Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
3.
Biopharm Drug Dispos ; 37(6): 379-83, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27417918

ABSTRACT

Cynomolgus monkeys are used frequently in preclinical studies for new drug development due to their evolutionary closeness to humans. An antiretroviral drug, efavirenz, is a typical probe substrate for human cytochrome P450 (P450) 2B6, but is mainly metabolized by cynomolgus monkey P450 2C9. In this study, plasma concentrations of efavirenz were assessed in six cynomolgus monkeys genotyped for P450 2C9 c.334 A > C (I112L) (three wild-type, one heterozygote and two homozygotes) by high performance liquid chromatography with tandem mass spectrometry. After intravenous administration at a dose of 1.0 mg/kg, biphasic plasma elimination curves of efavirenz were seen in these cynomolgus monkeys. The mean plasma concentration of the primary metabolite 8-hydroxyefavirenz (1 h after treatment, with hydrolysis by ß-glucuronidase) in the wild-type group was significantly higher (4.0-fold) than the combined heterozygous and homozygous group mean. The area under the plasma concentration-time curve value of efavirenz in the homozygous group after oral administration at a dose of 2.0 mg/kg was significantly higher (2.0-fold) than the combined wild-type and heterozygous group. These results collectively indicated that P450 2C9 c.334 A > C (I112L) variation was associated with efavirenz metabolic clearance in vivo. Cynomolgus P450 2C9 polymorphism might account for interindividual variations of efavirenz metabolism in cynomolgus monkeys. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Benzoxazines/pharmacokinetics , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Reverse Transcriptase Inhibitors/pharmacokinetics , Alkynes , Animals , Anti-HIV Agents/blood , Benzoxazines/blood , Benzoxazines/metabolism , Cyclopropanes , Genotype , Macaca fascicularis , Reverse Transcriptase Inhibitors/blood
4.
Biopharm Drug Dispos ; 37(5): 310-3, 2016 Jul.
Article in English | MEDLINE | ID: mdl-26581561

ABSTRACT

Cynomolgus monkeys are widely used in drug developmental stages as non-human primate models. Previous studies used 89 compounds to investigate species differences associated with cytochrome P450 (P450 or CYP) function that reported monkey specific CYP2C76 cleared 19 chemicals, and homologous CYP2C9 and CYP2C19 metabolized 17 and 30 human CYP2C9 and/or CYP2C19 substrates/inhibitors, respectively. In the present study, 22 compounds selected from viewpoints of global drug interaction guidances and guidelines were further evaluated to seek potential substrates for monkey CYP2C8, which is highly homologous to human CYP2C8 (92%). Amodiaquine, montelukast, quercetin and rosiglitazone, known as substrates or competitive inhibitors of human CYP2C8, were metabolically depleted by recombinant monkey CYP2C8 at relatively high rates. Taken together with our reported findings of the slow eliminations of amodiaquine and montelukast by monkey CYP2C9, CYP2C19 and CYP2C76, the present results suggest that these at least four chemicals may be good marker substrates for monkey CYP2C8. Copyright © 2016 John Wiley & Sons, Ltd.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Acetates/pharmacology , Amodiaquine/pharmacology , Animals , Cyclopropanes , Macaca fascicularis , Pharmaceutical Preparations/metabolism , Quercetin/pharmacology , Quinolines/pharmacology , Rosiglitazone , Species Specificity , Substrate Specificity , Sulfides , Thiazolidinediones/pharmacology
5.
Biopharm Drug Dispos ; 36(9): 636-43, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26348733

ABSTRACT

Cynomolgus monkeys are used widely in preclinical studies as non-human primate species. The amino acid sequence of cynomolgus monkey cytochrome P450 (P450 or CYP) 2C19 is reportedly highly correlated to that of human CYP2C19 (92%) and CYP2C9 (93%). In the present study, 89 commercially available compounds were screened to find potential substrates for cynomolgus monkey CYP2C19. Of 89 drugs, 34 were metabolically depleted by cynomolgus monkey CYP2C19 with relatively high rates. Among them, 30 compounds have been reported as substrates or inhibitors of, either or both, human CYP2C19 and CYP2C9. Several compounds, including loratadine, showed high selectivity to cynomolgus monkey CYP2C19, and all of these have been reported as human CYP2C19 and/or CYP2C9 substrates. In addition, cynomolgus monkey CYP2C19 formed the same loratadine metabolite as human CYP2C19, descarboethoxyloratadine. These results suggest that cynomolgus monkey CYP2C19 is generally similar to human CYP2C19 and CYP2C9 in its substrate recognition functionality.


Subject(s)
Cytochrome P-450 CYP2C19/metabolism , Macaca fascicularis/metabolism , Xenobiotics/metabolism , Animals , Biotransformation , Chromatography, High Pressure Liquid , Chromatography, Reverse-Phase , Cytochrome P-450 CYP2C19/genetics , Cytochrome P-450 CYP2C9/genetics , Cytochrome P-450 CYP2C9/metabolism , Histamine H1 Antagonists, Non-Sedating/chemistry , Histamine H1 Antagonists, Non-Sedating/metabolism , Humans , Isoenzymes/genetics , Isoenzymes/metabolism , Loratadine/analogs & derivatives , Loratadine/chemistry , Loratadine/metabolism , Molecular Structure , Oxidation-Reduction , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity , Tandem Mass Spectrometry , Xenobiotics/chemistry
6.
Drug Metab Dispos ; 43(7): 1119-22, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25948712

ABSTRACT

Cynomolgus monkeys are widely used as primate models in preclinical studies, because of their evolutionary closeness to humans. In humans, the cytochrome P450 (P450) 2C enzymes are important drug-metabolizing enzymes and highly expressed in livers. The CYP2C enzymes, including CYP2C9, are also expressed abundantly in cynomolgus monkey liver and metabolize some endogenous and exogenous substances like testosterone, S-mephenytoin, and diclofenac. However, comprehensive evaluation regarding substrate specificity of monkey CYP2C9 has not been conducted. In the present study, 89 commercially available drugs were examined to find potential monkey CYP2C9 substrates. Among the compounds screened, 20 drugs were metabolized by monkey CYP2C9 at a relatively high rates. Seventeen of these compounds were substrates or inhibitors of human CYP2C9 or CYP2C19, whereas three drugs were not, indicating that substrate specificity of monkey CYP2C9 resembled those of human CYP2C9 or CYP2C19, with some differences in substrate specificities. Although efavirenz is known as a marker substrate for human CYP2B6, efavirenz was not oxidized by CYP2B6 but by CYP2C9 in monkeys. Liquid chromatography-mass spectrometry analysis revealed that monkey CYP2C9 and human CYP2B6 formed the same mono- and di-oxidized metabolites of efavirenz at 8 and 14 positions. These results suggest that the efavirenz 8-oxidation could be one of the selective markers for cynomolgus monkey CYP2C9 among the major three CYP2C enzymes tested. Therefore, monkey CYP2C9 has the possibility of contributing to limited specific differences in drug oxidative metabolism between cynomolgus monkeys and humans.


Subject(s)
Anti-HIV Agents/metabolism , Benzoxazines/metabolism , Cytochrome P-450 CYP2C9/metabolism , Alkynes , Animals , Cyclopropanes , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , Humans , Kinetics , Macaca fascicularis , Oxidation-Reduction , Pharmaceutical Preparations/metabolism , Substrate Specificity
7.
Drug Metab Dispos ; 43(1): 27-33, 2015 Jan.
Article in English | MEDLINE | ID: mdl-25318994

ABSTRACT

Cynomolgus monkeys are widely used in preclinical studies during drug development because of their evolutionary closeness to humans, including their cytochrome P450s (P450s). Most cynomolgus monkey P450s are almost identical (≥90%) to human P450s; however, CYP2C76 has low sequence identity (approximately 80%) to any human CYP2Cs. Although CYP2C76 has no ortholog in humans and is partly responsible for species differences in drug metabolism between cynomolgus monkeys and humans, a broad evaluation of potential substrates for CYP2C76 has not yet been conducted. In this study, a screening of 89 marketed compounds, including human CYP2C and non-CYP2C substrates or inhibitors, was conducted to find potential CYP2C76 substrates. Among the compounds screened, 19 chemicals were identified as substrates for CYP2C76, including substrates for human CYP1A2 (7-ethoxyresorufin), CYP2B6 (bupropion), CYP2D6 (dextromethorphan), and CYP3A4/5 (dextromethorphan and nifedipine), and inhibitors for CYP2B6 (sertraline, clopidogrel, and ticlopidine), CYP2C8 (quercetin), CYP2C19 (ticlopidine and nootkatone), and CYP3A4/5 (troleandomycin). CYP2C76 metabolized a wide variety of the compounds with diverse structures. Among them, bupropion and nifedipine showed high selectivity to CYP2C76. As for nifedipine, CYP2C76 formed methylhydroxylated nifedipine, which was not produced by monkey CYP2C9, CYP2C19, or CYP3A4, as identified by mass spectrometry and estimated by a molecular docking simulation. This unique oxidative metabolite formation of nifedipine could be one of the selective marker reactions of CYP2C76 among the major CYP2Cs and CYP3As tested. These results suggest that monkey CYP2C76 contributes to bupropion hydroxylation and formation of different nifedipine oxidative metabolites as a result of its relatively large substrate cavity.


Subject(s)
Bupropion/metabolism , Cytochrome P-450 Enzyme System/metabolism , Macaca fascicularis/metabolism , Nifedipine/metabolism , Oxidoreductases/metabolism , Animals , Humans , Hydroxylation/physiology , Molecular Docking Simulation/methods
8.
Antimicrob Agents Chemother ; 58(7): 3837-42, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24752277

ABSTRACT

Onychomycosis is a common fungal nail disease that is difficult to treat topically due to the deep location of the infection under the densely keratinized nail plate. Keratin affinity of topical drugs is an important physicochemical property impacting therapeutic efficacy. To be effective, topical drugs must penetrate the nail bed and retain their antifungal activity within the nail matrix, both of which are adversely affected by keratin binding. We investigated these properties for efinaconazole, a new topical antifungal for onychomycosis, compared with those of the existing topical drugs ciclopirox and amorolfine. The efinaconazole free-drug concentration in keratin suspensions was 14.3%, significantly higher than the concentrations of ciclopirox and amorolfine, which were 0.7% and 1.9%, respectively (P < 0.001). Efinaconazole was released from keratin at a higher proportion than in the reference drugs, with about half of the remaining keratin-bound efinaconazole removed after washing. In single-dose in vitro studies, efinaconazole penetrated full-thickness human nails into the receptor phase and also inhibited the growth of Trichophyton rubrum under the nail. In the presence of keratin, efinaconazole exhibited fungicidal activity against Trichophyton mentagrophytes comparable to that of amorolfine and superior to that of ciclopirox. In a guinea pig onychomycosis model with T. mentagrophytes infection, an efinaconazole solution significantly decreased nail fungal burden compared to that of ciclopirox and amorolfine lacquers (P < 0.01). These results suggest that the high nail permeability of efinaconazole and its potent fungicidal activity in the presence of keratin are related to its low keratin affinity, which may contribute to its efficacy in onychomycosis.


Subject(s)
Antifungal Agents/pharmacokinetics , Antifungal Agents/therapeutic use , Keratins/metabolism , Nails/metabolism , Onychomycosis/drug therapy , Triazoles/pharmacokinetics , Triazoles/therapeutic use , Administration, Topical , Animals , Antifungal Agents/administration & dosage , Guinea Pigs , Humans , In Vitro Techniques , Microbial Sensitivity Tests , Tinea/drug therapy , Tinea/microbiology , Triazoles/administration & dosage , Trichophyton/drug effects
9.
J Vet Med Sci ; 76(12): 1647-50, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25649950

ABSTRACT

Cytochromes P450 (P450) are important for not only drug metabolism and toxicity, but also biosynthesis and metabolism of cholesterol and bile acids, and steroid synthesis. In cynomolgus macaques, widely used in biomedical research, we have characterized P450 cDNAs, which were isolated as expressed sequence tags of cynomolgus macaque liver. In this study, cynomolgus CYP7A1, CYP17A1, CYP20A1, CYP27A1 and CYP51A1 cDNAs were characterized by sequence analysis, phylogenetic analysis and tissue expression pattern. By sequence analysis, these five cynomolgus P450s had high sequence identities (94-99%) to the human orthologs in amino acids. By phylogenetic analysis, each cynomolgus P450 was more closely related to the human ortholog as compared with the dog or rat ortholog. By quantitative polymerase chain reaction, among the 10 tissue types, CYP7A1 and CYP17A1 mRNAs were preferentially expressed in liver and adrenal gland, respectively. Cynomolgus CYP27A1 and CYP51A1 mRNAs were most abundantly expressed in liver and testis, respectively. Cynomolgus CYP20A1 mRNA was expressed in all the tissues, including brain and liver. Tissue expression patterns of each cynomolgus P450 were generally similar to that of the human ortholog. These results suggest the molecular similarities of CYP7A1, CYP17A1, CYP20A1, CYP27A1 and CYP51A1 between cynomolgus macaques and humans.


Subject(s)
Cholesterol 7-alpha-Hydroxylase/genetics , Macaca fascicularis/genetics , Phylogeny , Steroid 17-alpha-Hydroxylase/genetics , Sterol 14-Demethylase/genetics , Adrenal Glands/metabolism , Animals , Base Sequence , Cholesterol 7-alpha-Hydroxylase/metabolism , Liver/metabolism , Macaca fascicularis/metabolism , Molecular Sequence Data , Sequence Analysis, DNA , Sequence Homology , Species Specificity , Steroid 17-alpha-Hydroxylase/metabolism , Sterol 14-Demethylase/metabolism
10.
Xenobiotica ; 43(12): 1037-42, 2013 Dec.
Article in English | MEDLINE | ID: mdl-23679834

ABSTRACT

Caffeine (1,3,7-trimethylxanthine) is a phenotyping substrate for human cytochrome P450 1A2. 3-N-Demethylation of caffeine is the main human metabolic pathway, whereas monkeys extensively mediate the 7-N-demethylation of caffeine to form pharmacological active theophylline. Roles of monkey P450 enzymes in theophylline formation from caffeine were investigated using individual monkey liver microsomes and 14 recombinantly expressed monkey P450 enzymes, and the results were compared with those for human P450 enzymes. Caffeine 7-N-demethylation activity in microsomes from 20 monkey livers was not strongly inhibited by α-naphthoflavone, quinidine or ketoconazole, and was roughly correlated with diclofenac 4'-hydroxylation activities. Monkey P450 2C9 had the highest activity for caffeine 7-N-demethylation. Kinetic analysis revealed that monkey P450 2C9 had a high Vmax/Km value for caffeine 7-N-demethylation, comparable to low Km value for monkey liver microsomes. Caffeine could dock favorably with monkey P450 2C9 modeled for 7-N-demethylation and with human P450 1A2 for 3-N-demethylation. The primary metabolite theophylline was oxidized to 8-hydroxytheophylline in similar ways by liver microsomes and by recombinant P450s in both humans and monkeys. These results collectively suggest a high activity for monkey liver P450 2C9 toward caffeine 7-N-demethylation, whereas, in humans, P450 1A2-mediated caffeine 3-N-demethylation is dominant.


Subject(s)
Caffeine/metabolism , Cytochrome P-450 Enzyme System/metabolism , Haplorhini/metabolism , Liver/enzymology , Theophylline/metabolism , Animals , Caffeine/chemistry , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Humans , Hydroxylation/drug effects , Liver/drug effects , Metabolic Networks and Pathways/drug effects , Methylation/drug effects , Microsomes, Liver/enzymology , Molecular Docking Simulation , Oxidation-Reduction/drug effects , Recombinant Proteins/metabolism , Theophylline/chemistry
11.
Arch Biochem Biophys ; 466(1): 98-105, 2007 Oct 01.
Article in English | MEDLINE | ID: mdl-17689485

ABSTRACT

Cynomolgus monkey CYP2C76 does not have a corresponding ortholog in humans, and it is at least partly responsible for differences in drug metabolism between monkeys and humans. To determine if CYP2C76 is the only monkey-specific CYP gene, we identified cynomolgus monkey cDNAs for CYP2A23, CYP2A24, CYP2E1, CYP2J2, CYP3A5, CYP3A8, CYP4A11, CYP4F3, CYP4F11, CYP4F12, and CYP4F45. These CYP cDNAs showed a high sequence identity (93-96%) to the homologous human CYP cDNAs. The monkey CYPs were preferentially expressed in liver among the analyzed tissues. Moreover, all five analyzed monkey CYPs (CYP2A23, CYP2A24, CYP2E1, CYP3A5, and CYP3A8) metabolized typical substrates for human CYPs in the corresponding subfamilies. These results suggest that these 11 monkey CYP cDNAs are closely related to the human CYP cDNAs and thus, unlike CYP2C76, are not apparent monkey-specific cDNAs.


Subject(s)
Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/physiology , Amino Acid Sequence , Animals , Base Sequence , DNA, Complementary/genetics , Humans , Macaca fascicularis , Molecular Sequence Data , Organ Specificity , Sequence Homology , Species Specificity , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...