IV and oral fosfomycin pharmacokinetics in neonates with suspected clinical sepsis

Zoe Kane, Silke Gastine, Christina Obiero, Phoebe Williams, Sheila Murunga, Johnstone Thitiri, Sally Ellis, Erika Correia, Borna Nyaoke, Karin Kipper, John van den Anker, Mike Sharland, James A Berkley, Joseph F Standing, Zoe Kane, Silke Gastine, Christina Obiero, Phoebe Williams, Sheila Murunga, Johnstone Thitiri, Sally Ellis, Erika Correia, Borna Nyaoke, Karin Kipper, John van den Anker, Mike Sharland, James A Berkley, Joseph F Standing

Abstract

Background: Fosfomycin has the potential to be re-purposed as part of a combination therapy to treat neonatal sepsis where resistance to current standard of care (SOC) is common. Limited data exist on neonatal fosfomycin pharmacokinetics and estimates of bioavailability and CSF/plasma ratio in this vulnerable population are lacking.

Objectives: To generate data informing the appropriate dosing of IV and oral fosfomycin in neonates using a population pharmacokinetic analysis of plasma and CSF data.

Methods: The NeoFosfo study (NCT03453177) was a randomized trial that examined the safety and pharmacokinetics of fosfomycin comparing SOC versus SOC plus fosfomycin. Sixty-one neonates received fosfomycin (100 mg/kg IV q12h for 48 h) and then they converted to oral therapy at the same dose. Two plasma pharmacokinetic samples were taken following the first IV and oral doses, sample times were randomized to cover the whole pharmacokinetic profile and opportunistic CSF pharmacokinetic samples were collected. A population pharmacokinetic model was developed in NONMEM and simulations were performed.

Results: In total, 238 plasma and 15 CSF concentrations were collected. A two-compartment disposition model, with an additional CSF compartment and first-order absorption, best described the data. Bioavailability was estimated as 0.48 (95% CI = 0.347-0.775) and the CSF/plasma ratio as 0.32 (95% CI = 0.272-0.409). Allometric weight and postmenstrual age (PMA) scaling was applied; additional covariates included postnatal age (PNA) on clearance and CSF protein on CSF/plasma ratio.

Conclusions: Through this analysis a population pharmacokinetic model has been developed that can be used alongside currently available pharmacodynamic targets to select a neonatal fosfomycin dose based on an infant's PMA, PNA and weight.

© The Author(s) 2021. Published by Oxford University Press on behalf of the British Society for Antimicrobial Chemotherapy.

Figures

Figure 1.
Figure 1.
Observed CSF and plasma concentration versus time data for all subjects. The dashed lines represent mean concentrations, which are 37.6 mg/L in CSF and 70.1 and 201.7 mg/L in plasma following oral and IV dosing, respectively; the solid lines represent the loess smooth curves. PO, oral.
Figure 2.
Figure 2.
VPCs showing the observed data (black circles) and the 2.5th, 50th and 97.5th percentiles of the observed data (black lines) compared with the 95% CIs of the corresponding simulations (prediction intervals) from the final model (shaded areas). The top panel shows plasma following IV dosing, the middle panel shows plasma following oral dosing and the bottom panel shows CSF. The 2.5th and 97.5th percentiles, and corresponding prediction intervals, are not presented in the bottom panel due to the size of the CSF dataset evaluated.
Figure 3.
Figure 3.
Visualization of the PNA effect on clearance; individual predicted clearances have already been scaled for PMA and weight. Data points are grouped by subject ID (n =60); the left-hand panel shows individual fractional clearance at time = 0 and the right-hand panel shows individual fractional clearance at all PK sampling timepoints. The solid black line in each panel represents the model estimated PNA maturation function.
Figure 4.
Figure 4.
TA plots for various dose schemes using the full simulation population. The top row presents predicted AUC/MIC ratio in plasma following IV dosing, while the bottom row shows results following oral dosing. A comparison of 100, 150 and 200 mg/kg q12h is given for IV and 100, 200 and 300 mg/kg q12h for oral. The continuous black line is the predicted AUC/MIC ratio achieved by 95% of the population (5th percentile), while the typical patient (50th percentile) is shown by the dotted line. AUC/MIC target ratios for stasis (19.3), 1 log reduction (87.5) and resistance suppression (3136) are shown by the grey horizontal reference lines. The grey vertical reference lines highlight MIC values of 4 and 32 mg/L. BID, twice daily; PO, oral.

References

    1. Laxminarayan R, Matsoso P, Pant S. et al. Access to effective antimicrobials: a worldwide challenge. Lancet 2016; 387: 168–75.
    1. Chaurasia S, Sivanandan S, Agarwal R. et al. Neonatal sepsis in South Asia: huge burden and spiralling antimicrobial resistance. BMJ 2019; 364: k5314.
    1. Iroh Tam PY, Musicha P, Kawaza K. et al. Emerging resistance to empiric antimicrobial regimens for pediatric bloodstream infections in Malawi (1998-2017). Clin Infect Dis 2019; 69: 61–8.
    1. Hendlin D, Stapley EO, Jackson M. et al. Phosphonomycin, a new antibiotic produced by strains of streptomyces. Science 1969; 166: 122–3.
    1. Li G, Standing JF, Bielicki J. et al. The potential role of fosfomycin in neonatal sepsis caused by multidrug-resistant bacteria. Drugs 2017; 77: 941–50.
    1. Williams PCM, Waichungo J, Gordon NC. et al. The potential of fosfomycin for multi-drug resistant sepsis: an analysis of in vitro activity against invasive paediatric Gram-negative bacteria. J Med Microbiol 2019; 68: 711–9.
    1. Molina MA, Olay T, Quero J.. Pharmacodynamic data on fosfomycin in underweight infants during the neonatal period. Chemotherapy 1977; 23 Suppl 1: 217–22.
    1. Guggenbichler JP, Kienel G, Frisch H.. Fosfomycin, a new antibiotic drug (author’s transl). Padiatr Padol 1978; 13: 429–36.
    1. Guibert M, Magny JF, Poudenx F. et al. Comparative pharmacokinetics of fosfomycin in the neonate: 2 modes of administration. Pathol Biol (Paris) 1987; 35: 750–2.
    1. Suzuki S, Murayama Y, Sugiyama E. et al. Dose estimation for renal-excretion drugs in neonates and infants based on physiological development of renal function. Yakugaku Zasshi 2009; 129: 829–42.
    1. Shimizu K. Fosfomycin: absorption and excretion. Chemotherapy 1977; 23 Suppl 1: 153–8.
    1. Bundgaard H. Acid-catalyzed hydrolysis of fosfomycin and its implication in oral absorption of the drug. Int J Pharmaceut 1980; 6: 1–9.
    1. Gobernado M, Garcia J, Santos M. et al. Renal insufficiency and fosfomycin. Chemotherapy 1977; 23 Suppl 1: 200–3.
    1. Wenzler E, Ellis-Grosse EJ, Rodvold KA.. Pharmacokinetics, safety, and tolerability of single-dose intravenous (ZTI-01) and oral fosfomycin in healthy volunteers. Antimicrob Agents Chemother 2017; 61: e00775-17.
    1. Bergan T, Thorsteinsson SB, Albini E.. Pharmacokinetic profile of fosfomycin trometamol. Chemotherapy 1993; 39: 297–301.
    1. Kuhnen E, Pfeifer G, Frenkel C.. Penetration of fosfomycin into cerebrospinal fluid across non-inflamed and inflamed meninges. Infection 1987; 15: 422–4.
    1. Pfausler B, Spiss H, Dittrich P. et al. Concentrations of fosfomycin in the cerebrospinal fluid of neurointensive care patients with ventriculostomy-associated ventriculitis. J Antimicrob Chemother 2004; 53: 848–52.
    1. Traunmuller F, Popovic M, Konz KH. et al. A reappraisal of current dosing strategies for intravenous fosfomycin in children and neonates. Clin Pharmacokinet 2011; 50: 493–503.
    1. Stockmann C, Barrett JS, Roberts JK. et al. Use of modeling and simulation in the design and conduct of pediatric clinical trials and the optimization of individualized dosing regimens. CPT Pharmacometrics Syst Pharmacol 2015; 4: 630–40.
    1. Ogungbenro K, Aarons L.. How many subjects are necessary for population pharmacokinetic experiments? Confidence interval approach. Eur J Clin Pharmacol 2008; 64: 705–13.
    1. EMA. Guideline on Bioanalytical Method Validation (EMEA/CHMP/EWP/192217/2009). 2011. .
    1. Rhodin MM, Anderson BJ, Peters AM. et al. Human renal function maturation: a quantitative description using weight and postmenstrual age. Pediatr Nephrol 2009; 24: 67–76.
    1. Germovsek E, Kent A, Metsvaht T. et al. Development and evaluation of a gentamicin pharmacokinetic model that facilitates opportunistic gentamicin therapeutic drug monitoring in neonates and infants. Antimicrob Agents Chemother 2016; 60: 4869–77.
    1. Germovsek E, Lutsar I, Kipper K. et al. Plasma and CSF pharmacokinetics of meropenem in neonates and young infants: results from the NeoMero studies. J Antimicrob Chemother 2018; 73: 1908–16.
    1. Anderson BJ, Holford NHG.. Negligible impact of birth on renal function and drug metabolism. Paediatr Anaesth 2018; 28: 1015–21.
    1. Savic RM, Cowan MJ, Dvorak CC. et al. Effect of weight and maturation on busulfan clearance in infants and small children undergoing hematopoietic cell transplantation. Biol Blood Marrow Transplant 2013; 19: 1608–14.
    1. Ceriotti F, Boyd JC, Klein G. et al. Reference intervals for serum creatinine concentrations: assessment of available data for global application. Clin Chem 2008; 54: 559–66.
    1. Valitalo P, Kumpulainen E, Manner M. et al. Plasma and cerebrospinal fluid pharmacokinetics of naproxen in children. J Clin Pharmacol 2012; 52: 1516–26.
    1. Kumpulainen E, Valitalo P, Kokki M. et al. Plasma and cerebrospinal fluid pharmacokinetics of flurbiprofen in children. Br J Clin Pharmacol 2010; 70: 557–66.
    1. Johanson CE, Duncan JA, Klinge PM. et al. Multiplicity of cerebrospinal fluid functions: new challenges in health and disease. Cerebrospinal Fluid Res 2008; 5: 10.
    1. R Foundation for Statistical Computing - Core Team. R: A Language and Environment for Statistical Computing. 2013. .
    1. Lindbom L, Pihlgren P, Jonsson EN. et al. PsN-Toolkit–a collection of computer intensive statistical methods for non-linear mixed effect modeling using NONMEM. Comput Methods Programs Biomed 2005; 79: 241–57.
    1. Jonsson EN, Karlsson MO.. Xpose–an S-PLUS based population pharmacokinetic/pharmacodynamic model building aid for NONMEM. Comput Methods Programs Biomed 1999; 58: 51–64.
    1. Upton RN. Calculating the hybrid (macro) rate constants of a three-compartment mamillary pharmacokinetic model from known micro-rate constants. J Pharmacol Toxicol Methods 2004; 49: 65–8.
    1. Rich B. linpk: Generate Concentration-Time Profiles From Linear PK Systems. 2018. .
    1. Merino-Bohorquez V, Docobo-Perez F, Sojo J. et al. Population pharmacokinetics and pharmacodynamics of fosfomycin in non-critically ill patients with bacteremic urinary infection caused by multidrug-resistant Escherichia coli. Clin Microbiol Infect 2018; 24: 1177–83.
    1. Lepak AJ, Zhao M, VanScoy B. et al. In vivo pharmacokinetics and pharmacodynamics of ZTI-01 (fosfomycin for injection) in the neutropenic murine thigh infection model against Escherichia coli, Klebsiella pneumoniae, and Pseudomonas aeruginosa. Antimicrob Agents Chemother 2017; 61: e00476-17.
    1. Docobo-Perez F, Drusano GL, Johnson A. et al. Pharmacodynamics of fosfomycin: insights into clinical use for antimicrobial resistance. Antimicrob Agents Chemother 2015; 59: 5602–10.
    1. Standing JF, Ongas MO, Ogwang C. et al. Dosing of ceftriaxone and metronidazole for children with severe acute malnutrition. Clin Pharmacol Ther 2018; 104: 1165–74.
    1. Zeitlinger MA, Sauermann R, Traunmuller F. et al. Impact of plasma protein binding on antimicrobial activity using time-killing curves. J Antimicrob Chemother 2004; 54: 876–80.
    1. Kirby WM. Pharmacokinetics of fosfomycin. Chemotherapy 1977; 23 Suppl 1: 141–51.
    1. Botwinski CA, Falco GA.. Transition to postnatal renal function. J Perinat Neonatal Nurs 2014; 28: 150–4.
    1. Sulemanji M, Vakili K.. Neonatal renal physiology. Semin Pediatr Surg 2013; 22: 195–8.
    1. Benet LZ, Broccatelli F, Oprea TI.. BDDCS applied to over 900 drugs. AAPS J 2011; 13: 519–47.
    1. Ishizawa T, Hayashi M, Awazu S.. Paracellular and transcellular permeabilities of fosfomycin across small intestinal membrane of rat and rabbit by voltage-clamp method. J Pharmacobiodyn 1991; 14: 583–9.
    1. Ishizawa T, Sadahiro S, Hosoi K. et al. Mechanisms of intestinal absorption of the antibiotic, fosfomycin, in brush-border membrane vesicles in rabbits and humans. J Pharmacobiodyn 1992; 15: 481–9.
    1. Borsa F, Leroy A, Fillastre JP. et al. Comparative pharmacokinetics of tromethamine fosfomycin and calcium fosfomycin in young and elderly adults. Antimicrob Agents Chemother 1988; 32: 938–41.
    1. Cadorniga R, Diaz Fierros M, Olay T.. Pharmacokinetic study of fosfomycin and its bioavailability. Chemotherapy 1977; 23 Suppl 1: 159–74.
    1. Catassi C, Bonucci A, Coppa GV. et al. Intestinal permeability changes during the first month: effect of natural versus artificial feeding. J Pediatr Gastroenterol Nutr 1995; 21: 383–6.
    1. Saleem B, Okogbule-Wonodi AC, Fasano A. et al. Intestinal barrier maturation in very low birthweight infants: relationship to feeding and antibiotic exposure. J Pediatr 2017; 183: 31–6.
    1. Nau R, Sorgel F, Eiffert H.. Penetration of drugs through the blood-cerebrospinal fluid/blood-brain barrier for treatment of central nervous system infections. Clin Microbiol Rev 2010; 23: 858–83.
    1. Wang J, Kumar SS, Sherwin CM. et al. Renal clearance in newborns and infants: predictive performance of population-based modeling for drug development. Clin Pharmacol Ther 2019; 105: 1462–70.

Source: PubMed

3
Abonnere