DOI QR코드

DOI QR Code

Formulation of a rational dosage regimen of ceftiofur hydrochloride oily suspension by pharmacokinetic-pharmacodynamic (PK-PD) model for treatment of swine Streptococcus suis infection

  • Luo, Wanhe (National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues) ;
  • Wang, Dehai (College of Animal Science and Technology-College of Veterinary Medicine, Huazhong Agricultural University) ;
  • Qin, Hua (Beijing TEAM Junwei Healthcare Technology Development Co., Ltd.) ;
  • Chen, Dongmei (National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues) ;
  • Pan, Yuanhu (National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues) ;
  • Qu, Wei (National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues) ;
  • Huang, Lingli (National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues) ;
  • Xie, Shuyu (National Reference Laboratory of Veterinary Drug Residues (HZAU) and MARA Key Laboratory for Detection of Veterinary Drug Residues)
  • 투고 : 2020.10.13
  • 심사 : 2021.04.08
  • 발행 : 2021.11.30

초록

Background: Our previously prepared ceftiofur (CEF) hydrochloride oily suspension shows potential wide applications for controlling swine Streptococcus suis infections, while the irrational dose has not been formulated. Objectives: The rational dose regimens of CEF oily suspension against S. suis were systematically studied using a pharmacokinetic-pharmacodynamic model method. Methods: The healthy and infected pigs were intramuscularly administered CEF hydrochloride oily suspension at a single dose of 5 mg/kg, and then the plasma and pulmonary epithelial lining fluid (PELF) were collected at different times. The minimum inhibitory concentration (MIC), minimal bactericidal concentration, mutant prevention concentration (MPC), post-antibiotic effect (PAE), and time-killing curves were determined. Subsequently, the area under the curve by the MIC (AUC0-24h/MIC) values of desfuroylceftiofur (DFC) in the PELF was obtained by integrating in vivo pharmacokinetic data of the infected pigs and ex vivo pharmacodynamic data using the sigmoid Emax (Hill) equation. The dose was calculated based on the AUC0-24h/MIC values for bacteriostatic action, bactericidal action, and bacterial elimination. Results: The peak concentration, the area under the concentration-time curve, and the time to peak for PELF's DFC were 24.76 ± 0.92 ㎍/mL, 811.99 ± 54.70 ㎍·h/mL, and 8.00 h in healthy pigs, and 33.04 ± 0.99 ㎍/mL, 735.85 ± 26.20 ㎍·h/mL, and 8.00 h in infected pigs, respectively. The MIC of PELF's DFC against S. suis strain was 0.25 ㎍/mL. There was strong concentration-dependent activity as determined by MPC, PAE, and the time-killing curves. The AUC0-24h/MIC values of PELF's DFC for bacteriostatic activity, bactericidal activity, and virtual eradication of bacteria were 6.54 h, 9.69 h, and 11.49 h, respectively. Thus, a dosage regimen of 1.94 mg/kg every 72 h could be sufficient to reach bactericidal activity. Conclusions: A rational dosage regimen was recommended, and it could assist in increasing the treatment effectiveness of CEF hydrochloride oily suspension against S. Suis infections.

키워드

과제정보

The article is financially supported by the National Key Research and Development Program (2016YFD0501309).

참고문헌

  1. Wang Y, Wang Y, Sun L, Grenier D, Yi L. Streptococcus suis biofilm: regulation, drug-resistance mechanisms, and disinfection strategies. Appl Microbiol Biotechnol. 2018;102(21):9121-9129. https://doi.org/10.1007/s00253-018-9356-z
  2. Agass MJ, Willoughby CP, Bron AJ, Mitchell CJ, Mayon-White RT. Meningitis and endophthalmitis caused by Streptococcus suis type II (group R). BMJ. 1977;2(6080):167-168. https://doi.org/10.1136/bmj.2.6080.167
  3. Higgins R, Gottschalk M. Distribution of Streptococcus suis capsular types in 1994. Can Vet J. 1995;36(5):320.
  4. Perch B, Pedersen KB, Henrichsen J. Serology of capsulated streptococci pathogenic for pigs: six new serotypes of Streptococcus suis. J Clin Microbiol. 1983;17(6):993-996. https://doi.org/10.1128/jcm.17.6.993-996.1983
  5. Tian Y, Aarestrup FM, Lu CP. Characterization of Streptococcus suis serotype 7 isolates from diseased pigs in Denmark. Vet Microbiol. 2004;103(1-2):55-62. https://doi.org/10.1016/j.vetmic.2004.07.009
  6. Galina L, Collins JE, Pijoan C. Porcine Streptococcus suis in Minnesota. J Vet Diagn Invest. 1992;4(2):195-196. https://doi.org/10.1177/104063879200400216
  7. Heath PJ, Hunt B, Harwood DJ, Welchman DD. Isolation and identification of Streptococcus suis serotypes 3, 4 and 7. Vet Rec. 1995;136(21):547. https://doi.org/10.1136/vr.136.21.547
  8. Zhang CP, Ning YB, Zhang ZQ, Song L, Qiu HS, Gao HY, et al. Distributions of pathogenic capsular types and in vitro antimicrobial susceptibility of different serotypes of Streptococcus suis isolated from clinically healthy sows from 10 provinces in China. Zhonghua Liu Xing Bing Xue Za Zhi. 2009;30(3):235-238.
  9. Goyette-Desjardins G, Auger JP, Xu J, Segura M, Gottschalk M. Streptococcus suis, an important pig pathogen and emerging zoonotic agent-an update on the worldwide distribution based on serotyping and sequence typing. Emerg Microbes Infect. 2014;3(6):e45.
  10. Salmon SA, Watts JL, Yancey RJ Jr. In vitro activity of ceftiofur and its primary metabolite, desfuroylceftiofur, against organisms of veterinary importance. J Vet Diagn Invest. 1996;8(3):332-336. https://doi.org/10.1177/104063879600800309
  11. Risco CA, Hernandez J. Comparison of ceftiofur hydrochloride and estradiol cypionate for metritis prevention and reproductive performance in dairy cows affected with retained fetal membranes. Theriogenology. 2003;60(1):47-58. https://doi.org/10.1016/S0093-691X(02)01299-2
  12. Xie S, Zhang X, Luo W, et al. Formulation, characterization and pharmacokinetics of long-acting ceftiofur hydrochloride suspension. Curr Drug Deliv. 2021;18(2):224-233. https://doi.org/10.2174/1567201817666200903165119
  13. Luo W, Chen D, Wu M, Li Z, Tao Y, Liu Q, et al. Pharmacokinetics/pharmacodynamics models of veterinary antimicrobial agents. J Vet Sci. 2019;20(5):e40. https://doi.org/10.4142/jvs.2019.20.e40
  14. Mitchell JD, McKellar QA, McKeever DJ. Evaluation of antimicrobial activity against Mycoplasma mycoides subsp. mycoides small colony using an in vitro dynamic dilution pharmacokinetic/pharmacodynamic model. J Med Microbiol. 2013;62(Pt 1):56-61. https://doi.org/10.1099/jmm.0.045971-0
  15. Marchetti S, Schellens JH. The impact of FDA and EMEA guidelines on drug development in relation to phase 0 trials. Br J Cancer. 2007;97(5):577-581. https://doi.org/10.1038/sj.bjc.6603925
  16. Nan J, Hao H, Xie S, Pan Y, Xi C, Mao F, et al. Pharmacokinetic and pharmacodynamic integration and modeling of acetylkitasamycin in swine for Clostridium perfringens. J Vet Pharmacol Ther. 2017;40(6):641-655. https://doi.org/10.1111/jvp.12404
  17. Mir O, Broutin S, Desnoyer A, Delahousse J, Chaput N, Paci A. Pharmacokinetics/pharmacodynamic (PK/PD) relationship of therapeutic monoclonal antibodies used in oncology: what's new? Eur J Cancer. 2020;128:103-106. https://doi.org/10.1016/j.ejca.2020.01.004
  18. Hohnstein FS, Meurer M, de Buhr N, von Kockritz-Blickwede M, Baums CG, Alber G, et al. Analysis of porcine pro- and anti-inflammatory cytokine induction by S. suis in vivo and in vitro. Pathogens. 2020;9(1):40. https://doi.org/10.3390/pathogens9010040
  19. Lee JY, Park HJ, Kim YK, Yu S, Chong YP, Kim SH, et al. Cellular profiles of bronchoalveolar lavage fluid and their prognostic significance for non-HIV-infected patients with Pneumocystis jirovecii pneumonia. J Clin Microbiol. 2015;53(4):1310-1316. https://doi.org/10.1128/JCM.03494-14
  20. Zhang M, Yang F, Yu HJ, Kang TJ, Ding YH, Yu ML, et al. Pharmacokinetics of ceftiofur sodium in cats following a single intravenous and subcutaneous injection. J Vet Pharmacol Ther. 2019;42(6):602-608. https://doi.org/10.1111/jvp.12814
  21. Wang J, Hao H, Huang L, Liu Z, Chen D, Yuan Z. Pharmacokinetic and pharmacodynamic integration and modeling of enrofloxacin in swine for Escherichia coli. Front Microbiol. 2016;7:36.
  22. Gastine S, Rashed AN, Hsia Y, Jackson C, Barker CI, Mathur S, et al. GAPPS (grading and assessment of pharmacokinetic-pharmacodynamic studies) a critical appraisal system for antimicrobial PKPD studies - development and application in pediatric antibiotic studies. Expert Rev Clin Pharmacol. 2019;12(12):1091-1098. https://doi.org/10.1080/17512433.2019.1695600
  23. Fernandez-Varon E, Carceles-Garcia C, Serrano-Rodriguez JM, Carceles-Rodriguez CM. Pharmacokinetics (PK), pharmacodynamics (PD), and PK-PD integration of ceftiofur after a single intravenous, subcutaneous and subcutaneous-LA administration in lactating goats. BMC Vet Res. 2016;12(1):232. https://doi.org/10.1186/s12917-016-0863-9
  24. Craigmill AL, Brown SA, Wetzlich SE, Gustafson CR, Arndt TS. Pharmacokinetics of ceftiofur and metabolites after single intravenous and intramuscular administration and multiple intramuscular administrations of ceftiofur sodium to sheep. J Vet Pharmacol Ther. 1997;20(2):139-144. https://doi.org/10.1046/j.1365-2885.1997.00820.x
  25. Olson SC, Beconi-Barker MG, Smith EB, Martin RA, Vidmar TJ, Adams LD. In vitro metabolism of ceftiofur in bovine tissues. J Vet Pharmacol Ther. 1998;21(2):112-120. https://doi.org/10.1046/j.1365-2885.1998.00118.x
  26. Zhou YF, Yu Y, Sun J, Tao MT, Zhou WJ, Li X, et al. Ex vivo pharmacokinetic/pharmacodynamic relationship of valnemulin against Clostridium perfringens in plasma, the small intestinal and caecal contents of rabbits. Anaerobe. 2016;39:150-157. https://doi.org/10.1016/j.anaerobe.2016.04.005
  27. Yan L, Xie S, Chen D, Pan Y, Tao Y, Qu W, et al. Pharmacokinetic and pharmacodynamic modeling of cyadox against Clostridium perfringens in swine. Sci Rep. 2017;7(1):4064. https://doi.org/10.1038/s41598-017-03970-9