Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
1.
Drug Metab Dispos ; 42(5): 918-31, 2014 May.
Article in English | MEDLINE | ID: mdl-24608633

ABSTRACT

The disposition of a single oral dose of 5 mg (100 µCi) of [(14)C]axitinib was investigated in fasted healthy human subjects (N = 8). Axitinib was rapidly absorbed, with a median plasma Tmax of 2.2 hours and a geometric mean Cmax and half-life of 29.2 ng/ml and 10.6 hours, respectively. The plasma total radioactivity-time profile was similar to that of axitinib but the AUC was greater, suggesting the presence of metabolites. The major metabolites in human plasma (0-12 hours), identified as axitinib N-glucuronide (M7) and axitinib sulfoxide (M12), were pharmacologically inactive, and with axitinib comprised 50.4%, 16.2%, and 22.5% of the radioactivity, respectively. In excreta, the majority of radioactivity was recovered in most subjects by 48 hours postdose. The median radioactivity excreted in urine, feces, and total recovery was 22.7%, 37.0%, and 59.7%, respectively. The recovery from feces was variable across subjects (range, 2.5%-60.2%). The metabolites identified in urine were M5 (carboxylic acid), M12 (sulfoxide), M7 (N-glucuronide), M9 (sulfoxide/N-oxide), and M8a (methylhydroxy glucuronide), accounting for 5.7%, 3.5%, 2.6%, 1.7%, and 1.3% of the dose, respectively. The drug-related products identified in feces were unchanged axitinib, M14/15 (mono-oxidation/sulfone), M12a (epoxide), and an unidentified metabolite, comprising 12%, 5.7%, 5.1%, and 5.0% of the dose, respectively. The proposed mechanism to form M5 involved a carbon-carbon bond cleavage via M12a, followed by rearrangement to a ketone intermediate and subsequent Baeyer-Villiger rearrangement, possibly through a peroxide intermediate. In summary, the study characterized axitinib metabolites in circulation and primary elimination pathways of the drug, which were mainly oxidative in nature.


Subject(s)
Imidazoles/pharmacokinetics , Indazoles/pharmacokinetics , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Receptors, Vascular Endothelial Growth Factor/antagonists & inhibitors , Adult , Axitinib , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Feces/chemistry , Humans , Imidazoles/blood , Imidazoles/metabolism , Imidazoles/urine , Indazoles/blood , Indazoles/metabolism , Indazoles/urine , Magnetic Resonance Spectroscopy , Male , Mass Spectrometry , Middle Aged , Molecular Structure , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/metabolism , Protein Kinase Inhibitors/urine
2.
Drug Metab Dispos ; 40(3): 539-55, 2012 Mar.
Article in English | MEDLINE | ID: mdl-22180047

ABSTRACT

Sunitinib is an oral multitargeted tyrosine kinase inhibitor approved for the treatment of advanced renal cell carcinoma, imatinib-refractory gastrointestinal stromal tumor, and advanced pancreatic neuroendocrine tumors. The current studies were conducted to characterize the pharmacokinetics, distribution, and metabolism of sunitinib after intravenous and/or oral administrations of [(14)C]sunitinib in rats (5 mg/kg i.v., 15 mg/kg p.o.), monkeys (6 mg/kg p.o.), and humans (50 mg p.o.). After oral administration, plasma concentration of sunitinib and total radioactivity peaked from 3 to 8 h. Plasma terminal elimination half-lives of sunitinib were 8 h in rats, 17 h in monkeys, and 51 h in humans. The majority of radioactivity was excreted to the feces with a smaller fraction of radioactivity excreted to urine in all three species. The bioavailability in female rats was close to 100%, suggesting complete absorption of sunitinib. Whole-body autoradioluminography suggested radioactivity was distributed throughout rat tissues, with the majority of radioactivity cleared within 72 h. Radioactivity was eliminated more slowly from pigmented tissues. Sunitinib was extensively metabolized in all species. Many metabolites were detected both in urine and fecal extracts. The main metabolic pathways were N-de-ethylation and hydroxylation of indolylidene/dimethylpyrrole. N-Oxidation/hydroxylation/desaturation/deamination of N,N'-diethylamine and oxidative defluorination were the minor metabolic pathways. Des-ethyl metabolite M1 was the major circulating metabolite in all three species.


Subject(s)
Indoles/metabolism , Indoles/pharmacokinetics , Pyrroles/metabolism , Pyrroles/pharmacokinetics , Absorption/physiology , Administration, Oral , Adult , Animals , Biological Availability , Carbon Isotopes/administration & dosage , Carbon Isotopes/metabolism , Carbon Isotopes/urine , Carbon Radioisotopes , Feces/chemistry , Female , Half-Life , Humans , Indoles/administration & dosage , Indoles/urine , Injections, Intravenous/methods , Macaca fascicularis , Male , Middle Aged , Pyrroles/administration & dosage , Pyrroles/urine , Rats , Rats, Sprague-Dawley , Sunitinib , Tissue Distribution , Young Adult
3.
J Pharm Biomed Anal ; 28(1): 23-30, 2002 Apr 01.
Article in English | MEDLINE | ID: mdl-11861105

ABSTRACT

A column trapping system has been incorporated into high performance liquid chromatography-nuclear magnetic resonance-mass spectrometry (HPLC-NMR-MS) to reduce data acquisition time of NMR experiments. The system uses a trapping column to capture analytes after the HPLC column and back flush trapped analyte to the flow cell of the NMR probe for detection. A dilution solvent is mixed with eluent from HPLC column to reduce the influence of the organic content in the mobile phase before column trapping. The trapping column is also coupled with a mass spectrometer (MS) to get complementary MS data on the same peak. Studies on 1-hydroxylated 9-amino-1,2,3,4-tetrahydro-acridine (1-OH tacrine), indomethacin and testosterone with the column trapping system showed good recovery of analytes and over 3-fold mean increase in UV-VIS signal intensity. The time saving on NMR experiments with the column trapping system was demonstrated by the analysis of dog microsomal incubate with tacrine.


Subject(s)
Microsomes/metabolism , Tacrine/analysis , Tacrine/metabolism , Animals , Cholinesterase Inhibitors/analysis , Cholinesterase Inhibitors/chemistry , Cholinesterase Inhibitors/metabolism , Chromatography, High Pressure Liquid/methods , Dogs , Mass Spectrometry , Nuclear Magnetic Resonance, Biomolecular , Tacrine/chemistry
4.
Drug Metab Lett ; 1(1): 61-5, 2007 Jan.
Article in English | MEDLINE | ID: mdl-19356020

ABSTRACT

Six tetra- and two penta-oxygenated capravirine metabolites observed in rats, dogs and humans represent the maximum numbers of isomers that can be predicted since oxygenations are restricted at the pyridinyl nitrogen (N-oxidation), sulfur (sulfoxidation), and isopropyl group (hydroxylation), exemplifying a unique case that is very unusual for sequential drug metabolism.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Imidazoles/pharmacokinetics , Oxygen/metabolism , Sulfur Compounds/pharmacokinetics , Administration, Oral , Animals , Chromatography, High Pressure Liquid , Dogs , Humans , Isomerism , Mass Spectrometry , Rats , Rats, Sprague-Dawley , Species Specificity
5.
Rapid Commun Mass Spectrom ; 21(20): 3317-22, 2007.
Article in English | MEDLINE | ID: mdl-17879390

ABSTRACT

Carbamazepine (CBZ)-induced idiosyncratic toxicities are commonly believed to be related to the formation of reactive metabolites. CBZ is metabolized primarily into carbamazepine-10,11-epoxide (CBZE), 2-hydroxycarbamazepine (2-OHCBZ) and 3-hydroxycarbamazepine (3-OHCBZ), in human liver microsomes (HLM). Over the past two decades, the 2,3-arene oxidation has been commonly assumed to be the major bioactivation pathway of CBZ. Recently, CBZE has been also confirmed to be chemically reactive. In order to identify other possible primary and sequential CBZ bioactivation pathways, individual HLM incubations of CBZ, CBZE, 2-OHCBZ and 3-OHCBZ were conducted in the presence of glutathione (GSH). In the CBZ incubation, a variety of GSH adducts were formed via individual or combined pathways of 10,11-epoxidation, arene oxidation and iminoquinone formation. In the CBZE incubation, the only detected GSH adducts were CBZE-SG1 and CBZE-SG2, which represented the two most abundant conjugates observed in the CBZ incubation. In the incubation of either 2-OHCBZ or 3-OHCBZ, a number of sequential GSH adducts were observed. However, none of the 2-OHCBZ-derived GSH adducts were detected in the CBZ incubation. Meanwhile, several GSH adducts were only observed in the CBZ incubation. In conclusion, CBZ can be bioactivated in HLM via 10,11-epoxidation, 2,3-arene oxidation, and several other pathways. In addition, the sequential bioactivation of 3-OHCBZ appeared to play a more important role than that of either CBZE or 2-OHCBZ in the overall bioactivation of CBZ in HLM. The identification of several new bioactivation pathways of CBZ in HLM demonstrates that possible CBZ bioactivation can be more complex than previously thought.


Subject(s)
Anticonvulsants/pharmacokinetics , Carbamazepine/pharmacokinetics , Chromatography, High Pressure Liquid , Microsomes, Liver/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Biotransformation , Glutathione/metabolism , Humans
6.
Drug Metab Dispos ; 35(9): 1593-602, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17567732

ABSTRACT

Metabolism of [(14)C]capravirine was studied via both in vitro and in vivo means in rats and dogs. Mass balance was achieved in rats and dogs, with mean total recovery of radioactivity >86% for each species. Capravirine was well absorbed in rats but only moderately so in dogs. The very low levels of recovered unchanged capravirine and the large number of metabolites observed in rats and dogs indicate that capravirine was eliminated predominantly by metabolism in both species. Capravirine underwent extensive metabolism via oxygenation reactions (predominant pathways in both species), depicolylation and carboxylation in rats, and decarbamation in dogs. The major circulating metabolites of capravirine were two depicolylated products in rats and three decarbamated products in dogs. However, none of the five metabolites was observed in humans, indicating significant species differences in terms of identities and relative abundances of circulating capravirine metabolites. Because the majority of in vivo oxygenated metabolites of capravirine were observed in liver microsomal incubations, the in vitro models provided good insight into the in vivo oxygenation pathways. In conclusion, the diversity (i.e., hydroxylation, sulfoxidation, sulfone formation, and N-oxidation), multiplicity (i.e., mono-, di-, tri-, and tetraoxygenations), and high enzymatic specificity (>90% contribution by CYP3A4 in humans, CYP3A1/2 in rats, and CYP3A12 in dogs) of the capravirine oxygenation reactions observed in humans, rats, and dogs in vivo and in vitro suggest that capravirine can be a useful CYP3A substrate for probing catalytic mechanisms and kinetics of CYP3A enzymes in humans and animal species.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Cytochrome P-450 CYP3A/metabolism , Imidazoles/pharmacokinetics , Sulfur Compounds/pharmacokinetics , Animals , Anti-HIV Agents/pharmacology , Antibodies, Blocking/pharmacology , Biotransformation , Chromatography, Liquid , Cytochrome P-450 CYP3A Inhibitors , Dogs , In Vitro Techniques , Male , Mass Spectrometry , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Rats , Rats, Sprague-Dawley , Recombinant Proteins/metabolism , Ritonavir/pharmacology , Species Specificity , Substrate Specificity
7.
Drug Metab Dispos ; 34(11): 1798-802, 2006 Nov.
Article in English | MEDLINE | ID: mdl-16914510

ABSTRACT

Capravirine, a new non-nucleoside reverse transcriptase inhibitor, undergoes extensive oxygenation reactions, including N-oxidation, sulfoxidation, sulfonation, and hydroxylation in humans. Numerous primary (mono-oxygenated) and sequential (di-, tri-, and tetraoxygenated) metabolites of capravirine are formed via the individual or combined oxygenation pathways. In this study, cytochrome P450 enzymes responsible for the primary and sequential oxygenation reactions of capravirine in human liver microsomes were identified at the specific pathway level. The total oxygenation of capravirine is mediated predominantly (>90%) by CYP3A4 and marginally (<10%) by CYP2C8, 2C9, and 2C19 in humans. Specifically, each of the two major mono-oxygenated metabolites C23 (sulfoxide) and C26 (N-oxide), is mediated predominantly (>90%) by CYP3A4 and slightly (<10%) by CYP2C8, the minor tertiary hydroxylated metabolite C19 by CYP3A4, 2C8, and 2C19, and the minor primary hydroxylated metabolite C20 by CYP3A4, 2C8, and 2C9. However, all sequential oxygenation reactions are mediated exclusively by CYP3A4. Due to their relatively insignificant contributions of C19 and C20 to total capravirine metabolism, no attempt was made to determine relative contributions of cytochrome P450 enzymes to the formation of the two minor metabolites.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Imidazoles/pharmacokinetics , Microsomes, Liver/enzymology , Sulfur Compounds/pharmacokinetics , Biotransformation , Humans , In Vitro Techniques , Microsomes, Liver/metabolism , Molecular Structure , Oxidation-Reduction
8.
Drug Metab Dispos ; 33(10): 1438-45, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16006566

ABSTRACT

Capravirine, a non-nucleoside reverse transcriptase inhibitor for the treatment of human immunodeficiency virus type 1, undergoes extensive oxygenations to numerous sequential metabolites in humans. Because several possible oxygenation pathways may be involved in the formation and/or sequential metabolism of a single metabolite, it is very difficult or even impossible to determine the definitive pathways and their relative contributions to the overall metabolism of capravirine using conventional approaches. For this reason, a human liver microsome-based "sequential incubation" method has been developed to deconvolute the complicated sequential metabolism of capravirine. In brief, the method includes three fundamental steps: 1) 30-min primary incubation of [(14)C]capravirine, 2) isolation of (14)C metabolites from the primary incubate, and 3) 30-min sequential incubation of each isolated (14)C metabolite supplemented with an ongoing (30 min) microsomal incubation with nonlabeled capravirine. Based on the extent of both the disappearance of the isolated precursor (14)C metabolites and the formation of sequential (14)C metabolites, definitive oxygenation pathways of capravirine were assigned. In addition, the percentage contribution of a precursor metabolite to the formation of each of its sequential metabolites (called sequential contribution) and the percentage contribution of a sequential metabolite formed from each of its precursor metabolites (called precursor contribution) were determined. An advantage of this system is that the sequential metabolism of each isolated (14)C metabolite can be monitored selectively by radioactivity in the presence of all relevant metabolic components (i.e., nonlabeled parent and its other metabolites). This methodology should be applicable to mechanistic studies of other compounds involving complicated sequential metabolic reactions when radiolabeled materials are available.


Subject(s)
Imidazoles/metabolism , Microsomes, Liver/metabolism , Reverse Transcriptase Inhibitors/metabolism , Biotransformation , Carbon Radioisotopes , Humans , Imidazoles/chemistry , In Vitro Techniques , Methods , Sulfur Compounds
9.
Drug Metab Dispos ; 33(12): 1920-4, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16204461

ABSTRACT

The clinical use of carbamazepine (CBZ), an anticonvulsant, is associated with a variety of idiosyncratic adverse reactions that are likely related to the formation of chemically reactive metabolites. CBZ-10,11-epoxide (CBZE), a pharmacologically active metabolite of CBZ, is so stable in vitro and in vivo that the potential for the epoxide to covalently interact with macromolecules has not been fully explored. In this study, two glutathione (GSH) adducts were observed when CBZE was incubated with GSH in the absence of biological matrices and cofactors (e.g., liver microsomes and NADPH). The chemical reactivity of CBZE was further confirmed by the in vitro finding that [14C]CBZE formed covalent protein adducts in human plasma as well as in human liver microsomes (HLMs) without NADPH. The two GSH adducts formed in the chemical reaction of CBZE were identical to the two major GSH adducts observed in the HLM incubation of CBZ, indicating that the 10,11-epoxidation represents a bioactivation pathway of CBZ. The two GSH adducts were isolated and identified as two diastereomers of 10-hydroxy-11-glutathionyl-CBZ by NMR. In addition, the covalent binding of [14C]CBZE was significantly increased in the HLM incubation upon addition of NADPH, indicating that CBZE can be further bioactivated by HLMs. To our knowledge, this is the first time the metabolite CBZE has been confirmed for its ability to form covalent protein adducts and the identity of the two CBZE-glutathionyl adducts has been confirmed by NMR. These represent important findings in the bioactivation mechanism of CBZ.


Subject(s)
Carbamazepine/analogs & derivatives , Carbamazepine/metabolism , Glutathione/metabolism , Humans , Microsomes, Liver/metabolism , Protein Binding
10.
Drug Metab Dispos ; 32(7): 689-98, 2004 Jul.
Article in English | MEDLINE | ID: mdl-15205383

ABSTRACT

Metabolism and disposition of capravirine, a new non-nucleoside reverse transcriptase inhibitor, were studied in healthy male volunteers who were randomly divided into two groups (A and B) with five subjects in each group. Group A received a single oral dose of [(14)C]capravirine (1400 mg) and group B received multiple oral doses of ritonavir (100 mg), followed by a single oral dose of [(14)C]capravirine (1400 mg). Mean total recoveries of radioactivity for groups A and B were 86.3% and 79.0%, respectively, with a mean cumulative recovery in urine comparable with that in feces for both groups. Excretion of unchanged capravirine was negligible in urine and low in feces for both groups. The results suggest that capravirine was well absorbed, with metabolism as the principal mechanism of clearance. Capravirine underwent extensive metabolism to a variety of metabolites via oxygenations (mono-, di-, tri-, and tetra-) representing the predominant pathway, glucuronidation, and sulfation in humans. No useful plasma profiles of group A were obtained due to extremely low levels of plasma radioactivity. Analysis of group B plasma indicated that unchanged capravirine was the major radiochemical component, with three monooxygenated products and a glucuronide of capravirine as the major circulating metabolites. Nineteen metabolites were identified using liquid chromatography-multistage ion-trap mass spectrometry methodologies. In summary, coadministration of low-dose ritonavir (a potent CYP3A4 inhibitor) drastically decreased the levels of sequential oxygenated metabolites and markedly increased the levels of the parent drug and primary oxygenated metabolites overall in plasma, urine, and feces.


Subject(s)
Anti-HIV Agents/metabolism , Cytochrome P-450 Enzyme Inhibitors , Reverse Transcriptase Inhibitors/metabolism , Ritonavir/pharmacology , Administration, Oral , Anti-HIV Agents/blood , Carbon Radioisotopes , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP3A , Drug Interactions , Feces/chemistry , Humans , Imidazoles , Male , Reverse Transcriptase Inhibitors/blood , Spectrometry, Mass, Electrospray Ionization , Sulfur Compounds , Time Factors
11.
Drug Metab Dispos ; 32(6): 639-46, 2004 Jun.
Article in English | MEDLINE | ID: mdl-15155556

ABSTRACT

Troglitazone (TGZ), the first glitazone used for the treatment of type II diabetes mellitus and removed from the market for liver toxicity, was shown to bind covalently to microsomal protein and glutathione (GSH) following activation by cytochrome P450 (P450). The covalent binding of (14)C-TGZ in dexamethasone-induced rat liver microsomes was NADPH-dependent and required the active form of P450; it was completely inhibited by ketoconazole (10 microM) and GSH (4 mM). The covalent binding in P450 3A4 Supersomes (9.2 nmol of TGZ Eq/nmol P450) was greater than that with P450 1A2 (0.7), 2C8 (3.7), 2C19 (1.4), 2E1 (0.6), and 2D6 (1.1) and 3A5 (3.0). The covalent binding in liver microsomes from rats pretreated with dexamethasone (5.3 nmol of TGZ Eq bound/nmol P450) was greater than that from rats pretreated with vehicle (3.5), beta-naphthoflavone (0.4), phenobarbital (1.1), or pyridine (2.5). A TGZ-GSH adduct was detected by liquid chromatography-tandem mass spectrometry and radioactivity detection with a deprotonated quasi-molecular ion [M-H](-) at m/z 745, with fragment ions at m/z 438 (deprotonated TGZ moiety), and at m/z 306 (deprotonated GSH moiety). The TGZ-GSH adduct was determined to be 5-glutathionyl-5-[4-(6-hydroxy-2,5,7,8-tetramethylchroman-2-ylmethoxy)benzyl]-thiazolidine-2,4-dione based on collision-induced dissociation fragmentation, and one- and two-dimensional NMR analysis of the isolated adduct. The synthetic 5-hydroxy TGZ and the benzylidene derivative of TGZ did not react with GSH or GSH ethyl ester. The mechanisms for metabolic activation of TGZ may involve an ultimate reactive sulfonium ion which could be formed from an initial sulfoxide followed by a formal Pummerer rearrangement, or a C5 thiazolidinedione radical or a sulfur cation radical.


Subject(s)
Chromans/pharmacokinetics , Hypoglycemic Agents/pharmacokinetics , Microsomes, Liver/metabolism , Thiazolidinediones/pharmacokinetics , Animals , Biotransformation , Carbon Radioisotopes , Cells, Cultured , Chromans/metabolism , Cytochrome P-450 Enzyme System/metabolism , Dexamethasone/pharmacology , Glutathione/chemistry , Glutathione/metabolism , Hepatocytes/enzymology , Hepatocytes/metabolism , Humans , Hypoglycemic Agents/metabolism , Isoenzymes/metabolism , Magnetic Resonance Spectroscopy , Mass Spectrometry , Microsomes, Liver/enzymology , Protein Binding , Rats , Rats, Inbred F344 , Thiazolidinediones/metabolism , Troglitazone
SELECTION OF CITATIONS
SEARCH DETAIL