Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Drug Metab Dispos ; 36(8): 1659-69, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18505789

ABSTRACT

The disposition and metabolism of paraherquamide (PHQ), a potent and broad-spectrum anthelminthic, were examined in sheep, dogs, and gerbils. The metabolism of PHQ in these species was extensive and marked by significant species differences both in vitro and in vivo. In sheep and gerbils, PHQ metabolism occurs mainly at the pyrrolidine moiety, generating several metabolites that, for the most part, retained nematodicidal activity in vitro. In dogs, the dioxepene group was also extensively metabolized, ultimately resulting in formation of a catechol and loss of pharmacological activity. After oral administration of [3H]PHQ to intact sheep, gerbils, and dogs, the majority of the administered radioactivity was recovered in feces. Intact PHQ accounted for 0% (dogs) to approximately 30% (sheep and gerbils) of drug-related material in feces. A detailed investigation of the composition of the intestinal content of sheep indicated that a significant amount of the dose was still present in the rumen 24 h after dose and that PHQ underwent significant dehydration in the cecum. The oral pharmacokinetic parameters of PHQ in sheep and dogs suggest that its absorption is rapid in both species but that its apparent elimination rate is significantly higher in the dog (t(1/2) approximately 1.5 h) than it is in sheep (t(1/2) approximately 8.5 h). The short elimination half-life and the absence of PHQ or other active components in the dog gastrointestinal tract provide a potential explanation of the lack of efficacy of PHQ in this species.


Subject(s)
Indolizines/pharmacokinetics , Spiro Compounds/pharmacokinetics , Administration, Oral , Animals , Dogs , Feces/chemistry , Gerbillinae , Half-Life , Indolizines/administration & dosage , Sheep , Spiro Compounds/administration & dosage , Tritium
2.
Drug Metab Dispos ; 35(4): 521-4, 2007 Apr.
Article in English | MEDLINE | ID: mdl-17220240

ABSTRACT

Two novel metabolites of the dipeptidyl peptidase inhibitor sitagliptin (MK-0431, (2R)-4-oxo-4-[3-(trifluoromethyl)-5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl]-1-(2,4,5-trifluorophenyl)-butan-2-amine), were identified after purification from dog urine. The metabolites (referred to as M2 and M5) were characterized by hydrogen/deuterium exchange tandem mass spectrometry and NMR spectroscopy nuclear Overhauser effect experiments as the cis and trans stereoisomers formed by cyclization of the primary amino group with the alpha carbon of the piperazine ring, following oxidative desaturation.


Subject(s)
Enzyme Inhibitors/metabolism , Hypoglycemic Agents/metabolism , Pyrazines/metabolism , Triazoles/metabolism , Animals , Biotransformation , Cyclization , Dipeptidyl Peptidase 4/metabolism , Dipeptidyl-Peptidase IV Inhibitors , Dogs , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/urine , Hypoglycemic Agents/pharmacology , Hypoglycemic Agents/urine , Magnetic Resonance Spectroscopy , Molecular Structure , Oxidation-Reduction , Pyrazines/pharmacology , Pyrazines/urine , Sitagliptin Phosphate , Spectrometry, Mass, Electrospray Ionization , Stereoisomerism , Tandem Mass Spectrometry , Triazoles/pharmacology , Triazoles/urine
3.
Drug Metab Dispos ; 34(6): 1004-11, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16510541

ABSTRACT

[14C]7-(1,1-Dimethylethyl)-6-(2-ethyl-2H-1,2,4-triazol-3-ylmethoxy)-3-(2-fluorophenyl)-1,2,4-triazolo[4,3-b]pyridazine ([14C]-TPA023; 99 microCi/dose) was administered to five young, healthy, fasted male subjects as a single oral dose (3.0 mg) in solution (propylene glycol/water, 10:90 v/v). The parent compound was rapidly absorbed (plasma Tmax approximately 2 h), exhibited an apparent terminal half-life of 6.7 h, and accounted for approximately 53% of the total radioactivity in plasma. After 7 days of collection, the mean total recovery of radioactivity in the excreta was 82.6%, with 53.2% and 29.4% in urine and feces, respectively. Radiochromatographic analysis of the excreta revealed that TPA023 was metabolized extensively, and only trace amounts of unchanged parent were recovered. Radiochromatograms of urine and feces showed that TPA023 underwent metabolism via three pathways (t-butyl hydroxylation, N-deethylation, and direct N-glucuronidation). The products of t-butyl hydroxylation and N-deethylation, together with their corresponding secondary metabolites, accounted for the majority of the radioactivity in the excreta. In addition, approximately 10.3% of the dose was recovered in urine as the triazolo-pyridazine N1-glucuronide of TPA023. The t-butyl hydroxy and N-desethyl metabolites of TPA023, the TPA023 N1-glucuronide, and the triazolo-pyridazine N1-glucuronide of N-desethyl TPA023 were present in plasma. In healthy male subjects, therefore, TPA023 is well absorbed and is metabolized extensively (t-butyl hydroxylation and N-deethylation > glucuronidation), and the metabolites are excreted in urine and feces.


Subject(s)
Anti-Anxiety Agents/pharmacokinetics , GABA-A Receptor Agonists , Pyridazines/pharmacokinetics , Triazoles/pharmacokinetics , Administration, Oral , Anti-Anxiety Agents/administration & dosage , Biotransformation , Carbon Radioisotopes , Chromatography, Liquid , Feces/chemistry , Glucuronides/blood , Glucuronides/metabolism , Glucuronides/urine , Humans , Hydroxylation , Male , Mass Spectrometry , Pyridazines/administration & dosage , Receptors, GABA-A , Triazoles/administration & dosage
4.
Drug Metab Dispos ; 32(8): 848-61, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15258111

ABSTRACT

Compound A (3-[2-oxo-3-[3-(5,6,7,8-tetrahydro-[1,8]naphthyrindin-2-yl)propyl]-imidazolidin-1-yl]-3(S)-(6-methoxy-pyridin-3-yl)-propionic acid), a potent and selective antagonist of integrin alpha(v)beta(3) receptor, is under development for treatment of osteoporosis. This study describes metabolism and excretion of A in vivo in rats, dogs, and monkeys, and metabolism of A in vitro in primary hepatocytes from rats, dogs, monkeys, and humans. In all three animal species studied, A was primarily excreted as unchanged drug and, to a lesser degree, as phase I and phase II metabolites. Major biotransformation pathways of A included glucuronidation/glucosylation on the carboxylic group to form acyl-linked glucuronides/glucosides; and oxidation on the tetrahydronaphthyridine moiety to generate a carbinolamine and its further metabolized products. Minor pathways involved O-demethylation and hydroxylations on the alkyl chain. Only in rats, a glutathione adduct of A was also observed, and its formation is proposed to be via an iminium intermediate on the tetrahydronaphthyridine ring. Similar metabolic pathways were observed in the incubates of hepatocytes from the corresponding animals as well as from humans. CYP 3A and 2D subfamilies were capable of metabolizing A to its oxidative products. Overall, these in vitro and in vivo findings should provide useful insight on possible biotransformation pathways of A in humans.


Subject(s)
Integrin alphaVbeta3/antagonists & inhibitors , Integrin alphaVbeta3/metabolism , Animals , Dogs , Female , Integrin alphaVbeta3/analysis , Macaca mulatta , Male , Rats , Rats, Sprague-Dawley
5.
Chem Res Toxicol ; 16(12): 1598-605, 2003 Dec.
Article in English | MEDLINE | ID: mdl-14680374

ABSTRACT

The guanidine group present in the amino acid arginine was found to react with the lipid hydroperoxide-derived bifunctional electrophile, 4-oxo-2-nonenal. The reaction between N(alpha)-tert-butoxycarbony-l-arginine and 4-oxo-2-nonenal resulted in the formation of an adduct (adduct A) that subsequently dehydrated on heating to adduct B. Liquid chromatography/mass spectrometry and nuclear magnetic resonance spectroscopy were used to assign the structure of adduct B as (N(delta),N(omega)(')-etheno-2'-heptanon-2' '-one)-N(alpha)-t-Boc-arginine. The reaction proceeded from initial reaction of the primary N(omega)-amino group at the C-1 aldehyde of 4-oxo-2-nonenal. Subsequently, an intramolecular Michael addition of a secondary N(delta)-amino group occurring at C-3 resulted in formation of the cyclic carbinolamine adduct A. Dehydration and rearrangement of the exocyclic imine resulted in the formation of adduct B, which contained a stable imidazole ring. The tetra peptide LRDE reacted with 4-oxo-2-nonenal primarily at arginine rather than at the amino terminus. This suggests that arginine-containing proteins can react with lipid hydroperoxide-derived 4-oxo-2-nonenal to form a novel imidazole modification.


Subject(s)
Arginine/analogs & derivatives , Lipid Peroxides/chemistry , Aldehydes/chemistry , Arginine/chemistry , Arginine/metabolism , Borohydrides/chemistry , Chromatography, Liquid/methods , Guanidine/chemistry , Mass Spectrometry/methods , Nuclear Magnetic Resonance, Biomolecular , Oligopeptides/chemistry , Oxidation-Reduction
6.
Drug Metab Dispos ; 31(11): 1398-408, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14570773

ABSTRACT

The metabolism of rofecoxib, a potent and selective inhibitor of cyclooxygenase-2, was examined in vitro using human liver subcellular fractions. The biotransformation of rofecoxib was highly dependent on the subcellular fraction and the redox system used. In liver microsomal incubations, NADPH-dependent oxidation of rofecoxib to 5-hydroxyrofecoxib predominated, whereas NADPH-dependent reduction of rofecoxib to the 3,4-dihydrohydroxy acid metabolites predominated in cytosolic incubations. In incubations with S9 fractions, metabolites resulting from both oxidative and reductive pathways were observed. In contrast to microsomes, the oxidation of rofecoxib to 5-hydroxyrofecoxib by S9 fractions followed two pathways, one NADPH-dependent and one NAD+-dependent (non-cytochrome P450), with the latter accounting for about 40% of total activity. The 5-hydroxyrofecoxib thus formed was found to undergo NADPH-dependent reduction ("back reduction") to rofecoxib in incubations with liver cytosolic fractions. In incubations with dialyzed liver cytosol, net hydration of rofecoxib to form 3,4-dihydro-5-hydroxyrofecoxib was observed, whereas the 3,4-dihydrohydroxy acid derivatives were formed when NADPH was present. Although 3,4-dihydro-5-hydroxyrofecoxib could be reduced to the 3,4-dihydrohydroxy acid by cytosol in the presence of NADPH, the former species does not appear to serve as an intermediate in the overall reductive pathway of rofecoxib metabolism. In incubations of greater than 2 h with S9 fractions, net reductive metabolism predominated over oxidative metabolism. These in vitro results are consistent with previous findings on the metabolism of rofecoxib in vivo in human and provide a valuable insight into mechanistic aspects of the complex metabolism of this drug.


Subject(s)
Lactones/metabolism , Liver/cytology , Liver/metabolism , Cytosol/metabolism , Dose-Response Relationship, Drug , Humans , Lactones/chemistry , Subcellular Fractions/metabolism , Sulfones
7.
Chem Res Toxicol ; 16(7): 893-900, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12870892

ABSTRACT

Analysis of the reaction between 2'-deoxycytidine and 4-oxo-2-nonenal by LC/MS revealed the presence of three major products (adducts A(1), A(2), and B; [M + H](+) = 364). Adducts A(1) and A(2) were isomeric, and each dehydrated to form adduct B. The structure of adduct B was shown by LC/MS and NMR spectroscopy to be an etheno-2'-deoxycytidine adduct 1' '-[1-(2'-deoxy-beta-d-erythro-pentofuranosyl)-1H-imidazo[2,1-c]pyrimidin-2-oxo-4-yl]heptane-2' '-one. A time course experiment performed at 65 degrees C (pH 5-8) showed that the transformation of both A(1) and A(2) was pH-dependent. In acidic conditions, adducts A(1) and A(2) dehydrated primarily to adduct B. In contrast, in basic conditions, adducts A(1) and A(2) hydrolyzed primarily to dCyd. The data are consistent with adducts A(1) and A(2) being substituted ethano adducts that dehydrate to adduct B, a substituted 3,N(4)-etheno-2'-deoxycytidine adduct.


Subject(s)
Aldehydes/chemistry , Deoxycytidine/analogs & derivatives , Lipid Peroxidation , Aldehydes/analysis , Chromatography, Liquid , Deoxycytidine/analysis , Hydrogen-Ion Concentration , Isomerism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Molecular Structure , Temperature
8.
Biopolymers ; 68(4): 512-27, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12666176

ABSTRACT

Melanocortin receptor 4 (MC-4R) is involved in the regulation of energy balance and body weight, and recognizes alpha-, beta-, and gamma-melanocyte stimulating hormones (alpha-, beta-, gamma-MSH). In the search for compounds that regulate food intake and body weight, two synthetic lactam-derivative ligands of alpha-MSH were discovered, MTII and SHU9119. MTII is an agonist and reduces food intake in rats, whereas SHU9119 is an antagonist, and increases food intake and body weight in rats. MTII and SHU9119 are nonselective compounds to MC-4R. To enhance the potency and selectivity at the human MC-4R (hMC-4R), we recently synthesized analogs of SHU9119 (M. A. Bednarek, T. MacNeil, R. N. Kalyani, R. Tang, Van der L. H. T. Ploeg, and D. H. Weinberg, Journal of Medicinal Chemistry, 2001, Vol. 44, pp. 401-409), wherein compound 1 was the most selective for hMC-4R. Replacement of D-Nal by L-Nal in compound 1 made compound 2 weakly active. Comparison of the structures by NMR and molecular modeling of compounds 1 and 2 vs SHU9119 and MTII indicated that, even though they existed as an average of several conformations in solution, there were distinctions in their structures. The gamma-methylene protons of Arg in compound 1 were nonequivalent and shielded probably by the aromatic ring of Nal. The NHi-NHi+1 NOE cross peaks and the temperature coefficients of the amide protons around the "essential core" Nal/Phe7-Arg8-Trp9, required for high affinity and high selectivity at hMC-4R, were indicative of a possible turn structure for these compounds but with differences in their NOE strengths and temperature coefficient values. Molecular modeling of these compounds based on their NMR data showed that the essential core appeared as a "V" shape with two different orientations, one for compound 1 and some of the conformers of SHU9119 and MTII, and the other for compound 2 and some other conformers of SHU9119 and MTII. The remaining conformers of SHU9119 and MTII, which did not map to compound 1 or 2, suggested that they were outside of the hMC-4R binding envelop. These observations may lead to conjectures as to why compound 1 is highly active and selective toward hMC-4R.


Subject(s)
Oligopeptides/chemistry , Peptides, Cyclic/chemistry , Receptors, Corticotropin/antagonists & inhibitors , Humans , Magnetic Resonance Spectroscopy/methods , Models, Molecular , Oligopeptides/chemical synthesis , Oligopeptides/pharmacology , Peptides, Cyclic/chemical synthesis , Peptides, Cyclic/pharmacology , Protein Conformation , Receptor, Melanocortin, Type 4 , Thermodynamics
9.
Chem Res Toxicol ; 16(2): 198-207, 2003 Feb.
Article in English | MEDLINE | ID: mdl-12588191

ABSTRACT

Compound I, (2-[3-[(2,2-difluoro-2(2-pyridyl)ethyl)amino]-6-methyl-2-oxohydropyrazinyl]-N-[(3-fluoro(2-pyridyl))methyl]acetamide, is a potent competitive inhibitor of thrombin that reacts stoichiometrically with the protease. Compounds of this class possess therapeutic potential as anticoagulation agents. During the metabolic characterization of compound I, evidence was obtained for extensive metabolic activation of the pyrazinone ring system. Following administration of (14)C-labeled I to rats, significant levels of irreversibly bound radioactivity to proteins were detected in rat plasma and liver. LC/MS/MS analysis of metabolites formed in rat and human liver microsomes fortified with glutathione (GSH) revealed the presence of two structurally distinct GSH adducts. It is proposed that the first of these GSH conjugates derives from a two electron oxidation of the 6-methyl-2-oxo-3-aminopyrazinone moiety to afford an electrophilic imine-methide intermediate, while the second is formed by addition of GSH to an epoxide formed by P-450-mediated oxidation of the double bond at the 5-6 position of the pyrazinone ring. The addition of GSH to the proposed epoxide facilitates opening of the pyrazinone ring and a rearrangement to afford a stable, rearranged imidazole-containing metabolite. Elucidation of the metabolic activation pathways of I provides structural guidance for the design of thrombin inhibitors with decreased potential for the generation of chemically reactive intermediates.


Subject(s)
Protease Inhibitors/pharmacokinetics , Pyrazines/pharmacokinetics , Thrombin/antagonists & inhibitors , Administration, Oral , Animals , Binding, Competitive , Biotransformation , Carbon Radioisotopes , Epoxy Compounds/chemistry , Epoxy Compounds/metabolism , Glutathione/chemistry , Glutathione/metabolism , Humans , Imines/chemistry , Imines/metabolism , Microsomes, Liver/metabolism , NADP/metabolism , Nuclear Magnetic Resonance, Biomolecular , Oxidation-Reduction , Protease Inhibitors/chemistry , Protease Inhibitors/pharmacology , Protein Binding , Pyrazines/chemistry , Pyrazines/pharmacology , Rats , Spectrometry, Mass, Electrospray Ionization/methods
10.
Angew Chem Int Ed Engl ; 37(17): 2368-2370, 1998 Sep 18.
Article in English | MEDLINE | ID: mdl-29710963

ABSTRACT

The special pair of the bacterial photosystem has been modeled with a porphyrin dimer (the partial structure is shown). As with the natural system, only one pyrrole ring from each monomer subunit participates in π overlap.

SELECTION OF CITATIONS
SEARCH DETAIL
...