Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 107
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Biomed Chromatogr ; 35(2): e4970, 2021 Feb.
Article in English | MEDLINE | ID: mdl-32840903

ABSTRACT

As hypoxia-inducible factor stabilizers (HIFs) can artificially enhance an athlete's erythropoiesis, the World Anti-Doping Agency prohibits their use at all times. Every urine sample for doping control analysis has to be evaluated for the presence of HIFs and therefore sensitive methods that allow high sample throughput are needed. Samples suspicious for the presence of HIFs need to be confirmed following the identification criteria established by the World Anti-Doping Agency. Previous work has shown the advantages of using turbulent flow online solid-phase extraction (SPE) procedures to reduce matrix effects and retention time shifts. Furthermore, the use of online SPE allows for automation and high sample throughput. Both an initial testing procedure (ITP) and a confirmation method were developed and validated, using online SPE liquid chromatography-tandem mass spectrometry (LC-MS/MS), with limits of detection between 0.1 ng/ml (or possibly lower) and 4 ng/ml (or higher for GSK360a) and limits of identification between 0.1 ng/ml (or possibly lower) and 1.17 ng/ml. The ITP only takes 6.5 min per sample. To the best of our knowledge, these are the first ITP and confirmation methods that include more than three HIFs without the need for manual sample preparation.


Subject(s)
Doping in Sports , Hypoxia-Inducible Factor 1/chemistry , Pyrazoles/urine , Substance Abuse Detection/methods , Triazoles/urine , Chromatography, Liquid/methods , Glycine/analogs & derivatives , Glycine/chemistry , Glycine/urine , Humans , Isoquinolines/chemistry , Isoquinolines/urine , Limit of Detection , Linear Models , Pyrazoles/chemistry , Reproducibility of Results , Solid Phase Extraction/methods , Tandem Mass Spectrometry/methods , Triazoles/chemistry
2.
Biomed Chromatogr ; 35(6): e5075, 2021 Jun.
Article in English | MEDLINE | ID: mdl-33458843

ABSTRACT

Analytical methods to determine the potential misuse of the ghrelin mimetics capromorelin (CP-424,391), macimorelin (macrilen, EP-01572) and tabimorelin (NN703) in sports were developed. Therefore, different extraction strategies, i.e. solid-phase extraction, protein precipitation, as well as a "dilute-and-inject" approach, from urine and EDTA-plasma were assessed and comprehensive in vitro/in vivo experiments were conducted, enabling the identification of reliable target analytes by means of high resolution mass spectrometry. The drugs' biotransformation led to the preliminary identification of 51 metabolites of capromorelin, 12 metabolites of macimorelin and 13 metabolites of tabimorelin. Seven major metabolites detected in rat urine samples collected post-administration of 0.5-1.0 mg of a single oral dose underwent in-depth characterization, facilitating their implementation into future confirmatory test methods. In particular, two macimorelin metabolites exhibiting considerable abundances in post-administration rat urine samples were detected, which might contribute to an improved sensitivity, specificity, and detection window in case of human sports drug testing programs. Further, the intact drugs were implemented into World Anti-Doping Agency-compliant initial testing (limits of detection 0.02-0.60 ng/ml) and confirmation procedures (limits of identification 0.18-0.89 ng/ml) for human urine and blood matrices. The obtained results allow extension of the test spectrum of doping agents in multitarget screening assays for growth hormone-releasing factors from human urine.


Subject(s)
Dipeptides , Doping in Sports , Indoles , Piperidines , Pyrazoles , Tryptophan/analogs & derivatives , Animals , Biomarkers/metabolism , Biomarkers/urine , Chromatography, Liquid/methods , Dipeptides/metabolism , Dipeptides/urine , Female , Ghrelin , Humans , Indoles/metabolism , Indoles/urine , Limit of Detection , Male , Piperidines/metabolism , Piperidines/urine , Pyrazoles/metabolism , Pyrazoles/urine , Rats , Reproducibility of Results , Solid Phase Extraction , Tandem Mass Spectrometry/methods , Tryptophan/metabolism , Tryptophan/urine
3.
Clin Chem Lab Med ; 54(2): 275-83, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26167981

ABSTRACT

BACKGROUND: The utility of measuring non-vitamin K antagonist oral anticoagulants (NOACs) in plasma, serum and urine samples and with the point-of-care test (POCT) on urine samples should be analysed in an international laboratory study. METHODS: The study was performed to determine the inter-laboratory variance of data from two chromogenic assays each for the NOACs rivaroxaban, apixaban and dabigatran, and to analyse the sensitivity and specificity of the POCT assays for factor Xa- and thrombin inhibitors. Plasma, serum and urine samples were taken from six patients in each group on treatment with a NOAC. RESULTS: The inter-laboratory variances, which can be identified best by the coefficient of variation, ranged from 46% to 59% for apixaban, 63% to 73% for rivaroxaban and 39% to 104% for dabigatran using plasma, serum or urine samples and two chromogenic assays for each NOAC. The concentrations were about 20% higher in serum compared to plasma samples for apixaban and rivaroxaban, and 60% lower for dabigatran. The concentration in urine samples was five-fold (apixaban), 15-fold (rivaroxaban) and 50-fold (dabigatran) higher. Sensitivity and specificity of POCT for apixaban, rivaroxaban, and dabigatran were all >94%. CONCLUSIONS: The inter-laboratory study showed the feasibility of measurement of apixaban, rivaroxaban, and dabigatran in plasma, serum and urine samples of patients on treatment. Dabigatran was determined at far lower levels in serum compared to plasma samples. Concentrations of NOACs in urine were much higher compared to plasma. The POCT was highly sensitive and specific for all three NOACs.


Subject(s)
Anticoagulants/analysis , Dabigatran/analysis , Enzyme Assays , Factor Xa Inhibitors/analysis , Pyrazoles/analysis , Pyridones/analysis , Rivaroxaban/analysis , Anticoagulants/blood , Anticoagulants/urine , Chromogenic Compounds/chemistry , Dabigatran/blood , Dabigatran/urine , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/urine , Humans , Laboratories/standards , Point-of-Care Systems , Pyrazoles/blood , Pyrazoles/urine , Pyridones/blood , Pyridones/urine , Rivaroxaban/blood , Rivaroxaban/urine
4.
Drug Metab Dispos ; 43(2): 289-97, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25488930

ABSTRACT

The absorption, metabolism, and excretion of ibrutinib were investigated in healthy men after administration of a single oral dose of 140 mg of ¹4C-labeled ibrutinib. The mean (S.D.) cumulative excretion of radioactivity of the dose was 7.8% (1.4%) in urine and 80.6% (3.1%) in feces with <1% excreted as parent ibrutinib. Only oxidative metabolites and very limited parent compound were detected in feces, and this indicated that ibrutinib was completely absorbed from the gastrointestinal tract. Metabolism occurred via three major pathways (hydroxylation of the phenyl (M35), opening of the piperidine (M25 and M34), and epoxidation of the ethylene on the acryloyl moiety with further hydrolysis to dihydrodiol (PCI-45227, and M37). Additional metabolites were formed by combinations of the primary metabolic pathways or by further metabolism. In blood and plasma, a rapid initial decline in radioactivity was observed along with long terminal elimination half-life for total radioactivity. The maximum concentration (Cmax) and area under the concentration-time curve (AUC) for total radioactivity were higher in plasma compared with blood. The main circulating entities in blood and plasma were M21 (sulfate conjugate of a monooxidized metabolite on phenoxyphenyl), M25, M34, M37 (PCI-45227), and ibrutinib. At Cmax of radioactivity, 12% of total radioactivity was accounted for by covalent binding in human plasma. More than 50% of total plasma radioactivity was attributed to covalently bound material from 8 hours onward; as a result, covalent binding accounted for 38% and 51% of total radioactivity AUC(0-24 h) and AUC(0-72 h), respectively. No effect of CYP2D6 genotype was observed on ibrutinib metabolism. Ibrutinib was well-tolerated by healthy participants.


Subject(s)
Antineoplastic Agents/pharmacokinetics , Intestinal Absorption , Protein Kinase Inhibitors/pharmacokinetics , Protein-Tyrosine Kinases/antagonists & inhibitors , Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Adenine/analogs & derivatives , Adenine/analysis , Adenine/blood , Adenine/urine , Administration, Oral , Adult , Agammaglobulinaemia Tyrosine Kinase , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/adverse effects , Antineoplastic Agents/analysis , Biotransformation , Carbon Radioisotopes , Feces/chemistry , Half-Life , Humans , Hydrolysis , Hydroxylation , Intestinal Elimination , Male , Metabolic Clearance Rate , Middle Aged , Oxidation-Reduction , Piperidines , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/analysis , Protein-Tyrosine Kinases/metabolism , Pyrazoles/administration & dosage , Pyrazoles/adverse effects , Pyrazoles/analysis , Pyrazoles/blood , Pyrazoles/urine , Pyrimidines/administration & dosage , Pyrimidines/adverse effects , Pyrimidines/analysis , Renal Elimination
5.
Environ Sci Technol ; 49(16): 10038-47, 2015 Aug 18.
Article in English | MEDLINE | ID: mdl-26196357

ABSTRACT

Phenylpyrazole insecticides such as fipronil have been used as replacements for organophosphates. The wide application of fipronil raises concern about environmental contamination and risk for fish, birds, and other nontargeted beings as well as human health. A sensitive, competitive indirect heterologous enzyme-linked immunosorbent assay (ELISA) was developed. Antibodies with different specificities to fipronil and its metabolites were produced. Two ELISAs having IC50 values of 0.58 ± 0.06 and 2.6 ± 0.4 ng/mL were developed. Design of different haptens and coating antigens resulted in two assays with distinct cross-reactivity patterns for structurally related compounds: 96, 38, and 101% versus 39, 1.4, and 25% for fipronil-sulfide, fipronil-detrifluoromethylsulfonyl, and fipronil-desulfinyl, respectively. Performance of the immunoassays was demonstrated by a recovery study from spiked water and human serum and urine matrices, giving recovery values in the range of 85-111% for different concentrations. The assays demonstrated good correlation in fipronil recovery with conventional LC-MS/MS analysis. The generic assay 2265 has the sensitivity to measure fipronil and its analogs in serum at levels relevant for exposure monitoring. The assays were used to analyze human urine samples obtained from exposure studies and serum samples from rats treated with a fipronil-containing diet.


Subject(s)
Immunoassay/methods , Insecticides/analysis , Pyrazoles/analysis , Animals , Antibodies/immunology , Antigens/metabolism , Chromatography, Liquid , Cross Reactions/immunology , Enzyme-Linked Immunosorbent Assay/methods , Haptens/chemistry , Haptens/immunology , Humans , Immune Sera/immunology , Insecticides/chemistry , Male , Pyrazoles/blood , Pyrazoles/chemistry , Pyrazoles/urine , Rats , Reproducibility of Results , Tandem Mass Spectrometry , Water/chemistry
6.
Drug Metab Dispos ; 39(2): 256-64, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21071521

ABSTRACT

Apixaban, a potent and highly selective factor Xa inhibitor, is currently under development for treatment of arterial and venous thrombotic diseases. The distribution, metabolism, and elimination of [(14)C]apixaban were investigated in male, female, pregnant, and lactating rats after single oral doses. Tissue distribution of radioactivity in rats was measured using quantitative whole-body autoradiography. After a single oral administration, radioactivity distributed quickly in rats with C(max) at 1 h for most tissues. The elimination t(1/2) of radioactivity in blood was 1.7 to 4.2 h. The blood area under the plasma concentration-time curve of radioactivity was similar between male and female rats and was slightly higher in pregnant rats and lower in lactating rats. The radioactivity concentration in tissues involved in elimination was greater than that in blood with the highest concentration in the gastrointestinal tract, liver, and urinary bladder/contents and lowest level in brains. In pregnant rats, the whole-body autoradiogram showed that low levels of radioactivity were present in fetal blood, liver, and kidney and were much lower than the radioactivity in the respective maternal organs. The fecal route was the major pathway (74% of dose), and the urinary route was the minor pathway (14%) for apixaban elimination. After single oral doses of [(14)C]apixaban to lactating rats, apixaban exhibited extensive lacteal excretion with apixaban as the major component. In summary, tissue distribution of apixaban in rats was extensive but with limited transfer to fetal and brain tissues and extensive secretion into rat milk with the parent drug as the major component. Milk excretion could account for 10% of apixaban dose, which was comparable to urinary elimination in rats. Tissue distribution and drug excretion of apixaban are consistent with those for a moderately permeable drug that is a substrate for P-glycoprotein and breast cancer resistance protein efflux transporters.


Subject(s)
Fibrinolytic Agents/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Administration, Oral , Animals , Carbon Radioisotopes , Feces/chemistry , Female , Fibrinolytic Agents/blood , Fibrinolytic Agents/urine , Male , Maternal-Fetal Exchange , Metabolic Clearance Rate , Milk/chemistry , Pregnancy , Pyrazoles/blood , Pyrazoles/urine , Pyridones/blood , Pyridones/urine , Rats , Rats, Sprague-Dawley , Tissue Distribution
7.
Drug Metab Dispos ; 39(9): 1734-46, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21646437

ABSTRACT

The metabolism and disposition of eltrombopag, the first-in-class small molecule human thrombopoietin receptor agonist, were studied in six healthy men after a single oral administration of a solution dose of [(14)C]eltrombopag (75 mg, 100 µCi). Eltrombopag was well tolerated. The drug was quickly absorbed and was the predominant circulating component in plasma (accounting for 63% of the total plasma radioactivity). A mono-oxygenation metabolite (M1) and acyl glucuronides (M2) of eltrombopag were minor circulating components. The predominant route of elimination of radioactivity was fecal (58.9%). Feces contained approximately 20% of dose as glutathione-related conjugates (M5, M6, and M7) and another 20% as unchanged eltrombopag. The glutathione conjugates were probably detoxification products of a p-imine methide intermediate formed by metabolism of M1, which arises through cytochrome P450-dependent processes. Low levels of covalently bound drug-related intermediates to plasma proteins, which could result from the reaction of the imine methide or acyl glucuronide conjugates with proteins, were detected. The bound material contributes to the longer plasma elimination half-life of radioactivity. Renal elimination of conjugates of hydrazine cleavage metabolites (mostly as M3 and M4) accounted for 31% of the radiodose, with no unchanged eltrombopag detected in urine.


Subject(s)
Benzoates/pharmacokinetics , Hydrazines/pharmacokinetics , Pyrazoles/pharmacokinetics , Receptors, Thrombopoietin/agonists , Administration, Oral , Adult , Benzoates/blood , Benzoates/metabolism , Benzoates/urine , Biotransformation , Blood Proteins/metabolism , Cytochrome P-450 Enzyme System/metabolism , Feces/chemistry , Glucuronides/blood , Glutathione/metabolism , Half-Life , Humans , Hydrazines/blood , Hydrazines/metabolism , Hydrazines/urine , Male , Middle Aged , Protein Binding , Pyrazoles/blood , Pyrazoles/metabolism , Pyrazoles/urine , Receptors, Thrombopoietin/metabolism
8.
Drug Metab Dispos ; 39(3): 383-93, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21098644

ABSTRACT

The objective of this study was to assess the physiologically based pharmacokinetic (PBPK) model for predicting plasma concentration-time profiles of orally available cMet kinase inhibitors, (R)-3-[1-(2,6-dichloro-3-fluoro-phenyl)-ethoxy]-5-(1-piperidin-4-yl-1H-pyrazol-4-yl)-pyridin-2-ylamine (PF02341066) and 2-[4-(3-quinolin-6-ylmethyl-3H-[1,2,3]triazolo[4,5-b]pyrazin-5-yl)-pyrazol-1-yl]-ethanol (PF04217903), in humans. The prediction accuracy of pharmacokinetics (PK) by PBPK modeling was compared with that of a traditional one-compartment PK model based on allometric scaling. The predicted clearance values from allometric scaling with the correction for the interspecies differences in protein binding were used as a representative comparison, which showed more accurate PK prediction in humans than the other methods. Overall PBPK modeling provided better prediction of the area under the plasma concentration-time curves for both PF02341066 (1.2-fold error) and PF04217903 (1.3-fold error) compared with the one-compartment PK model (1.8- and 1.9-fold errors, respectively). Of more importance, the simulated plasma concentration-time profiles of PF02341066 and PF04217903 by PBPK modeling seemed to be consistent with the observed profiles showing multiexponential declines, resulting in more accurate prediction of the apparent half-lives (t(1/2)): the observed and predicted t(1/2) values were, respectively, 10 and 12 h for PF02341066 and 6.6 and 6.3 h for PF04217903. The predicted t(1/2) values by the one-compartment PK model were 17 h for PF02341066 and 1.9 h for PF04217903. Therefore, PBPK modeling has the potential to be more useful and reliable for the PK prediction of PF02341066 and PF04217903 in humans than the traditional one-compartment PK model. In summary, the present study has shown examples to indicate that the PBPK model can be used to predict PK profiles in humans.


Subject(s)
Drug Evaluation, Preclinical/methods , Models, Molecular , Protein Kinase Inhibitors/pharmacokinetics , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Administration, Oral , Adult , Animals , Biological Availability , Cells, Cultured , Crizotinib , Dogs , Half-Life , Hepatocytes/metabolism , Humans , Macaca fascicularis , Male , Metabolic Clearance Rate , Microsomes, Liver/metabolism , Middle Aged , Protein Kinase Inhibitors/administration & dosage , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/urine , Pyrazines/administration & dosage , Pyrazines/blood , Pyrazines/pharmacokinetics , Pyrazines/urine , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrazoles/pharmacokinetics , Pyrazoles/urine , Pyridines/administration & dosage , Pyridines/blood , Pyridines/pharmacokinetics , Pyridines/urine , Rats , Rats, Sprague-Dawley , Triazoles/administration & dosage , Triazoles/blood , Triazoles/pharmacokinetics , Triazoles/urine , Young Adult
10.
Drug Metab Dispos ; 38(11): 2023-31, 2010 Nov.
Article in English | MEDLINE | ID: mdl-20699411

ABSTRACT

The metabolism, excretion, and pharmacokinetics of 3-(4-(7H-pyrrolo[2,3-d]pyrimidin-4-yl)-1H-pyrazol-1-yl)-3-cyclopentylpropanenitrile (INCB018424), a potent, selective inhibitor of Janus tyrosine kinase1/2 and the first investigational drug of its class in phase III studies for the treatment of myelofibrosis, were investigated in healthy human subjects given a single oral 25-mg dose of [(14)C]INCB018424 as an oral solution. INCB018424 and total radioactivity were absorbed rapidly (mean time to reach the maximal drug concentration <1 h), declining in a monophasic or biphasic fashion (mean t(1/2) of 2.32 and 5.81 h, respectively). Recovery of administered radioactivity was fairly rapid (>70% within 24 h postdose) with 74 and 22% recovered in urine and feces, respectively. Parent compound was the predominant entity in the circulation, representing 58 to 74% of the total radioactivity up to 6 h postdose, indicating that the overall circulating metabolite burden was low (<50% of parent). Two metabolite peaks in plasma (M18 and a peak containing M16/M27, both hydroxylations on the cyclopentyl moiety) were identified as major (30 and 14% of parent based on area under the curve from 0 to 24 h). The exposures of other circulating INCB018424-related peaks were <10% of parent, consisting of mono- and dihydroxylated metabolites. The profiles in urine and feces consisted of hydroxyl and oxo metabolites and subsequent glucuronide conjugates with parent drug accounting for <1% of the excreted dose, strongly suggesting that after an oral dose, INCB018424 was >95% absorbed. In healthy subjects administered daily oral doses of unlabeled INCB018424, there were minimal differences in parent and metabolite concentrations between day 1 and day 10, indicating a lack of accumulation of parent or metabolites between single and multiple dosing.


Subject(s)
Enzyme Inhibitors/pharmacokinetics , Janus Kinase 1/antagonists & inhibitors , Janus Kinase 2/antagonists & inhibitors , Pyrazoles/pharmacokinetics , Chromatography, High Pressure Liquid , Dose-Response Relationship, Drug , Double-Blind Method , Enzyme Inhibitors/blood , Enzyme Inhibitors/metabolism , Enzyme Inhibitors/urine , Feces/chemistry , Female , Humans , Male , Metabolic Clearance Rate , Molecular Structure , Nitriles , Pyrazoles/blood , Pyrazoles/metabolism , Pyrazoles/urine , Pyrimidines , Tandem Mass Spectrometry
11.
Drug Metab Dispos ; 38(5): 789-800, 2010 May.
Article in English | MEDLINE | ID: mdl-20124396

ABSTRACT

Lersivirine [UK-453,061, 5-((3,5-diethyl-1-(2-hydroxyethyl)(3,5-14C2)-1H-pyrazol-4-yl)oxy)benzene-1,3-dicarbonitrile] is a next-generation non-nucleoside reverse transcriptase inhibitor, with a unique binding interaction within the reverse transcriptase binding pocket. Lersivirine has shown antiviral activity and is well tolerated in HIV-infected and healthy subjects. This open-label, Phase I study investigated the absorption, metabolism, and excretion of a single oral 500-mg dose of [14C]lersivirine (parent drug) and characterized the plasma, fecal, and urinary radioactivity of lersivirine and its metabolites in four healthy male volunteers. Plasma C(max) for total radioactivity and unchanged lersivirine typically occurred between 0.5 and 3 h postdose. The majority of radioactivity was excreted in urine (approximately 80%) with the remainder excreted in the feces (approximately 20%). The blood/plasma ratio of total drug-derived radioactivity [area under the plasma concentration-time profile from time zero extrapolated to infinite time (AUC(inf))] was 0.48, indicating that radioactive material was distributed predominantly into plasma. Lersivirine was extensively metabolized, primarily by UDP glucuronosyltransferase- and cytochrome P450-dependent pathways, with 22 metabolites being identified in this study. Analysis of precipitated plasma revealed that the lersivirine-glucuronide conjugate was the major circulating component (45% of total radioactivity), whereas unchanged lersivirine represented 13% of total plasma radioactivity. In vitro studies showed that UGT2B7 and CYP3A4 are responsible for the majority of lersivirine metabolism in humans.


Subject(s)
Nitriles/metabolism , Pyrazoles/metabolism , Reverse Transcriptase Inhibitors/metabolism , Adult , Anti-HIV Agents/adverse effects , Anti-HIV Agents/metabolism , Anti-HIV Agents/pharmacokinetics , Anti-HIV Agents/urine , Area Under Curve , Biocatalysis , Cytochrome P-450 CYP3A/metabolism , Dealkylation , Feces/chemistry , Glucuronidase/metabolism , Glucuronides/analysis , Glucuronides/metabolism , Glucuronosyltransferase/metabolism , Humans , Hydrolysis , Hydroxylation , Kinetics , Male , Microsomes, Liver/enzymology , Middle Aged , Molecular Structure , Nitriles/adverse effects , Nitriles/pharmacokinetics , Nitriles/urine , Oxidation-Reduction , Pyrazoles/adverse effects , Pyrazoles/pharmacokinetics , Pyrazoles/urine , Recombinant Proteins/metabolism , Reverse Transcriptase Inhibitors/adverse effects , Reverse Transcriptase Inhibitors/pharmacokinetics , Reverse Transcriptase Inhibitors/urine , Sulfates/metabolism , Tandem Mass Spectrometry
12.
Pharmacol Res Perspect ; 8(2): e00579, 2020 04.
Article in English | MEDLINE | ID: mdl-32314550

ABSTRACT

The pharmacokinetics (PK), metabolism, excretion, mass balance, and tissue distribution of [14 C]praliciguat were evaluated following oral administration of a 3-mg/kg dose in Sprague-Dawley rats and in a quantitative whole-body autoradiography (QWBA) study conducted in male Long-Evans rats. Plasma Tmax was 1 hour and the t1/2 of total plasma radioactivity was 23.7 hours. Unchanged praliciguat accounted for 87.4%, and a minor metabolite (N-dealkylated-praliciguat) accounted for 7.6% of the total radioactivity in plasma through 48 hours (AUC0-48 ). Tissues with the highest exposure ratios relative to plasma were liver, intestines, adrenal gland, and adipose, and those with the lowest values were seminal vesicle, blood, CNS tissues, lens of the eye, and bone. Most of the [14 C]praliciguat-derived radioactivity was excreted within 48 hours after oral administration. Mean cumulative recovery of the administered radioactivity in urine and feces over 168 hours was 3.7% and 95.7%, respectively. Unchanged praliciguat was not quantifiable in urine or bile of cannulated rats; however, based on the total radioactivity in these fluids, a minimum of approximately 82% of the orally administered dose was absorbed. [14 C]Praliciguat was metabolized via oxidative and glucuronidation pathways and the most abundant metabolites recovered in bile were praliciguat-glucuronide and hydroxy-praliciguat-glucuronide. These results indicate that praliciguat had rapid absorption, high bioavailability, extensive tissue distribution, and elimination primarily via hepatic metabolism.


Subject(s)
Pyrazoles/pharmacokinetics , Pyrimidines/pharmacokinetics , Animals , Bile/metabolism , Feces/chemistry , Male , Pyrazoles/blood , Pyrazoles/urine , Pyrimidines/blood , Pyrimidines/urine , Rats, Long-Evans , Rats, Sprague-Dawley , Soluble Guanylyl Cyclase , Tissue Distribution
13.
Basic Clin Pharmacol Toxicol ; 127(3): 221-233, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32248614

ABSTRACT

The absorption, distribution, metabolism and excretion of molidustat were investigated in healthy male participants. In study 1, a mass balance study, radiolabelled molidustat 25 mg (3.57 MBq) was administered as an oral solution (n = 4). Following rapid absorption, molidustat-related radioactivity was predominantly distributed in plasma rather than in red blood cells. The total recovery of the administered radioactivity was 97.0%, which was mainly excreted renally (90.7%). Metabolite M-1, produced by N-glucuronidation, was the dominant component in plasma (80.2% of the area under the concentration-time curve for total radioactivity) and was primarily excreted via urine (~85% of dose). Only minor amounts of unchanged molidustat were excreted in urine (~4%) and faeces (~6%). Study 2 investigated the absolute bioavailability and pharmacodynamics of molidustat (part 1, n = 12; part 2, n = 16). Orally administered molidustat immediate release tablets had an absolute bioavailability of 59%. Following intravenous administration (1, 5 and 25 mg), total body clearance of molidustat was 28.7-34.5 L/h and volume of distribution at steady state was 39.3-50.0 L. All doses of molidustat transiently elevated endogenous erythropoietin levels, irrespective of the route of administration. Molidustat was considered safe and well tolerated at the administered doses.


Subject(s)
Pyrazoles/metabolism , Pyrazoles/pharmacokinetics , Triazoles/metabolism , Triazoles/pharmacokinetics , Administration, Oral , Adult , Biological Availability , Drug Evaluation, Preclinical , Healthy Volunteers , Humans , Infusions, Intravenous , Male , Metabolic Clearance Rate , Middle Aged , Pyrazoles/blood , Pyrazoles/urine , Tissue Distribution , Triazoles/blood , Triazoles/urine
14.
Eur J Drug Metab Pharmacokinet ; 45(1): 101-111, 2020 Feb.
Article in English | MEDLINE | ID: mdl-31673875

ABSTRACT

BACKGROUND AND OBJECTIVES: Erdafitinib, an oral selective pan-fibroblast growth factor receptor (FGFR) kinase inhibitor, is primarily metabolized by cytochrome P450 (CYP) 2C9 and 3A4. The aim of this phase 1 study was to assess the pharmacokinetics and safety of erdafitinib in healthy participants when coadministered with fluconazole (moderate CYP2C9 and CYP3A inhibitor), and itraconazole (a strong CYP3A4 and P-glycoprotein inhibitor). The effect of CYP2C9 genotype variants (*1/*1, *1/*2, *1/*3) on the pharmacokinetics of erdafitinib was also investigated. METHODS: In this open-label, parallel-group, single-center study, eligible healthy adults were randomized by CYP2C9 genotype to receive Treatment A (single oral dose of erdafitinib 4 mg) on day 1, Treatment B (fluconazole 400 mg/day orally) on days 1-11, or Treatment C (itraconazole 200 mg/day orally) on days 1-11. Healthy adults randomized to Treatment B and C received a single oral 4-mg dose of erdafitinib on day 5. The pharmacokinetic parameters, including mean maximum plasma concentration (Cmax), area under the curve (AUC) from time 0 to 168 h (AUC168h), AUC from time 0 to the last quantifiable concentration (AUClast), and AUC from time 0 to infinity (AUC∞) were calculated from individual plasma concentration-time data using standard non-compartmental methods. RESULTS: Coadministration of erdafitinib with fluconazole increased Cmax of erdafitinib by approximately 21%, AUC168h by 38%, AUClast by 49%, and AUC∞ by 48% while coadministration with itraconazole resulted in no change in erdafitinib Cmax and increased AUC168h by 20%, AUClast by 33% and AUC∞ by 34%. Erdafitinib exposure was comparable between participants with CYP2C9 *1/*2 or *1/*3 and with wild-type CYP2C9 genotype. The ratio of total amount of erdafitinib excreted in the urine (inhibited to non-inhibited) was 1.09, the ratio of total amount of excreted metabolite M6 was 1.21, and the ratio of the metabolite to parent ratio in the urine was 1.11, when coadministration of erdafitinib with itraconazole was compared with single-dose erdafitinib. Treatment-emergent adverse events (TEAEs) were generally Grade 1 or 2 in severity; the most commonly reported TEAE was headache. No safety concerns were identified with single-dose erdafitinib when administered alone and in combination with fluconazole or itraconazole in healthy adults. CONCLUSION: Coadministration of fluconazole or itraconazole or other moderate/strong CYP2C9 or CYP3A4 inhibitors may increase exposure to erdafitinib in healthy adults and thus may warrant erdafitinib dose reduction or use of alternative concomitant medications with no or minimal CYP2C9 or CYP3A4 inhibition potential. TRIAL REGISTRATION: ClinicalTrials.gov identifier number: NCT03135106.


Subject(s)
Cytochrome P-450 Enzyme Inhibitors/pharmacology , Drug Interactions , Fluconazole/pharmacology , Itraconazole/pharmacology , Protein Kinase Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Quinoxalines/pharmacokinetics , Adult , Area Under Curve , Cytochrome P-450 CYP2C9/genetics , Drug Combinations , Female , Healthy Volunteers , Humans , Male , Middle Aged , Protein Kinase Inhibitors/adverse effects , Protein Kinase Inhibitors/blood , Protein Kinase Inhibitors/urine , Pyrazoles/adverse effects , Pyrazoles/blood , Pyrazoles/urine , Quinoxalines/adverse effects , Quinoxalines/blood , Quinoxalines/urine , Receptors, Fibroblast Growth Factor/antagonists & inhibitors
15.
Thromb Haemost ; 120(1): 132-140, 2020 Jan.
Article in English | MEDLINE | ID: mdl-31705521

ABSTRACT

The rapid determination of the presence of direct oral anticoagulants (DOACs) in a patient remains a major challenge in emergency medicine and for rapid medical treatment decisions. All DOACs are excreted into urine. A sensitive and specific point-of-care test has been developed to determine whether they are present in patient urine samples. This prospective multicenter study aimed to demonstrate at least 95% correct positive and negative predictive results for factor Xa and thrombin inhibitors in urine samples using DOAC Dipstick pads compared with liquid chromatography-tandem mass spectrometry (LC-MS/MS) (NCT03182829). Nine hundred and fourteen subjects were included and 880 were evaluated per protocol (factor Xa inhibitors apixaban, edoxaban, and rivaroxaban: n = 451, thrombin inhibitor dabigatran: n = 429) at 18 centers. The sensitivity, specificity, accuracy, and predictive values and agreement between methods for determination of factor Xa inhibitors were at least noninferior to 95% with a 0.5% margin and of thrombin inhibitor superior to 97.5%. These results were compared with LC-MS/MS results in the intention-to-analyze cohort (all p < 0.05). The receiver operating curve showed c-values of 0.989 (factor Xa inhibitors) and 0.995 (thrombin inhibitor). Visual evaluation of the factor Xa and thrombin inhibitor pads was not different between centers. Qualitative determination of both types of DOACs was accurate using the DOAC Dipstick compared with using LC-MS/MS. The high predictive values may impact laboratory and clinical decision-making processes.


Subject(s)
Antithrombins/urine , Dabigatran/urine , Factor Xa Inhibitors/urine , Pyrazoles/urine , Pyridines/urine , Pyridones/urine , Rivaroxaban/urine , Thiazoles/urine , Aged , Aged, 80 and over , Chromatography, Liquid , Factor Xa/metabolism , Female , Humans , Male , Middle Aged , Point-of-Care Systems , Predictive Value of Tests , ROC Curve , Reproducibility of Results , Sensitivity and Specificity , Tandem Mass Spectrometry
16.
Anal Chem ; 81(12): 4734-41, 2009 Jun 15.
Article in English | MEDLINE | ID: mdl-19462990

ABSTRACT

Nonsteroidal anti-inflammatory drugs (NSAIDs) are commonly used to treat rheumatoid arthritis, osteoarthritis, acute pain, and fever. However, NSAIDs have side effects that include gastric erosions, ulceration, bleeding, and perforation, etc. Selective cyclooxygenase (COX)-2 inhibitors have been developed to avoid the adverse drug reaction of traditional NSAIDs. The COX-2 inhibitors have a different mechanism of action from nonselective COX inhibitors. In this study, pattern recognition analysis of the (1)H nuclear magnetic resonance (NMR) spectra of urine was performed to develop surrogate biomarkers related to the gastrointestinal (GI) damage induced by NSAIDs in rats. Urine was collected for 5 h after administering the following NSAIDs at high doses: celecoxib (133 mg kg(-1), p.o.), a COX-2-selective inhibitor; and indomethacin (25 mg kg(-1), p.o.) or ibuprofen (800 mg kg(-1), p.o.), nonselective COX inhibitors. The urine was analyzed using 600 M (1)H NMR for spectral binning and targeted profiling. The level of gastric damage in each animal was also determined. Indomethacin and ibuprofen caused severe gastric damage, but no lesions were observed in the celecoxib-treated rats. The (1)H NMR urine spectra were divided into spectral bins (0.04 ppm) for global profiling, and 36 endogenous metabolites were assigned for targeted profiling. Multivariate data analyses were carried out to recognize the spectral pattern of endogenous metabolites related to NSAIDs using partial least-squares discrimination analysis (PLS-DA). There were different clusterings of (1)H NMR spectra according to the gastric damage scores in global profiling. In targeted profiling, a few endogenous metabolites of allantoin, taurine, and dimethylamine were selected as putative biomarkers for the gastric damage induced by NSAIDs. The results of global and targeted profilings suggest that the gastric damage induced by NSAIDs can be screened in the preclinical stage of drug development using a current metabolomics study. In addition, the putative biomarkers might also be useful for predicting the risk of adverse effects caused by NSAIDs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/toxicity , Magnetic Resonance Spectroscopy/methods , Metabolomics/methods , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/urine , Celecoxib , Cyclooxygenase 2 Inhibitors/metabolism , Cyclooxygenase 2 Inhibitors/toxicity , Cyclooxygenase 2 Inhibitors/urine , Ibuprofen/metabolism , Ibuprofen/toxicity , Ibuprofen/urine , Indomethacin/metabolism , Indomethacin/pharmacology , Indomethacin/toxicity , Least-Squares Analysis , Male , Pattern Recognition, Automated , Predictive Value of Tests , Pyrazoles/metabolism , Pyrazoles/toxicity , Pyrazoles/urine , Rats , Rats, Sprague-Dawley , Stomach/pathology , Sulfonamides/metabolism , Sulfonamides/toxicity , Sulfonamides/urine
17.
Drug Metab Dispos ; 37(8): 1738-48, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19420130

ABSTRACT

The metabolism and disposition of [(14)C]apixaban, a potent, reversible, and direct inhibitor of coagulation factor Xa, were investigated in mice, rats, rabbits, dogs, and humans after a single oral administration and in incubations with hepatocytes. In plasma, the parent compound was the major circulating component in mice, rats, dogs, and humans. O-Demethyl apixaban sulfate (M1) represented approximately 25% of the parent area under the time curve in human plasma. This sulfate metabolite was present, but in lower amounts relative to the parent, in plasma from mice, rats, and dogs. Rabbits showed a plasma metabolite profile distinct from that of other species with apixaban as a minor component and M2 (O-demethyl apixaban) and M14 (O-demethyl apixaban glucuronide) as prominent components. The fecal route was a major elimination pathway, accounting for >54% of the dose in animals and >46% in humans. The urinary route accounted for <15% of the dose in animals and 25 to 28% in humans. Apixaban was the major component in feces of every species and in urine of all species except rabbit. M1 and M2 were common prominent metabolites in urine and feces of all species as well as in bile of rats and humans. In vivo metabolite profiles showed quantitative differences between species and from in vitro metabolite profiles, but all human metabolites were found in animal species. After intravenous administration of [(14)C]apixaban to bile duct-cannulated rats, the significant portion (approximately 22%) of the dose was recovered as parent drug in the feces, suggesting direct excretion of the drug from gastrointestinal tracts of rats. Overall, apixaban was effectively eliminated via multiple elimination pathways in animals and humans, including oxidative metabolism, and direct renal and intestinal excretion.


Subject(s)
Fibrinolytic Agents/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyridones/pharmacokinetics , Administration, Oral , Adolescent , Adult , Animals , Area Under Curve , Bile/metabolism , Biotransformation , Carbon Radioisotopes , Dealkylation , Dogs , Factor Xa Inhibitors , Feces/chemistry , Female , Fibrinolytic Agents/administration & dosage , Fibrinolytic Agents/blood , Fibrinolytic Agents/urine , Glucuronides/metabolism , Hepatocytes/metabolism , Humans , Infusions, Intravenous , Macaca fascicularis , Male , Metabolomics/methods , Mice , Mice, Inbred ICR , Middle Aged , Oxidation-Reduction , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrazoles/urine , Pyridones/administration & dosage , Pyridones/blood , Pyridones/urine , Rabbits , Rats , Rats, Sprague-Dawley , Species Specificity , Young Adult
18.
Chem Res Toxicol ; 22(9): 1603-12, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19697924

ABSTRACT

Compound A (Cmpd A) was previously reported to form p-chlorophenyl isocyanate (CPIC), which was trapped by GSH to yield S- (N- [p-chlorophenyl] carbamoyl) glutathione adduct (SCPG) in the presence of human liver microsomes. In this study, P450 3A4 and 2C9 were demonstrated to be the enzymes mediating the activation of Cmpd A to CPIC in human liver microsomes based on inhibitory and correlation studies. Enzyme kinetics studies indicated that P450 3A4 was the primary enzyme involved in the activation of Cmpd A. In silico P450 3A4 active site docking of Cmpd A exhibited a low energy pose that orientated the pyrazole ring proximate to the heme iron atom, in which the distance between the C-3 and potential activated oxygen species was shown to be 3.4 A. Quantum molecular calculations showed that the electron density on C-3 was relatively higher than those on C-4 and C-5. These measurements suggested that the C-3 of Cmpd A was the preferred site of oxidation and hence predisposed Cmpd A in forming CPIC as previously proposed. The in silico prediction was corroborated by studies with the C-3 substituted analogue (methyl at C-3), which showed minimal conversion to CPIC in human liver microsomes. These results demonstrated a pivotal role for P450 3A4 in bioactivating Cmpd A by oxidizing at C-3 of the pyrazoline, hence facilitating the CPIC formation. Evidence of the bioactivation to CPIC in vivo was obtained by liquid chromatography-mass spectrometry (LC/MS) analysis of urine samples from human subjects administered a structural analogue of Cmpd A. The presence of S-(N-[p-chlorophenyl] carbamoyl) N-acetyl l-cysteine (SCPAC) as well as p-chlorophenyl aniline (CPA) was unequivocally demonstrated in the urine samples. The chemical scaffold, 4,5-dihydropyrazole-1-carboxylic acid-[(4-chlorophenyl)-amide], was demonstrated to possess potential metabolic liability in forming a reactive intermediate, CPIC, in humans. Bioactivation to CPIC may cause undesirable side effects through its reactivity and subsequent conversion to CPA, an established rodent carcinogen.


Subject(s)
Chlorobenzenes/metabolism , Cytochrome P-450 CYP3A/metabolism , Isocyanates/metabolism , Microsomes, Liver/metabolism , Pyrazoles/metabolism , Pyrones/metabolism , Catalytic Domain , Chlorobenzenes/chemistry , Chlorobenzenes/urine , Chromatography, High Pressure Liquid , Computer Simulation , Cytochrome P-450 CYP3A Inhibitors , Enzyme Inhibitors/chemistry , Enzyme Inhibitors/pharmacology , Humans , Isocyanates/chemistry , Kinetics , Pyrazoles/chemistry , Pyrazoles/urine , Pyrones/chemistry , Pyrones/urine , Quantum Theory , Spectrometry, Mass, Electrospray Ionization
19.
Eur J Drug Metab Pharmacokinet ; 44(6): 747-759, 2019 Dec.
Article in English | MEDLINE | ID: mdl-31571146

ABSTRACT

BACKGROUND AND OBJECTIVES: Darolutamide is a novel androgen receptor (AR) antagonist approved for the treatment of nonmetastatic castration-resistant prostate cancer (nmCRPC). Accordingly, the drug-drug interaction (DDI) potential of darolutamide was investigated in both nonclinical and clinical studies. METHODS: In vitro studies were performed to determine the potential for darolutamide to be a substrate, inducer or inhibitor for cytochrome P450 (CYP) isoforms, other metabolizing enzymes and drug transporters. A phase I drug-interaction study in healthy volunteers evaluated the impact of co-administering rifampicin [CYP3A4 and P-glycoprotein (P-gp) inducer] and itraconazole [CYP3A4, P-gp and breast cancer resistance protein (BCRP) inhibitor] on the pharmacokinetics of darolutamide. Two further phase I studies assessed the impact of co-administering oral darolutamide on the pharmacokinetics of midazolam (sensitive CYP3A4 substrate) and dabigatran etexilate (P-gp substrate) and the impact on the pharmacokinetics of co-administered rosuvastatin [a substrate for BCRP, organic anion-transporting polypeptide (OATP)1B1, OATP1B3 and organic anion transporter (OAT)3]. RESULTS: In vitro, darolutamide was predominantly metabolized via oxidative biotransformation catalyzed by CYP3A4 and was identified as a substrate for P-gp and BCRP. The enzymatic activity of nine CYP isoforms was not inhibited or slightly inhibited in vitro with darolutamide, and a rank order and mechanistic static assessment indicated that risk of clinically relevant DDIs via CYP inhibition is very low. In vitro, darolutamide exhibited no relevant induction of CYP1A2 or CYP2B6 activity. Inhibition of BCRP-, P-gp-, OAT3-, MATE1-, MATE2-K-, OATP1B1- and OATP1B3-mediated transport was observed in vitro. Phase I data showed that darolutamide exposure increased 1.75-fold with co-administered itraconazole and decreased by 72% with rifampicin. Co-administration of darolutamide with CYP3A4/P-gp substrates showed no effect or only minor effects. Rosuvastatin exposure increased 5.2-fold with darolutamide because of BCRP and probably also OATPB1/OATPB3 inhibition. CONCLUSIONS: Darolutamide has a low potential for clinically relevant DDIs with drugs that are substrates for CYP or P-gp; increased exposure of BCRP and probably OATP substrates was the main interaction of note.


Subject(s)
Cytochrome P-450 Enzyme System/drug effects , Drug Interactions , Pyrazoles/pharmacology , Pyrazoles/pharmacokinetics , Aged , Cells, Cultured , Cytochrome P-450 CYP3A Inducers/pharmacology , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Dabigatran/pharmacokinetics , Enzyme Induction/drug effects , Female , Humans , Itraconazole/pharmacology , Male , Membrane Transport Proteins/drug effects , Microsomes, Liver/drug effects , Midazolam/pharmacokinetics , Middle Aged , Pyrazoles/blood , Pyrazoles/urine , Rifampin/pharmacology , Rosuvastatin Calcium/pharmacokinetics
20.
Clin Pharmacokinet ; 58(9): 1155-1163, 2019 09.
Article in English | MEDLINE | ID: mdl-30828771

ABSTRACT

OBJECTIVES: The aim of this study was to prove the suitability of simultaneously administered microdoses of the factor Xa inhibitors (FXaIs) rivaroxaban, apixaban and edoxaban (100 µg in total). To evaluate drug-drug interactions, the impact of ketoconazole, a known strong inhibitor of cytochrome P450 3A4 and P-glycoprotein, was studied. METHODS: In a crossover clinical trial, 18 healthy volunteers were randomized to the two treatments using microdoses of rivaroxaban, apixaban and edoxaban alone and when coadministered with ketoconazole. Plasma and urine concentrations of microdosed apixaban, edoxaban and rivaroxaban were quantified using a validated ultra-performance liquid chromatography-tandem mass spectrometry assay with a lower limit of quantification of 2.5 pg/ml. RESULTS: The microdosed FXaI cocktail showed similar pharmacokinetic parameters compared with published data, using normal therapeutic doses of each FXaI. Ketoconazole significantly increased exposure, with geometric mean AUC ratios of 1.90 (apixaban), 2.35 (edoxaban) and 2.27 (rivaroxaban). CONCLUSION: The microdosed FXaI cocktail approach was able to precisely predict the drug interaction with ketoconazole. This is the first study that has been conducted to evaluate drug-drug interactions with a drug class, and the low administered doses also allow evaluation in vulnerable target populations. STUDY PROTOCOL: EudraCT 2016-003024-23.


Subject(s)
Drug Interactions , Factor Xa Inhibitors/pharmacokinetics , Pyrazoles/pharmacokinetics , Pyridines/pharmacokinetics , Pyridones/pharmacokinetics , Rivaroxaban/pharmacokinetics , Thiazoles/pharmacokinetics , Administration, Oral , Adolescent , Adult , Atrial Fibrillation/drug therapy , Case-Control Studies , Chromatography, Liquid/instrumentation , Cross-Over Studies , Cytochrome P-450 CYP3A Inhibitors/pharmacokinetics , Cytochrome P-450 CYP3A Inhibitors/pharmacology , Factor Xa Inhibitors/administration & dosage , Factor Xa Inhibitors/blood , Factor Xa Inhibitors/urine , Female , Humans , Ketoconazole/pharmacokinetics , Ketoconazole/pharmacology , Male , Middle Aged , Pyrazoles/administration & dosage , Pyrazoles/blood , Pyrazoles/urine , Pyridines/administration & dosage , Pyridines/blood , Pyridines/urine , Pyridones/administration & dosage , Pyridones/blood , Pyridones/urine , Rivaroxaban/administration & dosage , Rivaroxaban/blood , Rivaroxaban/urine , Tandem Mass Spectrometry/methods , Thiazoles/administration & dosage , Thiazoles/blood , Thiazoles/urine , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL