Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Vet Pharmacol Ther ; 44(1): 36-46, 2021 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-32757313

RESUMEN

The in vivo metabolism and pharmacokinetics of flunixin meglumine and phenylbutazone have been extensively characterized; however, there are no published reports describing the in vitro metabolism, specifically the enzymes responsible for the biotransformation of these compounds in horses. Due to their widespread use and, therefore, increased potential for drug-drug interactions and widespread differences in drug disposition, this study aims to build on the limited current knowledge regarding P450-mediated metabolism in horses. Drugs were incubated with equine liver microsomes and a panel of recombinant equine P450s. Incubation of phenylbutazone in microsomes generated oxyphenbutazone and gamma-hydroxy phenylbutazone. Microsomal incubations with flunixin meglumine generated 5-OH flunixin, with a kinetic profile suggestive of substrate inhibition. In recombinant P450 assays, equine CYP3A97 was the only enzyme capable of generating oxyphenbutazone while several members of the equine CYP3A family and CYP1A1 were capable of catalyzing the biotransformation of flunixin to 5-OH flunixin. Flunixin meglumine metabolism by CYP1A1 and CYP3A93 showed a profile characteristic of biphasic kinetics, suggesting two substrate binding sites. The current study identifies specific enzymes responsible for the metabolism of two NSAIDs in horses and provides the basis for future study of drug-drug interactions and identification of reasons for varying pharmacokinetics between horses.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Clonixina/análogos & derivados , Sistema Enzimático del Citocromo P-450/metabolismo , Caballos/metabolismo , Fenilbutazona/farmacocinética , Animales , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Clonixina/química , Clonixina/metabolismo , Clonixina/farmacocinética , ADN Complementario/genética , ADN Complementario/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Microsomas Hepáticos/metabolismo , Estructura Molecular , Fenilbutazona/química , Fenilbutazona/metabolismo
2.
J Dairy Sci ; 102(12): 11465-11469, 2019 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-31629517

RESUMEN

Flunixin is a nonsteroidal anti-inflammatory drug and the most commonly prescribed analgesic in cattle in the United States. Recently, the US Food and Drug Administration (FDA) approved a transdermal formulation of flunixin for control of pyrexia associated with bovine respiratory disease and the control of pain associated with foot rot. The transdermal formulation is not currently approved for use in lactating dairy cattle in the United States, but extra-label use in dairy cattle is permissible under US regulations. The objectives of this study were to determine the pharmacokinetics in milk of dairy cows treated with transdermal flunixin and determine an appropriate withdrawal time for milk. Ten lactating Holstein cows were enrolled into the study in mid lactation. Following treatment, cows were milked 3 times per day through 144 h. Milk samples were collected for drug analysis using ultra-high-pressure liquid chromatography coupled with a triple quadrupole mass spectrometer. The geometric mean maximum concentration for flunixin in milk was 0.010 µg/mL and was 0.061 µg/mL for the active metabolite, 5-hydroxyflunixin. The geometric mean terminal half-life was 20.71 h for flunixin and 22.62 h for 5-hydroxyflunixin. Calculations to approximate a withdrawal time in milk following transdermal flunixin administration were accomplished using a statistical tolerance limit procedure. This analysis indicated that it would be prudent to observe a withdrawal period of 96 h following the last treatment. This is more than twice as long as the labeled withdrawal period of 36 h following use of the injectable formulation. The withdrawal period suggested by this work should be applied carefully, as this study was not conducted under the full quality control practices required by the US FDA for a full drug approval study. Caution should be taken when applying this withdrawal time to diseased animals, animals that are milked with different milking frequencies, and those in different stages of production as these have all been shown to affect drug depletion from milk.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Clonixina/análogos & derivados , Leche/metabolismo , Administración Cutánea , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Bovinos , Cromatografía Líquida de Alta Presión , Clonixina/administración & dosificación , Clonixina/metabolismo , Clonixina/farmacocinética , Femenino , Lactancia , Espectrometría de Masas
3.
J Vet Pharmacol Ther ; 42(5): 572-579, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31353535

RESUMEN

The aim of this study was to determine the pharmacokinetics and prostaglandin E2 (PGE2 ) synthesis inhibiting effects of intravenous (IV) and transdermal (TD) flunixin meglumine in eight, adult, female, Huacaya alpacas. A dose of 2.2 mg/kg administered IV and 3.3 mg/kg administered TD using a cross-over design. Plasma flunixin concentrations were measured by LC-MS/MS. Prostaglandin E2 concentrations were determined using a commercially available ELISA. Pharmacokinetic (PK) analysis was performed using noncompartmental methods. Plasma PGE2 concentrations decreased after IV flunixin meglumine administration but there was minimal change after TD application. Mean t1/2 λz after IV administration was 4.531 hr (range 3.355 to 5.571 hr) resulting from a mean Vz of 570.6 ml/kg (range, 387.3 to 1,142 ml/kg) and plasma clearance of 87.26 ml kg-1  hr-1 (range, 55.45-179.3 ml kg-1  hr-1 ). The mean Cmax, Tmax and t1/2 λz for flunixin following TD administration were 106.4 ng/ml (range, 56.98 to 168.6 ng/ml), 13.57 hr (range, 6.000-34.00 hr) and 24.06 hr (18.63 to 39.5 hr), respectively. The mean bioavailability for TD flunixin was calculated as 25.05%. The mean 80% inhibitory concentration (IC80 ) of PGE2 by flunixin meglumine was 0.23 µg/ml (range, 0.01 to 1.38 µg/ml). Poor bioavailability and poor suppression of PGE2 identified in this study indicate that TD flunixin meglumine administered at 3.3 mg/kg is not recommended for use in alpacas.


Asunto(s)
Camélidos del Nuevo Mundo/sangre , Clonixina/análogos & derivados , Administración Cutánea , Administración Intravenosa , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/farmacocinética , Área Bajo la Curva , Clonixina/administración & dosificación , Clonixina/sangre , Clonixina/metabolismo , Clonixina/farmacocinética , Dinoprostona/sangre , Dinoprostona/metabolismo , Femenino , Semivida
4.
J Vet Pharmacol Ther ; 42(5): 505-510, 2019 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-31090076

RESUMEN

Medication control in greyhound racing requires information from administration studies that measure drug levels in the urine as well as plasma, with time points that extend into the terminal phase of excretion. To characterize the plasma and the urinary pharmacokinetics of flunixin and enable regulatory advice for greyhound racing in respect of both medication and residue control limits, flunixin meglumine was administered intravenously on one occasion to six different greyhounds at the label dose of 1 mg/kg and the levels of flunixin were measured in plasma for up to 96 hr and in urine for up to 120 hr. Using the standard methodology for medication control, the irrelevant plasma concentration was determined as 1 ng/ml and the irrelevant urine concentration was determined as 30 ng/ml. This information can be used by regulators to determine a screening limit, detection time and a residue limit. The greyhounds with the highest average urine pH had far greater flunixin exposure compared with the greyhounds that had the lowest. This is entirely consistent with the extent of ionization predicted by the Henderson-Hasselbalch equation. This variability in the urine pharmacokinetics reduces with time, and at 72 hr postadministration, in the terminal phase, the variability in urine and plasma flunixin concentrations are similar and should not affect medication control.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Clonixina/análogos & derivados , Perros/sangre , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/orina , Área Bajo la Curva , Clonixina/sangre , Clonixina/química , Clonixina/metabolismo , Clonixina/farmacocinética , Clonixina/orina , Perros/orina , Residuos de Medicamentos , Femenino , Semivida , Concentración de Iones de Hidrógeno , Infusiones Intravenosas , Masculino , Deportes , Orina/química
5.
Drug Metab Dispos ; 47(5): 516-524, 2019 05.
Artículo en Inglés | MEDLINE | ID: mdl-30858238

RESUMEN

Flunixin meglumine is a nonsteroidal anti-inflammatory drug (NSAID) widely used in veterinary medicine. It is indicated to treat inflammatory processes, pain, and pyrexia in farm animals. In addition, it is one of the few NSAIDs approved for use in dairy cows, and consequently gives rise to concern regarding its milk residues. The ABCG2 efflux transporter is induced during lactation in the mammary gland and plays an important role in the secretion of different compounds into milk. Previous reports have demonstrated that bovine ABCG2 Y581S polymorphism increases fluoroquinolone levels in cow milk. However, the implication of this transporter in the secretion into milk of anti-inflammatory drugs has not yet been studied. The objective of this work was to study the role of ABCG2 in the secretion into milk of flunixin and its main metabolite, 5-hydroxyflunixin, using Abcg2(-/-) mice, and to investigate the implication of the Y581S polymorphism in the secretion of these compounds into cow milk. Correlation with the in vitro situation was assessed by in vitro transport assays using Madin-Darby canine kidney II cells overexpressing murine and the two variants of the bovine transporter. Our results show that flunixin and 5-hydroxyflunixin are transported by ABCG2 and that this protein is responsible for their secretion into milk. Moreover, the Y581S polymorphism increases flunixin concentration into cow milk, but it does not affect milk secretion of 5-hydroxyflunixin. This result correlates with the differences in the in vitro transport of flunixin between the two bovine variants. These findings are relevant to the therapeutics of anti-inflammatory drugs.


Asunto(s)
Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/metabolismo , Antiinflamatorios/metabolismo , Clonixina/análogos & derivados , Leche/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2/genética , Transportadoras de Casetes de Unión a ATP/genética , Transportadoras de Casetes de Unión a ATP/metabolismo , Animales , Antibacterianos/metabolismo , Transporte Biológico/genética , Transporte Biológico/fisiología , Bovinos , Línea Celular , Clonixina/metabolismo , Perros , Femenino , Lactancia/metabolismo , Células de Riñón Canino Madin Darby , Ratones , Polimorfismo Genético/genética
6.
Res Vet Sci ; 123: 118-123, 2019 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-30641470

RESUMEN

The majority of cattle found to have violative liver residues of flunixin (FNX) in the United States are dairy cows. It has been hypothesized that illness of cows decreases the rate of FNX metabolism, resulting in violative residues at slaughter. Another contributing factor might be an age-related decrease in FNX metabolism, as dairy cull cows are typically older at slaughter than cattle raised for beef, rather than milk production. In order to investigate this possibility, subcellular fractions were prepared from liver slices from steers (n = 6) and heifers (n = 5) <30 months of age, and cows (n = 8) >48 mos of age. Cytochrome P450 (P450), NADPH-P450 reductase and glucose-6-phosphate dehydrogenase (G6PDH) activity and rate of 5-hydroxy FNX (5-OH FNX) formation were measured in liver homogenate, cytosolic, microsomal, and S9 fractions. Cows had lower concentrations of P450, NADPH-P450 reductase activity, and 5-OH FNX formation (P ≤ 0. 02), supporting the theory that advanced age may contribute to the higher incidence of violative FNX residues in dairy cows.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Bovinos/metabolismo , Clonixina/análogos & derivados , Hígado/metabolismo , Animales , Antiinflamatorios no Esteroideos/metabolismo , Clonixina/metabolismo , Clonixina/farmacocinética , Sistema Enzimático del Citocromo P-450/metabolismo , Femenino , Masculino , Técnicas de Cultivo de Tejidos
7.
J Dairy Sci ; 100(11): 8670-8679, 2017 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-28918155

RESUMEN

Casein proteins (αS1-, αS2-, ß- and κ-casein) account for 80% of the total protein content in bovine milk and form casein micelles (average diameter = 130 nm, approximately 1015 micelles/mL). The affinity of native casein micelles with the 3 hydrophobic active pharmaceutical ingredients (API), meloxicam [351.4 g/mol; log P = 3.43; acid dissociation constant (pKa) = 4.08], flunixin (296.2 g/mol; log P = 4.1; pKa = 5.82), and thiabendazole (201.2 g/mol; log P = 2.92; pKa = 4.64), was evaluated in bovine milk collected from dosed Holstein cows. Native casein micelles were separated from raw bovine milk by mild techniques such as ultracentrifugation, diafiltration, isoelectric point precipitation (pH 4.6), and size exclusion chromatography. Acetonitrile extraction of hydrophobic API was then done, followed by quantification using HPLC-UV. For the API or metabolites meloxicam, 5-hyroxy flunixin and 5-hydroxy thiabendazole, 31 ± 3.90, 31 ± 1.3, and 28 ± 0.5% of the content in milk was associated with casein micelles, respectively. Less than ∼5.0% of the recovered hydrophobic API were found in the milk fat fraction, and the remaining ∼65% were associated with the whey/serum fraction. A separate in vitro study showed that 66 ± 6.4% of meloxicam, 29 ± 0.58% of flunixin, 34 ± 0.21% of the metabolite 5-hyroxy flunixin, 50 ± 4.5% of thiabendazole, and 33 ± 3.8% of metabolite 5-hydroxy thiabendazole was found partitioned into casein micelles. Our study supports the hypothesis that casein micelles are native carriers for hydrophobic compounds in bovine milk.


Asunto(s)
Caseínas/metabolismo , Clonixina/análogos & derivados , Leche/química , Tiabendazol/metabolismo , Tiazinas/metabolismo , Tiazoles/metabolismo , Animales , Bovinos , Clonixina/metabolismo , Suplementos Dietéticos , Femenino , Interacciones Hidrofóbicas e Hidrofílicas , Meloxicam , Micelas , Unión Proteica , Suero Lácteo/metabolismo , Proteína de Suero de Leche/metabolismo
8.
Vet Surg ; 45(8): 1087-1094, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27731498

RESUMEN

OBJECTIVE: Determine the effects of nonsteroidal anti-inflammatory drugs (NSAID) on platelet function and thromboxane synthesis immediately after drug administration and following 5 days of NSAID administration in healthy horses. STUDY DESIGN: Randomized cross-over study. ANIMALS: Healthy adult horses (n=9; 6 geldings and 3 mares). METHODS: Horses received either flunixin meglumine (1.1 mg/kg IV every 12 hours), phenylbutazone (2.2 mg/kg IV every 12 hours), or firocoxib (loading dose of 0.27 mg/kg IV on day 1, then 0.09 mg/kg IV every 24 hours for 4 days) for a total of 5 days. Blood samples were collected prior to drug administration (day 0), 1 hour after initial NSAID administration (day 1), and then 1 hour post-NSAID administration on day 5. Platelet function was assessed using turbidimetric aggregometry and a platelet function analyzer. Serum thromboxane B2 concentrations were determined by commercial ELISA kit. A minimum 14 day washout period occurred between trials. RESULTS: At 1 hour and 5 days postadministration of firocoxib, flunixin meglumine, or phenylbutazone, there was no significant effect on platelet aggregation or function using turbidimetric aggregometry or a platelet function analyzer. There was, however, a significant decrease in thromboxane synthesis at 1 hour and 5 days postadministration of flunixin meglumine and phenylbutazone that was not seen with firocoxib. CONCLUSION: Preoperative administration of flunixin meglumine, phenylbutazone, or firocoxib should not inhibit platelet function based on our model. The clinical implications of decreased thromboxane B2 synthesis following flunixin meglumine and phenylbutazone administration are undetermined.


Asunto(s)
4-Butirolactona/análogos & derivados , Antiinflamatorios no Esteroideos/administración & dosificación , Plaquetas/efectos de los fármacos , Clonixina/análogos & derivados , Caballos/metabolismo , Fenilbutazona/administración & dosificación , Sulfonas/administración & dosificación , Tromboxanos/metabolismo , 4-Butirolactona/administración & dosificación , 4-Butirolactona/metabolismo , Animales , Antiinflamatorios no Esteroideos/metabolismo , Clonixina/administración & dosificación , Clonixina/metabolismo , Estudios Cruzados , Femenino , Masculino , Fenilbutazona/metabolismo , Sulfonas/metabolismo
9.
Am J Vet Res ; 77(6): 634-40, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27227502

RESUMEN

OBJECTIVE To describe plasma pharmacokinetic parameters and tissue elimination of flunixin in veal calves. ANIMALS 20 unweaned Holstein calves between 3 and 6 weeks old. PROCEDURES Each calf received flunixin (2.2 mg/kg, IV, q 24 h) for 3 days. Blood samples were collected from all calves before the first dose and at predetermined times after the first and last doses. Beginning 24 hours after injection of the last dose, 4 calves were euthanized each day for 5 days. Plasma and tissue samples were analyzed by ultraperformance liquid chromatography. Pharmacokinetic parameters were calculated by compartmental and noncompartmental methods. RESULTS Mean ± SD plasma flunixin elimination half-life, residence time, and clearance were 1.32 ± 0.94 hours, 12.54 ± 10.96 hours, and 64.6 ± 40.7 mL/h/kg, respectively. Mean hepatic and muscle flunixin concentrations decreased to below FDA-established tolerance limits (0.125 and 0.025 µg/mL, respectively) for adult cattle by 3 and 2 days, respectively, after injection of the last dose of flunixin. Detectable flunixin concentrations were present in both the liver and muscle for at least 5 days after injection of the last dose. CONCLUSIONS AND CLINICAL RELEVANCE The labeled slaughter withdrawal interval for flunixin in adult cattle is 4 days. Because administration of flunixin to veal calves represents extralabel drug use, any detectable flunixin concentrations in edible tissues are considered a violation. Results indicated that a slaughter withdrawal interval of several weeks may be necessary to ensure that violative tissue residues of flunixin are not detected in veal calves treated with that drug.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Bovinos/sangre , Clonixina/análogos & derivados , Animales , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Área Bajo la Curva , Clonixina/sangre , Clonixina/química , Clonixina/metabolismo , Clonixina/farmacocinética , Residuos de Medicamentos , Semivida , Hígado/química , Músculo Esquelético/química , Músculo Esquelético/metabolismo
10.
J Food Prot ; 77(2): 292-8, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24490924

RESUMEN

A simple, cow-side test for the presence of drug residues in live animal fluids would provide useful information for tissue drug residue avoidance programs. This work describes adaptation and evaluation of rapid screening tests to detect drug residues in serum and urine. Medicated heifers had urine, serum, and tissue biopsy samples taken while on drug treatment. Samples were tested by rapid methods and high-performance liquid chromatography (HPLC). The adapted microbial inhibition method, kidney inhibition swab test, was useful in detecting sulfadimethoxine in serum, and its response correlated with the prescribed withdrawal time for the drug, 5 to 6 days posttreatment. The lateral flow screening method for flunixin and beta-lactams, adapted for urine, was useful in predicting flunixin in liver detected by HPLC, 96 h posttreatment. The same adapted methods were not useful to detect ceftiofur in serum or urine due to a lack of sensitivity at the levels of interest. These antemortem screening test studies demonstrated that the method selected, and the sampling matrix chosen (urine or serum), will depend on the drug used and should be based on animal treatment history if available. The live animal tests demonstrated the potential for verification that an individual animal is free of drug residues before sale for human consumption.


Asunto(s)
Antibacterianos/análisis , Bovinos/metabolismo , Residuos de Medicamentos/análisis , Animales , Antibacterianos/sangre , Antibacterianos/orina , Cefalosporinas/metabolismo , Cromatografía Líquida de Alta Presión/métodos , Clonixina/análogos & derivados , Clonixina/metabolismo , Femenino , Riñón/química , Hígado/química , beta-Lactamas/análisis
11.
Artículo en Inglés | MEDLINE | ID: mdl-23789859

RESUMEN

5-Hydroxy-flunixin concentrations in milk samples were quantified by two commercially available screening assays--CHARM® and enzyme-linked immunoabsorbant assay (ELISA)--to determine whether any concentrations could be detected above the tolerance limit of 2 ng g⁻¹ from different regions in the United States. Milk samples came from large tanker trucks hauling milk to processing plants, and had already been screened for antibiotics. Positive results for flunixin residues based on a screening assay were confirmed by ultra-HPLC with mass spectrometric detection. Of the 500 milk samples analysed in this study, one sample was found to have a 5-hydroxy-flunixin concentration greater than the tolerance limit. The results of this study indicate that flunixin residues in milk are possible. Regulatory agencies should be aware that such residues can occur, and should consider incorporating or expanding flunixin screening tests as part of routine drug monitoring in milk. Larger studies are needed to determine the true prevalence of flunixin residues in milk from other regions in the United States as well as different countries.


Asunto(s)
Antiinflamatorios no Esteroideos/metabolismo , Clonixina/análogos & derivados , Residuos de Medicamentos/análisis , Contaminación de Alimentos , Inspección de Alimentos , Leche/química , Animales , Bovinos , Cromatografía Líquida de Alta Presión , Clonixina/análisis , Clonixina/metabolismo , Industria Lechera/legislación & jurisprudencia , Ensayo de Inmunoadsorción Enzimática , Contaminación de Alimentos/legislación & jurisprudencia , Manipulación de Alimentos , Inspección de Alimentos/legislación & jurisprudencia , Adhesión a Directriz , Legislación Alimentaria , Leche/normas , Espectrometría de Masa por Ionización de Electrospray , Transportes , Estados Unidos , United States Department of Agriculture , United States Food and Drug Administration
12.
J Vet Pharmacol Ther ; 36(6): 571-5, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23398483

RESUMEN

Residues of flunixin [and its marker residue 5-hydroxyflunixin (5OHFLU)] were determined in milk from cows that intravenously received therapeutic doses of the drug. The samples were collected during each milking (every 12 h) for six consecutive days, and concentrations of flunixin and its metabolites were determined by the method with and without enzymatic hydrolysis (beta-glucuronidase). The highest flunixin concentration in milk was observed 12 h after dosing (2.4 ± 1.42 µg/kg, mean ± SD). Flunixin concentrations in the samples determined with enzymatic hydrolysis were significantly higher (P < 0.05), which suggests the transfer of flunixin glucuronide to the milk. Additionally, unambiguous identification of flunixin glucuronide in the bovine milk was performed with linear ion-trap mass spectrometry. The 5OHFLU concentrations analyzed without enzymatic hydrolysis (22.3 ± 16.04 µg/kg) were similar to this obtained with enzymatic hydrolysis. Flunixin and 5OHFLU concentrations dropped below the limits of detection at 48 h after last dosing.


Asunto(s)
Bovinos/metabolismo , Clonixina/análogos & derivados , Residuos de Medicamentos , Leche/química , Animales , Clonixina/sangre , Clonixina/química , Clonixina/metabolismo , Clonixina/farmacocinética , Femenino
14.
J Vet Pharmacol Ther ; 27(2): 65-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15096102

RESUMEN

The plasma and urine pharmacokinetics of flunixin-meglumine (FNX) in cats were examined using a total of 12 adult animals. After an intravenous injection of FNX (2 mg/kg), the plasma concentration time curves showed a profile of a two-compartment open model with an elimination half-life of 6.6 h. In spite of high plasma protein binding (>99%), the V(d)beta was unusually large, 0.7 L/kg. Although the recovery of FNX from urine was only 0.4% of the dose, the estimated inherent renal clearance closely corresponded to the renal plasma flow rate, indicating that a renal active tubular secretion was involved in the pharmacokinetics of FNX. Cholestyramine (ChSA), an anion exchanger, was orally administered immediately before the FNX injection in order to determine the involvement of enterohepatic circulation in FNX pharmacokinetics. The elimination phase of the profile of FNX was prevented by the concomitant administration of ChSA, so it was concluded that the drug undergoes enterohepatic circulation in cats. Pravastatin (PV) is a specific substrate of the type-2 organic anion transporting polypeptide transporter (OATP-2) in human liver cells. The effect of a concomitant intravenous injection of PV with FNX was examined in order to determine the involvement of OATP-2 like transporter in the pharmacokinetics. The V1 and total body clearance were decreased after the injection of PV. In conclusion, at least two active transport mechanisms are involved in the pharmacokinetics of FNX in cats. One pathway is renal tubular secretion and the other is sinusoidal active uptake by liver cells. The latter may be responsible for the enterohepatic circulation of FNX in cats.


Asunto(s)
Resinas de Intercambio Aniónico/farmacología , Antiinflamatorios no Esteroideos/farmacocinética , Resina de Colestiramina/farmacología , Clonixina/análogos & derivados , Clonixina/farmacocinética , Circulación Enterohepática/efectos de los fármacos , Animales , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/farmacología , Área Bajo la Curva , Transporte Biológico Activo , Proteínas Sanguíneas/metabolismo , Gatos , Clonixina/metabolismo , Clonixina/farmacología , Interacciones Farmacológicas , Semivida
15.
J Chromatogr A ; 768(1): 67-71, 1997 Apr 18.
Artículo en Inglés | MEDLINE | ID: mdl-9175276

RESUMEN

This paper demonstrates the use of micellar electrokinetic capillary chromatography (MECC) to monitor enzyme reaction conditions. The hydrolysis reactions of model conjugated substrates (morphine and reduced flunixin glucuronides, napthyl sulfate), by proprietary beta-glucuronidase preparations, were studied under varied experimental conditions. Reactions were carried out in autosampler vials with incubation in a thermostatted CE autosampler tray. MECC was performed using borax buffer (17.5 mM, pH 9.3) modified with sodium dodecyl sulfate (70 mM). Repetitive injections were made from the sample vial throughout the course of the reactions at a frequency of up to 10 h-1. MECC provided a rapid and reproducible assay for the model substrates. Baseline interference from the enzymes prevented measurement of product increase, therefore substrate decrease was measured from the peak areas. Monitoring of reactions in this way has proved valuable in the optimisation of hydrolysis conditions used in sample preparation for drug analysis. beta-Glucuronidase preparations from Helix pomatia were found to give the best performance of those evaluated in terms of deconjugation efficiency.


Asunto(s)
Electroforesis Capilar/métodos , Glucuronatos/análisis , Glucuronidasa/metabolismo , Sulfatos/análisis , Animales , Clonixina/análogos & derivados , Clonixina/metabolismo , Glucuronatos/metabolismo , Caracoles Helix/enzimología , Concentración de Iones de Hidrógeno , Hidrólisis , Cinética , Naftalenos/análisis , Sulfatos/metabolismo , Ésteres del Ácido Sulfúrico/metabolismo , Temperatura
16.
Equine Vet J Suppl ; (11): 62-5, 1992 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-9109964

RESUMEN

The effects of access to hay and of restricted feeding on the pharmacokinetics of flunixin administered orally to six healthy ponies were compared in a cross-over study. No access to feed for a few hours before and after flunixin administration resulted in rapid absorption with a mean peak plasma concentration of 2.84 +/- 0.28 micrograms/ml attained in an average time of 0.76 +/- 0.18 h, followed by an exponential decline in plasma concentration. A lower peak plasma concentration was obtained when ponies had free access to hay before and after drug dosing. The mean maximum concentration (Cmax) was 1.30 +/- 0.23 micrograms/ml and maximum time (tmax) was prolonged to a mean time of 7.66 +/- 1.74 h. Free access to hay reduced and delayed the peak plasma concentration resulting in two or three separate concentration peaks in some ponies. The mean area under the plasma concentration-time curve was not significantly different for the two feeding regimens.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Clonixina/análogos & derivados , Ingestión de Alimentos/fisiología , Caballos/metabolismo , Absorción Intestinal/fisiología , Administración Oral , Alimentación Animal/normas , Animales , Antiinflamatorios no Esteroideos/administración & dosificación , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/metabolismo , Recolección de Muestras de Sangre/veterinaria , Cromatografía Líquida de Alta Presión/veterinaria , Clonixina/administración & dosificación , Clonixina/sangre , Clonixina/metabolismo , Clonixina/farmacocinética , Estudios Cruzados , Femenino , Masculino
17.
J Vet Pharmacol Ther ; 13(1): 93-104, 1990 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-2319641

RESUMEN

A study of the effects of intravenous administration of either 150 mg or 250 mg of furosemide to standardbred mares pre-treated with other drugs was undertaken to determine whether a unique pattern of drug elimination into urine and from plasma for each compound occurred. Furosemide significantly reduced the plasma concentrations of codeine compared to control 2-6 h after furosemide administration. In contrast, the plasma concentrations of theophylline, phenylbutazone, pentazocine, guaifenesin and flunixin were not markedly altered by furosemide. In the case of acepromazine, clenbuterol and fentanyl, the data generated were insufficient to state with certainty whether or not furosemide affected the plasma concentrations of these three drugs. A significant reduction was noted in the urinary concentrations of guaifenesin, acepromazine, clenbuterol, phenylbutazone, flunixin, fentanyl and pentazocine within 1-4 h of furosemide administration. The urinary concentrations of theophylline remained reduced as long as 8 h after furosemide injection. Furosemide administration to horses pre-treated with codeine resulted in depression of urinary morphine concentrations 2-4 h and 9-12 h after furosemide injection. A lower furosemide dose (150 mg) produced changes in drug urinary excretion and plasma elimination equivalent to the higher dose (250 mg). It is evident that furosemide affects the urinary and plasma concentrations of other co-administered drugs but not in a predictable fashion, which limits the extrapolation of these results to as yet untested drugs.


Asunto(s)
Furosemida/farmacología , Caballos/metabolismo , Premedicación/veterinaria , Acepromazina/metabolismo , Animales , Clenbuterol/metabolismo , Clonixina/análogos & derivados , Clonixina/metabolismo , Codeína/metabolismo , Interacciones Farmacológicas , Femenino , Fentanilo/metabolismo , Furosemida/administración & dosificación , Caballos/orina , Inyecciones Intravenosas/veterinaria , Morfina/metabolismo , Pentazocina/metabolismo , Fenilbutazona/metabolismo , Esfuerzo Físico , Teofilina/metabolismo
18.
Equine Vet J ; 19(3): 201-6, 1987 May.
Artículo en Inglés | MEDLINE | ID: mdl-3608957

RESUMEN

The efficacy of low doses of flunixin meglumine in reducing eicosanoid generation and clinical signs in response to experimentally induced endotoxaemia was investigated. Thromboxane B2 and 6-keto-prostaglandin F1 alpha were measured in serum and plasma by radioimmunoassay. Plasma flunixin concentrations were determined by high performance liquid chromatography and pharmacokinetic parameters derived non-compartmentally. In horses administered flunixin meglumine before endotoxin challenge, a significant suppression in plasma thromboxane B2 and 6-keto-prostaglandin F1 alpha generation was observed. Elevations in blood lactate were significantly suppressed in horses pretreated with 0.25 mg/kg bodyweight flunixin meglumine. Reduction of the clinical signs of endotoxaemia by flunixin meglumine was dose dependent. Low doses of flunixin inhibited eicosanoid production without masking all of the physical manifestations of endotoxaemia necessary for accurate clinical evaluation of the horse's status.


Asunto(s)
6-Cetoprostaglandina F1 alfa/sangre , Clonixina/farmacología , Enfermedades de los Caballos/metabolismo , Ácidos Nicotínicos/farmacología , Tromboxano B2/sangre , Toxemia/veterinaria , Animales , Clonixina/análogos & derivados , Clonixina/metabolismo , Clonixina/uso terapéutico , Endotoxinas , Enfermedades de los Caballos/tratamiento farmacológico , Caballos , Cinética , Lactatos/sangre , Distribución Aleatoria , Toxemia/tratamiento farmacológico , Toxemia/metabolismo
19.
Ann Rech Vet ; 17(4): 353-62, 1986.
Artículo en Francés | MEDLINE | ID: mdl-3493724

RESUMEN

Flunixin is a non-steroidal anti-inflammatory agent, with a potent analgesic activity and a slight toxicity. It is largely used in horses, in the form of meglumine salt, for the treatment of inflammatory diseases or colics, and often identified in dopage cases. Physical and chemical properties of the drug, its pharmacological and toxicological properties, and its use in equine species are depicted.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Clonixina/uso terapéutico , Cólico/veterinaria , Enfermedades de los Caballos/tratamiento farmacológico , Ácidos Nicotínicos/uso terapéutico , Animales , Fenómenos Químicos , Química , Clonixina/efectos adversos , Clonixina/análogos & derivados , Clonixina/metabolismo , Cólico/tratamiento farmacológico , Doping en los Deportes/métodos , Caballos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...