Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 24
Filter
Add more filters










Publication year range
1.
J Vet Pharmacol Ther ; 40(1): 88-91, 2017 Jan.
Article in English | MEDLINE | ID: mdl-27292121

ABSTRACT

The objective of this study was to determine the pharmacokinetics of single- and multi-dose ceftiofur crystalline-free acid (CCFA) administered subcutaneously at a dose of 13.2 mg/kg to 12 neonatal foals 1-3 days of age. Six foals received a single subcutaneous dose, while 6 additional foals received 4 doses of CCFA at 48-h intervals. Blood samples were collected at pre-determined times following drug administration, and plasma concentrations of ceftiofur free acid equivalents (CFAE) were measured using high-performance liquid chromatography. Following single-dose administration of CCFA, the mean ± standard deviation maximum observed plasma concentration was 3.1 ± 0.6 µg/mL and observed time to maximal plasma concentration was 14.0 ± 4.9 h. Following multi-dose administration of CCFA, the mean ±standard deviation times above CFAE concentrations of ≥0.5 µg/mL and ≥2.0 µg/mL were 192.95 ± 15.86 h and 78.80 ± 15.31 h, respectively. The mean ± standard deviation area under the concentration vs time curve (AUC0→∝ ) was 246.2 ± 30.7 h × µg/mL and 172.7 ± 27.14 h × µg/mL following single- and multi-dose CCFA administrations, respectively. Subcutaneous administration of CCFA at 13.2 mg/kg in neonatal foals was clinically well- tolerated and resulted in plasma concentrations sufficient for the treatment of most bacterial pathogens associated with neonatal foal septicemia. Multi-dose administration of four doses at dosing interval of 48 h between treatments maintains appropriate therapeutic concentrations in neonatal foals.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Cephalosporins/pharmacokinetics , Animals , Animals, Newborn/metabolism , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Cephalosporins/administration & dosage , Cephalosporins/blood , Chromatography, High Pressure Liquid/veterinary , Delayed-Action Preparations , Drug Administration Schedule , Female , Horses/metabolism , Injections, Subcutaneous/veterinary , Male
2.
Avian Dis ; 58(1): 8-15, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24758107

ABSTRACT

Ring-necked pheasants raised on propagation farms can be severely parasitized with Syngamus trachea (gapeworm) and other parasitic worms. Fenbendazole is a highly effective benzimidazole-class anthelmintic that is not currently approved for game bird species in the United States. The objective of this work was to provide target animal safety data to support a label claim for fenbendazole in pheasants at 100 parts per million (ppm) in the feed for 7 consecutive days. Demonstration of safety in young pheasants and a separate demonstration of reproductive safety in adult birds were required. In the young bird study, 160 Chinese ring-necked pheasants (Phasianus colchicus, 80 males and 80 females) were fed a commercial game bird starter ration containing no antibiotics, growth promoters, or coccidiostats until day 0 of the study (approximately 21 days of age). On day 0 the birds were placed on their respective study diets containing fenbendazole at 0, 100, 300, and 500 ppm for 21 days (three times the normal treatment duration). Clinical observations were recorded twice daily. Feed consumption, feed conversion rate, and body weights were determined for each pen. Three birds from each pen were randomly selected for necropsy, histopathology, and clinical pathology. Birds were carefully examined for feathering abnormalities immediately following euthanasia. The remaining birds in each pen were submitted for drug concentration analysis so that concentrations (for low vs. high treatment levels) could be correlated with clinical observations, clinical pathology, and histologic findings. There no morbidities or mortalities after study day--1. There were no statistically significant treatment-related differences in feed consumption, feed conversion rates, body weights, serum biochemistry profiles, hematologic profiles, gross necropsy findings, histopathologic examination, and feathering. Allowable liver and muscle concentrations of fenbendazole sulfone in turkeys are 6 and 2 ppm, respectively, with a 6-hr feed withdrawal. Analysis of fenbendazole concentrations in kidney, liver, leg/thigh, and breast muscle and skin with associated fat revealed that, even at the highest dose level used and with no feed withdrawal, fenbendazole concentrations were relatively low in these tissues. These findings indicate that fenbendazole has a relatively wide margin of safety in young pheasants and that the proposed dose of 100 ppm in the feed for 7 consecutive days is well within the margin of safety. In the reproductive safety study, two large game bird farms fed fendbendazole at 100 ppm for 7 days and collected data on hatching percentage of pheasant eggs before and after treatment. Reproductive performance in hen pheasants was not adversely affected.


Subject(s)
Antinematodal Agents/adverse effects , Antinematodal Agents/metabolism , Fenbendazole/adverse effects , Fenbendazole/metabolism , Galliformes/metabolism , Animal Feed/analysis , Animal Nutritional Physiological Phenomena , Animals , Body Weight/drug effects , Chromatography, High Pressure Liquid/veterinary , Dose-Response Relationship, Drug , Energy Metabolism/drug effects , Feeding Behavior/drug effects , Female , Male , Reproduction/drug effects , Tissue Distribution
3.
J Vet Pharmacol Ther ; 37(2): 205-8, 2014 Apr.
Article in English | MEDLINE | ID: mdl-23889045

ABSTRACT

Eight adult female dairy goats received one subcutaneous administration of tulathromycin at a dosage of 2.5 mg/kg body weight. Blood and milk samples were assayed for tulathromycin and the common fragment of tulathromycin, respectively, using liquid chromatography/mass spectrometry. Pharmacokinetic disposition of tulathromycin was analyzed by a noncompartmental approach. Mean plasma pharmacokinetic parameters (±SD) following single-dose administration of tulathromycin were as follows: C(max) (121.54 ± 19.01 ng/mL); T(max) (12 ± 12-24 h); area under the curve AUC(0→∞) (8324.54 ± 1706.56 ng·h/mL); terminal-phase rate constant λz (0.01 ± 0.002 h⁻¹); and terminal-phase rate constant half-life t1/2λz (67.20 h; harmonic). Mean milk pharmacokinetic parameters (±SD) following 45 days of sampling were as follows: Cmax (1594 ± 379.23 ng/mL); Tmax (12 ± 12-36 h); AUC(0→∞) (72,250.51 ± 18,909.57 ng·h/mL); λz (0.005 ± 0.001 h⁻¹); and t(1/2λz) (155.28 h; harmonic). All goats had injection-site reactions that diminished in size over time. The conclusions from this study were that tulathromycin residues are detectable in milk samples from adult goats for at least 45 days following subcutaneous administration, this therapeutic option should be reserved for cases where other treatment options have failed, and goat milk should be withheld from the human food chain for at least 45 days following tulathromycin administration.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Disaccharides/pharmacokinetics , Goats/blood , Heterocyclic Compounds/pharmacokinetics , Milk/chemistry , Animals , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/chemistry , Area Under Curve , Disaccharides/blood , Disaccharides/chemistry , Drug Residues/chemistry , Drug Residues/metabolism , Female , Half-Life , Heterocyclic Compounds/blood , Heterocyclic Compounds/chemistry
4.
J Vet Pharmacol Ther ; 36(1): 59-67, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22449008

ABSTRACT

The objective of this study was to determine the pharmacokinetics of CCFA in mares with placentitis and evaluate the disposition of the drug in fetal fluids, fetal membranes, colostrum, and serum of foals. A secondary objective was to obtain pilot data regarding the efficacy of CCFA for improving foal survival in mares with placentitis. Twelve pregnant pony mares were enrolled in the study, inoculated with Streptococcus zooepidemicus, intracervically and assigned to one of three groups: CEFT (n = 3; administered CCFA only; 6.6 mg/kg, i.m., q96h); COMBO (n = 6; administered combination therapy of CCFA, altrenogest, and pentoxifylline); UNTREAT (n = 3, no treatment). Treatment was initiated at the onset of clinical signs. Concentrations of desfuroylceftiofur acetamide (DCA), the acetamide derivative of ceftiofur and desfuroylceftiofur metabolites, were measured using high-performance liquid chromatography. Maximum and minimum serum concentrations of DCA at steady state in treated mares were 2.40±0.40 µg/mL and 1.06±0.29 µg/mL, respectively. Concentration of DCA in colostrum was 1.51±0.60 µg/mL. DCA concentrations in placenta and fetal tissues were very low (median = 0.03 µg/mL) and below the minimum inhibitory concentration of relevant pathogens. DCA was not detected in amniotic fluid or foal serum. Treatment did not appear to improve foal survival (CEFT: 0/3; COMBO: 2/6; UNTREAT: 2/3). Bacteria were recovered from the uterus of most mares postpartum and from blood cultures of most foals regardless of treatment.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Cephalosporins/analysis , Cephalosporins/pharmacokinetics , Placenta Diseases/veterinary , Animals , Anti-Bacterial Agents/analysis , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/therapeutic use , Cephalosporins/blood , Cephalosporins/therapeutic use , Colostrum/chemistry , Extraembryonic Membranes/chemistry , Female , Fetus/chemistry , Horses/metabolism , Placenta/chemistry , Placenta Diseases/drug therapy , Pregnancy
5.
J Vet Pharmacol Ther ; 36(2): 122-9, 2013 Apr.
Article in English | MEDLINE | ID: mdl-22486358

ABSTRACT

Six adult male alpacas received one subcutaneous administration of ceftiofur crystalline free acid (CCFA) at a dosage of 6.6 mg/kg. After a washout period, the same alpacas received three subcutaneous doses of 6.6 mg/kg CCFA at 5-day intervals. Blood samples collected from the jugular vein before and at multiple time points after each CCFA administration were assayed for ceftiofur- and desfuroylceftiofur-related metabolite concentrations using high-performance liquid chromatography. Pharmacokinetic disposition of CCFA was analyzed by a noncompartmental approach. Mean pharmacokinetic parameters (± SD) following single-dose administration of CCFA were Cmax (2.7 ± 0.9 µg/mL); Tmax (36 ± 0 h); area under the curve AUC0→∞ (199.2 ± 42.1 µg·h/mL); terminal phase rate constant λz (0.02 ± 0.003/h); and terminal phase rate constant half-life t1/2λz (44.7 h; harmonic). Mean terminal pharmacokinetic parameters (±SD) following three administrations of CCFA were Cmax (2.0 ± 0.4 µg/mL); Tmax (17.3 ± 16.3 h); AUC0→∞ (216.8 ± 84.5 µg·h/mL); λz (0.01 ± 0.003/h); and t1/2λz (65.9 h; harmonic). The terminal phase rate constant and the Tmax were significantly different between single and multiple administrations. Local reactions were noted in two alpacas following multiple CCFA administrations.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Camelids, New World/metabolism , Cephalosporins/pharmacokinetics , Animals , Anti-Bacterial Agents/administration & dosage , Area Under Curve , Bacteria/drug effects , Cephalosporins/administration & dosage , Drug Administration Schedule , Half-Life , Injections, Subcutaneous/veterinary , Male , Microbial Sensitivity Tests
6.
J Vet Pharmacol Ther ; 35(2): 113-20, 2012 Apr.
Article in English | MEDLINE | ID: mdl-21762403

ABSTRACT

Tulathromycin is a macrolide antimicrobial labeled for treatment of bacterial pneumonia in cattle and swine. The purpose of the present research was to evaluate tissue concentrations of tulathromycin in the caprine species. A tandem mass spectrometry regulatory analytical method that detects the common fragment of tulathromycin in cattle and swine was validated with goat tissues. The method was used to study tulathromycin depletion in goat tissues (liver, kidney, muscle, fat, injection site, and lung) over time. In two different studies, six juvenile and 25 market-age goats received a single injection of 2.5 mg/kg of tulathromycin subcutaneously; in a third study, 18 juvenile goats were treated with 2.5, 7.5, or 12.5 mg/kg tulathromycin weekly with three subcutaneous injections. Mean tulathromycin tissue concentrations were highest at injection site samples in all studies and all doses. Lung tissue concentrations were greatest at day 5 in market-age goats while in the multi-dose animals concentrations demonstrated dose-dependent increases. Concentrations were below limit of quantification in injection site and lung by day 18 and in liver, kidney, muscle, and fat at all time points. This study demonstrated that tissue levels in goats are very similar to those seen in swine and cattle.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Disaccharides/administration & dosage , Disaccharides/pharmacokinetics , Goats/metabolism , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/pharmacokinetics , Adipose Tissue , Animals , Anti-Bacterial Agents/blood , Disaccharides/blood , Dose-Response Relationship, Drug , Drug Administration Schedule , Drug Residues , Female , Heterocyclic Compounds/blood , Infusions, Subcutaneous , Kidney , Liver , Lung , Male , Muscle, Skeletal , Reproducibility of Results , Tissue Distribution
7.
J Vet Pharmacol Ther ; 34(5): 448-54, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21366621

ABSTRACT

Tulathromycin, a novel triamilide in the macrolide class, is labeled for treatment of bacterial pneumonia in cattle and swine. This manuscript evaluates pharmacokinetics of tulathromycin in goats. In two different studies, six juvenile and ten market-age goats received a single injection of 2.5 mg/kg of tulathromycin subcutaneously; in a third study, 18 juvenile goats were treated with 2.5, 7.5, or 12.5 mg/kg tulathromycin weekly with three subcutaneous injections. Pharmacokinetic parameters estimated from the plasma concentrations from single injections were similar between the two groups of goats and to previously reported parameters in cattle and swine. Mean terminal half-lives were 59.1 ± 7.6 and 61.2 ± 8.7 h for juvenile and market-age goats, respectively. In the multi-dose study, pharmacokinetic parameters estimated from plasma concentrations demonstrated significant differences at P < 0.05 among repeated injections but not among doses. Overall, pharmacokinetic parameters in goats are similar to those reported in cattle and swine, and tulathromycin may prove a useful drug for treating respiratory disease in goats.


Subject(s)
Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/pharmacokinetics , Disaccharides/administration & dosage , Disaccharides/pharmacokinetics , Goats/blood , Heterocyclic Compounds/administration & dosage , Heterocyclic Compounds/pharmacokinetics , Aging , Animals , Anti-Bacterial Agents/blood , Area Under Curve , Disaccharides/blood , Drug Administration Schedule , Female , Half-Life , Heterocyclic Compounds/blood , Male
8.
J Vet Pharmacol Ther ; 34(4): 403-9, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21083666

ABSTRACT

Ceftiofur, a third generation cephalosporin, demonstrates in vitro efficacy against microorganisms isolated from septicemic neonatal foals. This pharmacokinetic study evaluated the intravenous and subcutaneous administration of ceftiofur sodium (5 mg/kg body weight; n = 6 per group) and subcutaneous administration of ceftiofur crystalline free acid (6.6 mg/kg body weight; n = 6) in healthy foals. Plasma ceftiofur- and desfuroylceftiofur-related metabolite concentrations were measured using high performance liquid chromatography following drug administration. Mean (±SD) noncompartmental pharmacokinetic parameters for i.v. and s.c. ceftiofur sodium were: AUC(0→∝) (86.4 ± 8.5 and 91 ± 22 h·µg/mL for i.v. and s.c., respectively), terminal elimination half-life (5.82 ± 1.00 and 5.55 ± 0.81 h for i.v. and s.c., respectively), C(max(obs)) (13 ± 1.9 µg/mL s.c.), T(max(obs)) (0.75 ± 0.4 h for s.c.). Mean (± SD) noncompartmental pharmacokinetic parameters for s.c. ceftiofur crystalline free acid were: AUC(0→∝) (139.53 ± 22.63 h·µg/mL), terminal elimination half-life (39.7 ± 14.7), C(max(obs)) (2.52 ± 0.35 µg/mL) and t(max(obs)) (11.33 ± 1.63 h). No adverse effects attributed to drug administration were observed in any foal. Ceftiofur- and desfuroylceftiofur-related metabolites reached sufficient plasma concentrations to effectively treat common bacterial pathogens isolated from septicemic foals.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Cephalosporins/pharmacokinetics , Horses/metabolism , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Cephalosporins/administration & dosage , Cephalosporins/blood , Chromatography, High Pressure Liquid/veterinary , Female , Injections, Intravenous/veterinary , Injections, Subcutaneous/veterinary , Male
9.
J Vet Pharmacol Ther ; 33(6): 605-9, 2010 Dec.
Article in English | MEDLINE | ID: mdl-21062314

ABSTRACT

Progesterone (P4)-impregnated intravaginal controlled internal drug-releasing devices (CIDRs) have been used worldwide for estrus synchronization in ruminants. CIDRs serve to place all treated animals in the luteal phase of the estrous cycle. The objectives of this study were to compare P4 concentrations in milk from normal reproductively cycling, CIDR-treated, and pregnant goats. CIDRs were placed in treatment goats on day 0 and removed on day 19. Milk was collected daily from day 0 to day 21 from control and CIDR-treated goats and for 5 consecutive days between 40 and 60 days of gestation from pregnant does. Milk P4 was plotted against time (in days) for each individual, and the area under the curve (AUC) was calculated as an estimate of total milk P4. The AUC(day 0-21) for control and CIDR-treated goats were 29.5 ± 11.9 and 33.7 ± 6.6 d·ng/mL, respectively (P = 0.77). The highest single-day and highest 5-day average P4 values for each animal were also compared among groups. Single-day peak P4 levels were 4.8 ± 1.5, 4.0 ± 1.0, and 6.0 ± 0.4 ng/mL for control, CIDR-treated, and pregnant goats (P = 0.42). The highest 5-day average P4 concentrations were 3.6 ± 1.3, 2.9 ± 1.8, and 4.2 ± 0.3 for control, CIDR-treated, and pregnant goats (P = 0.56). The results of this study show that intravaginal P4 CIDR devices inserted for 19 days in healthy goats resulted in milk P4 levels similar to or less than those endogenously produced during diestrus or pregnancy.


Subject(s)
Goats/metabolism , Milk/chemistry , Progesterone/administration & dosage , Animals , Drug Implants , Drug Residues/analysis , Estrus/drug effects , Female , Pregnancy , Pregnancy Outcome , Progesterone/analysis , Progesterone/pharmacokinetics , Radioimmunoassay/veterinary
10.
J Aquat Anim Health ; 21(1): 8-13, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19485120

ABSTRACT

Nile tilapia Oreochromis niloticus, walleye Sander vitreus, and hybrid striped bass (female white bass Morone chrysops x male striped bass M. saxatilis) were medicated with florfenicol (AQUAFLOR type A medicated article; Schering-Plough Animal Health, Summit, New Jersey) via a medicated ration of 10 mg florfenicol x kg fish body weight(-1) d(-1) for 10 d to compare the elimination kinetics of the test article. This study was part of a larger effort in support of a species grouping concept that could contribute to the regulatory approval process for therapeutic compounds for cultured fishes. The trials in this study were conducted at the ideal water temperature for each species and at the temperature 5 degrees C lower than the ideal. The test temperatures were 30 degrees C and 25 degrees C for Nile tilapia, 25 degrees C and 20 degrees C for both walleyes and hybrid striped bass. In all cases, the elimination kinetics of florfenicol were more rapid at higher temperatures. The time to reach the tolerance of 1 microg/g in muscle-skin, as set by the U.S. Food and Drug Administration for channel catfish Ictalurus punctatus and salmonids, ranged from 6.1 to 4.1 d for Nile tilapia, from 12.6 to 9.7 d for walleyes, and from 2.6 to 0.7 d for hybrid striped bass at temperatures between 20 degrees C and 30 degrees C.


Subject(s)
Anti-Bacterial Agents/metabolism , Drug Residues , Perciformes/metabolism , Thiamphenicol/analogs & derivatives , Administration, Oral , Animal Feed , Animals , Species Specificity , Thiamphenicol/metabolism
11.
J Aquat Anim Health ; 21(1): 14-7, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19485121

ABSTRACT

Nile tilapia Oreochromis niloticus were medicated with florfenicol (AQUAFLOR type A medicated article; Schering-Plough Animal Health, Summit, New Jersey) via a medicated ration at 15 mg florfenicol x kg fish body weight(-1) d(-1) for 10 d to compare the elimination kinetics of the test article in different size fish held at 25 degrees C. The groups of fish used in the study had mean weights of approximately 100, 250, and 500 g. In each trial, the fish were provided the medicated ration and 15 fish were processed at each of seven time points postfeeding for determination of the florfenicol concentration in serum and the florfenicol residue in the edible portion (composite muscle and skin). There was a trend toward shorter half-lives of elimination in the smaller fish. The elimination times in muscle-skin (times to reach the established tolerance concentration for channel catfish Ictalurus punctatus and salmonids of 1.0 microg florfenicol residue/g) and half-lives were 9.2 and 1.2 d (100 g), 8.6 and 1.7 d (250 g), and 12.7 and 2.2 d (500 g), respectively.


Subject(s)
Anti-Bacterial Agents/metabolism , Body Weight/physiology , Cichlids/physiology , Drug Residues , Thiamphenicol/analogs & derivatives , Administration, Oral , Animal Feed , Animals , Species Specificity , Thiamphenicol/metabolism
12.
J Vet Pharmacol Ther ; 32(6): 552-65, 2009 Dec.
Article in English | MEDLINE | ID: mdl-20444010

ABSTRACT

A physiologically based pharmacokinetic (PBPK) model was developed for midazolam in the chicken and extended to three other species. Physiological parameters included organ weights obtained from 10 birds of each species and blood flows obtained from the literature. Partition coefficients for midazolam in tissues vs. plasma were estimated from drug residue data obtained at slaughter. The avian models include separate compartments for venous plasma, liver, kidney, muscle, fat and all other tissues. An estimate of total body clearance from an earlier in vitro study was used as a starting value in the model, assuming almost complete removal of the parent compound by liver metabolism. The model was optimized for the chicken with plasma and tissue data from a pharmacokinetic study after intravenous midazolam (5 mg/kg) dose. To determine which parameters had the most influence on the goodness of fit, a sensitivity analysis was performed. The optimized chicken model was then modified for the turkey, pheasant and quail. The models were validated with midazolam plasma and tissue residue data in the turkey, pheasant and quail. The PBPK models in the turkey, pheasant and quail provided good predictions of the observed tissue residues in each species, in particular for liver and kidney.


Subject(s)
GABA Modulators/pharmacokinetics , Galliformes , Midazolam/pharmacokinetics , Adipose Tissue/metabolism , Animals , Drug Residues , GABA Modulators/blood , Kidney/metabolism , Liver/metabolism , Midazolam/blood , Models, Biological , Muscle, Skeletal/metabolism , Reproducibility of Results , Sensitivity and Specificity , Tissue Distribution
14.
J Vet Pharmacol Ther ; 30(5): 429-36, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17803735

ABSTRACT

In vivo plasma pharmacokinetics of midazolam hydrochloride (5 mg/kg i.v.) were determined in commercially raised broiler chickens, turkeys, ring-necked pheasants and bobwhite quail. Pharmacokinetic profiles of midazolam were similar for all four species, especially with regard to the area under the plasma drug concentration-time curve. Estimates of the half-life of elimination of midazolam were 0.42, 1.45, 1.90, and 9.71 h for turkeys, chickens, bobwhite quail, and pheasant, respectively. This was similar to the major metabolite (1-hydroxymidazolam). Elimination half-lives for 1-hydroxymidazolam were 1.35, 1.86, 1.97, and 13.97 h for turkey, chicken, bobwhite quail and pheasant, respectively. Elimination half-lives for 4-hydroxymidazolam were 0.76, 1.23, 2.85, and 13.82 h for chicken, turkey, pheasant, and bobwhite quail, respectively. In addition to traditional pharmacokinetic approaches to parameter estimation, a bootstrapping technique was employed to attempt to achieve more realistic approximations of the concentrations at later time-points.


Subject(s)
Birds/metabolism , Hypnotics and Sedatives/pharmacokinetics , Midazolam/pharmacokinetics , Animals , Area Under Curve , Chickens/metabolism , Female , Hypnotics and Sedatives/administration & dosage , Hypnotics and Sedatives/blood , Injections, Intravenous , Male , Midazolam/administration & dosage , Midazolam/blood , Quail/metabolism , Turkeys/metabolism
15.
J Vet Pharmacol Ther ; 28(6): 505-13, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16343282

ABSTRACT

The purpose of this study was to determine whether Japanese quail (Coturnix japonica) would serve as a pharmacokinetic animal model for two small companion parrots: cockatiels (Nymphicus hollandicus) and Poicephalus parrots. Oxytetracycline (OTC) was the pharmacologic agent chosen for this study as it is eliminated primarily by renal glomerular filtration and undergoes minimal metabolism. A single intravenous injection of 20 mg/kg oxytetracycline hydrochloride was administered to the three study groups and blood samples were obtained at 5, 10, 15, and 30 min post-OTC injection as well as 1, 2, 4, 8, 12 and 24 h post-OTC injection. Quantification of plasma OTC was accomplished using a standardized microbial inhibition assay. Naïve-pooled data (NPD) analysis of the plasma concentration-time profile of OTC best fit a two-compartment open model for all three avian species. Noncompartmental analysis of the mean data yielded the following parameters for quail, cockatiels and Poicephalus parrots respectively: lambda(z) = 3.14, 4.57, 3.71 h; AUC = 38.9, 42.7, 49.6 microg x h/mL; and Cl = 514, 468, 403 mL/h/kg. Based on the similarity of these pharmacokinetic parameters, it appears that quail could be used as a model species to predict the appropriate OTC dosing regimen for small psittacine birds. A bootstrap procedure was also applied to these sparse data sets for both compartmental and noncompartmental analysis. The bootstrap procedure allowed for the calculation of variability of parameters; however, the estimates of the parameters were very similar to those calculated using the NPD and the data mean values.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Coturnix/metabolism , Oxytetracycline/pharmacokinetics , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Anti-Bacterial Agents/urine , Area Under Curve , Cockatoos/metabolism , Female , Glomerular Filtration Rate , Injections, Intravenous/veterinary , Kidney/metabolism , Male , Models, Animal , Oxytetracycline/administration & dosage , Oxytetracycline/blood , Oxytetracycline/urine , Parrots/metabolism
16.
J Vet Pharmacol Ther ; 27(4): 191-6, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15305846

ABSTRACT

Pharmacokinetic parameters of florfenicol were determined in 10 adult sheep (five wethers and five ewes) after a single 40 mg/kg intravenous (i.v.) dose, and three daily subcutaneous (s.c.) doses of 40 mg/kg of a commercial preparation (Nuflor((R))). The concentration of florfenicol in serum samples was assayed using a proprietary HPLC assay method, and pharmacokinetic parameters derived for individual animal data by each route using compartmental and noncompartmental approaches. Two animals (one male and one female) were excluded due to observed i.v. dosing problems, and a biexponential model was found to fit the i.v. data well for six of the other eight animals. Data from two males showed prolonged low concentrations of florfenicol in serum and were better fit by a three-compartment model. The mean +/- SD for the half-lives of the distribution and elimination phases for the six sheep best fit with a two-compartment model were 0.069 +/- 0.018 and 1.01 +/- 0.09 h respectively, and for the V(d(ss)) and clearances were 0.503 +/- 0.035 L/kg and 366 +/- 53 mL/h/kg respectively. The data collected during the s.c. multiple dose study were analyzed using noncompartmental methods only. The bioavailability (F%) after s.c. dosing was calculated in three ways to compare estimation methods as steady-state had not been reached and single dose s.c. data were not obtained past 24 h. Using the AUC(0--24) and AUC(0--> infinity ) from the first dose, the F% values averaged 27 and 40% respectively. Using the AUC(0--> infinity ) for all doses, the F% was 65%. Calculations of the mean time during which the serum concentration exceeded 0.5 and 1.0 microg/mL were 105 +/- 3.9 and 74.7 +/- 12.2 h respectively.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Sheep/metabolism , Thiamphenicol/analogs & derivatives , Thiamphenicol/pharmacokinetics , Animals , Anti-Bacterial Agents/administration & dosage , Area Under Curve , Female , Injections, Intravenous/veterinary , Injections, Subcutaneous/veterinary , Male , Thiamphenicol/administration & dosage
17.
J Vet Pharmacol Ther ; 25(1): 25-32, 2002 Feb.
Article in English | MEDLINE | ID: mdl-11874523

ABSTRACT

Separate groups of goats were used to determine drug depletion patterns in serum (n=10), tissue (n=20) and milk (n=8) following a single intramuscular (i.m.) dose of 20 mg/kg of a long-acting oxytetracycline (OTC) formulation (Liquamycin LA-200). Milk residues were also determined following a subcutaneous (s.c.) administration of the same product at the same dose. Serum samples were taken for 24 h post-treatment and tissues (fat, liver, kidney, muscle and injection site) collected at 4, 7, 14, 21 and 28 days following injection. Milk from lactating goats was collected every 12 h for 8 days following both the i.m. and s.c. treatments utilizing an intervening 5-week washout period. Residues in serum and tissue were measured using a microbial inhibition assay, while milk residues were measured using both a microbial inhibition assay and a validated HPLC method. The serum pharmacokinetic parameters of OTC in goats were determined, with a mean AUC=67.4 microg h/mL, mean terminal half-life=14.4 h, and apparent clearance=0.33 L/kg h. Tissue half-lives could not be determined with confidence because the collection times provided only two points at which residues could be measured for most tissues. Oxytetracycline residues in all goat tissue samples measured less then cattle tissue tolerance by 96 h postdosing. One-compartment model describing milk depletion data for i.m. and s.c. dosing had terminal slope half-lives of 20.1 and 36.1 h, respectively. By 96 h post-treatment none of the milk samples contained OTC residues in excess of the cattle milk tolerance (0.3 p.p.m.). For both milk and tissue, the upper-bound 99% confidence intervals for the samples taken from goats 96 h postdosing were lower than approved cow milk and tissue tolerances.


Subject(s)
Anti-Bacterial Agents/pharmacokinetics , Goats/metabolism , Milk/metabolism , Oxytetracycline/pharmacokinetics , Animals , Anti-Bacterial Agents/administration & dosage , Anti-Bacterial Agents/blood , Area Under Curve , Chromatography, High Pressure Liquid/veterinary , Delayed-Action Preparations , Drug Residues/pharmacokinetics , Female , Injections, Intramuscular/veterinary , Injections, Subcutaneous/veterinary , Oxytetracycline/administration & dosage , Oxytetracycline/blood , Tissue Distribution
18.
J Vet Pharmacol Ther ; 23(6): 345-52, 2000 Dec.
Article in English | MEDLINE | ID: mdl-11168911

ABSTRACT

The pharmacokinetics of a long-acting oxytetracycline (OTC) formulation (Liquamycin LA-200) injected intramuscularly (i.m.) at a dose of 20 mg/kg were determined in four calves and 24 sheep to determine if the approved label dose for cattle provided a similar serum time/concentration profile in sheep. The AUC for the calves was 168+/-14.6 (microg ? h/mL) and was significantly less than the AUC for sheep (209+/-43 microg ? h/mL). Using the standard two-stage approach and a one-compartment model, the mean Cmax for the calves was 5.2+/-0.8 microg /mL, and for the sheep was 6.1+/-1.3 microg /mL. The mean terminal phase rate constants were 0.031 and 0.033 h, and the Vdss were 3.3 and 3.08 L/kg for the calves and sheep respectively. Analysis of the data using the standard two-stage approach, the naive pooled-data approach and a population model gave very similar results for both the cattle and sheep data. Sheep tissue residues of OTC in serum, liver, kidney, fat, muscle and injection site were measured at 1, 2, 3, 5, 7 and 14 days after a single i.m. injection of 20 mg/kg OTC. Half-lives of OTC residues in the tissues were 38.6, 33.4, 28.6, 25.4, 21.3, and 19.9 h for injection site, kidney, muscle, liver, mesenteric fat and renal fat, respectively. The ratio of tissue to serum concentration was fairly consistent at all slaughter times, except for the fat and injection sites. The mean ratios were 1.72, 4.19, 0.11, 0.061, 0.84 and 827 for the liver, kidney, renal fat, mesenteric fat, muscle and injection sites, respectively. The tissue concentrations of OTC residues were below the established cattle tolerances for OTC in liver (6 p.p.m.), muscle (2 p.p.m.) and kidney (12 p.p.m.) by 48 h, and in injection site muscle by 14 days after the single i.m. injection of 20 mg/kg.


Subject(s)
Oxytetracycline/pharmacokinetics , Animals , Area Under Curve , Cattle , Delayed-Action Preparations , Female , Half-Life , Injections, Intramuscular , Male , Oxytetracycline/blood , Sheep , Tissue Distribution
19.
J Vet Pharmacol Ther ; 21(2): 85-91, 1998 Apr.
Article in English | MEDLINE | ID: mdl-9597644

ABSTRACT

The pharmacokinetics of ceftiofur sodium were determined in domestic chicks, turkey poults, adult cockatiels (Nymphicus hollandicus), and adult orange-winged Amazon parrots (Amazona amazonica) after subcutaneous (chicks and turkey poults and intramuscular (i.m.) dosing (cockatiels and Amazon parrots). Turkey poult data were best fit to a single exponential model with disappearance half-lives (t1/2) of 8.6, 7.4 and 5.6 h after doses of 0.12, 0.24 and 0.48 mg ceftiofur free acid equivalents (CFAE)/poult, respectively. Data from chicks were best fit to a biexponential model with primary and secondary half-lives of 2.2 and 7.5, 3.7 and 6.8, and 3.8 and 5.3 h after doses of 0.04, 0.08 and 0.16 mg CFAE/chick, respectively. Cockatiel and Amazon parrot data were best fit to a biexponential model with primary and secondary half-lives of 0.28 and 2.5, and 0.93 and 7.9 h, respectively, after doses of 10 mg CFAE/kg body weight. The maximum concentration (Cmax) and area under the concentration time curve (AUC) in chicks and poults were dose-proportional. The Cmax for cockatiels was 5.2 micrograms/mL and for Amazon parrots was 11 micrograms/mL. Clearance in cockatiels and Amazon parrots were 11.3 and 3.8 mL/min/kg, respectively, and reflected the much greater AUC seen in Amazon parrots. Clearance values of ceftiofur were similar in chicks and Amazon parrots, slightly greater in turkey poults and greatest in cockatiels. These results indicate that pharmacokinetic differences must be considered when establishing dosage regimens for different avian species.


Subject(s)
Cephalosporins/pharmacokinetics , Chickens/metabolism , Parrots/metabolism , Turkeys/metabolism , Animals , Area Under Curve , Cephalosporins/administration & dosage , Data Interpretation, Statistical , Female , Half-Life , Male , Species Specificity
20.
J Vet Pharmacol Ther ; 20(5): 368-73, 1997 Oct.
Article in English | MEDLINE | ID: mdl-9350257

ABSTRACT

Twelve (12) lactating dairy goats (46-71 kg body wt at study initiation) were divided into four treatment groups and dosed with ceftiofur sodium at 1.1 mg ceftiofur free acid equivalents (CFAE)/kg or 2.2 CFAE/kg using a complete two route (intravenous, i.v.; intramuscular, i.m.), two-period crossover design, with a 2-week washout between injections. After another 2-week washout period, the goats were dosed with ceftiofur sodium i.m. for 5 consecutive days at either 1.1 or 2.2 mg CFAE/kg. The goats from the 2.2 mg/kg multiple dose group were dried off and the i.v. kinetic study repeated. After all injections, blood samples were obtained serially for determination of combined serum concentrations of ceftiofur and metabolites. After intravenous doses of 1.1 and 2.2 mg/kg, the harmonic means of the terminal phase half-lives were 171.8 and 233 min, respectively, for lactating does. The harmonic mean of the terminal phase half-life after an i.v. dose of 2.2 mg/kg in non-lactating does was 254 min. The AUC0-infinity was significantly less and the clearance significantly greater during lactation. After i.m. doses of 1.1 and 2.2 mg/kg, the harmonic mean terminal phase half-lives were 163 and 156 min, respectively. The i.m. bioavailability of ceftiofur sodium in goats was 100%, and the AUC0-infinity was dose-proportional from 1.1-2.2 mg CFAE/kg body weight. After five daily i.m. doses of ceftiofur sodium at either 1.1 or 2.2 mg CFAE, there was minimal accumulation of drug in serum as assessed by Cmax, and serum concentrations were dose-proportional after the multiple dosing regimen.


Subject(s)
Cephalosporins/pharmacokinetics , Goats/metabolism , Animals , Area Under Curve , Biological Availability , Cephalosporins/administration & dosage , Cephalosporins/blood , Cross-Over Studies , Dairying , Dose-Response Relationship, Drug , Female , Half-Life , Injections, Intramuscular/veterinary , Injections, Intravenous/veterinary , Lactation
SELECTION OF CITATIONS
SEARCH DETAIL
...