CL of drugs. We have successfully tested this hypothesis in rats with rosuvastatin, which is transported by hepatic Organic anion transporting polypeptides (OATPs). Here, we tested this hypothesis for another drug and another transporter; namely, organic cation transporter (OCT)1-mediated hepatic distributional CL of metformin. First, we estimated the in vivo metformin hepatic sinusoidal uptake CL (CLh,s,in) of metformin by reanalysis of previously published human positron emission tomography imaging data. Next, using the REF approach, we predicted the in vivo metformin CLh,s,in using OCT1-transporter-expressing HEK293 cells or plated human hepatocytes. Finally, we compared this REF-based prediction with that using the MGPGL approach. The REF approach accurately predicted the in vivo metformin hepatic CLh,s,in, whereas the MGPGL approach considerably underpredicted the in vivo metformin CLh,s,in Based on these and previously published data, the REF approach appears to be superior to the MGPGL approach for a diverse set of drugs transported by different transporters. SIGNIFICANCE STATEMENT: This study is the first to use organic cation transporter 1-expressing cells and plated hepatocytes to compare proteomics-informed REF approach with the traditional MGPGL approach to predict hepatic uptake CL of metformin in humans. The proteomics-informed REF approach, which corrected for plasma membrane abundance, accurately predicted the positron emission tomography-imaged metformin hepatic uptake CL, whereas the MGPGL approach consistently underpredicted this CL.
Collapse
Affiliation(s)
- Madhav Sachar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Vineet Kumar
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Lars C Gormsen
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Ole Lajord Munk
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| | - Jashvant D Unadkat
- Department of Pharmaceutics, University of Washington, Seattle, Washington (M.S., V.K., J.D.U.) and Department of Nuclear Medicine and PET Center, Aarhus University Hospital, Aarhus N, Denmark (L.C.G., O.L.M.)
| |
Collapse
|
44
|
Li N, Badrinarayanan A, Li X, Roberts J, Hayashi M, Virk M, Gupta A. Comparison of In Vitro to In Vivo Extrapolation Approaches for Predicting Transporter-Mediated Hepatic Uptake Clearance Using Suspended Rat Hepatocytes. Drug Metab Dispos 2020; 48:861-872. [PMID: 32759366 DOI: 10.1124/dmd.120.000064] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2020] [Accepted: 07/08/2020] [Indexed: 12/15/2022] Open
Abstract
Clearance (CL) prediction remains a significant challenge in drug discovery, especially when complex processes such as drug transporters are involved. The present work explores various in vitro to in vivo extrapolation (IVIVE) approaches to predict hepatic CL driven by uptake transporters in rat. Broadly, two different IVIVE methods using suspended rat hepatocytes were compared: initial uptake CL (PSu,inf) and intrinsic metabolic CL (CLint,met) corrected by unbound hepatocytes to medium partition coefficient (Kpuu). Kpuu was determined by temperature method (Temp Kpuu,ss), homogenization method (Hom Kpuu,ss), and initial rate method (Kpuu,V0). In addition, the impact of bovine serum albumin (BSA) on each of these methods was investigated. Twelve compounds, which are known substrates of organic anion-transporting polypeptides representing diverse chemical matter, were selected for these studies. As expected, CLint,met alone significantly underestimated hepatic CL for all the test compounds. Overall, predicted hepatic CL using PSu,inf with BSA, Hom Kpuu,ss with BSA, and Temp Kpuu,ss showed the most robust correlation with in vivo rat hepatic CL. Adding BSA improved hepatic CL prediction for selected compounds when using the PSu,inf and Hom Kpuu,ss methods, with minimal impact on the Temp Kpuu,ss and Kpuu,V0 methods. None of the IVIVE approaches required an empirical scaling factor. These results suggest that supplementing rat hepatocyte suspension with BSA may be essential in drug discovery research for novel chemical matters to improve CL prediction. SIGNIFICANCE STATEMENT: The current investigation demonstrates that hepatocyte uptake assay supplemented with 4% bovine serum albumin is a valuable tool for estimating unbound hepatic uptake clearance (CL) and Kpuu. Based upon the extended clearance concept, direct extrapolation from these in vitro parameters significantly improved the overall hepatic CL prediction for organic anion-transporting polypeptide substrates in rat. This study provides a practical in vitro to in vivo extrapolation strategy for predicting transporter-mediated hepatic CL in early drug discovery.
Collapse
Affiliation(s)
- Na Li
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| | - Akshay Badrinarayanan
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| | - Xingwen Li
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| | - John Roberts
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| | - Mike Hayashi
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| | - Manpreet Virk
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| | - Anshul Gupta
- Department of Pharmacokinetics and Drug Metabolism, Amgen Research, Amgen Inc., Cambridge, Massachusetts
| |
Collapse
|
45
|
Kwon JH, Lee HJ, Escher BI. Bioavailability of hydrophobic organic chemicals on an in vitro metabolic transformation using rat liver S9 fraction. Toxicol In Vitro 2020; 66:104835. [DOI: 10.1016/j.tiv.2020.104835] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 03/07/2020] [Accepted: 03/22/2020] [Indexed: 10/24/2022]
|
46
|
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
|
47
|
Back HM, Yun HY, Kim SK, Kim JK. Beyond the Michaelis-Menten: Accurate Prediction of In Vivo Hepatic Clearance for Drugs With Low K M. Clin Transl Sci 2020; 13:1199-1207. [PMID: 32324332 PMCID: PMC7719389 DOI: 10.1111/cts.12804] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2019] [Accepted: 04/12/2020] [Indexed: 02/03/2023] Open
Abstract
Clearance (CL) is the major pharmacokinetic parameter for evaluating systemic exposure of drugs in the body and, thus, for developing new drugs. To predict in vivo CL, the ratio between the maximal rate of metabolism and Michaelis‐Menten constant (Vmax/KM estimated from in vitro metabolism study has been widely used. This canonical approach is based on the Michaelis‐Menten equation, which is valid only when the KM value of a drug is much higher than the hepatic concentration of the enzymes, especially cytochrome P450, involved in its metabolism. Here, we find that such a condition does not hold for many drugs with low KM, and, thus, the canonical approach leads to considerable error. Importantly, we propose an alternative approach, which incorporates the saturation of drug metabolism when concentration of the enzymes is not sufficiently lower than KM. This new approach dramatically improves the accuracy of prediction for in vivo CL of high‐affinity drugs with low KM. This indicates that the proposed approach in this study, rather than the canonical approach, should be used to predict in vivo hepatic CL for high‐affinity drugs, such as midazolam and propafenone.
Collapse
Affiliation(s)
- Hyun-Moon Back
- Department of Pharmaceutics, Ernest Mario School of Pharmacy, Rutgers, The State University of New Jersey, Piscataway, New Jersey, USA
| | - Hwi-Yeol Yun
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Sang Kyum Kim
- College of Pharmacy, Chungnam National University, Daejeon, Republic of Korea
| | - Jae Kyoung Kim
- Department of Mathematical Sciences, Korean Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
48
|
Sodhi JK, Wang HJ, Benet LZ. Are There Any Experimental Perfusion Data that Preferentially Support the Dispersion and Parallel-Tube Models over the Well-Stirred Model of Organ Elimination? Drug Metab Dispos 2020; 48:537-543. [PMID: 32305951 DOI: 10.1124/dmd.120.090530] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 03/31/2020] [Indexed: 12/18/2022] Open
Abstract
In reviewing previously published isolated perfused rat liver studies, we find no experimental data for high-clearance metabolized drugs that reasonably or unambiguously support preference for the dispersion and parallel-tube models versus the well-stirred model of organ elimination when only entering and exiting drug concentrations are available. It is likely that the investigators cited here may have been influenced by: 1) the unphysiologic aspects of the well-stirred model, which may have led them to undervalue the studies that directly test the various hepatic disposition models for high-clearance drugs (for which model differences are the greatest); 2) experimental assumptions made in the last century, which are no longer valid today, related to the predictability of in vivo outcomes from in vitro measures of drug elimination and the influence of albumin in hepatic drug uptake; and 3) a lack of critical review of previously reported experimental studies, resulting in inappropriate interpretation of the available experimental data. The number of papers investigating the theoretical aspects of the dispersion, parallel-tube, and well-stirred models of hepatic elimination greatly outnumber the papers that actually examine the experimental evidence available to substantiate these models. When all experimental studies that measure organ elimination using entering and exiting drug concentrations at steady state are critically reviewed, the simple but unphysiologic well-stirred model is the only model that can describe all trustworthy published available data. SIGNIFICANCE STATEMENT: Although the dispersion model of hepatic elimination more adequately reflects physiologic reality, there are no convincing experimental data that unambiguously favor this model. The well-stirred model can describe all well-designed perfusion studies with high-clearance drugs and nondrug substrates, but the field has not recognized this because of hesitation to accept a nonphysiologic model and flawed attempts to utilize in vitro-in vivo extrapolation approaches.
Collapse
Affiliation(s)
- Jasleen K Sodhi
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (J.K.S., L.Z.B.) and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan (H.-J.W.)
| | - Hong-Jaan Wang
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (J.K.S., L.Z.B.) and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan (H.-J.W.)
| | - Leslie Z Benet
- Department of Bioengineering and Therapeutic Sciences, Schools of Pharmacy and Medicine, University of California San Francisco, San Francisco, California (J.K.S., L.Z.B.) and School of Pharmacy, National Defense Medical Center, Taipei, Taiwan (H.-J.W.)
| |
Collapse
|
49
|
Alluri RV, Li R, Varma MVS. Transporter–enzyme interplay and the hepatic drug clearance: what have we learned so far? Expert Opin Drug Metab Toxicol 2020; 16:387-401. [DOI: 10.1080/17425255.2020.1749595] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Affiliation(s)
- Ravindra V. Alluri
- Clinical Pharmacology and Safety Sciences, R&D, AstraZeneca, Cambridge, UK
| | - Rui Li
- Modeling and Simulations, Medicine Design, Worldwide Research and Development, Pfizer Inc., Cambridge, MA, USA
| | - Manthena V. S. Varma
- ADME Sciences, Medicine Design, Worldwide Research and Development, Pfizer Inc., Groton, CT, USA
| |
Collapse
|
50
|
Nozaki Y, Izumi S. Recent advances in preclinical in vitro approaches towards quantitative prediction of hepatic clearance and drug-drug interactions involving organic anion transporting polypeptide (OATP) 1B transporters. Drug Metab Pharmacokinet 2020; 35:56-70. [DOI: 10.1016/j.dmpk.2019.11.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 09/29/2019] [Accepted: 11/02/2019] [Indexed: 12/26/2022]
|