Simultaneous pharmacokinetic/pharmacodynamic (PKPD) assessment of ampicillin and gentamicin in the treatment of neonatal sepsis

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

Objectives: This study aimed to simultaneously investigate the pharmacokinetics of ampicillin and gentamicin, currently the WHO standard of care for treating neonatal sepsis.

Methods: Pharmacokinetic data were collected in 59 neonates receiving ampicillin and gentamicin for suspected or proven sepsis in the NeoFosfo trial (NCT03453177). A panel of 23 clinical Escherichia coli isolates from neonates with sepsis, resistant to either ampicillin, gentamicin or both, were tested for susceptibility using chequerboards. Pharmacokinetic/pharmacodynamic (PKPD) modelling and simulations were used to compare single-agent (EUCAST MIC) and combination (chequerboard MIC) target attainment with standard dosing regimens.

Results: A model was established that simultaneously estimated parameters of a one-compartment ampicillin model and a two-compartment gentamicin model. A common clearance for both drugs was used (6.89 L/h/70 kg) relating to glomerular filtration (CLGFR), with an additional clearance term added for ampicillin (5.3 L/h/70 kg). Covariate modelling included a priori allometric weight and post-menstrual age scaling of clearance. Further covariate relationships on renal clearance were postnatal age and serum creatinine.Simulation-based PKPD assessments suggest good Gram-positive (MIC ≤ 0.25 mg/L) cover. However, less than one-quarter of neonates were predicted to receive efficacious coverage against Enterobacterales (MIC ≤ 2 mg/L). The benefit of the ampicillin/gentamicin combination was limited, with only 2/23 E. coli clinical strains showing FIC index < 0.5 (synergy) and most in the range 0.5-1 (suggesting additivity). Simulations showed that feasible dosing strategies would be insufficient to cover resistant strains.

Conclusions: PKPD simulations showed ampicillin and gentamicin combination therapy was insufficient to cover Enterobacterales, suggesting the need for alternative empirical treatment options for neonatal sepsis.

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

Figures

Figure 1.
Figure 1.
Observed plasma concentration versus time since last dose for ampicillin (left) and gentamicin (middle). Each line represents a single patient’s profile. Correlation of observed ampicillin and gentamicin concentrations is shown on the right.
Figure 2.
Figure 2.
VPC for joint ampicillin and gentamicin model. Black dots, observed values; solid line, median; dashed lines, 5th and 95th percentiles for the observed values; grey areas, 95% prediction interval of respective median and percentiles.
Figure 3.
Figure 3.
Target attainment as %fT>MIC against MICs for simulated ampicillin regimen. Solid line, population median; grey area, 90% prediction interval; dashed lines, EUCAST breakpoints for Enterobacterales and CoNS.
Figure 4.
Figure 4.
Target attainment as Cmax/MIC ratio against MICs for simulated gentamicin regimen. Solid line, population median; grey area, 90% prediction interval; dashed line, EUCAST breakpoint for Enterobacterales.
Figure 5.
Figure 5.
Resistance testing, FICI results and PTA for coverage of MICs for neonatal clinical specimens with the respective combination treatment. Light grey bars, gentamicin; dark grey bars, ampicillin.

References

    1. UNICEF, WHO, World Bank Group and United Nations. Levels and trends in child mortality. Report 2020. Estimates developed by the UN Inter-agency group for child mortality estimation. .
    1. Bielicki JA, Lundin R, Sharland M et al. Antibiotic resistance prevalence in routine bloodstream isolates from children’s hospitals varies substantially from adult surveillance data in Europe. Pediatr Infect Dis J 2015; 34: 734–41.
    1. Droz N, Hsia Y, Ellis S et al. Bacterial pathogens and resistance causing community acquired paediatric bloodstream infections in low- and middle-income countries: a systematic review and meta-analysis. Antimicrob Resist Infect Control 2019; 8: 207.
    1. Williams PCM, Isaacs D, Berkley JA. Antimicrobial resistance among children in sub-Saharan Africa. Lancet Infect Dis 2018; 18: e33–44.
    1. WHO. Pocket book of hospital care for children: guidelines for the management of common childhood illnesses: second edition. 2013. .
    1. Hsia Y, Lee BR, Versporten A et al. Use of the WHO Access, Watch, and Reserve classification to define patterns of hospital antibiotic use (AWaRe): an analysis of paediatric survey data from 56 countries. Lancet Glob Health 2019; 7: e861–71.
    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. Sharland M, Pulcini C, Harbarth S et al. Classifying antibiotics in the WHO Essential Medicines List for optimal use - be AWaRe. Lancet Infect Dis 2018; 18: 18–20.
    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. Soto E, Shoji S, Muto C et al. Population pharmacokinetics of ampicillin and sulbactam in patients with community-acquired pneumonia: evaluation of the impact of renal impairment. Br J Clin Pharmacol 2014; 77: 509–21.
    1. Nahata MC, Vashi VI, Swanson RN et al. Pharmacokinetics of ampicillin and sulbactam in pediatric patients. Antimicrob Agents Chemother 1999; 43: 1225–9.
    1. Kane Z, Gastine S, Obiero C et al. IV and oral fosfomycin pharmacokinetics in neonates with suspected clinical sepsis. J Antimicrob Chemother 2021; 76: 1855–64.
    1. Holford N, Heo YA, Anderson B. A pharmacokinetic standard for babies and adults. J Pharm Sci 2013; 102: 2941–52.
    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, 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. 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. Standing JF, Readman J, Hamawandi A et al. Identifying carbapenem and amikacin-sparing first-line combination therapy for neonatal sepsis/meningitis in centres with high prevalence of extended-spectrum beta-lactamase-producing Enterobacteriaceae. ECCMID 2019, Amsterdam, The Netherlands, 2019. Abstract O0827.
    1. Andrews JM. Determination of minimum inhibitory concentrations. J Antimicrob Chemother 2001; 48 Suppl 1: 5–16.
    1. Bonapace CR, Bosso JA, Friedrich LV et al. Comparison of methods of interpretation of checkerboard synergy testing. Diagn Microbiol Infect Dis 2002; 44: 363–6.
    1. Mir AR, Bashir Y, Dar FA et al. Identification of genes coding aminoglycoside modifying enzymes in E. coli of UTI patients in India. ScientificWorldJournal 2016; 2016: 1875865.
    1. Dallenne C, Da Costa A, Decre D et al. Development of a set of multiplex PCR assays for the detection of genes encoding important beta-lactamases in Enterobacteriaceae. J Antimicrob Chemother 2010; 65: 490–5.
    1. Gastine S, Hsia Y, Clements M et al. Variation in target attainment of beta-lactam antibiotic dosing between international pediatric formularies. Clin Pharmacol Ther 2021; 109: 958–70.
    1. EUCAST. Clinical breakpoint tables, Version 11. 2021. .
    1. Guilhaumou R, Benaboud S, Bennis Y et al. Optimization of the treatment with beta-lactam antibiotics in critically ill patients - guidelines from the French Society of Pharmacology and Therapeutics (Societe Francaise de Pharmacologie et Therapeutique-SFPT) and the French Society of Anaesthesia and Intensive Care Medicine (Societe Francaise d’Anesthesie et Reanimation-SFAR). Crit Care 2019; 23: 104.
    1. Moore RD, Lietman PS, Smith CR. Clinical response to aminoglycoside therapy: importance of the ratio of peak concentration to minimal inhibitory concentration. J Infect Dis 1987; 155: 93–9.
    1. Tumuhamye J, Sommerfelt H, Bwanga F et al. Neonatal sepsis at Mulago national referral hospital in Uganda: etiology, antimicrobial resistance, associated factors and case fatality risk. PLoS One 2020; 15: e0237085.
    1. Gezmu AM, Bulabula ANH, Dramowski A et al. Laboratory-confirmed bloodstream infections in two large neonatal units in sub-Saharan Africa. Int J Infect Dis 2021; 103: 201–7.
    1. Sands K, Carvalho MJ, Portal E et al. Characterization of antimicrobial-resistant Gram-negative bacteria that cause neonatal sepsis in seven low- and middle-income countries. Nat Microbiol 2021; 6: 512–23.
    1. Jatsho J, Nishizawa Y, Pelzom D et al. Clinical and bacteriological profile of neonatal sepsis: a prospective hospital-based study. Int J Pediatr 2020; 2020: 1835945.
    1. Foulds G. Pharmacokinetics of sulbactam/ampicillin in humans: a review. Rev Infect Dis 1986; 8 Suppl 5: S503–11.
    1. Sjovall J, Westerlund D, Alvan G. Renal excretion of intravenously infused amoxycillin and ampicillin. Br J Clin Pharmacol 1985; 19: 191–201.
    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.
    1. Lonsdale DO, Baker EH, Kipper K et al. Scaling beta-lactam antimicrobial pharmacokinetics from early life to old age. Br J Clin Pharmacol 2019; 85: 316–46.
    1. Wammanda RD, Adamu SA, Joshua HD et al. Implementation of the WHO guideline on treatment of young infants with signs of possible serious bacterial infection when hospital referral is not feasible in rural Zaria, Nigeria: challenges and solutions. PLoS One 2020; 15: e0228718.
    1. WHO. Guidelines for the prevention and control of carbapenem-resistant Enterobacteriaceae, Acinetobacter baumannii and Pseudomonas aeruginosa in health care facilities. 2017. .
    1. Tacconelli E, Cataldo MA, Dancer SJ et al. ESCMID guidelines for the management of the infection control measures to reduce transmission of multidrug-resistant Gram-negative bacteria in hospitalized patients. Clin Microbiol Infect 2014; 20 Suppl 1: 1–55.

Source: PubMed

3
Iratkozz fel