323
Views
10
CrossRef citations to date
0
Altmetric
Review

Neonatal medicines research: challenges and opportunities

, , & , MD PhD

Bibliography

  • Kearns GL, Abdel-Rahman SM, Alander SW, et al. Developmental pharmacology—drug disposition, action, and therapy in infants and children. N Engl J Med 2003;349:1157–67
  • Allegaert K, Verbesselt R, Naulaers G, et al. Developmental pharmacology: neonates are not just small adults…. Acta Clin Belg 2008;63:16–24
  • Turner MA. Neonatal drug development. Early Hum Dev 2011;87:763–8
  • Stiers JL, Ward RM. Newborns, one of the last therapeutic orphans to be adopted. JAMA Pediatr 2014;168:106–8
  • Laughon MM, Avant D, Tripathi N, et al. Drug labeling and exposure in neonates. JAMA Pediatr 2014;168:130–6
  • Turner MA, Catapano M, Hirschfeld S, et al. Paediatric drug development: the impact of evolving regulations. Adv Drug Deliv Rev 2014;73:2–13
  • Allegaert K. Tailored tools to improve pharmacotherapy in infants. Expert Opin Drug Metab Toxicol 2014;10:1069–78
  • Anderson BJ, van Lingen RA, Hansen TG, et al. Acetaminophen developmental pharmacokinetics in premature neonates and infants: a pooled population analysis. Anesthesiology 2002;96:1336–45
  • Choonara I. Percutaneous drug absorption and administration. Arch Dis Child 1994;71:F73–4
  • de Cock RF, Allegaert K, Sherwin CM, et al. A neonatal amikacin covariate model can be used to predict ontogeny of other drugs eliminated through glomerular filtration in neonates. Pharm Res 2014;31:754–67
  • Allegaert K, Langhendries JP, van den Anker JN. Educational paper: do we need neonatal clinical pharmacologists? Eur J Pediatr 2013;172:429–35
  • Roberts JA, Pea F, Lipman J. The clinical relevance of plasma protein binding changes. Clin Pharmacokinet 2013;52:1–8
  • Smits A, Kulo A, Verbesselt R, et al. Cefazolin plasma protein binding and its covariates in neonates. Eur J Clin Microbiol Infect Dis 2012;31:3359–65
  • Smits A, Roberts JA, Vella-Brincat JW, et al. Cefazolin plasma protein binding in different human populations: more than cefazolin-albumin interaction. Int J Antimicrob Agents 2014;43:199–200
  • Hines RN. Developmental expression of drug metabolizing enzymes: impact on disposition in neonates and young children. Int J Pharm 2013;452:3–7
  • de Wildt SN. Profound changes in drug metabolism enzymes and possible effects on drug therapy in neonates and children. Expert Opin Drug Metab Toxicol 2011;7:935–48
  • 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
  • De Cock RF, Allegaert K, Schreuder MF, et al. Maturation of the glomerular filtration rate in neonates, as reflected by amikacin clearance. Clin Pharmacokinet 2012;51:105–17
  • Smits A, Kulo A, de Hoon JN, et al. Pharmacokinetics of drugs in neonates: pattern recognition beyond compound specific observations. Curr Pharm Des 2012;18:3119–46
  • Schreuder MF, Bueters RR, Allegaert K. The interplay between drugs and the kidney in premature neonates. Pediatr Nephrol 2014;29:2083–91
  • Mulla H. Understanding developmental pharmacodynamics: importance for drug development and clinical practice. Paediatr Drugs 2010;12:223–33
  • Stephenson T. How children’s responses to drugs differ from adults. Br J Clin Pharmacol 2005;59:670–3
  • Bajcetic M, Uzelac TV, Jovanovic I. Heart failure pharmacotherapy: differences between adult and paediatric patients. Curr Med Chem 2014;21:3108–20
  • McWilliam SJ, Antoine DJ, Sabbisetti V, et al. Mechanism-based urinary biomarkers to identify the potential for aminoglycoside-Induced nephrotoxicity in premature neonates: a proof-of-concept study. PLoS ONE 2012;7:e43809
  • Kent A, Turner MA, Sharland M, et al. Aminoglycoside toxicity in neonates: something to worry about? Expert Rev Anti Infect Ther 2014;12:319–31
  • Allegaert K, Pauwels S, Smits A, et al. Enzymatic isotope dilution mass spectrometry (IDMS) traceable serum creatinine is preferable over Jaffe in neonates and young infants. Clin Chem Lab Med 2014;52:e107–9
  • Allegaert K, Mekahli D, van den Anker J. Cystatin C in newborns: a promising renal biomarker in search for standardization and validation. J Matern Fetal Neonatal Med 2014;5:1–6
  • Iacovidou N, Syggelou A, Chalkias A, et al. Metabolomics applied in neonatology. Bioanalysis 2014;6:403–10
  • Whittaker A, Currie AE, Turner MA, et al. Toxic additives in medication for preterm infants. Arch Dis Child Fetal Neonatal Ed 2009;94:F236–40
  • Standing JF, Tuleu C. Paediatric formulations–Getting to the heart of the problem. Int J Pharm 2005;300:56–66
  • Lass J, Naelapää K, Shah U, et al. Hospitalised neonates in Estonia commonly receive potentially harmful excipients. BMC Pediatr 2012;12:136
  • Turner MA, Duncan J, Shah U, et al. European Study of Neonatal Exposure to Excipients: an update. Int J Pharm 2013;457:357–8
  • . Salunke S, Brandys B, Giacoia G, et al.The STEP. Safety and Toxicity of Excipients for Paediatrics) database: part 2 – the pilot version. Int J Pharm 2013;457:310–22
  • De Cock RF, Knibbe CA, Kulo A, et al. Developmental pharmacokinetics of propylene glycol in preterm and term neonates. Br J Clin Pharmacol 2013;75:162–71
  • Mulla H, Yakkundi S, McElnay J, et al. An observational study of blood concentrations and kinetics of methyl- and propyl-parabens in neonates. Pharm Res 2015;32(3):1084–93
  • Allegaert K, Vanhaesebrouck S, Kulo A, et al. Prospective assessment of short-term propylene glycol tolerance in neonates. Arch Dis Child 2010;95:1054–8
  • Allegaert K, Anderson BJ, Vrancken M, et al. Impact of a paediatric vial on the magnitude of systematic medication errors in neonates. Paed Perinatal Drug Ther 2006;7:59–63
  • EMA. Guideline on the investigation of medicinal products in the term and preterm neonate. 2009. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2009/09/WC500003750.pdf [last accessed 28 January 2015]
  • Patel P, Mulla H, Kairamkonda V, et al. Dried blood spot assay and sparse sampling: a practical approach to estimating pharmacokinetic parameters of caffeine in preterm infants. Br J Clin Pharmacol 2013;75:805–13
  • Suyagh M, Collier PS, Millerschip JS, et al. Metronidazole population pharmacokinetics in preterm neonates using dried blood-spot sampling. Pediatrics 2011;127:e367–74
  • Villanelli F, Giocaliere E, Malvagia S, et al. Dried blood spots for the quantification of phenytoin using Liquid Chromatography-Mass Spectrometry. Clin Chim Acta 2014;440C:31–5
  • Suyagh M, Hawwa AF, Collier PS, et al. Potassium canrenoate treatment in paediatric patients: a population pharmacokinetic study using novel dried blood spot sampling. J Hypertens 2013;31:1901–8
  • Della Bona ML, Malvagia S, Villanelli F, et al. A rapid liquid chromatography tandem mass spectrometry-based method for measuring propranolol on dried blood spots. J Pharm Biomed Anal 2013;78-79:34–8
  • Patel P, Tanna S, Mulla H, et al. Dexmethasone quantification in dried blood spot samples using LC-MS: the potential for application to neonatal pharmacokinetic studies. J Chromatogr B Analyt Technol Biomed Life Sci 2010;878:3277–82
  • Bakhireva LN, Leeman L, Savich RD, et al. The validity of phosphatidylethanol in dried blood spots of newborns for the identification of prenatal alcohol exposure. Alcohol Clin Exp Res 2014;38:1078–85
  • Ma WL, Gao C, Bell EM, et al. Analysis of polychlorinated biphenyls and organochlorine pesticides in archived dried blood spots and its application to track temporal trends of environmental chemicals in newborns. Environ Res 2014;133:204–10
  • Yakkundi S, McElnay J, Millership J, et al. Use of dried blood spots to study excipient kinetics in neonates. Bioanalysis 2011;3:2691–3
  • Vu DH, Koster RA, Bolhuis MS, et al. Simultaneous determination of rifampicin, clarithromycin and their metabolites in dried blood spots using LC-MS/MS. Talanta 2014;121:9–17
  • Vu DH, Bolhuis MS, Koster RA, et al. Dried blood spot analysis for therapeutic drug monitoring of linezolid in patients with multidrug-resistant tuberculosis. Antimicrob Agents Chemother 2012;56:5758–63
  • La Marca G, Malvagia S, Filippi L, et al. Rapid assay of topiramate in dried blood spots by a new liquid chromatography-tandem mass spectrometric method. J Pharm Biomed Anal 2008;48:1392–6
  • Beaumont C, Young GC, Cavalier T, et al. Human absorption, distribution, metabolism and excretion properties of drug molecules: a plethora of approaches. Br J Clin Pharmacol 2014;78:1185–200
  • Vuong LT, Blood AB, Vogel JS, et al. Applications of accelerator MS in pediatric drug evaluation. Bioanalysis 2012;4:1871–82
  • Garner RC, Park BK, French NS, et al. Observational infant exploratory [14C] paracetamol pharmacokinetic microdose/therapeutic dose study with accelerator mass spectrometry bioanalysis. Br J Clin Pharmacol 2015. [Epub ahead of print]
  • Gordi T, Baillie R, Vuong Le T, et al. Pharmacokinetic analysis of 14C-ursodiol in newborn infants using accelerator mass spectrometry. J Clin Pharmacol 2014;54:1031–7
  • Mooij MG, van Duijn E, Knibbe CA, et al. Pediatric microdose study of [14C] paracetamol to study drug metabolism using accelerated mass spectrometry: proof of concept. Clin Pharmacokinet 2014;53:1045–51
  • Schierbeek H, van den Akker CH, Fay LB, et al. High-precision mass spectrometric analysis using stable isotopes in studies of children. Mass Spectrom Rev 2012;31:312–30
  • Himebauch AS, Zuppa A. Methods for pharmacokinetic analysis in young children. Expert Opin Drug Metab Toxicol 2014;10:497–509
  • Laughon MM, Benjamin DKJr, Capparelli EV, et al. Innovative clinical trial design for pediatric therapeutics. Expert Rev Clin Pharmacol 2011;4:643–52
  • De Cock RF, Piana C, Krekels EH, et al. The role of population PK-PD modelling in paediatric clinical research. Eur J Clin Pharmacol 2011;67(Suppl 1):5–16
  • Barrett JS, Della Casa Alberighi O, Läer S, et al. Physiologically based pharmacokinetic (PBPK) modeling in children. Clin Pharmacol Ther 2012;92:40–9
  • Anderson BJ, Allegaert K, Holford NH. Population clinical pharmacology of children: general principles. Eur J Pediatr 2006;165:741–6
  • Maharaj AR, Edginton AN. Physiologically based pharmacokinetic modeling and simulation in pediatric drug development. CPT Pharmacometrics Syst Pharmacol 2014;3:e150
  • Abduljalil K, Jamei M, Rostami-Hodjegan A, et al. Changes in individual drug-independent system parameters during virtual paediatric pharmacokinetic trials: introducing time-varying physiology into a paediatric PBPK model. AAPS J 2014;16:568–76
  • Manolis E, Osman TE, Herold R, et al. Role of modeling and simulation in pediatric investigation plans. Paediatr Anaesth 2011;21:214–21
  • Zisowsky J, Krause A, Dingemanse J. Drug development for pediatric populations: regulatory aspects. Pharmaceutics 2010;2:364–88
  • European Medicines Agency. European Medicines Agency modelling and simulation working group plan. 2014. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Other/2014/06/WC500167816.pdf [last accessed 15 February 2015]
  • Khalil F, Läer S. Physiologically based pharmacokinetic models in the prediction of oral drug exposure over the entire pediatric age range-sotalol as a model drug. AAPS J 2014;16:226–39
  • Yu G, Zheng QS, Li GF. Similarities and differences in gastrointestinal physiology between neonates and adults: a physiologically based pharmacokinetic modeling perspective. AAPS J 2014;16:1162–6
  • Zane RN, Thakker DR. A physiologically based pharmacokinetic model for voriconazole disposition predicts intestinal first-pass metabolism in children. Clin Pharmacokinetic 2014;53:1171–82
  • Björkman S. Prediction of drug disposition in infants and children by means of physiologically based pharmacokinetic (PBPK) modelling: theophylline and midazolam as model drugs. Br J Clin Pharmacol 2005;59:691–704
  • Ginsberg G, Hattis D, Russ A, et al. Physiologically based pharmacokinetics- (PBPK) modeling of caffeine and theophylline in neonates and adults: implications for assessing children’s risks from environmental agents. J Toxicol Environ Health A 2004;67:297–329
  • Johnson TN, Rostami-Hodjegan A, Tucker GT. Prediction of the clearance of eleven drugs and associated variability in neonates, infants and children. Clin Pharmacokinet 2006;45:931–56
  • Claassen K, Willmann S, Thelen K, et al. Physiology-based simulations of amikacin pharmacokinetics in preterm neonates. Annual meeting of the Population Approach Group in Europe (PAGE). Berlin, 2010; abstract 1859
  • Claassen K, Willmann S, Thelen K, et al. Evaluation of a PBPK model for preterm neonates by predicting paracetamol pharmacokinetics. Annual meeting of the Population Approach Group in Europe (PAGE). Athens, 2011; abstract 1978
  • Krekels EH, Neely M, Panoilia E, et al. From pediatric covariate model to semiphysiological function for maturation: part I-extrapolation of a covariate model from morphine to zidovudine. CPT Pharmacometrics Syst Pharmacol 2012;1:e9
  • European Medicines Agency. 5-year Report to the European Commission, General report on the experience acquired as a result of the application of the Paediatric Regulation. 2012. EMA/428172/2012 Available from: http://ec.europa.eu/health/files/paediatrics/2012-09_pediatric_report-annex1-2_en.pdf [last accessed 28 January 2015]
  • FDA. Neonatology Subcommittee of the Pediatric Advisory Committee Meeting. 15 March 2013. Available from: http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/PediatricAdvisoryCommittee/UCM342926.pdf [last accessed 28 January 2015]
  • Olski TM, Lampus SF, Gjerarducci G, Saint Raymond A. Three years of paediatric regulation in the European Union. Eur J Clin Pharmacol 2011;67:245–52
  • Carleer J, Karres J. Juvenile animal studies and paediatric drug development: a European regulator experience. Birth Defects Res B Dev Reprod Toxicol 2011;92:254–60
  • Bax R, Tomasi P. Neonatal pharmacotherapy: legal and regulatory issues: 108-123. In: Mimouni FB, van den Anker JN, Editors Pediatric and Adolescent Medicine, volume 18: Neonatal pharmacology and nutrition update. Karger; Basel: 2015
  • Turner MA. Clinical trials of medicines in neonates: the influence of ethical and practical issues on design and conduct. Br J Clin Pharmacol 2015;79(3):370–8
  • Sosenko IR, Bancalari E. NO for preterm infants at risk for bronchopulmonary dysplasia. Lancet 2010;376:308–10
  • Ruggieri L, Giannuzzi V, Baiardi P, et al. Successful private-public funding of paediatric medicines research: lessons from the EU programme to fund research into off-patent medicines. Eur J Pediatr 2014;174(4):481–91
  • Jacqz-Aigrain E. Drug policy in Europe Research and funding in neonates: current challenges, future perspectives, new opportunities. Early Hum Dev 2011;87(Suppl 1):S27–30
  • Reed MD. Reversing the myths obstructing the determination of optimal age- and disease-based drug dosing in pediatrics. J Pediatr Pharmacol Ther 2011;16:4–13
  • Available from: http://www.fda.gov/AdvisoryCommittees/Calendar/ucm341119.htm [last accessed 28 January 2015]
  • De Boeck K, Bulteel V, Tiddens H, et al. Guideline on the design and conduct of cystic fibrosis clinical trials: the European Cystic Fibrosis Society-Clinical Trials Network (ECFS-CTN). J Cyst Fibros 2011;10(Suppl 2):S67–74
  • Ruperto N, Martini A. Networking in paediatrics: the example of Paediatric Rheumatology International Trials Organisation (PRINTO). Arch Dis Child 2011;96:596–601
  • Rose AC, van’t Hoff W, Beresford MW, et al. NIHR Medicines for Children Research Network: improving children’s health through clinical research. Expert Rev Clin Pharmacol 2013;6:581–7
  • Van’t Hoff W, Offringa M. StaR Child Health: developing evidence-based guidance for the design, conduct and reporting of paediatric trials. Arch Dis Child 2015;100(2):189–92
  • European Medicines Agency. Concept paper on extrapolation of efficacy and safety in medicine development Draft. 2012. Available from: http://www.ema.europa.eu/docs/en_GB/document_library/Scientific_guideline/2012/06/WC500129285.pdf [Last accessed January 2015]
  • Cools F, Askie LM, Offringa M, et al. Elective high-frequency oscillatory versus conventional ventilation in preterm infants: a systematic review and meta-analysis of individual patients’ data. Lancet 2010;375:2082–91
  • Askie LM, Ballard RA, Cutter GR, et al. Inhaled nitric oxide in preterm infants: an individual-patient data meta-analysis of randomized trials. Pediatrics 2011;128:729–39
  • Zhao W, Kaguelidou F, Biran V, et al. External evaluation of population pharmacokinetic models of vancomycin in neonates: the transferability of published models to different clinical settings. Br J Clin Pharmacol 2013;75:1068–80
  • Desfrere L, Zohar S, Morville P, et al. Dose-finding study of ibuprofen in patent ductus arteriosus using the continual reassessment method. J Clin Pharm Ther 2005;30:121–32
  • Thall PF, Nguyen HQ, Zohar S, et al. Optimizing sedative dose in preterm infants undergoing treatment for respiratory distress syndrome. J Am Stat Assoc 2014;109:931–43

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.