Skip to main content

Advertisement

Log in

Model-informed precision dosing of antimicrobial drugs in pediatrics: experiences from a pilot scale program

  • RESEARCH
  • Published:
European Journal of Pediatrics Aims and scope Submit manuscript

Abstract

Antibiotics are among the most utilized drugs in pediatrics. Nonetheless, there is a lack in pharmacokinetics information for this population, and dosing criteria may vary between healthcare centers. Physiological variability associated with maturation in pediatrics makes it challenging to reach a consensus on adequate dosing, which is further accentuated in more vulnerable groups, such as critically ill or oncology patients. Model-informed precision dosing is a useful practice that allows dose optimization and attainment of antibiotic-specific pharmacokinetic/pharmacodynamic targets. The aim of this study was to evaluate the needs of model-informed precision dosing of antibiotics in a pediatrics unit, at a pilot scale. Pediatric patients under antibiotic treatment were monitored with either a pharmacokinetic/pharmacodynamic optimized sampling scheme or through opportunistic sampling. Clindamycin, fluconazole, linezolid, meropenem, metronidazole, piperacillin, and vancomycin plasma concentrations were quantified through a liquid chromatography coupled to mass spectrometry method. Pharmacokinetic parameters were estimated using a Bayesian approach to verify pharmacokinetic/pharmacodynamic target attainment. A total of 23 pediatric patients aged 2 to 16 years were included, and 43 dosing regimens were evaluated; 27 (63%) of them required adjustments as follows: 14 patients were underdosed, 4 were overdosed, and 9 patients needed infusion rate adjustments. Infusion rate adjustments were mostly recommended for piperacillin and meropenem; daily doses were augmented for vancomycin and metronidazole, meanwhile linezolid was adjusted for under- and overdosing. Clindamycin and fluconazole regimens were not adjusted at all.

  Conclusion: Results showcase a lack of antibiotic pharmacokinetic/pharmacodynamic target attainment (particularly for linezolid, vancomycin, meropenem, and piperacillin), and the need for model-informed precision dosing in pediatrics. This study provides pharmacokinetic evidence which can further improve antibiotic dosing practices.

What is Known:

• Model-informed precision dosing is performed in pediatrics to optimize the treatment of antimicrobial drugs such as vancomycin and aminoglycosides, while its usefulness is debated for other groups (beta-lactams, macrolides, etc.).

What is New:

• Vulnerable pediatric subpopulations, such as critically ill or oncology patients, can benefit the most from model-informed precision dosing of antibiotics.

• Model-informed precision dosing of linezolid, meropenem, piperacillin, and vancomycin is particularly useful in pediatrics, and further research may improve dosing practices altogether.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1

Similar content being viewed by others

Data availability

The datasets generated and analysed during the current study are available from the corresponding author on request.

Abbreviations

α1-agp:

α1-Acid glycoprotein

AUC24h :

24-h area under de curve

Cmax :

Maximum plasma concentration

ECOFF:

Epidemiological cut-off

EUCAST:

European Committee on Antimicrobial Susceptibility Testing

fC50ss :

Mid free drug concentration at steady state

FDA:

Food and Drug Administration

fT > MIC:

Fraction of dosing interval free drug concentration is above the minimum inhibitory concentration (%)

HIV:

Human immunodeficiency virus

HPLC:

High performance liquid chromatography

LC:

Liquid chromatography

MIC:

Minimum inhibitory concentration

MIPD:

Model-informed precision dosing

MS/MS:

Tandem mass spectrometry

PD:

Pharmacodynamic/pharmacodynamics

PK:

Pharmacokinetic/pharmacokinetics

PopPK:

Population pharmacokinetics

QD:

Once daily

QID:

Four times a day

TID:

Three times a day

UPLC:

Ultra high performance liquid chromatography

References

  1. Dadgostar P (2019) Antimicrobial resistance: implications and costs. Infect Drug Resist 12:3903–10. Available from: https://www.dovepress.com/antimicrobial-resistance-implications-and-costs-peer-reviewed-fulltext-article-IDR

  2. de Moraes-Pinto MI, Ferrarini MAG (2020) Opportunistic infections in pediatrics: when to suspect and how to approach. Elsevier Editora Ltda [cited 2021 Feb 10]. Jornal de Pediatria 96:47–57. Available from: https://pubmed.ncbi.nlm.nih.gov/31790645/

  3. Downes KJ, Hahn A, Wiles J et al (2013) Dose optimisation of antibiotics in children: application of pharmacokinetics/pharmacodynamics in paediatrics. Int J Antimicrob Agents. Available from: https://doi.org/10.1016/j.ijantimicag.2013.11.006

  4. Le J, Bradley JS (2017) Optimizing antibiotic drug therapy in pediatrics: current state and future needs. J Clin Pharmacol 2018(58):S108–S122

    Google Scholar 

  5. Novak E, Allen PJ (2007) Prescribing medications in pediatrics: concerns regarding FDA approval and pharmacokinetics. Pediatr Nurs 33(1):64–70

    PubMed  Google Scholar 

  6. Food and Drug Administration (2022) Population Pharmacokinetics. Guidance for Industry. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/population-pharmacokinetics

  7. Rashed AN, Jackson C, Gastine S et al (2019) Pediatric pharmacokinetics of the antibiotics in the access and watch groups of the 2019 WHO model list of essential medicines for children: a systematic review. Expert Rev Clin Pharmacol 12(12):1099–106. Available from: https://doi.org/10.1080/17512433.2019.1693257

  8. Hartman SJF, Brüggemann RJ, Orriëns L et al (2020) Pharmacokinetics and target attainment of antibiotics in critically Ill children: a systematic review of current literature. Vol. 59, Clinical Pharmacokinetics. Springer International Publishing pp. 173–205. Available from: https://doi.org/10.1007/s40262-019-00813-w

  9. Reeves D, Lovering A, Thomson A (2016) Therapeutic drug monitoring in the past 40 years of the Journal of Antimicrobial Chemotherapy. J Antimicrob Chemother 71(12):3330–3332

    Article  CAS  PubMed  Google Scholar 

  10. Ates HC, Roberts JA, Lipman J et al (2020) On-site therapeutic drug monitoring. Trends Biotechnol. Available from: https://doi.org/10.1016/j.tibtech.2020.03.001

  11. Imani S, Buscher H, Marriott D et al (2017) Too much of a good thing: a retrospective study of β-lactam concentration-toxicity relationships. J Antimicrob Chemother [cited 2021 May 20] 72(10):2891–7. Available from: https://academic.oup.com/jac/article/72/10/2891/3952632

  12. Roger C, Louart B (2021) Beta-lactams toxicity in the intensive care unit: an underestimated collateral damage? Microorganism 9(7):1505. Available from: https://www.mdpi.com/2076-2607/9/7/1505/htm

  13. Schwartz GJ, Haycock GB, Edelmann CM et al (1976) A simple estimate of glomerular filtration rate in children derived from body length and plasma creatinine. Pediatrics 58(2):259–63. Available from: https://pubmed.ncbi.nlm.nih.gov/951142/

  14. Smith MJ, Gonzalez D, Goldman JL et al (2017) Pharmacokinetics of clindamycin in obese and nonobese children. Antimicrob Agents Chemother 61(4). Available from: https://doi.org/10.1128/AAC.02014-16

  15. Gonzalez D, Melloni C, Yogev R et al (2014) Use of opportunistic clinical data and a population pharmacokinetic model to support dosing of clindamycin for premature infants to adolescents. Clin Pharmacol Ther 96(4):429–37. Available from: https://doi.org/10.1038/clpt.2014.134

  16. Autmizguine J, Guptill JT, Cohen-Wolkowiez M et al (2014) Pharmacokinetics and pharmacodynamics of antifungals in children: clinical implications. Vol. 74, Drugs. Springer International Publishing, p. 891–909

  17. Van Der Elst KCM, Pereboom M, Van Den Heuvel ER et al (2014) Insufficient fluconazole exposure in pediatric cancer patients and the need for therapeutic drug monitoring in critically Ill children. Clin Infect Dis 59(11):1527–1533

    Article  PubMed  Google Scholar 

  18. Seay RE, Larson TA, Toscano JP et al (1995) Pharmacokinetics of fluconazole in immune-compromised children with leukemia or other hematologic disease. Pharmacother J Hum Pharmacol Drug Ther 15(1):52–58

    CAS  Google Scholar 

  19. Dou L, Meng D, Dong Y et al (2020) Dosage regimen and toxicity risk assessment of linezolid in sepsis patients. Int J Infect Dis 1(96):105–111

    Article  Google Scholar 

  20. Alsultan A (2019) Determining therapeutic trough ranges for linezolid. Saudi Pharm J 27(8):1061–1063

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Boak LM, Rayner CR, Grayson ML et al (2014) Clinical population pharmacokinetics and toxicodynamics of linezolid. Antimicrob Agents Chemother 58(4):2334–43. Available from: https://journals.asm.org/journal/aac

  22. Garcia-Prats AJ, Schaaf HS, Draper HR et al (2019) Pharmacokinetics, optimal dosing, and safety of linezolid in children with multidrug-resistant tuberculosis: combined data from two prospective observational studies. PLOS Med 16(4):e1002789. Available from: https://doi.org/10.1371/journal.pmed.1002789

  23. Dhaese S, Van Vooren S, Boelens J et al (2020) Therapeutic drug monitoring of β-lactam antibiotics in the ICUm, pp. 1155–64. Available from: https://doi.org/10.1080/14787210.2020.1788387

  24. Hassan HE, Ivaturi V, Gobburu J et al (2020) Dosage regimens for meropenem in children with pseudomonas infections do not meet serum concentration targets. Clin Transl Sci 13(2):301–8. Available from: https://doi.org/10.1111/cts.12710

  25. Child J, Chen X, Mistry RD et al (2019) Pharmacokinetic and pharmacodynamic properties of metronidazole in pediatric patients with acute appendicitis: a prospective study. J Pediatric Infect Dis Soc 8(4):297–302

    Article  PubMed  Google Scholar 

  26. Cohen-Wolkowiez M, Ouellet D, Smith PB et al (2012) Population pharmacokinetics of metronidazole evaluated using scavenged samples from preterm infants. Antimicrob Agents Chemother 56(4):1828–1837

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sprandel KA, Drusano GL, Hecht DW et al (2006) Population pharmacokinetic modeling and Monte Carlo simulation of varying doses of intravenous metronidazole. Diagn Microbiol Infect Dis 55(4):303–309

    Article  CAS  PubMed  Google Scholar 

  28. Thibault C, Lavigne J, Litalien C et al (2019) Population pharmacokinetics and safety of piperacillin-tazobactam extended infusions in infants and children. Antimicrob Agents Chemother 63(11):1-12. Available from: https://journals.asm.org/doi/10.1128/AAC.01260-19

  29. Béranger A, Benaboud S, Urien S et al (2018) Piperacillin population pharmacokinetics and dosing regimen optimization in critically Ill children with normal and augmented renal clearance. Clin Pharmacokinet 58(2):223–33. Available from: https://doi.org/10.1007/s40262-018-0682-1

  30. Thorsted A, Kristoffersson AN, Maarbjerg SF et al (2019) Population pharmacokinetics of piperacillin in febrile children receiving cancer chemotherapy: the impact of body weight and target on an optimal dosing regimen. J Antimicrob Chemother 74(10):2984–2993

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Zhang T, Cheng H, Pan Z et al (2020) Desired vancomycin trough concentration to achieve an AUC0–24/MIC ≥400 in Chinese children with complicated infectious diseases. Basic Clin Pharmacol Toxicol 126(1):75–85. Available from: https://doi.org/10.1111/bcpt.13303

  32. Le J, Ny P, Capparelli E et al (2015) Pharmacodynamic characteristics of nephrotoxicity associated with vancomycin use in children. J Pediatric Infect Dis Soc 4(4):e109–16. Available from: https://academic.oup.com/jpids/article/4/4/e109/2580148

  33. Smit C, Goulooze SC, Brüggemann RJM et al (2021) Dosing recommendations for vancomycin in children and adolescents with varying levels of obesity and renal dysfunction: a population pharmacokinetic study in 1892 children aged 1–18 years. AAPS J 23(3):1–10. Available from: https://doi.org/10.1208/s12248-021-00577-x

  34. Alsultan A, Abouelkheir M, Alqahtani S et al (2018) Optimizing vancomycin monitoring in pediatric patients. Pediatr Infect Dis J 37(9):880–5. Available from: https://journals.lww.com/pidj/Fulltext/2018/09000/Optimizing_Vancomycin_Monitoring_in_Pediatric.8.aspx

  35. Santos Buelga D, Del Mar Fernandez De Gatta M, Herrera EV et al (2005) Population pharmacokinetic analysis of vancomycin in patients with hematological malignancies. Antimicrob Agents Chemother 49(12):4934–41. Available from: https://journals.asm.org/journal/aac

  36. Food and Drug Administration (2018) Bioanalytical Method Validation. Guidance for Industry. Available from: https://www.fda.gov/regulatory-information/search-fda-guidance-documents/bioanalytical-method-validation-guidance-industry

  37. Patel BM, Paratz J, See NC et al (2012) Therapeutic drug monitoring of beta-lactam antibiotics in burns patients-a one-year prospective study. Ther Drug Monit 34(2):160–4. Available from: https://journals.lww.com/drug-monitoring/Fulltext/2012/04000/Therapeutic_Drug_Monitoring_of_Beta_Lactam.8.aspx

  38. Maharaj AR, Gonzalez D, Cohen-Wolkowiez M et al (2018) Improving pediatric protein binding estimates: an evaluation of α1-acid glycoprotein maturation in healthy and infected subjects HHS Public Access. Clin Pharmacokinet 57(5):577–589

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Kays MB, White RL, Gatti G et al (1992) Ex vivo protein binding of clindamycin in sera with normal and elevated α1-acid glycoprotein concentrations. Pharmacother J Hum Pharmacol Drug Ther 12(1):50–5. Available from: https://doi.org/10.1002/j.1875-9114.1992.tb02671.x

  40. EUCAST. MIC Breakpoints. [cited 14 Apr 2021]. Available from: https://mic.eucast.org

  41. Burns B, Hartenstein M, Lin A et al (2019) Optimizing time to antibiotic administration in children with possible febrile neutropenia through quality improvement methodologies. Pediatr Qual Saf 4(6):e236

    Article  PubMed  PubMed Central  Google Scholar 

  42. White L, Ybarra M (2017) Neutropenic fever. Hematol Oncol Clin North Am 31(6):981–93. Available from: https://doi.org/10.1016/j.hoc.2017.08.004

  43. Cohen-Wolkowiez M, Benjamin DK (2014) Fluconazole therapeutic drug monitoring in children with cancer: not today. Vol. 59, Clinical Infectious Diseases. Oxford University Press; p. 1534–6

  44. Stockmann C, Constance JE, Roberts JK et al (2014) Pharmacokinetics and pharmacodynamics of antifungals in children and their clinical implications. Vol. 53, Clinical Pharmacokinetics. Springer International Publishing; p. 429–54

  45. Costenaro P, Minotti C, Cuppini E et al (2020) Optimizing antibiotic treatment strategies for neonates and children: does implementing extended or prolonged infusion provide any advantage? Vol. 9, Antibiotics. MDPI AG; p. 1–20

  46. Chongcharoenyanon T, Wacharachaisurapol N, Anugulruengkitt S et al (2021) Comparison of piperacillin plasma concentrations in a prospective randomised trial of extended infusion versus intermittent bolus of piperacillin/tazobactam in paediatric patients. Int J Infect Dis 1(108):102–108

    Article  Google Scholar 

  47. Steffens NA, Zimmermann ES, Nichelle SM et al (2021) Meropenem use and therapeutic drug monitoring in clinical practice: a literature review. J Clin Pharm Ther 6(3):610–621. Available from: https://doi.org/10.1111/jcpt.13369

    Article  CAS  Google Scholar 

  48. Germovsek E, Lutsar I, Kipper K et al (2018) Plasma and CSF pharmacokinetics of meropenem in neonates and young infants: results from the NeoMero studies. J Antimicrob Chemother 73(7):1908–16. Available from: https://academic.oup.com/jac/article/73/7/1908/4978317

  49. Huttner A, Harbarth S, Hope WW et al (2015) Therapeutic drug monitoring of the β-lactam antibiotics: what is the evidence and which patients should we be using it for? J Antimicrob Chemother 70(12):3178–83. Available from: https://academic.oup.com/jac/article/70/12/3178/2363621

  50. Scharf C, Paal M, Schroeder I et al (2020) Therapeutic drug monitoring of meropenem and piperacillin in critical illness—experience and recommendations from one year in routine clinical practice. Antibiotics 9(3):131. Available from: www.mdpi.com/journal/antibiotics

  51. Guilhaumou R, Marsot A, Dupouey J et al (2016) Pediatric patients with solid or hematological tumor disease: vancomycin population pharmacokinetics and dosage optimization. Ther Drug Monit 38(5):559–66. Available from: https://pubmed.ncbi.nlm.nih.gov/27631462/

  52. Zhao W, Zhang D, Fakhoury M et al (2014) Population pharmacokinetics and dosing optimization of vancomycin in children with malignant hematological disease. Antimicrob Agents Chemother 58(6):3191–9. Available from: https://journals.asm.org/journal/aac

  53. Marsot A, Gallais F, Galambrun C et al (2018) Vancomycin in pediatric patients with solid or hematological malignant disease: predictive performance of a population pharmacokinetic model and new optimized dosing regimens. Paediatr Drug 20(4):375–81. Available from: https://pubmed.ncbi.nlm.nih.gov/29736878/

Download references

Acknowledgements

The authors would like to thank the residents and nurses of the Pediatrics division at Hospital Central “Dr Ignacio Morones Prieto” for their support during patient recruitment, as well as the patients and their family members who agreed to participate in the study.

Funding

The present study was supported by Mexico’s Office of Public Education through the government program: “Programa para el Desarrollo Profesional Docente, PRODEP” (511-6/2020-8585). A master’s fellowship was granted to Rodrigo Velarde-Salcedo by the Technological Research Council of Science (CONACyT) from Mexico (Grant 782280).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the study conception and design. Material preparation, data collection, and analysis were performed by Rodrigo Velarde-Salcedo, Susanna Edith Medellín-Garibay, Silvia Romano-Moreno, Rosa del Carmen Milán Segovia, Luis Fernando Pérez-González, Francisco Javier Arriaga García, and Ana Socorro Rodríguez-Báez. The first draft of the manuscript was written by Rodrigo Velarde-Salcedo, and all authors commented on previous versions of the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Susanna Edith Medellín-Garibay.

Ethics declarations

Ethics approval

This study was carried out in line with ethical principles of the Declaration of Helsinki. Approval was granted by the Research and Ethics Committee of Hospital Central “Dr. Ignacio Morones Prieto, SLP, México (Registration number 69-21; August 26th, 2021).

Consent to participate

Written informed consent was obtained from patients’ parents, and informed assent was obtained from patients over 12 years of age.

Competing interests

The authors declare no competing interests.

Additional information

Communicated by Peter de Winter

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Velarde-Salcedo, R., Pérez-González, L.F., Rodríguez-Báez, A.S. et al. Model-informed precision dosing of antimicrobial drugs in pediatrics: experiences from a pilot scale program. Eur J Pediatr 182, 4143–4152 (2023). https://doi.org/10.1007/s00431-023-05103-z

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00431-023-05103-z

Keywords

Navigation