Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 254
Filter
1.
Acta Pharmacol Sin ; 42(9): 1535-1546, 2021 Sep.
Article in English | MEDLINE | ID: mdl-33244163

ABSTRACT

Vicagrel, a novel irreversible P2Y12 receptor inhibitor, is undergoing phase III trials for the treatment of acute coronary syndromes in China. In this study, we evaluated the pharmacokinetics, mass balance, and metabolism of vicagrel in six healthy male Chinese subjects after a single oral dose of 20 mg [14C]vicagrel (120 µCi). Vicagrel absorption was fast (Tmax = 0.625 h), and the mean t1/2 of vicagrel-related components was ~38.0 h in both plasma and blood. The blood-to-plasma radioactivity AUCinf ratio was 0.55, suggesting preferential distribution of drug-related material in plasma. At 168 h after oral administration, the mean cumulative excreted radioactivity was 96.71% of the dose, including 68.03% in urine and 28.67% in feces. A total of 22 metabolites were identified, and the parent vicagrel was not detected in plasma, urine, or feces. The most important metabolic spot of vicagrel was on the thiophene ring. In plasma pretreated with the derivatization reagent, M9-2, which is a methylated metabolite after thiophene ring opening, was the predominant drug-related component, accounting for 39.43% of the radioactivity in pooled AUC0-8 h plasma. M4, a mono-oxidation metabolite upon ring-opening, was the most abundant metabolite in urine, accounting for 16.25% of the dose, followed by M3-1, accounting for 12.59% of the dose. By comparison, M21 was the major metabolite in feces, accounting for 6.81% of the dose. Overall, renal elimination plays a crucial role in vicagrel disposition, and the thiophene ring is the predominant metabolic site.


Subject(s)
Phenylacetates/metabolism , Phenylacetates/pharmacokinetics , Purinergic P2Y Receptor Antagonists/metabolism , Purinergic P2Y Receptor Antagonists/pharmacokinetics , Thiophenes/metabolism , Thiophenes/pharmacokinetics , Administration, Oral , Adult , Clopidogrel , Humans , Male , Phenylacetates/blood , Phenylacetates/chemistry , Purinergic P2Y Receptor Antagonists/blood , Purinergic P2Y Receptor Antagonists/chemistry , Thiophenes/blood , Thiophenes/chemistry
2.
BMC Vet Res ; 16(1): 355, 2020 Sep 25.
Article in English | MEDLINE | ID: mdl-32988403

ABSTRACT

BACKGROUND: Robenacoxib (Onsior™) is a non-steroidal anti-inflammatory drug developed for canine and feline use for the control of pain and inflammation. It is available as both tablets and solution for injection. The objective of this study was to evaluate the safety of the interchangeable use of commercially available robenacoxib formulations when administered to cats orally using 6 mg tablets and subcutaneously using a solution for injection containing 20 mg/mL. Thirty-four naïve healthy 4-month old cats were enrolled in this 37-day study and were randomized to four groups (three robenacoxib and one control). One robenacoxib group received the maximum recommended dose (MRD) rate of each formulation, while the other two received two and three times this dose rate. The cats underwent three 10-day treatment cycles comprised of seven days of once daily oral administration followed by three days of subcutaneous administration. The third cycle was followed by an additional seven days of oral treatment. The control group received oral empty gelatin capsules or subcutaneous saline injections. Assessment of safety was based on general health observations, clinical observations, physical, ophthalmic, electrocardiographic and neurological examinations, clinical pathology evaluations, food consumption, body weight, and macroscopic and microscopic examinations. Blood samples were collected for toxicokinetic evaluation. RESULTS: Blood concentrations of robenacoxib confirmed systemic exposure of all treated cats. All cats were in good health through study termination and there were no serious adverse events during the study. There were no changes in body weight, food consumption, ophthalmic, physical or neurological examinations during the study. Treatment-related abnormalities were of low occurrence at all doses and included injection site changes (transient edema with minimal or mild, subacute/chronic inflammation histologically) and prolongation of the QT interval. These findings were consistent with previously observed findings in studies with robenacoxib administered separately orally or subcutaneously in cats. Thus, there were no adverse effects that could be attributed specifically to the interchangeable use of oral and injectable robenacoxib. CONCLUSIONS: This 37-day laboratory study supports the safety of interchanging robenacoxib injection at a daily dose of 2 mg/kg with robenacoxib tablets at a daily dose of 1 mg/kg, or vice versa.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Diphenylamine/analogs & derivatives , Phenylacetates/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Cats , Diphenylamine/administration & dosage , Diphenylamine/adverse effects , Diphenylamine/blood , Diphenylamine/pharmacokinetics , Electrocardiography/drug effects , Electrocardiography/veterinary , Female , Injections, Subcutaneous/veterinary , Male , Phenylacetates/adverse effects , Phenylacetates/blood , Phenylacetates/pharmacokinetics , Tablets/administration & dosage
3.
Molecules ; 25(6)2020 Mar 11.
Article in English | MEDLINE | ID: mdl-32168894

ABSTRACT

Rubusoside is a natural sweetener and the active component of Rubus suavissimus. The preventive and therapeutic effect of rubusoside on high-fat diet-induced (HFD) serum metabolite changes in golden hamsters was analyzed by 1H-NMR metabolomics to explore the underlying mechanism of lipid metabolism regulation. 1H-NMR serum metabolomics analyses revealed a disturbed amino acid-, sugar-, fat-, and energy metabolism in HFD animals. Animals supplemented with rubusoside can partly reverse the metabolism disorders induced by high-fat diet and exerted good anti-hypertriglyceridemia effect by intervening in some major metabolic pathways, involving amino acid metabolism, synthesis of ketone bodies, as well as choline and 4-hydroxyphenylacetate metabolism. This study indicates that rubusoside can interfere with and normalize high-fat diet-induced metabolic changes in serum and could provide a theoretical basis to establish rubusoside as a potentially therapeutic tool able to revert or prevent lipid metabolism disorders.


Subject(s)
Diet, High-Fat/adverse effects , Diterpenes, Kaurane/pharmacology , Energy Metabolism/drug effects , Glucosides/pharmacology , Hyperlipidemias/prevention & control , Obesity/prevention & control , Rubus/chemistry , Amino Acids/blood , Animals , Carbohydrate Metabolism/drug effects , Choline/blood , Hyperlipidemias/blood , Hyperlipidemias/etiology , Hyperlipidemias/pathology , Lipid Metabolism , Magnetic Resonance Spectroscopy , Mesocricetus , Metabolomics/methods , Obesity/blood , Obesity/etiology , Obesity/pathology , Phenylacetates/blood
4.
Mol Genet Metab ; 125(3): 251-257, 2018 11.
Article in English | MEDLINE | ID: mdl-30217721

ABSTRACT

INTRODUCTION: Glycerol phenylbutyrate (GPB) is approved in the US and EU for the chronic management of patients ≥2 months of age with urea cycle disorders (UCDs) who cannot be managed by dietary protein restriction and/or amino acid supplementation alone. GPB is a pre-prodrug, hydrolyzed by lipases to phenylbutyric acid (PBA) that upon absorption is beta-oxidized to the active nitrogen scavenger phenylacetic acid (PAA), which is conjugated to glutamine (PAGN) and excreted as urinary PAGN (UPAGN). Pharmacokinetics (PK) of GPB were examined to see if hydrolysis is impaired in very young patients who may lack lipase activity. METHODS: Patients 2 months to <2 years of age with UCDs from two open label studies (n = 17, median age 10 months) predominantly on stable doses of nitrogen scavengers (n = 14) were switched to GPB. Primary assessments included traditional plasma PK analyses of PBA, PAA, and PAGN, using noncompartmental methods with WinNonlin™. UPAGN was collected periodically throughout the study up to 12 months. RESULTS: PBA, PAA and PAGN rapidly appeared in plasma after GPB dosing, demonstrating evidence of GPB cleavage with subsequent PBA absorption. Median concentrations of PBA, PAA and PAGN did not increase over time and were similar to or lower than the values observed in older UCD patients. The median PAA/PAGN ratio was well below one over time, demonstrating that conjugation of PAA with glutamine to form PAGN did not reach saturation. Covariate analyses indicated that age did not influence the PK parameters, with body surface area (BSA) being the most significant covariate, reinforcing current BSA based dosing recommendations as seen in older patients. CONCLUSION: These observations demonstrate that UCD patients aged 2 months to <2 years have sufficient lipase activity to adequately convert the pre-prodrug GPB to PBA. PBA is then converted to its active moiety (PAA) providing successful nitrogen scavenging even in very young children.


Subject(s)
Glycerol/analogs & derivatives , Lipase/blood , Phenylbutyrates/administration & dosage , Prodrugs/administration & dosage , Urea Cycle Disorders, Inborn/drug therapy , Child , Child, Preschool , Female , Glutamine/blood , Glycerol/administration & dosage , Glycerol/blood , Glycerol/pharmacokinetics , Humans , Infant , Male , Nitrogen/blood , Nitrogen/metabolism , Phenylacetates/blood , Phenylbutyrates/blood , Phenylbutyrates/pharmacokinetics , Prodrugs/pharmacokinetics , Urea Cycle Disorders, Inborn/blood , Urea Cycle Disorders, Inborn/pathology
5.
J Cardiovasc Pharmacol ; 72(5): 252-258, 2018 11.
Article in English | MEDLINE | ID: mdl-30358688

ABSTRACT

Vicagrel, a novel acetate analogue of clopidogrel, exerts more potent antiplatelet effect than clopidogrel in rodents. Relevant evidence indicated that aspirin and vicagrel are the drug substrate for carboxylesterase 2. Accordingly, it is deduced that concomitant use of aspirin could attenuate the bioactivation of and platelet response to vicagrel. To clarify whether there could be such an important drug-drug interaction, the differences in both the formation of vicagrel active metabolite H4 and the inhibition of adenosine diphosphate-induced platelet aggregation by vicagrel were measured and compared between mice treated with vicagrel alone or in combination with aspirin. The plasma H4 concentration was determined by liquid chromatography-tandem mass spectrometry, and the inhibition of platelet aggregation by vicagrel was assessed by whole-blood platelet aggregation. Compared with vicagrel (2.5 mg·kg) alone, concurrent use of aspirin (5, 10, or 20 mg·kg) significantly decreased systemic exposure of H4, an average of 38% and 41% decrease in Cmax and AUC0-∞ in mice when in combination with aspirin at 10 mg·kg, respectively. Furthermore, concomitant use of aspirin (10 mg·kg) and vicagrel (2.5 mg·kg) resulted in an average of 66% reduction in the inhibition of adenosine diphosphate-induced platelet aggregation by vicagrel. We conclude that aspirin significantly attenuates the formation of vicagrel active metabolite H4 and platelet response to vicagrel in mice, and that such an important drug-drug interaction would appear in clinical settings if vicagrel is taken with aspirin concomitantly when marketed in the future.


Subject(s)
Aspirin/pharmacology , Blood Platelets/drug effects , Phenylacetates/pharmacology , Platelet Aggregation Inhibitors/pharmacology , Platelet Aggregation/drug effects , Thiophenes/pharmacology , Activation, Metabolic , Animals , Aspirin/metabolism , Blood Platelets/metabolism , Carboxylesterase , Carboxylic Ester Hydrolases/metabolism , Chromatography, Liquid , Drug Interactions , Male , Mice, Inbred C57BL , Phenylacetates/blood , Phenylacetates/pharmacokinetics , Platelet Aggregation Inhibitors/blood , Platelet Aggregation Inhibitors/pharmacokinetics , Platelet Function Tests , Tandem Mass Spectrometry , Thiophenes/blood , Thiophenes/pharmacokinetics
6.
BMC Vet Res ; 14(1): 242, 2018 Aug 17.
Article in English | MEDLINE | ID: mdl-30119677

ABSTRACT

BACKGROUND: Robenacoxib is a non-steroidal anti-inflammatory drug available for canine and feline use for the control of pain and inflammation marketed as Onsior™. The aim of this target animal safety study was to evaluate the 6-month safety profile of oral robenacoxib administration. It was a randomized, negative-controlled, parallel group study. Thirty-two healthy, young, experimentally naïve, purebred Beagle dogs were administered 0 (sham control, Group 1), 2, 6, and 10 mg/kg robenacoxib (corresponding to the upper end of the dosage range [1X, Group 2] and multiples thereof [3X and 5X, Group 3 and 4]), orally once daily for 6 months. Assessment of safety included general health and clinical observations, physical, neurological, ophthalmological and electrocardiographic examinations, gross and histopathological examinations and clinical pathology evaluations. Blood samples were collected for toxicokinetic assessment of robenacoxib. RESULTS: No serious adverse events were reported. When compared with control, no treatment effect was observed for body weight, feed or water consumption, clinical pathology, urinalysis and fecal examination parameters. There were no treatment-related changes in stifle joint tissues and microscopic/histopathology examinations of all tissues/organs were normal. Salivation and soft feces were noted in all groups but observed more frequently in the treated groups as compared with control. On Day 178, increased buccal mucosal bleeding times were observed in two treated animals (Group 3 and 4) and one dog in Group 4 displayed a retinal change. Decreased hopping and conscious proprioception was noted in four treated dogs. One dog in Group 2 had ventricular premature complexes. Post-mortem changes included mild, red foci on the cecum in one dog (Group 3) and minimal duodenal discoloration in one dog (Group 4), with no corresponding histological findings in either dog. Ovarian weights were decreased in females from Group 3 and 4 with no gross or histological changes in the ovaries. Blood concentrations of robenacoxib confirmed systemic exposure of treated dogs. Exposure increased with increasing doses and there were no accumulation of robenacoxib in blood. CONCLUSIONS: Robenacoxib was well tolerated at doses from 2 to 10 mg/kg/day and this 6-month study supports the safe use of Onsior™ (robenacoxib) tablets in dogs for the intended dosing regimen.


Subject(s)
Diphenylamine/analogs & derivatives , Dogs , Phenylacetates/administration & dosage , Phenylacetates/adverse effects , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Diphenylamine/administration & dosage , Diphenylamine/adverse effects , Diphenylamine/blood , Dose-Response Relationship, Drug , Female , Male , Organ Size/drug effects , Ovary/drug effects , Phenylacetates/blood , Tablets
7.
J Proteome Res ; 16(7): 2516-2526, 2017 07 07.
Article in English | MEDLINE | ID: mdl-28585834

ABSTRACT

Flavan-3-ols and methylxanthines have potential beneficial effects on human health including reducing cardiovascular risk. We performed a randomized controlled crossover intervention trial to assess the acute effects of consumption of flavan-3-ol-enriched dark chocolate, compared with standard dark chocolate and white chocolate, on the human metabolome. We assessed the metabolome in urine and blood plasma samples collected before and at 2 and 6 h after consumption of chocolates in 42 healthy volunteers using a nontargeted metabolomics approach. Plasma samples were assessed and showed differentiation between time points with no further separation among the three chocolate treatments. Multivariate statistics applied to urine samples could readily separate the postprandial time points and distinguish between the treatments. Most of the markers responsible for the multivariate discrimination between the chocolates were of dietary origin. Interestingly, small but significant level changes were also observed for a subset of endogenous metabolites. 1H NMR revealed that flavan-3-ol-enriched dark chocolate and standard dark chocolate reduced urinary levels of creatinine, lactate, some amino acids, and related degradation products and increased the levels of pyruvate and 4-hydroxyphenylacetate, a phenolic compound of bacterial origin. This study demonstrates that an acute chocolate intervention can significantly affect human metabolism.


Subject(s)
Chocolate/analysis , Flavonoids/administration & dosage , Metabolome/physiology , Phytochemicals/administration & dosage , Amino Acids/blood , Amino Acids/urine , Creatinine/blood , Creatinine/urine , Cross-Over Studies , Female , Flavonoids/blood , Flavonoids/urine , Humans , Lactic Acid/blood , Lactic Acid/urine , Male , Metabolomics/methods , Phenylacetates/blood , Phenylacetates/urine , Phytochemicals/blood , Phytochemicals/urine , Postprandial Period , Pyruvic Acid/blood , Pyruvic Acid/urine , Sex Factors
8.
Mol Genet Metab ; 122(3): 39-45, 2017 11.
Article in English | MEDLINE | ID: mdl-28888854

ABSTRACT

Urea cycle disorders (UCDs) are genetic conditions characterized by nitrogen accumulation in the form of ammonia and caused by defects in the enzymes required to convert ammonia to urea for excretion. UCDs include a spectrum of enzyme deficiencies, namely n-acetylglutamate synthase deficiency (NAGS), carbamoyl phosphate synthetase I deficiency (CPS1), ornithine transcarbamylase deficiency (OTC), argininosuccinate lyase deficiency (ASL), citrullinemia type I (ASS1), and argininemia (ARG). Currently, sodium phenylbutyrate and glycerol phenylbutyrate are primary medications used to treat patients with UCDs, and long-term monitoring of these compounds is critical for preventing drug toxic levels. Therefore, a fast and simple ultra-performance liquid chromatography (UPLC-MS/MS) method was developed and validated for quantification of phenylbutyrate (PB), phenylacetate (PA), and phenylacetylglutamine (PAG) in plasma and urine. The separation of all three analytes was achieved in 2min, and the limits of detection were <0.04µg/ml. Intra-precision and inter-precision were <8.5% and 4% at two quality control concentrations, respectively. Average recoveries for all compounds ranged from 100% to 106%. With the developed assay, a strong correlation between PA and the PA/PAG ratio and an inverse correlation between PA/PAG ratio and plasma glutamine were observed in 35 patients with confirmed UCDs. Moreover, all individuals with a ratio ≥0.6 had plasma glutamine levels<1000µmol/l. Our data suggest that a PA/PAG ratio in the range of 0.6-1.5 will result in a plasma glutamine level<1000µmol/l without reaching toxic levels of PA.


Subject(s)
Chromatography, Liquid/methods , Glutamine/analogs & derivatives , Glutamine/blood , Phenylacetates/metabolism , Phenylbutyrates/blood , Phenylbutyrates/metabolism , Ammonia/metabolism , Argininosuccinic Aciduria/physiopathology , Female , Glutamine/metabolism , Glutamine/urine , Glycerol/analogs & derivatives , Glycerol/therapeutic use , Humans , Limit of Detection , Male , Middle Aged , Ornithine Carbamoyltransferase Deficiency Disease/physiopathology , Phenylacetates/blood , Phenylacetates/urine , Phenylbutyrates/therapeutic use , Phenylbutyrates/urine , Tandem Mass Spectrometry , Urea/metabolism , Urea Cycle Disorders, Inborn/blood
9.
BMC Vet Res ; 13(1): 359, 2017 Nov 28.
Article in English | MEDLINE | ID: mdl-29179750

ABSTRACT

BACKGROUND: Robenacoxib (Onsior™) is a non-steroidal anti-inflammatory drug developed for canine and feline use for the control of pain and inflammation. It is available as both tablets and solution for injection. The objective of this safety study was to investigate the interchangeable use of two robenacoxib formulations in dogs using a novel study design alternating between oral tablets and subcutaneous injections. Thirty-two naïve healthy 4-month dogs were enrolled in this 88-day study and were randomized among four groups to be untreated or to receive robenacoxib at the highest recommended or elevated dose rates. The dogs were administered three 20-day treatment cycles each separated by a 14-day washout period. Each 20-day cycle was comprised of 10 days of once daily oral administration, 3 days of subcutaneous administration, followed by further 7 days of oral administration (Groups 2 to 4). The control group (Group 1) received oral empty gelatin capsules or subcutaneous saline injections. Assessment of safety was based on general health observations, clinical observations, physical and neurological examinations including ophthalmological examinations, electrocardiographic examinations and clinical pathology evaluations, food and water consumption, body weight, and macroscopic and microscopic examinations. Blood samples were collected for pharmacokinetic evaluation. RESULTS: Blood concentrations of robenacoxib confirmed systemic exposure of all treated dogs. All dogs were in good health through study termination and there were no serious adverse events during the course of the study. No changes in body weight, food consumption, ophthalmic, neurological examinations, electrocardiograms, buccal mucosal blood times, clinical pathology or organ weight were attributable to robenacoxib formulation administration. Primary treatment-related abnormalities were of low incidence at all doses. They were confined to macroscopic and microscopic changes observed locally at the subcutaneous injection sites and microscopic findings within the gastrointestinal tract. These findings were as expected based on previous studies with robenacoxib solution for injection alone and the known properties of this class of compound and mode of administration. There were no adverse effects which could be attributed specifically to the interchangeable use of oral and injectable robenacoxib. CONCLUSIONS: Alternating regimens of robenacoxib tablets and solution for injection were well tolerated in healthy young dogs.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Diphenylamine/analogs & derivatives , Phenylacetates/administration & dosage , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Body Weight/drug effects , Diphenylamine/administration & dosage , Diphenylamine/adverse effects , Diphenylamine/blood , Diphenylamine/pharmacokinetics , Dogs , Drug Administration Schedule/veterinary , Eating/drug effects , Electrocardiography/drug effects , Female , Injections, Subcutaneous/veterinary , Male , Phenylacetates/adverse effects , Phenylacetates/blood , Phenylacetates/pharmacokinetics , Tablets
10.
J Vet Pharmacol Ther ; 40(2): 148-157, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27493016

ABSTRACT

The objective of the study was to establish the dose-response relationship for robenacoxib, a selective cyclooxygenase (COX)-2 inhibitor, in a urate crystal model of acute synovitis. In a randomized partial Latin square design trial, 12 beagle dogs were administered orally single doses of robenacoxib (0.5, 1, 2, 4 and 8 mg/kg), placebo and the positive control meloxicam (0.1 mg/kg), 3 h after injection of sodium urate crystals into a stifle joint. Dogs were assessed for weight bearing on a force plate and by subjective clinical orthopaedic observations. Robenacoxib produced dose-dependent improvement in weight bearing, and decreased pain on palpation and joint swelling, over the dose range 0.5-2 mg/kg with no further increase in effect over the range 2-8 mg/kg. For weight bearing on the force plate, the ED50 of robenacoxib was 0.6-0.8 mg/kg. The onset of action and time to peak effect of robenacoxib were faster (respectively, 2-2.5 h and 3-5 h) than for meloxicam (respectively, 3 h and 6 h). Robenacoxib significantly inhibited COX-2 at all doses, with dose-related activity. Robenacoxib did not inhibit COX-1 over the dose range 0.5-4 mg/kg, but produced transient inhibition at 8 mg/kg. In conclusion, oral administration of robenacoxib over the dose range 0.5-8 mg/kg demonstrated significant analgesic and anti-inflammatory efficacy in dogs.


Subject(s)
Diphenylamine/analogs & derivatives , Dog Diseases/chemically induced , Phenylacetates/therapeutic use , Stifle/drug effects , Synovitis/veterinary , Uric Acid/toxicity , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/therapeutic use , Cyclooxygenase 1/metabolism , Cyclooxygenase 2/metabolism , Cyclooxygenase Inhibitors/blood , Cyclooxygenase Inhibitors/pharmacokinetics , Cyclooxygenase Inhibitors/therapeutic use , Diphenylamine/blood , Diphenylamine/pharmacokinetics , Diphenylamine/therapeutic use , Dog Diseases/drug therapy , Dogs , Lameness, Animal , Phenylacetates/blood , Phenylacetates/pharmacokinetics
11.
Pharm Res ; 31(1): 238-51, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24037619

ABSTRACT

PURPOSE: To investigate the contributions of intestine and plasma to the presystemic bioconversion of vicagrel, and track its subsequent bioconversion to 2-oxo-clopidogrel in vivo and in vitro to rationalize the design of vicagrel, an acetate analogue of clopidogrel. METHODS: The concentration-time profiles of 2-oxo-clopidogrel and active metabolite (AM) in presystem and circulation system was determined in the cannulated rats. Also, the rat intestinal S9 and human intestinal microsomes were conducted to examine the formation of 2-oxo-clopidogrel and AM. Meanwhile, the esterases in plasma and intestinal fractions responsible for the bioconversion of vicagrel to 2-oxo-clopidogrel were screened by the esterase inhibition and recombinant esterases. RESULTS: The intestine was responsible for the formation of 2-oxo-clopidogrel and AM in vivo and in vitro, where carboxylesterases 2 (CE2) contributed greatly to the vicagrel cleavage during absorption. Other related esterases in plasma were paraoxonases (PON), carboxylesterases 1 (CE1) and butyrylcholine esterases (BChE). CONCLUSION: The findings rationalized the prodrug design hypothesis that vicagrel could overcome the extensive invalid hydrolysis of clopidogrel by the hepatic CE1 but experience the extensive hydrolysis to 2-oxo-clopidogrel and subsequent oxidation to AM in the intestine. This also supported the theory of improved pharmacological activity through facilitated formation of 2-oxo-clopidogrel, thus warranting much needed future clinical pharmacokinetic studies of vicagrel.


Subject(s)
Acetates/metabolism , Intestinal Mucosa/metabolism , Phenylacetates/blood , Phenylacetates/metabolism , Thiophenes/blood , Thiophenes/metabolism , Ticlopidine/analogs & derivatives , Animals , Clopidogrel , Esterases/metabolism , Humans , Hydrolysis , Microsomes/metabolism , Prodrugs/metabolism , Rats , Rats, Sprague-Dawley , Ticlopidine/blood , Ticlopidine/metabolism
12.
J Vet Pharmacol Ther ; 37(4): 354-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24628410

ABSTRACT

Robenacoxib and ketoprofen are acidic nonsteroidal anti-inflammatory drugs (NSAIDs). Both are licensed for once daily administration in the cat, despite having short blood half-lives. This study reports the pharmacokinetic/pharmacodynamic (PK/PD) modelling of each drug in a feline model of inflammation. Eight cats were enrolled in a randomized, controlled, three-period cross-over study. In each period, sterile inflammation was induced by the injection of carrageenan into a subcutaneously implanted tissue cage, immediately before the subcutaneous injection of robenacoxib (2 mg/kg), ketoprofen (2 mg/kg) or placebo. Blood samples were taken for the determination of drug and serum thromboxane (Tx)B2 concentrations (measuring COX-1 activity). Tissue cage exudate samples were obtained for drug and prostaglandin (PG)E2 concentrations (measuring COX-2 activity). Individual animal pharmacokinetic and pharmacodynamic parameters for COX-1 and COX-2 inhibition were generated by PK/PD modelling. S(+) ketoprofen clearance scaled by bioavailability (CL/F) was 0.114 L/kg/h (elimination half-life = 1.62 h). For robenacoxib, blood CL/F was 0.684 L/kg/h (elimination half-life = 1.13 h). Exudate elimination half-lives were 25.9 and 41.5 h for S(+) ketoprofen and robenacoxib, respectively. Both drugs reduced exudate PGE2 concentration significantly between 6 and 36 h. Ketoprofen significantly suppressed (>97%) serum TxB2 between 4 min and 24 h, whereas suppression was mild and transient with robenacoxib. In vivo IC50 COX-1/IC50 COX-2 ratios were 66.9:1 for robenacoxib and 1:107 for S(+) ketoprofen. The carboxylic acid nature of both drugs may contribute to the prolonged COX-2 inhibition in exudate, despite short half-lives in blood.


Subject(s)
Cat Diseases/chemically induced , Diphenylamine/analogs & derivatives , Inflammation/drug therapy , Ketoprofen/pharmacology , Ketoprofen/pharmacokinetics , Phenylacetates/pharmacology , Phenylacetates/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Anti-Inflammatory Agents, Non-Steroidal/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Carrageenan/toxicity , Cat Diseases/drug therapy , Cats , Diffusion Chambers, Culture , Diphenylamine/blood , Diphenylamine/chemistry , Diphenylamine/pharmacokinetics , Diphenylamine/pharmacology , Female , Ketoprofen/blood , Ketoprofen/chemistry , Male , Molecular Structure , Phenylacetates/blood , Phenylacetates/chemistry
13.
Vet J ; 307: 106200, 2024 Oct.
Article in English | MEDLINE | ID: mdl-39095012

ABSTRACT

Robenacoxib (RX) is a non-steroidal anti-inflammatory drug (NSAID) of the coxib class. This study aimed to evaluate the plasma dispositions and faecal excretion profiles of RX in Alpine and Saanen goats following oral and subcutaneous routes. Two different goat breeds were allocated into two treatment groups concerning the breed. RX was administered subcutaneously to animals at a dose of 4 mg/kg b.w. Following a one-week washout period, RX was administered by oral route to the same animals at the same dose. Heparinized blood samples were collected from all animals before drug administration (0 h) and subsequently up to 24 h. Faecal samples were collected at various times between 8 h and 36 h. The concentrations of RX in plasma and faeces were determined by HPLC. The plasma half-life (T1/2λz) of RX in Saanen goats (1.21 h) was significantly longer (P < 0.017) than in Alpine goats (0.90 h) after subcutaneous administration. In both goat breeds, statistical differences were observed between subcutaneous and oral administration of RX for T1/2λz, Tlast, Cmax, AUC0-∞, and MRT0-∞. Faecal Cmax and Tmax parameters following oral administrations were 0.92 µg/g and 0.85 µg/g at 30 h and at 24 h in Alpine and Saanen goats, respectively. The difference in plasma protein ratio between Alpine and Saanen goats may have affected the T1/2λz of the drug. NSAIDs are among the drug groups frequently detected in aquatic and terrestrial ecosystems around the world and there are data on the effects of NSAID residues on wildlife and aquatic species. Therefore, revealing the excretion of NSAIDs, which are frequently used in the veterinary field, in faeces and urine should be considered for ecological sustainability.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal , Feces , Goats , Phenylacetates , Animals , Goats/metabolism , Goats/blood , Feces/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/blood , Injections, Subcutaneous/veterinary , Administration, Oral , Phenylacetates/pharmacokinetics , Phenylacetates/blood , Phenylacetates/administration & dosage , Half-Life , Diphenylamine/pharmacokinetics , Diphenylamine/analogs & derivatives , Diphenylamine/administration & dosage , Female , Male , Area Under Curve
14.
Mol Genet Metab ; 110(4): 446-53, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24144944

ABSTRACT

BACKGROUND: Phenylacetic acid (PAA) is the active moiety in sodium phenylbutyrate (NaPBA) and glycerol phenylbutyrate (GPB, HPN-100). Both are approved for treatment of urea cycle disorders (UCDs) - rare genetic disorders characterized by hyperammonemia. PAA is conjugated with glutamine in the liver to form phenylacetyleglutamine (PAGN), which is excreted in urine. PAA plasma levels ≥ 500 µg/dL have been reported to be associated with reversible neurological adverse events (AEs) in cancer patients receiving PAA intravenously. Therefore, we have investigated the relationship between PAA levels and neurological AEs in patients treated with these PAA pro-drugs as well as approaches to identifying patients most likely to experience high PAA levels. METHODS: The relationship between nervous system AEs, PAA levels and the ratio of plasma PAA to PAGN were examined in 4683 blood samples taken serially from: [1] healthy adults [2], UCD patients of ≥ 2 months of age, and [3] patients with cirrhosis and hepatic encephalopathy (HE). The plasma ratio of PAA to PAGN was analyzed with respect to its utility in identifying patients at risk of high PAA values. RESULTS: Only 0.2% (11) of 4683 samples exceeded 500 µg/ml. There was no relationship between neurological AEs and PAA levels in UCD or HE patients, but transient AEs including headache and nausea that correlated with PAA levels were observed in healthy adults. Irrespective of population, a curvilinear relationship was observed between PAA levels and the plasma PAA:PAGN ratio, and a ratio>2.5 (both in µg/mL) in a random blood draw identified patients at risk for PAA levels>500 µg/ml. CONCLUSIONS: The presence of a relationship between PAA levels and reversible AEs in healthy adults but not in UCD or HE patients may reflect intrinsic differences among the populations and/or metabolic adaptation with continued dosing. The plasma PAA:PAGN ratio is a functional measure of the rate of PAA metabolism and represents a useful dosing biomarker.


Subject(s)
Glutamine/analogs & derivatives , Hepatic Encephalopathy/blood , Phenylacetates/blood , Urea Cycle Disorders, Inborn/blood , Biomarkers/blood , Drug-Related Side Effects and Adverse Reactions/blood , Drug-Related Side Effects and Adverse Reactions/etiology , Glutamine/administration & dosage , Glutamine/blood , Glycerol/administration & dosage , Glycerol/analogs & derivatives , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/pathology , Humans , Liver/drug effects , Liver/metabolism , Neoplasms/complications , Neoplasms/drug therapy , Phenylacetates/administration & dosage , Phenylbutyrates/administration & dosage , Randomized Controlled Trials as Topic , Urea Cycle Disorders, Inborn/epidemiology , Urea Cycle Disorders, Inborn/etiology , Urea Cycle Disorders, Inborn/pathology
15.
Artif Organs ; 37(4): 409-16, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23330821

ABSTRACT

Protein-bound uremic toxins, such as phenylacetic acid, indoxyl sulfate, and p-cresyl sulfate, contribute substantially to the progression of chronic kidney disease (CKD) and cardiovascular disease (CVD). However, based on their protein binding, these hydrophobic uremic toxins are poorly cleared during conventional dialysis and thus accumulate in CKD-5D patients. Therefore, we investigated whether hydrophobic and cationic adsorbers are more effective for removal of protein-bound, hydrophobic uremic toxins than conventional high-flux hemodialyzer. Five CKD-5D patients were treated using the fractionated plasma separation, adsorption, and dialysis (FPAD) system for 5 h. A control group of five CKD patients was treated with conventional high-flux hemodialysis. Plasma concentrations of phenylacetic acid, indoxyl sulfate, and p-cresyl sulfate were measured. Removal rates of FPAD treatment in comparison to conventional high-flux hemodialysis were increased by 130% for phenylacetic acid, 187% for indoxyl sulfate, and 127% for p-cresol. FPAD treatment was tolerated well in terms of clinically relevant biochemical parameters. However, patients suffered from mild nausea 2 h after the start of the treatment, which persisted until the end of treatment. Due to the high impact of protein-bound, hydrophobic uremic toxins on progression of CKD and CVD in CKD-5D patients, the use of an adsorber in combination with dialysis membranes may be a new therapeutic option to increase the removal rate of these uremic toxins. However, larger, long-term prospective clinical trials are needed to demonstrate the impact on clinical outcome.


Subject(s)
Cresols/isolation & purification , Indican/isolation & purification , Phenylacetates/isolation & purification , Plasmapheresis/methods , Renal Dialysis/methods , Sulfuric Acid Esters/isolation & purification , Uremia/therapy , Adsorption , Blood Proteins/metabolism , Cresols/blood , Cresols/metabolism , Humans , Indican/blood , Indican/metabolism , Phenylacetates/blood , Phenylacetates/metabolism , Pilot Projects , Protein Binding , Sulfuric Acid Esters/blood , Sulfuric Acid Esters/metabolism , Uremia/blood , Uremia/metabolism
16.
J Hepatol ; 56(1): 109-14, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21835138

ABSTRACT

BACKGROUND & AIMS: Ornithine phenylacetate (OP) is a new drug that has been proposed for the treatment of hepatic encephalopathy (HE) because it decreases plasma ammonia. We performed a study to assess if OP would impact on neuronal function. METHODS: Motor-evoked potentials (MEP), a surrogate of hepatic encephalopathy, were assessed (without anesthesia) in rats with portacaval anastomosis (PCA) that received gastrointestinal blood (GIB). Rats were pre-treated with OP prior to GIB. Ammonia and related metabolites (plasma, urine, and brain microdialysis) were assessed by HPLC and mass spectroscopy. RESULTS: OP (one dose or 3 days) prevented disturbances in MEP induced by GIB in PCA rats. In rats treated with OP for 3 days, the amplitude and latency of MEP remained stable (-1% and +1%), while in the control group the amplitude decreased -21% and the latency increased +12% (p<0.01). OP attenuated the rise of ammonia in plasma by 45%, ammonia in brain microdialysate by 48%, induced a faster glutamine rise and the appearance of phenylacetylglutamine in plasma and urine. In addition, OP was associated with a lower concentration of ammonia and glutamate in brain microdialysate (approx. 50%). CONCLUSIONS: OP prevents abnormalities in MEP precipitated by GIB in a model of HE. This is probably due to the enhancement of glutamine synthesis and metabolism, which results in a lower rise of plasma ammonia and the prevention of changes in glutamate in microdialysate. Thus, OP may be a good drug to prevent HE precipitated by gastrointestinal bleeding.


Subject(s)
Evoked Potentials, Motor/drug effects , Hepatic Encephalopathy/drug therapy , Ornithine/analogs & derivatives , Ornithine/administration & dosage , Ornithine/pharmacology , Phenylacetates/administration & dosage , Amino Acids/metabolism , Ammonia/metabolism , Animals , Brain/metabolism , Disease Models, Animal , Glutamine/analogs & derivatives , Glutamine/blood , Hepatic Encephalopathy/blood , Hepatic Encephalopathy/etiology , Hepatic Encephalopathy/physiopathology , Male , Phenylacetates/blood , Portacaval Shunt, Surgical/adverse effects , Rats , Rats, Sprague-Dawley
17.
Mol Genet Metab ; 107(3): 308-14, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22958974

ABSTRACT

UNLABELLED: We have analyzed pharmacokinetic data for glycerol phenylbutyrate (also GT4P or HPN-100) and sodium phenylbutyrate with respect to possible dosing biomarkers in patients with urea cycle disorders (UCD). STUDY DESIGN: These analyses are based on over 3000 urine and plasma data points from 54 adult and 11 pediatric UCD patients (ages 6-17) who participated in three clinical studies comparing ammonia control and pharmacokinetics during steady state treatment with glycerol phenylbutyrate or sodium phenylbutyrate. All patients received phenylbutyric acid equivalent doses of glycerol phenylbutyrate or sodium phenylbutyrate in a cross over fashion and underwent 24-hour blood samples and urine sampling for phenylbutyric acid, phenylacetic acid and phenylacetylglutamine. RESULTS: Patients received phenylbutyric acid equivalent doses of glycerol phenylbutyrate ranging from 1.5 to 31.8 g/day and of sodium phenylbutyrate ranging from 1.3 to 31.7 g/day. Plasma metabolite levels varied widely, with average fluctuation indices ranging from 1979% to 5690% for phenylbutyric acid, 843% to 3931% for phenylacetic acid, and 881% to 1434% for phenylacetylglutamine. Mean percent recovery of phenylbutyric acid as urinary phenylacetylglutamine was 66.4 and 69.0 for pediatric patients and 68.7 and 71.4 for adult patients on glycerol phenylbutyrate and sodium phenylbutyrate, respectively. The correlation with dose was strongest for urinary phenylacetylglutamine excretion, either as morning spot urine (r = 0.730, p < 0.001) or as total 24-hour excretion (r = 0.791 p<0.001), followed by plasma phenylacetylglutamine AUC(24-hour), plasma phenylacetic acid AUC(24-hour) and phenylbutyric acid AUC(24-hour). Plasma phenylacetic acid levels in adult and pediatric patients did not show a consistent relationship with either urinary phenylacetylglutamine or ammonia control. CONCLUSION: The findings are collectively consistent with substantial yet variable pre-systemic (1st pass) conversion of phenylbutyric acid to phenylacetic acid and/or phenylacetylglutamine. The variability of blood metabolite levels during the day, their weaker correlation with dose, the need for multiple blood samples to capture trough and peak, and the inconsistency between phenylacetic acid and urinary phenylacetylglutamine as a marker of waste nitrogen scavenging limit the utility of plasma levels for therapeutic monitoring. By contrast, 24-hour urinary phenylacetylglutamine and morning spot urine phenylacetylglutamine correlate strongly with dose and appear to be clinically useful non-invasive biomarkers for compliance and therapeutic monitoring.


Subject(s)
Ammonia/urine , Glutamine/analogs & derivatives , Glycerol/analogs & derivatives , Phenylacetates/urine , Phenylbutyrates/urine , Urea Cycle Disorders, Inborn/drug therapy , Urea Cycle Disorders, Inborn/urine , Adolescent , Adult , Ammonia/blood , Biomarkers, Pharmacological/blood , Biomarkers, Pharmacological/urine , Child , Cross-Over Studies , Drug Administration Schedule , Female , Glutamine/blood , Glutamine/urine , Glycerol/blood , Glycerol/pharmacokinetics , Glycerol/urine , Humans , Male , Phenylacetates/blood , Phenylbutyrates/blood , Phenylbutyrates/pharmacokinetics , Urea Cycle Disorders, Inborn/blood
18.
J Vet Pharmacol Ther ; 35(3): 290-300, 2012 Jun.
Article in English | MEDLINE | ID: mdl-21736587

ABSTRACT

The safety of robenacoxib, a nonsteroidal anti-inflammatory drug with high selectivity for inhibition of the cyclooxygenase (COX)-2 isoform of COX, was investigated in the cat in two randomized, blinded, placebo-controlled, parallel-group studies. Robenacoxib was administered orally to healthy young domestic short-hair cats at dosages of 0 (placebo), 5 and 10 mg/kg once daily for 28 days (study 1) and at 0 (placebo), 2, 6 and 10 mg/kg twice daily for 42 days (study 2). The recommended minimum dosage for robenacoxib tablets in cats is 1 mg/kg once daily (range 1-2.4 mg/kg). Relative to placebo treatment, no toxicologically significant effects of robenacoxib were recorded in either study, based on general observations of health, haematological and clinical chemistry variables and urinalyses in life, and by post mortem organ weight, gross pathology and histopathology assessments. Pharmacokinetic-pharmacodynamic simulations indicated that all dosages of robenacoxib were associated with marked inhibition of COX-2 at peak effect (median I(max) 97.8-99.4% inhibition) with lesser inhibition of COX-1 (median I(max) 26.8-58.3% inhibition). Inhibition of the COXs was short lasting, with >10% median inhibition persisting for 4.0 h for COX-2 and 1.5 h for COX-1. These levels of inhibition of COX-1 and COX-2 twice daily with robenacoxib were not associated with any detectable toxicity, suggesting that, as previously described in dogs, the high safety index of robenacoxib in cats may be related to a combination of its high COX-2 selectivity and short residence time in the central compartment.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/adverse effects , Diphenylamine/analogs & derivatives , Phenylacetates/adverse effects , Administration, Oral , Animals , Anti-Inflammatory Agents, Non-Steroidal/administration & dosage , Anti-Inflammatory Agents, Non-Steroidal/blood , Cats , Cyclooxygenase 2 Inhibitors/administration & dosage , Cyclooxygenase 2 Inhibitors/adverse effects , Cyclooxygenase Inhibitors/administration & dosage , Cyclooxygenase Inhibitors/adverse effects , Diphenylamine/administration & dosage , Diphenylamine/adverse effects , Diphenylamine/blood , Dose-Response Relationship, Drug , Female , Male , Phenylacetates/administration & dosage , Phenylacetates/blood
19.
J Vet Pharmacol Ther ; 35(1): 19-32, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21767277

ABSTRACT

Robenacoxib is a novel nonsteroidal anti-inflammatory drug developed for use in cats. It is a highly selective COX-2 inhibitor. Results from previous feline studies showed that, despite a short half-life in blood, the effect of robenacoxib persisted for 24 h in clinical studies. A tissue cage model of acute inflammation was used to determine robenacoxib's pharmacokinetics and its ex vivo and in vivo selectivity for COX-1 and COX-2 using serum TxB(2) and exudate PGE(2) as surrogate markers for enzyme activity, respectively. After intravenous, subcutaneous and oral administration (2 mg/kg), the clearance of robenacoxib from blood was rapid (0.54-0.71 L·h/kg). The mean residence time (MRT) in blood was short (0.4, 1.9 and 3.3 h after intravenous, subcutaneous and oral administration, respectively), but in exudate MRT was approximately 24 h regardless of the route of administration. Robenacoxib inhibition of COX-1 in blood was transient, occurring only at high concentrations, but inhibition of COX-2 in exudate persisted to 24 h. The potency ratio (IC(50) COX-1: IC(50) COX-2) was 171:1, and slopes of the concentration-effect relationship were 1.36 and 1.12 for COX-1 and COX-2, respectively. These data highlight the enzymatic selectivity and inflamed tissue selectivity of robenacoxib and support the current recommendation of once-daily administration.


Subject(s)
Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacokinetics , Diphenylamine/analogs & derivatives , Inflammation/drug therapy , Phenylacetates/pharmacology , Phenylacetates/pharmacokinetics , Animals , Anti-Inflammatory Agents, Non-Steroidal/blood , Area Under Curve , Cat Diseases/blood , Cat Diseases/drug therapy , Cat Diseases/metabolism , Cats , Cross-Over Studies , Diffusion Chambers, Culture , Dinoprostone/blood , Dinoprostone/metabolism , Diphenylamine/blood , Diphenylamine/pharmacokinetics , Diphenylamine/pharmacology , Female , Half-Life , Male , Phenylacetates/blood , Prostaglandins/blood , Thromboxane B2/blood , Thromboxane B2/metabolism
20.
JCI Insight ; 6(9)2021 05 10.
Article in English | MEDLINE | ID: mdl-33986195

ABSTRACT

Gut microbe-derived metabolites influence human physiology and disease. However, establishing mechanistic links between gut microbial metabolites and disease pathogenesis in animal models remains challenging. The major route of absorption for microbe-derived small molecules is venous drainage via the portal vein to the liver. In the event of presystemic hepatic metabolism, the route of metabolite administration becomes critical. To our knowledge, we describe here a novel portal vein cannulation technique using a s.c. implanted osmotic pump to achieve continuous portal vein infusion in mice. We first administered the microbial metabolite trimethylamine (TMA) over 4 weeks, during which increased peripheral plasma levels of TMA and its host liver-derived cometabolite, trimethylamine-N-oxide, were observed when compared with a vehicle control. Next, 4-hydroxyphenylacetic acid (4-HPAA), a microbial metabolite that undergoes extensive presystemic hepatic metabolism, was administered intraportally to examine effects on hepatic gene expression. As expected, hepatic levels of 4-HPAA were elevated when compared with the control group while peripheral plasma 4-HPAA levels remained the same. Moreover, significant changes in the hepatic transcriptome were revealed by an unbiased RNA-Seq approach. Collectively, to our knowledge this work describes a novel method for administering gut microbe-derived metabolites via the portal vein, mimicking their physiologic delivery in vivo.


Subject(s)
Gastrointestinal Microbiome , Infusions, Intravenous/methods , Liver/metabolism , Methylamines/administration & dosage , Phenylacetates/administration & dosage , Portal Vein , Animals , Gene Expression/drug effects , Methylamines/blood , Methylamines/metabolism , Methylamines/pharmacology , Mice , Phenylacetates/blood , Phenylacetates/metabolism , Phenylacetates/pharmacology , RNA-Seq , Transcriptome/drug effects
SELECTION OF CITATIONS
SEARCH DETAIL