Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Dig Dis Sci ; 67(11): 5107-5115, 2022 11.
Article in English | MEDLINE | ID: mdl-35122589

ABSTRACT

BACKGROUND AND AIMS: To determine prevalence and clinical utility of pathogenic germline variants (PGV) in gastric and esophageal cancer patients using universal genetic testing approach. METHODS: We undertook a prospective study of germline sequencing using an > 80 gene next-generation sequencing platform among patients with gastric and esophageal cancers receiving care at Mayo Clinic Cancer Center between April 1, 2018, and March 31, 2020. Patients were not selected based on cancer stage, family history of cancer, ethnicity, or age. Family cascade testing was offered at no cost. RESULTS: A total of 96 patients were evaluated. Median age was 66 years, 80.2% were male, 89.6% were white. Nearly 39% of the cohort had esophageal cancer, 35.4% gastric cancer and 26% gastroesophageal junction cancers. Approximately half (52%) of the patients had metastatic disease. Pathogenic germline variants (PGV) were detected in 15.6% (n = 15) patients. The prevalence of PGV was 10.8% in esophageal cancer, 17.6% in gastric cancer and 20% in gastroesophageal cancer. Eighty percent of patients with a positive result would not have been detected by screening with standard guidelines for genetic testing. Most PGV detected included genes with high and moderate penetrance related to DNA damage response including BRCA1, BRCA2, PALB2 and ATM. CONCLUSIONS: Universal multi-gene panel testing in gastric and esophageal cancers was associated with detection of heritable mutations in 15% of patients. The majority of PGV would not be detected with current screening guidelines and are related to DNA damage response.


Subject(s)
Esophageal Neoplasms , Stomach Neoplasms , Humans , Male , Aged , Female , Prospective Studies , Esophageal Neoplasms/diagnosis , Esophageal Neoplasms/epidemiology , Esophageal Neoplasms/genetics , Stomach Neoplasms/diagnosis , Stomach Neoplasms/epidemiology , Stomach Neoplasms/genetics , Germ-Line Mutation , Genetic Testing , Germ Cells , Genetic Predisposition to Disease
2.
Clin Pharmacol Ther ; 95(6): 653-62, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24569517

ABSTRACT

Fluoxetine and its circulating metabolite norfluoxetine comprise a complex multiple-inhibitor system that causes reversible or time-dependent inhibition of the cytochrome P450 (CYP) family members CYP2D6, CYP3A4, and CYP2C19 in vitro. Although significant inhibition of all three enzymes in vivo was predicted, the areas under the concentration-time curve (AUCs) for midazolam and lovastatin were unaffected by 2-week dosing of fluoxetine, whereas the AUCs of dextromethorphan and omeprazole were increased by 27- and 7.1-fold, respectively. This observed discrepancy between in vitro risk assessment and in vivo drug-drug interaction (DDI) profile was rationalized by time-varying dynamic pharmacokinetic models that incorporated circulating concentrations of fluoxetine and norfluoxetine enantiomers, mutual inhibitor-inhibitor interactions, and CYP3A4 induction. The dynamic models predicted all DDIs with less than twofold error. This study demonstrates that complex DDIs that involve multiple mechanisms, pathways, and inhibitors with their metabolites can be predicted and rationalized via characterization of all the inhibitory species in vitro.


Subject(s)
Antidepressive Agents, Second-Generation/pharmacology , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP2D6/metabolism , Cytochrome P-450 CYP3A/metabolism , Fluoxetine/analogs & derivatives , Adult , Antidepressive Agents, Second-Generation/administration & dosage , Antidepressive Agents, Second-Generation/pharmacokinetics , Area Under Curve , Aryl Hydrocarbon Hydroxylases/antagonists & inhibitors , Biotransformation , Computer Simulation , Cytochrome P-450 CYP2C19 , Cytochrome P-450 CYP2D6 Inhibitors , Cytochrome P-450 CYP3A Inhibitors , Dextromethorphan/pharmacokinetics , Drug Interactions , Female , Fluoxetine/administration & dosage , Fluoxetine/pharmacokinetics , Fluoxetine/pharmacology , Half-Life , Hepatocytes/drug effects , Hepatocytes/metabolism , Humans , Lovastatin/pharmacokinetics , Male , Midazolam/pharmacokinetics , Models, Statistical , Omeprazole/pharmacokinetics , Stereoisomerism
3.
Clin Pharmacol Ther ; 89(6): 888-95, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21490593

ABSTRACT

An endogenous probe for CYP3A activity would be useful for early identification of in vivo cytochrome P450 (CYP) 3A4 inhibitors. The aim of this study was to determine whether formation clearance (CL(f)) of the sum of 6ß-hydroxycortisol and 6ß-hydroxycortisone is a useful probe of CYP3A4 inhibition in vivo. In human liver microsomes (HLMs), the formation of 6ß-hydroxycortisol and 6ß-hydroxycortisone was catalyzed by CYP3A4, and itraconazole inhibited these reactions with half maximal inhibitory concentration (IC(50))(,u) values of 3.1 nmol/l and 3.4 nmol/l, respectively. The in vivo IC(50,u) value of itraconazole for the combined CL(f) of 6ß-hydroxycortisone and 6ß-hydroxycortisol was 1.6 nmol/l. The greater inhibitory potency in vivo is probably due to circulating inhibitory itraconazole metabolites. The maximum in vivo inhibition was 59%, suggesting that f(m,CYP3A4) for cortisol and cortisone 6ß-hydroxylation is ~60%. Given the significant decrease in CL(f) of 6ß-hydroxycortisone and 6ß-hydroxycortisol after 200-mg and 400-mg single doses of itraconazole, this endogenous probe can be used to detect moderate and potent CYP3A4 inhibition in vivo.


Subject(s)
Cortisone/analogs & derivatives , Cytochrome P-450 CYP3A Inhibitors , Cytochrome P-450 CYP3A/biosynthesis , Hydrocortisone/analogs & derivatives , Molecular Probes/metabolism , Cortisone/antagonists & inhibitors , Cortisone/metabolism , Cytochrome P-450 CYP3A/metabolism , Drug Combinations , Drug Evaluation, Preclinical/methods , Humans , Hydrocortisone/antagonists & inhibitors , Hydrocortisone/biosynthesis , Hydrocortisone/metabolism , Itraconazole/metabolism , Itraconazole/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Reproducibility of Results
4.
Clin Pharmacol Ther ; 88(4): 499-505, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20739919

ABSTRACT

Inhibitory drug metabolites may contribute to drug-drug interactions (DDIs). The aim of this study was to determine the importance of inhibitory metabolites of itraconazole (ITZ) in in vivo cytochrome P450 (CYP) 3A4 inhibition. The pharmacokinetics of ITZ and midazolam (MDZ) were determined in six healthy volunteers in four sessions after administration of MDZ with and without oral ITZ. After doses of 50, 200, and 400 mg of ITZ, the clearance of orally administered MDZ decreased by 27, 74, and 83%, respectively. The in vivo half maximal inhibitory concentration (IC(50)) for ITZ ranged from 5 to 132 nmol/l in the six subjects. The metabolites of ITZ were estimated to account for ~50% of the total CYP3A4 inhibition, with the relative contribution increasing with time after ITZ dosing. Of the total of 18 interactions observed, 15 (84%) could be predicted within a twofold error margin, with improved accuracy observed when ITZ metabolites were included in the predictions. This study shows that the metabolites of ITZ contribute to CYP3A4 inhibition and need to be accounted for in quantitative rationalization of ITZ-mediated DDIs.


Subject(s)
Cytochrome P-450 CYP3A Inhibitors , Enzyme Inhibitors/pharmacology , Itraconazole/pharmacology , Midazolam/pharmacokinetics , Adult , Area Under Curve , Cytochrome P-450 CYP3A , Dose-Response Relationship, Drug , Drug Interactions , Female , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Male , Metabolic Clearance Rate , Young Adult
5.
Clin Pharmacol Ther ; 83(1): 77-85, 2008 Jan.
Article in English | MEDLINE | ID: mdl-17495874

ABSTRACT

Itraconazole (ITZ) is metabolized in vitro to three inhibitory metabolites: hydroxy-itraconazole (OH-ITZ), keto-itraconazole (keto-ITZ), and N-desalkyl-itraconazole (ND-ITZ). The goal of this study was to determine the contribution of these metabolites to drug-drug interactions caused by ITZ. Six healthy volunteers received 100 mg ITZ orally for 7 days, and pharmacokinetic analysis was conducted at days 1 and 7 of the study. The extent of CYP3A4 inhibition by ITZ and its metabolites was predicted using this data. ITZ, OH-ITZ, keto-ITZ, and ND-ITZ were detected in plasma samples of all volunteers. A 3.9-fold decrease in the hepatic intrinsic clearance of a CYP3A4 substrate was predicted using the average unbound steady-state concentrations (C(ss,ave,u)) and liver microsomal inhibition constants for ITZ, OH-ITZ, keto-ITZ, and ND-ITZ. Accounting for circulating metabolites of ITZ significantly improved the in vitro to in vivo extrapolation of CYP3A4 inhibition compared to a consideration of ITZ exposure alone.


Subject(s)
Antifungal Agents/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Enzyme Inhibitors/pharmacology , Itraconazole/analogs & derivatives , Itraconazole/pharmacology , Liver/drug effects , Administration, Oral , Adult , Antifungal Agents/administration & dosage , Antifungal Agents/pharmacokinetics , Biotransformation , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Drug Interactions , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacokinetics , Female , Glucuronides/metabolism , Humans , Itraconazole/administration & dosage , Itraconazole/pharmacokinetics , Liver/enzymology , Male , Models, Biological
6.
Epilepsia ; 43(7): 691-6, 2002 Jul.
Article in English | MEDLINE | ID: mdl-12102670

ABSTRACT

PURPOSE: To evaluate the potential pharmacokinetic interactions between topiramate (TPM) and phenytoin (PHT) in patients with epilepsy by studying their pharmacokinetics (PK) after monotherapy and concomitant TPM/PHT treatment. METHODS: Twelve patients with epilepsy stabilized on PHT monotherapy were enrolled in this study, with 10 and seven patients completing the phases with 400 and 800 mg TPM daily doses, respectively. TPM was added at escalating doses, and after stabilization at the highest tolerated TPM dose, PHT doses were tapered. Serial blood and urine samples were collected for PK analysis during the monotherapy phase or the lowest PHT dose after taper and the concomitant TPM/PHT phase. Potential metabolic interaction between PHT and TPM also was studied in vitro in human liver microsomal preparations. RESULTS: In nine of the 12 patients, PHT plasma concentrations remained stable, with a mean (+/-SD) area under the curve (AUC) ratio (combination therapy/monotherapy) of 1.13 +/- 0.17 (range, 0.89-1.23). Three patients had AUC ratios of 1.25, 1.39, and 1.55, respectively, and with the addition of TPM (800, 400, and 400 mg daily, respectively), their peak PHT plasma concentrations increased from 15 to 21 mg/L, 28 to 36 mg/L, and 27 to 41 mg/L, respectively. Human liver microsomal studies with S-mephenytoin showed that TPM partially inhibited CYP2C19 at very high concentrations of 300 microM (11% inhibition) and 900 microM (29% inhibition). Such high plasma concentrations would correspond to doses in humans that are 5 to 15 times higher than the recommended dose (200-400 mg). TPM clearance was approximately twofold higher during concomitant TPM/PHT therapy CONCLUSIONS: This study provides evidence that the addition of TPM to PHT generally does not cause clinically significant PK interaction. PHT induces the metabolism of TPM, causing increased TPM clearance, which may require TPM dose adjustments when PHT therapy is added or is discontinued. TPM may affect PHT concentrations in a few patients because of inhibition by TPM of the CYP2C19-mediated minor metabolic pathway of PHT.


Subject(s)
Anticonvulsants/pharmacokinetics , Aryl Hydrocarbon Hydroxylases , Epilepsy/drug therapy , Fructose/pharmacokinetics , Phenytoin/pharmacokinetics , Adolescent , Adult , Anticonvulsants/therapeutic use , Cytochrome P-450 CYP2C19 , Cytochrome P-450 Enzyme System/drug effects , Cytochrome P-450 Enzyme System/metabolism , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Interactions , Drug Therapy, Combination , Epilepsy/metabolism , Female , Fructose/analogs & derivatives , Fructose/therapeutic use , Humans , Male , Middle Aged , Mixed Function Oxygenases/drug effects , Mixed Function Oxygenases/metabolism , Phenytoin/therapeutic use , Topiramate
7.
Clin Pharmacol Ther ; 70(5): 415-24, 2001 Nov.
Article in English | MEDLINE | ID: mdl-11719727

ABSTRACT

OBJECTIVE: Several reports indicate that fluvoxamine decreases the clearance of cytochrome P4501A2 (CYP1A2) substrates. This study compared in vitro and in vivo inhibition potencies of fluvoxamine toward CYP1A2 with an approach based on inhibition constants (K(i)) determined in vitro and in vivo. METHODS: In vitro inhibition constant values were determined with human liver microsomes and complementary deoxyribonucleic acid-expressed CYP1A2 (supersomes). Fluvoxamine in vivo inhibition constants (K(i)iv) for CYP1A2 were obtained from an investigation of single-dose theophylline (250 mg) disposition in 9 healthy volunteers receiving steady-state (9 days) fluvoxamine at 3 doses (0, 25, or 75 mg/d) in a randomized crossover design. RESULTS: In vitro K(i) values based on total inhibitor concentrations were 177 +/- 56 nmol/L, 121 +/- 21 nmol/L, and 52 +/- 13 nmol/L in human liver microsomes with 1 mg/ml protein and 0.5 mg/ml protein and in supersomes with 0.3 mg/ml protein, respectively. The corresponding in vitro K(i) values based on unbound fluvoxamine concentrations were 35 nmol/L, 36 nmol/L, and 36 nmol/L. The ratio of 1-methyluric acid formation clearances (control/inhibited) in 8 subjects was positively correlated with fluvoxamine concentration (r (2) = 0.87; P <.001) with an intercept near 1. Mean values for K(i)iv based on total and unbound plasma concentrations at steady state were 25.3 nmol/L (range, 14-39 nmol/L) and 3.6 nmol/L (range, 2.4-5.9 nmol/L), respectively. CONCLUSION: Comparison of in vitro and in vivo K(i) values based on unbound fluvoxamine concentrations suggests that fluvoxamine inhibition potency is approximately 10 times greater in vivo than in vitro.


Subject(s)
Cytochrome P-450 CYP1A2 Inhibitors , Enzyme Inhibitors/pharmacology , Fluvoxamine/pharmacology , Theophylline/pharmacokinetics , Adult , Cross-Over Studies , Drug Interactions , Fluvoxamine/metabolism , Humans , In Vitro Techniques
8.
Biochemistry ; 40(43): 12925-31, 2001 Oct 30.
Article in English | MEDLINE | ID: mdl-11669629

ABSTRACT

An extensive body of research on the structural properties of cytochrome P450 enzymes has established that these proteins possess a b-type heme prosthetic group which is noncovalently bound at the active site. Coordinate, electrostatic, and hydrogen bond interactions between the protein backbone and heme functional groups are readily overcome upon mild acid treatment of the enzyme, which releases free heme from the protein. In the present study, we have used a combination of HPLC, LC/ESI-MS, and SDS-PAGE techniques to demonstrate that members of the mammalian CYP4B, CYP4F, and CYP4A subfamilies bind their heme in an unusually tight manner. HPLC chromatography of CYP4B1 on a POROS R2 column under mild acidic conditions caused dissociation of less than one-third of the heme from the protein. Moreover, heme was not substantially removed from CYP4B1 under electrospray or electrophoresis conditions that readily release the prosthetic group from other non-CYP4 P450 isoforms. This was evidenced by an intact protein mass value of 59,217 +/- 3 amu for CYP4B1 (i.e., apoprotein plus heme) and extensive staining of this approximately 60 kDa protein with tetramethylbenzidine/H(2)O(2) following SDS-PAGE. In addition, treatment of CYP4B1, CYP4F3, and CYP4A5/7 with strong base generated a new, chromatographically distinct, polar heme species with a mass of 632.3 amu rather than 616.2 amu. This mass shift is indicative of the incorporation of an oxygen atom into the heme nucleus and is consistent with the presence of a novel covalent ester linkage between the protein backbone of the CYP4 family of mammalian P450s and their heme catalytic center.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/chemistry , Cytochrome P-450 Enzyme System/metabolism , Heme/chemistry , Amino Acid Sequence , Animals , Baculoviridae/metabolism , Benzidines/pharmacology , Binding Sites , Catalytic Domain , Chromatography , Chromatography, High Pressure Liquid , Cytochrome P-450 CYP4A , Cytochrome P450 Family 4 , Electrophoresis, Polyacrylamide Gel , Gas Chromatography-Mass Spectrometry , Heme/metabolism , Histidine/chemistry , Humans , Hydrogen Bonding , Hydrogen Peroxide/pharmacology , Mixed Function Oxygenases/chemistry , Mixed Function Oxygenases/metabolism , Molecular Sequence Data , Protein Binding , Protein Isoforms , Rabbits , Sequence Homology, Amino Acid , Spectrophotometry , Ultraviolet Rays
9.
Pharm Res ; 17(3): 299-305, 2000 Mar.
Article in English | MEDLINE | ID: mdl-10801218

ABSTRACT

PURPOSE: The intestinal metabolism of some CYP3A substrates can be altered profoundly by co-administration of the potent inhibitor, ketoconazole. The present research was conducted to test the hypothesis that, unlike the inhibition kinetics observed with isolated microsomes, inhibition of CYP3A4 by ketoconazole in an intestinal cell monolayer is time-dependent and slowly reversible. METHODS: Confluent, 1alpha,25-dihydroxy Vitamin D3-treated Caco-2 cells were exposed to 1 microM ketoconazole for two hours (Phase I) and then washed three times with culture medium containing no inhibitor. This was followed by a second incubation period (Phase II) that varied in the composition of the apical and basolateral culture medium: Condition 1. apical/basolateral differentiation medium (DM); Condition 2, apical/ basolateral DM + basolateral 2g/dL Human Serum Albumin (HSA); Condition 3, apical/basolateral DM + apical/basolateral 2 g/dL HSA. After various lengths of time for the second phase (0 to 4 hours), both apical and basolateral medium were exchanged with fresh DM. Midazolam (6 microM) was included in the apical medium for determination of CYP3A4 activity (Phase III). RESULTS: Two-way ANOVA of the data revealed persistent inhibition of CYP3A4 under Conditions 1 and 2 (p < 0.001). In contrast, cells treated under Condition 3 exhibited rapid reversal of CYP3A4 inhibition. The level of CYP3A4 activity observed was inversely correlated with the amount of ketoconazole remaining in the cell monolayer at the end of Phase II. CONCLUSIONS: These studies provide mechanistic evidence that ketoconazole can be sequestered into the intestinal mucosa after oral administration, producing a persistent inhibition of first-pass CYP3A4 activity.


Subject(s)
Antifungal Agents/pharmacology , Caco-2 Cells/enzymology , Cytochrome P-450 Enzyme Inhibitors , Ketoconazole/pharmacology , Mixed Function Oxygenases/antagonists & inhibitors , Caco-2 Cells/drug effects , Cytochrome P-450 CYP3A , Enzyme Activation/drug effects , GABA Modulators/pharmacology , Humans , Hydroxylation , Midazolam/pharmacology , Vitamin D/analogs & derivatives , Vitamin D/pharmacology
10.
Drug Metab Dispos ; 27(5): 596-9, 1999 May.
Article in English | MEDLINE | ID: mdl-10220488

ABSTRACT

The purpose of this work was to evaluate the effect of mutual unbound inhibitor and unbound enzyme depletion on the potency of three antifungal cytochrome P-450 (CYP)3A inhibitors with over 1000-fold range in enzyme affinity. Incubations were performed with human liver microsomal protein concentrations that varied from 25 to 1000 microg/ml. The effect of each inhibitor was evaluated using midazolam as a CYP3A probe. Clotrimazole was found to be a tight binding inhibitor of CYP3A with a Ki of 250 pM. Analysis of percent inhibition data by stepwise linear regression for the matrix of inhibitor and enzyme concentrations used showed that protein concentrations predicted the percent inhibition by clotrimazole (r2 = 0.60, p <.001). When clotrimazole concentrations were added to the model, the r2 improved to 0.81, p =.003. Clotrimazole concentrations alone were not a significant predictor of percent inhibition (r2 = 0. 21, p =.08). For ketoconazole, protein concentrations provided a weak prediction of the percent inhibition (r2 = 0.39, p =.006). Conversely, ketoconazole concentrations alone were a good predictor of percent inhibition (r2 = 0.55, p <.001). In contrast to results with clotrimazole and ketoconazole, percent inhibition by fluconazole was not dependent on protein concentrations (r2 = 0.06, p =.39). We conclude that microsomal inhibitory potency can be affected by incubation conditions that deplete the unbound concentration of inhibitor available to the enzyme. This may introduce serious error into a quantitative prediction of an in vivo drug-drug interaction based on an in vitro derived Ki value.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Clotrimazole/metabolism , Clotrimazole/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Cytochrome P-450 Enzyme System/metabolism , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/pharmacology , Oxidoreductases, N-Demethylating/antagonists & inhibitors , Oxidoreductases, N-Demethylating/metabolism , Anti-Infective Agents/metabolism , Anti-Infective Agents/pharmacology , Binding, Competitive , Cytochrome P-450 CYP3A , Humans , Kinetics , Microsomes, Liver/drug effects , Microsomes, Liver/enzymology
11.
J Pharmacol Exp Ther ; 289(2): 1134-42, 1999 May.
Article in English | MEDLINE | ID: mdl-10215697

ABSTRACT

Cytochrome P-450 (CYP) 3A4 accounts for approximately 50% of all P-450s found in the small intestine (Paine et al., 1997) and contributes to the extensive and variable first-pass extraction of drugs such as cyclosporine and saquinavir. We recently demonstrated that CYP3A4 expression in a differentiated Caco-2 subclone is increased when cell monolayers are treated with 1alpha,25-dihydroxy-vitamin-D3 (Schmiedlin-Ren et al., 1997). This improved metabolic capacity permits the in vitro modeling of first-pass intestinal metabolic kinetics. Midazolam (MDZ) 1'-hydroxylation was used as a specific probe for CYP3A-mediated metabolism in modified Caco-2 monolayers. Caco-2 cells were grown to confluence on laminin-coated culture inserts, and then for two additional weeks in the presence of 1alpha,25-dihydroxy vitamin-D3. Cell monolayers were subsequently exposed to MDZ for varying lengths of time and concentrations. The amount of MDZ in the monolayer increased rapidly after apical drug administration, reaching a pseudo steady state within 6 min. The cellular uptake rate was considerably slower after a basolateral dose. By either route of administration, the rate of 1'-hydroxymidazolam formation was stable and linear for 2 h. Under basolateral sink conditions and low apical MDZ dosing concentration (1-8 microM), the first-pass extraction ratio was found to be approximately 15%. Higher dosing concentrations led to saturation of the hydroxylation reaction and reduction in the extraction ratio. The modified Caco-2 cell monolayer is an excellent model for studying drug absorption and first-pass intestinal metabolic kinetic processes. In this system, the selective CYP3A probe MDZ was rapidly absorbed, yet extensively metabolized, as is observed in vivo.


Subject(s)
Calcitriol/pharmacology , Midazolam/metabolism , Biotransformation , Blood Proteins/metabolism , Blotting, Western , Caco-2 Cells , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/metabolism , Electric Conductivity , Extracellular Space/metabolism , Humans , Kinetics , Midazolam/analogs & derivatives , Midazolam/blood , Midazolam/pharmacokinetics , Mixed Function Oxygenases/metabolism , Permeability , Reproducibility of Results
12.
J Pharmacol Exp Ther ; 289(2): 1143-50, 1999 May.
Article in English | MEDLINE | ID: mdl-10215698

ABSTRACT

It has been suggested that the binding of a drug to plasma proteins will influence the intestinal extraction efficiency when drug is delivered to the mucosal epithelium via either the gut lumen or vasculature. We evaluated this hypothesis using cytochrome P-450 (CYP)3A4-expressing Caco-2 monolayers as a model for the intestinal epithelial barrier and midazolam as a CYP3A-specific enzyme probe. The rate of 1'-hydroxylation was measured following apical or basolateral midazolam administration to monolayers incubated in the presence or absence of 4 g/dl of human serum albumin (HSA) in the basolateral compartment medium. The midazolam-free fraction in culture medium containing HSA was 3.3%. Inclusion of HSA in the basolateral medium decreased peak intracellular midazolam accumulation after an apical midazolam dose (3 microM) by 35% and reduced the 1'-hydroxymidazolam formation rate by approximately 20%. Because of the accelerated diffusion of midazolam through the cell monolayer and into the basolateral compartment, there was a 61% reduction in the first-pass metabolic extraction ratio: 13.3 +/- 0. 12% for control versus 5.2 +/- 1% with HSA. Compared with control, addition of HSA resulted in a 91% decrease in the peak intracellular midazolam level and a 86% decrease in the rate of 1'-hydroxylation after the administration of midazolam into basolateral medium. These findings suggest that, in vivo, binding of a drug to plasma proteins will impact both first-pass and systemic intestinal midazolam extraction efficiency. Furthermore, the effect will be more pronounced for a drug that is delivered to mucosal enterocytes by way of arterial blood, compared with oral drug delivery.


Subject(s)
Extracellular Matrix Proteins/metabolism , Midazolam/metabolism , Algorithms , Caco-2 Cells , Electric Conductivity , Extracellular Space/metabolism , Humans , Hydroxylation , Midazolam/analogs & derivatives , Midazolam/pharmacokinetics , Protein Binding , Serum Albumin/metabolism
13.
Drug Metab Dispos ; 27(2): 180-7, 1999 Feb.
Article in English | MEDLINE | ID: mdl-9929500

ABSTRACT

The purpose of this study was to compare the kinetics of intestinal and hepatic cytochrome P-450 3A (CYP3A) inhibition by using microsomal midazolam 1'-hydroxylation as a marker of enzyme activity. The effect of two antifungal agents commonly implicated in CYP3A drug-drug interactions was examined. Inhibition type and affinities were determined for human liver and intestinal microsomes screened for the presence or absence of CYP3A4 and CYP3A5, as well as for cDNA-expressed CYP3A4 and CYP3A5 microsomes. Ketoconazole and fluconazole were found to be noncompetitive inhibitors of both enzymes. Ketoconazole exhibited a Ki for cDNA-expressed CYP3A4 of 26. 7 +/- 1.71 nM, whereas the Ki for cDNA expressed CYP3A5 was 109 +/- 19.7 nM. Corresponding Ki values for fluconazole were 9.21 +/- 0.51 microM and 84.6 +/- 12.9 microM. For liver and intestinal microsomes that contained only CYP3A4, the average ketoconazole Ki was found to be 14.9 +/- 6.7 nM and 17.0 +/- 7.9 nM, respectively, whereas fluconazole yielded mean respective Ki values of 10.7 +/- 4.2 microM and 10.4 +/- 2.9 microM. Liver and intestinal microsomes that contained an equal or greater amount of CYP3A5, in addition to CYP3A4, were less susceptible to inhibition by both ketoconazole and fluconazole. These findings suggest that there can be significant differences in the affinity of these two enzymes for inhibitors. This may further broaden interindividual variability with respect to the magnitude of in vivo drug-drug interactions. We also conclude that there is no significant difference in inhibition type and affinity of ketoconazole and fluconazole for hepatic versus intestinal CYP3A4.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors , Intestine, Small/enzymology , Microsomes, Liver/enzymology , Microsomes/enzymology , Mixed Function Oxygenases/antagonists & inhibitors , Algorithms , Antifungal Agents/pharmacology , Blotting, Western , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/biosynthesis , DNA, Complementary/metabolism , Drug Interactions , Enzyme Inhibitors/pharmacology , Fluconazole/pharmacology , Humans , In Vitro Techniques , Ketoconazole/pharmacology , Kinetics , Midazolam/metabolism , Mixed Function Oxygenases/biosynthesis
14.
J Chromatogr B Biomed Sci Appl ; 708(1-2): 75-85, 1998 Apr 24.
Article in English | MEDLINE | ID: mdl-9653949

ABSTRACT

A method for the quantification of subnanomolar levels of in vitro metabolites of caffeine by an isotope dilution gas chromatographic-mass spectrometric (GC-MS) assay has been developed and applied. Trideuteromethylated analogs of each primary metabolite were synthesized and added after incubations of caffeine with human liver microsomes high in cytochrome P4501A2. HPLC separation of the metabolites prior to GC-MS quantification allowed the isolation of theobromine and paraxanthine which coeluted by GC and enabled quantification over a larger dynamic range. Quantitative analysis was performed on the n-propylated derivatives by selected-ion monitoring of either the M+. ions for the dimethylxanthines or [M-C3H6]+. ions for 1,3,7-trimethyluric acid. For the least abundant metabolite (1,3,7-trimethyluric acid), the detection level on column was 200 pg. Replicate analyses exhibited intra- and inter-day variability of 4.2 and 7.9%, respectively. This assay has been successfully used in the quantification of caffeine's primary metabolites in more than 180 incubations, at varying substrate concentrations and with multiple enzyme sources.


Subject(s)
Caffeine/metabolism , Gas Chromatography-Mass Spectrometry/methods , Chromatography, High Pressure Liquid , Humans , In Vitro Techniques , Microsomes, Liver/metabolism , Sensitivity and Specificity
15.
Biochemistry ; 37(20): 7407-19, 1998 May 19.
Article in English | MEDLINE | ID: mdl-9585555

ABSTRACT

The rapid loss of human CYP1A2 (cytochrome P450 1A2) activity caused by the 8-methylxanthine furafylline is investigated with the aim of determining whether a stable covalent adduct of the xanthine to the enzyme could be identified. Metabolic studies employing expressed CYP1A2 with radiolabeled furafylline and a close analogue, cyclohexylline, where the furan ring is replaced with cyclohexane, indicate that these xanthines are bound in a 1:1 ratio to CYP1A2 protein. This result, combined with earlier kinetic studies, verifies that these compounds are mechanism-based inhibitors of the enzyme. The 8'-methyl carbinols are the only metabolites formed by CYP1A2, and substantial (70-80%) incorporation of oxygen from the medium into the carbinols is observed. Carbinol formation is further characterized by high intramolecular isotope effects (kH/kD > 9) and low intermolecular isotope effects (DV/K < 2). Overall partition ratios are low (5.0 and 7.6, respectively), confirming our previous conclusion that furafylline is an efficient inactivator. By contrast, the N7-methyl-8-methylxanthines are good substrates for CYP1A2 but are not themselves inactivating agents. In addition to other metabolic products, the 8'-methyl carbinols of these N7-methyl-8-methylxanthines are formed in substantial amounts with equally high intramolecular isotope effects; however, the carbinol oxygen is derived exclusively from molecular oxygen. We conclude that oxidation of the 8-methyl group of furafylline and cyclohexylline, but not their N7-methyl analogues, by CYP1A2 promotes a major fraction of the inactivating xanthines to a two electron oxidized intermediate which either terminates enzyme activity by reaction with an active site amino acid or is decomposed by reaction with the medium to give carbinol.


Subject(s)
Cytochrome P-450 CYP1A2 Inhibitors , Imidazoles/metabolism , Theophylline/analogs & derivatives , Cell Line, Transformed , Cytochrome P-450 CYP1A2/metabolism , Deuterium , Electrophoresis, Polyacrylamide Gel , Enzyme Activation/drug effects , Humans , Microsomes, Liver/enzymology , Oxygen/metabolism , Theophylline/metabolism , Theophylline/pharmacology , Xanthines/pharmacology
16.
Clin Pharmacol Ther ; 62(5): 490-504, 1997 Nov.
Article in English | MEDLINE | ID: mdl-9390105

ABSTRACT

OBJECTIVE: The spectrum of cytochrome P450 inhibition of stiripentol, a new anticonvulsant, was characterized in vitro and in vivo. METHODS: Stiripentol was incubated in vitro with (R)-warfarin, coumarin, (S)-warfarin, (S)-mephenytoin, bufuralol, p-nitrophenol, and carbamazepine as probes for CYPs 1A2, 2A6, 2C9, 2C19, 2D6, 2E1, and 3A4, respectively. Caffeine demethylation and the 6 beta-hydroxycortisol/cortisol ratio were monitored in vivo before and after 14 days of treatment with stiripentol as measures of CYP1A2 and CYP3A4 activity, and dextromethorphan O- and N-demethylation were used to measure CYP2D6 and CYP3A4 activity, respectively. In vivo inhibition constants for CYP3A4 were calculated with use of data that previously documented the interaction between stripentol and carbamazepine. RESULTS: In vitro, stiripentol inhibited CYPs 1A2, 2C9, 2C19, 2D6, and 3A4, with inhibition constant values at or slightly higher than therapeutic (total) concentrations of stiripentol, but it did not inhibit CYPs 2A6 and 2E1 even at tenfold therapeutic concentrations. In vivo inhibition of caffeine demethylation and dextromethorphan N-demethylation were consistent with inhibition of CYP1A2 and CYP3A4, respectively. The 6 beta-hydroxycortisol/cortisol ratio did not provide a reliable index of CYP3A4 inhibition. Inhibition of CYP2D6-mediated O-demethylation was not observed in vivo. With use of carbamazepine, in vivo inhibition constants for CYP3A4 ranged between 12 and 35 mumol/L, whereas the corresponding in vitro value was 80 mumol/L. CONCLUSIONS: Stiripentol appears to inhibit several CYP450 enzymes in vitro and in vivo. In vivo inhibition constants show that stiripentol inhibition of CYP3A4 is linearly related to plasma concentration in patients with epilepsy.


Subject(s)
Anticonvulsants/pharmacology , Cytochrome P-450 Enzyme Inhibitors , Dioxolanes/pharmacology , Adult , Anticonvulsants/chemistry , Caffeine , Carbon Dioxide/analysis , Carbon Isotopes , Cytochrome P-450 CYP3A , Dextromethorphan , Dioxolanes/chemistry , Epilepsy/drug therapy , Epilepsy/enzymology , Humans , Hydrocortisone , In Vitro Techniques , Mixed Function Oxygenases/antagonists & inhibitors , Reference Values , Time Factors
17.
Anesthesiology ; 87(1): 36-50, 1997 Jul.
Article in English | MEDLINE | ID: mdl-9232132

ABSTRACT

BACKGROUND: There is considerable unexplained variability in alfentanil pharmacokinetics, particularly systemic clearance. Alfentanil is extensively metabolized in vivo, and thus systemic clearance depends on hepatic biotransformation. Cytochrome P450 3A4 was previously shown to be the predominant P450 isoform responsible for human liver microsomal alfentanil metabolism in vitro. This investigation tested the hypothesis that P450 3A4 is responsible for human alfentanil metabolism and clearance in vivo. METHODS: Nine healthy male volunteers who provided institutionally approved written informed consent were studied in a three-way randomized crossover design. Each subject received alfentanil (20 micrograms/kg given intravenously) 30 min after midazolam (1 mg injected intravenously) on three occasions: control; high P450 3A4 activity (rifampin induction); and low P450 3A4 activity (selective inhibition by troleandomycin). Midazolam is a validated selective in vivo probe for P450 3A4 activity. Venous blood was sampled for 24 h and plasma concentrations of midazolam and alfentanil and their primary metabolites 1'-hydroxymidazolam and noralfentanil were measured by gas chromatography-mass spectrometry. Pharmacokinetic parameters were determined by two-stage analysis using both noncompartmental and three-compartment models. RESULTS: Plasma alfentanil concentration-time profiles depended significantly on P450 3A4 activity. Alfentanil noncompartmental clearance was 5.3 +/- 2.3, 14.6 +/- 3.8, and 1.1 +/- 0.5 ml.kg-1.min-1, and elimination half-life was 58 +/- 13, 35 +/- 7, and 630 +/- 374 min, respectively, in participants with normal (controls), high (rifampin), and low (troleandomycin) P450 3A4 activity (means +/- SD; P < 0.05 compared with controls). Multicompartmental modeling suggested a time-dependent inhibition-resynthesis model for troleandomycin effects on P450 3A4 activity, characterized as k10(t) = k10[1-phi e-alpha(t-tzero)], where k10(t) is the apparent time-dependent rate constant, k10 is the uninhibited rate constant, phi is the fraction of P450 3A4 inhibited, and alpha is the apparent P450 3A4 reactivation rate. Alfentanil clearance was calculated as V1 k10 for controls and men receiving rifampin, and as V1.average k10(t) for men receiving troleandomycin. This clearance was 4.9 +/- 2.1, 13.2 +/- 3.6, and 1.5 +/- 0.8 ml.kg-1.min-1, respectively, in controls and in men receiving rifampin or troleandomycin. There was a significant correlation (r = 0.97, P < 0.001) between alfentanil systemic clearance and P450 3A4 activity. CONCLUSIONS: Modulation of P450 3A4 activity by rifampin and troleandomycin significantly altered alfentanil clearance and disposition. These results strongly suggest that P450 3A4 is the major isoform of P450 responsible for clinical alfentanil metabolism and clearance. This observation, combined with the known population variability in P450 3A4 activity, provides a mechanistic explanation for the interindividual variability in alfentanil disposition. Furthermore, known susceptibility of human P450 3A4 activity to induction and inhibition provides a conceptual framework for understanding and predicting clinical alfentanil drug interactions. Finally, human liver microsomal alfentanil metabolism in vitro is confirmed as an excellent model for human alfentanil metabolism in vivo.


Subject(s)
Alfentanil/pharmacokinetics , Anesthetics, Intravenous/pharmacokinetics , Cytochrome P-450 Enzyme System/metabolism , Mixed Function Oxygenases/metabolism , Adult , Alfentanil/administration & dosage , Cytochrome P-450 CYP3A , Drug Interactions , Humans , Male
18.
J Pharmacol Exp Ther ; 283(3): 1552-62, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9400033

ABSTRACT

Cytochrome P450 3A (CYP3A) metabolizes a diverse array of clinically important drugs. For some of these (e.g., cyclosporine, verapamil, midazolam), CYP3A in the intestinal mucosa contributes to their extensive and variable first-pass extraction. To further characterize this phenomenon, we measured CYP3A content and catalytic activity toward the probe substrate midazolam in mucosa isolated from duodenal, jejunal and ileal sections of 20 human donor intestines. For comparison, the same measurements were performed for 20 human donor livers, eight of which were obtained from the same donors as eight of the intestines. Excellent correlations existed between homogenate and microsomal CYP3A content for the three intestinal regions. Median microsomal CYP3A content was greatest in the duodenum and lowest in the ileum (31 vs. 17 pmol/mg of protein). With respect to midazolam 1'-hydroxylation kinetics, the median Km for each intestinal region was similar to the median hepatic Km, approximately 4 microM. In contrast, the median Vmax decreased from liver to duodenum to jejunum to ileum (850 vs. 644 vs. 426 vs. 68 pmol/min/mg). Intrinsic clearance (Vmax/Km) followed a similar trend for the intestinal regions; median duodenal intrinsic clearance was comparable to hepatic intrinsic clearance (157 and 200 microl/min/mg, respectively). Vmax correlated with CYP3A content for all tissues except the ileum. Duodenal and jejunal Vmax and CYP3A content varied by >30-fold among donors. Microsomes prepared from every other 1-foot section of six intestines were also analyzed for CYP3A as well as for two coenzymes. In general, CYP3A activity, CYP3A content and CYP reductase activity rose slightly from duodenum to middle jejunum and then declined to distal jejunum and ileum. Cytochrome b5 content and cytochrome b5 reductase activity varied little throughout the intestinal tract. Regional intrinsic midazolam 1'-hydroxylation clearance was greatest for the jejunum, followed by the duodenum and ileum (144, 50 and 19 ml/min, respectively). Collectively, these results demonstrate that the upper small intestine serves as the major site for intestinal CYP3A-mediated first-pass metabolism and provides a rationale for interindividual differences in oral bioavailability for some CYP3A substrates.


Subject(s)
Aryl Hydrocarbon Hydroxylases , Cytochrome P-450 Enzyme System/physiology , Intestinal Mucosa/metabolism , Oxidoreductases, N-Demethylating/physiology , Cytochrome P-450 CYP3A , Cytochrome P-450 Enzyme System/analysis , Cytochromes b5/analysis , Humans , Hydroxylation , Liver/metabolism , Metabolic Clearance Rate , Midazolam/metabolism , Oxidoreductases, N-Demethylating/analysis
19.
Ther Drug Monit ; 18(6): 666-71, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8946663

ABSTRACT

To assess the reliability of predicting plasma concentrations of enoxacin, ciprofloxacin, and theophylline from drug concentrations in saliva, six healthy volunteers received single oral doses of enoxacin, ciprofloxacin, and theophylline administered in combination on each of four separate study days, with different, doses separated by at least 5 days. Drug concentrations were determined by a newly developed high-performance liquid chromatography (HPLC) assay, which could measure simultaneously all three drugs in plasma or saliva. Saliva data from the postabsorptive phase after drug administration were used to minimize the effects of variation in absorption. There were good correlations between saliva and plasma concentrations of enoxacin, ciprofloxacin, and theophylline (r = 0.91, 0.88, and 0.98, respectively). The mean (+/-SD) saliva-to-plasma (S/P) ratio for theophylline was 0.63 +/- 0.06 with a coefficient of variation (CV) of 7.9 +/- 2.7%. In contrast, the S/P ratios and CV values for enoxacin and ciprofloxacin were 0.72 +/- 0.21 and 28.9 +/- 11.1%, and 0.58 +/- 0.15 and 25.3 +/- 6.7%, respectively. Because of the large inter- and intraindividual variability, saliva concentrations of enoxacin and ciprofloxacin are not reliable for predicting plasma concentrations. However, saliva may be used reliably for predicting plasma concentrations of theophylline.


Subject(s)
Anti-Infective Agents/pharmacology , Bronchodilator Agents/metabolism , Ciprofloxacin/pharmacology , Enoxacin/pharmacology , Saliva/chemistry , Theophylline/metabolism , Adult , Anti-Infective Agents/blood , Bronchodilator Agents/blood , Ciprofloxacin/blood , Drug Interactions , Enoxacin/blood , Female , Humans , Male , Protein Binding/drug effects , Theophylline/blood
20.
Toxicol Appl Pharmacol ; 141(2): 595-606, 1996 Dec.
Article in English | MEDLINE | ID: mdl-8975785

ABSTRACT

The combined presence of CYP1A2 and 3A4, both of which oxidize aflatoxin B1 (AFB1) to the reactive aflatoxin B1-8,9-epoxide (AFBO) and to hydroxylated inactivation products aflatoxin M1 (AFM1) and aflatoxin Q1 (AFQ1), substantially complicates the kinetic analysis of AFB1 oxidation in human liver microsomes. In the present study, we examine the reaction kinetics of AFB1 oxidation in human liver microsomes (HLMs, N = 3) and in human CYP3A4 and CYP1A2 cDNA-expressed lymphoblastoid microsomes for the purpose of identifying the CYP isoform(s) responsible for AFB1 oxidation at low substrate concentrations approaching those potentially encountered in the diet. AFBO formation by cDNA-expressed human CYP1A2 followed Michaelis-Menten kinetics (Km = 41 microM, Vmax = 2.63 nmol/min/nmol P450). Furthermore, the portion of AFBO formed in HLMs which was eliminated by furafylline, a specific mechanism-based inhibitor of CYP1A2, also followed Michaelis-Menten kinetics (Km = 32-47 microM, Vmax = 0.36-0.69 nmol/min/nmol P450). The formation of AFBO (activation product) and AFQ1 (detoxification product) in cDNA-expressed human CYP3A4 microsomes was sigmoidal and consistent with the kinetics of substrate activation. Accordingly, application of a sigmoid Vmax model equivalent to the Hill equation produced excellent fits to the cDNA-expressed CYP3A4 data and also to the data from HLMs pretreated with furafylline to remove CYP1A2. The Hill model predicted that two substrate binding sites are involved in CYP3A4-mediated AFB1 catalysis and that the average affinity of AFB1 for the two sites was 140-180 microM. Vmax values for AFQ1 formation were 10-fold greater than those for AFBO, and total substrate turnover to both was 67 nmol/min/nmol CYP3A4. Using the derived kinetic parameters for CYP1A2 and 3A4 to model the in vitro rates of AFB activation at low substrate concentrations, it was predicted that CYP1A2 contributes to over 95% of AFB activation in human liver microsomes at 0.1 microM AFB. The important role of CYP1A2 in the in vitro activation of AFB at low substrate concentrations was supported by DNA binding studies. AFB1-DNA binding in control HLMs (reflecting the contribution of CYP1A2 and CYP3A4) and furafylline-pretreated microsomes (reflecting the contribution of CYP3A4 only) catalyzed the binding of 1.71 and 0.085 pmol equivalents of AFB1 to DNA, respectively, indicating that CYP1A2 was responsible for 95% of AFB1-DNA adduct formation at 0.133 microM AFB. These results demonstrate that CYP1A2 dominates the activation of AFB in human liver microsomes in vitro at submicromolar concentrations and support the hypothesis that CYP1A2 is the predominant enzyme responsible for AFBO activation in human liver in vivo at the relatively low dietary concentrations encountered in the human diet, even in high AFB exposure regions of the world. However, because the actual concentrations of AFB in liver in vivo following dietary exposures are uncertain, additional studies in exposed human populations are needed. Quantitative data on the relative rates of AFM1 and AFQ1 excretion (potential biomarkers for CYP1A2 and 3A4 activity, respectively) in humans would be useful to validate the actual contributions of these two enzymes to AFB1 oxidation in vivo.


Subject(s)
Aflatoxin B1/metabolism , Cytochrome P-450 CYP1A2/physiology , Cytochrome P-450 Enzyme System/physiology , Microsomes, Liver/metabolism , Mixed Function Oxygenases/physiology , Adult , Aflatoxins/metabolism , Cytochrome P-450 CYP3A , DNA Adducts/metabolism , Humans , Kinetics , Male , Middle Aged , Oxidation-Reduction , Recombinant Proteins/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL
...