Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
Add more filters










Publication year range
1.
J Pharm Sci ; 112(12): 3224-3232, 2023 12.
Article in English | MEDLINE | ID: mdl-37722451

ABSTRACT

Remdesivir (RDV) and tenofovir alafenamide (TAF) are prodrugs designed to be converted to their respective active metabolites. Plasma protein binding (PPB) determination of these prodrugs is important for patients with possible alteration of free fraction of the drugs due to plasma protein changes in renal impairment, hepatic impairment, or pregnancy. However, the prodrugs' instability in human plasma presents a challenge for accurate PPB determination. In this research work, two approaches were used in the method development and qualification for PPB assessment of RDV and TAF. For RDV, dichlorvos was used to inhibit esterase activity to stabilize the prodrug in plasma during equilibrium dialysis (ED). The impact of dichlorvos on protein binding was evaluated and determined to be insignificant by comparing the unbound fraction (fu) determined by the ED method with dichlorvos present and the fu determined by an ultrafiltration method without dichlorvos. In contrast to RDV, TAF degradation in plasma is ∼3-fold slower, and TAF stability cannot be improved by dichlorvos. Fit-for-purpose acceptance criteria for the TAF PPB method were chosen, and an ED method was developed based on these criteria. These two methods were then qualified and applied for PPB determinations in clinical studies.


Subject(s)
Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Anti-HIV Agents , HIV Infections , Prodrugs , Humans , Tenofovir , Anti-HIV Agents/therapeutic use , Protein Binding , Prodrugs/metabolism , Dichlorvos/therapeutic use , Adenine , Blood Proteins/metabolism , HIV Infections/drug therapy
2.
J Pharm Biomed Anal ; 212: 114646, 2022 Apr 01.
Article in English | MEDLINE | ID: mdl-35180564

ABSTRACT

SBECD (Captisol®) with an average degree of substitution of 6.5 sulfobutylether functional groups (SBE = 6.5), is a solubility enhancer for remdesivir (RDV) and a major component in Veklury, which was approved by FDA for the treatment of patients with COVID-19 over 12 years old and weighing over 40 kg who require hospitalization. SBECD is cleared mainly by renal filtration, thus, potential accumulation of SBECD in the human body is a concern for patients dosed with Veklury with compromised renal function. An LC-MS/MS method was developed and validated for specific, accurate, and precise determination of SBECD concentrations in human plasma. In this method, the hexa-substituted species, SBE6, was selected for SBECD quantification, and the mass transition from its dicharged molecular ion [(M-2H)/2]2-, Molecular (parent) Ion (Q1)/Molecular (parent) Ion (Q3) of m/z 974.7/974.7, was selected for quantitative analysis of SBECD. Captisol-G (SBE-γ-CD, SBE = 3) was chosen as the internal standard. With 25 µL of formic-acid-treated sample and with a calibration range of 10.0-1000 µg/mL, the method was validated with respect to pre-established criteria based on regulatory guidelines and was applied to determine SBECD levels in plasma samples collected from pediatric patients during RDV clinical studies.


Subject(s)
COVID-19 Drug Treatment , beta-Cyclodextrins , Adenosine Monophosphate/analogs & derivatives , Alanine/analogs & derivatives , Child , Chromatography, Liquid , Humans , SARS-CoV-2 , Sodium , Tandem Mass Spectrometry/methods
3.
Anal Biochem ; 593: 113611, 2020 03 15.
Article in English | MEDLINE | ID: mdl-32035040

ABSTRACT

Tenofovir disoproxil fumarate (TDF) and tenofovir alafenamide (TAF) are both tenofovir (TFV) prodrugs, with the same active intracellular metabolite, TFV-diphosphate (TFV-DP). TAF delivers TFV-DP to target cells more efficiently and at lower doses than TDF, thereby substantially reducing systemic exposure to TFV, which results in improved bone and renal safety relative to TDF. As such, the method developed for the determination of TFV following TAF administration involved two key differences from determination of TFV following TDF administration. First, human plasma samples (500 µL) immediately upon collection were treated with 20% formic acid (40 µL) (plasma: formic acid ratio of 100:8) to minimize hydrolysis of TAF to TFV, and thereby avoided overestimation of TFV concentrations. Second, various TFV validation tests were conducted in the presence of TAF to mimic the high TAF:TFV ratios in clinical samples collected within ~2 h after dosing. The method for determination of TFV was developed and validated at a US lab and followed FDA and EMA guidelines. To support global clinical studies of TAF, the method was cross-validated (one-way) between the US lab and a China lab and was successfully used for TFV determination in plasma samples from a clinical study that involved healthy Chinese subjects.


Subject(s)
Adenine/analogs & derivatives , Anti-HIV Agents/pharmacokinetics , Chromatography, Liquid/methods , Tandem Mass Spectrometry/methods , Tenofovir/blood , Adenine/pharmacokinetics , Alanine , Formates/chemistry , Humans , Prodrugs/pharmacokinetics
4.
Anal Biochem ; 585: 113399, 2019 11 15.
Article in English | MEDLINE | ID: mdl-31437427

ABSTRACT

A fluorescent quantitation method to determine PBMC-derived DNA amounts using purified human genomic DNA (gDNA) as the reference standard was developed and validated. gDNA was measured in a fluorescence-based assay using a DNA intercalant, SYBR green. The fluorescence signal was proportional to the amount (mass) of DNA in the sample. The results confirmed a linear fit from 0.0665 to 1.17 µg/µL for gDNA, corresponding to 2.0 × 106 to 35.0 × 106 cells/PBMC sample. Intra-batch and inter-batch accuracy (%RE) was within ±15%, and precision (%CV) was <15%. Benchtop stability, freeze/thaw stability and long term storage stability of gDNA in QC sample matrix, PBMC pellets samples, and pellet debris samples, respectively, as well as dilution linearity had been established. Consistency between hemocytometry cell counting method and gDNA-based counting method was established. 6 out of 6 evaluated PBMC lots had hemocytometry cell counts that were within ±20% of the cell counts determined by the gDNA method. This method was used in conjunction with a validated LC-MS/MS method to determine the level of tenofovir diphosphate (TFV-DP), the active intracellular metabolite of the prodrugs tenofovir alafenamide (TAF) and tenofovir disoproxil fumarate (TDF), measured in PBMCs in clinical trials of TAF or TDF-containing fixed dose combinations.


Subject(s)
Adenine/analogs & derivatives , DNA/chemistry , Leukocytes, Mononuclear/metabolism , Organophosphates/analysis , Adenine/analysis , Adenine/metabolism , Alanine , Cell Count/methods , Chromatography, High Pressure Liquid , Fluorescent Dyes/chemistry , Genomics , Humans , Image Cytometry , Intercalating Agents/chemistry , Prodrugs/metabolism , Tandem Mass Spectrometry , Tenofovir/metabolism
5.
Expert Opin Drug Metab Toxicol ; 14(8): 781-802, 2018 Aug.
Article in English | MEDLINE | ID: mdl-30010446

ABSTRACT

INTRODUCTION: Peripheral blood mononuclear cells (PBMCs) are a critical component of the immune system and the target cells for human immunodeficiency virus, type 1 (HIV-1) infection. Nucleoside/nucleotide analogs for the treatment of HIV infection are prodrugs that require cellular activation to triphosphate (TP) metabolites for antiviral activity. A reliable method of PBMC isolation and subsequent cell counting, as well as an accurate bioanalytical determination of the TPs in PBMCs are important for understanding the intracellular pharmacokinetic (PK) of the TPs and its correlation with plasma PK, the drug effect, and dose determination. Areas covered: The authors review the challenges and solutions in PBMC sample collection, sample processing, cell lysis, cell counting methods, analyte extraction, and liquid chromatography/tandem mass spectrometry (LC-MS/MS) quantitative analysis of the nucleoside reverse transcriptase inhibitor-triphosphate (NRTI-TP) metabolites, and analogs. Expert opinion: Analyzing large numbers of clinical PBMC samples for determination of NRTI-TPs and analogs in PBMCs requires not only a validated LC-MS/MS bioanalytical method but also reliable methods for PBMC isolation, counting, cell lysis, and analyte recovery, and an approach for assessing analyte stability. Furthermore, a simple, consistent, and validated cell counting method often involves DNA quantitation of the PBMCs samples collected from clinical studies.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Leukocytes, Mononuclear/metabolism , Reverse Transcriptase Inhibitors/pharmacokinetics , Anti-HIV Agents/administration & dosage , Cell Count , Chromatography, Liquid/methods , HIV Infections/drug therapy , Humans , Polyphosphates/metabolism , Reproducibility of Results , Reverse Transcriptase Inhibitors/administration & dosage , Tandem Mass Spectrometry/methods
6.
Clin Pharmacokinet ; 57(11): 1449-1457, 2018 11.
Article in English | MEDLINE | ID: mdl-29520729

ABSTRACT

BACKGROUND: The pharmacokinetics and safety of velpatasvir, a potent pangenotypic hepatitis C virus NS5A inhibitor, were evaluated in two hepatic impairment studies: a phase I study in hepatitis C virus-uninfected subjects and a phase III study (ASTRAL-4) in hepatitis C virus-infected patients. METHODS: In the phase I study, subjects with moderate or severe hepatic impairment (Child-Pugh-Turcotte Class B or C), and demographically matched subjects with normal hepatic function received a single dose of velpatasvir 100 mg. Pharmacokinetics and safety assessments were performed, and pharmacokinetic parameters were calculated using non-compartmental methods and summarized using descriptive statistics and compared statistically by geometric least-squares mean ratios and 90% confidence intervals. In ASTRAL-4, subjects with decompensated cirrhosis (Child-Pugh-Turcotte Class B) were randomized to receive treatment with either sofosbuvir/velpatasvir ± ribavirin for 12 weeks or sofosbuvir/velpatasvir for 24 weeks. Pharmacokinetic and safety assessments were performed and pharmacokinetic parameters were calculated using a non-compartmental analysis and summarized using descriptive statistics and were compared to pharmacokinetics from ASTRAL-1 [subjects without cirrhosis or with compensated (Child-Pugh-Turcotte Class A) cirrhosis]. RESULTS: In the phase I study, plasma exposures (area under the concentration-time curve) were similar in subjects with Child-Pugh-Turcotte Class B (n = 10) or Child-Pugh-Turcotte Class C hepatic impairment (n = 10) compared with normal hepatic function (n = 13). Percent free velpatasvir was similar in subjects without or with any degree of hepatic impairment. In the phase III study, velpatasvir overall exposure (area under the concentration-time curve over the 24-h dosing interval; AUCtau) was similar and sofosbuvir exposures were higher (~ 100%) for patients with Child-Pugh-Turcotte Class B hepatic impairment compared with the ASTRAL-1 population, which was not considered clinically relevant. CONCLUSIONS: No sofosbuvir/velpatasvir dose modification is warranted for patients with any degree of hepatic impairment.


Subject(s)
Carbamates/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Sofosbuvir/pharmacokinetics , Adolescent , Adult , Aged , Antiviral Agents/adverse effects , Antiviral Agents/blood , Antiviral Agents/pharmacokinetics , Antiviral Agents/therapeutic use , Carbamates/adverse effects , Carbamates/blood , Drug Combinations , Drug Therapy, Combination/statistics & numerical data , Female , Hepatic Insufficiency/blood , Hepatic Insufficiency/drug therapy , Hepatitis C/blood , Hepatitis C/complications , Hepatitis C/drug therapy , Heterocyclic Compounds, 4 or More Rings/adverse effects , Heterocyclic Compounds, 4 or More Rings/blood , Humans , Liver Cirrhosis/blood , Liver Cirrhosis/complications , Male , Middle Aged , Ribavirin/therapeutic use , Sofosbuvir/adverse effects , Uridine/analogs & derivatives , Uridine/blood , Uridine/pharmacokinetics , Young Adult
7.
Clin Infect Dis ; 67(6): 934-940, 2018 08 31.
Article in English | MEDLINE | ID: mdl-29522076

ABSTRACT

Background: Combining antiviral regimens in the hepatitis C virus (HCV)/human immunodeficiency virus (HIV)-coinfected population can be complex as they share overlapping mechanisms for elimination that may result in drug interactions. The pharmacokinetics, safety, and tolerability of sofosbuvir/velpatasvir (SOF/VEL) with multiple antiretroviral (ARV) regimens were evaluated. Methods: Healthy volunteers were enrolled into 2 phase 1, open-label, randomized, multiple-dose, cross-over studies. SOF/VEL and ARV regimens were administered alone and in combination; ARVs (and pharmacokinetic enhancers) included atazanavir (ATV), cobicistat (COBI), darunavir (DRV), dolutegravir (DTG), efavirenz (EFV), elvitegravir (EVG), emtricitabine (FTC), lopinavir (LPV), raltegravir (RAL), rilpivirine (RPV), ritonavir (RTV), tenofovir alafenamide (TAF), and tenofovir disoproxil fumarate (TDF). Geometric least squares means ratios (coadministration:alone) and 90% confidence intervals were constructed for area under the plasma concentration-time curve over the dosing interval, maximum concentration, and trough, for all analytes. Safety and tolerability were also evaluated. Results: In total, 237 participants were enrolled. No clinically relevant differences in the pharmacokinetics (PK) of SOF, SOF metabolite GS-331007, or VEL were observed other than an approximate 50% decrease in VEL exposure when administered with EFV/FTC/TDF. No clinically relevant differences in the PK of ARVs were observed when administered with SOF/VEL. Study treatments were well tolerated, including no observed creatinine clearance changes during evaluation of TDF-containing regimens. Conclusions: SOF/VEL and ARV regimens including ATV, COBI, DRV, DTG, EVG, FTC, LPV, RAL, RPV, RTV, TAF, or TDF may be coadministered without dose adjustment. Use of SOF/VEL with EFV-containing regimens is not recommended due to an approximate 50% reduction in VEL exposure.


Subject(s)
Anti-HIV Agents/pharmacokinetics , Carbamates/pharmacokinetics , Heterocyclic Compounds, 4 or More Rings/pharmacokinetics , Sofosbuvir/pharmacokinetics , Adolescent , Adult , Anti-HIV Agents/adverse effects , Carbamates/adverse effects , Cross-Over Studies , Drug Interactions , Female , Healthy Volunteers , Heterocyclic Compounds, 4 or More Rings/adverse effects , Humans , Male , Middle Aged , Sofosbuvir/adverse effects , Tenofovir/adverse effects , Tenofovir/pharmacokinetics , Young Adult
8.
Expert Opin Drug Metab Toxicol ; 8(4): 467-85, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22394356

ABSTRACT

INTRODUCTION: Idiosyncratic drug reactions (IDRs) that involve the formation of toxic metabolites followed by covalent binding to cellular proteins often go undiscovered until after post-marketing. The goal of this article is to review the current status of IDRs, potential mechanisms and the challenges associated with predicting drug toxicity. AREAS COVERED: The authors review the metabolic pathways of five select classes of sulfur-containing drugs (captopril, troglitazone, tienilic acid, zileuton, methimazole and sudoxicam) suggesting that bioactivation plays a crucial role in the occurrence of IDRs. The reader will gain further awareness that the sulfur atom can propagate as the bioactivation site for the formation of reactive and conceivably toxic metabolites. As such, it is the body's capacity to detoxify these drug products that may determine whether IDRs occur. EXPERT OPINION: Incomplete understanding of mechanisms culminating in IDR occurrence represents a monumental impediment toward their abrogation. Moreover, current technology utilized to predict their manifestation (including structure-toxicity relationships) is not infallible and thus, development of novel tools and strategies is indispensible. In an attempt to streamline clinical development and drug approval processes, consortiums have been instated under the US FDA Critical Path Initiative. Collectively, these parameters along with the availability of validated biomarkers and new/updated regulatory guidance could positively influence the outcome of drug toxicity profiles and direct future drug development.


Subject(s)
Drug-Related Side Effects and Adverse Reactions , Sulfur Compounds/adverse effects , Animals , Biomarkers , Biotransformation/physiology , Humans , Metabolic Networks and Pathways , Pharmaceutical Preparations/metabolism , Pharmacokinetics , Structure-Activity Relationship , Sulfur Compounds/pharmacokinetics , United States , United States Food and Drug Administration
9.
Drug Metab Dispos ; 37(7): 1355-70, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19359406

ABSTRACT

Time-dependent inhibition (TDI) of cytochrome P450 (P450) enzymes caused by new molecular entities (NMEs) is of concern because such compounds can be responsible for clinically relevant drug-drug interactions (DDI). Although the biochemistry underlying mechanism-based inactivation (MBI) of P450 enzymes has been generally understood for several years, significant advances have been made only in the past few years regarding how in vitro time-dependent inhibition data can be used to understand and predict clinical DDI. In this article, a team of scientists from 16 pharmaceutical research organizations that are member companies of the Pharmaceutical Research and Manufacturers of America offer a discussion of the phenomenon of TDI with emphasis on the laboratory methods used in its measurement. Results of an anonymous survey regarding pharmaceutical industry practices and strategies around TDI are reported. Specific topics that still possess a high degree of uncertainty are raised, such as parameter estimates needed to make predictions of DDI magnitude from in vitro inactivation parameters. A description of follow-up mechanistic experiments that can be done to characterize TDI are described. A consensus recommendation regarding common practices to address TDI is included, the salient points of which include the use of a tiered approach wherein abbreviated assays are first used to determine whether NMEs demonstrate TDI or not, followed by more thorough inactivation studies for those that do to define the parameters needed for prediction of DDI.


Subject(s)
Cytochrome P-450 Enzyme System/metabolism , Drug Industry , Drug Interactions , Microsomes, Liver/metabolism , Aryl Hydrocarbon Hydroxylases/metabolism , Cytochrome P-450 CYP2B6 , Cytochrome P-450 CYP3A/metabolism , Drug Design , Glucuronosyltransferase , Humans , Microsomes, Liver/enzymology , Oxidoreductases, N-Demethylating/metabolism , Pharmaceutical Preparations/metabolism , Structure-Activity Relationship , Substrate Specificity , Time Factors
10.
Expert Opin Drug Deliv ; 2(5): 891-908, 2005 Sep.
Article in English | MEDLINE | ID: mdl-16296785

ABSTRACT

Ophthalmic drugs typically achieve < 10% ocular bioavailability. A drug applied to the surface of the eye may cross ocular-blood barriers where it may encounter metabolising enzymes and cellular transporters before it distributes to the site of action. Characterisation of ocular enzyme systems and cellular transporters and their respective substrate selectivity have provided new insight into the roles these proteins may play in ocular drug delivery and distribution. Altered metabolism and transport have been proposed to contribute to a number of ocular disease processes including inflammation, glaucoma, cataract, dry eye and neurodegeneration. As ocular enzyme and transport systems are better characterised, their properties become an integral consideration in drug design and development.


Subject(s)
Drug Delivery Systems , Eye/cytology , Eye/metabolism , Ophthalmic Solutions , Animals , Carrier Proteins/genetics , Carrier Proteins/metabolism , Eye/enzymology , Eye Diseases/drug therapy , Humans , Structure-Activity Relationship
11.
Invest Ophthalmol Vis Sci ; 46(12): 4697-706, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16303967

ABSTRACT

PURPOSE: Cytochrome P450 3A (CYP3A) is an enzyme of paramount importance to drug metabolism. The expression and activity of CYP3A, an enzyme responsible for active androgen clearance, was investigated in the rabbit lacrimal gland. METHODS: Analysis of CYP3A expression and activity was performed on lacrimal gland tissues obtained from naïve untreated and treated New Zealand White rabbits. For 5 days, treated rabbits received daily administration of vehicle or 0.1% or 1.0% dexamethasone, in the lower cul-de-sac of each eye. Changes in mRNA expression were monitored by real-time RT-PCR. Protein expression was confirmed by Western blot. Functional activity was measured by monitoring the metabolism of CYP3A probe substrates-namely, 7-benzyloxyquinoline (BQ) and [3H]testosterone. RESULTS: Cytochrome P450 heme protein was detected at a concentration of 44.6 picomoles/mg protein, along with its redox partner NADPH reductase and specifically CYP3A6 in the naïve rabbit lacrimal gland. Genes encoding CYP3A6, in addition to the pregnane-X-receptor (PXR) and P-glycoprotein (P-gp) were expressed in the untreated tissue. BQ dealkylation was measured in the naïve rabbit lacrimal gland at a rate of 14 +/- 7 picomoles/mg protein per minute. Changes in CYP3A6, P-gp, and androgen receptor mRNA expression levels were detected after dexamethasone treatment. In addition, dexamethasone treatment resulted in significant increases in BQ dealkylation and CYP3A6-mediated [3H]testosterone metabolism. Concomitant increases in CYP3A6-mediated hydroxylated testosterone metabolites were observed in the treated rabbits. Furthermore, ketoconazole, all-trans retinoic acid, and cyclosporine inhibited CYP3A6 mediated [3H]testosterone 6beta hydroxylation in a concentration-dependent manner, with IC50 ranging from 3.73 to 435 microM. CONCLUSIONS: The results demonstrate, for the first time, the expression and activity of CYP3A6 in the rabbit lacrimal gland. In addition, this pathway was shown to be subject to modulation by a commonly prescribed glucocorticoid and can be inhibited by known CYP3A inhibitors.


Subject(s)
Aryl Hydrocarbon Hydroxylases/genetics , Dexamethasone/pharmacology , Gene Expression Regulation, Enzymologic/physiology , Glucocorticoids/pharmacology , Lacrimal Apparatus/enzymology , Testosterone/metabolism , ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Animals , Aryl Hydrocarbon Hydroxylases/metabolism , Blotting, Western , Chromatography, High Pressure Liquid , Enzyme Inhibitors/pharmacology , Hydroxylation , Lacrimal Apparatus/drug effects , Male , Microsomes/enzymology , Pregnane X Receptor , Quinolines/metabolism , RNA, Messenger/metabolism , Rabbits , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Cytoplasmic and Nuclear/metabolism , Receptors, Steroid/genetics , Receptors, Steroid/metabolism , Reverse Transcriptase Polymerase Chain Reaction
12.
J Pharm Sci ; 94(10): 2246-55, 2005 Oct.
Article in English | MEDLINE | ID: mdl-16136553

ABSTRACT

Oral tazarotene, an acetylenic retinoid, is in clinical development for the treatment of psoriasis. The disposition and biotransformation of tazarotene were investigated in six healthy male volunteers, following a single oral administration of a 6 mg (100 microCi) dose of [14C]tazarotene, in a gelatin capsule. Blood levels of radioactivity peaked 2 h postdose and then rapidly declined. Total recovery of radioactivity was 89.2+/-8.0% of the administered dose, with 26.1+/-4.2% in urine and 63.0+/-7.0% in feces, within 7 days of dosing. Only tazarotenic acid, the principle active metabolite formed via esterase hydrolysis of tazarotene, was detected in blood. One major urinary oxidative metabolite, tazarotenic acid sulfoxide, accounted for 19.2+/-3.0% of the dose. The majority of radioactivity recovered in the feces was attributed to tazarotenic acid representing 46.9+/-9.9% of the dose and only 5.82+/-3.84% of dose was excreted as unchanged tazarotene. Thus following oral administration, tazarotene was rapidly absorbed and underwent extensive hydrolysis to tazarotenic acid, the major circulating species in the blood that was then excreted unchanged in feces. A smaller fraction of tazarotenic acid was further metabolized to an inactive sulfoxide that was excreted in the urine.


Subject(s)
Dermatologic Agents/pharmacokinetics , Nicotinic Acids/pharmacokinetics , Prodrugs/pharmacokinetics , Administration, Oral , Adolescent , Adult , Biotransformation , Carbon Radioisotopes , Dermatologic Agents/blood , Dermatologic Agents/urine , Feces/chemistry , Humans , Male , Middle Aged , Nicotinic Acids/blood , Nicotinic Acids/urine , Time Factors
13.
J Lipid Res ; 46(12): 2745-51, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16150817

ABSTRACT

Prostaglandin F2alpha 1-ethanolamide (prostamide F2alpha) is a potent ocular hypotensive agent in animals and represents a new class of fatty acid amide compounds. Accumulated evidence indicated that anandamide, an endogenous bioactive ligand for cannabinoid receptors, may serve as a common substrate to produce all prostamides, including prostamide F2alpha. After incubation of anandamide with cyclooxygenase 2 (COX-2), the reaction mixture was profiled by HPLC and an intermediate metabolite was discovered and characterized as a cyclic endoperoxide ethanolamide using HPLC-tandem mass spectrometry. Formation of prostamide F2alpha was also demonstrated when the intermediate metabolite was isolated and incubated with prostaglandin F synthase (PGF synthase). These results suggest that the biosynthesis of prostamide F2alpha proceeds in two consecutive steps: oxidation of anandamide to form an endoperoxide intermediate by COX-2, and reduction of the endoperoxide intermediate to form prostamide F2alpha by PGF synthase. This endoperoxide ethanolamide intermediate has been proposed as prostamide H2.


Subject(s)
Arachidonic Acids/metabolism , Cyclooxygenase 2/metabolism , Dinoprostone/analogs & derivatives , Hydroxyprostaglandin Dehydrogenases/metabolism , Arachidonic Acids/chemistry , Chromatography, High Pressure Liquid , Dinoprostone/chemistry , Dinoprostone/metabolism , Endocannabinoids , Humans , Mass Spectrometry , Molecular Structure , Polyunsaturated Alkamides
14.
J Lipid Res ; 45(4): 757-63, 2004 Apr.
Article in English | MEDLINE | ID: mdl-14729864

ABSTRACT

We investigated the formation of PGF(2alpha) 1-ethanolamide, PGE(2) 1-ethanolamide, and PGD(2) 1-ethanolamide (prostamides F(2alpha), E(2), and D(2), respectively) in liver, lung, kidney, and small intestine after a single intravenous bolus administration of 50 mg/kg of anandamide to normal and fatty acid amide hydrolase knockout (FAAH -/-) male mice. One group of three normal mice was not dosed (naïve) while another group of three normal mice received a bolus intravenous injection of 50 mg/kg of anandamide. Three FAAH -/- mice also received an intravenous injection of 50 mg/kg of anandamide. After 30 min, the lung, liver, kidney, and small intestine were harvested and processed by liquid-liquid extraction. The concentrations of prostamide F(2alpha), prostamide E(2), prostamide D(2), and anandamide were determined by HPLC-tandem mass spectrometry. Prostamide F(2alpha) was detected in tissues in FAAH -/- mice after administration of anandamide. Concentrations of anandamide, prostamide E(2), and prostamide D(2) in liver, kidney, lung, and small intestine were much higher in the anandamide-treated FAAH -/- mice than those of the anandamide-treated control mice. This report demonstrates that prostamides, including prostamide F(2alpha), were formed in vivo from anandamide, potentially by the cyclooxygenase-2 pathway when the competing FAAH pathway is lacking.


Subject(s)
Amidohydrolases/deficiency , Arachidonic Acids/pharmacokinetics , Dinoprostone/analogs & derivatives , Prostaglandins/biosynthesis , Amidohydrolases/metabolism , Animals , Arachidonic Acids/administration & dosage , Biotransformation , Chromatography, High Pressure Liquid , Cyclooxygenase 2 , Dinoprostone/analysis , Dinoprostone/biosynthesis , Endocannabinoids , Intestine, Small/chemistry , Isoenzymes/metabolism , Kidney/chemistry , Liver/chemistry , Lung/chemistry , Mass Spectrometry , Mice , Mice, Knockout , Polyunsaturated Alkamides , Prostaglandin-Endoperoxide Synthases/metabolism , Prostaglandins/analysis , Tissue Distribution
15.
Drug Metab Dispos ; 31(4): 476-81, 2003 Apr.
Article in English | MEDLINE | ID: mdl-12642475

ABSTRACT

Upon oral administration, tazarotene is rapidly converted to tazarotenic acid by esterases. The main circulating agent, tazarotenic acid is subsequently oxidized to the inactive sulfoxide metabolite. Therefore, alterations in the metabolic clearance of tazarotenic acid may have significant effects on its systemic exposure. The objective of this study was to identify the human liver microsomal enzymes responsible for the in vitro metabolism of tazarotenic acid. Tazarotenic acid was incubated with 1 mg/ml pooled human liver microsomes, in 100 mM potassium phosphate buffer (pH 7.4), at 37 degrees C, over a period of 30 min. The microsomal enzymes that may be involved in tazarotenic acid metabolism were identified through incubation with microsomes containing cDNA-expressed human microsomal isozymes. Chemical inhibition studies were then conducted to confirm the identity of the enzymes potentially involved in tazarotenic acid metabolism. Reversed-phase high performance liquid chromatography was used to quantify the sulfoxide metabolite, the major metabolite of tazarotenic acid. Upon incubation of tazarotenic acid with microsomes expressing CYP2C8, flavin-containing monooxygenase 1 (FMO1), or FMO3, marked formation of the sulfoxide metabolite was observed. The involvement of these isozymes in tazarotenic acid metabolism was further confirmed by inhibition of metabolite formation in pooled human liver microsomes by specific inhibitors of CYP2C8 or FMO. In conclusion, the in vitro metabolism of tazarotenic acid to its sulfoxide metabolite in human liver microsomes is mediated by CYP2C8 and FMO.


Subject(s)
Aryl Hydrocarbon Hydroxylases/metabolism , Nicotinic Acids/metabolism , Oxygenases/metabolism , Safrole/analogs & derivatives , Teratogens/metabolism , Angiogenesis Inhibitors/metabolism , Antithyroid Agents/metabolism , Aryl Hydrocarbon Hydroxylases/genetics , Chromatography, High Pressure Liquid/instrumentation , Chromatography, High Pressure Liquid/methods , Cytochrome P-450 CYP2C8 , Dose-Response Relationship, Drug , Humans , In Vitro Techniques , Inhibitory Concentration 50 , Mass Spectrometry/instrumentation , Mass Spectrometry/methods , Methimazole/metabolism , Microsomes, Liver , Nicotinic Acids/chemistry , Paclitaxel/metabolism , Safrole/metabolism , Teratogens/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...