Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 20
Filter
1.
Toxicol Sci ; 167(2): 458-467, 2019 02 01.
Article in English | MEDLINE | ID: mdl-30289550

ABSTRACT

TAK-875 (fasiglifam), a GPR40 agonist in development for the treatment of type 2 diabetes (T2D), was voluntarily terminated in Phase III trials due to adverse liver effects. The potential mechanisms of TAK-875 toxicity were explored by combining in vitro experiments with quantitative systems toxicology (QST) using DILIsym, a mathematical representation of drug-induced liver injury. In vitro assays revealed that bile acid transporters were inhibited by both TAK-875 and its metabolite, TAK-875-Glu. Experimental data indicated that human bile salt export pump (BSEP) inhibition by TAK-875 was mixed whereas sodium taurocholate co-transporting polypeptide (NTCP) inhibition by TAK-875 was competitive. Furthermore, experimental data demonstrated that both TAK-875 and TAK-875-Glu inhibit mitochondrial electron transport chain (ETC) enzymes. These mechanistic data were combined with a physiologically based pharmacokinetic (PBPK) model constructed within DILIsym to estimate liver exposure of TAK-875 and TAK-875-Glu. In a simulated population (SimPops) constructed to reflect T2D patients, 16/245 (6.5%) simulated individuals developed alanine aminotransferase (ALT) elevations, an incidence similar to that observed with 200 mg daily dosing in clinical trials. Determining the mode of bile acid transporter inhibition (Ki) was critical to accurate predictions. In addition, simulations conducted on a sensitive subset of individuals (SimCohorts) revealed that when either BSEP or ETC inhibition was inactive, ALT elevations were not predicted to occur, suggesting that the two mechanisms operate synergistically to produce the observed clinical response. These results demonstrate how utilizing QST methods to interpret in vitro experimental results can lead to an improved understanding of the clinically relevant mechanisms underlying drug-induced toxicity.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 11/antagonists & inhibitors , Benzofurans/toxicity , Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/etiology , Liver/drug effects , Mitochondria/drug effects , Models, Biological , Sulfones/toxicity , Benzofurans/pharmacokinetics , Chemical and Drug Induced Liver Injury/metabolism , Computer Simulation , Humans , Liver/metabolism , Mitochondria/metabolism , Sulfones/pharmacokinetics
2.
Curr Drug Metab ; 17(5): 430-55, 2016.
Article in English | MEDLINE | ID: mdl-26728264

ABSTRACT

BACKGROUND: ABCB1 is a key ABC efflux transporter modulating the pharmacokinetics of a large percentage of drugs. ABCB1 is also a site of transporter mediated drug-drug interactions (tDDI). It is the transporter most frequently tested for tDDIs both in vitro and in the clinic. OBJECTIVE: Understanding the limitations of various in vitro and in vivo models, therefore, is crucial. In this review we cover regulatory aspects of ABCB1 mediated drug transport as well as inhibition and the available models and methods. We also discuss protein structure and mechanistic aspects of transport as ABCB1 displays complex kinetics that involves multiple binding sites, potentiation of transport and probe-dependent IC50 values. RESULTS: Permeability of drugs both passive and mediated by transporters is also a covariate that modulates apparent kinetic values. Levels of expression as well as lipid composition of the expression system used in in vitro studies have also been acknowledged as determinates of transporter activity. ABCB1-mediated clinical tDDIs are often complex as multiple transporters as well as metabolic enzymes may play a role. This complexity often masks the role of ABCB1 in tDDIs. CONCLUSION: It is expected that utilization of in vitro data will further increase with the refinement of simulations. It is also anticipated that transporter humanized preclinical models have a significant impact and utility.


Subject(s)
Drug Interactions , ATP Binding Cassette Transporter, Subfamily B/metabolism , Animals , Biological Assay , Drug Approval , Humans , Pharmaceutical Preparations/metabolism
3.
J Membr Biol ; 248(6): 967-77, 2015 Dec.
Article in English | MEDLINE | ID: mdl-25926125

ABSTRACT

ATP-binding cassette sub-family B member 1 (ABCB1) [P-glycoprotein (P-gp), multidrug resistance protein 1 (MDR1)] can affect the pharmacokinetics, safety, and efficacy of drugs making it important to identify compounds that interact with ABCB1. The ATPase assay and vesicular transport (VT) assay are membrane based assays that can be used to measure the interaction of compounds with ABCB1 at a lower cost and higher throughput compared to cellular-based assays and therefore can be used earlier in the drug development process. To that end, we tested compounds previously identified as ABCB1 substrates and inhibitors for interaction with ABCB1 using the ATPase and VT assays. All compounds tested interacted with ABCB1 in both the ATPase and VT assays. All compounds previously identified as ABCB1 substrates activated ABCB1-mediated ATPase activity in the ATPase assay. All compounds previously identified as ABCB1 inhibitors inhibited the ABCB1-mediated transport in the VT assay. Interestingly, six of the ten compounds previously identified as ABCB1 inhibitors activated the basal ATPase activity in activation assays suggesting that the compounds are substrates of ABCB1 but can inhibit ABCB1 in inhibition assays. Importantly, for ATPase activators the EC50 of activation correlated with the IC50 values from the VT assay showing that interactions of compounds with ABCB1 can be measured with similar levels of potency in either assay. For ATPase nonactivators the IC50 values from the ATPase inhibition and VT inhibition assay showed correlation. These results demonstrate the utility of membrane assays as tools to detect and rank order drug-transporter interactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Cell Membrane/metabolism , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Adenosine Triphosphatases/antagonists & inhibitors , Adenosine Triphosphatases/metabolism , Animals , Biological Transport , Cell Line , Colchicine/pharmacology , Dose-Response Relationship, Drug , Enzyme Activation , Humans , Inhibitory Concentration 50 , Kinetics , Paclitaxel/pharmacology
4.
Eur J Pharm Sci ; 49(4): 773-81, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23684934

ABSTRACT

Accurate determination of potential drug-drug interaction mediated by efflux transporters (tDDI) is crucial to assess the risk of pharmacokinetic interaction and toxicity of drugs. Passive permeability and uptake transporter mediated transport are important covariates of cell-based inhibition assays that need to be taken into consideration when performing kinetic analysis of data. Vesicular uptake inhibition has been considered by regulatory agencies as a viable alternative for testing tDDI potential of low passive permeability drugs in particular. Membranes prepared from a P-gp overexpressing human cell line has superior transport properties over membranes prepared from Sf9 cells and cholesterol enriched Sf9 membranes. P-gp expressed in this membrane effluxes N-methyl-quinidine (NMQ) with high affinity (K(m) is 3.65 µM) and a high rate (V(max) is 656 pmol/mg protein/min). Digoxin, vinblastine and paclitaxel, established P-gp substrates inhibited transport of NMQ with estimated K(i) values of 250, 0.1 and 0.6 µM, respectively. A panel of 11 drugs that have been listed by regulatory agencies as reference inhibitors were used to validate the assay to predict clinical inhibition potential. All the drugs that have been implicated in P-gp mediated DDI were found to be inhibitors in a relevant concentration range.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Biological Assay , Drug Interactions , Quinolines/metabolism , Animals , Biological Transport , Cell Line , Humans , Insecta , K562 Cells , Pharmaceutical Preparations/metabolism , Reproducibility of Results , Transport Vesicles
5.
Drug Metab Dispos ; 41(7): 1347-66, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620485

ABSTRACT

A P-glycoprotein (P-gp) IC50 working group was established with 23 participating pharmaceutical and contract research laboratories and one academic institution to assess interlaboratory variability in P-gp IC50 determinations. Each laboratory followed its in-house protocol to determine in vitro IC50 values for 16 inhibitors using four different test systems: human colon adenocarcinoma cells (Caco-2; eleven laboratories), Madin-Darby canine kidney cells transfected with MDR1 cDNA (MDCKII-MDR1; six laboratories), and Lilly Laboratories Cells--Porcine Kidney Nr. 1 cells transfected with MDR1 cDNA (LLC-PK1-MDR1; four laboratories), and membrane vesicles containing human P-glycoprotein (P-gp; five laboratories). For cell models, various equations to calculate remaining transport activity (e.g., efflux ratio, unidirectional flux, net-secretory-flux) were also evaluated. The difference in IC50 values for each of the inhibitors across all test systems and equations ranged from a minimum of 20- and 24-fold between lowest and highest IC50 values for sertraline and isradipine, to a maximum of 407- and 796-fold for telmisartan and verapamil, respectively. For telmisartan and verapamil, variability was greatly influenced by data from one laboratory in each case. Excluding these two data sets brings the range in IC50 values for telmisartan and verapamil down to 69- and 159-fold. The efflux ratio-based equation generally resulted in severalfold lower IC50 values compared with unidirectional or net-secretory-flux equations. Statistical analysis indicated that variability in IC50 values was mainly due to interlaboratory variability, rather than an implicit systematic difference between test systems. Potential reasons for variability are discussed and the simplest, most robust experimental design for P-gp IC50 determination proposed. The impact of these findings on drug-drug interaction risk assessment is discussed in the companion article (Ellens et al., 2013) and recommendations are provided.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Digoxin/pharmacokinetics , Risk Assessment , Animals , Biological Transport , Caco-2 Cells , Dogs , Drug Interactions , Humans , Inhibitory Concentration 50 , LLC-PK1 Cells , Principal Component Analysis , Swine
6.
Drug Metab Dispos ; 41(7): 1367-74, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23620486

ABSTRACT

In the 2012 Food and Drug Administration (FDA) draft guidance on drug-drug interactions (DDIs), a new molecular entity that inhibits P-glycoprotein (P-gp) may need a clinical DDI study with a P-gp substrate such as digoxin when the maximum concentration of inhibitor at steady state divided by IC50 ([I1]/IC50) is ≥0.1 or concentration of inhibitor based on highest approved dose dissolved in 250 ml divide by IC50 ([I2]/IC50) is ≥10. In this article, refined criteria are presented, determined by receiver operating characteristic analysis, using IC50 values generated by 23 laboratories. P-gp probe substrates were digoxin for polarized cell-lines and N-methyl quinidine or vinblastine for P-gp overexpressed vesicles. Inhibition of probe substrate transport was evaluated using 15 known P-gp inhibitors. Importantly, the criteria derived in this article take into account variability in IC50 values. Moreover, they are statistically derived based on the highest degree of accuracy in predicting true positive and true negative digoxin DDI results. The refined criteria of [I1]/IC50 ≥ 0.03 and [I2]/IC50 ≥ 45 and FDA criteria were applied to a test set of 101 in vitro-in vivo digoxin DDI pairs collated from the literature. The number of false negatives (none predicted but DDI observed) were similar, 10 and 12%, whereas the number of false positives (DDI predicted but not observed) substantially decreased from 51 to 40%, relative to the FDA criteria. On the basis of estimated overall variability in IC50 values, a theoretical 95% confidence interval calculation was developed for single laboratory IC50 values, translating into a range of [I1]/IC50 and [I2]/IC50 values. The extent by which this range falls above the criteria is a measure of risk associated with the decision, attributable to variability in IC50 values.


Subject(s)
Digoxin/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Decision Trees , Drug Interactions , Humans , ROC Curve , United States , United States Food and Drug Administration
7.
Curr Protoc Toxicol ; Chapter 23: Unit 23.4, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23169269

ABSTRACT

The canalicular membrane of hepatocytes contains several transport proteins that use the energy of ATP to efflux potentially toxic molecules to the bile. Probably the two most important proteins at this location are MRP2 and BSEP, which transport phase II conjugates of xenobiotics and endobiotics and conjugated bile salts, respectively. The impaired function of either of these transporter proteins reduces the clearance of the toxic conjugates, resulting in their accumulation in the hepatocytes and eventually the plasma. Conjugated bile salts and phase II metabolites are compounds with low passive permeability; therefore, the most commonly used test system to investigate MRP2- and BSEP-mediated transport processes is the vesicular transport assay. The concentration of probe substrates and inhibitors used in the experiment is close to their free concentration in the hepatocytes, providing an advantage when calculating kinetic parameters (K(m), K(i), V(max)). The protocols aim to assist scientists to set up a transport assay for a known or potential substrate and test small molecule inhibition of the transporters.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/metabolism , Multidrug Resistance-Associated Proteins/antagonists & inhibitors , Multidrug Resistance-Associated Proteins/metabolism , Transport Vesicles/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , Benzbromarone , Bile Acids and Salts , Biological Transport, Active/drug effects , Biological Transport, Active/physiology , Cell Membrane/metabolism , Cyclosporine , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Multidrug Resistance-Associated Protein 2 , Reproducibility of Results , Substrate Specificity , Transport Vesicles/drug effects
8.
Drug Metab Pharmacokinet ; 27(3): 349-53, 2012.
Article in English | MEDLINE | ID: mdl-22790065

ABSTRACT

We are showing that chlorothiazide, a diuretic, is an ABCG2 substrate. It is a Biopharmaceutics Classification System/Biopharmaceutics Drug Distribution and Classification System (BCS/BDDCS) Class IV drug with low bioavailability. Therefore, we tested if chlorothiazide interacts with major apically located intestinal efflux transporters. Our data show that chlorothiazide is transported by ABCG2 with a Km value of 334.6 µM and does not interact with ABCB1 or ABCC2. The chlorothiazide-ABCG2 interaction results in a vectorial transport in MDCKII-BCRP and Caco-2 cells with efflux ratios of 36 and 8.1 respectively. Inhibition of ABCG2 in Caco-2 cells reduced the efflux ratio to 1.4, suggesting that ABCG2 plays a role in limiting chlorothiazide bioavailability in humans.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cell Membrane Permeability , Chlorothiazide/metabolism , Diuretics/metabolism , Enterocytes/metabolism , Neoplasm Proteins/metabolism , Sodium Chloride Symporter Inhibitors/metabolism , ATP Binding Cassette Transporter, Subfamily B , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphate/antagonists & inhibitors , Adenosine Triphosphate/metabolism , Animals , Biological Transport/drug effects , Caco-2 Cells , Cell Membrane Permeability/drug effects , Dogs , Enterocytes/drug effects , Estrone/analogs & derivatives , Estrone/metabolism , Humans , Intestinal Absorption/drug effects , Kinetics , Madin Darby Canine Kidney Cells , Membrane Transport Modulators/pharmacology , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/genetics , Multidrug Resistance-Associated Proteins/metabolism , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/genetics , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Transport Vesicles/drug effects , Transport Vesicles/metabolism
9.
Toxicol In Vitro ; 26(8): 1294-9, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22120137

ABSTRACT

Bile salt export pump (BSEP, ABC11) is a membrane protein that is localized in the cholesterol-rich canalicular membrane of hepatocytes. Its function is to eliminate unconjugated and conjugated bile acids/salts from hepatocyte into the bile. In humans there is no compensatory mechanism for the loss of this transporter. Mutations of BSEP result in a genetic disease, called progressive familial intrahepatic cholestasis type 2 (PFIC2), that is characterized with decreased biliary bile salt secretion, leading to decreased bile flow and accumulation of bile salts inside the hepatocyte, inflicting damage. BSEP inhibitor drugs produce similar bile salt retention that may lead to severe cholestasis and liver damage. Drug-induced liver injury is a relevant clinical issue, in severe cases ending in liver transplantation. Therefore, measurement of BSEP inhibition by candidate drugs has high importance in drug discovery and development. Although several methods are suitable to detect BSEP-drug interactions, due to interspecies differences in bile acid composition, differences in hepatobiliary transporter modulation, they have limitations. This review summarizes appropriate in vitro methods that could be able to predict BSEP-drug candidate interactions in humans before the start of clinical phases.


Subject(s)
ATP-Binding Cassette Transporters/antagonists & inhibitors , Chemical and Drug Induced Liver Injury/etiology , Cholestasis/chemically induced , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/genetics , Animals , Bile Acids and Salts/metabolism , Chemical and Drug Induced Liver Injury/physiopathology , Cholestasis/physiopathology , Cholestasis, Intrahepatic/genetics , Cholestasis, Intrahepatic/physiopathology , Drug Design , Humans , Severity of Illness Index , Species Specificity
10.
J Biomol Screen ; 16(8): 886-94, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21832259

ABSTRACT

This study provides evidence that quinidine can be used as a probe substrate for ABCB1 in multiple experimental systems both in vitro and in vivo relevant to the blood-brain barrier (BBB). The combination of quinidine and PSC-833 (valspodar) is an effective tool to assess investigational drugs for interactions on ABCB1. Effects of quinidine and substrate-inhibitor interactions were tested in a membrane assay and in monolayer assays. The authors compared quinidine and digoxin as ABCB1 probes in the in vitro assays and found that quinidine was more potent and at least as specific as digoxin in ATPase and monolayer efflux assays employing MDCKII-MDR1 and the rat brain microcapillary endothelial cell system. Brain exposure to quinidine was tested in dual-/triple-probe microdialysis experiments in rats by assessing levels of quinidine in blood and brain. Comparing quinidine levels in dialysate samples from valspodar-treated and control animals, it is evident that systemic/local administration of the inhibitor diminishes the pumping function of ABCB1 at the BBB, resulting in an increased brain penetration of quinidine. In sum, quinidine is a good probe to study ABCB1 function at the BBB. Moreover, quinidine/PSC-833 is an ABCB1-specific substrate/inhibitor combination applicable to many assay systems both in vitro and in vivo.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B/metabolism , Blood-Brain Barrier/drug effects , Brain/drug effects , Drug Evaluation, Preclinical/methods , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , High-Throughput Screening Assays , ATP Binding Cassette Transporter, Subfamily B/antagonists & inhibitors , Animals , Biological Products/analysis , Blood-Brain Barrier/metabolism , Brain/cytology , Brain/metabolism , Cell Line , Chromatography, High Pressure Liquid , Coculture Techniques , Cyclosporins/pharmacology , Digoxin/pharmacology , Dogs , Drug Combinations , Drug Interactions , Endothelial Cells/cytology , Endothelial Cells/metabolism , Endothelium, Vascular/cytology , Endothelium, Vascular/metabolism , Enzyme Inhibitors/pharmacology , Immunosuppressive Agents/pharmacology , Male , Microdialysis , Quinidine/pharmacology , Rats , Rats, Wistar
11.
J Biomol Screen ; 16(1): 112-9, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21057029

ABSTRACT

The multidrug resistance protein 1 (MDR1) transporter is the most abundantly investigated adenosine triphosphate (ATP)-Binding Cassette (ABC) transporter protein. Multiple assay systems were developed to study MDR1-mediated transport and possible drug-drug interactions. Yet, as different probe substrates are used in these assays, it is difficult to directly compare the results. In this study, a common probe substrate was applied in 3 assay systems developed to study MDR1: the cellular dye efflux assay, the ATPase assay, and the vesicular transport assay. This probe substrate is calcein acetoxymethyl ester (calcein AM), the acetoxymethyl ester derivative of the fluorescent dye, calcein. Using a common probe allows the investigation of the effect of passive permeability on the result obtained by testing various compounds. In this study, 22 compounds with different logP values were tested in the above-mentioned 3 assay systems. The vesicular transport assay proved most sensitive, detecting 18 of 22 interactions with the protein. The ATPase assay detected 15 interactions, whereas the cellular dye efflux assay was the least sensitive with only 10 hits. A correlation was found between the hydrophobicity of the compound and the ratio of cellular and vesicular transport IC(50) values, indicating the effect of passive permeability on the result. Based on hydrophobicity, the current study provides guidelines on applying the most correct tool for studying MDR1 interactions.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Biological Assay , Biological Transport, Active/drug effects , Fluoresceins/metabolism , Permeability/drug effects , Fluorescent Dyes/metabolism , Humans , Hydrophobic and Hydrophilic Interactions , Inhibitory Concentration 50 , Protein Binding
12.
Drug Metab Rev ; 42(3): 402-36, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20082599

ABSTRACT

ABCC2/Abcc2 (MRP2/Mrp2) is expressed at major physiological barriers, such as the canalicular membrane of liver cells, kidney proximal tubule epithelial cells, enterocytes of the small and large intestine, and syncytiotrophoblast of the placenta. ABCC2/Abcc2 always localizes in the apical membranes. Although ABCC2/Abcc2 transports a variety of amphiphilic anions that belong to different classes of molecules, such as endogenous compounds (e.g., bilirubin-glucuronides), drugs, toxic chemicals, nutraceuticals, and their conjugates, it displays a preference for phase II conjugates. Phenotypically, the most obvious consequence of mutations in ABCC2 that lead to Dubin-Johnson syndrome is conjugate hyperbilirubinemia. ABCC2/Abcc2 harbors multiple binding sites and displays complex transport kinetics.


Subject(s)
Multidrug Resistance-Associated Proteins/metabolism , Animals , Biological Transport, Active , Cloning, Molecular , Drug Resistance, Multiple , Humans , Kinetics , Mice , Mice, Knockout , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/biosynthesis , Multidrug Resistance-Associated Proteins/chemistry , Multidrug Resistance-Associated Proteins/genetics , Protein Conformation , Xenobiotics/metabolism
14.
Drug Metab Dispos ; 37(9): 1878-86, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19520776

ABSTRACT

The efflux transporter responsible for the canalicular elimination of bile salts from the hepatocytes is the bile salt export pump (BSEP, ABCB11). Absence or inhibition of this transporter leads to bile salt retention in the hepatocyte and in turn can lead to cholestatic liver disease. We expressed the BSEP/Bsep protein from three species (human, rat, and mouse) in a baculovirus-infected Sf9 system. Vesicles prepared from these cells were used to evaluate bile salt transport of four conjugated bile salts. Because the Sf9 system contains less membrane cholesterol than the liver canalicular membrane, the effect of added cholesterol on the kinetics of BSEP/Bsep-mediated bile salt transport was also investigated. Cholesterol treatment increased the V(max) values in all the species, with the most pronounced effect observed in the rat transporter. In contrast, K(m) values, with the exception of glycochenodeoxycholate, remained largely unchanged. The species-specific bile salt transport inhibition potential of three compounds known to cause clinical cholestasis was investigated in vesicles containing BSEP/Bsep. Troglitazone and glibenclamide inhibited the BSEP/Bsep-mediated transport of different bile salts with similar affinities, whereas the potential of cyclosporine A to inhibit bile salt transport showed species- and bile salt-specific variations. In conclusion, the cholesterol-loaded Sf9 vesicles overexpressing BSEP/Bsep seem to be a useful system for the identification of potential cholestatic compounds and can also be used for the investigation of species specificity. We observed greater differences in IC(50) values for inhibitors than in K(m) values for substrates between species.


Subject(s)
ATP-Binding Cassette Transporters/metabolism , Cholesterol/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , Bile Acids and Salts/metabolism , Biological Transport, Active , Blotting, Western , Cell Line , Cell Membrane/metabolism , Chromans/metabolism , Cyclosporine/metabolism , Electrophoresis, Polyacrylamide Gel , Glyburide/metabolism , Humans , Hypoglycemic Agents/metabolism , Immunosuppressive Agents/metabolism , Insecta/metabolism , Kinetics , Mice , Rats , Species Specificity , Substrate Specificity , Thiazolidinediones/metabolism , Troglitazone , Vesicular Transport Proteins/metabolism
15.
J Biomol Screen ; 14(1): 10-5, 2009 Jan.
Article in English | MEDLINE | ID: mdl-19029016

ABSTRACT

The mouse ortholog of the human bile salt export pump (BSEP) transporter was expressed in a baculovirus-infected insect cell (Sf9) system to study the effect of membrane cholesterol content on the transporter function. The transport activity of cholesterol-loaded mouse Bsep-HAM-Sf9 vesicles was determined in a vesicular transport assay with taurochenodeoxycholate (TCDC), a known BSEP substrate. Mouse Bsep transports TCDC at a high rate that can be sensitively detected in the ATPase assay. Cholesterol upload of the Sf9 membrane potentiates both TCDC transport and TCDC-stimulated ATPase activities. Inhibitory effect of BSEP interactors on probe substrate transport was tested in both vesicular transport and ATPase assays using cholesterol-loaded membrane vesicles. A good rank order correlation was found between IC(50) values measured in TCDC-stimulated mBsep ATPase assay and in the human BSEP vesicular transport assay utilizing taurocholate (TC) as probe substrate. This upgraded form of the mouse Bsep-HAM ATPase assay is a user friendly, sensitive, nonradioactive method for early high-throughput screening of drugs with BSEP-related cholestatic potential. It may complement the human BSEP-mediated taurocholate vesicular transport inhibition assay.


Subject(s)
ATP-Binding Cassette Transporters/analysis , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphatases/analysis , Adenosine Triphosphatases/metabolism , Cholesterol/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 11 , ATP-Binding Cassette Transporters/antagonists & inhibitors , ATP-Binding Cassette Transporters/genetics , Adenosine Triphosphatases/antagonists & inhibitors , Animals , Biological Transport/drug effects , Cell Line , Cholestasis/drug therapy , Cholesterol/pharmacology , Mice , Radioligand Assay , Spodoptera
16.
Expert Opin Drug Metab Toxicol ; 4(6): 721-32, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18611113

ABSTRACT

BACKGROUND: The last 15 years have marked an expansion in our understanding of how ABC transporters modulate the pharmacokinetic properties of drugs. Assays based on different membrane preparations were one of the first methods developed to study ABC transporters. Later, they turned out to be valuable tools to gain insight into the nature of drug-ABC transporter interactions. OBJECTIVES: Membranes prepared from different sources have been used and characterized; based on the biochemical characteristics of the transport process, a number of different assay types have been developed. METHODS: This review focuses on the current experiences on how different membrane-based assays can be utilized in pharmaceutical R&D. Sources of membrane preparations, available assay types and correlation studies between different in-vitro and in-vivo methods are discussed. RESULTS/CONCLUSION: Membrane-based assays are valuable tools in drug discovery to characterize drug-ABC transporter interactions.


Subject(s)
ATP-Binding Cassette Transporters/drug effects , Biological Assay/methods , Cell Membrane/drug effects , ATP-Binding Cassette Transporters/metabolism , Adenosine Triphosphate/metabolism , Animals , Biological Transport/drug effects , Cell Membrane/metabolism , Dose-Response Relationship, Drug , Humans , Hydrolysis , Kinetics , Recombinant Proteins/drug effects , Reproducibility of Results , Subcellular Fractions
17.
J Biomol Screen ; 13(4): 295-301, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18349419

ABSTRACT

MRP2 (ABCC2) is an efflux transporter expressed on the apical membrane of polarized cells. This protein has a major role in the biliary elimination of toxic compounds from the liver. As MRP2 transports many endogenous compounds, including LTC4 as well as xenobiotics and toxic phase II metabolites, blockade of this transporter may cause the accumulation of these compounds in the hepatocyte, resulting in hepatotoxicity. The vesicular transport assay is a great tool to study drug-drug and drug-endogenous compound interactions of ABC transporters. In this assay, inside-out membrane vesicles are used, so the test compound can readily access the transporter. As MRP2 transports many ionic compounds that are difficult to investigate in a whole-cell system because of permeability reasons, the vesicular transport assay is a good choice for screening MRP2-mediated interactions. LTC4 is not an optimal substrate for high-throughput screening for MRP2 interactors, even though it is an important MRP2 substrate. Therefore, the transport of a drug surrogate, 5(6)-carboxy-2,'7'-dichlorofluorescein (CDCF), by MRP2 was characterized using the vesicular transport assay. The data indicate that CDCF proves to be an ideal substrate for MRP2 vesicular transport assay with its optimal detection and transport properties.


Subject(s)
Fluoresceins/metabolism , Fluorescent Dyes/metabolism , Leukotriene C4/metabolism , Multidrug Resistance-Associated Proteins/metabolism , Biological Transport , Humans , Kinetics , Multidrug Resistance-Associated Protein 2 , Reproducibility of Results
18.
J Pept Sci ; 12(11): 714-20, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16967433

ABSTRACT

Analogs of the decapeptide, gonadotropin-releasing hormone (GnRH), used in the treatment of hormone-dependent tumors, contain numerous unnatural amino acids, giving rise to many adverse effects. lGnRH-III, a natural isoform of GnRH isolated from the sea lamprey, is a weak agonist of GnRH in the pituitary, but inhibits the growth of human cancer cells in micromolar concentrations. As lGnRH-III is not a natural ligand in humans, it is possible that a more potent peptide, also containing only natural amino acids, can be synthesized. A positional scanning peptide library, focused on the variable region of the GnRH family of peptides, residues 5-8, was synthesized. The synthesized peptides were analyzed in competitive binding experiments and six new analogs were designed on the basis of the results. Their biological activities were evaluated in cell growth experiments. The only natural sequence selected was chicken GnRH-II. The synthetic library did not yield a more potent peptide than lGnRH-III.


Subject(s)
Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/analogs & derivatives , Oligopeptides/pharmacology , Peptides/chemistry , Pyrrolidonecarboxylic Acid/analogs & derivatives , Antineoplastic Agents/metabolism , Breast Neoplasms/pathology , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Female , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/metabolism , Gonadotropin-Releasing Hormone/pharmacology , Humans , Oligopeptides/chemistry , Peptide Library , Peptides/metabolism , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/pharmacology , Tumor Cells, Cultured
19.
Peptides ; 26(3): 419-22, 2005 Mar.
Article in English | MEDLINE | ID: mdl-15652648

ABSTRACT

Naturally occurring isoforms of the decapeptide gonadotropin-releasing hormone (GnRH) share residues 1-4 and 9-10. lGnRH-III, the third isoform isolated in the sea lamprey has no endocrine effect in mammals but shows a direct antiproliferative effect on human breast, prostate and endometrial cancer cell lines. To investigate these features, residues 5-8 of lGnRH-III were systematically replaced with Ala. The ability of the synthetic analogs to interact with receptors on MDA-MB 231 human breast cancer cells and their effect on the growth of the same cell line were investigated. [Ala6]lGnRH-III and [Ala7]lGnRH-III have neither receptor binding nor antiproliferative activity. Replacement of His5 with Ala resulted in an analog that binds to the receptor but does not have antiproliferative activity. The results are in agreement with previous reports that modifications of Lys at position 8 are well tolerated.


Subject(s)
Breast Neoplasms/drug therapy , Gonadotropin-Releasing Hormone/chemistry , Gonadotropin-Releasing Hormone/metabolism , Oligopeptides/chemistry , Oligopeptides/metabolism , Pyrrolidonecarboxylic Acid/analogs & derivatives , Pyrrolidonecarboxylic Acid/chemistry , Pyrrolidonecarboxylic Acid/metabolism , Alanine/chemistry , Animals , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Histidine/chemistry , Humans , Inhibitory Concentration 50 , Lampreys , Lysine/chemistry , Peptides/therapeutic use , Protein Binding , Protein Isoforms , Protein Structure, Tertiary , Radioligand Assay
20.
FEBS Lett ; 556(1-3): 199-203, 2004 Jan 02.
Article in English | MEDLINE | ID: mdl-14706850

ABSTRACT

Gastrin (G17) and N-carboxymethylgastrin (G17-Gly) have been shown to stimulate the growth of colon cancer cells both in vivo and in vitro. The identity of the receptor mediating these effects is controversial. A recent study demonstrated the presence of a low affinity binding site for G17 and G17-Gly on the DLD-1 human colon cancer cell line. The goal of the current study was to further investigate the role of this receptor in mediating the growth-promoting effects of gastrin peptides. Binding of [Leu(15)]G17 and [Leu(15)]G17-Gly to DLD-1 cell membranes in competition with [(3)H]G17-Gly was examined. Binding of [(3)H]cholecystokinin-8 (CCK8) to DLD-1 cell membranes was also assessed. Whole cell binding experiments were carried out using [(125)I-Tyr(12),Leu(15)]G17-Gly. In addition, the ability of [Leu(15)]G17 and [Leu(15)]G17-Gly to stimulate cell growth, as determined by cell counting, was tested. [Leu(15)]G17 and [Leu(15)]G17-Gly competed with [(3)H]G17-Gly at both a high and a low affinity site on DLD-1 membranes. The IC(50) values for [Leu(15)]G17 were 6.0 x 10(-8) M and 6.9 x 10(-6) M while those for [Leu(15)]G17-Gly were 3.2 x 10(-9) M and 4.9 x 10(-6) M. [(3)H]CCK8 did not bind to either site. [Leu(15)]G17-Gly also competed with [(125)I-Tyr(12),Leu(15)]G17-Gly at both a high and a low affinity site on DLD-1 cells with similar affinities as observed with membranes. [Leu(15)]G17 and [Leu(15)]G17-Gly significantly stimulated the growth of DLD-1 cells in a dose-dependent and biphasic manner. The binding profiles of the peptides tested suggest that these sites are different from previously identified wild-type and mutant CCK(1) or CCK(2) receptors.


Subject(s)
Colorectal Neoplasms/metabolism , Gastrins/physiology , Mitogens/physiology , Receptor, Cholecystokinin B/metabolism , Animals , CHO Cells , Cell Line, Tumor , Cell Membrane/metabolism , Colorectal Neoplasms/pathology , Cricetinae , Dose-Response Relationship, Drug , Gastrins/chemistry , Humans , Mitogens/chemistry , Radioligand Assay , Receptors, Cholecystokinin/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...