Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 12 de 12
Filter
Add more filters










Publication year range
1.
Chem Res Toxicol ; 25(10): 2067-82, 2012 Oct 15.
Article in English | MEDLINE | ID: mdl-22931300

ABSTRACT

Drug-induced liver injury is the most common cause of market withdrawal of pharmaceuticals, and thus, there is considerable need for better prediction models for DILI early in drug discovery. We present a study involving 223 marketed drugs (51% associated with clinical hepatotoxicity; 49% non-hepatotoxic) to assess the concordance of in vitro bioactivation data with clinical hepatotoxicity and have used these data to develop a decision tree to help reduce late-stage candidate attrition. Data to assess P450 metabolism-dependent inhibition (MDI) for all common drug-metabolizing P450 enzymes were generated for 179 of these compounds, GSH adduct data generated for 190 compounds, covalent binding data obtained for 53 compounds, and clinical dose data obtained for all compounds. Individual data for all 223 compounds are presented here and interrogated to determine what level of an alert to consider termination of a compound. The analysis showed that 76% of drugs with a daily dose of <100 mg were non-hepatotoxic (p < 0.0001). Drugs with a daily dose of ≥100 mg or with GSH adduct formation, marked P450 MDI, or covalent binding ≥200 pmol eq/mg protein tended to be hepatotoxic (∼ 65% in each case). Combining dose with each bioactivation assay increased this association significantly (80-100%, p < 0.0001). These analyses were then used to develop the decision tree and the tree tested using 196 of the compounds with sufficient data (49% hepatotoxic; 51% non-hepatotoxic). The results of these outcome analyses demonstrated the utility of the tree in selectively terminating hepatotoxic compounds early; 45% of the hepatotoxic compounds evaluated using the tree were recommended for termination before candidate selection, whereas only 10% of the non-hepatotoxic compounds were recommended for termination. An independent set of 10 GSK compounds with known clinical hepatotoxicity status were also assessed using the tree, with similar results.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Drug Evaluation, Preclinical/methods , Drug-Related Side Effects and Adverse Reactions/metabolism , Liver/drug effects , Pharmaceutical Preparations/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Decision Trees , Glutathione/metabolism , Humans , Liver/metabolism , Protein Binding
2.
Xenobiotica ; 42(9): 880-90, 2012 Sep.
Article in English | MEDLINE | ID: mdl-22509823

ABSTRACT

OATP1A2 is expressed in the luminal membrane of human blood-brain barrier (BBB). The human tissue with the highest OATP1A2 mRNA expression is the brain. We have established a robust BacMam2-OATP1A2 transduced HEK293 system. Among the 36 central nervous system (CNS) marketed drugs tested, hydrophilic triptans, 5-HT(1B/1D) receptor agonists for the treatment of migraine attacks, were identified as OATP1A2 substrates. Kinetics (K(m) and V(max)) were determined for six marketed triptans. Structure-activity relationship (SAR) obtained from 18 triptan structural analogs revealed that the positively charged basic amine atom was essential for efficient OATP1A2-mediated triptan uptake and uptake rate was in the order of tertiary > secondary > primary. Preliminary quantitative SAR analysis of the triptan analogs demonstrated positive correlation between OATP1A2-mediated uptake rate and van der Waals volume (vdw_vol). OATP1A2 was specifically expressed on the apical side of MDCKII monolayer after BacMam2-OATP1A2 transduction and can facilitate transport of triptans across the MDCKII monolayer from apical to basolateral side. Involvement of OATP1A2 for brain penetration of triptans in human requires further investigation.


Subject(s)
Blood-Brain Barrier/metabolism , Migraine Disorders/drug therapy , Organic Anion Transporters/metabolism , Serotonin 5-HT1 Receptor Agonists/metabolism , Tryptamines/metabolism , Animals , Baculoviridae , Dogs , Genetic Vectors/genetics , HEK293 Cells , Humans , Immunohistochemistry , Madin Darby Canine Kidney Cells , Real-Time Polymerase Chain Reaction , Serotonin 5-HT1 Receptor Agonists/therapeutic use , Structure-Activity Relationship , Tryptamines/therapeutic use
3.
Drug Metab Dispos ; 38(2): 260-9, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19889884

ABSTRACT

From previous fits of drug transport kinetics across confluent Madin-Darby canine kidney II cell line overexpressing human multidrug resistance 1 cell monolayers, we found that a drug's binding constant to P-glycoprotein (P-gp) was significantly smaller than its IC(50) when that drug was used as an inhibitor against another P-gp substrate. We tested several IC(50) candidate functions, including the standard function, the Kalvass-Pollack function, and the efflux ratio, to determine whether any of them yielded an IC(50) = K(I), as would be expected for water-soluble enzymes. For the confluent cell monolayer, the IC(50)/K(I) ratio is greater than 1 for all candidate functions tested. From the mass action kinetic model, we have derived a simple approximate equation that shows how the IC(50)/K(I) ratio depends on the elementary rate constants from our mass action model. Thus, the IC(50) will differ between cell lines and tissues, for the same probe substrate and inhibitor, if there are different membrane concentrations of P-gp, or the probe substrate's elementary rate constants, partition coefficient, binding constant to P-gp, passive permeability, and ability to access the other transporters (if any) in the two cell lines. The mass action model and the approximate equation for the IC(50)/K(I) ratio derived here can be used to estimate the elementary rate constants needed to extrapolate in vitro drug-drug interactions for compounds to the in vivo environment.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Animals , Biological Transport/drug effects , Cell Line , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Membrane Permeability/drug effects , Computer Simulation , Digoxin/metabolism , Digoxin/pharmacokinetics , Dogs , Drug Interactions , Genes, MDR , Humans , Kidney/metabolism , Models, Biological , Protein Binding/drug effects , Quinidine/metabolism , Quinidine/pharmacokinetics , Thermodynamics
4.
Mol Pharm ; 6(1): 11-8, 2009.
Article in English | MEDLINE | ID: mdl-19248229

ABSTRACT

The Biopharmaceutics Classification System (BCS) is the scientific basis for classifying drugs based on their aqueous solubility and intestinal permeability that supports in vivo bioavailability and bioequivalence waivers for immediate-release solid dosage form drugs. One requirement of the BCS is that the permeability method must be validated. In order to accommodate the variety of in vitro/in situ permeability models, the BCS Guidance gives a general framework for the validation requirements, necessitating implemented experimental details to be selected by the applicant laboratory. The objective of this work was to define the parameters for a cell based in vitro permeability method (e.g., cell type, pH, transport direction, time, and concentration) and validate the method to support formal BCS classification of drugs. Twenty reference drugs were selected and permeability values determined using the Madin-Darby canine kidney type II cell line heterologously expressing the human P-glycoprotein transporter (MDCKII-MDR1). A rank order relationship was established between the in vitro permeability value and human intestinal absorption values. This relationship was as predicted and validates the MDCKII-MDR1 permeability method as defined by the BCS Guidance. The final validated in vitro permeability method employs the MDCKII-MDR1 cell line incubated with the Pgp inhibitor GF120918. It is a unidirectional apical-to-basolateral transport assay performed at apical pH values of 5.5 and 7.4 and a basolateral pH of 7.4. Four reference standards (metoprolol, pindolol, labetalol and ranitidine) dosed and analyzed as a single cassette are included in each experiment. A strategy on selection of drug concentrations and on how to deal with problematic compounds (i.e., those suffering from poor mass balance) is discussed.


Subject(s)
Biopharmaceutics/classification , Cell Membrane Permeability , Pharmaceutical Preparations/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line , Humans , Hydrogen-Ion Concentration , Time Factors
5.
Eur J Pharm Sci ; 34(2-3): 149-63, 2008 Jul 03.
Article in English | MEDLINE | ID: mdl-18467078

ABSTRACT

The objective of this investigation is to characterize the role of complex biophase distribution kinetics in the pharmacokinetic-pharmacodynamic correlation of a wide range of opioids. Following intravenous infusion of morphine, alfentanil, fentanyl, sufentanil, butorphanol and nalbuphine the time course of the EEG effect was determined in conjunction with blood concentrations. Different biophase distribution models were tested for their ability to describe hysteresis between blood concentration and effect. In addition, membrane transport characteristics of the opioids were investigated in vitro, using MDCK:MDR1 cells and in silico with QSAR analysis. For morphine, hysteresis was best described by an extended-catenary biophase distribution model with different values for k1e and keo of 0.038+/-0.003 and 0.043+/-0.003 min(-1), respectively. For the other opioids hysteresis was best described by a one-compartment biophase distribution model with identical values for k1e and keo. Between the different opioids, the values of k1e ranged from 0.04 to 0.47 min(-1). The correlation between concentration and EEG effect was successfully described by the sigmoidal Emax pharmacodynamic model. Between opioids significant differences in potency (EC50 range 1.2-451 ng/ml) and intrinsic activity (alpha range 18-109 microV) were observed. A statistically significant correlation was observed between the values of the in vivo k1e and the apparent passive permeability as determined in vitro in MDCK:MDR1 monolayers. It can be concluded that unlike other opioids, only morphine displays complex biophase distribution kinetics, which can be explained by its relatively low passive permeability and the interaction with active transporters at the blood-brain barrier.


Subject(s)
Analgesics, Opioid/pharmacology , Analgesics, Opioid/pharmacokinetics , Electroencephalography/drug effects , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Algorithms , Animals , Cell Line , Diffusion , Dogs , Male , Models, Statistical , Permeability , Quantitative Structure-Activity Relationship , Rats , Rats, Wistar , Tissue Distribution
6.
Drug Metab Dispos ; 36(2): 452-60, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17967933

ABSTRACT

A robust screen for compound interaction with P-glycoprotein (P-gp) has some obvious requirements, such as a cell line expressing P-gp and a probe substrate that is transported solely by P-gp and passive permeability. It is actually difficult to prove that a particular probe substrate interacts only with P-gp in the chosen cell line. Using a confluent monolayer of MDCKII-hMDR1 cells, we have determined the elementary rate constants for the P-gp efflux of amprenavir, digoxin, loperamide, and quinidine. For amprenavir and quinidine, transport was fitted with just P-gp and passive permeability. For digoxin and loperamide, fitting required a basolateral transporter (p < 0.01), which was inhibited by the P-gp inhibitor N-(4-[2-(1,2,3,4-tetrahydro-6,7-dimethoxy-2-isoquinolinyl)ethyl]-phenyl)-9,10-dihydro-5-methoxy-9-oxo-4-acridine carboxamide (GF120918). This means that when digoxin is used as a probe substrate and a compound is shown to inhibit digoxin flux, it could be that the inhibition occurs at the basolateral transporter rather than at P-gp. Digoxin basolateral>apical efflux also required an apical importer (p < 0.05). We propose that amprenavir and quinidine are robust probe substrates for assessing P-gp interactions using the MDCKII-hMDR1 confluent cell monolayer. Usage of another cell line, e.g., LLC-hMDR1 or Caco-2, would require the same kinetic validation to ensure that the probe substrate interacts only with P-gp. Attempts to identify the additional digoxin and loperamide transporters using a wide range of substrates/inhibitors of known epithelial transporters (organic cation transporters, organic anion transporters, organic ion-transporting polypeptide, uric acid transporter, or multidrug resistance-associated protein) failed to inhibit the digoxin or loperamide transport through their basolateral transporter.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Digoxin/metabolism , Loperamide/metabolism , Animals , Carbamates/metabolism , Cell Line , Cell Membrane/metabolism , Dogs , Furans , Kinetics , Quinidine/metabolism , Sulfonamides/metabolism
7.
Biochemistry ; 45(51): 15505-19, 2006 Dec 26.
Article in English | MEDLINE | ID: mdl-17176072

ABSTRACT

The multidrug resistance transporter P-glycoprotein (P-gp) effluxes a wide range of substrates and can be affected by a wide range of inhibitors or modulators. Many studies have presented classifications for these binding interactions, within either the context of equilibrium binding or the Michaelis-Menten enzyme analysis of the ATPase activity of P-gp. Our approach is to study P-gp transport and its inhibition using a physiologically relevant confluent monolayer of hMDR1-MDCKII cells. We measure the elementary rate constants for P-gp efflux of substrates and study inhibition using pairwise combinations with a different unlabeled substrate acting as the inhibitor. Our current kinetic model for P-gp has only a single binding site, because a previous study proved that the mass-action kinetics of efflux of a single substrate were not sensitive to whether there are one or more substrate-binding and efflux sites. In this study, using this one-site model, we found that, with "high" concentrations of either a substrate or an inhibitor, the elementary rate constants fitted independently for each of the substrates alone quantitatively predicted the efflux curves, simply applying the assumption that binding at the "one site" was competitive. On the other hand, at "low" concentrations of both the substrate and inhibitor, we found no inhibition of the substrate efflux, despite the fact that both the substrate and inhibitor were being well-effluxed. This was not an effect of excess "empty" P-gp molecules, because the competitive efflux model takes site occupancy into account. Rather, it is quantitative evidence that the substrate and inhibitor are being effluxed by multiple pathways within P-gp. Remarkably, increasing the substrate concentration above the "low" concentration, caused the inhibition to become competitive; i.e., the inhibitor became effective. These data and their analysis show that the binding of these substrates must be cooperative, either positive or negative.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Signal Transduction , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Animals , Binding, Competitive/genetics , Biological Transport, Active/drug effects , Biological Transport, Active/genetics , Carbamates/antagonists & inhibitors , Carbamates/metabolism , Cell Line , Cell Membrane Permeability/genetics , Dogs , Furans , Humans , Loperamide/antagonists & inhibitors , Loperamide/metabolism , Protein Binding/genetics , Quinidine/pharmacology , Signal Transduction/genetics , Substrate Specificity/drug effects , Substrate Specificity/genetics , Sulfonamides/antagonists & inhibitors , Sulfonamides/metabolism
8.
Drug Metab Dispos ; 34(6): 1063-9, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16531474

ABSTRACT

Drug-induced changes in expression of cytochrome P450 (P450) genes are a significant issue in the preclinical development of pharmaceuticals. For example, preclinically, P450 induction can affect safety studies by reducing the systemic exposure of a compound undergoing toxicological evaluation, thus limiting the exposure that can be safely investigated in patients. Therefore, the induction potential of candidate drugs has been studied as part of the drug development process, typically using protein and/or catalytic end points. However, measuring changes in the levels of mRNA using TaqMan technology offers the opportunity to investigate this issue with the advantages of better dynamic range and specific enzyme identification. Here, we describe the TaqMan application to study ex vivo the P450 gene induction in the rat. Initially, livers from rats dosed with the prototypic P450 inducers beta-napthoflavone (BNF), phenobarbital (PB), dexamethasone (DEX), and clofibric acid (CLO) were analyzed for mRNA levels of CYP1A1, 1A2, 2B1, 2B2, 2E1, 3A2, 3A23, and 4A1 and compared with control animals. The maximum fold induction of mRNA varied: 2500-fold for CYP1A1 with BNF, 680-fold for CYP2B1 with PB, 59-fold for CYP3A23 with DEX, and 16-fold for CYP4A1 with CLO. This method was then applied to estimate the inductive potential of putative drug candidates undergoing rodent toxicological evaluation. We present a summary of these data that demonstrates the sensitivity and specificity of the TaqMan assay to distinguish between inducers and noninducers and that offers a highly specific alternative to the quantification of drug effects on P450 expression using immunodetection and substrate metabolism.


Subject(s)
Cytochrome P-450 Enzyme System/biosynthesis , Liver/metabolism , Reverse Transcriptase Polymerase Chain Reaction/methods , Animals , Aryl Hydrocarbon Hydroxylases/biosynthesis , Aryl Hydrocarbon Hydroxylases/genetics , Catalysis , Cytochrome P-450 CYP1A1/biosynthesis , Cytochrome P-450 CYP1A1/genetics , Cytochrome P-450 CYP2B1/biosynthesis , Cytochrome P-450 CYP2B1/genetics , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/genetics , Dexamethasone/pharmacology , Drug Evaluation, Preclinical , Enzyme Induction , Liver/drug effects , Liver/enzymology , Male , Phenobarbital/pharmacology , RNA, Messenger/analysis , RNA, Messenger/metabolism , Rats , Rats, Sprague-Dawley , Reproducibility of Results , Taq Polymerase , beta-Naphthoflavone/pharmacology
9.
Pharm Res ; 22(10): 1667-77, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16180124

ABSTRACT

PURPOSE: Typically, the kinetics of membrane transport is analyzed using the steady-state Michaelis-Menten (or Eadie-Hofstee or Hanes) equations. This approach has been successful when the substrate is picked up from the aqueous phase, like a water-soluble enzyme, for which the Michaelis-Menten steady-state analysis was developed. For membrane transporters whose substrate resides in the lipid bilayer of the plasma membrane, like P-glycoprotein (P-gp), there has been no validation of the accuracy of the steady-state analysis because the elementary rate constants for transport were not known. METHODS: Recently, we fitted the mass action elementary kinetic rate constants of P-gp transport of three different drugs through a confluent monolayer of MDCKII-hMDR1 cells. With these elementary rate constants in hand, we use computer simulations to assess the accuracy of the steady-state Michaelis-Menten parameters. This limits the simulation to parameter ranges known to be physiologically relevant. RESULTS: Using over 2,300 different vectors of initial elementary parameters spanning the space bounded by the three drugs, which defines 2,300 "virtual substrates", the concentrations of substrate transported were calculated and fitted to Eadie-Hofstee plots. Acceptable plots were obtained for 1,338 cases. CONCLUSION: The fitted steady-state Vmax values from the analysis correlated to within a factor of 2-3 with the values predicted from the elementary parameters. However, the fitted Km value could be generated by a wide range of underlying "molecular" Km values. This is because of the convolution of the drug passive permeability kinetics into the fitted Km. This implies that Km values measured in simpler systems, e.g., microsomes or proteoliposomes, even if accurate, would not predict the Km values for the confluent monolayer system or, by logical extension, in vivo. Reliable in vitro-in vivo extrapolation seems to require using the elementary rate constants rather than the Michaelis-Menten steady-state parameters.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Cell Membrane Permeability , Models, Biological , Pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Acridines/pharmacokinetics , Animals , Biological Transport, Active/physiology , Carbamates , Cell Line , Computer Simulation , Dogs , Furans , Humans , Loperamide/pharmacokinetics , Quinidine/pharmacokinetics , Reproducibility of Results , Sulfonamides/pharmacokinetics , Tetrahydroisoquinolines/pharmacokinetics
10.
Bioorg Med Chem Lett ; 15(19): 4370-4, 2005 Oct 01.
Article in English | MEDLINE | ID: mdl-16039851

ABSTRACT

The novel 8-piperazinyl-2,3-dihydropyrroloisoquinoline template was synthesized in nine steps. The template was N-substituted to give a series of compounds showing binding to human cloned 5-HT1A, 5-HT1B and 5-HT1D receptors with pKi's greater than 9 and selectivities up to 1000-fold against other serotonin, dopamine and adrenergic receptors. Several compounds were shown to possess weak partial agonist activity in cloned receptors, which translated to antagonism in in vitro studies.


Subject(s)
Isoquinolines/chemical synthesis , Receptors, Serotonin/drug effects , Serotonin Antagonists/chemical synthesis , Serotonin Receptor Agonists/chemical synthesis , Administration, Oral , Animals , Biological Availability , Brain Chemistry , Isoquinolines/pharmacokinetics , Isoquinolines/pharmacology , Ligands , Rats , Receptor, Serotonin, 5-HT1A , Receptor, Serotonin, 5-HT1B , Receptor, Serotonin, 5-HT1D , Serotonin Antagonists/pharmacokinetics , Serotonin Antagonists/pharmacology , Serotonin Receptor Agonists/pharmacokinetics , Serotonin Receptor Agonists/pharmacology , Structure-Activity Relationship
11.
Biophys J ; 88(1): 715-38, 2005 Jan.
Article in English | MEDLINE | ID: mdl-15501934

ABSTRACT

The human multi-drug resistance membrane transporter, P-glycoprotein, or P-gp, has been extensively studied due to its importance to human health and disease. Thus far, the kinetic analysis of P-gp transport has been limited to steady-state Michaelis-Menten approaches or to compartmental models, neither of which can prove molecular mechanisms. Determination of the elementary kinetic rate constants of transport will be essential to understanding how P-gp works. The experimental system we use is a confluent monolayer of MDCKII-hMDR1 cells that overexpress P-gp. It is a physiologically relevant model system, and transport is measured without biochemical manipulations of P-gp. The Michaelis-Menten mass action reaction is used to model P-gp transport. Without imposing the steady-state assumptions, this reaction depends upon several parameters that must be simultaneously fitted. An exhaustive fitting of transport data to find all possible parameter vectors that best fit the data was accomplished with a reasonable computation time using a hierarchical algorithm. For three P-gp substrates (amprenavir, loperamide, and quinidine), we have successfully fitted the elementary rate constants, i.e., drug association to P-gp from the apical membrane inner monolayer, drug dissociation back into the apical membrane inner monolayer, and drug efflux from P-gp into the apical chamber, as well as the density of efflux active P-gp. All three drugs had overlapping ranges for the efflux active P-gp, which was a benchmark for the validity of the fitting process. One novel finding was that the association to P-gp appears to be rate-limited solely by drug lateral diffusion within the inner monolayer of the plasma membrane for all three drugs. This would be expected if P-gp structure were open to the lipids of the apical membrane inner monolayer, as has been suggested by recent structural studies. The fitted kinetic parameters show how P-gp efflux of a wide range of xenobiotics has been maximized.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/chemistry , Algorithms , Animals , Antibiotics, Antitubercular/pharmacology , Antidiarrheals/pharmacology , Biological Transport , Carbamates , Cell Line , Cell Membrane/metabolism , Cytoplasm/metabolism , Diffusion , Dogs , Dose-Response Relationship, Drug , Enzyme Inhibitors/pharmacology , Furans , Kinetics , Liposomes/chemistry , Loperamide/pharmacology , Models, Chemical , Protein Binding , Quinidine/pharmacology , Software , Sulfonamides/pharmacology , Time Factors , Xenobiotics/pharmacology
12.
J Pharm Sci ; 93(8): 2108-23, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15236458

ABSTRACT

Knowledge of the passive permeability coefficient for new drugs is useful for estimating the fraction absorbed across the gastrointestinal tract. The commonly used approximate formula for the passive permeability coefficient is based on the initial rate of permeation across cell monolayers, requires measurement during the linear phase of permeation, and is not applicable when there is significant back flux of compound or mass balance problem. To develop a rigorous equation that can be used at any time point, i.e., that is valid outside of the linear phase, the mass action equations were integrated for a standard single barrier model of passive permeability. The simple analytical solution found also allows correction for both loss of drug (e.g., due to binding and/or hydrolysis) and sampling volume loss for multiple time point experiments. To test this equation, we measured the passive permeation of three well characterized drugs (amprenavir, quinidine, and loperamide) across confluent monolayers of MDCKII-hMDR1 cells. The potent P-glycoprotein inhibitor GF120918 was used to inhibit P-glycoprotein activity, so only passive permeability was determined. Dramatically different time-dependent behavior was observed for the three compounds, with loperamide showing significant loss of compound, and loperamide and quinidine causing plasma membrane modifications over time. The simple and exact equation for the permeability coefficient developed here works from start of transport to equilibrium, being valid when the commonly used approximate equation may not be. Thus, the exact equation is safer to use in any context, even for single time point estimates in high-throughput permeability assays.


Subject(s)
Pharmacokinetics , Animals , Cell Line , Cell Membrane Permeability , Chromatography, High Pressure Liquid , Dogs
SELECTION OF CITATIONS
SEARCH DETAIL
...