Pharmacokinetics and Tissue Residues of Sulfathiazole and Sulfamethazine in Pigs

1994 ◽  
Vol 57 (9) ◽  
pp. 796-801 ◽  
Author(s):  
LIEVE S. G. VAN POUCKE ◽  
CARLOS H. VAN PETEGHEM

The plasma pharmacokinetics and tissue penetration of sulfathiazole (ST) and sulfamethazine (SM) after intravenous and intramuscular injection in pigs were studied. Following a single intravenous dose of 40 mg ST/kg of bodyweight or 80 mg SM/kg of bodyweight, the plasma ST and SM concentrations were best fitted to a two-compartment model. The areas under the curve were 447 ± 39 and 1485 ± 41 mg/h/L, clearances were 0.090 ± 0.007 and 0.054 ± 0.001 L/kg/h, volumes of distribution were 1.16 ± 0.16 and 0.77 ± 0.06 L/kg, half-lifes in distribution phase were l.18 ± 0.57 and 0.23 ± 0.16 h and half-lifes in eliminations phase were 9.0 ± l.6 and 9.8 ± 0.6 h. When the two compounds were administered simultaneously as a single intravenous injection, the pharmacokinetic parameters for ST were not significantly different. The values for SM show statistical differences for some important parameters: α, β and the AUC0–>∞ were significantly decreased and t1/2α, Vd and CIB were significantly increased. It can be concluded that after a single intravenous injection of 40 mg/kg, sulfathiazole has a high tl/2β resulting in higher tissue concentrations. This half-life, which is higher than what is reported in the literature, is not influenced by the simultaneous presence of sulfamethazine. The tl/2β for sulfamethazine after a single intravenous injection of 80 mg/kg is comparable to the data from the literature and is not influenced by the presence of sulfathiazole. Sulfathiazole and SM were also administered simultaneously as an intramuscular injection to healthy pigs at a dosage of 40 and 80 mg/kg bodyweight. Pharmacokinetic experiments were conducted on three pigs. From this pharmacokinetic study it can be concluded that upon a single intramuscular administration of 40 mg/kg of ST and 80 mg/kg of SM the absolute bioavailability in pigs is 0.92 ± 0.04 for ST and l.01 ± 0.07 for SM. Six pigs received five intramuscular im) injections as a single dose of ST and SM every 24 h for five consecutive days for the residue study. The pigs were slaughtered at different times after the last dose was given and samples were taken from various tissues and organs. Concentrations were determined by a microbiological method and a HPTLC method. No edible tissue contained more than 100 μg/kg of the individual sulfonamides after 10 days of withdrawal. It means that adult animals which have a shorter half-life and thus lower tissue concentrations will certainly meet the economic community EC) maximum residue limits after a 10 days withdrawal period.

2017 ◽  
Vol 20 (2) ◽  
pp. 261-268
Author(s):  
A. Burmańczuk ◽  
T. Grabowski ◽  
T. Błądek ◽  
C. Kowalski ◽  
P. Dębiak

Abstract The aim of the study was to carry out retrospective and prospective comparative analyses of the pharmacokinetics of CEF after single intramammary (IMM) administration in cows. The prospective study (study A) was conducted on 9 dairy cows of the Polish Black-White race with clinical mastitis during the lactation period. Milk samples were collected at 2, 4, 6, 8, 10, 24, 36, 48, 72 and 84 h after single IMM administration of 250 mg of CEF to one quarter. Drug concentrations in milk samples were determined by HPLC-MS/MS technique and the results of the pharmacokinetic analysis were compared to those obtained in previous studies based on the microbiological (study B) and HPLC-UV methods (study C and D). Pharmacokinetic parameters were calculated based on adapted two-compartment model of drug distribution. One of the findings of the comparison of the analysed investigations is that the CEF kinetics determined with the microbiological method is consistent with the results obtained by the authors of this paper. Both studies yielded similar results of the key pharmacokinetic parameters related to the level of the drug distribution to tissues and elimination half-life. In the pharmacodynamic analysis, the observations in all four studies were entirely consistent and have shown lower values of T>MIC90 in healthy animals and significantly higher values in infected dairy cows. The comparison of studies A, B, C, and D revealed that the time of complete CEF wash-out of 90.90% varied and amounted to 5.7, 8.0, 2.2, and 2.2 days after administration of the drug, respectively. It was confirmed that not only the type of the analytical method but also correct sampling have a significant impact on determination of the correct value of the drug half-life after IMM administration. The comparative analysis of studies in which the milk yield was high and low allows a conclusion that this parameter in the case of CEF has no significant effect on T>MIC90.


2019 ◽  
Vol 104 (6) ◽  
pp. e4.2-e4
Author(s):  
G Salis ◽  
N Medlicott ◽  
D Reith

BackgroundGentamicin is commonly used in the NICU setting and is often administered via long lines, which increases variability in the rate of administration. We aimed to model drug delivery pharmacokinetic parameters for intravenous gentamicin administered via umbilical venous catheters (UVCs).MethodsData was modelled from infusion simulations of gentamicin delivery using UVCs with a background flow rate of 0.5 ml/h.1 Different combinations of dose (2 mg, 5 mg) were given by bolus injection over 3–5 minutes, followed by a normal saline flush (1 ml, 2 ml). Gentamicin levels were measured at 5 minute intervals over an hour via high pressure liquid chromatography.Phoenix Certara (version 8.1) was used for modelling. An extravascular model with clearance removed was used to predict parameters: absorption constant (Ka), time lag (Tlag), and bioavailability (F). F was used to enable an estimate of the variability in dose administered. Different error models were tested to ascertain which best described the data.ResultsAn extravascular one compartment model with first order absorption and additive error best described the data. Estimates for the model with a 2 mg dose and 1 ml flush were Ka 0.34L/min, Tlag 1.28min, F 0.97, standard deviation (stdev) 0.14. For 2 mg, 2 ml flush, estimates were Ka 0.86L/min, Tlag 3.01min, F 0.87, stdev 0.01. For 5 mg, 1 ml flush, estimates were Ka 0.48L/min, Tlag 3.13min, F 1.03, stdev 0.12. For 5 mg, 2 ml flush, estimates were Ka 0.83L/min, Tlag 3.29min, F 1.09, stdev 0.02. For each model epsshrinkage and nshrinkage for Tlag and F were low, however nshrinkage for ka was 0.9999.ConclusionThis is the first known modelling of gentamicin delivery kinetics. The studies all had high nshrinkage for Ka, therefore the individual estimates of ka may be unreliable. Further studies with a higher number of replicates would provide more favourable data for estimating Ka.ReferenceLala AC ( 2016). Variability in neonatal gentamicin administration influencing drug delivery kinetics (Thesis, Master of Medical Science). University of Otago.Disclosure(s)No conflict of interest declared. Funding for research via the Freemasons Society of New Zealand.


2016 ◽  
Vol 19 (10) ◽  
pp. 992-997 ◽  
Author(s):  
Gabriela A Albarellos ◽  
Laura Montoya ◽  
Sabrina M Passini ◽  
Martín P Lupi ◽  
Paula M Lorenzini ◽  
...  

Objectives The aim of this study was to determine the plasma pharmacokinetic profile, tissue concentrations and urine elimination of cefazolin in cats under surgical conditions after a single intravenous dose of 20 mg/kg. Methods Intravenous cefazolin (20 mg/kg) was administered to nine young mixed-breed cats 30 mins before they underwent surgical procedures (ovariectomy or orchiectomy). After antibiotic administration, samples from blood, some tissues and urine were taken. Cefazolin concentrations were determined in all biological matrices and pharmacokinetic parameters were estimated. Results Initial plasma concentrations were high (Cp(0), 134.80 ± 40.54 µg/ml), with fast and moderately wide distribution (distribution half-life [t½(d)] 0.16 ± 0.15 h; volume of distribution at steady state [V(d[ss])] 0.29 ± 0.10 l/kg) and rapid elimination (body clearance [ClB], 0.21 ± 0.06 l/h/kg; elimination half-life [t½], 1.18 ± 0.27 h; mean residence time 1.42 ± 0.36 h). Thirty to 60 mins after intravenous administration, cefazolin tissue concentrations ranged from 9.24 µg/ml (subcutaneous tissue) to 26.44 µg/ml (ovary). The tissue/plasma concentration ratio ranged from 0.18 (muscle) to 0.58 (ovary). Cefazolin urine concentrations were high with 84.2% of the administered dose being eliminated in the first 6 h postadministration. Conclusions and relevance Cefazolin plasma concentrations remained above a minimum inhibitory concentration of ⩽2 µg/ml up to 4 h in all the studied cats. This suggests that a single intravenous dose of 20 mg/kg cefazolin would be adequate for perioperative prophylactic use in cats.


1963 ◽  
Vol 44 (2) ◽  
pp. 250-258 ◽  
Author(s):  
Pavo Hedner ◽  
Yngve Einerth

ABSTRACT A single intravenous injection of 10 U. S. P. units of a purified ACTH preparation in man produced a maximal effect for at least 4 hours, and the effect was still significant 8 hours after the injection. The biological half life of this preparation was calculated and found to be about 1 hour. When given as an infusion over 8 hours this preparation was calculated to give a maximally active plasma ACTH level for 9½ hours. A significantly increased plasma corticosteroid level was present 12 hours after the beginning of the infusion, and a comparable response was obtained by the same amount of ACTH twice injected intravenously with 4 hours interval. A maximal adrenocortical activity was obtained with 50 U. S. P. units of the same ACTH injected intramuscularly and this persisted for at least 8 hours. The result of this investigation is not consistent with the view often held that the biological half life of exogenous ACTH is short and this discrepancy may be explained by the purified ACTH preparation used here. The relation between different methods of ACTH administration and their significance in clinical therapy is discussed.


Author(s):  
Fan Yang ◽  
Han Wang ◽  
Zhe-wen Song ◽  
Meng-li Yu ◽  
Mei Zhang ◽  
...  

Background: Ceftiofur is a third-generation cephalosporin antibiotic developed exclusively for veterinary applications. Although not approved in China, ceftiofur is being used extensively in an extra-label manner to treat poultry infections. Black-bone silky fowl is a unique chicken breed which is different from the common chicken breeds in both morphology and other physiology. These differences may result in varied pharmacokinetic profiles. Therefore, the present study aimed to investigate the pharmacokinetics of ceftiofur (measured by ceftiofur and its active metabolites concentrations) in black-bone silky fowl following a single intravenous or intramuscular injection of ceftiofur sodium.Methods: Ceftiofur sodium was intravenously and intramuscularly given to six healthy black-bone silky fowl at the dose of 10 mg/kg body weight. A derivatization method was used to quantify the concentrations of ceftiofur and its active metabolites (expressed as desfuroylceftiofur acetamide) in plasma samples. A non-compartmental method was used to calculate the pharmacokinetics parameters. Results: The terminal half-life (t1/2lz) was calculated as 3.19±0.28 and 3.36±0.17 h following intravenous and intramuscular injections, respectively. After intravenous treatment, the total body clearance (Cl) and volume of distribution at steady state (VSS) were determined as 73.79±5.83 ml/h/kg and 318.65±30.06 ml/kg, respectively. After intramuscular injection, the peak concentration (Cmax; 22.55±0.89 ìg/ml) was observed at 1.67±0.26 h and the absorption half-life (t1/2ka) and absolute bioavailability (F) were calculated as 0.40±0.13 h and 93.03%±7.07%, respectively. The current results demonstrated the rapid and complete absorption, however, poor distribution and rapid elimination of ceftiofur and its active metabolites in black-bone silky fowl.


1996 ◽  
Vol 14 (5) ◽  
pp. 1581-1588 ◽  
Author(s):  
M Sandström ◽  
A Freijs ◽  
R Larsson ◽  
P Nygren ◽  
M L Fjällskog ◽  
...  

PURPOSE The aim of this study was to investigate the covariance between the pharmacokinetics of the three components of the FEC regimen, epirubicin (EPI), fluorouracil (5-FU), and the cyclophosphamide (CP) metabolite 4-hydroxycyclophosphamide (4-OHCP), in breast cancer patients. PATIENTS AND METHODS Data from 21 women were collected over a total of 35 cycles. 5-FU (300 to 600 mg/m2) and CP (300 to 600 mg/m2) were administered as bolus injections, whereas EPI (15 to 60 mg/m2) was administered either as a bolus injection or as an infusion. The pharmacokinetics of the component drugs were monitored using a limited sampling scheme. Population pharmacokinetic models for each of the three drugs were developed using the program NONMEM. RESULTS The data for 5-FU were best described by a one-compartment model with nonlinear elimination, where the maximal rate of elimination (Vmax) and the concentration at which the elimination was half-maximal (Km) were 105 mg/L.h and 27 mg/L, respectively. EPI concentration-time profiles showed a triexponential decline, with a mean terminal half-life of 24 hours and a clearance (CL) of 59 L/h. The elimination of 4-OHCP was monoexponential, with a mean half-life of 7 hours. The interindividual coefficients of variation (CVs) in CL were 30%, 22%, and 41% for 5-FU, EPI, and 4-OHCP, respectively. The corresponding values for intrapatient course-to-course variability in CL were 11%, 8%, and 27%. No significant correlation in any of the pharmacokinetic parameters between the drugs was found. CONCLUSION Individualization of dosing of the FEC regimen using therapeutic drug monitoring and attempts to find concentration-response relationships may be successful, but requires that the exposure of all three drugs is considered simultaneously.


Blood ◽  
2004 ◽  
Vol 104 (11) ◽  
pp. 1031-1031
Author(s):  
Wolfhart Kreuz ◽  
Inmarculada Martinez-Saguer ◽  
Hildegard Stoll ◽  
Sigurd Knaub ◽  
Thomas Klingebiel

Abstract Hereditary and acquired deficiencies in C1 inhibitor (C1-INH) function can result in potentially life-threatening attacks of hereditary angioedema (HAE). A highly purified and pasteurized C1-INH concentrate has been used effectively as prophylaxis against and treatment for angioedema attacks in patients with hereditary C1-INH deficiencies, but relatively little is known about its pharmacokinetic properties. Objective: To evaluate the pharmacokinetics and in vivo recovery (IVR) of C1-INH concentrate (Berinert®P) in two groups of patients with hereditary angioedema (HAE) receiving this preparation either as individual replacement therapy (IRT, regular, immediate treatment of first HAE symptoms in patients with frequent and severe attacks) or as on-demand treatment. Methods: Forty subjects (15 under IRT, 25 under on-demand treatment) with HAE received intravenous injections of C1-INH (542–1,617 U) in an attack-free interval in a prospective, open, uncontrolled, single-center study. Blood was sampled for determination of C1-INH with a commercially available functional chromogenic assay for up to 72 hours after dosing. Pharmacokinetic parameters were calculated using a single-compartment model and IVR was determined using standard methods. Results: The mean (± SD) time to maximum plasma concentration (Tmax) for C1-INH administered in patients under IRT was 1.3 ± 2.1 hours, the area under the time versus plasma concentration curve (AUC) was 20.5 ± 19.1 hour•U/mL, the elimination half-life (t½) was 33.3 ± 19.8 hours, mean residence time (MRT) was 48.0 ± 28.5 hours, total body clearance (Cl) was 1.1 ± 0.6 mL/kg/hour, and volume of distribution at steady state (Vss) was 39.5 ± 9.9 mL/kg. The respective values for patients treated on demand were 2.9 ± 6.5 hours, 20.0 ± 14.5 hour•U/mL, 43.9 ± 22.4 hours, 63.4 ± 32.3 hours, 1.2 ± 1.0 mL/kg•hour, and 51.4 ± 10.9 mL/kg. The mean IVRs for IRT and on-demand treatment were 108.2 ± 48.3% and 85.8 ± 28.3%, respectively. Children tended to have slightly lower half-life and a slightly higher Vss compared to adults. Conclusions: C1-INH concentrate has a short Tmax and a long, t½ and MRT consistent with the rapid onset of clinical efficacy for this preparation in subjects suffering angioedema attacks and the ability to effectively carry out IRT with injections administered every 2–5 days. This analysis provides to our knowledge the most comprehensive pharmacokinetic evaluation in subjects with HAE.


Blood ◽  
1986 ◽  
Vol 67 (3) ◽  
pp. 646-649 ◽  
Author(s):  
SE Steinberg ◽  
JF Garcia ◽  
GR Matzke ◽  
J Mladenovic

Abstract Detailed studies to analyze the early events of erythropoietin (Ep) secretion and clearance were performed in a rat model using a double antibody radioimmunoassay. Ep clearance was determined following intravenous injection of 1 mL of Ep-rich plasma, 1,080 mU/mL, obtained from phlebotomized rats. Analysis revealed a disappearance curve that conformed to a two-compartment model with an alpha half-life t1/2 of 3.6 minutes and a beta t1/2 of 86 minutes. The volume of distribution was similar to the calculated plasma volume. In anephric animals, there was no change in the plasma clearance rate or the volume of distribution. Rapid Ep secretion was elicited by a single 15 mL/kg phlebotomy (hematocrit decrement 45% to 30%), so that levels reached 20 to 30 times baseline (524 +/- 76 v 24 +/- 7 mU/mL) at five hours, whereas they plateaued for at least 33 hours. The increase in the rate of secretion was geometric, from 9.9 mU/h baseline secretion to 429 mU/h. These data identify a very sensitive and rapidly responsive system for Ep modulation in the rat.


2021 ◽  
Vol 19 (suplemento) ◽  
Author(s):  
A Anadón

The aim of this study was to evaluate the pharmacokinetic behaviour and the absolute bioavailability of marbofloxacin (MFX) in adult water buffaloes and to estimate the pharmacokinetic parameters for calculating the therapeutic dose in this animal species. Six adult buffaloes (3 males and 3 females) where treated by intravenous (IV) and subcutaneous (SC) route with a 10% experimental MFX injectable formulation at the dose of 2 mg/kg. After administration blood samples were drawn at pre-established times and MFX plasma concentrations where determined by microbiologic method. The pharmacokinetic analysis was made by compartmental analysis. After IV administration MFX presented a clearance of 198.4 ± 21.0 mL.kg.h and a half-life of elimination of 7.64 ± 3.29 h. After SC administration marbofloxacin presented a half-life of elimination of 8.5 ± 2.42 h, reaching it maximum plasma concentration (1.67 ± 0.516 μg/mL) at 1.69 ± 0.231 h, with a bioavailability of 80.8 ± 11.2 %. The estimated values of clearance and bioavailability will be employed in further studies for calculating the therapeutic dose of MFX in water buffaloes.    


Sign in / Sign up

Export Citation Format

Share Document