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.
Toxicol Appl Pharmacol ; 212(1): 35-44, 2006 Apr 01.
Article in English | MEDLINE | ID: mdl-16051291

ABSTRACT

Idiosyncratic liver injury occurs in a small fraction of people on certain drug regimens. The cause of idiosyncratic hepatotoxicity is not known; however, it has been proposed that environmental factors such as concurrent inflammation initiated by bacterial lipopolysaccharide (LPS) increase an individual's susceptibility to drug toxicity. Ranitidine (RAN), a histamine-2 receptor antagonist, causes idiosyncratic liver injury in humans. In a previous report, idiosyncrasy-like liver toxicity was created in rats by cotreating them with LPS and RAN. In the present study, the ability of metabonomic techniques to distinguish animals cotreated with LPS and RAN from those treated with each agent individually was investigated. Rats were treated with LPS or its vehicle and with RAN or its vehicle, and urine was collected for nuclear magnetic resonance (NMR)- and mass spectroscopy-based metabonomic analyses. Blood and liver samples were also collected to compare metabonomic results with clinical chemistry and histopathology. NMR metabonomic analysis indicated changes in the pattern of metabolites consistent with liver damage that occurred only in the LPS/RAN cotreated group. Principal component analysis of urine spectra by either NMR or mass spectroscopy produced a clear separation of the rats treated with LPS/RAN from the other three groups. Clinical chemistry (serum alanine aminotransferase and aspartate aminotransferase activities) and histopathology corroborated these results. These findings support the potential use of a noninvasive metabonomic approach to identify drug candidates with potential to cause idiosyncratic liver toxicity with inflammagen coexposure.


Subject(s)
Chemical and Drug Induced Liver Injury/metabolism , Histamine H2 Antagonists/toxicity , Lipopolysaccharides/toxicity , Ranitidine/toxicity , Alanine Transaminase/blood , Animals , Aspartate Aminotransferases/blood , Chemical and Drug Induced Liver Injury/pathology , Discriminant Analysis , Liver/chemistry , Liver/pathology , Liver Function Tests , Magnetic Resonance Spectroscopy , Male , Principal Component Analysis , Rats , Rats, Sprague-Dawley , Spectrometry, Mass, Electrospray Ionization
2.
Drug Metab Dispos ; 31(11): 1448-55, 2003 Nov.
Article in English | MEDLINE | ID: mdl-14570778

ABSTRACT

A pharmacokinetics and metabolism study was conducted in eight healthy human volunteers. After oral administration of [14C]eplerenone (EP) at a dose of 100 mg per person as an aqueous solution, blood, saliva, breath, urine, and fecal samples were collected at various time points. All matrices were analyzed for total radioactivity and/or for EP and its open-lactone-ring form (EPA). EP was well absorbed, and a mean EP Cmax of 1.72 mug/ml was achieved 1.2 h postdose. After the Cmax, plasma concentrations of EP declined with a half-life of 3.0 h. Plasma concentrations of EPA were much lower than EP concentrations, and the area under the plasma-concentration time curve (AUC) for EPA was only 4% of the EP AUC. Plasma protein binding was moderate (33-60%) but concentration-dependent over the therapeutic concentration range. EP and its metabolites did not preferentially partition into the red blood cells and blood concentrations of total radioactivity were lower than plasma concentrations. Approximately 66.6% and 32.0% of the radioactive dose were excreted in urine and feces, respectively. The majority of urinary and fecal radioactivity was due to metabolites, indicating extensive metabolism of EP. The major metabolic pathways were 6beta- and/or 21-hydroxylation and 3-keto reduction. There was no evidence for any alteration of the 9,11-epoxide ring or the methyl ester. As a percentage of dose, the primary metabolic products excreted in urine and feces included 6beta-hydroxy-EP (6beta-OHEP) (32.0%), 6beta,21-OHEP (20.5%), 21-OHEP (7.89%), and 2alpha,3beta,21-OHEP (5.96%). The amounts of the other metabolites excreted were less than 5% each.


Subject(s)
Spironolactone/analogs & derivatives , Spironolactone/administration & dosage , Spironolactone/pharmacokinetics , Administration, Oral , Adult , Carbon Radioisotopes/pharmacokinetics , Eplerenone , Humans , Male , Middle Aged , Spironolactone/chemistry
3.
Toxicol Appl Pharmacol ; 187(3): 137-46, 2003 Mar 15.
Article in English | MEDLINE | ID: mdl-12662897

ABSTRACT

The role that metabonomics has in the evaluation of xenobiotic toxicity studies is presented here together with a brief summary of published studies. To provide a comprehensive assessment of this approach, the Consortium for Metabonomic Toxicology (COMET) has been formed between six pharmaceutical companies and Imperial College of Science, Technology and Medicine (IC), London, UK. The objective of this group is to define methodologies and to apply metabonomic data generated using (1)H NMR spectroscopy of urine and blood serum for preclinical toxicological screening of candidate drugs. This is being achieved by generating databases of results for a wide range of model toxins which serve as the raw material for computer-based expert systems for toxicity prediction. The project progress on the generation of comprehensive metabonomic databases and multivariate statistical models for prediction of toxicity, initially for liver and kidney toxicity in the rat and mouse, is reported. Additionally, both the analytical and biological variation which might arise through the use of metabonomics has been evaluated. An evaluation of intersite NMR analytical reproducibility has revealed a high degree of robustness. Second, a detailed comparison has been made of the ability of the six companies to provide consistent urine and serum samples using a study of the toxicity of hydrazine at two doses in the male rat, this study showing a high degree of consistency between samples from the various companies in terms of spectral patterns and biochemical composition. Differences between samples from the various companies were small compared to the biochemical effects of the toxin. A metabonomic model has been constructed for urine from control rats, enabling identification of outlier samples and the metabolic reasons for the deviation. Building on this success, and with the completion of studies on approximately 80 model toxins, first expert systems for prediction of liver and kidney toxicity have been generated.


Subject(s)
Metabolism/genetics , Toxicology/methods , Xenobiotics/toxicity , Animals , Databases, Factual , Drug Evaluation, Preclinical , Humans , Magnetic Resonance Spectroscopy , Mice , Rats , Toxicology/standards , Xenobiotics/blood , Xenobiotics/urine
4.
Drug Metab Dispos ; 31(4): 491-501, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12642477

ABSTRACT

The pharmacokinetics and metabolism of valdecoxib, a potent cyclooxygenase-2 selective inhibitor, were investigated in mice. Valdecoxib was extensively metabolized after a single 5 mg/kg oral administration of [(14)C]valdecoxib and elimination of unchanged drug was minor (less than 1%) in male and female mice. The total mean percentage of administered radioactive dose recovered was 99.8% in the male mice and 94.7% in the female mice. Sixteen metabolites were identified in mouse plasma, red blood cells, urine, and feces. The main phase I metabolic pathway of valdecoxib in mice involved the oxidation of the 5-methyl group to form the active hydroxymethyl metabolite M1. M1 was further oxidized to the carboxylic acid metabolite M4, which underwent opening of the isoxazole ring to form M6 and M13. Phase II metabolism included glucuronide, glucoside, and methyl sulfone conjugations. M1 was also conjugated with glucuronic acid and glucose to yield M-G and M1-glucose, respectively. Three novel methylsulfone conjugates M20, M21, and M21-G were detected in blood or urine. Valdecoxib and M1 were the major radioactive components in plasma and red blood cells. The plasma area under the curve from zero to infinity (AUC(0-infinity)) values for valdecoxib and M1 were 3.58 and 0.850 microg. h/ml in males and 2.08 and 1.63 microg. h/ml in females, respectively. The RBC AUC(0-infinity) values for valdecoxib and M1 were 12.1 and 22.6 microg. h/g in males and 6.42 and 35.2 microg. h/g in females, respectively.


Subject(s)
Isoenzymes/antagonists & inhibitors , Isoxazoles/pharmacokinetics , Sulfonamides/pharmacokinetics , Animals , Biotransformation , Carbon Isotopes/blood , Carbon Isotopes/pharmacokinetics , Carbon Isotopes/urine , Carbon Radioisotopes , Chromatography, Liquid/instrumentation , Chromatography, Liquid/methods , Cyclooxygenase 2 , Dose-Response Relationship, Drug , Erythrocytes/drug effects , Erythrocytes/metabolism , Female , Gas Chromatography-Mass Spectrometry/instrumentation , Gas Chromatography-Mass Spectrometry/methods , Glucuronidase/metabolism , Hydrolysis , Isoxazoles/blood , Isoxazoles/metabolism , Isoxazoles/urine , Magnetic Resonance Imaging/instrumentation , Magnetic Resonance Imaging/methods , Male , Metabolic Clearance Rate , Mice , Mice, Inbred Strains , Plasma/drug effects , Plasma/metabolism , Prostaglandin-Endoperoxide Synthases , Sulfonamides/blood , Sulfonamides/metabolism , Sulfonamides/urine , Time Factors
5.
Drug Metab Dispos ; 30(9): 1013-21, 2002 Sep.
Article in English | MEDLINE | ID: mdl-12167567

ABSTRACT

Valdecoxib is a potent and specific inhibitor of cyclooxygenase-2, which is used for the treatment of rheumatoid arthritis, osteoarthritis, and the dysmenorrhea pain. Eight male human subjects each received a single 50-mg oral dose of [(14)C]valdecoxib. Urine, feces, and blood samples were collected after administration of the radioactive dose. Most of the radioactivity in plasma was associated with valdecoxib and the hydroxylated metabolite of valdecoxib (M1). The estimated terminal half-life for valdecoxib was about 7 h. About 76.1% of the radioactive dose was recovered in urine and 18% of the radioactive dose was recovered in feces. Valdecoxib was extensively metabolized in human, and nine phase I metabolites were identified. The primary oxidative metabolic pathways of valdecoxib involved hydroxylation at either the methyl group to form M1 or N-hydroxylation at the sulfonamide moiety to form M2. Further oxidation of M1 led to the formation of several other phase I metabolites. Oxidative breakdown of the N-hydroxy sulfonamide function group in M2 led to the formation of corresponding sulfinic acid and sulfonic acid metabolites. The O-glucuronide conjugate of M1 and N-glucuronide conjugate of valdecoxib were the major urinary metabolites, which accounted for 23.3 and 19.5% of the total administered dose, respectively. The remaining urinary metabolites were glucuronide conjugates of other phase I metabolites. Only 3% of the administered dose was recovered in urine as unchanged parent, suggesting that renal clearance is insignificant for valdecoxib. Absorption of valdecoxib was excellent since the recovery of unchanged valdecoxib in feces was <1% of the administered dose.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antipruritics/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacokinetics , Isoenzymes/metabolism , Isoxazoles/pharmacokinetics , Prostaglandin-Endoperoxide Synthases/metabolism , Sulfonamides/pharmacokinetics , Acute Disease , Administration, Oral , Anti-Inflammatory Agents, Non-Steroidal/urine , Antipruritics/blood , Antipruritics/urine , Chromatography, High Pressure Liquid , Cyclooxygenase 2 , Cyclooxygenase 2 Inhibitors , Cyclooxygenase Inhibitors/blood , Cyclooxygenase Inhibitors/urine , Feces/chemistry , Glucuronidase/metabolism , Humans , Isoxazoles/blood , Isoxazoles/urine , Male , Mass Spectrometry , Membrane Proteins , Pain/metabolism , Sulfonamides/blood , Sulfonamides/urine
SELECTION OF CITATIONS
SEARCH DETAIL
...