Skip to main content
Erschienen in: Clinical Pharmacokinetics 5/2021

Open Access 13.03.2021 | Letter to the Editor

Comment on: "Preterm Physiologically Based Pharmacokinetic Model, Part I and Part II”

verfasst von: Swantje Völler, Robert B. Flint, Sinno H. P. Simons, Catherijne A. J. Knibbe

Erschienen in: Clinical Pharmacokinetics | Ausgabe 5/2021

download
DOWNLOAD
print
DRUCKEN
insite
SUCHEN
Hinweise
The original online version of this article was revised due to a retrospective Open Access cancellation order.
An author's reply to this comment is available online at https://​doi.​org/​10.​1007/​s40262-021-00995-2.
A correction to this article is available online at https://​doi.​org/​10.​1007/​s40262-022-01117-2.
Dear Editor
We read with interest the two recent articles in Clinical Pharmacokinetcs by Abduljalil et al. describing a preterm physiologically based pharmacokinetic model and evaluate its performance for eight hepatically and renally cleared drugs [1, 2].
The lack of adequate dosing regimens mostly due to off-label use and the high interindividual variability in both pharmacokinetics (PK) and pharmacodynamics (PD) make these infants more prone to suffer from overexposure, adverse effects and treatment failure [3]. To solve these issues, most recent papers focus on using state-of-the-art population modelling and simulation approaches to describe the pharmacokinetics of one individual drug in plasma. However, we need to reach a better understanding of the rapid physiological changes during this early phase of life, as conducting studies for each drug puts a high additional burden on both patients and their parents. Physiology based models can be a tool that can help us understand the underlying processes and to choose the dose for future First-in-Neonate  trials.
We therefore highly value the efforts by Abduljalil et al. to get one step closer to being able to predict the PK in preterm neonates; however, we believe this can only be a first stepping stone towards better PK predictions, and some points deserve further attention before the model can be used in practice.
Abduljalil and colleagues used postmenstrual age (PMA) to describe all relevant maturation processes in preterm neonates. While this might seem a smart approach as one can describe the data of all preterm neonates with one function, it has one major drawback—it does not take into account the effect of birth on maturational processes. Birth leads to complex physiological changes in multiple organ systems, independent from gestational age, including the cardiovascular, respiratory, hepatic and renal systems, all of which impact the maturation of the PK of drugs [46]. The effect of birth has been repeatedly shown in population PK models for both hepatically cleared [79] and renally cleared drugs [10, 11]. In those studies, the combination of gestational age or birthweight with postnatal age, and not PMA, were the best predictors of PK in a large heterogeneous group of preterm neonates. When it comes to renal function, the impact of birth on the maturation of glomerular filtration rate has recently been clearly shown by Salem et al. [12].
We therefore believe that, for a physiologically based PK (PBPK) model, it is crucial to take into account the different maturation rates for different gestational ages. To this end, PMA does not appear to be the best predictor of maturation for the population of preterm neonates with varying PMA of between 28 and 44 weeks.
We observed that due to the lack of available data in preterms, the developed  model was based on many assumptions. Although we fully understand that very limited and sometimes conflicting information is available, these assumptions might heavily influence the predictions of the model. For example, preterm neonates were assumed to have the same tissue blood flow to organs as full-term neonates and cardiac output was calculated based on bodyweight only, which does not seem very likely. In addition, a significantly higher chance of a patent ductus arteriosus (PDA) in children born at lower gestational ages [6] makes it likely that cardiac output also differs in children with and without PDA. Due to this, blood flow to organs might also vary.
Another drawback is the way in which cytochrome P450 (CYP) metabolism has been implemented into the model. We agree with the authors that much about the ontogeny still remains unknown; however, not implementing CYP3A7 metabolism, which is the major route of hepatic elimination in neonates [13], into the model because of the lack of data on its ontogeny can be a major pitfall. Although Abduljalil et al. acknowledge this problem and state that ontogeny functions can be manually adjusted, their second paper shows the major drawback of their approach. The PK of the four hepatically cleared drugs studied (alfentanil, midazolam, caffeine, ibuprofen) could not be adequately predicted, while the fit for the two renally cleared drugs (gentamicin, vancomycin) looked satisfactory. The authors extensively discuss the drawbacks of each of the clinical studies used, but are, in our opinion, less critical towards the drawbacks of their own model design, most importantly the lack of CYP3A7 metabolism in the model.
In their model, Abduljalil et al. did not consider extremely premature neonates with gestational ages below 28 weeks, however, these neonates have the highest need for intensive care and pharmacological treatment. Furthermore, these very young infants are so small that repetitive blood sampling is difficult, and they would benefit most from insights gained from a PBPK model. Unfortunately, the authors do not discuss why they excluded this important subpopulation, but we believe that possible explanations could be the lack of physiological data in this population and/or the poor performance of their model in this age group. Another population that has not been taken into account in this model is infants who are small for gestational age. Due to intrauterine growth restrictions, these children may develop at a different pace compared with children who are appropriate for their gestational age. We believe that, for the future, it would be extremely valuable to also include these two subpopulations to cover the whole preterm population.
With this PBPK model, the authors have made a valuable step towards better prediction of the PK of drugs in preterm infants. However, potential users need to be very aware of its limitations and can only use the model to get a very rough idea of the potential concentrations in a subpopulation of preterm infants. Including preterm infants with a gestational age below 28 weeks and infants who are small for gestational age in the next version would already make the model applicable for the whole preterm population. After that, crucial next steps would be to first take the effect of birth into account in the maturation function, thereby recognizing the separate influence of intra- and extrauterine maturation, and then to obtain a better description of hepatic elimination.

Declarations

Conflict of interest

The authors declare that there is no conflict of interest.

Funding

The authors received no specific funding.
download
DOWNLOAD
print
DRUCKEN

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Abduljalil K, Pan X, Pansari A, Jamei M, Johnson TN. Preterm physiologically based pharmacokinetic model. Part II: applications of the model to predict drug pharmacokinetics in the preterm population. Clin Pharmacokinet. 2020;59(4):501–18.CrossRefPubMed Abduljalil K, Pan X, Pansari A, Jamei M, Johnson TN. Preterm physiologically based pharmacokinetic model. Part II: applications of the model to predict drug pharmacokinetics in the preterm population. Clin Pharmacokinet. 2020;59(4):501–18.CrossRefPubMed
2.
Zurück zum Zitat Abduljalil K, Pan X, Pansari A, Jamei M, Johnson TN. A Preterm physiologically based pharmacokinetic model. Part I: physiological parameters and model building. Clin Pharmacokinet. 2020;59(4):485–500.CrossRefPubMed Abduljalil K, Pan X, Pansari A, Jamei M, Johnson TN. A Preterm physiologically based pharmacokinetic model. Part I: physiological parameters and model building. Clin Pharmacokinet. 2020;59(4):485–500.CrossRefPubMed
3.
Zurück zum Zitat Allegaert K, Van Den Anker JN. Adverse drug reactions in neonates and infants: a population-tailored approach is needed. Br J Clin Pharmacol. 2015;80(4):788–95.CrossRefPubMedPubMedCentral Allegaert K, Van Den Anker JN. Adverse drug reactions in neonates and infants: a population-tailored approach is needed. Br J Clin Pharmacol. 2015;80(4):788–95.CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Bartelink IH, Rademaker CMA, Schobben AFAM, Van Den Anker JN. Guidelines on paediatric dosing on the basis of developmental physiology and pharmacokinetic considerations. Clin Pharmacokinet. 2006;45(11):1077–97.CrossRefPubMed Bartelink IH, Rademaker CMA, Schobben AFAM, Van Den Anker JN. Guidelines on paediatric dosing on the basis of developmental physiology and pharmacokinetic considerations. Clin Pharmacokinet. 2006;45(11):1077–97.CrossRefPubMed
5.
Zurück zum Zitat Vrancken SL, van Heijst AF, de Boode WP. Neonatal hemodynamics: from developmental physiology to comprehensive monitoring. Front Pediatr. 2018;6:87.CrossRefPubMedPubMedCentral Vrancken SL, van Heijst AF, de Boode WP. Neonatal hemodynamics: from developmental physiology to comprehensive monitoring. Front Pediatr. 2018;6:87.CrossRefPubMedPubMedCentral
6.
Zurück zum Zitat de Klerk JCA, Engbers AGJ, van Beek F, Flint RB, Reiss IKM, Völler S, et al. Spontaneous closure of the ductus arteriosus in preterm infants: a systematic review. Front Pediatr. 2020;8:541.CrossRefPubMedPubMedCentral de Klerk JCA, Engbers AGJ, van Beek F, Flint RB, Reiss IKM, Völler S, et al. Spontaneous closure of the ductus arteriosus in preterm infants: a systematic review. Front Pediatr. 2020;8:541.CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Völler S, Flint RB, Stolk LM, Degraeuwe PLJ, Simons SHP, Pokorna P, et al. Model-based clinical dose optimization for phenobarbital in neonates: an illustration of the importance of data sharing and external validation. Eur J Pharm Sci. 2017;109S:S90-97.CrossRefPubMed Völler S, Flint RB, Stolk LM, Degraeuwe PLJ, Simons SHP, Pokorna P, et al. Model-based clinical dose optimization for phenobarbital in neonates: an illustration of the importance of data sharing and external validation. Eur J Pharm Sci. 2017;109S:S90-97.CrossRefPubMed
8.
Zurück zum Zitat Völler S, Flint RB, Andriessen P, Allegaert K, Zimmermann LJI, Liem KD, et al. Rapidly maturing fentanyl clearance in preterm neonates. Arch Dis Child Fetal Neonatal Ed. 2019;104(6):F598–603.CrossRefPubMed Völler S, Flint RB, Andriessen P, Allegaert K, Zimmermann LJI, Liem KD, et al. Rapidly maturing fentanyl clearance in preterm neonates. Arch Dis Child Fetal Neonatal Ed. 2019;104(6):F598–603.CrossRefPubMed
10.
Zurück zum Zitat De Cock RFW, Allegaert K, Brussee JM, Sherwin CMT, Mulla H, De Hoog M, et al. Simultaneous pharmacokinetic modeling of gentamicin, tobramycin and vancomycin clearance from neonates to adults: towards a semi-physiological function for maturation in glomerular filtration. Pharm Res. 2014;31(10):2643–54.CrossRefPubMedPubMedCentral De Cock RFW, Allegaert K, Brussee JM, Sherwin CMT, Mulla H, De Hoog M, et al. Simultaneous pharmacokinetic modeling of gentamicin, tobramycin and vancomycin clearance from neonates to adults: towards a semi-physiological function for maturation in glomerular filtration. Pharm Res. 2014;31(10):2643–54.CrossRefPubMedPubMedCentral
11.
Zurück zum Zitat Janssen EJH, Välitalo PAJ, Allegaert K, de Cock RFW, Simons SHP, Sherwin CMT, et al. Towards rational dosing algorithms for vancomycin in neonates and infants based on population pharmacokinetic modeling. Antimicrob Agents Chemother. 2015;60(2):1013–21.CrossRefPubMed Janssen EJH, Välitalo PAJ, Allegaert K, de Cock RFW, Simons SHP, Sherwin CMT, et al. Towards rational dosing algorithms for vancomycin in neonates and infants based on population pharmacokinetic modeling. Antimicrob Agents Chemother. 2015;60(2):1013–21.CrossRefPubMed
12.
Zurück zum Zitat Salem F, Johnson TN, Hodgkinson ABJ, Ogungbenro K, Rostami-Hodjegan A. Does, “Birth” as an event impact maturation trajectory of renal clearance via glomerular filtration? Reexamining data in preterm and full-term neonates by avoiding the creatinine bias. J Clin Pharmacol. 2021;61(2):159–71.CrossRefPubMed Salem F, Johnson TN, Hodgkinson ABJ, Ogungbenro K, Rostami-Hodjegan A. Does, “Birth” as an event impact maturation trajectory of renal clearance via glomerular filtration? Reexamining data in preterm and full-term neonates by avoiding the creatinine bias. J Clin Pharmacol. 2021;61(2):159–71.CrossRefPubMed
13.
Zurück zum Zitat Lacroix D, Sonnier M, Moncion A, Cheron G, Cresteil T. Expression of CYP3A in the human liver—evidence that the shift between CYP3A7 and CYP3A4 occurs immediately after birth. Eur J Biochem. 1997;247(2):625–34.CrossRefPubMed Lacroix D, Sonnier M, Moncion A, Cheron G, Cresteil T. Expression of CYP3A in the human liver—evidence that the shift between CYP3A7 and CYP3A4 occurs immediately after birth. Eur J Biochem. 1997;247(2):625–34.CrossRefPubMed
Metadaten
Titel
Comment on: "Preterm Physiologically Based Pharmacokinetic Model, Part I and Part II”
verfasst von
Swantje Völler
Robert B. Flint
Sinno H. P. Simons
Catherijne A. J. Knibbe
Publikationsdatum
13.03.2021
Verlag
Springer International Publishing
Erschienen in
Clinical Pharmacokinetics / Ausgabe 5/2021
Print ISSN: 0312-5963
Elektronische ISSN: 1179-1926
DOI
https://doi.org/10.1007/s40262-021-00993-4

Weitere Artikel der Ausgabe 5/2021

Clinical Pharmacokinetics 5/2021 Zur Ausgabe