Current Ceftriaxone Dose Recommendations are Adequate for Most Critically Ill Children: Results of a Population Pharmacokinetic Modeling and Simulation Study

Stan J F Hartman, Parth J Upadhyay, Nienke N Hagedoorn, Ron A A Mathôt, Henriëtte A Moll, Michiel van der Flier, Michiel F Schreuder, Roger J Brüggemann, Catherijne A Knibbe, Saskia N de Wildt, Stan J F Hartman, Parth J Upadhyay, Nienke N Hagedoorn, Ron A A Mathôt, Henriëtte A Moll, Michiel van der Flier, Michiel F Schreuder, Roger J Brüggemann, Catherijne A Knibbe, Saskia N de Wildt

Abstract

Background and objective: Ceftriaxone is a cornerstone antibiotic for critically ill children with severe infections. Despite its widespread use, information on the pharmacokinetics of ceftriaxone is lacking in this population. We aimed to determine ceftriaxone pharmacokinetics in critically ill children and to propose ceftriaxone dosing guidelines resulting in adequate target attainment using population pharmacokinetic modeling and simulation.

Methods: Critically ill children (aged 0-18 years) treated with intravenous ceftriaxone (100 mg/kg once daily, infused in 30 minutes) and a central or arterial line in place were eligible. Opportunistic blood sampling for total and unbound ceftriaxone concentrations was used. Population pharmacokinetic analysis was performed using non-linear mixed-effects modeling on NONMEM™ Version 7.4.3. Simulations were performed to select optimal doses using probability of target attainment for two pharmacokinetic targets of the minimum inhibitory concentration (MIC) reflecting the susceptibility of pathogens (f T > MIC 100% and fT > 4 × MIC 100%).

Results: Two hundred and five samples for total and 43 time-matched samples for unbound plasma ceftriaxone concentrations were collected from 45 patients, median age 2.5 (range 0.1-16.7) years. A two-compartment model with bodyweight as the co-variate for volume of distribution and clearance, and creatinine-based estimated glomerular filtration rate as an additional covariate for clearance, best described ceftriaxone pharmacokinetics. For a typical patient (2.5 years, 14 kg) with an estimated glomerular filtration rate of 80 mL/min/1.73 m2, the current 100-mg/kg once-daily dose results in a probability of target attainment of 96.8% and 60.8% for a MIC of 0.5 mg/L and 4 × MIC (2 mg/L), respectively, when using fT > MIC 100% as a target. For a 50-mg/kg twice-daily regimen, the probability of target attainment was 99.9% and 93.4%, respectively.

Conclusions: The current dosing regimen of ceftriaxone provides adequate exposure for susceptible pathogens in most critically ill children. In patients with an estimated glomerular filtration rate of > 80 mL/min/1.73 m2 or in areas with a high prevalence of less-susceptible pathogens (MIC ≥ 0.5 mg/L), a twice-daily dosing regimen of 50 mg/kg can be considered to improve target attainment.

Clinical trial registration: POPSICLE study (ClinicalTrials.gov, NCT03248349, registered 14 August, 2017), PERFORM study (ClinicalTrials.gov, NCT03502993, registered 19 April, 2018).

Conflict of interest statement

Roger J. Brüggemann has no conflicts of interest that are directly relevant to the content of this article. Outside of this work, he has served as consultant to and has received unrestricted research grants from Astellas Pharma Inc., F2G, Amplyx, Gilead Sciences, Merck Sharpe and Dohme Corp., and Pfizer Inc. All payments were invoiced by the Radboud University Medical Centre. Saskia N. de Wildt has no conflicts of interest that are directly relevant to the content of this article. Outside of this work she has served as consultant to and had received unrestricted research grant or in kind support from UCB Pharma, Spingotec, and Pfizer Inc. Saskia N. de Wildt is the director of the Dutch Pediatric Formulary and its internationally licensed versions (Stichting Nederlands Kenniscentrum Pharmacotherapy voor Kinderen & Kinderformularium B.V.) All payments were invoiced by the Radboud University Medical Centre. Stan J.F. Hartman, Parth J. Upadhyay, Nienke N. Hagedoorn, Ron A.A. Mathôt, Henriëtte A. Moll, Michiel van der Flier, Michiel F. Schreuder, and Catherijne A. Knibbe have no conflicts of interest that are directly relevant to the content of this article.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
Goodness of fit of predicted total (gray) and unbound ceftriaxone (white) concentrations. Diagnostic plots for ceftriaxone: observed ceftriaxone concentrations vs population predictions and individual predictions and conditional weighted residuals vs time after the last dose and population predictions. Dots represent individual total (gray) and unbound (white) ceftriaxone concentrations. The red line represents the regression line, the blue line represents the correlation of our observations and predictions. CWRES conditional weighted residuals
Fig. 2
Fig. 2
Impact of the final covariates weight and estimated glomerular filtration (eGFR) on ceftriaxone clearance. Dots represent the individual estimated clearance values, with colors and shapes representing different categories of median eGFRcreat during the study period (red squares <30 mL/min/1.73 m2, yellow dots 30–80 mL/min/1.73 m2, blue triangles 80–120 mL/min/1.73 m2, and green diamonds > 120 mL/min/1.73 m2. Solid lines represent the range of weight in each eGFR category observed in our dataset. Dotted lines represent extrapolated clearance for each category for a weight of 0–80 kg
Fig. 3
Fig. 3
Probability of target attainment (PTA) for different ceftriaxone dosing regimens for patients with a median estimated glomerular filtration rate for different MICs. The PTA for patients with p50 estimated glomerular filtration rate (85.22 mL/min/1.73 m2) and albumin levels (27 g/L) for the four tested dosing regimens (red circles 100 mg/kg once daily; yellow squares 50 mg/kg once daily; blue diamonds 80 mg/kg once daily, and green triangles 50 mg/kg twice daily). The dashed horizontal line represents target PTA of 90%. Shaded areas represent MICs selected for the primary (0.5 mg/L, gray) and high (2.0 mg/L, blue) MIC target

References

    1. Steele RW, Eyre LB, Bradsher RW, Weinfeld RE, Patel IH, Spicehandler J. Pharmacokinetics of ceftriaxone in pediatric patients with meningitis. Antimicrob Agents Chemother. 1983;23(2):191–194. doi: 10.1128/AAC.23.2.191.
    1. Fusco NM, Parbuoni KA, Morgan JA. Time to first antimicrobial administration after onset of sepsis in critically ill children. J Pediatr Pharmacol Ther. 2015;20(1):37–44. doi: 10.5863/1551-6776-20.1.37.
    1. Hartman SJF, Brüggemann RJ, Orriëns L, Dia N, Schreuder MF, de Wildt SN. Pharmacokinetics and target attainment of antibiotics in critically ill children: a systematic review of current literature. Clin Pharmacokinet. 2020;59(2):173–205. doi: 10.1007/s40262-019-00813-w.
    1. Roberts JA, Abdul-Aziz MH, Lipman J, Mouton JW, Vinks AA, Felton TW, et al. Individualised antibiotic dosing for patients who are critically ill: challenges and potential solutions. Lancet Infect Dis. 2014;14(6):498–509. doi: 10.1016/S1473-3099(14)70036-2.
    1. De Waele JJ, Lipman J, Akova M, Bassetti M, Dimopoulos G, Kaukonen M, et al. Risk factors for target non-attainment during empirical treatment with beta-lactam antibiotics in critically ill patients. Intensive Care Med. 2014;40(9):1340–1351. doi: 10.1007/s00134-014-3403-8.
    1. Schaad UB, Stoeckel K. Single-dose pharmacokinetics of ceftriaxone in infants and young children. Antimicrob Agents Chemother. 1982;21(2):248–253. doi: 10.1128/aac.21.2.248.
    1. Ollivier J, Carrie C, d'Houdain N, Djabarouti S, Petit L, Xuereb F, et al. Are standard dosing regimens of ceftriaxone adapted for critically ill patients with augmented creatinine clearance? Antimicrob Agents Chemother. 2019;63(3):e02134–e2218. doi: 10.1128/AAC.02134-18.
    1. Udy AA, De Waele JJ, Lipman J. Augmented renal clearance and therapeutic monitoring of beta-lactams. Int J Antimicrob Agents. 2015;45(4):331–333. doi: 10.1016/j.ijantimicag.2014.12.020.
    1. Dhont E, Van Der Heggen T, De Jaeger A, Vande Walle J, De Paepe P, De Cock PA. Augmented renal clearance in pediatric intensive care: are we undertreating our sickest patients? Pediatr Nephrol. 2020;35(1):25–39. doi: 10.1007/s00467-018-4120-2.
    1. Kearns GL, Abdel-Rahman SM, Alander SW, Blowey DL, Leeder JS, Kauffman RE. Developmental pharmacology: drug disposition, action, and therapy in infants and children. N Engl J Med. 2003;349(12):1157–1167. doi: 10.1056/NEJMra035092.
    1. Cies JJ, Moore WS, 2nd, Enache A, Chopra A. Beta-lactam therapeutic drug management in the PICU. Crit Care Med. 2018;46(2):272–279. doi: 10.1097/CCM.0000000000002817.
    1. Slater A, Shann F, Group APS The suitability of the Pediatric Index of Mortality (PIM), PIM2, the Pediatric Risk of Mortality (PRISM), and PRISM III for monitoring the quality of pediatric intensive care in Australia and New Zealand. Pediatr Crit Care Med. 2004;5(5):447–454. doi: 10.1097/01.PCC.0000138557.31831.65.
    1. Pollack MM, Patel KM, Ruttimann UE. PRISM III: an updated Pediatric Risk of Mortality score. Crit Care Med. 1996;24(5):743–752. doi: 10.1097/00003246-199605000-00004.
    1. Leteurtre S, Duhamel A, Salleron J, Grandbastien B, Lacroix J, Leclerc F, et al. PELOD-2: an update of the PEdiatric logistic organ dysfunction score. Crit Care Med. 2013;41(7):1761–1773. doi: 10.1097/CCM.0b013e31828a2bbd.
    1. Schwartz GJ, Schneider MF, Maier PS, Moxey-Mims M, Dharnidharka VR, Warady BA, et al. Improved equations estimating GFR in children with chronic kidney disease using an immunonephelometric determination of cystatin C. Kidney Int. 2012;82(4):445–453. doi: 10.1038/ki.2012.169.
    1. Kinderformularium. Dutch Pediatric Formulary: ceftriaxone. 2020. . Accessed 6 May 2021.
    1. Nickolai DJ, Lammel CJ, Byford BA, Morris JH, Kaplan EB, Hadley WK, et al. Effects of storage temperature and pH on the stability of eleven beta-lactam antibiotics in MIC trays. J Clin Microbiol. 1985;21(3):366–370. doi: 10.1128/jcm.21.3.366-370.1985.
    1. Tsai D, Lipman J, Roberts JA. Pharmacokinetic/pharmacodynamic considerations for the optimization of antimicrobial delivery in the critically ill. Curr Opin Crit Care. 2015;21(5):412–420. doi: 10.1097/MCC.0000000000000229.
    1. De Cock RF, Smits A, Allegaert K, de Hoon J, Saegeman V, Danhof M, et al. Population pharmacokinetic modelling of total and unbound cefazolin plasma concentrations as a guide for dosing in preterm and term neonates. J Antimicrob Chemother. 2014;69(5):1330–1338. doi: 10.1093/jac/dkt527.
    1. Khan MW, Wang YK, Wu YE, Tang BH, Kan M, Shi HY, et al. Population pharmacokinetics and dose optimization of ceftriaxone for children with community-acquired pneumonia. Eur J Clin Pharmacol. 2020;76(11):1547–1556. doi: 10.1007/s00228-020-02939-4.
    1. Grupper M, Kuti JL, Nicolau DP. Continuous and prolonged intravenous beta-lactam dosing: implications for the clinical laboratory. Clin Microbiol Rev. 2016;29(4):759–772. doi: 10.1128/CMR.00022-16.
    1. Abdul-Aziz MH, Alffenaar JC, Bassetti M, Bracht H, Dimopoulos G, Marriott D, et al. Antimicrobial therapeutic drug monitoring in critically ill adult patients: a position paper. Intensive Care Med. 2020;46(6):1127–1153. doi: 10.1007/s00134-020-06050-1.
    1. Roberts JA, Lipman J. Pharmacokinetic issues for antibiotics in the critically ill patient. Crit Care Med. 2009;37(3):840–851. doi: 10.1097/CCM.0b013e3181961bff.
    1. European Committee on Antimicrobial Susceptibility Testing. Breakpoint tables for interpretation of MICs and zone diameters. Version 10.0, 2020. . Accessed 6 May 2021.
    1. Schleibinger M, Steinbach CL, Topper C, Kratzer A, Liebchen U, Kees F, et al. Protein binding characteristics and pharmacokinetics of ceftriaxone in intensive care unit patients. Br J Clin Pharmacol. 2015;80(3):525–533. doi: 10.1111/bcp.12636.
    1. Garot D, Respaud R, Lanotte P, Simon N, Mercier E, Ehrmann S, et al. Population pharmacokinetics of ceftriaxone in critically ill septic patients: a reappraisal. Br J Clin Pharmacol. 2011;72(5):758–767. doi: 10.1111/j.1365-2125.2011.04005.x.
    1. Hayton WL, Stoeckel K. Age-associated changes in ceftriaxone pharmacokinetics. Clin Pharmacokinet. 1986;11(1):76–86. doi: 10.2165/00003088-198611010-00005.
    1. Soveri I, Berg UB, Bjork J, Elinder CG, Grubb A, Mejare I, et al. Measuring GFR: a systematic review. Am J Kidney Dis. 2014;64(3):411–424. doi: 10.1053/j.ajkd.2014.04.010.
    1. Guignard JP, Drukker A. Why do newborn infants have a high plasma creatinine? Pediatrics. 1999;103(4):e49. doi: 10.1542/peds.103.4.e49.
    1. De Cock PA, Standing JF, Barker CI, de Jaeger A, Dhont E, Carlier M, et al. Augmented renal clearance implies a need for increased amoxicillin-clavulanic acid dosing in critically ill children. Antimicrob Agents Chemother. 2015;59(11):7027–7035. doi: 10.1128/AAC.01368-15.
    1. De Cock PA, van Dijkman SC, de Jaeger A, Willems J, Carlier M, Verstraete AG, et al. Dose optimization of piperacillin/tazobactam in critically ill children. J Antimicrob Chemother. 2017;72(7):2002–2011. doi: 10.1093/jac/dkx093.
    1. Béranger A, Benaboud S, Urien S, Moulin F, Bille E, Lesage F, et al. Piperacillin population pharmacokinetics and dosing regimen optimization in critically ill children with normal and augmented renal clearance. Clin Pharmacokinet. 2019;58(2):223–233. doi: 10.1007/s40262-018-0682-1.
    1. Béranger A, Oualha M, Urien S, Genuini M, Renolleau S, Aboura R, et al. Population pharmacokinetic model to optimize cefotaxime dosing regimen in critically ill children. Clin Pharmacokinet. 2018;57(7):867–875. doi: 10.1007/s40262-017-0602-9.
    1. Cies JJ, Shankar V, Schlichting C, Kuti JL. Population pharmacokinetics of piperacillin/tazobactam in critically ill young children. Pediatr Infect Dis J. 2014;33(2):168–173. doi: 10.1097/INF.0b013e3182a743c7.
    1. Jones AE, Barnes ND, Tasker TC, Horton R. Pharmacokinetics of intravenous amoxycillin and potassium clavulanate in seriously ill children. J Antimicrob Chemother. 1990;25(2):269–274. doi: 10.1093/jac/25.2.269.
    1. Pottel H, Vrydags N, Mahieu B, Vandewynckele E, Croes K, Martens F. Establishing age/sex related serum creatinine reference intervals from hospital laboratory data based on different statistical methods. Clin Chim Acta. 2008;396(1–2):49–55.

Source: PubMed

3
Iratkozz fel