Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 31
Filter
1.
Xenobiotica ; 50(3): 245-251, 2020 Mar.
Article in English | MEDLINE | ID: mdl-31039046

ABSTRACT

Clopidogrel is widely prescribed in patients with cardiovascular disease. Most research has focused on the role of hepatic CYP450 metabolism as the primary source of response variability despite 85-90% of clopidogrel being hydrolyzed by human carboxylesterase-1 (CES1).The purpose of this study is to determine the effects of the known CES1 inhibitor alcohol on clopidogrel metabolism: (1) in vitro in human recombinant CES1 and human liver S9 (HLS9) fractions and (2) in a plasma carboxylesterase deficient mouse (Es1e) strain administered 25 mg/kg oral clopidogrel alone and with 3 g/kg alcohol.Alcohol significantly inhibited the hydrolysis of clopidogrel (IC50 161 mM) and 2-oxo-clopidogrel (IC50 6 mM). In HLS9, alcohol treatment formed ethylated metabolites via transesterification and an increased formation of the H4 active metabolite. These results were replicated in Es1e mice as alcohol increased clopidogrel (91%) and H4 (22%) AUC and reduced formation of the clopidogrel (48%) and 2-oxo-clopidogrel (42%) carboxylate metabolites.Clopidogrel metabolism is highly sensitive to alterations in CES1 activity. The Es1e mouse may represent a suitable model of human CES1 drug metabolism that can be used to rapidly assess how alterations in CES1 function impact the disposition of substrate drugs.


Subject(s)
Carboxylesterase/metabolism , Clopidogrel/metabolism , Animals , Carboxylic Ester Hydrolases , Enzyme Inhibitors , Humans , Inactivation, Metabolic , Liver/metabolism , Mice , Ticlopidine/analogs & derivatives
2.
Br J Clin Pharmacol ; 83(12): 2687-2694, 2017 12.
Article in English | MEDLINE | ID: mdl-28758694

ABSTRACT

AIMS: To determine the disposition and effects of caffeine after administration using a new dosage form (AeroShot) that delivers caffeine by inspiration of a fine powder into the oral cavity and compare it to an equivalent dose of an oral solution (energy drink) as the reference standard. METHODS: Healthy human subjects (n = 17) inspired a 100 mg caffeine dose using the AeroShot device or consumed an energy drink on separate study days. Heart rate, blood pressure and subject assessments of effects were measured over an 8-h period. Plasma concentrations of caffeine and its major metabolites were determined by liquid chromatography-mass spectrometry. Pharmacokinetic, cardiovascular and perceived stimulant effects were compared between AeroShot and energy drink phases using a paired t test and standard bioequivalency analysis. RESULTS: Caffeine disposition was similar after caffeine administration by the AeroShot device and energy drink: peak plasma concentration 1790 and 1939 ng ml-1 , and area under the concentration-time curve (AUC) 15 579 and 17 569 ng ml-1 × h, respectively, but they were not bioequivalent: AeroShot AUC of 80.3% (confidence interval 71.2-104.7%) and peak plasma concentration of 86.3% (confidence interval 62.8-102.8%) compared to the energy drink. Female subjects did have a significantly larger AUC compared to males after consumption of the energy drink. The heart rate and blood pressure were not significantly affected by the 100 mg caffeine dose, and there were no consistently perceived stimulant effects by the subjects using visual analogue scales. CONCLUSION: Inspiration of caffeine as a fine powder using the AeroShot device produces a similar caffeine profile and effects compared to administration of an oral solution (energy drink).


Subject(s)
Caffeine/administration & dosage , Caffeine/pharmacokinetics , Energy Drinks , Administration, Inhalation , Administration, Oral , Adolescent , Adult , Area Under Curve , Blood Pressure/drug effects , Caffeine/adverse effects , Energy Drinks/adverse effects , Female , Heart Rate/drug effects , Humans , Male , Middle Aged , Powders , Sex Factors , Tennessee , Therapeutic Equivalency , Young Adult
3.
Bioanalysis ; 9(5): 469-483, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28176533

ABSTRACT

AIM: Optimal dosing of antibiotics in critically ill patients is complicated by the development of resistant organisms requiring treatment with multiple antibiotics and alterations in systemic exposure due to diseases and extracorporeal drug removal. Developing guidelines for optimal antibiotic dosing is an important therapeutic goal requiring robust analytical methods to simultaneously measure multiple antibiotics. METHODS: An LC-MS/MS assay using protein precipitation for cleanup followed by a 6-min gradient separation was developed to simultaneously determine five antibiotics in human plasma. RESULTS: The precision and accuracy were within the 15% acceptance range. The formic acid concentration was an important determinant of signal intensity, peak shape and matrix effects. CONCLUSION: The method was designed to be simple and successfully applied to a clinical pharmacokinetic study.


Subject(s)
Anti-Bacterial Agents/blood , Formates/chemistry , Tandem Mass Spectrometry , Chromatography, High Pressure Liquid , Critical Illness , Humans , Limit of Detection
4.
Biomed Chromatogr ; 31(6)2017 Jun.
Article in English | MEDLINE | ID: mdl-27864843

ABSTRACT

Caffeine is a mild stimulant with significant potential for abuse, being consumed in larger doses with the widespread availability of energy drinks and by novel routes of administration such as inspired powder, oral sprays and electronic cigarettes. How these recent changes in caffeine consumption affecting caffeine disposition and abuse potential is of growing concern. In the study of caffeine disposition in humans, it is common to only measure the caffeine concentration; however, caffeine's three major metabolites (paraxanthine, theobromine and theophylline) retain central nervous system stimulant activity that may contribute to the overall pharmacological activity and toxicity. Therefore, it would be scientifically more rigorous to measure caffeine and its major metabolites in the evaluation of caffeine disposition in human subjects. Herein, we report a method for the simultaneous quantification of caffeine and its three major metabolites in human plasma by high-performance liquid chromatography coupled to electrospray tandem mass spectrometry (HPLC-ESI-MS/MS). Human plasma samples were treated by simple protein precipitation and the analytes were separated using a 6 min gradient program. Precision and accuracy were well within in the 15% acceptance range. The simple sample preparation, short runtime, sensitivity and the inclusion of caffeine's major metabolites make this assay methodology optimal for the study of caffeine's pharmacokinetics and pharmacodynamics in human subjects.


Subject(s)
Caffeine/blood , Chromatography, High Pressure Liquid/methods , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Humans
5.
Pharmacotherapy ; 36(12): 1229-1237, 2016 12.
Article in English | MEDLINE | ID: mdl-27862103

ABSTRACT

STUDY OBJECTIVE: High-dose continuous venovenous hemofiltration (CVVH) is a continuous renal replacement therapy (CRRT) used frequently in patients with burns. However, antibiotic dosing is based on inference from studies assessing substantially different methods of CRRT. To address this knowledge gap for imipenem/cilastatin (I/C), we evaluated the systemic and extracorporeal clearances (CLs) of I/C in patients with burns undergoing high-dose CVVH. DESIGN: Prospective clinical pharmacokinetic study. PATIENTS: Ten adult patients with burns receiving I/C for a documented infection and requiring high-dose CVVH were studied. METHODS: Blood and effluent samples for analysis of I/C concentrations were collected for up to 6 hours after the I/C infusion for calculation of I/C total CL (CLTotal ), CL by CVVH (CLHF ), half-life during CVVH, volume of distribution at steady state (Vdss ), and the percentage of drug eliminated by CVVH. RESULTS: In this patient sample, the mean age was 50 ± 17 years, total body surface area burns was 23 ± 27%, and 80% were male. Nine patients were treated with high-dose CVVH for acute kidney injury and one patient for sepsis. The mean delivered CVVH dose was 52 ± 14 ml/kg/hour (range 32-74 ml/kg/hr). The imipenem CLHF was 3.27 ± 0.48 L/hour, which accounted for 23 ± 4% of the CLTotal (14.74 ± 4.75 L/hr). Cilastatin CLHF was 1.98 ± 0.56 L/hour, which accounted for 45 ± 19% of the CLTotal (5.16 + 2.44 L/hr). The imipenem and cilastatin half-lives were 1.77 ± 0.38 hours and 4.21 ± 2.31 hours, respectively. Imipenem and cilastatin Vdss were 35.1 ± 10.3 and 32.8 ± 13.8 L, respectively. CONCLUSION: Efficient removal of I/C by high-dose CVVH, a high overall clearance, and a high volume of distribution in burn intensive care unit patients undergoing this CRRT method warrant aggressive dosing to treat serious infections effectively depending on the infection site and/or pathogen.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Burns/drug therapy , Cilastatin/pharmacokinetics , Hemofiltration/methods , Imipenem/pharmacokinetics , Acute Kidney Injury/therapy , Adult , Aged , Anti-Bacterial Agents/administration & dosage , Bacterial Infections/drug therapy , Burns/complications , Burns/pathology , Cilastatin/administration & dosage , Cilastatin, Imipenem Drug Combination , Drug Combinations , Female , Half-Life , Humans , Imipenem/administration & dosage , Intensive Care Units , Male , Middle Aged , Prospective Studies , Tissue Distribution , Young Adult
6.
Eur J Drug Metab Pharmacokinet ; 41(6): 689-703, 2016 Dec.
Article in English | MEDLINE | ID: mdl-26254911

ABSTRACT

BACKGROUND AND OBJECTIVE: Amiodarone (AMD) is one of the most effective drugs for rhythm control of atrial fibrillation. The use of AMD is also associated with adverse effects in multiple tissues. Both the parent compound and its major metabolite desethylamiodarone (DEA) contribute to the drug's therapeutic and toxic action. The present study aimed to build a whole-body physiologically based pharmacokinetic (PBPK) model for AMD and DEA in rats. METHODS: Pharmacokinetic data from multiple studies were collected. Some of the data were pooled together to develop the PBPK model; others were used to evaluate the model. Development of the model also involved in vitro to in vivo extrapolation based on in vitro metabolism data. RESULTS: The final model consisted of 11 tissue compartments, including therapeutic target organs and those to which AMD and DEA may be harmful. Model simulations were in good agreement with the observed time courses of the drug-metabolite pair in tissues, under various dosing scenarios. The key pharmacokinetic properties of AMD, such as extensive tissue distribution, substantial storage in the fat tissue, and long half-lives in many tissues, were closely reflected. CONCLUSION: The developed PBPK model can be regarded as the first step towards a PBPK-pharmacodynamic model that can used to mechanistically evaluate and explain the high adverse event rate and potentially to determine which factors are the primary drives for experiencing an adverse event.


Subject(s)
Amiodarone/analogs & derivatives , Amiodarone/pharmacokinetics , Anti-Arrhythmia Agents/pharmacokinetics , Cytochrome P-450 Enzyme Inhibitors/pharmacokinetics , Ether-A-Go-Go Potassium Channels/antagonists & inhibitors , Models, Biological , Potassium Channel Blockers/pharmacokinetics , Algorithms , Amiodarone/administration & dosage , Amiodarone/adverse effects , Amiodarone/blood , Amiodarone/metabolism , Animals , Anti-Arrhythmia Agents/administration & dosage , Anti-Arrhythmia Agents/adverse effects , Anti-Arrhythmia Agents/metabolism , Biotransformation , Blood-Brain Barrier/metabolism , Cytochrome P-450 Enzyme Inhibitors/administration & dosage , Cytochrome P-450 Enzyme Inhibitors/adverse effects , Cytochrome P-450 Enzyme Inhibitors/metabolism , Dose-Response Relationship, Drug , Ether-A-Go-Go Potassium Channels/metabolism , Half-Life , Infusions, Intravenous , Injections, Intravenous , Male , Metabolic Clearance Rate , Microsomes, Liver/enzymology , Microsomes, Liver/metabolism , Potassium Channel Blockers/administration & dosage , Potassium Channel Blockers/adverse effects , Potassium Channel Blockers/metabolism , Rats , Tissue Distribution
7.
Clin Pharmacokinet ; 54(6): 627-38, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25511794

ABSTRACT

BACKGROUND AND OBJECTIVE: Human carboxylesterase-1 (CES1) and human carboxylesterase-2 (CES2) play an important role in metabolizing many medications. Alcohol is a known inhibitor of these enzymes but the relative effect on CES1 and CES2 is unknown. The aim of this study was to determine the impact of alcohol on the metabolism of specific probes for CES1 (oseltamivir) and CES2 (aspirin). METHODS: The effect of alcohol on CES1- and CES2-mediated probe drug hydrolysis was determined in vitro using recombinant human carboxylesterase. To characterize the in vivo effects of alcohol, healthy volunteers received each probe drug alone and in combination with alcohol followed by blood sample collection and determination of oseltamivir, aspirin, and respective metabolite pharmacokinetics. RESULTS: Alcohol significantly inhibited oseltamivir hydrolysis by CES1 in vitro but did not affect aspirin metabolism by CES2. Alcohol increased the oseltamivir area under the plasma concentration-time curve (AUC) from 0 to 6 h (AUC0 → 6 h) by 27% (range 11-46%, p = 0.011) and decreased the metabolite/oseltamivir AUC0 → 6 h ratio by 34% (range 25-41%, p < 0.001). Aspirin pharmacokinetics were not affected by alcohol. CONCLUSIONS: Alcohol significantly inhibited the hydrolysis of oseltamivir by CES1 both in vitro and in humans, but did not affect the hydrolysis of aspirin to salicylic acid by CES2. These results suggest that alcohol's inhibition of CES1 could potentially result in clinically significant drug interactions with other CES1-substrate drugs, but it is unlikely to significantly affect CES2-substrate drug hydrolysis.


Subject(s)
Antiviral Agents/pharmacokinetics , Carboxylesterase/metabolism , Carboxylic Ester Hydrolases/metabolism , Ethanol/pharmacology , Oseltamivir/pharmacokinetics , Adult , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Antiviral Agents/administration & dosage , Antiviral Agents/blood , Aspirin/administration & dosage , Aspirin/blood , Aspirin/pharmacokinetics , Carboxylesterase/antagonists & inhibitors , Carboxylic Ester Hydrolases/antagonists & inhibitors , Cross-Over Studies , Enzyme Inhibitors/blood , Enzyme Inhibitors/pharmacology , Ethanol/blood , Female , Humans , Hydrolysis/drug effects , Male , Middle Aged , Oseltamivir/administration & dosage , Oseltamivir/blood
8.
PLoS One ; 9(10): e109300, 2014.
Article in English | MEDLINE | ID: mdl-25330096

ABSTRACT

OBJECTIVES: We aim to obtain the intra-subject coefficient of variability of a highly variable antidepressant agomelatine in humans, and propose an adjusted bioequivalence assessment strategy. METHODS: A single-dose, randomized crossover design was conducted in four periods (reference administered thrice, placebo administered once) separated by seven days. A validated LC-MS/MS assay was used to measure drug concentrations in serial blood samples. RESULTS: The intra-subject coefficient of variability was calculated using the residual variance of ANOVA analysis, and the results for Cmax and AUC0-t was 78.34% and 43.52%, respectively, in Chinese healthy subjects. The sample size required for standard BE study were 124(192, 340) if the expected deviation between the reference and generic products was set to 0 (5%, 10%). CONCLUSIONS: Agomelatine meets the criteria for highly variable drug in Chinese healthy male subjects, and the traditional BE criteria for agomelatine needs to be adjusted to alleviate the resource and ethical burden of using a large numbers of subjects in clinical trials. Our clinical data on the intra-subject variability of agomelatine PK in Chinese healthy population enables to adjust bioequivalence (BE) assessment approach for agomelatine based on the RSABE approaches recommended by regulatory agencies. TRIAL REGISTRATION: ChiCTR.org ChiCTR-TTRCC-13003835.


Subject(s)
Acetamides/pharmacokinetics , Asian People , Healthy Volunteers , Adolescent , Adult , Dose-Response Relationship, Drug , Humans , Male , Reference Standards , Therapeutic Equivalency , Time Factors , Young Adult
9.
Clin Pharmacokinet ; 53(9): 825-36, 2014 Sep.
Article in English | MEDLINE | ID: mdl-25103325

ABSTRACT

BACKGROUND AND OBJECTIVE: Human carboxylesterase-1 (CES1) is an enzyme that is primarily expressed in the liver, where it plays an important role in the metabolism of many commonly used medications. Ethanol (alcohol)-mediated inhibition of CES1 and loss-of-function polymorphisms in the CES1 gene can markedly reduce this enzyme's function. Such alterations in CES1 activity may have important effects on the disposition of substrate drugs. The aim of this study is to develop a physiologically based pharmacokinetic (PBPK) model to predict changes in CES1 substrate drug exposure in humans with CES1 activity impaired by ethanol or loss-of-function CES1 genetic polymorphisms. METHODS: The antiviral drug oseltamivir, an ethyl ester prodrug that is rapidly converted in vivo to the active metabolite oseltamivir carboxylate (OSC) by CES1 was used as a probe drug for CES1 activity. Oseltamivir PBPK models integrating in vitro and in vivo data were developed and refined. Then the changes in oseltamivir and OSC exposure in humans with CES1 impaired by ethanol or polymorphisms were simulated using a PBPK model incorporating in vitro inhibition and enzyme kinetic data. Model assumptions were verified by comparison of simulations with observed and published data. A sensitivity analysis was performed to gain a mechanistic understanding of the exposure changes of oseltamivir and OSC. RESULTS: The simulated changes in oseltamivir and OSC exposures in humans with CES1 impaired by ethanol or polymorphism were similar to the observed data. The observed exposures to oseltamivir were increased by 46 and 37 % for the area under the plasma concentration-time curve from time zero to 6 h (AUC6) and from time zero to 24 h (AUC24), respectively, with co-administration of ethanol 0.6 g/kg. In contrast, only a slight change was observed in OSC exposure. The simulated data show the same trend as evidenced by greater change in exposures to oseltamivir (27 and 26 % for AUC(6) and AUC(24), [corrected] respectively) than OSC (≤6 %). CONCLUSIONS: The PBPK model of impaired CES1 activity correctly predicts observed human data. This model can be extended to predict the effects of drug interactions and other factors affecting the pharmacokinetics of other CES1 substrate drugs.


Subject(s)
Antiviral Agents/pharmacokinetics , Carboxylic Ester Hydrolases/metabolism , Models, Biological , Oseltamivir/pharmacokinetics , Adult , Antiviral Agents/blood , Carboxylic Ester Hydrolases/genetics , Computer Simulation , Humans , Male , Oseltamivir/analogs & derivatives , Oseltamivir/blood , Polymorphism, Genetic
10.
Rapid Commun Mass Spectrom ; 28(11): 1285-92, 2014 Jun 15.
Article in English | MEDLINE | ID: mdl-24760569

ABSTRACT

RATIONALE: Clopidogrel (CLO) is a prodrug used to prevent ischemic events in patients undergoing percutaneous coronary intervention or with myocardial infarction. A previous study found ethyl clopidogrel (ECLO) is formed by transesterification of CLO when incubated with alcohol in human liver microsomes. We hypothesize that ECLO will be subject to further metabolism and developed an assay to identify its metabolites. METHODS: A liquid chromatography/triple quadrupole mass spectrometry (LC/MS/MS) method was developed to identify metabolites of ECLO. According to the predicted metabolic pathway of ECLO, precursor-product ion pairs were used to screen the possible metabolites of ECLO in human liver S9 fractions. Subsequently, the detected metabolites were characterized by the results of product ion scan. RESULTS: In the presence of alcohol, CLO was tranesterified to ECLO, which was further oxidized to form ethylated 2-oxo-clopidogrel and several ethylated thiol metabolites including the ethylated form of the H4 active metabolite. CONCLUSIONS: The ECLO formed by transesterification with alcohol is subject to metabolism by CYP450 enzymes producing ethylated forms of 2-oxo-clopidogrel and the active H4 thiol metabolite.


Subject(s)
Alcohols/metabolism , Chromatography, High Pressure Liquid/methods , Mass Spectrometry/methods , Ticlopidine/analogs & derivatives , Alcohols/analysis , Clopidogrel , Esterification , Humans , Microsomes, Liver/chemistry , Microsomes, Liver/metabolism , Molecular Structure , Ticlopidine/chemistry , Ticlopidine/metabolism
11.
Br J Pharmacol ; 171(11): 2778-89, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24471734

ABSTRACT

BACKGROUND AND PURPOSE: Alvespimycin, a new generation of heat shock protein 90 (Hsp90) inhibitor in clinical trial, is a promising therapeutic agent for cancer. Pharmacokinetic models of alvespimycin would help in the understanding of drug disposition, predicting drug exposure and interpreting dose-response relationship. In the present study we aimed to develop a physiologically based pharmacokinetic (PBPK) model of alvespimycin in mice and evaluate the utility of the model for predicting alvespimycin disposition in other species. EXPERIMENTAL APPROACH: A literature search was performed to collect pharmacokinetic data for alvespimycin. A PBPK model was initially constructed to demonstrate the disposition of alvespimycin in mice, and then extrapolated to rats and humans by taking into account the interspecies differences in physiological- and chemical-specific parameters. KEY RESULTS: A PBPK model, employing a permeability-limited model structure and saturable tissue binding, was built in mice. It successfully characterized the time course of the disposition of alvespimycin in mice. After extrapolation to rats, the model simulated the alvespimycin concentration-time profiles in rat tissues with acceptable accuracies. Likewise, a reasonable match was found between the observed and simulated human plasma pharmacokinetics of alvespimycin. CONCLUSIONS AND IMPLICATIONS: The PBPK model described here is beneficial to the understanding and prediction of the effects of alvespimycin in different species. It also provides a good basis for further development, which necessitates additional studies, especially those needed to clarify the in-depth mechanism of alvespimycin elimination. A refined PBPK model would benefit the understanding of dose-response relationships and optimization of dosing regimens.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Benzoquinones/pharmacokinetics , Lactams, Macrocyclic/pharmacokinetics , Models, Biological , Animals , Antineoplastic Agents/blood , Benzoquinones/blood , Humans , Lactams, Macrocyclic/blood , Mice , Rats , Tissue Distribution
12.
Drug Metab Dispos ; 42(2): 201-6, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24212379

ABSTRACT

Dabigatran etexilate (DABE) is an oral prodrug that is rapidly converted to the active thrombin inhibitor, dabigatran (DAB), by serine esterases. The aims of the present study were to investigate the in vitro kinetics and pathway of DABE hydrolysis by human carboxylesterase enzymes, and the effect of alcohol on these transformations. The kinetics of DABE hydrolysis in two human recombinant carboxylesterase enzymes (CES1 and CES2) and in human intestinal microsomes and human liver S9 fractions were determined. The effects of alcohol (a known CES1 inhibitor) on the formation of DABE metabolites in carboxylesterase enzymes and human liver S9 fractions were also examined. The inhibitory effect of bis(4-nitrophenyl) phosphate on the carboxylesterase-mediated metabolism of DABE and the effect of alcohol on the hydrolysis of a classic carboxylesterase substrate (cocaine) were studied to validate the in vitro model. The ethyl ester of DABE was hydrolyzed exclusively by CES1 to M1 (Km 24.9 ± 2.9 µM, Vmax 676 ± 26 pmol/min per milligram protein) and the carbamate ester of DABE was exclusively hydrolyzed by CES2 to M2 (Km 5.5 ± 0.8 µM; Vmax 71.1 ± 2.4 pmol/min per milligram protein). Sequential hydrolysis of DABE in human intestinal microsomes followed by hydrolysis in human liver S9 fractions resulted in complete conversion to DAB. These results suggest that after oral administration of DABE to humans, DABE is hydrolyzed by intestinal CES2 to the intermediate M2 metabolite followed by hydrolysis of M2 to DAB in the liver by CES1. Carboxylesterase-mediated hydrolysis of DABE was not inhibited by alcohol.


Subject(s)
Antithrombins/metabolism , Benzimidazoles/metabolism , Carboxylesterase/metabolism , Intestines/enzymology , Liver/enzymology , Prodrugs/metabolism , Pyridines/metabolism , Administration, Oral , Antithrombins/administration & dosage , Benzimidazoles/administration & dosage , Biotransformation , Carboxylesterase/antagonists & inhibitors , Carboxylic Ester Hydrolases/antagonists & inhibitors , Carboxylic Ester Hydrolases/metabolism , Cocaine/metabolism , Dabigatran , Enzyme Inhibitors/pharmacology , Humans , Hydrolysis , Intestines/drug effects , Kinetics , Liver/drug effects , Microsomes, Liver/enzymology , Prodrugs/administration & dosage , Pyridines/administration & dosage , Recombinant Proteins/metabolism , Substrate Specificity
13.
Curr Drug Metab ; 15(8): 761-6, 2014.
Article in English | MEDLINE | ID: mdl-25705907

ABSTRACT

Drug-drug interaction (DDI) is one important topic in drug discovery, drug development and clinical practice. Recently, a novel approach, in vivo information-guided prediction (IVIP), was introduced for predicting the magnitude of pharmacokinetic DDIs which are caused by changes in cytochrome P450 (CYP) activity. This approach utilizes two parameters, i.e. CR (the apparent contribution of the target metabolizing enzyme to the clearance of the substrate drug) and IX (the apparent effect of a perpetrator on the target CYP) to describe the magnitude of DDI between a perpetrator and a victim drug. The essential concept of this method assumes that at a given dose level, the IX for a given perpetrator remains constant whatever the victim drug is. Usually, this IVIP method is only based on information from clinical studies and does not need in vitro information. In this review, basic concept, application and extension, as well as pros and cons of the IVIP method were presented. How to apply this approach was also discussed. Thus far, this method displayed good performance in predicting DDIs associated with CYPs, and can be used to forecast the magnitude of a large number of possible DDIs, of which only a small portion have been investigated in clinical studies. The key concept of this static approach could even be implemented in dynamic modeling to assess risks of DDIs involving drug transporters.


Subject(s)
Drug Interactions , Models, Biological , Pharmacokinetics , Computer Simulation , Dose-Response Relationship, Drug , Drug Design , Humans , Pharmaceutical Preparations/administration & dosage , Pharmaceutical Preparations/metabolism
15.
Br J Pharmacol ; 171(4): 1043-53, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24283665

ABSTRACT

BACKGROUND AND PURPOSE: In vitro inhibitory potency (Ki )-based predictions of P-glycoprotein (P-gp)-mediated drug-drug interactions (DDIs) are hampered by the substantial variability in inhibitory potency. In this study, in vivo-based [I]/Ki values were used to predict the DDI risks of a P-gp substrate dabigatran etexilate (DABE) using physiologically based pharmacokinetic (PBPK) modelling. EXPERIMENTAL APPROACH: A baseline PBPK model was established with digoxin, a known P-gp substrate. The Km (P-gp transport) of digoxin in the baseline PBPK model was adjusted to Km (i) to fit the change of digoxin pharmacokinetics in the presence of a P-gp inhibitor. Then 'in vivo' [I]/Ki of this P-gp inhibitor was calculated using Km (i) /Km . Baseline PBPK model was developed for DABE, and the 'in vivo' [I]/Ki was incorporated into this model to simulate the static effect of P-gp inhibitor on DABE pharmacokinetics. This approach was verified by comparing the observed and the simulated DABE pharmacokinetics in the presence of five different P-gp inhibitors. KEY RESULTS: This approach accurately predicted the effects of five P-gp inhibitors on DABE pharmacokinetics (98-133% and 89-104% for the ratios of AUC and Cmax respectively). The effects of 16 other P-gp inhibitors on the pharmacokinetics of DABE were also confidently simulated. CONCLUSIONS AND IMPLICATIONS: 'In vivo' [I]/Ki and PBPK modelling, used in combination, can accurately predict P-gp-mediated DDIs. The described framework provides a mechanistic basis for the proper design of clinical DDI studies, as well as avoiding unnecessary clinical DDI studies.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Benzimidazoles/pharmacokinetics , Models, Biological , Pyridines/pharmacokinetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/antagonists & inhibitors , Amiodarone/analogs & derivatives , Amiodarone/pharmacology , Benzimidazoles/blood , Clarithromycin/pharmacology , Dabigatran , Digoxin/blood , Digoxin/pharmacokinetics , Dronedarone , Drug Interactions , Humans , Pyridines/blood , Quinidine/pharmacology , Verapamil/pharmacology , beta-Alanine/analogs & derivatives , beta-Alanine/blood
16.
Acta Pharmacol Sin ; 34(11): 1437-48, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24056706

ABSTRACT

AIM: To investigate the pharmacokinetics and disposition of simmitecan (L-P) that was a water-soluble ester prodrug of chimmitecan (L-2-Z) with potent anti-tumor activities in different experimental animals, and to assess its drug-drug interaction potential. METHODS: SD rats were injected with a single iv bolus doses of L-P (3.75, 7.5 and 15 mg/kg). The pharmacokinetics, tissue distribution, excretion and metabolism of L-P and its active metabolite L-2-Z were studied through quantitative measurements and metabolite profiling with LC/MS. The binding of L-P and L-2-Z to rat plasma proteins was examined using an ultrafiltration method. Systemic exposures of beagle dogs to L-P as well as drug distribution in tumors of the nude mice xenograft model of human hepatic cancer SMMC-7721 cells were also examined. The metabolism of L-P by liver mcirosomal carboxylesterase in vitro was investigated in different species. The effects of L-P and L-2-Z on cytochrome P450 enzymes were examined using commercial screening kits. RESULTS: The in vivo biotransformation of L-P to L-2-Z showed a significant species difference, with a mean elimination half-life t1/2 of approximately 1.4 h in rats and 1.9 h in dogs. The systemic exposure levels of L-P and L-2-Z were increased in a dose-dependent manner. In rats, approximately 66% of L-P and 79% of L-2-Z were bound to plasma proteins. In rats and the nude mice bearing human hepatic cancers, most organ tissues had significantly higher concentrations of L-P than the corresponding plasma levels. In the tumor tissues, the L-P levels were comparable to those of plasma, whereas the L-2-Z levels were lower than the L-P levels. In rats, L-P was eliminated mainly via biliary excretion, but metabolism played an important role in elimination of the intact L-P. Finally, L-P and L-2-Z exerted moderate inhibition on the activity of CYP3A4 in vitro. CONCLUSION: L-P and L-2-Z have relatively short elimination half-lives and L-P is mainly eliminated via biliary excretion. The species difference in the conversion of L-P to L-2-Z and potential drug-drug interactions due to inhibition of CYP3A4 should be considered in further studies.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Camptothecin/analogs & derivatives , Enzyme Inhibitors/pharmacokinetics , Animals , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/pharmacology , Camptothecin/administration & dosage , Camptothecin/pharmacokinetics , Camptothecin/pharmacology , Carboxylesterase/metabolism , Cytochrome P-450 CYP3A , Cytochrome P-450 CYP3A Inhibitors , Dogs , Dose-Response Relationship, Drug , Drug Interactions , Enzyme Inhibitors/administration & dosage , Enzyme Inhibitors/pharmacology , Female , Half-Life , Humans , Injections, Intravenous , Liver Neoplasms, Experimental/metabolism , Male , Mice , Mice, Nude , Microsomes, Liver/metabolism , Prodrugs , Rats , Rats, Sprague-Dawley , Species Specificity , Tissue Distribution , Xenograft Model Antitumor Assays
17.
J Mass Spectrom ; 48(8): 945-50, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23893642

ABSTRACT

A hydrophilic interaction chromatography/mass spectrometry (HILIC-MS)-based assay for imipenem (IMP) and cilastatin (CIL) was recently reported. This orthogonal electrospray ion source-based (ORS) assay utilized nonvolatile salt (unremovable) to stabilize IMI in plasma. Unfortunately, this method was not applicable to conventional MS with off-axis spray (OAS-MS) because MS sensitivity was rapidly deteriorated by the nonvolatile salt. Therefore, we aimed to find a nonvolatile salt- and ion suppression-free approach to stabilize and measure the analytes in plasma using OAS-MS. Acetonitrile and methanol were tested to stabilize the analytes in the plasma samples. The recoveries, matrix effects and stabilities of the analytes in the stabilizer-treated samples were studied. The variations in MS signal intensities were used as the indicator of the assay ruggedness. The results show that a mixture of methanol and acetonitrile (1:1) is best for the storage and measurement of IMP and CIL in human plasma. Utilization of this precipitant not only blocked the hydrolysis of the analytes in plasma but also resulted in an ion suppression-free, fast (120 s per sample) and sensitive detection. The sensitivity obtained using the less sensitive OAS-MS (API3000, 4 pg on column) is much greater than that of the published ORS-MS-based assay (API4000, 77 pg on column). The ruggedness of the assay was demonstrated by its constant MS signal intensity. In conclusion, an improved HILIC/MS-based assay for IMP and CIL was established. The approach presented here provides a simple solution to the challenge of analyzing hydrolytically unstable ß-lactam antibiotics in biological samples.


Subject(s)
Chromatography, Liquid/methods , Cilastatin/blood , Imipenem/blood , Spectrometry, Mass, Electrospray Ionization/methods , Acetonitriles/chemistry , Cilastatin/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Imipenem/chemistry , Linear Models , Methanol , Oseltamivir/analogs & derivatives , Oseltamivir/blood , Oseltamivir/chemistry , Reproducibility of Results , Sensitivity and Specificity
18.
Clin Pharmacokinet ; 52(6): 433-41, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23494981

ABSTRACT

BACKGROUND AND OBJECTIVE: Organic anion-transporting polypeptide (OATP)-mediated drug-drug interactions (DDIs) are among the most important classes of clinically relevant DDIs. Accurate prediction of the OATP-mediated DDIs is not successful due to the sequential disposition pathways of OATP substrates in humans. Intestinal and hepatic uptake transporters, efflux transporters, and cytochrome P450 (CYP) enzymes are often involved in the sequential disposition pathways of typical OATP substrates. The aim of this proof-of-concept study is to develop and validate a novel approach which can be used to predict OATP-mediated DDIs with significantly increased accuracy and decreased false-negatives. METHODS: The feasibility of using a disposition pathway-dependent prediction (DPDP) approach to predict the ratios of the area under the plasma concentration-time curve (AUC(R)) in the presence and absence of the inhibitor was investigated. A total of 62 clinical DDI studies were included in this feasibility study. The disposition pathways governing the outcome of DDIs were first identified for each substrate using the information within learning sets, and then substrate-specific algorithms were used to predict the DDI risks of the external validation set (51 DDIs). RESULTS: The method predicted AUC(R) within 50-200 % for 50 studies (98 %), and the false-negative rate was 9.8 %. The DPDP approach showed significant improvement over an existing approach and was used to forecast the magnitude of 198 DDIs that have not been studied. CONCLUSION: This approach can be used to avoid unnecessary clinical DDI studies during new drug development.


Subject(s)
Drug Interactions , Models, Biological , Organic Anion Transporters/metabolism , Pharmaceutical Preparations/metabolism , Tissue Distribution , Drug-Related Side Effects and Adverse Reactions , Forecasting , Humans , Pharmaceutical Preparations/blood , Substrate Specificity
19.
J Pharm Biomed Anal ; 72: 245-50, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23000242

ABSTRACT

Although liquid chromatography/electrospray ionization tandem mass spectrometry-based assays have been reported for the measurement of the antiviral oseltamivir (OS) in human samples, these assays either involve complicated sample pretreatment or lack sensitivity. Here we introduce a straightforward approach to improve the assay performance for OS and its metabolite oseltamivir carboxylate (OSC) in human plasma. A very low concentration of mobile phase modifier can improve the ionization efficiency of both analytes, thus enabling a high sensitivity without any matrix effect. The fast LC gradient further increases the sensitivity by narrowing the peak width (6-9s) and eluting the analytes at higher organic content. The increased ionization efficiency and minimized matrix effects enabled us to introduce a one-step protein precipitation for sample clean-up without compromising the sensitivity. The lower limit of quantification was 0.34 ng/mL for both analytes, which was at least 3 times more sensitive than published assays that involve complicated sample pretreatment. The assay involves measurement of analytes and their stable-isotope internal standards in small-volume (30-µL) plasma. Sodium fluoride was utilized to prevent the hydrolysis of OS during and after sampling. The calibration curve was linear over the range of 0.34-1000 ng/mL. Accuracy was 95-110% and the precision was 2.2-11.0%. This method was applied successfully to the human pharmacokinetic study of OS, and can estimate the relevant pharmacokinetic parameters of OS with more accuracy. The approach utilized in the optimization of assay performance can be extended to the measurement of other drugs in biomatrices.


Subject(s)
Oseltamivir/analogs & derivatives , Oseltamivir/blood , Oseltamivir/chemistry , Chromatography, High Pressure Liquid/methods , Humans , Oseltamivir/metabolism , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods
20.
Anal Bioanal Chem ; 405(5): 1695-704, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23239178

ABSTRACT

Dabigatran etexilate (DABE) is an oral prodrug that is rapidly converted by esterases to dabigatran (DAB), a direct inhibitor of thrombin. To elucidate the esterase-mediated metabolic pathway of DABE, a high-performance liquid chromatography/mass spectrometry based metabolite identification and semi-quantitative estimation approach was developed. To overcome the poor full-scan sensitivity of conventional triple quadrupole mass spectrometry, precursor-product ion pairs were predicted to search for the potential in vitro metabolites. The detected metabolites were confirmed by the product ion scan. A dilution method was introduced to evaluate the matrix effects on tentatively identified metabolites without chemical standards. Quantitative information on detected metabolites was obtained using "metabolite standards" generated from incubation samples that contain a high concentration of metabolite in combination with a correction factor for mass spectrometry response. Two in vitro metabolites of DABE (M1 and M2) were identified, and quantified by the semi-quantitative estimation approach. It is noteworthy that CES1 converts DABE to M1 while CES2 mediates the conversion of DABE to M2. M1 and M2 were further metabolized to DAB by CES2 and CES1, respectively. The approach presented here provides a solution to a bioanalytical need for fast identification and semi-quantitative estimation of CES metabolites in preclinical samples.


Subject(s)
Benzimidazoles/metabolism , Carboxylesterase/metabolism , Prodrugs/metabolism , Pyridines/metabolism , Tandem Mass Spectrometry/methods , Animals , Antithrombins/metabolism , Chromatography, Liquid/methods , Dabigatran , Humans , Plasma/metabolism , Rats , beta-Alanine/analogs & derivatives , beta-Alanine/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...