editorial board
past issues
contributors
feedback
contact us

 

printer friendly page    email page

2010, Vol. 5 No. 2, Article 66

 

Pharmacokinetics and Bioavailability of
Ceftriaxone in Patanwadi Sheep

Swati, S. Tiwari, U. D. Patel, S. K. Bhavsar and A. M. Thaker*

 

Department of Pharmacology and Toxicology,
College of Veterinary Science and A.H., Anand Agricultural University, Anand 388 001

 

*Corresponding Author; e-mail address: [email protected]

 


ABSTRACT

Pharmacokinetics of ceftriaxone following single dose intravenous and intramuscular administration (10 mg/kg) in six Patanwadi sheep was determined. The plasma concentration-time curves were characteristic of a two and one-compartment open model following intravenous and intramuscular administration, respectively. Following intravenous administration the area under plasma drug concentration-time curve (AUC), apparent volume of distribution (Vdarea), elimination half-life (t1/2b) and total body clearance (ClB) were calculated to be 42.65 ± 1.36 µg.h/mL, 0.41 ± 0.02 L/kg, 1.21 ± 0.05 h and 3.91 ± 0.13 mL/min/kg, respectively. Peak plasma drug concentration of 15.53 ± 0.71 µg/mL was obtained at 0.5 h after intramuscular administration. The area under plasma drug concentration-time curve (AUC), apparent volume of distribution (Vdarea), elimination half-life and total body clearance (ClB) were 47.68 ± 1.31 µg.h/mL, 0.43 ± 0.02 L/kg, 2.24 ± 0.07 h and 2.22 ± 0.12 mL/min/kg, respectively. The intramuscular bioavailability of the drug was 64.0 ± 2.0%. Pharmacokinetic profile and moderate bioavailability of ceftriaxone indicated that the drug can be used intramuscularly to treat susceptible bacterial infections in sheep.

KEY WORDS

Pharmacokinetics, ceftriaxone, intravenous, intramuscular, sheep.

INTRODUCTION

Ceftriaxone is a member of third generation cephalosporin having excellent activity against aerobic gram-negative and gram-positive bacteria; however it shows variable activity against anaerobic bacteria. Ceftriaxone distributes in a wide variety of tissues and body fluids such as pleural fluid, peritoneal fluid, bile, bronchial mucosa, myometrium and bone. It crosses not only the inflamed, but also the healthy blood-cerebrospinal fluid barrier in horses and man (Richards et al., 1984 and Ringger et al., 1996). The drug is associated with very low incidence of adverse effects and development of resistance.
The disposition kinetics and dosage schedule of ceftriaxone have been determined in goats (Ismail, 2005; Tiwari et al., 2009), sheep (Guerrini et al., 1985; Goudah et al., 2006), cow calves (Soback and Ziv, 1988; Johal and Srivastava, 1998, 1999; Maradiya, 2004), buffalo calves (Dardi et al., 2004; Gohil et al., 2009), camels (Goudah, 2008), horses (Gardner and Aucoin, 1994; Ringger et al., 1996, 1998) and dogs (Rebuelto et al., 2002). Certain bacterial diseases are major causes of neonatal mortality in sheep where ceftriaxone can be used for the treatment. As ceftriaxone has great potential for clinical use in veterinary medicine, the data on its pharmacokinetics in Patanwadi breed of sheep is not well determined. The present study was therefore conducted to determine the pharmacokinetics of ceftriaxone following single intravenous and intramuscular administration at the dose of 10 mg/kg in Patanwadi breed of sheep.

MATERIALS AND METHODS

Experimental animals and drug administration
The experiment was conducted on six healthy female Patanwadi sheep (20-24 months of age) weighing 20.1-31.5 kg. The animals were kept under constant observation for two weeks before commencement of the experiment at Instructional farm of the college. The animals were examined clinically to evaluate health status and to rule out the possibility of any diseases. Each animal was housed in a separate pen and provided standard ration. Water was provided ad libitum. All animals were randomly grouped to receive either intravenous or intramuscular injection of ceftriaxone sodium (Vetaceph, Unichem Pharmaceuticals Ltd., India) at the dose rate of 10 mg/kg. A washout period of 3 weeks was observed between treatments.
Collection of samples
Blood samples (4 mL each) were collected in heparinized glass test tubes through the intravenous catheter fixed in contra-lateral jugular vein before and after administration of the drug. The samples were collected at 2, 5, 10, 15, 30 min before administration and 1, 2, 4, 6, 8, 12, 24, 36 and 48 h after intravenous administration (through jugular vein) and intramuscular administration ( in deep gluteal muscle). Plasma was separated by centrifugation at 3,000 revolutions per min for 10 min at room temperature and stored at –20°C and assayed with in 24 h.
Ceftriaxone assay and pharmacokinetic analysis
Plasma ceftriaxone concentration was determined by the high performance liquid chromatography (HPLC) method (Hakim et al., 1988) as modified by (Tiwari et al., 2009). Briefly, the HPLC system (Merck-Hitachi LaChrom) consists of isocratic pump (L-7110) with an online degasser (L-7612), interface (D-7000), UV detector (7400), autosampler (7200), sample cooler (L-7200), chromatography data station software (D-7000) and multi HSM-manager. Chromatographic separation was done using Lichrocart RP-18 column (250 mm X 4 mm) at room temperature.
Samples (250 µl) were deproteinized by addition of acetonitrile (500 µl), vortexed for one minute followed by centrifugation for 10 min at 5,000 revolutions per minute. A clear supernatant fluid was decanted in a glass insert (automatic sampler vessels) from which 50 µl was injected into the HPLC system. The mobile phase consisted of a mixture of buffer and acetonitrile (62:38). The buffer was prepared by dissolving 1.78 g of di-sodium hydrogen phosphate dihydrate and 1.0 g of N-acetyl -N, N, N-trimethyl ammonium bromide in 950 mL of Milli Q water, pH (7.0) was adjusted with orthophosphoric acid. Mobile phase was filtered through 0.45µ Millipore filter. Mobile phase was pumped through column at a flow rate of 1.0 mL/min, at an ambient temperature of 25°C. The elute was monitored at a wavelength of 254 nm. All chemicals used in the present study were of HPLC grade.
Ceftriaxone standards (0.19, 0.26, 0.52, 1.68, 4.93, 14.94, 49.79, 76.59, 90.11, 100.12 µg/mL) were prepared by serial dilutions of stock solution of the pure ceftriaxone in drug-free plasma of goats. Calibration curve was prepared for drug concentrations ranging from 0.19 to 100.12 µg/mL and was used to quantify the drug concentration in samples. The calibration curve was prepared daily and it had a R2 value ≥ 0.99. The assay was linear for drug concentrations of 0.19 to 100.12 µg/mL. The lower limit of quantification of assay was 0.19 µg/mL. Different pharmacokinetic parameters were calculated as described by Gibaldi and Perrier (1982) and Notari (1987).

RESULTS

Following intravenous and intramuscular administration, the data was best fitted to two and one compartment open model, respectively. The drug was detected in plasma up to 6 and 12 h following intravenous and intramuscular administration, respectively. Comparative disposition of ceftriaxone following single dose intravenous and intramuscular administration in sheep is shown on semilogarithmic scale in figure 1. Following intravenous administration, concentration of ceftriaxone 0.48 µg/mL of plasma was maintained up to 6 h. Following single dose intramuscular administration, the therapeutically effective plasma concentration (> 0.2 µg/mL) was detected at 0.083 h (5 minutes) and maintained for 12 h. Peak plasma drug concentration of 15.53 ± 0.71 µg/mL was obtained at 0.5 h after intramuscular administration. The bioavailability of ceftriaxone was 64.0 ± 2.0 per cent following intramuscular administration. Pharmacokinetic parameters determined following intravenous and intramuscular administration of the drug are depicted in table 1.

DISCUSSION

Following intravenous administration, the drug was distributed fast as evidenced by short distribution half-life. Similar pattern of distribution of ceftriaxone after intravenous administration has been observed in goats (Tiwari et al., 2009) and buffalo calves (Dardi et al., 2004; Gohil et al., 2009). The drug was moderately distributed following intravenous administration in sheep as evidenced by Vdarea. Apparent volume of distribution of 0.30 L/kg (Guerrini et al., 1985) in sheep and 0.58 ± 0.04 L/kg in goats (Tiwari et al., 2009) has earlier been reported which supports the present observations. Total body clearance of the drug observed in the present study was similar to clearance of the drug reported in calves, sheep, goats and horses (Guerrini et al, 1985; Soback and Ziv 1988; Gardener and Aucoin, 1994; Ismail, 2005; Goudah et al, 2006; Gohil et al., 2009; Tiwari et al., 2009). Elimination half-life and total body clearance suggests faster elimination of cetriaxone in sheep. However, longer elimination half-life of 2.57 ± 0.52 h has been reported in camel (Goudah, 2008) which may be due to slower total body clearance (1.83 ± 0.16 mL/min/kg) of the drug in camel.
Peak plasma concentration (Cmax) of 15.53 ± 0.71 µg/mL observed at 0.5 h following intramuscular injection, was lower than that observed in goats (23.6 ± 1.2 and 21.51 ± 0.61 µg/mL); and sheep (23.16 ± 2.94 µg/mL) (Ismail, 2005; Goudah et al, 2006; Tiwari et al., 2009). Short absorption half-life following intramuscular administration indicates rapid drug absorption from the site of injection. Rapid absorption following intramuscular injection has also been reported for ceftriaxone in goats (Tiwari et al., 2009) and cow calves (Maradiya, 2004). The longer elimination half-life and slow clearance of the drug observed during present investigation indicates that drug may remain in the body of sheep for long time as compared to cow calves (1.94 ± 0.12 h) and goats (1.44 h; 2.03 ± 0.09 h) (Soback and Ziv 1988; Ismail, 2005; Tiwari et al., 2009). The intramuscular bioavailability of ceftriaxone determined in the present study was in agreement with bioavailability (59.0 ± 4.0 %) of the drug reported in goats (Tiwari et al., 2009). However, higher bioavailability of ceftriaxone following intramuscular administration has been reported in cow calves (78%), buffalo calves (86.7%), goats (85%), sheep (83.6 ± 20.53%) and camel (93.42 ± 21.4%) (Soback and Ziv, 1988; Dardi et al, 2004; Ismail, 2005; Goudah et al, 2006; Goudah, 2008).
The optimal outcome of therapy with bactericidal drug requires attainment of high concentrations and the success of therapy correlates with the AUC/MIC ratio, while prevention of the development of resistance correlates with the Cmax/MIC ratio (Shojaee Aliabadi and Lees, 2000). The effective use of the antibacterial drugs against clinically important pathogens depends on designing dosages that attain a Cmax/MIC ratio > 8-10 and an AUC/MIC ratio > 100-125 (Lode et al, 1998). Thus values of AUC/MIC and Cmax/MIC after intramuscular administration were calculated using MIC90 (0.2 µg/mL) of ceftriaxone against Salmonella spp., Escherichia coli and Pasteurella multocida isolates (Soback and Ziv, 1988). The calculated value of Cmax/MIC (77.65) and AUC/MIC (238.4) indicates that ceftriaxone may have outstanding clinical and bacteriological efficacy against gram-negative infections in sheep. The pharmacokinetic characteristics of ceftriaxone indicates that the drug may be used intramuscularly to treat susceptible bacterial infections in sheep.

REFERENCES

  1. Dardi MS, Sharma SK, Srivastava AK. Pharmacokinetics and dosage regimen of ceftriaxone in buffalo calves. Vet Res Commun 2004, 28: 331-338.

  2. Gardner SY, Aucoin DP. Pharmacokinetics of ceftriaxone in mares. J Vet Pharmacol Ther 1994; 17, 155-156.

  3. Gibaldi M, Perrier D. Pharmacokinetics. (2nd Edn.), Marcel-Dekker Inc., New York, 1982, pp. 45-109.

  4. Gohil, PV, Patel UD, Bhavsar SK, Thaker AM. Pharmacokinetics of ceftriaxone in buffalo calves (Bubalus bubalis) following intravenous and intramuscular administration. Iranian J Vet Res 2009; 10: 33-37.

  5. Goudah A, Shin HC, Shim AJ, Abd El-Aty AM. Charecterization of the relationship between serum and milk residue disposition of ceftriaxone in lactating ewes. J Vet Pharmacol Ther 2006; 29, 307-312.

  6. Goudah A. Pharmacokinetic parameters of ceftriaxone after single intravenous and intramuscular administration in camels (Camelus Dromedarius). Res Vet Sci 2008; 84, 483–489

  7. Guerrini VH, Filippich LJ, Cao GR, English PB, Bourne DWA. Pharmacokinetics of cefaronide, ceftriaxone and cefoperazone in sheep. J Vet Pharmacol Ther 1985; 8, 120-127.

  8. Hakim L, Bourne DW, Triggs EJ. High performance liquid chromatographic assay of cefotaxime, desacetylcefotaxime and ceftriaxone in rat plasma. J Chromatogr 1988; 424:111-117.

  9. Ismail MM. Pharmacokinetics, urinary and mammary excretion of ceftriaxone in lactating goats. J Vet Med A : Physiol Pathol Clin Med 2005; 52, 354-358.

  10. Johal B, Srivastava AK. Disposition kinetics and dosage regimen of ceftriaxone in crossbred calves. Acta Vet Hung 1999; 47, 243-248.

  11. Johal B, Srivastava AK. Pharmacokinetics, urinary excretion and dosage regimen of ceftriaxone in crossbred cow calves following single intramuscular administration. Indian J Ani Sci 1998; 68: 1017-1019.

  12. Lode, H., Borner, K., Koeppe, P.: Pharmacodynamics of fluoroquinolones. Clin Infect Dis 1998; 27: 33-39.

  13. Maradiya JJ. Studies on ceftriaxone pharmacokinetics following intravenous and intramuscular administration in crossbred calves. MVSc. Thesis, Anand Agricultural University, Anand, Gujarat, India. 2004; PP: 67-82.

  14. Notari RE. Biopharmaceutics and clinical pharmacokinetics- An introduction. 4th Edn. Marcell dekker Inc. New York, 1987.

  15. Rebuelto M, Albarellos G, Ambros L, Kreil V, Montoya L, Bonafine R, Otero P, Hallu R. Pharmacokinetics of ceftriaxone administered by the intravenous, intramuscular or subcutaneous routes to dogs. J Vet Pharmacol Ther 2002; 25, 73-76.

  16. Richards DM, Heel RC, Brogden RN, Speight TM, Avery GS. Ceftriaxone. A review of its antibacterial activity, pharmacological properties and therapeutic use. Drugs 1984; 27, 469-527.

  17. Ringger NC, Brown MP, Kohlepp SJ, Gronwall RR, Merritt K. Pharmacokinetics of Ceftriaxone in Neonatal Foals. Equine Vet J 1998; 30: 163-165.

  18. Ringger, N.C., Pearson, E.G., Gronwall, R. and Kohlepp, S.J. (1996). Pharmacokinetics of Ceftriaxone in Healthy Horses. Equine Vet J 28: 476-479.

  19. Shojaee Aliabadi F, Lees P. Antibiotic treatment for animals: effect on bacterial population and dosage regimen optimisation. Intern J Antimicrob Agents 2000; 14: 307–313.

  20. Soback S, Ziv G. Pharmacokinetics and bioavailability of ceftriaxone administered intravenously and intramuscularly to calves. Am J Vet Res 1988; 49: 535-538.

  21. Tiwari S, Swati, Bhavsar SK, Patel UD, Thaker AM. (2009). Disposition of ceftriaxone in goats (Capra hircus). Vetscan. Vol4 No2


TABLES

Table 1: Pharmacokinetic parameters of ceftriaxone after single dose intravenous and intramuscular administration (10 mg/kg) in sheep (n = 6)

ceftriaxone sheep
 t1/2α
:  half-life of distribution phases; t1/2β: elimination half life;  t1/2K(a): absorption half-life; AUC(0-∞): total area under plasma drug concentration-time curve; AUMC: area under first moment of curve; Vd(area): apparent volume of distribution;  Vd(ss): volume of distribution at steady state;   Cl(B): total plasma clearance; MRT: mean residence time;  F: bioavailability; Cmax: maximum drug concentration; Tmax: time of maximum observed concentration in plasma

 

FIGURES



Figure 1.
Pharmacokinetics and Bioavailability of Ceftriaxone

 

 

 


Copyright © Vet Scan 2005-

All Right Reserved with VetScan
www.vetscan.co.in and www.kashvet.org
ISSN 0973-6980

 

Home | e-Learning |Resources | Alumni | Forum | Picture blog | Disclaimer

                   

 

 

powered by eMedia Services