1
|
Gardner I, Xu M, Han C, Wang Y, Jiao X, Jamei M, Khalidi H, Kilford P, Neuhoff S, Southall R, Turner DB, Musther H, Jones B, Taylor S. Non-specific binding of compounds in in vitro metabolism assays: a comparison of microsomal and hepatocyte binding in different species and an assessment of the accuracy of prediction models. Xenobiotica 2022; 52:943-956. [PMID: 36222269 DOI: 10.1080/00498254.2022.2132426] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Non-specific binding in in vitro metabolism systems leads to an underestimation of the true intrinsic metabolic clearance of compounds being studied. Therefore in vitro binding needs to be accounted for when extrapolating in vitro data to predict the in vivo metabolic clearance of a compound. While techniques exist for experimentally determining the fraction of a compound unbound in in vitro metabolism systems, early in drug discovery programmes computational approaches are often used to estimate the binding in the in vitro system.Experimental fraction unbound data (n = 60) were generated in liver microsomes (fumic) from five commonly used pre-clinical species (rat, mouse, dog, minipig, monkey) and humans. Unbound fraction in incubations with mouse, rat or human hepatocytes was determined for the same 60 compounds. These data were analysed to determine the relationship between experimentally determined binding in the different matrices and across different species. In hepatocytes there was a good correlation between fraction unbound in human and rat (r2=0.86) or mouse (r2=0.82) hepatocytes. Similar correlations were observed between binding in human liver microsomes and microsomes from rat, mouse, dog, Göttingen minipig or monkey liver microsomes (r2 of >0.89, n = 51 - 52 measurements in different species). Physicochemical parameters (logP, pKa and logD) were predicted for all evaluated compounds. In addition, logP and/or logD were measured for a subset of compounds.Binding to human hepatocytes predicted using 5 different methods was compared to the measured data for a set of 59 compounds. The best methods evaluated used measured microsomal binding in human liver microsomes to predict hepatocyte binding. The collated physicochemical data were used to predict the human fumic using four different in silico models for a set of 53-60 compounds. The correlation (r2) and root mean square error between predicted and observed microsomal binding was 0.69 & 0.20, 0.47 & 0.23, 0.56 & 0.21 and 0.54 & 0.26 for the Turner-Simcyp, Austin, Hallifax-Houston and Poulin models, respectively. These analyses were extended to include measured literature values for binding in human liver microsomes for a larger set of compounds (n=697). For the larger dataset of compounds, microsomal binding was well predicted for neutral compounds (r2=0.67 - 0.70) using the Poulin, Austin, or Turner-Simcyp methods but not for acidic or basic compounds (r2<0.5) using any of the models. While the lipophilicity-based models can be used, the in vitro binding should be measured for compounds where more certainty is needed, using appropriately calibrated assays and possibly established weak, moderate, and strong binders as reference compounds to allow comparison across databases.
Collapse
Affiliation(s)
| | - Mandy Xu
- Pharmaron Beijing Co. Ltd., Beijing, China
| | | | - Yi Wang
- Pharmaron Beijing Co. Ltd., Beijing, China
| | | | | | | | - Peter Kilford
- Certara UK Ltd., Sheffield, United Kingdom.,Labcorp Drug Development, Harrogate, United Kingdom
| | | | | | | | | | - Barry Jones
- Pharmaron UK, Hoddesdon, Hertfordshire, United Kingdom
| | - Simon Taylor
- Pharmaron UK, Hoddesdon, Hertfordshire, United Kingdom
| |
Collapse
|
2
|
Riede J, Wollmann BM, Molden E, Ingelman-Sundberg M. Primary human hepatocyte spheroids as an in vitro tool for investigating drug compounds with low clearance. Drug Metab Dispos 2021; 49:DMD-AR-2020-000340. [PMID: 34074732 DOI: 10.1124/dmd.120.000340] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2020] [Revised: 03/24/2021] [Accepted: 04/15/2021] [Indexed: 11/22/2022] Open
Abstract
Characterizing the pharmacokinetic properties of drug candidates represents an essential task during drug development. In the past, liver microsomes and primary suspended hepatocytes have been extensively used for this purpose, but their relatively short stability limits the applicability of such in vitro systems for drug compounds with low metabolic turnover. In the present study, we used 3D primary human hepatocyte spheroids to predict the hepatic clearance of seven drugs with low to intermediate clearance in humans. Our results indicate that hepatocyte spheroids maintain their in vivo like phenotype during prolonged incubations allowing to monitor the depletion of parent drug for seven days. In contrast, attempts to increase the relative metabolic capacity by pooling hepatocyte spheroids resulted in an immediate fusion of the spheroids followed by hepatocellular de-differentiation processes, demonstrating limited applicability of the pooling approach for quantitative pharmacokinetic studies. The hepatic clearance values obtained from incubations with individual spheroids were in close correlation with the clinical reference data with six out of seven drug compounds being predicted within a three-fold deviation and average fold and absolute average fold errors of 0.57 and 1.74, respectively. In conclusion, the hepatocyte spheroid model enables accurate hepatic clearance predictions for slowly metabolized drug compounds and represents a valuable tool for determining the pharmacokinetic properties of new drug candidates as well as for mechanistic pharmacokinetic studies. Significance Statement Traditional in vitro systems often fail to predict the hepatic clearance of slowly metabolized drug compounds. The current study demonstrates the ability of primary human hepatocyte spheroids to provide accurate projections on the hepatic clearance of drug compounds with low and intermediate clearance.
Collapse
Affiliation(s)
- Julia Riede
- Drug Metabolism and Pharmacokinetics, Novartis Institutes for Biomedical Research, Switzerland
| | | | - Espen Molden
- Dept. of Pharmacology, University of Oslo, Norway
| | | |
Collapse
|
3
|
Metabolism and pharmacokinetics characterization of metarrestin in multiple species. Cancer Chemother Pharmacol 2020; 85:805-816. [PMID: 32185484 DOI: 10.1007/s00280-020-04042-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2019] [Accepted: 02/17/2020] [Indexed: 01/17/2023]
Abstract
PURPOSE Metarrestin is a first-in-class pyrrolo-pyrimidine-derived small molecule targeting a marker of genome organization associated with metastasis and is currently in preclinical development as an anti-cancer agent. Here, we report the in vitro ADME characteristics and in vivo pharmacokinetic behavior of metarrestin. METHODS Solubility, permeability, and efflux ratio as well as in vitro metabolism of metarrestin in hepatocytes, liver microsomes and S9 fractions, recombinant cytochrome P450 (CYP) enzymes, and potential for CYP inhibition were evaluated. Single dose pharmacokinetic profiles after intravenous and oral administration in mice, rat, dog, monkey, and mini-pig were obtained. Simple allometric scaling was applied to predict human pharmacokinetics. RESULTS Metarrestin had an aqueous solubility of 150 µM at pH 7.4, high permeability in PAMPA and moderate efflux ratio in Caco-2 assays. The compound was metabolically stable in liver microsomes, S9 fractions, and hepatocytes from six species, including human. Metarrestin is a CYP3A4 substrate and, in mini-pigs, is also directly glucuronidated. Metarrestin did not show cytochrome P450 inhibitory activity. Plasma concentration-time profiles showed low to moderate clearance, ranging from 0.6 mL/min/kg in monkeys to 48 mL/min/kg in mice and moderate to high volume of distribution, ranging from 1.5 L/kg in monkeys to 17 L/kg in mice. Metarrestin has greater than 80% oral bioavailability in all species tested. The excretion of unchanged parent drug in urine was < 5% in dogs and < 1% in monkeys over collection periods of ≥ 144 h; in bile-duct cannulated rats, the excretion of unchanged drug was < 1% in urine and < 2% in bile over a collection period of 48 h. CONCLUSIONS Metarrestin is a low clearance compound which has good bioavailability and large biodistribution after oral administration. Biotransformation appears to be the major elimination process for the parent drug. In vitro data suggest a low drug-drug interaction potential on CYP-mediated metabolism. Overall favorable ADME and PK properties support metarrestin's progression to clinical investigation.
Collapse
|
4
|
Pharmacokinetics, Tissue Distribution and Excretion of a Novel Diuretic (PU-48) in Rats. Pharmaceutics 2018; 10:pharmaceutics10030124. [PMID: 30096833 PMCID: PMC6160999 DOI: 10.3390/pharmaceutics10030124] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Revised: 07/21/2018] [Accepted: 07/27/2018] [Indexed: 01/02/2023] Open
Abstract
Methyl 3-amino-6-methoxythieno [2,3-b] quinoline-2-carboxylate (PU-48) is a novel diuretic urea transporter inhibitor. The aim of this study is to investigate the profile of plasma pharmacokinetics, tissue distribution, and excretion by oral dosing of PU-48 in rats. Concentrations of PU-48 within biological samples are determined using a validated high performance liquid chromatography-tandem mass spectrometry (LC-MS/MS) method. After oral administration of PU-48 (3, 6, and 12 mg/kg, respectively) in self-nanomicroemulsifying drug delivery system (SNEDDS) formulation, the peak plasma concentrations (Cmax), and the area under the curve (AUC0⁻∞) were increased by the dose-dependent and linear manner, but the marked different of plasma half-life (t1/2) were not observed. This suggests that the pharmacokinetic profile of PU-48 prototype was first-order elimination kinetic characteristics within the oral three doses range in rat plasma. Moreover, the prototype of PU-48 was rapidly and extensively distributed into thirteen tissues, especially higher concentrations were detected in stomach, intestine, liver, kidney, and bladder. The total accumulative excretion of PU-48 in the urine, feces, and bile was less than 2%. This research is the first report on disposition via oral administration of PU-48 in rats, and it provides important information for further development of PU-48 as a diuretic drug candidate.
Collapse
|
5
|
Riede J, Camenisch G, Huwyler J, Poller B. Current In Vitro Methods to Determine Hepatic Kp uu : A Comparison of Their Usefulness and Limitations. J Pharm Sci 2017; 106:2805-2814. [DOI: 10.1016/j.xphs.2017.03.025] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2017] [Revised: 03/23/2017] [Accepted: 03/27/2017] [Indexed: 12/20/2022]
|
6
|
Chen S, Prieto Garcia L, Bergström F, Nordell P, Grime K. Intrinsic Clearance Assay Incubational Binding: A Method Comparison. Drug Metab Dispos 2017; 45:342-345. [PMID: 28122786 DOI: 10.1124/dmd.116.074138] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2016] [Accepted: 01/18/2017] [Indexed: 11/22/2022] Open
Abstract
The fraction of unbound drug (fuinc) in in vitro intrinsic clearance (CLint) incubation is an important parameter in the pursuit of accurate clearance predictions and is often predicted using algorithms based on drug lipophilicity measures. However, analysis of an AstraZeneca database suggests that simple lipophilicity alone is a relatively poor predictor of fuinc measured using equilibrium dialysis. He fuinc value can also be measured directly in CLint assays using multiple concentrations of hepatocytes or microsomal protein. Since this approach informs of the unbound drug concentration in the assay used to predict in vivo clearance, it should be considered the gold standard method. As a starting point for building better predictive algorithms we aimed to determine if equilibrium dialysis really is an appropriate assay for assessing fuinc Employing a large number of compounds with a wide range of lipophilicities, experiments were performed to measure fuinc using rat hepatocytes (RH) and human liver microsomes (HLM) in both assay formats. A high percentage (94% and 93% for HLM and RH, respectively) of the fuinc values were within 2-fold when the compound distribution coefficient describing the ratio of compound concentration in octanol and pH 7.4 buffer when the test system is at equilibrium (lipophilicity measure) (logD7.4) values were less than 3.5. However, with logD7.4 values greater than these, the agreement was considerably worse. Additional experimental data generated indicated that this discrepancy was likely due to failings in the direct method when drug binding is high. Thus, we conclude that unbound CLint can be indeed calculated indirectly by incorporating equilibrium dialysis data with measured CLint but that simple lipophilicity descriptors alone may be inadequate for predicting fuinc.
Collapse
Affiliation(s)
- Sofia Chen
- Respiratory, Inflammation and Autoimmunity Department of Drug Metabolism and Pharmacokinetics (K.G., S.C., L.P.G.), Drug Safety and Metabolism (P.N.), and Cardiovascular and Metabolic Diseases Department of Drug Metabolism and Pharmacokinetics (F.B.), Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Luna Prieto Garcia
- Respiratory, Inflammation and Autoimmunity Department of Drug Metabolism and Pharmacokinetics (K.G., S.C., L.P.G.), Drug Safety and Metabolism (P.N.), and Cardiovascular and Metabolic Diseases Department of Drug Metabolism and Pharmacokinetics (F.B.), Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Fredrik Bergström
- Respiratory, Inflammation and Autoimmunity Department of Drug Metabolism and Pharmacokinetics (K.G., S.C., L.P.G.), Drug Safety and Metabolism (P.N.), and Cardiovascular and Metabolic Diseases Department of Drug Metabolism and Pharmacokinetics (F.B.), Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Pär Nordell
- Respiratory, Inflammation and Autoimmunity Department of Drug Metabolism and Pharmacokinetics (K.G., S.C., L.P.G.), Drug Safety and Metabolism (P.N.), and Cardiovascular and Metabolic Diseases Department of Drug Metabolism and Pharmacokinetics (F.B.), Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| | - Ken Grime
- Respiratory, Inflammation and Autoimmunity Department of Drug Metabolism and Pharmacokinetics (K.G., S.C., L.P.G.), Drug Safety and Metabolism (P.N.), and Cardiovascular and Metabolic Diseases Department of Drug Metabolism and Pharmacokinetics (F.B.), Innovative Medicines and Early Development, AstraZeneca, Gothenburg, Sweden
| |
Collapse
|
7
|
Yamagata T, Zanelli U, Gallemann D, Perrin D, Dolgos H, Petersson C. Comparison of methods for the prediction of human clearance from hepatocyte intrinsic clearance for a set of reference compounds and an external evaluation set. Xenobiotica 2016; 47:741-751. [PMID: 27560606 DOI: 10.1080/00498254.2016.1222639] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
1. We compared direct scaling, regression model equation and the so-called "Poulin et al." methods to scale clearance (CL) from in vitro intrinsic clearance (CLint) measured in human hepatocytes using two sets of compounds. One reference set comprised of 20 compounds with known elimination pathways and one external evaluation set based on 17 compounds development in Merck (MS). 2. A 90% prospective confidence interval was calculated using the reference set. This interval was found relevant for the regression equation method. The three outliers identified were justified on the basis of their elimination mechanism. 3. The direct scaling method showed a systematic underestimation of clearance in both the reference and evaluation sets. The "Poulin et al." and the regression equation methods showed no obvious bias in either the reference or evaluation sets. 4. The regression model equation was slightly superior to the "Poulin et al." method in the reference set and showed a better absolute average fold error (AAFE) of value 1.3 compared to 1.6. A larger difference was observed in the evaluation set were the regression method and "Poulin et al." resulted in an AAFE of 1.7 and 2.6, respectively (removing the three compounds with known issues mentioned above). A similar pattern was observed for the correlation coefficient. Based on these data we suggest the regression equation method combined with a prospective confidence interval as the first choice for the extrapolation of human in vivo hepatic metabolic clearance from in vitro systems.
Collapse
Affiliation(s)
- Tetsuo Yamagata
- a Global Early Development/Quantitative Pharmacology and Drug Disposition (QPD), Merck KGaA , Grafing , Germany and
| | - Ugo Zanelli
- b Global Early Development/Quantitative Pharmacology and Drug Disposition (QPD), Merck KGaA , Darmstadt , Germany
| | - Dieter Gallemann
- a Global Early Development/Quantitative Pharmacology and Drug Disposition (QPD), Merck KGaA , Grafing , Germany and
| | - Dominique Perrin
- b Global Early Development/Quantitative Pharmacology and Drug Disposition (QPD), Merck KGaA , Darmstadt , Germany
| | - Hugues Dolgos
- b Global Early Development/Quantitative Pharmacology and Drug Disposition (QPD), Merck KGaA , Darmstadt , Germany
| | - Carl Petersson
- b Global Early Development/Quantitative Pharmacology and Drug Disposition (QPD), Merck KGaA , Darmstadt , Germany
| |
Collapse
|
8
|
Montelukast Disposition: No Indication of Transporter-Mediated Uptake in OATP2B1 and OATP1B1 Expressing HEK293 Cells. Pharmaceutics 2015; 7:554-64. [PMID: 26694455 PMCID: PMC4695834 DOI: 10.3390/pharmaceutics7040554] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Revised: 11/24/2015] [Accepted: 12/09/2015] [Indexed: 12/26/2022] Open
Abstract
Clinical studies with montelukast show variability in effect and polymorphic OATP2B1-dependent absorption has previously been implicated as a possible cause. This claim has been challenged with conflicting data and here we used OATP2B1-transfected HEK293 cells to clarify the mechanisms involved. For montelukast, no significant difference in cell uptake between HEK-OATP2B1 and empty vector cell lines was observed at pH 6.5 or pH 7.4, and no concentration-dependent uptake was detected. Montelukast is a carboxylic acid, a relatively potent inhibitor of OATP1B1, OATP1B3, and OATP2B1, and has previously been postulated to be actively transported into human hepatocytes. Using OATP1B1-transfected HEK293 cells and primary human hepatocytes in the presence of OATP inhibitors we demonstrate for the first time that active OATP-dependent transport is unlikely to play a significant role in the human disposition of montelukast.
Collapse
|
9
|
Fay KA, Nabb DL, Mingoia RT, Bischof I, Nichols JW, Segner H, Johanning K, Han X. Determination of Metabolic Stability Using Cryopreserved Hepatocytes from Rainbow Trout (
Oncorhynchus mykiss
). ACTA ACUST UNITED AC 2015; 65:4.42.1-4.42.29. [DOI: 10.1002/0471140856.tx0442s65] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Kellie A. Fay
- ORD/NHEERL/Mid‐Continent Ecology Division, U.S. EPA Duluth Minnesota
| | - Diane L. Nabb
- DuPont Haskell Global Centers for Health and Environmental Sciences Newark Delaware
| | - Robert T. Mingoia
- DuPont Haskell Global Centers for Health and Environmental Sciences Newark Delaware
| | - Ina Bischof
- Centre for Fish and Wildlife Health, University of Bern Bern Switzerland
- Fraunhofer Institute for Molecular Biology and Applied Ecology Schmallenberg Germany
| | - John W. Nichols
- ORD/NHEERL/Mid‐Continent Ecology Division, U.S. EPA Duluth Minnesota
| | - Helmut Segner
- Centre for Fish and Wildlife Health, University of Bern Bern Switzerland
| | - Karla Johanning
- KJ Scientific LCC, Texas Life Sciences Collaboration Center Georgetown Texas
| | - Xing Han
- DuPont Haskell Global Centers for Health and Environmental Sciences Newark Delaware
| |
Collapse
|
10
|
Nordell P, Winiwarter S, Hilgendorf C. Resolving the distribution-metabolism interplay of eight OATP substrates in the standard clearance assay with suspended human cryopreserved hepatocytes. Mol Pharm 2013; 10:4443-51. [PMID: 24102095 DOI: 10.1021/mp400253f] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Uptake transporters may act to elevate the intrahepatic exposure of drugs, impacting the route and rate of elimination, as well as the drug-drug interaction potential. We have here extended the assessment of metabolic drug stability in a standard human hepatocyte incubation to allow for elucidation of the distribution-metabolism interplay established for substrates of drug transporters. Cellular concentration-time profiles were obtained from incubations of eight known OATP substrates at 1 μM, each for two different 10-donor batches of suspended cryopreserved human hepatocytes. The kinetic data sets were analyzed using a mechanistic mathematical model that allowed for separate estimation of active uptake, bidirectional diffusion, metabolism and nonspecific extracellular and intracellular binding. The range of intrinsic clearances attributed to active uptake, diffusion and metabolism of the test set spanned more than 2 orders of magnitude each, with median values of 18, 5.3, and 0.5 μL/min/10(6) cells, respectively. This is to be compared with the values for the apparent clearance from the incubations, which only spanned 1 order of magnitude with a median of 2.6 μL/min/10(6) cells. The parameter estimates of the two pooled 10-donor hepatocyte batches investigated displayed only small differences in contrast to the variability associated with use of cells from individual donors reported in the literature. The active contribution to the total cellular uptake ranged from 55% (glyburide) to 96% (rosuvastatin), with an unbound intra-to-extracellular concentration ratio at steady state of 2.1 and 17, respectively. Principal component analysis showed that the parameter estimates of the investigated compounds were largely influenced by lipophilicity. Active cellular uptake in hepatocytes was furthermore correlated to pure OATP1B1-mediated uptake as measured in a transfected cell system. The presented approach enables the assessment of the key pathways regulating hepatic disposition of transporter and enzyme substrates from one single, reproducible and generally accessible human in vitro system.
Collapse
Affiliation(s)
- Pär Nordell
- Drug Safety and Metabolism, AstraZeneca R&D Mölndal , Pepparedsleden 1, SE-431 83 Mölndal, Sweden
| | | | | |
Collapse
|