Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Nanomedicine (Lond) ; 18(24): 1697-1701, 2023 Oct.
Article in English | MEDLINE | ID: mdl-37877695

ABSTRACT

Tweetable abstract Layered double hydroxide nanocarriers are capable of intercalating hydrophobic NSAIDs, such as mefenamic acid, which improves their pharmacokinetics and bioavailability.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Mefenamic Acid , Mefenamic Acid/pharmacokinetics , Hydroxides/chemistry , Biological Availability
2.
CPT Pharmacometrics Syst Pharmacol ; 10(2): 108-118, 2021 02.
Article in English | MEDLINE | ID: mdl-33439535

ABSTRACT

This analysis reports a quantitative modeling and simulation approach for oral dapagliflozin, a primarily uridine diphosphate-glucuronosyltransferase (UGT)-metabolized human sodium-glucose cotransporter 2 selective inhibitor. A mechanistic dapagliflozin physiologically based pharmacokinetic (PBPK) model was developed using in vitro metabolism and clinical pharmacokinetic (PK) data and verified for context of use (e.g., exposure predictions in pediatric subjects aged 1 month to 18 years). Dapagliflozin exposure is challenging to predict in pediatric populations owing to differences in UGT1A9 ontogeny maturation and paucity of clinical PK data in younger age groups. Based on the exposure-response relationship of dapagliflozin, twofold acceptance criteria were applied between model-predicted and observed drug exposures and PK parameters (area under the curve and maximum drug concentration) in various scenarios, including monotherapy in healthy adults (single/multiple dose), monotherapy in hepatically or renally impaired patients, and drug-drug interactions with UGT1A9 modulators, such as mefenamic acid and rifampin. The PBPK model captured the observed exposure within twofold of the observed monotherapy data in adults and adolescents and in special population. As a guide to determining dosing regimens in pediatric studies, the verified PBPK model, along with UGT enzyme ontogeny maturation understanding, was used for predictions of dapagliflozin monotherapy exposures in pediatric subjects aged 1 month to 18 years that best matched exposure in adult patients with a 10-mg single dose of dapagliflozin.


Subject(s)
Benzhydryl Compounds/pharmacokinetics , Glucosides/pharmacokinetics , Glucuronosyltransferase/metabolism , Mefenamic Acid/pharmacokinetics , Rifampin/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics , UDP-Glucuronosyltransferase 1A9/metabolism , Administration, Oral , Adolescent , Antibiotics, Antitubercular/administration & dosage , Antibiotics, Antitubercular/adverse effects , Antibiotics, Antitubercular/pharmacokinetics , Area Under Curve , Child , Child, Preschool , Computer Simulation , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/adverse effects , Cyclooxygenase Inhibitors/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Female , Healthy Volunteers/statistics & numerical data , Hepatic Insufficiency/drug therapy , Humans , Infant , Infant, Newborn , Male , Mefenamic Acid/administration & dosage , Mefenamic Acid/adverse effects , Models, Biological , Predictive Value of Tests , Renal Insufficiency/drug therapy , Rifampin/administration & dosage , Rifampin/adverse effects
3.
CPT Pharmacometrics Syst Pharmacol ; 10(2): 127-136, 2021 02.
Article in English | MEDLINE | ID: mdl-33314761

ABSTRACT

The sodium-glucose cotransporter 2 inhibitor ertugliflozin is metabolized by the uridine 5'-diphospho-glucuronosyltransferase (UGT) isozymes UGT1A9 and UGT2B4/2B7. This analysis evaluated the drug-drug interaction (DDI) following co-administration of ertugliflozin with the UGT inhibitor mefenamic acid (MFA) using physiologically-based pharmacokinetic (PBPK) modeling. The ertugliflozin modeling assumptions and parameters were verified using clinical data from single-dose and multiple-dose studies of ertugliflozin in healthy volunteers, and the PBPK fraction metabolized assignments were consistent with human absorption, distribution, metabolism, and excretion results. The model for MFA was developed using clinical data, and in vivo UGT inhibitory constant values were estimated using the results from a clinical DDI study with MFA and dapagliflozin, a UGT1A9 and UGT2B4/2B7 substrate in the same chemical class as ertugliflozin. Using the verified compound files, PBPK modeling predicted an ertugliflozin ratio of area under the plasma concentration-time curves (AUCR ) of 1.51 when co-administered with MFA. ClinicalTrials.gov identifier: NCT00989079.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Cyclooxygenase Inhibitors/pharmacokinetics , Glucuronosyltransferase/metabolism , Mefenamic Acid/pharmacokinetics , Sodium-Glucose Transporter 2 Inhibitors/pharmacokinetics , Adult , Area Under Curve , Bridged Bicyclo Compounds, Heterocyclic/administration & dosage , Bridged Bicyclo Compounds, Heterocyclic/metabolism , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/metabolism , Drug Interactions , Female , Healthy Volunteers , Humans , Male , Mefenamic Acid/administration & dosage , Mefenamic Acid/metabolism , Middle Aged , Models, Biological , Sodium-Glucose Transporter 2 Inhibitors/administration & dosage , Sodium-Glucose Transporter 2 Inhibitors/metabolism , UDP-Glucuronosyltransferase 1A9 , Uridine/metabolism
4.
J Chromatogr B Analyt Technol Biomed Life Sci ; 1122-1123: 49-57, 2019 Aug 01.
Article in English | MEDLINE | ID: mdl-31153131

ABSTRACT

Ertugliflozin (ERTU) is a novel, potent, and highly selective sodium glucose cotransporter 2 inhibitor that has been recently approved for the treatment of type 2 diabetes mellitus. We describe a novel bioanalytical method using high-performance liquid chromatography (HPLC) coupled with fluorescence detection for quantitative determination of ERTU in rat plasma. Acetonitrile-based protein precipitation method was used for sample preparation, and chromatographic separation was performed on a Kinetex® C18 column with an isocratic mobile phase comprising acetonitrile and 10 mM potassium phosphate buffer (pH 6.0). The eluent was monitored by a fluorescence detector at an optimized excitation/emission wavelength pair of 277/320 nm. The method was validated to demonstrate the selectivity, linearity (ranging from 4 to 2000 ng/mL), precision, accuracy, recovery, matrix effect, and stability in line with the current FDA guidelines. The newly developed method was successfully applied to investigate the pharmacokinetic interactions of ERTU with mefenamic acid (MEF) and ketoconazole (KET). The findings of the present study revealed that the pharmacokinetics of ERTU may be altered by concurrent administration of MEF and KET in rats. To our knowledge, the present study is the first to develop a validated bioanalytical method for quantification of ERTU using HPLC coupled with fluorescence detection and to assess the drug interaction potential of ERTU with non-steroidal anti-inflammatory (MEF) and azole antifungal (KET) drugs.


Subject(s)
Bridged Bicyclo Compounds, Heterocyclic/blood , Bridged Bicyclo Compounds, Heterocyclic/pharmacokinetics , Chromatography, High Pressure Liquid/methods , Ketoconazole/pharmacokinetics , Mefenamic Acid/pharmacokinetics , Spectrometry, Fluorescence/methods , Animals , Bridged Bicyclo Compounds, Heterocyclic/chemistry , Drug Interactions , Ketoconazole/blood , Ketoconazole/chemistry , Limit of Detection , Linear Models , Male , Mefenamic Acid/blood , Mefenamic Acid/chemistry , Rats , Rats, Sprague-Dawley , Reproducibility of Results
5.
Mol Pharm ; 16(7): 2884-2891, 2019 07 01.
Article in English | MEDLINE | ID: mdl-31120762

ABSTRACT

The purpose of this work was to investigate the use of the dimethylaminoethyl methacrylate-copolymer Eudragit EPO (EPO) in oral solubility-enabling formulations for anionic lipophilic drugs, aiming to guide optional formulation design and maximize oral bioavailability. We have studied the solubility, the permeability, and their interplay, using the low-solubility nonsteroidal anti-inflammatory drug mefenamic acid as a model drug. Then, we studied the biorelevant solubility enhancement of mefenamic acid from EPO-based formulations throughout the gastrointestinal tract (GIT), using the pH-dilution dissolution method. EPO allowed a profound and linear solubility increase of mefenamic acid, from 10 µg/mL without EPO to 9.41 mg/mL in the presence of 7.5% EPO (∼940-fold; 37 °C); however, a concomitant decrease of the drug permeability was obtained, both in vitro and in vivo in rats, indicating a solubility-permeability trade-off. In the absence of an excipient, the unstirred water layer (UWL) adjacent to the GI membrane was found to hinder the permeability of the drug, accounting for this UWL effect and revealing that the true membrane permeability allowed good prediction of the solubility-permeability trade-off as a function of EPO level using a direct relationship between the increased solubility afforded by a given EPO level and the consequent decreased permeability. Biorelevant dissolution studies revealed that EPO levels of 0.05 and 0.1% were insufficient to dissolve mefenamic acid dose during the entire dissolution time course, whereas 0.5 and 1% EPO allowed complete solubility with no drug precipitation. In conclusion, EPO may serve as a potent solubility-enabling excipient for BCS class II/IV acidic drugs; however, it should be used carefully. It is prudent to use the minimal EPO amounts just sufficient to dissolve the drug dose throughout the GIT and not more than that. Excess amounts of EPO provide no solubility gain and cause further permeability loss, jeopardizing the overall success of the formulation. This work may help the formulator to hit the optimal solubility-permeability balance, maximizing the oral bioavailability afforded by the formulation.


Subject(s)
Cell Membrane Permeability/drug effects , Chemistry, Pharmaceutical/methods , Excipients/chemistry , Intestinal Absorption/drug effects , Mefenamic Acid/chemistry , Mefenamic Acid/pharmacokinetics , Polymethacrylic Acids/chemistry , Administration, Oral , Animals , Biological Availability , Drug Compounding/methods , Drug Liberation , Membranes, Artificial , Rats , Rats, Wistar , Solubility
6.
Ther Deliv ; 10(1): 21-36, 2019 01.
Article in English | MEDLINE | ID: mdl-30730824

ABSTRACT

AIM: The current investigation is focused on solid self-microemulsifying drug-delivery systems (S-SMEDDS) of mefenamic acid (MFA) for improving pharmacodynamic activity. Methodology & results: Solubility assessment in various lipid excipients and optimization of pseudoternary plots were carried out for development of liquid SMEDDS. The optimized liquid SMEDD formulation was spray dried to solid dosage form and observed with enhanced amorphization or molecular dispersion of MFA in S-SMEDDS, as evident from x-ray diffractometry and differential scanning calorimetry studies. Enhanced in vitro dissolution rate of optimized formulation was observed, resulting in multifold enhancement in absorption profile of MFA, as compared with pure drug and marketed product. These studies further substantiate the dose reduction in SMEDDS by gaining equivalent therapeutic profile with marketed product. Enhanced analgesic and anti-inflammatory activity was observed with S-SMEDD formulations in acetic acid-induced writhings and carrageenan-induced paw edema models, respectively. CONCLUSION: The optimized S-SMEDD formulation holds great promise for enhancement of its physiochemical and biological attributes.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Drug Carriers/chemistry , Drug Compounding/methods , Mefenamic Acid/administration & dosage , Acetic Acid/toxicity , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Biological Availability , Carrageenan/immunology , Chemistry, Pharmaceutical , Disease Models, Animal , Drug Liberation , Edema/drug therapy , Edema/immunology , Emulsions , Excipients/chemistry , Humans , Male , Mefenamic Acid/pharmacokinetics , Mice , Pain/chemically induced , Pain/drug therapy , Particle Size , Rats , Solubility , Surface-Active Agents/chemistry
7.
Biochem Pharmacol ; 159: 25-31, 2019 01.
Article in English | MEDLINE | ID: mdl-30423313

ABSTRACT

The activities of hundreds, perhaps thousands, of metabolites are regulated by human cytosolic sulfotransferases (SULTs) - a 13-member family of disease relevant enzymes that catalyze transfer of the sulfuryl moiety (-SO3) from PAPS (3'-phosphoadenosine 5'-phosphosulfonate) to the hydroxyls and amines of acceptors. SULTs harbor two independent allosteric sites, one of which, the focus of this work, binds non-steroidal anti-inflammatory drugs (NSAIDs). The structure of the first NSAID-binding site - that of SULT1A1 - was elucidated recently and homology modeling suggest that variants of the site are present in all SULT isoforms. The objective of the current study was to assess whether the NSAID-binding site can be used to regulate sulfuryl transfer in humans in an isoform specific manner. Mefenamic acid (Mef) is a potent (Ki 27 nM) NSAID-inhibitor of SULT1A1 - the predominant SULT isoform in small intestine and liver. Acetaminophen (APAP), a SULT1A1 specific substrate, is extensively sulfonated in humans. Dehydroepiandrosterone (DHEA) is specific for SULT2A1, which we show here is insensitive to Mef inhibition. APAP and DHEA sulfonates are readily quantified in urine and thus the effects of Mef on APAP and DHEA sulfonation could be studied non-invasively. Compounds were given orally in a single therapeutic dose to a healthy, adult male human with a typical APAP-metabolite profile. Mef profoundly decreased APAP sulfonation during first pass metabolism and substantially decreased systemic APAP sulfonation without influencing DHEA sulfonation; thus, it appears the NSAID site can be used to control sulfonation in humans in a SULT-isoform specific manner.


Subject(s)
Acetaminophen/pharmacokinetics , Arylsulfotransferase/metabolism , Mefenamic Acid/pharmacokinetics , Sulfotransferases/metabolism , Acetaminophen/metabolism , Acetaminophen/urine , Allosteric Site , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Arylsulfotransferase/antagonists & inhibitors , Arylsulfotransferase/chemistry , Binding Sites , Dehydroepiandrosterone/administration & dosage , Dehydroepiandrosterone/metabolism , Dehydroepiandrosterone/urine , Drug Interactions , Humans , Isoenzymes/chemistry , Isoenzymes/metabolism , Magnetic Resonance Spectroscopy , Mefenamic Acid/metabolism , Mefenamic Acid/urine , Sulfotransferases/antagonists & inhibitors , Sulfotransferases/chemistry
8.
J Pharm Biomed Anal ; 149: 603-611, 2018 Feb 05.
Article in English | MEDLINE | ID: mdl-29202439

ABSTRACT

Mefenamic Acid (MFA) is a widely-used non-steroidal anti-inflammatory drug. MFA presents four possible crystal forms; Form I and Form II being the only two pure crystals that have been isolated and fully characterized. Both Form I and Form II were prepared following the literature and completely characterized by middle (MIR) and near (NIR) infrared spectroscopy, digital optical microscopy, differential scanning calorimetry, melting point and dissolution properties. In order to develop quantitative models to assess Form I in formulated products, two sets of samples, training (n=10) and validation (n=8) sets, were prepared by mixing both polymorphs and the matrix of excipient (simulating commercial tablets). The particle size of the samples was homogenized by sieving and samples were mechanically mixed. A batch of commercial tablets was gently disaggregated, sieved and mechanically mixed for further analysis. For each sample, full MIR and NIR spectra were acquired and used as input of partial least squares (PLS) algorithm separately. Method optimization and internal validation were performed by leave one out procedure. Full spectra and 5 PLS-factors were used for MIR; while, 5 PLS-factors and mean center spectra of full spectra were the optimal conditions for NIR. Accuracy and precision were assessed by evaluation of the actual vs. predicted curve of validation set; and by calculating validation set recoveries and deviations (104.3±8.2% and 100.4±1.0% for MIR and NIR respectively). Only NIR-PLS yielded acceptable results and low deviations during commercial samples evaluation (102.8±0.1%). The same behavior was observed when spiked tablets were analyzed (103.5±0.5%). Additionally, for the calibration set ten dissolution profiles (average of 6 curves each), were obtained under optimized test conditions (900 ml of buffer phosphate pH 9 with surfactant, apparatus II USP, 100rpm, detection at 342nm). A multiple linear regression (MLR) was carried out using dissolution profiles and Form I content. The developed MLR model could correlate dissolution profiles and polymorphic richness. This approach, coupled to previously developed NIR-PLS, may act as a valid tool to estimate dissolution profiles from solid forms.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Drug Liberation , Mefenamic Acid/pharmacokinetics , Models, Chemical , Spectrophotometry, Infrared/methods , Calibration , Chemistry, Pharmaceutical , Crystallization , Excipients/chemistry , Excipients/pharmacokinetics , Feasibility Studies , Least-Squares Analysis , Mefenamic Acid/chemistry , Multivariate Analysis , Particle Size , Solubility , Spectrophotometry, Infrared/instrumentation , Tablets/chemistry , Tablets/pharmacokinetics
9.
Drug Dev Ind Pharm ; 43(8): 1265-1275, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28398095

ABSTRACT

Drug load plays an important role in the development of solid dosage forms, since it can significantly influence both processability and final product properties. The percolation threshold of the active pharmaceutical ingredient (API) corresponds to a critical concentration, above which an abrupt change in drug product characteristics can occur. The objective of this study was to identify the percolation threshold of a poorly water-soluble drug with regard to the dissolution behavior from immediate release tablets. The influence of the API particle size on the percolation threshold was also studied. Formulations with increasing drug loads were manufactured via roll compaction using constant process parameters and subsequent tableting. Drug dissolution was investigated in biorelevant medium. The percolation threshold was estimated via a model dependent and a model independent method based on the dissolution data. The intragranular concentration of mefenamic acid had a significant effect on granules and tablet characteristics, such as particle size distribution, compactibility and tablet disintegration. Increasing the intragranular drug concentration of the tablets resulted in lower dissolution rates. A percolation threshold of approximately 20% v/v could be determined for both particle sizes of the API above which an abrupt decrease of the dissolution rate occurred. However, the increasing drug load had a more pronounced effect on dissolution rate of tablets containing the micronized API, which can be attributed to the high agglomeration tendency of micronized substances during manufacturing steps, such as roll compaction and tableting. Both methods that were applied for the estimation of percolation threshold provided comparable values.


Subject(s)
Drug Compounding/methods , Excipients/chemistry , Mefenamic Acid/pharmacokinetics , Tablets , Water/chemistry , Chemistry, Pharmaceutical , Drug Liberation , Kinetics , Mefenamic Acid/chemistry , Particle Size , Solubility
10.
Med Chem ; 12(6): 585-91, 2016.
Article in English | MEDLINE | ID: mdl-26740206

ABSTRACT

BACKGROUND: Non steroidal anti-inflammatory drugs are the most widely prescribed drugs to manage pain and inflammatory conditions, but their long term use is associated with gastrointestinal toxicity. OBJECTIVES: The study aimed to synthesize an ester-based prodrug of a non steroidal anti-inflammatory agent, mefenamic acid in order to improve the therapeutic index vis a vis to overcome the side effects such as gastrointestinal irritation and bleeding associated with the use of mefenamic acid. METHODS: The ester prodrug (MA-NH) was prepared by condensing mefenamic acid with N-hydroxymethylsuccinimide in the presence of Phosphorus oxychloride. The pharmacokinetic profile, including stability and release of mefenamic acid and N-hydroxymethylsuccinimide from the ester prodrug (MA-NH) was studied by RP- HPLC in acidic medium (pH 1.2), basic medium (pH 7.4), 80 % v/v human plasma, 10 % w/v rat intestinal homogenate and 10 % w/v rat liver homogenate (pH 7.4). RESULTS: The chemical structure of the title compound was characterized by using modern spectroscopic techniques. The prodrug was found to be stable in acid medium, but it hydrolyzed and released sufficient quantities of the drug in alkaline medium. The prodrug produced lesser number of ulcers and showed improved analgesic and anti-inflammatory activity as compared to the parent drug. CONCLUSION: The results indicate that the synthesized prodrug (MA-NH) is better in terms of analgesic and antiinflammatory activities and with less GI toxicity than the parent drug.


Subject(s)
Analgesics/therapeutic use , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Mefenamic Acid/analogs & derivatives , Mefenamic Acid/metabolism , Prodrugs/therapeutic use , Succinimides/therapeutic use , Analgesics/chemical synthesis , Analgesics/pharmacokinetics , Analgesics/toxicity , Animals , Anti-Inflammatory Agents, Non-Steroidal/chemical synthesis , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Female , Humans , Hydrolysis , Male , Mefenamic Acid/chemical synthesis , Mefenamic Acid/pharmacokinetics , Mefenamic Acid/therapeutic use , Mefenamic Acid/toxicity , Prodrugs/chemical synthesis , Prodrugs/pharmacokinetics , Prodrugs/toxicity , Rats , Rats, Wistar , Succinimides/chemical synthesis , Succinimides/chemistry , Succinimides/pharmacokinetics , Succinimides/toxicity , Ulcer/chemically induced
11.
Mater Sci Eng C Mater Biol Appl ; 58: 629-38, 2016 Jan 01.
Article in English | MEDLINE | ID: mdl-26478354

ABSTRACT

PURPOSE: The anionic form of the drug mefenamic acid intercalated into the nanocarrier layered double hydroxide (LDH-Mef) was evaluated by anti-inflammatory and antinociceptive assays. METHODS: The LDH-Mef material was characterized by a set of physicochemical techniques, which was supported by Density Functional Theory calculations. The pharmacological effects of LDH-Mef (40 wt% of drug) were evaluated by hemolytic, anti-inflammatory activity and antinociceptive assays. RESULTS: In vivo assays were conducted for the first time in order to assess the LDH-Mef potential. The hemolytic effects decreased for the intercalated Mef as demonstrated by the higher tolerated hemolytic concentration (1.83 mM) compared to mefenamic acid (MefH), 0.48 mM. Pretreatment of animals with MefH or LDH-Mef reduced carrageenan-, dextran sulfate- and PGE2-induced paw edema. MefH or LDH-Mef also decrease total leucocytes and neutrophil counts of the peritoneal cavity after inflammation induction with carrageenan. In the nociception model, oral pretreatment with LDH-Mef reduced mechanical hypernociception carrageenan-induced after 3-4h and also the number of writhings induced by acetic acid. CONCLUSIONS: This work shows the increase of the anti-inflammatory and antinociceptive potential of the drug confined into the LDH, as well as, its hemolytic effect.


Subject(s)
Analgesics/chemistry , Anti-Inflammatory Agents/chemistry , Drug Carriers/chemistry , Mefenamic Acid/chemistry , Nanoparticles/chemistry , Analgesics/pharmacokinetics , Analgesics/pharmacology , Analgesics/therapeutic use , Animals , Anti-Inflammatory Agents/pharmacokinetics , Anti-Inflammatory Agents/pharmacology , Anti-Inflammatory Agents/therapeutic use , Behavior, Animal/drug effects , Carrageenan , Drug Carriers/pharmacokinetics , Edema/chemically induced , Edema/drug therapy , Hemolysis/drug effects , Humans , Hydroxides/chemistry , Inflammation/chemically induced , Inflammation/drug therapy , Male , Mefenamic Acid/pharmacokinetics , Mefenamic Acid/pharmacology , Mefenamic Acid/therapeutic use , Mice , Nanoparticles/toxicity
12.
Biopharm Drug Dispos ; 36(8): 529-51, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26109199

ABSTRACT

Mefenamic acid (MEF) is a widely prescribed non-steroidal anti-inflammatory drug that has been found associated with rare but severe cases of hepatotoxicity, nephrotoxicity and gastrointestinal toxicity. The formation of protein-reactive acylating metabolites such as 1-O-acyl-MEF glucuronide (MEFG) and 3'-hydroxymethyl-MEF 1-O-acyl-glucuronide is one proposed cause. In addition to the well-reported 3'-hydroxymethyl-MEF, two mono-hydroxyl-MEF (OH-MEFs) were recently identified in vitro. However, in vivo evidence is lacking and whether these OH-MEFs would be further glucuronidated to the potentially reactive 1-O-acyl-glucuronides (OH-MEFGs) is unknown. Utilizing UPLC-Q-TOF/MS and LC-MS/MS, the current study identified, for the first time, four OH-MEFs and their corresponding OH-MEFGs from plasma after a single oral administration of MEF (40 mg/kg) to rats, including an OH-MEF that has not been reported previously. The systemic exposure of these identified metabolites was high, with metabolic to parent AUC0 → 24 h ratios reaching 23-52% (OH-MEFs) and 8-29% (OH-MEFGs). These metabolites also had a long systemic exposure time in both single and 5 day multiple oral MEF-treated rats, with elimination half-lives between 9 h and > 24 h. In addition to these novel metabolites, the previously reported MEFG was also identified and its systemic exposure was found to be doubled after multiple MEF administrations. These pharmacokinetic results suggest that systemic toxicities caused by the potentially reactive MEFG and OH-MEFGs could be considerable, especially after repeated MEF treatment. Nevertheless, MEFG and OH-MEFGs had negligible uptake in the brain, indicating a minimal risk of brain toxicities. Furthermore, an in situ intestinal perfusion study revealed that during MEF absorption, it was extensively metabolized to MEFG while < 5% was metabolized to OH-MEFs and OH-MEFGs.


Subject(s)
Brain/metabolism , Intestine, Small/metabolism , Mefenamic Acid/analogs & derivatives , Microsomes, Liver/metabolism , Administration, Oral , Animals , Biotransformation , Brain/drug effects , Chromatography, Liquid , Dose-Response Relationship, Drug , Glucuronates/blood , Glucuronates/metabolism , Glucuronates/pharmacokinetics , Glucuronates/toxicity , In Vitro Techniques , Intestinal Absorption , Male , Mefenamic Acid/blood , Mefenamic Acid/metabolism , Mefenamic Acid/pharmacokinetics , Mefenamic Acid/toxicity , Molecular Structure , Perfusion , Rats , Rats, Sprague-Dawley , Tandem Mass Spectrometry , Tissue Distribution
13.
J Ethnopharmacol ; 170: 106-16, 2015 Jul 21.
Article in English | MEDLINE | ID: mdl-25980420

ABSTRACT

ETHNOPHARMACOLOGICAL RELEVANCE: Scutellariae Radix (SR), the dried root of Scutellariae baicalensis Georgi, has a lot in common with non-steroidal anti-inflammatory drugs (NSAIDs). Their similarities in therapeutic action (anti-inflammation) and metabolic pathways (phase II metabolisms) may lead to co-administration by patients with the potential of pharmacokinetic and/or pharmacodynamic interactions. The current study aims to investigate the potential interactions between SR and an NSAID, mefenamic acid (MEF), on the overall pharmacokinetic dispositions, anti-inflammatory effects and adverse effects in rats. MATERIALS AND METHODS: The current study simultaneously monitored the pharmacokinetic and pharmacodynamic interactions in a single animal. Four groups of Sprague-Dawley rats (n=7 each) received oral doses of a standardized SR extract (300mg/kg, twice daily), MEF (40mg/kg, daily), combination of SR extract and MEF, and vehicle control, respectively, for 5 days. On Day 5, blood samples were collected after first dose over 24h for the determination of (1) plasma concentrations of SR bioactive components, MEF and its metabolites by LC-MS/MS, and (2) prostaglandin E2 (PGE2) production and cyclooxygenase-2 (COX-2) gene expression by ex vivo analyses using LPS-stimulated RAW264.7 macrophage cells, ELISA and real time-PCR. After the rats were sacrificed, stomachs were isolated to assess their gross mucosal damage. Statistical comparisons were conducted using ANOVA and t-test. RESULTS: Minimal pharmacokinetic interaction between SR extract and MEF was observed. Co-administration of SR extract and MEF did not significantly alter the plasma concentration-time profile or the pharmacokinetic parameters such as Cmax, AUC0→24, Tmax or clearance. Pharmacodynamic interaction via the COX-2 pathway was observed. The PGE2 level in LPS-stimulated RAW264.7 cells treated with plasma collected from control group over the 24h sampling (AUC0→24[PGE2]) was 191981±8789pg/mlhr, which was significantly reduced to 174,780±6531 and 46,225±1915pg/mlhr by plasma collected from rats administered with SR extract and MEF, respectively. Co-administration of SR extract and MEF further potentiated the PGE2 inhibition, with an AUC0→24[PGE2] of 37013±2354pg/mlhr (p<0.05, compared to SR or MEF group). By analyzing the COX-2 gene expression, SR extract significantly prolonged the COX-2 inhibitory effect of MEF over the 24h (p<0.05). Furthermore, the MEF-induced stomach ulcer after the 5-day treatment, as evidenced by the increased gross ulcer index and sum of lesion length (p<0.05, compared to control), could be alleviated by co-administration with SR extract (p<0.05). CONCLUSIONS: Co-administration of SR extract and MEF potentiated the anti-inflammatory effects, alleviated the MEF-induced stomach adverse effect while having minimal pharmacokinetic interactions. Our findings provide insight for combination therapy of SR extract and MEF against inflammatory diseases.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Herb-Drug Interactions , Mefenamic Acid/pharmacology , Plant Extracts/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/toxicity , Area Under Curve , Cell Line , Chromatography, Liquid , Gastric Mucosa/drug effects , Gastric Mucosa/pathology , Inflammation/drug therapy , Inflammation/pathology , Macrophages/drug effects , Macrophages/metabolism , Male , Mefenamic Acid/pharmacokinetics , Mefenamic Acid/toxicity , Mice , Rats , Rats, Sprague-Dawley , Scutellaria baicalensis , Tandem Mass Spectrometry
14.
Chem Res Toxicol ; 27(12): 2071-81, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25372302

ABSTRACT

Mefenamic acid (MFA) has been associated with rare but severe cases of hepatotoxicity, nephrotoxicity, gastrointestinal toxicity, and hypersensitivity reactions that are believed to result from the formation of reactive metabolites. Although formation of protein-reactive acylating metabolites by phase II metabolism has been well-studied and proposed to be the cause of these toxic side effects, the oxidative bioactivation of MFA has not yet been competely characterized. In the present study, the oxidative bioactivation of MFA was studied using human liver microsomes (HLM) and recombinant human P450 enzymes. In addition to the major metabolite 3'-OH-methyl-MFA, resulting from the benzylic hydroxylation by CYP2C9, 4'-hydroxy-MFA and 5-hydroxy-MFA were identified as metabolites resulting from oxidative metabolism of both aromatic rings of MFA. In the presence of GSH, three GSH conjugates were formed that appeared to result from GSH conjugation of the two quinoneimines formed by further oxidation of 4'-hydroxy-MFA and 5-hydroxy-MFA. The major GSH conjugate was identified as 4'-OH-5'-glutathionyl-MFA and was formed at the highest activity by CYP1A2 and to a lesser extent by CYP2C9 and CYP3A4. Two minor GSH conjugates resulted from secondary oxidation of 5-hydroxy-MFA and were formed at the highest activity by CYP1A2 and to a lesser extent by CYP3A4. Additionally, the ability of seven human glutathione S-transferases (hGSTs) to catalyze the GSH conjugation of the quinoneimines formed by P450s was also investigated. The highest increase of total GSH conjugation was observed with hGSTP1-1, followed by hepatic hGSTs hGSTA2-2 and hGSTM1-1. The results of this study show that, next to phase II metabolites, reactive quinoneimines formed by oxidative bioactivation might also contribute to the idiosyncratic toxicity of MFA.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Glutathione Transferase/metabolism , Imines/chemistry , Mefenamic Acid/pharmacokinetics , Quinones/metabolism , Activation, Metabolic , Base Sequence , Chromatography, High Pressure Liquid , DNA Primers , Humans , Mefenamic Acid/antagonists & inhibitors , Oxidation-Reduction , Proton Magnetic Resonance Spectroscopy
15.
Chem Res Toxicol ; 27(4): 576-86, 2014 Apr 21.
Article in English | MEDLINE | ID: mdl-24552538

ABSTRACT

NAD(P)H: quinone oxidoreductase 1 (NQO1) is an enzyme capable of reducing a broad range of chemically reactive quinones and quinoneimines (QIs) and can be strongly upregulated by Nrf2/Keap1-mediated stress responses. Several commonly used drugs implicated in adverse drug reactions (ADRs) are known to form reactive QI metabolites upon bioactivation by P450, such as acetaminophen (APAP), diclofenac (DF), and mefenamic acid (MFA). In the present study, the reductive activity of human NQO1 toward the QI metabolites derived from APAP and hydroxy-metabolites of DF and MFA was studied, using purified bacterial P450 BM3 (CYP102A1) mutant M11 as a bioactivation system. The NQO1-catalyzed reduction of the QI metabolites was quantified relative to spontaneous glutathione (GSH) conjugation. Addition of NQO1 to the incubations strongly reduced the formation of all corresponding GSH conjugates, and this activity could be prevented by dicoumarol, a selective NQO1 inhibitor. The GSH conjugation was strongly increased by adding human GSTP1-1 in a wide range of GSH concentrations. Still, NQO1 could effectively compete with the GST catalyzed GSH conjugation by reducing the QIs. In conclusion, we identified the QI metabolites of the 4'- and 5-hydroxy-metabolites of DF and MFA as novel substrates for human NQO1. NQO1-mediated reduction proves to be an effective pathway to detoxify these QI metabolites in addition to GSH conjugation. Genetically determined deficiency of NQO1 therefore might be a risk factor for ADRs induced by reactive QI drug metabolites.


Subject(s)
Diclofenac/pharmacokinetics , Mefenamic Acid/pharmacokinetics , NAD(P)H Dehydrogenase (Quinone)/metabolism , Quinones/antagonists & inhibitors , Activation, Metabolic , Catalysis , Cell Line , Glutathione/metabolism , Glutathione S-Transferase pi/metabolism , Humans , Imines/chemistry , Quinones/chemistry , Quinones/metabolism
16.
Mol Pharm ; 10(11): 3997-4004, 2013 Nov 04.
Article in English | MEDLINE | ID: mdl-23961831

ABSTRACT

Many pharmaceutically active compounds are weak electrolytes and are ionizable in the pH range experienced throughout the gastrointestinal tract. Changes in protonation state due to pH changes in the gut can have dramatic effects on solubility, dissolution, and permeation through biological barriers. Preclinical assessment of the pH-dependence of oral absorption is critical for compounds possessing pH-dependent solubility. Here we examine pH-dependent solubility and oral exposure in rat for three model compounds, dasatinib, ketoconazole, and mefenamic acid. Dasatinib and ketoconazole are both weak bases, while mefenamic acid is a carboxylic acid. The effects of gastric pH modulators, pentagastrin and famotidine, were investigated in rat PK studies to assess the applicability of using the rat to evaluate the risk of pH-dependent oral exposure for ionizable compounds. Dasatinib showed similar exposure between control and pentagastrin-pretreated groups, and 4.5-fold lower AUC in famotidine-pretreated rats. Ketoconazole showed a 2-fold increase in AUC in pentagastrin-treated rats relative to control, and 4.5-fold lower AUC in famotidine treated rats, relative to the pentagastrin group. Mefenamic acid showed highly similar exposures among control, pentagastrin-pretreated, and famotidine-pretreated groups. The rat model was shown to be useful for compounds displaying pH-dependent solubility and oral absorption that may be affected by gastric pH modulators.


Subject(s)
Administration, Oral , Animals , Dasatinib , Famotidine/administration & dosage , Famotidine/pharmacokinetics , Humans , Hydrogen-Ion Concentration , Ketoconazole/administration & dosage , Ketoconazole/pharmacokinetics , Male , Mefenamic Acid/administration & dosage , Mefenamic Acid/pharmacokinetics , Pentagastrin/administration & dosage , Pentagastrin/pharmacokinetics , Pyrimidines/administration & dosage , Pyrimidines/pharmacokinetics , Rats , Rats, Sprague-Dawley , Solubility , Thiazoles/administration & dosage , Thiazoles/pharmacokinetics
17.
Drug Metab Dispos ; 40(8): 1515-26, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22577085

ABSTRACT

Carboxylic acid-containing nonsteroidal anti-inflammatory drugs (NSAIDs) can be metabolized to chemically reactive acyl glucuronide and/or S-acyl-CoA thioester metabolites capable of transacylating GSH. We investigated the metabolism of the NSAID mefenamic acid (MFA) to metabolites that transacylate GSH, leading to MFA-S-acyl-GSH thioester (MFA-SG) formation in incubations with rat and human hepatocytes and in vivo in rat bile. Thus, incubation of MFA (1-500 µM) with rat hepatocytes led to the detection of MFA-1-ß-O-acyl glucuronide (MFA-1-ß-O-G), MFA-S-acyl-CoA (MFA-SCoA), and MFA-SG by liquid chromatography-tandem mass spectrometric analysis. The C(max) of MFA-SG (330 nM; 10-min incubation with 100 µM MFA) was 120- to 1400-fold higher than the C(max) of drug S-acyl-GSH adducts detected from studies with other carboxylic acid drugs to date. MFA-SG was also detected in incubations with human hepatocytes, but at much lower concentrations. Inhibition of MFA acyl glucuronidation in rat hepatocytes had no effect on MFA-SG formation, whereas a 58 ± 1.7% inhibition of MFA-SCoA formation led to a corresponding 66 ± 3.5% inhibition of MFA-SG production. Reactivity comparisons with GSH in buffer showed MFA-SCoA to be 80-fold more reactive than MFA-1-ß-O-G forming MFA-SG. MFA-SG was detected in MFA-dosed (100 mg/kg) rat bile, where 17.4 µg was excreted after administration. In summary, MFA exhibited bioactivation in rat and human hepatocytes and in vivo in rat, leading to reactive acylating derivatives that transacylate GSH. The formation of MFA-SG in hepatocytes was shown not to be mediated by reaction with MFA-1-ß-O-G, and not solely by MFA-SCoA, but perhaps also by intermediary MFA-acyl-adenylate formation, which is currently under investigation.


Subject(s)
Biotransformation , Glutathione/pharmacokinetics , Mefenamic Acid/pharmacokinetics , Animals , Chromatography, Liquid , Hepatocytes/metabolism , Humans , In Vitro Techniques , Rats , Tandem Mass Spectrometry
18.
Biomed Chromatogr ; 26(10): 1137-42, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22275128

ABSTRACT

In the present study, the development and validation of an LC-MS/MS method for quantifying mefenamic acid in human plasma is described. The method involves liquid-liquid extraction using diclofenac as an internal standard (IS). Chromatographic separation was achieved on a Thermo Hypurity C(18) , 50 × 4.6 mm, 5 µm column with a mobile phase consisting of 2 m m ammonium acetate buffer and methanol (pH 4.5 adjusted with glacial acetic acid; 15:85, v/v) at a flow-rate of 0.75 mL/min and the total run time was 1.75 min. Analyte was introduced to the LC-MS/MS using an atmospheric pressure ionization source. Both the drug and IS were detected in negative-ion mode using multiple reaction monitoring m/z 240.0 → 196.3 and m/z 294.0 → 250.2, respectively, with a dwell time of 200 ms for each of the transitions. The standard curve was linear from 20 to 6000 ng/mL. This assay allows quantification of mefenamic acid at a concentration as low as 20 ng/mL in human plasma. The observed mean recovery was 73% for the drug. The applicability of this method for pharmacokinetic studies has been established after successful application during a 12-subject bioavailabity study.


Subject(s)
Chromatography, High Pressure Liquid/methods , Mefenamic Acid/blood , Tandem Mass Spectrometry/methods , Acetates , Anticoagulants/pharmacology , Area Under Curve , Diclofenac , Drug Stability , Humans , Hydrogen-Ion Concentration , Linear Models , Liquid-Liquid Extraction , Male , Mefenamic Acid/chemistry , Mefenamic Acid/pharmacokinetics , Methanol , Reproducibility of Results , Sensitivity and Specificity
19.
Med Princ Pract ; 21(3): 223-7, 2012.
Article in English | MEDLINE | ID: mdl-22116543

ABSTRACT

OBJECTIVES: The objectives of this study were to assess the bioavailability of an optimized mephenamic acid (MFA) microspheres (test) against a Ponstan® capsule (reference) in healthy volunteers, and to establish a correlation with in vitro parameters. SUBJECTS AND METHODS: Four subjects received the test and reference (250 mg MFA each) in a randomized crossover design, separated by a 1-week washout period. The drug was analyzed in plasma by a specific high-performance liquid chromatographic method. The relevant pharmacokinetic parameters [maximum plasma concentration (C(max)), time of peak concentration (T(max)), area under plasma concentration-time curves from 0 to 12 h (AUC(0-12)) and area under plasma concentration-time curves from zero to ∞ (AUC(0-)∞)] were calculated from the plasma drug concentration-time data. RESULTS: The test product exhibited faster absorption (T(max) of 1.87 ± 0.482 vs. 2.14 ± 0.20 h; C(max) of 5.91 ± 0.604 vs. 3.58 ± 0.671 µg/ml) when compared to the reference. The relative bioavailability of the test compared to the reference capsule was 172%. Good correlations were established between the in vitro 90% dissolution (T90) and each of the AUC(0-12) and T(max), as well as between the percentage of drug released and plasma concentrations. CONCLUSION: The formulation of MFA microsphere with polyethylene glycol improved the dissolution rate and bioavailability of MFA, as evidenced by a higher C(max), AUC(0-12) and AUC(0-)∞, and shorter T(max) values. Good correlations between T90 and both AUC(0-12) and T(max) as well as between the percentage of drug released and plasma concentrations were achieved.


Subject(s)
Cyclooxygenase Inhibitors/pharmacokinetics , Mefenamic Acid/pharmacokinetics , Microspheres , Adult , Analysis of Variance , Area Under Curve , Biological Assay , Cyclooxygenase Inhibitors/administration & dosage , Humans , In Vitro Techniques , Male , Mefenamic Acid/administration & dosage , Statistics as Topic
20.
Pharm Dev Technol ; 16(5): 497-510, 2011 Oct.
Article in English | MEDLINE | ID: mdl-20550465

ABSTRACT

A suitable topical formulation of mefenamic acid was developed in order to eliminate the gastrointestinal disorders associated with its oral administration. Drug coprecipitates prepared with different polymers at various drug-to-polymer ratios improved drug solubility and dissolution compared to pure drug and physical mixtures. PVP polymers (ratio 1:4) produced the best results. Aqueous ionic cream, ointments of absorption and water soluble bases and gels of methylcellulose, carboxymethylcellulose sodium, HPMC, Carbopol® 934 and 940, and Pluronic® F127 bases containing 1-10% drug as coprecipitates of PVP polymers (1:4) were prepared. The highest drug release was achieved at 1% drug concentration from water soluble base and methylcellulose among cream/ointment and gel bases, respectively. Gels, in general yielded better release than creams/ointments. All tested medicated creams/ointments exhibited plastic flow while all gels conformed to pseudoplasticity. Most of them showed thixotropy, a desired property of topical preparations. Stability studies revealed that HPMC and methylcellulose had the smallest changes in drug content, viscosity, and pH among the formulations. Considering drug release, rheological properties, and stability, methylcellulose gel containing 1% drug as coprecipitates of PVP K90 was the best among the studied formulations, was promising for improving bioavailability of mefenamic acid and can be used in future studies.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/chemistry , Drug Delivery Systems/methods , Mefenamic Acid , Absorption , Acrylic Resins , Administration, Topical , Biological Availability , Drug Compounding , Drug Stability , Gels/chemistry , Humans , Mefenamic Acid/chemistry , Mefenamic Acid/pharmacokinetics , Methylcellulose/analogs & derivatives , Methylcellulose/chemistry , Ointment Bases/administration & dosage , Ointments/administration & dosage , Polyethylenes/chemistry , Polypropylenes/chemistry , Polyvinyls/chemistry , Pyrrolidines/chemistry , Rheology , Viscosity
SELECTION OF CITATIONS
SEARCH DETAIL
...