Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
1.
Drug Metab Dispos ; 44(1): 102-14, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26512042

ABSTRACT

N-Methyl-2-[3-((E)-2-pyridin-2-yl-vinyl)-1H-indazol-6-ylsulfanyl]-benzamide (axitinib) is an oral inhibitor of vascular endothelial growth factor receptors 1-3, which is approved for the treatment of advanced renal cell cancer. Human [(14)C]-labeled clinical studies indicate axitinib's primary route of clearance is metabolism. The aims of the in vitro experiments presented herein were to identify and characterize the enzymes involved in axitinib metabolic clearance. In vitro biotransformation studies of axitinib identified a number of metabolites including an axitinib sulfoxide, several less abundant oxidative metabolites, and glucuronide conjugates. The most abundant NADPH- and UDPGA-dependent metabolites, axitinib sulfoxide (M12) and axitinib N-glucuronide (M7) were selected for phenotyping and kinetic study. Phenotyping experiments with human liver microsomes (HLMs) using chemical inhibitors and recombinant human cytochrome P450s demonstrated axitinib was predominately metabolized by CYP3A4/5, with minor contributions from CYP2C19 and CYP1A2. The apparent substrate concentration at half-maximal velocity (Km) and Vmax values for the formation of axitinib sulfoxide by CYP3A4 or CYP3A5 were 4.0 or 1.9 µM and 9.6 or 1.4 pmol·min(-1)·pmol(-1), respectively. Using a CYP3A4-specific inhibitor (Cyp3cide) in liver microsomes expressing CYP3A5, 66% of the axitinib intrinsic clearance was attributable to CYP3A4 and 15% to CYP3A5. Axitinib N-glucuronidation was primarily catalyzed by UDP-glucuronosyltransferase (UGT) UGT1A1, which was verified by chemical inhibitors and UGT1A1 null expressers, with lesser contributions from UGTs 1A3, 1A9, and 1A4. The Km and Vmax values describing the formation of the N-glucuronide in HLM or rUGT1A1 were 2.7 µM or 0.75 µM and 8.9 or 8.3 pmol·min(-1)·mg(-1), respectively. In summary, CYP3A4 is the major enzyme involved in axitinib clearance with lesser contributions from CYP3A5, CYP2C19, CYP1A2, and UGT1A1.


Subject(s)
Angiogenesis Inhibitors/metabolism , Cytochrome P-450 CYP3A/metabolism , Glucuronosyltransferase/metabolism , Imidazoles/metabolism , Indazoles/metabolism , Microsomes, Liver/enzymology , Protein Kinase Inhibitors/metabolism , Axitinib , Cytochrome P-450 CYP1A2/metabolism , Cytochrome P-450 CYP2C19/metabolism , Cytochrome P-450 CYP3A/genetics , Cytochrome P-450 Enzyme Inhibitors/pharmacology , Female , Genotype , Glucuronides/metabolism , Glucuronosyltransferase/genetics , Humans , Inactivation, Metabolic , Kinetics , Male , Metabolic Clearance Rate , Microsomes, Liver/drug effects , Models, Biological , Oxidation-Reduction , Phenotype , Recombinant Proteins/metabolism , Substrate Specificity , Sulfoxides/metabolism
2.
Drug Metab Dispos ; 41(8): 1575-83, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23729661

ABSTRACT

Axitinib is an inhibitor of tyrosine kinase vascular endothelin growth factor receptors 1, 2, and 3. The ATP-binding cassette (ABC) and solute carrier (SLC) transport properties of axitinib were determined in selected cellular systems. Axitinib exhibited high passive permeability in all cell lines evaluated (Papp ≥ 6 × 10(-6) cm/s). Active efflux was observed in Caco-2 cells, and further evaluation in multidrug resistance gene 1 (MDR1) or breast cancer resistance protein (BCRP) transfected Madin-Darby canine kidney cells type 2 (MDCK) cells indicated that axitinib is at most only a weak substrate for P-glycoprotein (P-gp) but not BCRP. Axitinib showed incomplete inhibition of P-gp-mediated transport of digoxin in Caco-2 cells and BCRP transport of topotecan in BCRP-transfected MDCK cells with IC50 values of 3 µM and 4.4 µM, respectively. Axitinib (10 mg) did not pose a risk for systemic drug interactions with P-gp or BCRP per regulatory guidance. A potential risk for drug interactions through inhibition of P-gp and BCRP in the gastrointestinal tract was identified because an axitinib dose of 10 mg divided by 250 mL was greater than 10-fold the IC50 for each transporter. However, a GastroPlus simulation that considered the low solubility of axitinib resulted in lower intestinal concentrations and suggested a low potential for gastrointestinal interactions with P-gp and BCRP substrates. Organic anion transporting polypeptide 1B1 (OATP1B1) and OATP1B3 transfected human embryonic kidney 293 (HEK293) cells transported axitinib to a minor extent but uptake into suspended hepatocytes was not inhibited by rifamycin SV suggesting that high passive permeability predominates. Mouse whole-body autoradiography revealed that [(14)C]axitinib-equivalents showed rapid absorption and distribution to all tissues except the brain. This suggests that efflux transport of axitinib may occur at the mouse blood-brain barrier.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , ATP-Binding Cassette Transporters/physiology , Imidazoles/metabolism , Indazoles/metabolism , Liver/metabolism , Neoplasm Proteins/physiology , Protein Kinase Inhibitors/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , ATP Binding Cassette Transporter, Subfamily G, Member 2 , ATP-Binding Cassette Transporters/antagonists & inhibitors , Animals , Autoradiography , Axitinib , Caco-2 Cells , Drug Interactions , Hepatocytes/metabolism , Humans , Imidazoles/chemistry , Indazoles/chemistry , Mice , Neoplasm Proteins/antagonists & inhibitors , Permeability , Risk Assessment , Solubility
3.
Drug Metab Dispos ; 40(5): 943-51, 2012 May.
Article in English | MEDLINE | ID: mdl-22328583

ABSTRACT

CYP2J2, an arachidonic acid epoxygenase, is recognized for its role in the first-pass metabolism of astemizole and ebastine. To fully assess the role of CYP2J2 in drug metabolism, a selective substrate and potent specific chemical inhibitor are essential. In this study, we report amiodarone 4-hydoxylation as a specific CYP2J2-catalyzed reaction with no CYP3A4, or other drug-metabolizing enzyme, involvement. Amiodarone 4-hydroxylation enabled the determination of liver relative activity factor and intersystem extrapolation factor for CYP2J2. Amiodarone 4-hydroxylation correlated with astemizole O-demethylation but not with CYP2J2 protein content in a sample of human liver microsomes. To identify a specific CYP2J2 inhibitor, 138 drugs were screened using terfenadine and astemizole as probe substrates with recombinant CYP2J2. Forty-two drugs inhibited CYP2J2 activity by ≥50% at 30 µM, but inhibition was substrate-dependent. Of these, danazol was a potent inhibitor of both hydroxylation of terfenadine (IC(50) = 77 nM) and O-demethylation of astemizole (K(i) = 20 nM), and inhibition was mostly competitive. Danazol inhibited CYP2C9, CYP2C8, and CYP2D6 with IC(50) values of 1.44, 1.95, and 2.74 µM, respectively. Amiodarone or astemizole were included in a seven-probe cocktail for cytochrome P450 (P450) drug-interaction screening potential, and astemizole demonstrated a better profile because it did not appreciably interact with other P450 probes. Thus, danazol, amiodarone, and astemizole will facilitate the ability to determine the metabolic role of CYP2J2 in hepatic and extrahepatic tissues.


Subject(s)
Amiodarone/metabolism , Astemizole/metabolism , Cytochrome P-450 Enzyme System/metabolism , Danazol , Enzyme Inhibitors , Microsomes, Liver/enzymology , Terfenadine/metabolism , Amiodarone/chemistry , Astemizole/chemistry , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2J2 , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/genetics , Danazol/chemistry , Danazol/metabolism , Danazol/pharmacology , Drug Discovery , Drug Interactions , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Humans , Hydroxylation , In Vitro Techniques , Methylation , Microsomes, Liver/metabolism , Models, Biological , Molecular Structure , Substrate Specificity , Tandem Mass Spectrometry , Terfenadine/chemistry
4.
Drug Metab Dispos ; 38(2): 347-56, 2010 Feb.
Article in English | MEDLINE | ID: mdl-19923256

ABSTRACT

Several antihistamine drugs including terfenadine, ebastine, and astemizole have been identified as substrates for CYP2J2. The overall importance of this enzyme in drug metabolism has not been fully explored. In this study, 139 marketed therapeutic agents and compounds were screened as potential CYP2J2 substrates. Eight novel substrates were identified that vary in size and overall topology from relatively rigid structures (amiodarone) to larger complex structures (cyclosporine). The substrates displayed in vitro intrinsic clearance values ranging from 0.06 to 3.98 mul/min/pmol CYP2J2. Substrates identified for CYP2J2 are also metabolized by CYP3A4. Extracted ion chromatograms of metabolites observed for albendazole, amiodarone, astemizole, thioridazine, mesoridazine, and danazol showed marked differences in the regioselectivity of CYP2J2 and CYP3A4. CYP3A4 commonly metabolized compounds at multiple sites, whereas CYP2J2 metabolism was more restrictive and limited, in general, to a single site for large compounds. Although the CYP2J2 active site can accommodate large substrates, it may be more narrow than CYP3A4, limiting metabolism to moieties that can extend closer toward the active heme iron. For albendazole, CYP2J2 forms a unique metabolite compared with CYP3A4. Albendazole and amiodarone were evaluated in various in vitro systems including recombinant CYP2J2 and CYP3A4, pooled human liver microsomes (HLM), and human intestinal microsomes (HIM). The Michaelis-Menten-derived intrinsic clearance of N-desethyl amiodarone was 4.6 greater in HLM than in HIM and 17-fold greater in recombinant CYP3A4 than in recombinant CYP2J2. The resulting data suggest that CYP2J2 may be an unrecognized participant in first-pass metabolism, but its contribution is minor relative to that of CYP3A4.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Pharmaceutical Preparations/metabolism , Algorithms , Catalytic Domain , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP2J2 , Cytochrome P-450 CYP3A/chemistry , Cytochrome P-450 CYP3A/metabolism , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/chemistry , Enzyme Inhibitors/pharmacology , Humans , Intestinal Mucosa/metabolism , Isoenzymes/chemistry , Isoenzymes/metabolism , Kinetics , Liver/metabolism , Microsomes/metabolism , Models, Structural , Organ Specificity , Pharmaceutical Preparations/chemistry , Recombinant Proteins/metabolism , Spectrometry, Mass, Electrospray Ionization , Substrate Specificity , Tandem Mass Spectrometry
5.
Int J Pharm ; 308(1-2): 133-9, 2006 Feb 03.
Article in English | MEDLINE | ID: mdl-16352407

ABSTRACT

PK express module is a physiologically based model of first pass metabolism, which integrates in vitro data with an in silico physiologically based pharmacokinetic (PBPK) model to predict human bioavailability (F(H)). There are three required inputs: FDp (Fraction dose absorbed, final parameter from iDEA absorption module), protein binding (fu) and disappearance kinetics in human hepatocytes. Caco-2 permeability, aqueous solubility (at multiple pH's), estimated dose and chemical structure are inputs required for the estimation of FDp (Norris et al., 2000; Stoner et al., 2004) and were determined for all compounds in our laboratory or obtained from literature. Protein binding data was collected from literature references and/or Pfizer database. Human hepatocyte data was generated in-house using an automated human hepatocyte method (using Tecan Genesis Workstation) as described previously (). Sixteen compounds (commercial and Pfizer compounds) were chosen to evaluate the PK express model and the bioavailability predicted from the module was compared with known clinical endpoints. For majority of the 16 compounds (approximately 80%), the PK express model F(H) values were comparable to the known human bioavailability (F(H)) (within 23.7 units of the known human (true) F, except for PF 3, PF 4, PF 6). In conclusion, the PK express model integrates a number of key readily available discovery parameters and provides estimates of human performance by integrating in silico and experimental variables built on a physiological based pharmacokinetic model. Information from this model in conjunction with other ADME data (e.g., P450 inhibition) will enable progression of most promising compounds for further in vivo PK and/or efficacy studies.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Biological , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Biological Availability , Caco-2 Cells , Hepatocytes/metabolism , Humans , Hydrogen-Ion Concentration , In Vitro Techniques , Intestinal Absorption , Intestinal Mucosa/metabolism , Pharmaceutical Preparations/chemistry , Protein Binding , Reproducibility of Results , Retrospective Studies , Solubility
6.
J Pharm Biomed Anal ; 37(2): 319-26, 2005 Feb 23.
Article in English | MEDLINE | ID: mdl-15708673

ABSTRACT

An automated high throughput human hepatocyte assay has been established with a 96-well format using a Tecan Genesistrade mark Workstation. Validation of this assay was performed with nine commercially available compounds and an additional 10 Pfizer compounds with varying hepatic extraction ratios (E(H)) ranging from 0.02 to approximately 1. The incubation conditions in the automated assay are readily and precisely controlled and cell viability of over 80% was achieved in the automated assay further confirming its utility for absorption, distribution, metabolism, and excretion (toxicity) (ADME (T)) screening. The results of the nine commercial compounds correlate with both manually executed (R(2)=0.97) and literature reported experimental results (R(2)=0.93). Overall, measured E(H)s were within two-fold of the literature values for approximately 90% of the 19 compounds tested. Additionally, good inter- and intra-day reproducibility was observed for all the 19 compounds. In conclusion, an automated and robust assay suitable for simultaneously testing up to 48 compounds with multiple time points has been validated. Throughput of 192 compounds per run can be achieved using 384-well plates to meet increasing needs in drug discovery. Currently, this automated assay is used to support early discovery profiling towards lead optimization of various discovery targets/programs.


Subject(s)
Drug Evaluation, Preclinical/methods , Drugs, Investigational/pharmacokinetics , Hepatocytes/metabolism , Cell Survival , Cells, Cultured , Drug Evaluation, Preclinical/instrumentation , Humans , Inactivation, Metabolic , Metabolic Clearance Rate , Reference Standards
SELECTION OF CITATIONS
SEARCH DETAIL