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.
Biochem Pharmacol ; 77(10): 1642-54, 2009 May 15.
Article in English | MEDLINE | ID: mdl-19426702

ABSTRACT

Trabectedin (YONDELIS) is a potent anticancer agent which was recently approved in Europe for the treatment of soft tissue sarcoma. The drug is currently also in clinical development for the treatment of ovarian carcinoma. In vitro experiments were conducted to investigate the hepatic metabolism of [(14)C]trabectedin in Cynomolgus monkey and human liver subcellular fractions. The biotransformation of trabectedin was qualitatively similar in 12,000 x g supernatants of both species, and all human metabolites were also produced by the monkey. The trabectedin metabolites were identified by QTOF mass spectrometry, and HPLC co-chromatography with reference compounds. Trabectedin was metabolized via different biotransformation pathways. Most of the metabolic conversions occurred at the trabectedin A domain including mono-oxidation and di-oxidation, carboxylic acid formation with and without additional oxidation, and demethylation either without (N-demethylation to ET-729) or with additional mono-, di- or tri-oxidation. Another metabolite resulted from O-demethylation at the trabectedin C subunit, and in addition, aliphatic ring opening of the methylene dioxybridge at the B domain was detected. Overall, demethylation and oxidation played a major role in phase I metabolism of the drug. Human cDNA expressed CYPs 1A2, 2A6, 2B6, 2C8, 2C9, 2C18, 2D6, 2E1, 3A4 and 3A5 in E. coli membranes, but not CYP1B1, 2C19, and 4A11 were able to metabolize [(14)C]trabectedin. Experiments with chemical inhibitors and CYP inhibitory antibodies indicated that, at therapeutic levels, CYP3A4 is the main human CYP isoform involved in trabectedin's hepatic metabolism. In monkey and human liver microsomes, trabectedin was not substantially metabolized by glucuronidation.


Subject(s)
Antineoplastic Agents, Alkylating/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dioxoles/metabolism , Microsomes, Liver/metabolism , Tetrahydroisoquinolines/metabolism , Animals , Antineoplastic Agents, Alkylating/pharmacokinetics , Chromatography, High Pressure Liquid , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/biosynthesis , Dioxoles/pharmacokinetics , Enzyme Inhibitors/pharmacology , Escherichia coli/metabolism , Female , Humans , In Vitro Techniques , Macaca fascicularis , Male , Microsomes, Liver/enzymology , Phenotype , Species Specificity , Subcellular Fractions/enzymology , Subcellular Fractions/metabolism , Tetrahydroisoquinolines/pharmacokinetics , Trabectedin
2.
Drug Metab Dispos ; 37(4): 809-20, 2009 Apr.
Article in English | MEDLINE | ID: mdl-19131522

ABSTRACT

Absorption, metabolism, and excretion of darunavir, an inhibitor of human immunodeficiency virus protease, was studied in eight healthy male subjects after a single oral dose of 400 mg of [(14)C]darunavir given alone (unboosted subjects) or with ritonavir [100 mg b.i.d. 2 days before and 7 days after darunavir administration (boosted subjects)]. Plasma exposure to darunavir was 11-fold higher in boosted subjects. Total recoveries of radioactivity in urine and feces were 93.9 and 93.5% of administered radioactivity in unboosted and boosted subjects, respectively. The most radioactivity was recovered in feces (81.7% in unboosted subjects and 79.5% in boosted subjects, compared with 12.2 and 13.9% recovered in urine, respectively). Darunavir was extensively metabolized in unboosted subjects, mainly by carbamate hydrolysis, isobutyl aliphatic hydroxylation, and aniline aromatic hydroxylation and to a lesser extent by benzylic aromatic hydroxylation and glucuronidation. Total excretion of unchanged darunavir accounted for 8.0% of the dose in unboosted subjects. Boosting with ritonavir resulted in significant inhibition of carbamate hydrolysis, isobutyl aliphatic hydroxylation, and aniline aromatic hydroxylation but had no effect on aromatic hydroxylation at the benzylic moiety, whereas excretion of glucuronide metabolites was markedly increased but still represented a minor pathway. Total excretion of unchanged darunavir accounted for 48.8% of the administered dose in boosted subjects as a result of the inhibition of darunavir metabolism by ritonavir. Unchanged darunavir in urine accounted for 1.2% of the administered dose in unboosted subjects and 7.7% in boosted subjects, indicating a low renal clearance. Darunavir administered alone or with ritonavir was well tolerated.


Subject(s)
HIV Protease Inhibitors/pharmacokinetics , Ritonavir/pharmacokinetics , Sulfonamides/pharmacokinetics , Adult , Chromatography, High Pressure Liquid , Darunavir , Dose-Response Relationship, Drug , Feces , HIV Protease Inhibitors/administration & dosage , HIV Protease Inhibitors/blood , HIV Protease Inhibitors/urine , Humans , Male , Middle Aged , Reference Values , Ritonavir/administration & dosage , Sulfonamides/administration & dosage , Sulfonamides/blood , Sulfonamides/urine , Tandem Mass Spectrometry
3.
Antimicrob Agents Chemother ; 52(10): 3478-83, 2008 Oct.
Article in English | MEDLINE | ID: mdl-18644951

ABSTRACT

In this open-label, single-center study, eight healthy men each received a single 500-mg dose of [(14)C]doripenem, containing 50 microCi of [(14)C]doripenem, administered as a 1-h intravenous infusion. The concentrations of unchanged doripenem and its primary metabolite (doripenem-M-1) resulting from beta-lactam ring opening were measured in plasma and urine by a validated liquid chromatography method coupled to a tandem mass spectrometry assay. Total radioactivity was measured in blood, plasma, urine, and feces by liquid scintillation counting. Further metabolite profiling was conducted on urine samples using liquid chromatography coupled to radiochemical detection and high-resolution mass spectrometry. Unchanged doripenem and doripenem-M-1 accounted for means of 80.7% and 12.7% of the area under the plasma total-radioactivity-versus-time curve (area under the concentration-time curve extrapolated to infinity) and exhibited elimination half-lives of 1.1 and 2.5 h, respectively. Total clearance of doripenem was 16 liters/h, and renal clearance was 12.5 liters/h. At 7 days after the single dose, 95.3% of total doripenem-related radioactivity was recovered in urine and 0.72% in feces. A total mean of 97.2% of the administered dose was excreted in the urine as unchanged doripenem (78.7% +/- 5.7%) and doripenem-M-1 (18.5% +/- 2.6%). Most of the urinary recovery occurred within 4 h of dosing. Three additional minor metabolites were identified in urine: the glycine and taurine conjugates of doripenem-M-1 and oxidized doripenem-M-1. These results show that doripenem is predominantly eliminated in urine as unchanged drug, with only a fraction metabolized to doripenem-M-1 and other minor metabolites.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Carbapenems/administration & dosage , Carbapenems/pharmacokinetics , Adolescent , Adult , Anti-Bacterial Agents/adverse effects , Anti-Bacterial Agents/metabolism , Carbapenems/adverse effects , Carbapenems/metabolism , Carbon Radioisotopes , Chromatography, Liquid , Cohort Studies , Doripenem , Humans , Infusions, Intravenous , Male , Middle Aged , Safety , Tandem Mass Spectrometry
4.
Drug Metab Dispos ; 36(4): 769-79, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18227146

ABSTRACT

Absorption, metabolism, and excretion of paliperidone, an atypical antipsychotic, was studied in five healthy male subjects after a single dose of 1 mg of [(14)C]paliperidone oral solution ( approximately 16 microCi/subject). One week after dosing, 88.4 to 93.8% (mean 91.1%) of the administered radioactivity was excreted: 77.1 to 87.1% (mean 79.6%) in urine and 6.8 to 14.4% (mean 11.4%) in the feces. Paliperidone was the major circulating compound (97% of the area under the plasma concentration-time curve at 24 h). No metabolites could be detected in plasma. Renal excretion was the major route of elimination with 59% of the dose excreted unchanged in urine. About half of the renal excretion occurred by active secretion. Unchanged drug was not detected in feces. Four metabolic pathways were identified as being involved in the elimination of paliperidone, each of which accounted for up to a maximum of 6.5% of the biotransformation of the total dose. Biotransformation of the drug occurred through oxidative N-dealkylation (formation of the acid metabolite M1), monohydroxylation of the alicyclic ring (M9), alcohol dehydrogenation (formation of the ketone metabolite M12), and benzisoxazole scission (formation of M11), the latter in combination with glucuronidation (M16) or alicyclic hydroxylation (M10). Unchanged drug, M1, M9, M12, and M16 were detected in urine; M10 and M11 were detected in feces. The monohydroxylated metabolite M9 was solely present in urine samples of extensive CYP2D6 metabolizers, whereas M10, another metabolite monohydroxylated at the alicyclic ring system, was present in feces of poor metabolizers as well. In conclusion, paliperidone is not metabolized extensively and is primarily renally excreted.


Subject(s)
Biogenic Monoamines/antagonists & inhibitors , Intestinal Absorption/physiology , Isoxazoles/metabolism , Pyrimidines/metabolism , Adult , Biogenic Monoamines/metabolism , Feces/chemistry , Humans , Intestinal Absorption/drug effects , Isoxazoles/chemistry , Isoxazoles/pharmacology , Kidney/drug effects , Kidney/metabolism , Liver/drug effects , Liver/metabolism , Male , Metabolic Networks and Pathways/drug effects , Metabolic Networks and Pathways/physiology , Middle Aged , Paliperidone Palmitate , Pyrimidines/chemistry , Pyrimidines/pharmacology , Time Factors
5.
Expert Opin Drug Metab Toxicol ; 1(1): 75-90, 2005 Jun.
Article in English | MEDLINE | ID: mdl-16922654

ABSTRACT

Cell-based in vitro models are invaluable tools in elucidating the pharmacokinetic profile of a drug candidate during its drug discovery and development process. As biotransformation is one of the key determinants of a drug's disposition in the body, many in vitro models to study drug metabolism have been established, and others are still being developed and validated. This review is aimed at providing the reader with a concise overview of the characteristics and optimal application of established and emerging in vitro cell-based models to study human drug metabolism and induction of drug metabolising enzymes in the liver. The strengths and weaknesses of liver-derived models, such as primary hepatocytes, either freshly isolated or cryopreserved, and from adult or fetal donors, precision-cut liver slices, and cell lines, including immortalised cells, reporter cell lines, hepatocarcinoma-derived cell lines and recombinant cell lines, are discussed. Relevant cell culture configuration aspects as well as other models such as stem cell-derived hepatocyte-like cells and humanised animal models are also reviewed. The status of model development, their acceptance by health authorities and recommendations for the most appropriate use of the models are presented.


Subject(s)
Cells, Cultured , Liver/cytology , Liver/enzymology , Organ Culture Techniques , Pharmaceutical Preparations/metabolism , Animals , Enzyme Induction/drug effects , Enzyme Induction/physiology , Humans , Liver/metabolism , Models, Animal , Pharmaceutical Preparations/administration & dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...