Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Front Vet Sci ; 7: 586, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-33005646

RESUMEN

Castration and tail-docking of pre-wean piglets are common procedures that are known to induce pain and would benefit from pain mitigation. Flunixin meglumine (FM) is a non-steroidal anti-inflammatory drug currently approved in the United States for pyrexia in swine and lameness pain in cattle. The objective of this study was to establish the pharmacokinetic (PK) parameters resulting from intravenous (IV), intramuscular (IM), oral (PO) and transdermal (TD) administration of FM in pre-wean piglets. FM was administered to thirty-nine pre-wean piglets at a target dose of 2.2 mg/kg for IV and IM and 3.3 mg/kg for PO and TD route. Plasma was collected at twenty-seven time points from 0 to 9 days after FM administration and concentrations were determined using ultra-high performance liquid chromatography coupled with mass spectrometry (UPLC-MS). Pharmacokinetic data were analyzed using noncompartmental analysis (NCA) methods and nonlinear mixed-effects (NLME). Initial plasma concentration for IV (C0) 11,653 µg/L and mean peak plasma concentrations (Cmax) 6,543 µg/L (IM), 4,883 µg/L (PO), and 31.5 µg/L (TD) were measured. The time points of peak FM concentrations (tmax) were estimated 30 min, 1 h, and 24 h for IM, PO, and TD, respectively. The bioavailability (F) of PO and IM FM was estimated at >99%, while the bioavailability of TD FM was estimated to be 7.8%. The reported Cmax of FM after IM and PO administration is consistent with therapeutic concentration ranges that mitigate pain in other species and adult pigs. However, the low estimated concentration of FM after TD dosing is not expected to mitigate pain in pre-wean piglets. The low F of TD FM suggests that expanding the surface area of application is unlikely to be sufficient to establish an effective TD dose for pain, while the high bioavailability for PO FM should allow for an effective dose regimen to be established.

2.
BMC Vet Res ; 16(1): 211, 2020 Jun 22.
Artículo en Inglés | MEDLINE | ID: mdl-32571315

RESUMEN

BACKGROUND: Flunixin meglumine (FM) was investigated for the effectiveness of plasma, oral fluid, and urine concentrations to predict tissue residue depletion profiles in finishing-age swine, along with the potential for untreated pigs to acquire tissue residues following commingled housing with FM-treated pigs. Twenty pigs were housed in groups of three treated and one untreated control. Treated pigs received one 2.2 mg/kg dose of FM intramuscularly. Before treatment and at 1, 3, 6, 12, 24, 36, and 48 h (h) after treatment, plasma samples were taken. At 1, 4, 8, 12 and 16 days (d) post-treatment, necropsy and collection of plasma, urine, oral fluid, muscle, liver, kidney, and injection site samples took place. Analysis of flunixin concentrations using liquid chromatography/tandem mass spectrometry was done. A published physiologically based pharmacokinetic (PBPK) model for flunixin in cattle was extrapolated to swine to simulate the measured data. RESULTS: Plasma concentrations of flunixin were the highest at 1 h post-treatment, ranging from 1534 to 7040 ng/mL, and were less than limit of quantification (LOQ) of 5 ng/mL in all samples on Day 4. Flunixin was detected in the liver and kidney only on Day 1, but was not found 4-16 d post-treatment. Flunixin was either not seen or found less than LOQ in the muscle, with the exception of one sample on Day 16 at a level close to LOQ. Flunixin was found in the urine of untreated pigs after commingled housing with FM-treated pigs. The PBPK model adequately correlated plasma, oral fluid and urine concentrations of flunixin with residue depletion profiles in liver, kidney, and muscle of finishing-age pigs, especially within 24 h after dosing. CONCLUSIONS: Results indicate untreated pigs can be exposed to flunixin by shared housing with FM-treated pigs due to environmental contamination. Plasma and urine samples may serve as less invasive and more easily accessible biological matrices to predict tissue residue statuses of flunixin in pigs at earlier time points (≤24 h) by using a PBPK model.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Clonixina/análogos & derivados , Sus scrofa/fisiología , Animales , Antiinflamatorios no Esteroideos/sangre , Antiinflamatorios no Esteroideos/orina , Clonixina/sangre , Clonixina/farmacocinética , Clonixina/orina , Contaminación de Alimentos/análisis , Carne de Cerdo/análisis , Saliva/química
3.
Comp Med ; 69(5): 413-418, 2019 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-31581974

RESUMEN

Sheep are commonly used as animal models for human biomedical research, but descriptions of their use for studying the pharmacokinetics of carbapenem antimicrobials, such as ertapenem, are unavailable. Ertapenem is a critical antimicrobial for human infections, and the description of the pharmacokinetics of this drug is of value for research using ovine as models for human diseases, such as urinary tract infections (UTI). There are currently no ovine models for comparative biomedical research of UTI. The objective of this study was to report the pharmacokinetics of ertapenem in sheep after single and multiple dosing. In addition, we explored the effects of an immunomodulatory drug (Zelnate) on the pharmacokinetics of ertapenem in sheep. Eight healthy ewes (weight, 64.4 ± 7.7 kg) were used in an ovine bacterial cystitis model of human cystitis with Pseudomonas aeruginosa. After disease confirmation, each ewe received 1 g of ertapenem intravenously once every 24 h for 5 administrations. Blood was collected intensively (14 samples) during 24 h after the first and last administration. After multiple-dose administration, the volume of distribution was 84.5 mL/kg, clearance was 116.3 mL/h/kg, T1/2(λz) was 1.1 h, and the extraction ratio was 0.02. No significant differences in pharmacokinetic parameters or time points were found between groups treated with the immunostimulant and controls or after the 1st or 5th administration of ertapenem. No accumulation was noted from previous administration. Our ovine pharmacokinetic findings can be used to evaluate therapeutic strategies for ertapenem use (varying drug dosing schedules and combinations with other antimicrobials or immune modulators) in the context of UTI.


Asunto(s)
Antibacterianos/farmacocinética , Modelos Animales de Enfermedad , Ertapenem/farmacocinética , Pseudomonas aeruginosa/efectos de los fármacos , Ovinos , Animales , Humanos , Infecciones Urinarias/microbiología
4.
Vet Anaesth Analg ; 45(6): 849-857, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-30122634

RESUMEN

OBJECTIVE: To study the influence of pain on the pharmacokinetics and anti-inflammatory actions of transdermal flunixin administered at dehorning. STUDY DESIGN: Prospective, crossover, clinical study. ANIMALS: A total of 16 male Holstein calves, aged 6-8 weeks weighing 61.3 ± 6.6 kg. METHODS: Calves were randomly assigned to one of two treatments: transdermal flunixin and dehorning (PAIN) or transdermal flunixin and sham dehorning (NO PAIN). Flunixin meglumine (3.33 mg kg-1) was administered topically as a pour-on concurrently with hot iron dehorning or sham dehorning. The calves were subjected to the alternative treatment 14 days later. Blood samples were collected at predetermined time points up to 72 hours for measurement of plasma flunixin concentrations. Pharmacokinetics parameters were determined using noncompartmental analysis. Prostaglandin E2 (PGE2) concentration was determined using a commercial enzyme-linked immunosorbent assay. The 80% inhibition concentration (IC80) of PGE2 was determined using nonlinear regression. Pharmacokinetic data were statistically analyzed using paired t tests and Wilcoxon rank sums for nonparametric data. Flunixin and PGE2 concentrations were log transformed and analyzed using repeated measures. RESULTS: A total of 15 calves completed the study. Plasma half-life of flunixin was significantly longer in PAIN (10.09 hours) than NO PAIN (7.16 hours) (p = 0.0202). Bioavailability of transdermal flunixin was 30% and 37% in PAIN and NO PAIN, respectively (p = 0.097). Maximum plasma concentrations of flunixin were 0.95 and 1.16 µg mL-1 in PAIN and NO PAIN, respectively (p = 0.089). However, there was a treatment (PAIN versus NO PAIN) by time interaction (p = 0.0353). PGE2 concentrations were significantly lower in the PAIN treatment at 48 and 72 hours (p = 0.0092 and p = 0.0287, respectively). The IC80 of PGE2 by flunixin was similar in both treatments (p = 0.88). CONCLUSION AND CLINICAL RELEVANCE: Pain alters the pharmacokinetics and anti-inflammatory effects of transdermally administered flunixin.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacocinética , Bovinos/metabolismo , Clonixina/análogos & derivados , Dolor/veterinaria , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Bovinos/cirugía , Cauterización/veterinaria , Clonixina/farmacocinética , Clonixina/uso terapéutico , Cuernos/cirugía , Masculino , Dolor/metabolismo
5.
J Vet Pharmacol Ther ; 41(6): 848-860, 2018 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-29971798

RESUMEN

Ceftiofur (CEF) is a third-generation cephalosporin that is the most widely used antimicrobial in the dairy industry. Currently, violative meat residues in cull dairy cattle are commonly associated with CEF. One potential cause for violative residues is altered pharmacokinetics of the drug due to disease, which could increase the time needed for the residue to deplete. The objectives of this study were (a) to determine the absolute bioavailability of CEF crystalline-free acid (CFA) in healthy versus diseased cows; (b) to compare the plasma and interstitial fluid pharmacokinetics and plasma protein binding of CEF between healthy dairy cows and those with disease; and (c) to determine the CEF residue profile in tissues of diseased cows. For this trial, disease was induced through intramammary Escherichia coli infusion. Following disease induction and CEF CFA administration, for plasma concentrations, there was not a significant effect of treatment (p = 0.068), but the treatment-by-time interaction (p = 0.005) was significant. There was a significantly greater concentration of CEF in the plasma of the DIS cows at T2 hr (p = 0.002), T8 hr (p < 0.001), T12 hr (p = 0.001), and T16 hr (p = 0.002). For PK parameters in plasma, the slope of the terminal phase of the concentration versus time curve was significantly lower (p = 0.007), terminal half-life was significantly longer (p = 0.014), and apparent volume of distribution during the elimination phase was significantly higher (p = 0.028) diseased group. There was no difference in plasma protein binding of CEF and interstitial fluid pharmacokinetics. None of the cows had kidney CEF residues above the US tolerance level following observation of the drug's withdrawal period, but one cow with a larger apparent volume of distribution and longer terminal half-life had tissue residues slightly below the tolerance. Whereas these findings do not support the hypothesis that severely ill cows need longer withdrawal times, alterations in the terminal half-life suggest that it is theoretically possible.


Asunto(s)
Cefalosporinas/farmacocinética , Infecciones por Escherichia coli/veterinaria , Mastitis Bovina/microbiología , Animales , Disponibilidad Biológica , Bovinos , Cefalosporinas/uso terapéutico , Infecciones por Escherichia coli/sangre , Infecciones por Escherichia coli/tratamiento farmacológico , Femenino , Mastitis Bovina/sangre , Mastitis Bovina/tratamiento farmacológico , Distribución Tisular
6.
Am J Vet Res ; 79(5): 568-575, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29688783

RESUMEN

OBJECTIVE To determine the effect of age on the pharmacokinetics and pharmacodynamics of flunixin meglumine following IV and transdermal administration to calves. ANIMALS 8 healthy weaned Holstein bull calves. PROCEDURES At 2 months of age, all calves received an injectable solution of flunixin (2.2 mg/kg, IV); then, after a 10-day washout period, calves received a topical formulation of flunixin (3.33 mg/kg, transdermally). Blood samples were collected at predetermined times before and for 48 and 72 hours, respectively, after IV and transdermal administration. At 8 months of age, the experimental protocol was repeated except calves received flunixin by the transdermal route first. Plasma flunixin concentrations were determined by liquid chromatography-tandem mass spectroscopy. For each administration route, pharmacokinetic parameters were determined by noncompartmental methods and compared between the 2 ages. Plasma prostaglandin (PG) E2 concentration was determined with an ELISA. The effect of age on the percentage change in PGE2 concentration was assessed with repeated-measures analysis. The half maximal inhibitory concentration of flunixin on PGE2 concentration was determined by nonlinear regression. RESULTS Following IV administration, the mean half-life, area under the plasma concentration-time curve, and residence time were lower and the mean clearance was higher for calves at 8 months of age than at 2 months of age. Following transdermal administration, the mean maximum plasma drug concentration was lower and the mean absorption time and residence time were higher for calves at 8 months of age than at 2 months of age. The half maximal inhibitory concentration of flunixin on PGE2 concentration at 8 months of age was significantly higher than at 2 months of age. Age was not associated with the percentage change in PGE2 concentration following IV or transdermal flunixin administration. CONCLUSIONS AND CLINICAL RELEVANCE In calves, the clearance of flunixin at 2 months of age was slower than that at 8 months of age following IV administration. Flunixin administration to calves may require age-related adjustments to the dose and dosing interval and an extended withdrawal interval.


Asunto(s)
Administración Cutánea , Administración Intravenosa , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/farmacocinética , Clonixina/análogos & derivados , Factores de Edad , Animales , Antiinflamatorios no Esteroideos/sangre , Bovinos , Clonixina/sangre , Clonixina/farmacocinética , Clonixina/farmacología , Dinoprostona , Masculino
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...