Dosing of Ceftriaxone and Metronidazole for Children With Severe Acute Malnutrition

Joseph F Standing, Martin O Ongas, Caroline Ogwang, Nancy Kagwanja, Sheila Murunga, Shalton Mwaringa, Rehema Ali, Neema Mturi, Moline Timbwa, Christine Manyasi, Laura Mwalekwa, Victor L Bandika, Bernhards Ogutu, Joseph Waichungo, Karin Kipper, James A Berkley, FLACSAM-PK Study Group, Joseph F Standing, Martin O Ongas, Caroline Ogwang, Nancy Kagwanja, Sheila Murunga, Shalton Mwaringa, Rehema Ali, Neema Mturi, Moline Timbwa, Christine Manyasi, Laura Mwalekwa, Victor L Bandika, Bernhards Ogutu, Joseph Waichungo, Karin Kipper, James A Berkley, FLACSAM-PK Study Group

Abstract

Infants and young children with severe acute malnutrition (SAM) are treated with empiric broad-spectrum antimicrobials. Parenteral ceftriaxone is currently a second-line agent for invasive infection. Oral metronidazole principally targets small intestinal bacterial overgrowth. Children with SAM may have altered drug absorption, distribution, metabolism, and elimination. Population pharmacokinetics of ceftriaxone and metronidazole were studied, with the aim of recommending optimal dosing. Eighty-one patients with SAM (aged 2-45 months) provided 234 postdose pharmacokinetic samples for total ceftriaxone, metronidazole, and hydroxymetronidazole. Ceftriaxone protein binding was also measured in 190 of these samples. A three-compartment model adequately described free ceftriaxone, with a Michaelis-Menten model for concentration and albumin-dependent protein binding. A one-compartment model was used for both metronidazole and hydroxymetronidazole, with only 1% of hydroxymetronidazole predicted to be formed during first-pass. Simulations showed 80 mg/kg once daily of ceftriaxone and 12.5 mg/kg twice daily of metronidazole were sufficient to reach therapeutic targets.

© 2018 The Authors Clinical Pharmacology & Therapeutics published by Wiley Periodicals, Inc. on behalf of American Society for Clinical Pharmacology and Therapeutics.

Figures

Figure 1
Figure 1
Prediction corrected visual predictive check (VPC) for total and unbound ceftriaxone (top row) showing model simulated 95% confidence intervals for the simulated 2.5th, 50th, and 97.5th percentiles.
Figure 2
Figure 2
Prediction corrected visual predictive check (VPC) for metronidazole and hydroxymetronidazole showing model simulated 95% confidence intervals for the simulated 2.5th, 50th, and 97.5th percentiles.
Figure 3
Figure 3
ft > MIC for 50, 80, and 100 mg/kg once daily. The 5th, 10th, 20th, and 50th percentiles show the cutoffs for ft > MIC for 95, 90, 80, and 50% of patients, respectively. Lines at a MIC cutoff of 2 mg/L and ft > MIC of 0.5 show the proposed MIC target.
Figure 4
Figure 4
Simulations for free metronidazole plus hydroxymetronidazole under 10, 12.5, and 15 mg/kg twice daily dosing, with hydroxymetronidazole concentrations being multiplied by 0.65 to reflect the assumed lower activity. A break‐point of 8 mg/L is highlighted. (a) Yhe ft > MIC with a value of 0.9 highlighted; whereas (b) shows the AUC:MIC ratio with a cutoff of 70 mg.h/L highlighted. A comparison of 24‐ and 48‐hour target attainment is given in the upper and lower panels, respectively.
Figure 5
Figure 5
Boxplots to illustrate simulated covariate effects on translation to ft > MIC. Probability of target attainment reported beneath each box with a target of 50% of the dosing interval with concentrations above 2 mg/L at 24 hours for free ceftriaxone, and 90% of the dosing interval with concentrations of free metronidazole and 0.65 x hydroxymetronidazole above 8 mg/L at 48 hours. (a) Effect of edema score of 1 or more on ceftriaxone target attainment. (b) Effect of edema score of 2 or more on metronidazole target attainment. (c) Effect of serum creatinine on ceftriaxone target attainment.

References

    1. Tickell, K.D. & Denno, D.M. Inpatient management of children with severe acute malnutrition: a review of WHO guidelines. Bull. World Health Organ. 94, 642–651 (2016).
    1. Lazzerini, M. & Tickell, D. Antibiotics in severely malnourished children: systematic review of efficacy, safety and pharmacokinetics. Bull. World Health Organ. 89, 594–607 (2011).
    1. Williams, C. M. & Berkley, J. A. severe acute malnutrition update: Current WHO guidelines and the WHO essential medicine list for children. J. Clin. Pharmacol. <> (2016).
    1. Schleibinger, M. et al Protein binding characteristics and pharmacokinetics of ceftriaxone in intensive care unit patients. Br. J. Clin. Pharmacol. 80, 525–533 (2015).
    1. Organization, W.H. Pocket Book of Hospital Care for Children: Guidelines for the Management of Common Childhood Illnesses. (World Health Organization, 2013). <>
    1. Heikens, G.T. , Schofield, W.N. & Dawson, S. The Kingston Project. II. The effects of high energy supplement and metronidazole on malnourished children rehabilitated in the community: anthropometry. Eur. J. Clin. Nutr. 47, 160–173 (1993).
    1. Lares‐Asseff, I. , Cravioto, J. , Santiago, P. & Perez‐Ortiz, B. Pharmacokinetics of metronidazole in severely malnourished and nutritionally rehabilitated children. Clin. Pharmacol. Ther. 51, 42–50 (1992).
    1. Sprandel, K.A. et al Population pharmacokinetic modeling and Monte Carlo simulation of varying doses of intravenous metronidazole. Diagn. Microbiol. Infect. Dis. 55, 303–309 (2006).
    1. Germovsek, E. , Barker, C.I. , Sharland, M. & Standing, J.F. Scaling clearance in paediatric pharmacokinetics: All models are wrong, which are useful? Br. J. Clin. Pharmacol. 83, 777–790 (2017).
    1. Rhodin, M.M. et al Human renal function maturation: a quantitative description using weight and postmenstrual age. Pediatr. Nephrol. 24, 67–76 (2009).
    1. Upreti, V.V. & Wahlstrom, J.L. Meta‐analysis of hepatic cytochrome P450 ontogeny to underwrite the prediction of pediatric pharmacokinetics using physiologically based pharmacokinetic modeling. J. Clin. Pharmacol. 56, 266–283 (2016).
    1. Ceriotti, F. et al Reference intervals for serum creatinine concentrations: assessment of available data for global application. Clin. Chem. 54, 559–566 (2008).
    1. Perry, T.R. & Schentag, J.J. Clinical use of ceftriaxone: a pharmacokinetic‐pharmacodynamic perspective on the impact of minimum inhibitory concentration and serum protein binding. Clin. Pharmacokinet. 40, 685–694 (2001).
    1. Toutain, P.L. & Bousquet‐Melou, A. Free drug fraction vs free drug concentration: a matter of frequent confusion. J. Vet. Pharmacol. Ther. 25, 460–463 (2002).
    1. Germovsek, E. et al Development and evaluation of a gentamicin pharmacokinetic model that facilitates opportunistic gentamicin therapeutic drug monitoring in neonates and infants. Antimicrob. Agents Chemother. 60, 4869–4877 (2016).
    1. Johansson, A.M. et al A population pharmacokinetic/pharmacodynamic model of methotrexate and mucositis scores in osteosarcoma. Ther. Drug Monit. 33, 711–718 (2011).
    1. Hennig, S. , Standing, J.F. , Staatz, C.E. & Thomson, A.H. Population pharmacokinetics of tobramycin in patients with and without cystic fibrosis. Clin. Pharmacokinet. 52, 289–301 (2013).
    1. Savic, R. M. & Karlsson, M. O. Importance of shrinkage in empirical bayes estimates for diagnostics: problems and solutions. AAPS J 11, 558–569 (2009).
    1. Seaton, C. et al Population pharmacokinetics of a single daily intramuscular dose of gentamicin in children with severe malnutrition. J. Antimicrob. Chemother. 59, 681–689 (2007).
    1. Standing, J. F. , Anderson, B. J. , Holford, N. H. , Lutsar, I. & Metsvaht, T. Comment on pharmacokinetic studies in neonates: the utility of an opportunistic sampling design. Clin. Pharmacokinet. 54, 1287–1288 (2015).
    1. Garot, D. et al Population pharmacokinetics of ceftriaxone in critically ill septic patients: a reappraisal. Br. J. Clin. Pharmacol. 72, 758–767 (2011).
    1. Lodise, T.P. et al Comparison of the probability of target attainment between ceftriaxone and cefepime in the cerebrospinal fluid and serum against Streptococcus pneumoniae. Diagn. Microbiol. Infect. Dis. 58, 445–452 (2007).
    1. Fukumoto, K. , Aida, S. , Oishi, T. & Ueno, K. Pharmacokinetics of ceftriaxione, a third‐generation cephalosporin, in pediatric patients. Biol. Pharm. Bull. 32, 1139–1141 (2009).
    1. De Cock, R.F. et al Population pharmacokinetic modeling of total and unbound cefazolin plasma concentrations as a guide for dosing in preterm and term neonates. J. Antimicrob. Chemother. 69, 1330–1338 (2014).
    1. Schaad, U.B. The cephalosporin compounds in severe neonatal infection. Eur. J. Pediatr. 141, 143–146 (1984).
    1. Stoeckel, K. , McNamara, P.J. , Brandt, R. , Plozza‐Nottebrock, H. & Ziegler, W.H. Effects of concentration‐dependent plasma protein binding on ceftriaxone kinetics. Clin. Pharmacol. Ther. 29, 650–657 (1981).
    1. Kristoffersson, A.N. et al Simulation‐based evaluation of PK/PD indices for meropenem across patient groups and experimental designs. Pharm. Res. 33, 1115–1125 (2016).
    1. Barker, C.I. et al Pharmacokinetic/pharmacodynamic modeling approaches in paediatric infectious diseases and immunology. Adv. Drug Deliv. Rev. 73, 127–139 (2014).
    1. Huang, Y. , Carroll, K.C. , Cosgrove, S.E. & Tamma, P.D. Determining the optimal ceftriaxone MIC for triggering extended‐spectrum Î2‐lactamase confirmatory testing. J. Clin. Microbiol. 52, 2228–2230 (2014).
    1. Cheung, S.Y. , Yates, J.W. & Aarons, L. The design and analysis of parallel experiments to produce structurally identifiable models. J. Pharmacokinet. Pharmacodyn. 40, 93–100 (2013).
    1. Standing, J.F. et al Oseltamivir pharmacokinetics and clinical experience in neonates and infants during an outbreak of H1N1 influenza A virus infection in a neonatal intensive care unit. Antimicrob. Agents Chemother. 56, 3833–3840 (2012).
    1. Haller, I. In vitro activity of the two principal oxidative metabolites of metronidazole against Bacteroides fragilis and related species. Antimicrob. Agents Chemother. 22, 165–166 (1982).
    1. Cohen‐Wolkowiez, M. et al Population pharmacokinetics of metronidazole evaluated using scavenged samples from preterm infants. Antimicrob. Agents Chemother. 56, 1828–1837 (2012).
    1. Agudelo, M. & Vesga, O. Therapeutic equivalence requires pharmaceutical, pharmacokinetic, and pharmacodynamic identities: true bioequivalence of a generic product of intravenous metronidazole. Antimicrob. Agents Chemother. 56, 2659–2665 (2012).
    1. Ongas, M. et al Liquid chromatography–tandem mass spectrometry for the simultaneous quantitation of ceftriaxone, metronidazole and hydroxymetronidazole in plasma from seriously ill, severely malnourished children [version 1; referees: awaiting peer review]. Wellcome Open Res 2, 46 (2017).
    1. Price, P.S. et al Modeling interindividual variation in physiological factors used in PBPK models of humans. Crit. Rev. Toxicol. 33, 469–503 (2003).
    1. Pearce, R.E. , Cohen‐Wolkowiez, M. , Sampson, M.R. & Kearns, G.L. The role of human cytochrome P450 enzymes in the formation of 2‐hydroxymetronidazole: CYP2A6 is the high affinity (low Km) catalyst. Drug Metab. Dispos. 41, 1686–1694 (2013).
    1. Beal, S. , Sheiner, L.B. , Boeckmann, A. & Bauer, R.J. NONMEM User's Guides. (1989–2013). Icon Development Solutions, Ellicott City, MD: (2013).
    1. Lindbom, L. , Pihlgren, P. , Jonsson, E.N. & Jonsson, N. PsN‐Toolkit — a collection of computer intensive statistical methods for non‐linear mixed effect modeling using NONMEM. Comput. Methods Programs Biomed. 79, 241–257 (2005).

Source: PubMed

3
Abonnieren