Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 5 de 5
Filter
Add more filters










Database
Language
Publication year range
1.
Methods Mol Biol ; 691: 283-301, 2011.
Article in English | MEDLINE | ID: mdl-20972760

ABSTRACT

Xenobiotics, including therapeutic agents, can produce a variety of beneficial, as well as adverse, effects in mammals. One potential source of drug-mediated toxicity stems from metabolic activation of the parent compound, typically catalyzed by one or more members of the cytochrome P450 family of enzymes. The resulting electrophile, if not quenched by low molecular weight endogenous nucleophiles, can form covalent adducts to cellular proteins, potentially resulting in enzyme inactivation, cell death, or formation of an immunogenic species. The toxicological consequences of exposure to such reactive intermediates range from mild inflammation to organ failure, anaphylaxis, and death. At Merck Research Laboratories, the potential of drug candidates to bind covalently to proteins is evaluated at the lead optimization stage of drug discovery by incubating a radiolabeled analog of the compound in question with liver microsomal preparations (under oxidative conditions) or whole cells (full cellular metabolic capability), typically derived from rat and human liver. A semi-automated method based on the Brandel Harvester technique then is used to measure the formation of covalent adducts of the test compound to liver proteins. This assay is viewed as an important component of drug discovery programs, since the findings are employed to guide specific efforts to abrogate bioactivation issues through informed structural modification of lead compounds.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/metabolism , Liver/metabolism , Proteins/metabolism , Animals , Chromatography, High Pressure Liquid , Hepatocytes/cytology , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Liver/drug effects , Microsomes/drug effects , Microsomes/metabolism , Protein Binding/drug effects , Proteins/isolation & purification , Radiometry , Rats , Scintillation Counting , Solid Phase Extraction , Solutions , Toxicity Tests , Tritium/metabolism , Xenobiotics/metabolism , Xenobiotics/toxicity
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.
Drug Metab Dispos ; 34(9): 1457-61, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16772365

ABSTRACT

MK-0767 (KRP-297; 2-methoxy-5-(2,4-dioxo-5-thiazolidinyl)-N-[[4-(trifluoromethyl)phenyl] methyl]benzamide) is a thiazolidinedione (TZD)-containing dual agonist of the peroxisome proliferator-activated receptors alpha and gamma that has been studied as a potential treatment for patients with type 2 diabetes. The metabolism and excretion of [14C]MK-0767 were evaluated in six human volunteers after a 5-mg (200 microCi) oral dose. Excretion of 14C radioactivity was found to be nearly equal into the urine (approximately 50%) and feces (approximately 40%). Elimination of [14C]MK-0767 was primarily by metabolism, with minimal excretion of parent compound into the urine (<0.5% of dose) and feces (approximately 14% of the dose). [14C]MK-0767 was the major circulating compound-related entity (>96% of radioactivity) through 48 h postdose. It was also found that approximately 91% of the total radioactivity area under the curve was due to intact MK-0767. Several minor metabolites were detected in plasma (<1% of radioactivity, each), formed by cleavage of the TZD ring and subsequent S-methylation and oxidation. All the metabolites excreted into urine were formed by TZD cleavage, whereas the major metabolite in feces was the O-demethylated derivative of MK-0767.


Subject(s)
Hypoglycemic Agents/pharmacology , Thiazoles/pharmacokinetics , Absorption , Administration, Oral , Adolescent , Adult , Biotransformation , Carbon Radioisotopes , Feces/chemistry , Humans , Hypoglycemic Agents/administration & dosage , Hypoglycemic Agents/urine , Male , Middle Aged , Thiazoles/administration & dosage , Thiazoles/urine
4.
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
5.
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
SELECTION OF CITATIONS
SEARCH DETAIL
...