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










Publication year range
1.
Drug Metab Dispos ; 38(3): 459-73, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20016052

ABSTRACT

The pharmacokinetics and metabolism of anacetrapib (MK-0859), a novel cholesteryl ester transfer protein inhibitor, were examined in rats and rhesus monkeys. Anacetrapib exhibited a low clearance in both species and a moderate oral bioavailability of approximately 38% in rats and approximately 13% in monkeys. The area under the plasma concentration-time curve in both species increased in a less than dose-proportional manner over an oral dose range of 1 to 500 mg/kg. After oral administration of [(14)C]anacetrapib at 10 mg/kg, approximately 80 and 90% of the radioactive dose was recovered over 48 h postdose from rats and monkeys, respectively. The majority of the administered radioactive dose was excreted unchanged in feces in both species. Biliary excretion of radioactivity accounted for approximately 15% and urinary excretion for less than 2% of the dose. Thirteen metabolites, resulting from oxidative and secondary glucuronic acid conjugation, were identified in rat and monkey bile. The main metabolic pathways consisted of O-demethylation (M1) and hydroxylation on the biphenyl moiety (M2) and hydroxylation on the isopropyl side chain (M3); these hydroxylations were followed by O-glucuronidation of these metabolites. A glutathione adduct (M9), an olefin metabolite (M10), and a propionic acid metabolite (M11) also were identified. In addition to parent anacetrapib, M1, M2, and M3 metabolites were detected in rat but not in monkey plasma. Overall, it appears that anacetrapib exhibits a low-to-moderate degree of absorption after oral dosing and majority of the absorbed dose is eliminated via oxidation to a series of hydroxylated metabolites that undergo conjugation with glucuronic acid before excretion into bile.


Subject(s)
Anticholesteremic Agents/pharmacokinetics , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Oxazolidinones/pharmacokinetics , Animals , Anticholesteremic Agents/administration & dosage , Anticholesteremic Agents/analysis , Anticholesteremic Agents/chemistry , Bile/chemistry , Biotransformation , Blood Chemical Analysis , Drug Evaluation, Preclinical , Feces/chemistry , Glucuronides/analysis , Glucuronides/blood , Glucuronides/chemistry , Glucuronides/urine , Hydroxylation , Intestinal Absorption , Macaca mulatta , Magnetic Resonance Spectroscopy , Male , Molecular Structure , Oxazolidinones/administration & dosage , Oxazolidinones/analysis , Oxazolidinones/chemistry , Rats , Rats, Sprague-Dawley , Species Specificity , Tandem Mass Spectrometry , Urine/chemistry
2.
Drug Metab Dispos ; 38(3): 474-83, 2010 Mar.
Article in English | MEDLINE | ID: mdl-20016053

ABSTRACT

Anacetrapib is a novel cholesteryl ester transfer protein inhibitor being developed for the treatment of primary hypercholesterolemia and mixed dyslipidemia. The absorption, distribution, metabolism, and excretion of anacetrapib were investigated in an open-label study in which six healthy male subjects received a single oral dose of 150 mg and 165 microCi of [(14)C]anacetrapib. Plasma, urine, and fecal samples were collected at predetermined times for up to 14 days postdose and were analyzed for total radioactivity, the parent compound, and metabolites. The majority of the administered radioactivity (87%) was eliminated by fecal excretion, with negligible amounts present in urine (0.1%). The peak level of radioactivity in plasma (approximately 2 microM equivalents of [(14)C]anacetrapib) was achieved approximately 4 h postdose. The parent compound was the major radioactive component (79-94% of total radioactivity) in both plasma and feces. Three oxidative metabolites, M1, M2, and M3, were detected in plasma and feces and were identified as the O-demethylated species (M1) and two secondary hydroxylated derivatives of M1 (M2 and M3). Each metabolite was detected at low levels, representing

Subject(s)
Anticholesteremic Agents/pharmacokinetics , Cholesterol Ester Transfer Proteins/antagonists & inhibitors , Oxazolidinones/pharmacokinetics , Adolescent , Adult , Anticholesteremic Agents/adverse effects , Anticholesteremic Agents/metabolism , Biotransformation , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Cytochrome P-450 Enzyme System/metabolism , Feces/chemistry , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Male , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Middle Aged , Molecular Structure , Oxazolidinones/adverse effects , Oxazolidinones/metabolism , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Tandem Mass Spectrometry , Young Adult
3.
Bioorg Med Chem Lett ; 19(17): 5033-6, 2009 Sep 01.
Article in English | MEDLINE | ID: mdl-19631535

ABSTRACT

During our effort to design a receptor binding assay to aid in the elucidation of the molecular mechanism of ezetimibe, we prepared a sulfur-35 containing radioligand which exhibits improved potency over the glucuronide conjugate of ezetimibe in both native enterocyte brush border membranes and membranes from cells expressing recombinant NPC1L1. Herein, we describe the different synthetic strategies which were used to obtain this compound as well as its effectiveness in the aforementioned assay.


Subject(s)
Anticholesteremic Agents/chemistry , Azetidines/chemistry , Membrane Proteins/antagonists & inhibitors , Animals , Anticholesteremic Agents/chemical synthesis , Anticholesteremic Agents/pharmacology , Azetidines/chemical synthesis , Azetidines/pharmacology , Cell Line , Ezetimibe , Glucuronides/chemistry , Humans , Ligands , Membrane Proteins/metabolism , Membrane Transport Proteins , Mice , Protein Binding , Rats , Recombinant Proteins/antagonists & inhibitors , Recombinant Proteins/metabolism , Sulfur Radioisotopes/chemistry
5.
Drug Metab Dispos ; 36(2): 469-73, 2008 Feb.
Article in English | MEDLINE | ID: mdl-17998295

ABSTRACT

Recent clinical reports have suggested that the cyclooxygenase-2 inhibitor, lumiracoxib (Prexige), may cause a rare but serious hepatotoxicity in patients. In view of the close structural resemblance between lumiracoxib and diclofenac, a widely used nonsteroidal anti-inflammatory drug whose use also has been associated with rare cases of liver injury, it is possible that the toxicity of the two agents may share a common mechanism. Because it is believed that chemically reactive metabolites may play a role as mediators of diclofenac-mediated hepatotoxicity, the present in vitro study was carried out to test the hypothesis that lumiracoxib also undergoes metabolic activation when incubated with liver microsomal preparations and hepatocytes from rats and humans. By means of liquid chromatography tandem mass spectrometry and nuclear magnetic resonance spectrometry techniques, two previously unknown N-acetylcysteine (NAC) conjugates were identified, namely, 3'-NAC-4'-hydroxy lumiracoxib (M1) and 4'-hydroxy-6'-NAC-desfluoro lumiracoxib (M2), the structures of which reveal the intermediacy of an electrophilic quinone imine species. Based on the results of studies with immunoinhibitory antibodies, it was demonstrated that the formation of M1 and M2 in human liver microsomes was catalyzed by cytochrome P450 (P450) 2C9. These findings demonstrate that lumiracoxib is subject to P450-mediated bioactivation in both rat and human liver preparations, leading to the formation of a reactive intermediate analogous to species generated during the metabolism of diclofenac.


Subject(s)
Cyclooxygenase 2 Inhibitors/metabolism , Diclofenac/analogs & derivatives , Hepatocytes/metabolism , Microsomes, Liver/metabolism , Acetylcysteine/pharmacology , Animals , Cells, Cultured , Chromatography, Liquid , Diclofenac/metabolism , Humans , Rats , Tandem Mass Spectrometry
6.
J Pharmacol Exp Ther ; 321(2): 673-83, 2007 May.
Article in English | MEDLINE | ID: mdl-17314201

ABSTRACT

Sitagliptin, a selective dipeptidyl peptidase 4 inhibitor recently approved for the treatment of type 2 diabetes, is excreted into the urine via active tubular secretion and glomerular filtration in humans. In this report, we demonstrate that sitagliptin is transported by human organic anion transporter hOAT3 (Km=162 microM), organic anion transporting polypeptide OATP4C1, and multidrug resistance (MDR) P-glycoprotein (Pgp), but not by human organic cation transporter 2 hOCT2, hOAT1, oligopeptide transporter hPEPT1, OATP2B1, and the multidrug resistance proteins MRP2 and MRP4. Our studies suggested that hOAT3, OATP4C1, and MDR1 Pgp might play a role in transporting sitagliptin into and out of renal proximal tubule cells, respectively. Sitagliptin did not inhibit hOAT1-mediated cidofovir uptake, but it showed weak inhibition of hOAT3-mediated cimetidine uptake (IC50=160 microM). hOAT3-mediated sitagliptin uptake was inhibited by probenecid, ibuprofen, furosemide, fenofibric acid, quinapril, indapamide, and cimetidine with IC50 values of 5.6, 3.7, 1.7, 2.2, 6.2, 11, and 79 microM, respectively. Sitagliptin did not inhibit Pgp-mediated transport of digoxin, verapamil, ritonavir, quinidine, and vinblastine. Cyclosporine A significantly inhibited Pgp-mediated transport of sitagliptin (IC50=1 microM). Our data indicate that sitagliptin is unlikely to be a perpetrator of drug-drug interactions with Pgp, hOAT1, or hOAT3 substrates at clinically relevant concentrations. Renal secretion of sitagliptin could be inhibited if coadministered with OAT3 inhibitors such as probenecid. However, the magnitude of interactions should be low, and the effects may not be clinically meaningful, due to the high safety margin of sitagliptin.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Adenosine Deaminase Inhibitors , Dipeptidyl-Peptidase IV Inhibitors , Enzyme Inhibitors/metabolism , Glycoproteins/antagonists & inhibitors , Organic Anion Transporters, Sodium-Independent/physiology , Organic Anion Transporters/physiology , Pyrazines/metabolism , Triazoles/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Animals , Biological Transport , CHO Cells , Cricetinae , Cricetulus , Dipeptidyl Peptidase 4 , Humans , Male , Membrane Transport Proteins/physiology , Mice , Multidrug Resistance-Associated Protein 2 , Multidrug Resistance-Associated Proteins/physiology , Sitagliptin Phosphate
8.
Bioorg Med Chem Lett ; 16(6): 1692-5, 2006 Mar 15.
Article in English | MEDLINE | ID: mdl-16377185

ABSTRACT

The synthesis of the first high specific activity S-35-labeled hERG radioligand, [(35)S]MK-0499, for use in HTS assays of drug candidates for hERG interaction is described. The radioligand is prepared by [(35)S]sulfonylation of a high diastereomeric excess (de) aniline precursor prepared from unlabeled MK-0499.


Subject(s)
Benzopyrans/chemistry , Benzopyrans/pharmacology , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Piperidines/chemistry , Piperidines/pharmacology , Potassium Channel Blockers/chemical synthesis , Radiopharmaceuticals/chemical synthesis , Ether-A-Go-Go Potassium Channels/metabolism , Humans , Potassium Channel Blockers/pharmacology , Radioligand Assay , Radiopharmaceuticals/pharmacology , Sulfur Radioisotopes/chemistry
9.
Proc Natl Acad Sci U S A ; 102(23): 8132-7, 2005 Jun 07.
Article in English | MEDLINE | ID: mdl-15928087

ABSTRACT

Ezetimibe is a potent inhibitor of cholesterol absorption that has been approved for the treatment of hypercholesterolemia, but its molecular target has been elusive. Using a genetic approach, we recently identified Niemann-Pick C1-Like 1 (NPC1L1) as a critical mediator of cholesterol absorption and an essential component of the ezetimibe-sensitive pathway. To determine whether NPC1L1 is the direct molecular target of ezetimibe, we have developed a binding assay and shown that labeled ezetimibe glucuronide binds specifically to a single site in brush border membranes and to human embryonic kidney 293 cells expressing NPC1L1. Moreover, the binding affinities of ezetimibe and several key analogs to recombinant NPC1L1 are virtually identical to those observed for native enterocyte membranes. KD values of ezetimibe glucuronide for mouse, rat, rhesus monkey, and human NPC1L1 are 12,000, 540, 40, and 220 nM, respectively. Last, ezetimibe no longer binds to membranes from NPC1L1 knockout mice. These results unequivocally establish NPC1L1 as the direct target of ezetimibe and should facilitate efforts to identify the molecular mechanism of cholesterol transport.


Subject(s)
Azetidines/pharmacology , Membrane Proteins/metabolism , Membrane Transport Proteins/metabolism , Proteins/metabolism , Animals , Azetidines/chemistry , Binding Sites , Cell Line , Cell Membrane/metabolism , Enterocytes/cytology , Enterocytes/metabolism , Ezetimibe , Humans , Intestinal Mucosa/metabolism , Intestines/cytology , Macaca mulatta , Membrane Proteins/genetics , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/genetics , Mice , Mice, Inbred C57BL , Microvilli/metabolism , Niemann-Pick Diseases , Protein Binding , Proteins/genetics , Rats , Rats, Sprague-Dawley , Species Specificity
10.
Article in English | MEDLINE | ID: mdl-15686997

ABSTRACT

To support pharmacokinetic studies, a selective and sensitive liquid chromatography/tandem mass spectrometry (LC-MS/MS) method has been developed and validated for the simultaneous determination of a novel KDR kinase inhibitor (1) and its active metabolite (2) in human plasma. The method is fully automated using a Packard MultiPROBE II system and a TomTec Quadra 96 liquid handling workstation to perform sample preparation and solid-phase extraction (SPE). Following the extraction on a mixed-mode SPE using Oasis MCX 96-well plate, the analytes were separated on a Aquasil C18 column (50 mm x 2.1 mm, i.d., 3 microm) with a mobile phase consisting of acetonitrile/ammonium acetate buffer (5 mM, pH 5.0) (60/40, v/v). The run time for each injection was 4.5 min with the retention times of approximately 2.0 and 2.7 min for 1 and 2 respectively, at a flow rate of 0.25 mL/min. A tandem mass spectrometric detection was conducted using multiple reaction monitoring (MRM) under the positive ion mode with a turbo ion-spray interface. The linear ranges of the calibration curves were 0.05-400 ng/mL for 1 and 0.1-400 ng/mL for 2 on a PE Sciex API 4000 LC-MS/MS system. The lower limits of quantitation (LLOQ) of the assay were 0.05 and 0.1 ng/mL for 1 and 2 respectively, when 0.4 mL of plasma was processed. Intra-day assay precision (using five standard curves prepared by spiking compounds to five lots of plasma) was less than 4.9% for 1 and less than 9.6% for 2 on each concentration. Assay accuracy was found to be 95.1-104.6% of nominal for 1 standards and 93.5-105.6% for 2 standards. QC samples were stable when kept at room temperature for 4 h, at -70 degrees C for 10 days, and after three freeze-thaw cycles. The extraction recoveries were 80%, 83% and 84% for 1 and 2 and I.S. respectively, and no significant matrix effects were observed. The method was successfully applied to plasma samples from clinical studies after oral administration of compound 1.


Subject(s)
Chromatography, High Pressure Liquid/methods , Intracellular Signaling Peptides and Proteins/blood , Mass Spectrometry/methods , Oxides/metabolism , Humans , Reference Standards , Reproducibility of Results , Sensitivity and Specificity
12.
Eur J Pharmacol ; 499(1-2): 77-84, 2004 Sep 19.
Article in English | MEDLINE | ID: mdl-15363953

ABSTRACT

Compound A (N-[2-[4-(4,5-dihydro-1H-imidazol-2-yl)phenyl]ethyl]-2-[(2R)-1-(2-napthylsulfonyl)-3-oxo-1,2,3,4-tetrahydroquinoxalin-2-yl]acetamide) is a member of a new class of aryl sulfonamide dihydroquinoxalinone bradykinin B1 receptor antagonists that should be useful pharmacological tools. Here we report on some of the pharmacological properties of compound A as well as the characterization of [35S]compound A as the first nonpeptide bradykinin B1 receptor radioligand. Compound A inhibited tritiated peptide ligand binding to the cloned human, rabbit, dog, and rat bradykinin B1 receptors expressed in CHO cells with Ki values of 0.016, 0.050, 0.56, and 29 nM, respectively. It was inactive at 10 microM in binding assays with the cloned human bradykinin B2 receptor. In functional antagonist assays with the cloned bradykinin B1 receptors, compound A inhibited agonist-induced signaling with activities consistent with the competition binding results, but had no antagonist activity at the bradykinin B2 receptor. Compound A was also found to be a potent antagonist in a rabbit aorta tissue bath preparation and to effectively block des-Arg9 bradykinin depressor responses in lipopolysaccharide-treated rabbit following intravenous administration. The binding of [35S]compound A was evaluated with the cloned bradykinin B1 receptors. In assays with human, rabbit, and dog receptors, [35S]compound A labeled a single site with Kd values of 0.012, 0.064, and 0.37 nM, respectively, and with binding site densities equivalent to those obtained using the conventional tritiated peptide ligands. Binding assays with the cloned rat bradykinin B1 receptor were not successful, presumably due to the low affinity of the ligand for this species receptor. There was no specific binding of the ligand detected in CHO cells expressing the human bradykinin B2 receptor. In assays with the cloned human bradykinin B1 receptor, the pharmacologies of the binding of [35S]compound A and [3H][Leu9]des-Arg10-kallidin were the same. The high signal-to-noise ratio obtained with [35S]compound A will allow this ligand to be a very useful tool for future investigations of the bradykinin B1 receptor.


Subject(s)
Bradykinin B1 Receptor Antagonists , Kallidin/analogs & derivatives , Receptor, Bradykinin B1/metabolism , Animals , Aorta, Thoracic/drug effects , Aorta, Thoracic/physiology , Binding, Competitive/drug effects , Blood Pressure/drug effects , CHO Cells , Cricetinae , Cricetulus , Dogs , Dose-Response Relationship, Drug , Humans , Imidazoles/metabolism , Imidazoles/pharmacology , In Vitro Techniques , Kallidin/metabolism , Lipopolysaccharides/pharmacology , Male , Quinoxalines/metabolism , Quinoxalines/pharmacology , Rabbits , Radioligand Assay , Rats , Receptor, Bradykinin B1/genetics , Transfection , Tritium , Vasoconstriction/drug effects
13.
Drug Metab Dispos ; 32(6): 584-6, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155548

ABSTRACT

(-)-N-3-Benzyl-phenobarbital (NBPB) was reported to be a potent and selective inhibitor of CYP2C19. To validate the selectivity of NBPB toward CYP2C19 in human liver microsomes, the inhibitory effects on major cytochrome P450 isoform-specific reactions were evaluated in the present study. In human liver microsomes, NBPB showed potent competitive inhibition on CYP2C19-mediated S-mephenytoin 4'-hydroxylation with an IC(50) value of 0.25 microM and K(i) value of 0.12 microM, whereas weak inhibition was observed for CYP1A2-, CYP2A6-, CYP2B6-, CYP2C8-, CYP2C9-, CYP2D6-, and CYP3A4-mediated reactions with IC(50) values >100, >100, 62, 34, 19, >100, and 89 microM, respectively. Importantly, its selectivity toward CYP2C19 among the CYP2C subfamily was demonstrated. Therefore, NBPB can be used as a potent and selective inhibitor to establish the relative contribution of CYP2C19 for in vitro reaction phenotyping studies. This compound can also serve as a positive control inhibitor of CYP2C19 for routine screening of P450 reversible inhibition when human liver microsomes are used as the enzyme source.


Subject(s)
Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Microsomes, Liver/drug effects , Mixed Function Oxygenases/antagonists & inhibitors , Phenobarbital/analogs & derivatives , Phenobarbital/pharmacology , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2C19 , Enzyme Inhibitors/pharmacology , Female , Humans , In Vitro Techniques , Male , Mass Spectrometry , Microsomes, Liver/enzymology , Phenobarbital/chemical synthesis
14.
Toxicol Appl Pharmacol ; 194(1): 10-23, 2004 Jan 01.
Article in English | MEDLINE | ID: mdl-14728975

ABSTRACT

As a class, hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors can potentially cause skeletal myopathy. One statin, cerivastatin, has recently been withdrawn from the market due to an unacceptably high incidence of rhabdomyolysis. The mechanism underlying statin-induced myopathy is unknown. This paper sought to investigate the relationship among statin-induced myopathy, mitochondrial function, and muscle ubiquinone levels. Rats were administered cerivastatin at 0.1, 0.5, and 1.0 (mg/kg)/day or dose vehicle (controls) by oral gavage for 15 days. Samples of type I-predominant skeletal muscle (soleus) and type II-predominant skeletal muscle [quadriceps and extensor digitorum longus (EDL)], and blood were collected on study days 5, 10, and 15 for morphological evaluation, clinical chemistry, mitochondrial function tests, and analysis of ubiquinone levels. No histological changes were observed in any of the animals on study days 5 or 10, but on study day 15, mid- and high-dose animals had necrosis and inflammation in type II skeletal muscle. Elevated creatine kinase (CK) levels in blood (a clinical marker of myopathy) correlated with the histopathological diagnosis of myopathy. Ultrastructural characterization of skeletal muscle revealed disruption of the sarcomere and altered mitochondria only in myofibers with degeneration, while adjacent myofibers were unaffected and had normal mitochondria. Thus, mitochondrial effects appeared not to precede myofiber degeneration. Mean coenzyme Q9 (CoQ9) levels in all dose groups were slightly decreased relative to controls in type II skeletal muscle, although the difference was not significantly different in most cases. Mitochondrial function in skeletal muscle was not affected by the changes in ubiquinone levels. The ubiquinone levels in high-dose-treated animals exhibiting myopathy were not significantly different from low-dose animals with no observable toxic effects. Furthermore, ubiquinone levels did not correlate with circulating CK levels in treated animals. The results of this study suggest that neither mitochondrial injury, nor a decrease in muscle ubiquinone levels, is the primary cause of skeletal myopathy in cerivastatin-dosed rats.


Subject(s)
Mitochondria, Muscle/metabolism , Muscle, Skeletal/metabolism , Muscular Diseases/metabolism , Pyridines/toxicity , Ubiquinone/metabolism , Animals , Drug Evaluation, Preclinical/methods , Female , Mitochondria, Muscle/drug effects , Mitochondria, Muscle/pathology , Muscle, Skeletal/drug effects , Muscle, Skeletal/pathology , Muscular Diseases/chemically induced , Muscular Diseases/pathology , Rats , Rats, Sprague-Dawley , Ubiquinone/drug effects
15.
J Pharmacol Exp Ther ; 306(3): 903-13, 2003 Sep.
Article in English | MEDLINE | ID: mdl-12766251

ABSTRACT

An alpha4beta1/alpha4beta7 dual antagonist, 35S-compound 1, was used as a model ligand to study the effect of divalent cations on the activation state and ligand binding properties of alpha4 integrins. In the presence of 1 mM each Ca2+/Mg2+, 35S-compound 1 bound to several cell lines expressing both alpha4beta1 and alpha4beta7, but 2S-[(1-benzenesulfonyl-pyrrolidine-2S-carbonyl)-amino]-4-[4-methyl-2S-(methyl-[2-[4-(3-o-tolyl-ureido)-phenyl]-acetyl]-amino) pentanoylamino]-butyric acid (BIO7662), a specific alpha4beta1 antagonist, completely inhibited 35S-compound 1 binding, suggesting that alpha4beta1 was responsible for the observed binding. 35S-Compound 1 bound RPMI-8866 cells expressing predominantly alpha4beta7 with a KD of 1.9 nM in the presence of 1 mM Mn2+, and binding was inhibited only 29% by BIO7662, suggesting that the probe is a potent antagonist of activated alpha4beta7. With Ca2+/Mg2+, 35S-compound 1 bound Jurkat cells expressing primarily alpha4beta1 with a KD of 18 nM. In contrast, the binding of 35S-compound 1 to Mn2+-activated Jurkat cells occurred slowly, reaching equilibrium by 60 min, and failed to dissociate within another 60 min. The ability of four alpha4beta1/alpha4beta7 antagonists to block binding of activated alpha4beta1 or alpha4beta7 to vascular cell adhesion molecule-1 or mucosal addressin cell adhesion molecule-1, respectively, or to 35S-compound 1 was measured, and a similar rank order of potency was observed for native ligand and probe. Inhibition of 35S-compound 1 binding to alpha4beta1 in Ca2+/Mg2+ was used to identify nonselective antagonists among these four. These studies demonstrate that alpha4beta1 and alpha4beta7 have distinct binding properties for the same ligand, and binding parameters are dependent on the state of integrin activation in response to different divalent cations.


Subject(s)
Cations, Divalent/metabolism , Dipeptides/pharmacology , Integrin alpha4beta1/antagonists & inhibitors , Integrins/antagonists & inhibitors , Phenylalanine/pharmacology , Phenylurea Compounds/pharmacology , Binding Sites , Cell Line , Dipeptides/chemistry , Humans , Integrin alpha4beta1/metabolism , Integrins/metabolism , Jurkat Cells , K562 Cells , Kinetics , Ligands , Phenylalanine/analogs & derivatives , Phenylalanine/chemistry , Phenylurea Compounds/chemistry , Protein Binding , Radioligand Assay , Sulfur Radioisotopes , Tumor Cells, Cultured , Vascular Cell Adhesion Molecule-1/metabolism
16.
Drug Metab Dispos ; 31(4): 360-6, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12642460

ABSTRACT

This article is an invited report of a symposium sponsored by the Drug Metabolism Division of the American Society for Pharmacology and Experimental Therapeutics held at Experimental Biology 2002 in New Orleans. The impetus for the symposium was a perceived shortage in the supply of graduate students qualified for drug metabolism research positions in industry, academia, and government. For industry, recent hiring stems largely from the expansion of drug metabolism departments in an effort to keep pace with the demands of drug discovery and new technologies. In turn, regulatory scientists are needed to review and verify the results of the increased number and volume of studies required for drug development and approval. Thus the initial source of training, academia, has been forced to recognize these external hiring pressures while trying to attract and retain the faculty, postdoctoral scientists, and students necessary for active teaching and research programs. The trend of the expansion of the interdisciplinary nature of traditional drug metabolism to include emerging technologies such as pharmacogenetics, transporters, and proteomics and the implications for future needs in training and funding were acknowledged. There was also consensus on the value of partnerships between academia and industry for increasing student interest and providing training in disciplines directly applicable to industrial drug metabolism research. Factors affecting the sources of these trainees, such as federal funding, the number of trainees per institution, and recent issues with immigration restrictions that have limited the flow of scientists were also discussed.


Subject(s)
Education, Graduate/organization & administration , Technology, Pharmaceutical/education , Environment , Faculty , Government , Humans , Industry , Research Support as Topic/organization & administration , Technology, Pharmaceutical/organization & administration , Technology, Pharmaceutical/trends , Training Support/organization & administration
17.
Eur J Pharmacol ; 450(1): 19-28, 2002 Aug 16.
Article in English | MEDLINE | ID: mdl-12176104

ABSTRACT

A novel, potent nonpeptide oxytocin receptor antagonist (1-(1-(2-(2,2,2-trifluoroethoxy)-4-(1-methylsulfonyl-4-piperidinyloxy) phenylacetyl)-4-piperidinyl)-3,4-dihydro-2(1H)-quinolinone) has been identified that can be labeled to high specific activity with [35S]. In binding studies, this compound exhibits sub-nanomolar affinity and a high degree of selectivity (900-1800-fold) for human oxytocin receptors compared to human vasopressin receptors. This compound appears suitable for studying the pharmacology of oxytocin receptors in human and nonhuman primate tissues, for which there is currently a paucity of highly selective tools. It may also be useful as a nonlabeled competitor or as a radioligand in autoradiographic studies of oxytocin receptor localization in these tissues.


Subject(s)
Piperidines/pharmacology , Quinolones/pharmacology , Receptors, Oxytocin/antagonists & inhibitors , Receptors, Oxytocin/metabolism , Receptors, Vasopressin/metabolism , Animals , Antidiuretic Hormone Receptor Antagonists , Binding Sites , Binding, Competitive , Blood Platelets/metabolism , CHO Cells , Calcium/metabolism , Cricetinae , Humans , In Vitro Techniques , Kinetics , Ligands , Piperidines/chemical synthesis , Quinolones/chemical synthesis , Radioligand Assay
18.
Drug Metab Dispos ; 30(8): 937-43, 2002 Aug.
Article in English | MEDLINE | ID: mdl-12124313

ABSTRACT

[3R,5R,6S]-3-(2-cyclopropyloxy-5-trifluoromethoxyphenyl)-6-phenyl-1-oxa-7-azaspiro[4.5]decane is a substance P (Neurokinin 1 receptor) antagonist. Substance P antagonists are proven in concept to have excellent potential for the treatment of major depression, and they allow superior and sustained protection from acute and delayed chemotherapy-induced emesis. The metabolism of this compound was investigated in rat hepatocytes, and circulating rat plasma metabolites were identified following oral and intravenous dosing. The turnover in rat hepatocytes within 4 h was about 30%, and the major metabolites were identified as two nitrones and a lactam associated with the piperidine ring. Although these metabolites were also observed in rat plasma, the major circulating metabolite was a keto acid following oxidative de-amination of the piperidine ring. Liquid chromatography/tandem mass spectrometry and nuclear magnetic resonance were used to confirm the structure of the latter metabolite. A mechanism leading to the formation of the keto acid metabolite has been suggested, and most intermediates were observed in rat plasma.


Subject(s)
Aza Compounds/blood , Aza Compounds/metabolism , Hepatocytes/metabolism , Neurokinin-1 Receptor Antagonists , Spiro Compounds/blood , Spiro Compounds/metabolism , Substance P/antagonists & inhibitors , Administration, Oral , Animals , Aza Compounds/pharmacology , Chromatography, Liquid , In Vitro Techniques , Injections, Intravenous , Keto Acids/blood , Magnetic Resonance Spectroscopy , Male , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Spiro Compounds/pharmacology
19.
Biochemistry ; 41(20): 6548-60, 2002 May 21.
Article in English | MEDLINE | ID: mdl-12009920

ABSTRACT

35S-labeled derivatives of the insecticides nodulisporic acid and ivermectin were synthesized and demonstrated to bind with high affinity to a population of receptors in Drosophila head membranes that were previously shown to be associated with a glutamate-gated chloride channel. Nodulisporic acid binding was modeled as binding to a single population of receptors. Ivermectin binding was composed of at least two kinetically distinct receptor populations, only one of which was associated with nodulisporic acid binding. The binding of these two ligands was modulated by glutamate, ivermectin, and antagonists of invertebrate gamma-aminobutyric acid (GABA)ergic receptors. Because solubilized nodulisporic acid and ivermectin receptors comigrated as 230-kDa complexes by gel filtration, antisera specific for both the Drosophila glutamate-gated chloride channel subunit GluCl alpha (DmGluCl alpha) and the GABA-gated chloride channel subunit Rdl (DmRdl) proteins were generated and used to examine the possible coassembly of these two subunits within a single receptor complex. DmGluCl alpha antibodies immunoprecipitated all of the ivermectin and nodulisporic acid receptors solubilized by detergent from Drosophila head membranes. DmRdl antibodies also immunoprecipitated all solubilized nodulisporic receptors, but only approximately 70% of the ivermectin receptors. These data suggest that both DmGluCl alpha and DmRdl are components of nodulisporic acid and ivermectin receptors, and that there also exists a distinct class of ivermectin receptors that contains the DmGluCl alpha subunit but not the DmRdl subunit. This co-association of DmGluCl alpha and DmRdl represents the first biochemical and immunological evidence of coassembly of subunits from two different subclasses of ligand-gated ion channel subunits.


Subject(s)
Chloride Channels/metabolism , Drosophila Proteins/physiology , Glutamic Acid/physiology , Indoles/metabolism , Ivermectin/metabolism , Receptors, Drug/metabolism , Receptors, GABA-A/physiology , gamma-Aminobutyric Acid/physiology , Animals , Binding Sites , Cell Membrane/metabolism , Drosophila Proteins/chemistry , Drosophila melanogaster , Immune Sera/metabolism , Ion Channel Gating , Precipitin Tests , Radioligand Assay , Receptors, Drug/immunology , Solubility , Sulfur Radioisotopes/metabolism
20.
Biochemistry ; 41(22): 7125-41, 2002 Jun 04.
Article in English | MEDLINE | ID: mdl-12033947

ABSTRACT

Integrins alpha9beta1 and alpha4beta1 form a distinct structural class, but while alpha4beta1 has been subjected to extensive study, alpha9beta1 remains poorly characterized. We have used the small molecule N-(benzenesulfonyl)-(L)-prolyl-(L)-O-(1-pyrrolidinylcarbonyl)tyrosine (3) to investigate the biochemical properties of alpha9beta1 and directly compare these properties with those of alpha4beta1. Compound 3 has a high affinity for both integrins with K(D) values of < or =3 and 180 pM for alpha9beta1 in 1 mM Mn2+ (activating) and 1 mM Ca2+ and 1 mM Mg2+ (nonactivating) conditions and < or =5 and 730 pM for alpha4beta1 under the corresponding conditions. Ca2+ treatment promoted the binding of 3 to both integrins (EC50 = 30 microM Ca2+ in both cases). Compound 3 binding to both integrins was also stimulated by the addition of the activating monoclonal antibody TS2/16. These findings indicate that the mechanisms by which metal ions and TS2/16 regulate ligand binding to alpha9beta1 and alpha4beta1 are similar. The binding of 3 to both integrins induced the mAb 9EG7 LIBS epitope, a property consistent with occupancy of the receptor's ligand binding site by 3. But whereas EGTA treatment inhibited the binding of 9EG7 to alpha4beta1, it stimulated the binding of 9EG7 to alpha9beta1. The 9EG7 and TS2/16 effects point to contributions of the beta1-chains on binding. Cross-linking data revealed that the integrin alpha-chains are also involved in binding the small molecule, as stable linkages were observed on both the alpha9 chain of alpha9beta1 and the alpha4 chain of alpha4beta1. Extensive structure-activity analyses with natural and synthetic ligands indicate distinct features of the ligand binding pockets. Most notable was the estimated >1000-fold difference in the affinity of the integrins for VCAM-1, which binds alpha4beta1with an apparent K(D) of 10 nM and alpha9beta1 with an apparent K(D) of >10 microM. Differences were also seen in the binding of alpha9beta1 and alpha4beta1 to osteopontin. Compound 3 competed effectively for the binding of VCAM-1 and osteopontin to both integrins. While these studies show many similarities in the biochemical properties of alpha9beta1 and alpha4beta1, they identify important differences in their structure and function that can be exploited in the design of selective alpha9beta1 and alpha4beta1 inhibitors.


Subject(s)
Dipeptides/metabolism , Integrins/metabolism , Metals/metabolism , Receptors, Lymphocyte Homing/metabolism , Sialoglycoproteins/metabolism , Sulfones/metabolism , Vascular Cell Adhesion Molecule-1/metabolism , Binding, Competitive/physiology , Calcium/metabolism , Dipeptides/chemical synthesis , Humans , Integrin alpha4beta1 , Integrins/antagonists & inhibitors , Jurkat Cells , K562 Cells , Ligands , Magnesium/metabolism , Manganese/metabolism , Oligopeptides/metabolism , Osteopontin , Protein Binding/physiology , Receptors, Lymphocyte Homing/antagonists & inhibitors , Sialoglycoproteins/pharmacology , Sulfones/chemical synthesis , Vascular Cell Adhesion Molecule-1/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...